Guidelines For Basic Science Open Access
Copyright ©2009 Baishideng. All rights reserved.
World J Hepatol. Oct 31, 2009; 1(1): 17-27
Published online Oct 31, 2009. doi: 10.4254/wjh.v1.i1.17
Activins and follistatins: Emerging roles in liver physiology and cancer
Emanuel Kreidl, Deniz Öztürk, Thomas Metzner, Walter Berger, Michael Grusch, Institute of Cancer Research, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, Vienna A-1090, Austria
Author contributions: Kreidl E, Öztürk D, Metzner T and Grusch M performed the literature search; Kreidl E and Grusch M wrote the manuscript and prepared the figures; Öztürk D, Metzner T and Berger W read, discussed and improved the manuscript.
Supported by Herzfelder Foundation
Correspondence to: Michael Grusch, PhD, Institute of Cancer Research, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, Vienna A-1090, Austria. michael.grusch@meduniwien.ac.at
Telephone: +43-1-427765144 Fax: +43-1-42779651
Received: June 16, 2009
Revised: September 10, 2009
Accepted: September 17, 2009
Published online: October 31, 2009

Abstract

Activins are secreted proteins belonging to the TGF-β family of signaling molecules. Activin signals are crucial for differentiation and regulation of cell proliferation and apoptosis in multiple tissues. Signal transduction by activins relies mainly on the Smad pathway, although the importance of crosstalk with additional pathways is increasingly being recognized. Activin signals are kept in balance by antagonists at multiple levels of the signaling cascade. Among these, follistatin and FLRG, two members of the emerging family of follistatin-like proteins, can bind secreted activins with high affinity, thereby blocking their access to cell surface-anchored activin receptors. In the liver, activin A is a major negative regulator of hepatocyte proliferation and can induce apoptosis. The functions of other activins expressed by hepatocytes have yet to be more clearly defined. Deregulated expression of activins and follistatin has been implicated in hepatic diseases including inflammation, fibrosis, liver failure and primary cancer. In particular, increased follistatin levels have been found in the circulation and in the tumor tissue of patients suffering from hepatocellular carcinoma as well as in animal models of liver cancer. It has been argued that up-regulation of follistatin protects neoplastic hepatocytes from activin-mediated growth inhibition and apoptosis. The use of follistatin as biomarker for liver tumor development is impeded, however, due to the presence of elevated follistatin levels already during preceding stages of liver disease. The current article summarizes our evolving understanding of the multi-faceted activities of activins and follistatins in liver physiology and cancer.

Key Words: Activin, Inhibin, Follistatin, Follistatin-like protein, Transforming growth factor β, Liver cancer



INTRODUCTION
The activin family

Activins are cytokines belonging to the TGF-β family of growth and differentiation factors[1] and were named according to their first identification as activators of follicle-stimulating hormone (FSH) release from pituitary cells[2,3]. Like TGF-β, activins are formed via the covalent dimerization of two subunits[4]. So far, five different subunits participating in the formation of activins have been identified. The subunits activin beta A, beta B, beta C and beta E were found in humans as well as other mammalian species, while activin beta D has only been identified in Xenopus laevis[5]. The four mammalian beta subunits are each encoded by a single gene, called INHBA, INHBB, INHBC and INHBE respectively[6]. INHBC and INHBE are closely linked in several species and are thought to have arisen from tandem duplication of an ancestral gene[7].

The different activin subunits can form homo- as well as heterodimers. A homodimer of two beta A subunits is called activin A, while a heterodimer of a beta A and a beta B subunit is called activin AB. The nomenclature for dimers of the other subunits follows the same scheme. While activins AB and AC have been described under physiological conditions in vivo[8,9], we and others have demonstrated the formation of activins AE, BC and CE after ectopic expression of the respective cDNAs in various cell lines[10-12]. Activin subunits are synthesized as pro-proteins of 350 to 426 amino acids[6]. The proteins are glycosylated in the pro-domain region, but addition of the carbohydrate group seems to be dispensable for secretion. This is in contrast to the related inhibin alpha subunit, a member of the TGF-β family and dimerization partner of activin subunits (see below)[13]. Dimers are created by intermolecular disulphide bond formation between the sixth of nine conserved cysteines in the mature proteins. The other cysteines are involved in the formation of intramolecular disulphide bonds, creating the so-called cysteine knot, typical for members of the TGF-β family and required for their biological activity[14].

Following dimerization, the protein is cleaved by pro-protein convertases of the subtilisin/kexin family in the ER and Golgi, producing a mature peptide chain of 115 or 116 amino acids. While the biologically active protein is secreted as a dimer of the mature peptides only, it has been suggested that the pro-region is required for correct folding, dimer formation and secretion[15]. Unprocessed, dimeric activin A was found to be biologically inactive[16]. Monomers have been reported to retain some affinity for the receptors of dimeric activin A but do not cause activation[17]. In addition to dimerization with another beta subunit, activin beta A and activin beta B can form heterodimers with the inhibin alpha subunit, giving rise to inhibins A and B, both inhibiting FSH release[18]. It remains uncertain if inhibin C exists, as there was evidence for the formation of a dimer between activin beta C and inhibin alpha in some[19] but not all reports[10].

Activin signal transduction

Like other members of the TGF-β family, activins are believed to signal via single-pass transmembrane receptors with an intracellular Ser-Thr kinase domain. This has been proven for activins A, B and AB. Activin A first binds to dimers of the type II receptors ActR-II (aka ACVR2) or ActR-IIB (aka ACVR2B), leading to the (preferential) recruitment and phosphorylation of dimers of the type I receptor ALK4 (aka ActR-IB/ACVR1B)[20]. While binding to the same type II receptors, activins B and AB preferentially recruit ALK7 (ACVR1C) as type I receptor[21]. Upon ligand binding, receptors are typically internalized[22]. It has been questioned however, if this internalization is generally necessary for signal transduction[23]. As a consequence of activation, receptor-regulated Smads (R-Smads) are recruited to the receptor complex and phosphorylated by the type I receptor. This process is supported by accessory proteins like SARA and the motor protein kinesin-1. Depending on the identity of the receptor, either Smad 2 and Smad 3 (ALK4, ALK5, ALK7) or Smad 1, Smad 5 and Smad 8 (ALK1, ALK2, ALK3, ALK6) are recruited and activated[4]. For TGF-β it has been shown that the ligand can recruit different type I receptors, activating different subsets of Smads depending on the cell type[24]. So far, activins have only been shown to signal through Smad 2 and Smad 3[25]. R-Smads then form complexes with the common mediator Smad 4 and translocate to the nucleus where, together with cofactors, they are directly involved in regulation of gene expression.

In addition, recent evidence suggests Smad independent signaling of activin A via MAP kinases ERK 1/2 and p38[26] as well as the phosphatidylinositol 3'-kinase (PI3K)/Akt pathway[27]. Rho and JNK were also found to be stimulated by activin A[28].

ACTIVINS IN HEPATIC FUNCTION AND DYSFUNCTION
Beta A and beta B

Activin A represents the most extensively investigated activin. Multiple biological functions of activin A in a variety of cells and tissues have been described, including involvement in mesoderm induction[29], stem cell biology[30], reproductive biology[31], erythroid differentiation[32], systemic inflammation[33], cell death induction[34], wound healing[35], and fibrosis[36]. Knock-out mice for activin beta A show severe defects in craniofacial development and die shortly after birth[37]. Activin A potently inhibits mitogen-induced DNA synthesis in the liver and induces hepatocyte apoptosis in vivo and in vitro[38-40]. Activin beta A antisense oligonucleotides stimulated cell proliferation in the human hepatoma cell line HLF suggesting a growth inhibitory function of endogenous activin A[41]. In regenerating liver, activin beta A gene expression was reduced at time points when hepatocyte replication took place and was increased at time points when liver regeneration terminated[42]. Other studies, however, have described increased expression of beta A at earlier time points after partial hepatectomy[43,44].

Beside its effects on DNA synthesis and cell growth, activin A also regulates restoration of liver architecture after partial hepatectomy by stimulating collagen production in hepatic stellate cells (HSC) and tubulogenesis of sinusoidal endothelial cells[45,46]. Stimulation of HSC may also contribute to liver fibrosis and several investigations have found elevated levels of activin beta A in fibrotic and cirrhotic rat livers[47-50]. In hepatocytes, activin A was also demonstrated to stimulate the expression of connective tissue growth factor (CCTF/CCN2), an important regulator of liver fibrosis[51]. Elevated levels of circulating activin A were found in patients with acute liver failure, chronic viral hepatitis, alcohol induced liver cirrhosis and hepatocellular carcinoma (HCC)[52-57]. Elevated serum activin A was also reported in a study with patients suffering from non-alcoholic fatty liver disease (NAFLD), with particularly high levels in the subgroup with non-alcoholic steatohepatitis (NASH)[58]. These patients also had an increased activin beta A/follistatin mRNA ratio in liver tissue. In the same study activin A was shown in Huh7 hepatoma cells to promote collagen III and TGF-β 1 expression, matrix metalloproteinase (MMP) activity, induce mitochondrial beta-oxidation and down-regulate fatty acid synthase (FAS) activity. Together these findings suggest an involvement of activin A not only in fibrosis but also in lipid accumulation. A study from our group in contrast, has found reduced expression of activin beta A transcripts in tumor tissue from chemically-induced rat liver tumors[59]. In addition to a pro-apoptotic and a pro-fibrotic effect, activin A has also been linked to hepatic neoangiogenesis via stimulation of VEGF expression in human hepatoma cells[60]. With respect to hepatic differentiation, it has been shown that a gradient of activin/TGF-β signaling controls differentiation of hepatoblasts into hepatocytes and biliary cells in the mouse, with high signaling activity required for development into biliary cells[61]. The contributing activin/TGF-β ligands, however, have not been fully identified. Several studies have used activin A as part of protocols to differentiate human embryonic stem cells (hESC) into hepatocyte-like cells[62-65].

Like activin beta A, the beta B subunit is expressed in multiple tissues and organs[11,66]. Knock-out mice for beta B are viable but show defects in eyelid development and female reproduction[67]. When the coding region of the mature peptide of the beta A subunit gene was replaced with the corresponding region of the beta B subunit, the developmental defects of the beta A knock-out mice were only partially rescued indicating differences in receptor activation or downstream signals[68]. In the liver, the function of the beta B subunit is not well characterized. One reason for this might be the low expression level in normal rat liver, where we observed the beta B subunit to be the only activin subunit undetectable by RNAse protection assay[11]. By immunohistochemistry, however, weak staining of beta B was detected in hepatocytes of normal rat livers and in connective tissue septa in fibrotic livers[47]. Activin beta B mRNA was induced in stellate cells of CCl4 treated rat livers[47] and exposure to the peroxisome proliferator di-n-butyl phthalate led to a transient surge of beta B mRNA expression 6 h after treatment[69]. With respect to biological activities, recombinant activin B, in contrast to activins A and AB, did not inhibit EGF induced DNA synthesis in primary rat hepatocytes[70]. In contrast to the rat, beta A and beta B transcripts are expressed to similar levels in human liver (Rodgarkia-Dara, unpublished observation). Ectopic expression of ALK7, the preferred type two receptor for activins B and AB induced apoptosis in hepatoma cell lines in a Smad and MAPK- dependent manner[71]. Both activin B and ALK7 have been linked to obesity and diabetes, two well-known risk factors for HCC, via participation in regulatory circuits in adipose tissue and the pancreas[72-74].

Beta C and beta E

In contrast to beta A and beta B, whose expression level is the highest in reproductive organs, the liver is the organ where the beta C and the beta E subunit reach by far their highest expression levels. The activin beta C subunit was cloned from liver cDNA and demonstrated to be predominantly expressed in hepatocytes by Northern blot analysis and RNAse protection assays[11,44,75,76]. By immunohistochemistry, significant activin beta C expression has been detected in cells from additional organs, including the prostate, ovary, testes, and pituitary gland[10,77]. After partial hepatectomy, a transient down-regulation of activin beta C expression was observed by several studies[42,44,78,79]. We have found reduced activin beta C expression in HepG2 and Hep3B hepatoma cells versus normal liver tissue[80] and a drop of beta C expression was also described in rat hepatocytes during primary culture with and without EGF treatment[44]. In contrast, increased activin beta C expression was reported in rat liver during the development of CCl4-induced cirrhosis[48,81] and in response to treatment with the peroxisome proliferator bi-n-butyl phthalate[69]. The functions of the activin beta C subunit are controversial. Activin beta C knock-out mice developed normally and liver regeneration after partial hepatectomy proceeded similar in knock-out animals and wild-type littermates[82]. Studies from our group showed that ectopic expression of activin beta C induced apoptosis in human (HepG2, Hep3B) and rat (H4IIEC3) hepatoma cells and delayed liver regeneration in mice[80,83]. In contrast, in AML12 cells, an immortalized mouse hepatocyte cell line, and in primary rat hepatocytes activin beta C increased DNA synthesis[84]. Adenovirus-mediated expression of activin beta C accelerated liver regeneration after partial hepatectomy in rats[85] and association of activin beta C immunoreactivity with mitotic hepatocytes was observed in regenerating liver after partial hepatectomy[42]. Activin C does not activate activin A-responsive promoters and it was suggested that the beta C subunit down-regulates the levels of bioactive activin A via the formation of signaling-incompetent activin AC heterodimers in PC3 human prostate cancer cells[9,86]. In a recent study from the same group, it was shown that homodimeric activin C inhibited activin A-induced Smad2 phosphorylation and growth inhibition, and that activin beta C transgenic mice develop prostate, testis and liver pathologies suggestive of an activin A antagonistic effect[87]. In line with these observations, elevated beta C immunoreactivity was found in human prostate, testis and liver cancers[87].

Like beta C, also the beta E subunit is highly expressed in the liver, but has been detected at lower levels in several other tissues as well[7,11,88,89]. In the liver of the developing mouse, activin beta E expression could not be detected until the very late stages of embryonic development and peaked at birth[82]. The biological functions and molecular interaction partners of activin beta E remain largely unknown. Like beta C, beta E knock-out (as well as beta C, beta E double knock-out) mice developed normally and showed no impairment of liver function or regeneration[82]. In vitro, overexpression of activin beta E in the human hepatoma cell lines HepG2 and Hep3B, as well as in the murine hepatocyte cell line AML12, caused decreased proliferation and induced apoptosis[12,80]. In vivo, transient overexpression of activin beta E inhibited regenerative DNA synthesis in mouse liver[83], while mice constitutively overexpressing the protein showed impaired growth of pancreatic exocrine cells[90]. Following partial hepatectomy, activin beta E mRNA increased rapidly and decreased to near-basal levels after 48 h[82]. We observed a diurnal variation of beta E mRNA depending on food consumption in the rat liver and a surge of beta E expression in response to bacterial lipopolysaccharide (LPS) stimulation was also described[1,89]. Additionally, beta E expression was found and confirmed to be elevated in HepG2 cells as a consequence of phospholipidosis, a lipid storage disorder[91,92]. Expression of activin beta E was also significantly increased in the lung following airway inflammation[93] and in brains of rats infected with Borna disease virus[94]. Interestingly, a neuronal component has also been implied by recent work describing reduced anxiety-related behavior in mice overexpressing activin beta E[95]. Overexpression of the tumor suppressor RASSF1A stimulated expression of beta E, while knock-down of endogenous RASSF1A in nasopharyngeal epithelial cells resulted in beta E downregulation[96]. Finally, in gene chip analysis, mRNA levels from INHBE were found to be altered in HepG2 in response to hypoxia[97]. One possible mode of action for activin beta E was described by Chow et al[96], who demonstrated that the expression of Inhibitor of DNA binding 2 (Id2) protein is down-regulated in response to overexpression of activin beta E. Id2 is a known target of TGF-β and a potential oncogene[98]. Large scale analysis identified mutations in the INHBE gene in breast cancer[99]. An evaluation of single nucleotide polymorphisms (SNPs) in genes coding for activins in testicular cancer showed a correlation for the risk of disease and mutations in INHBA but not in INHBB, INHBC or INHBE[100].

FOLLISTATINS AND THEIR ROLE IN ACTIVIN ANTAGONISM AND LIVER DISEASE

Follistatin was discovered as antagonist of activin activity with respect to FSH release from pituitary cells[101]. Sequence analysis of follistatin revealed no homology to the TGF-β family, but the presence of three domains with a similar architecture, namely 10 cysteines spaced in a conserved fashion resulting in a characteristic pattern of intramolecular disulphide bond formation[102]. Accordingly, this domain was termed follistatin domain and bears resemblance to the Kazal domain of serine proteinase inhibitors. Follistatin domains have been identified in a number of additional extracellular proteins and some of these have been filed as follistatin-related proteins or follistatin-like proteins[103-105] (Figure 1). The connection of follistatin and follistatin-like proteins with activin signaling and their involvement in hepatic functions is discussed below. Additional regulation of activin signal transduction takes place at the receptor level by co-receptors, such as cripto, nodal, betaglycan, or BAMBI and intracellularly, for instance by the inhibitory Smad 6 and 7, and has been reviewed elsewhere[106,107].

Figure 1
Figure 1 Number and arrangement of follistatin/Kazal-like domains in follistatin-like proteins. SP: Signal peptide; FS: Follistatin/Kazal-like domain; EF: EF-hand domain; VWFC: von Willebrand factor type C repeat; IGFBP: IGF-binding protein N-terminal domain; Ig: Immunoglobulin-like domain. Text to the right shows the number of amino acids (aa), most common name(s) and Uniprot accession number.
Follistatin

The biological activities described for follistatin seem to depend largely on its interaction with activins and other members of the TGF-β family. Follistatin, which is expressed in most organs expressing activins[66,108], binds mature secreted activin A with high affinity[109-111]. Complex formation with follistatin completely abolished receptor binding of activin A, thus blocking activin signaling[110,112]. Two follistatin molecules embrace one activin dimer and bury one-third of its residues and its receptor binding sites[113]. Alternative splicing and protein processing of a single follistatin gene results in secretion of three major isoforms containing 288, 303, and 315 amino acids[109]. Of the three follistatin domains present in all follistatin isoforms[114], the first two, but not the third, are necessary for activin A binding[111,115]. Follistatin 288 binds to heparan sulfates, whereas this binding is blocked by an acidic tail in follistatin 315[109]. In addition to binding activins A, B, AB, and E, follistatin was also shown to bind and antagonize myostatin as well as BMPs 2, 4, 6, and 7[88,116-119]. Follistatin administration by intraportal infusion or adenovirus-mediated overexpression caused DNA synthesis and liver growth in the rat, presumably by antagonizing tonic inhibition of hepatocyte proliferation by activin A[120,121]. Following partial hepatectomy, follistatin expression was up-regulated after 24-48 h, the time period in which hepatocyte replication was increased[42]. Administration of exogenous follistatin after partial hepatectomy accelerated liver regeneration but led to impaired restoration of normal tissue architecture and compromised liver function[122-124]. Administration of follistatin in CCl4-treated rats attenuated the formation of liver fibrosis[125]. These results likely reflect the ability of follistatin to antagonize both growth-inhibitory and pro-fibrotic activities of activin A.

In mouse and rat models of chemically induced liver tumors, we found follistatin expression to be up-regulated in about 60% of tumor tissue samples[59,126]. Moreover, we demonstrated that administration of follistatin stimulated DNA synthesis in preneoplastic rat hepatocytes in an ex vivo system, whereas hepatoma cell lines were unresponsive to exogenous follistatin possibly due to autocrine production of follistatin or other activin antagonists[59,126-128]. Knock-out mice for parkin, an E3 ubiquitin ligase implicated in Parkinson’s disease and frequently deleted in HCC and hepatoma cell lines[129], develop liver tumors in a follistatin upregulation-dependent fashion[130]. In human HCC elevated follistatin levels were found in the tumor tissue and the circulation of patients[53,59,131]. However, follistatin had no benefit as surveillance biomarker for HCC development in patients with alcoholic and non-alcoholic liver disease (ALD and NAFLD) due to the already elevated levels in the underlying liver pathologies[131]. Whether or not, interaction of follistatin with TGF-β family members other than activins (myostatin, BMP, GDFs) or with angiogenin[132] plays a role in liver tumorigenesis remains to be explored.

Follistatin-like proteins

Follistatin-like 1 (fstl1, also called follistatin-related protein, FRP or Tsc-36) contains only a single follistatin domain and no activin-binding activity has been reported. In fact, the interaction partners of fstl1 on a molecular level have not been identified and its function is far from clear. Fstl1 itself was identified as a TGF-β inducible gene[133] and has been implicated in inflammation and cardioprotection[134-136]. It has been suggested to act as a potential tumor suppressor in epithelial cancers[137-140] but is over-expressed in astrocytic brain tumors[141]. Considering hepatoma cells, we recently demonstrated that the expression of fstl1 is low in HepG2 cells, which show an epithelial morphology/proteome pattern and high in Hep3B cells with fibroblastoid characteristics. These observations suggest fstl1 as potential indicator of epithelial-mesenchymal transition (EMT)[142].

The term follistatin-like 2 (fstl2) is only rarely used. It refers to a protein described to have IGF- (insulin-like growth factor) as well as activin-binding activity and sequence homology with follistatin[143,144]. This protein was also termed mac25 and angiomodulin but is better known as IGFBP7 (IGF binding protein 7) or IGFBP-rP1 (IGF binding protein-related protein 1)[145]. It has been suggested to act as tumor suppressor, because its expression is reduced in neoplastic tissues of different cancer types including liver tumors from SV40T/t antigen transgenic mice[146]. However, the biological relevance of IGFBP7 binding to activin is still unclear. In the course of evolution fish went through whole genome duplication and the term fstl2 has also been used (synonymously with fstl1b) to denote the second zebrafish orthologue of mammalian fstl1.

Among the follistatin like proteins follistatin-like 3 (fstl3), encoded by follistatin-related gene (FLRG), has the highest overall similarity with follistatin and shares its ability to bind TGF-β family proteins, but contains only two instead of three follistatin domains[147]. The FLRG gene was originally identified as a target of chromosomal rearrangement in leukemia[148]. The highest tissue expression of FLRG was found in placenta, whereas highest follistatin expression was found in ovary, testis, and pituitary[147,149]. In HepG2 hepatoma cells, expression of both FLRG and follistatin was induced in response to activin A treatment suggesting that they participate in a feedback loop to restrict activin A signals[150]. FLRG knock-out mice developed increased pancreatic islet number and size, beta cell hyperplasia, decreased visceral fat mass, and hepatic steatosis. This is in line with a physiological role of fstl3 in antagonizing activin and myostatin activity in the pancreas, adipose tissue and liver[151]. Elevated expression of FLRG has been linked to breast cancer[152] and we have found increased FLRG transcript levels in chemically induced rat liver tumors but not in human liver tumor specimens[59].

Recently follistatin-like 4 and 5 were identified as two additional follistatin-related proteins[153], but their expression pattern and function have yet to be worked out. In addition to the follistatin-like proteins, SPARC, agrin, tomoregulin and others contain one or more follistatin domains, but none of these have so far been connected to activin signal transduction[103,104].

CONCLUSION

Despite the apparent gaps in our knowledge, it is becoming increasingly clear that tightly regulated activin signals are of fundamental importance for the maintenance of liver architecture and cellular homeostasis. While still much has to be learned, especially about the less explored members of the activin and follistatin families, the pace of progress has appreciably sped up in recent years. Deregulated expression of activin A and/or follistatin has been consistently observed in liver cancer in human patients and in a growing number of animal models, and was shown to causally contribute to the inflammatory and fibrotic conditions that promote carcinogenesis (Figure 2). The picture that emerges is that inflammation-associated elevated activin A levels contribute to fibrotic tissue remodelling and cell death of normal hepatocytes, whereas preneoplastic and neoplastic hepatocytes become resistant to activin A-induced growth control, at least in part through overexpression of follistatin. Conditional and liver cell type-specific knock-out of activin beta A and follistatin in mouse hepatocarcinogenesis models could shed further light on the contribution of the activin-follistatin axis to liver cancer development. For the two activin subunits with predominant expression in hepatocytes, namely beta C and beta E, as well as for fstl1, 4 and 5 future efforts should be directed at elucidating their molecular interaction with cell surface receptors or secreted proteins as a prerequisite to better understand their biological activities. Although the complexity of the system may sometimes seem daunting, the hope is well founded that in the not-too-far future, the increasing knowledge on activins and follistatins will translate into improved diagnostic or therapeutic opportunities for patients suffering from chronic liver disease and HCC.

Figure 2
Figure 2 Reported effects of activin A on liver cells and inhibition by activin and follistatin family members. A potential function of the other family members, as agonists or antagonists of hepatic activin signal transduction or as regulators of activin A-independent activities in the liver, remains to be elucidated. ECM: Extracellular matrix; MMP: Matrix metalloproteinase.
Footnotes

Peer reviewer: Inci Alican, MD, PhD, Professor of Physiology, Marmara University School of Medicine, Department of Physiology, 34668, Haydarpasa, Istanbul, Turkey

S- Editor Zhang HN L- Editor Negro F E- Editor Ma WH

References
1.  Rodgarkia-Dara C, Vejda S, Erlach N, Losert A, Bursch W, Berger W, Schulte-Hermann R, Grusch M. The activin axis in liver biology and disease. Mutat Res. 2006;613:123-137.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Ling N, Ying SY, Ueno N, Shimasaki S, Esch F, Hotta M, Guillemin R. A homodimer of the beta-subunits of inhibin A stimulates the secretion of pituitary follicle stimulating hormone. Biochem Biophys Res Commun. 1986;138:1129-1137.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Ling N, Ying SY, Ueno N, Shimasaki S, Esch F, Hotta M, Guillemin R. Pituitary FSH is released by a heterodimer of the beta-subunits from the two forms of inhibin. Nature. 1986;321:779-782.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Schmierer B, Hill CS. TGFbeta-SMAD signal transduction: molecular specificity and functional flexibility. Nat Rev Mol Cell Biol. 2007;8:970-982.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Oda S, Nishimatsu S, Murakami K, Ueno N. Molecular cloning and functional analysis of a new activin beta subunit: a dorsal mesoderm-inducing activity in Xenopus. Biochem Biophys Res Commun. 1995;210:581-588.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Grusch M, Rodgarkia-Dara C, Bursch W, Schulte-Hermann R. Activins and the liver. TGF-β Superfamily Members in Normal and Tumor Biology. Totowa: Humana Press 2008; .  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Fang J, Wang SQ, Smiley E, Bonadio J. Genes coding for mouse activin beta C and beta E are closely linked and exhibit a liver-specific expression pattern in adult tissues. Biochem Biophys Res Commun. 1997;231:655-661.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Evans LW, Muttukrishna S, Knight PG, Groome NP. Development, validation and application of a two-site enzyme-linked immunosorbent assay for activin-AB. J Endocrinol. 1997;153:221-230.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Mellor SL, Ball EM, O’Connor AE, Ethier JF, Cranfield M, Schmitt JF, Phillips DJ, Groome NP, Risbridger GP. Activin betaC-subunit heterodimers provide a new mechanism of regulating activin levels in the prostate. Endocrinology. 2003;144:4410-4419.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Mellor SL, Cranfield M, Ries R, Pedersen J, Cancilla B, de Kretser D, Groome NP, Mason AJ, Risbridger GP. Localization of activin beta(A)-, beta(B)-, and beta(C)-subunits in humanprostate and evidence for formation of new activin heterodimers of beta(C)-subunit. J Clin Endocrinol Metab. 2000;85:4851-4858.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Vejda S, Cranfield M, Peter B, Mellor SL, Groome N, Schulte-Hermann R, Rossmanith W. Expression and dimerization of the rat activin subunits betaC and betaE: evidence for the ormation of novel activin dimers. J Mol Endocrinol. 2002;28:137-148.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Wada W, Medina JJ, Kuwano H, Kojima I. Comparison of the function of the beta(C) and beta(E) subunits of activin in AML12 hepatocytes. Endocr J. 2005;52:169-175.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Antenos M, Stemler M, Boime I, Woodruff TK. N-linked oligosaccharides direct the differential assembly and secretion of inhibin alpha- and betaA-subunit dimers. Mol Endocrinol. 2007;21:1670-1684.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Mason AJ. Functional analysis of the cysteine residues of activin A. Mol Endocrinol. 1994;8:325-332.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Gray AM, Mason AJ. Requirement for activin A and transforming growth factor--beta 1 pro-regions in homodimer assembly. Science. 1990;247:1328-1330.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Mason AJ, Farnworth PG, Sullivan J. Characterization and determination of the biological activities of noncleavable high molecular weight forms of inhibin A and activin A. Mol Endocrinol. 1996;10:1055-1065.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Husken-Hindi P, Tsuchida K, Park M, Corrigan AZ, Vaughan JM, Vale WW, Fischer WH. Monomeric activin A retains high receptor binding affinity but exhibits low biological activity. J Biol Chem. 1994;269:19380-19384.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Mason AJ, Hayflick JS, Ling N, Esch F, Ueno N, Ying SY, Guillemin R, Niall H, Seeburg PH. Complementary DNA sequences of ovarian follicular fluid inhibin show precursor structure and homology with transforming growth factor-beta. Nature. 1985;318:659-663.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Ushiro Y, Hashimoto O, Seki M, Hachiya A, Shoji H, Hasegawa Y. Analysis of the function of activin betaC subunit using recombinant protein. J Reprod Dev. 2006;52:487-495.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Attisano L, Wrana JL, Montalvo E, Massague J. Activation of signalling by the activin receptor complex. Mol Cell Biol. 1996;16:1066-1073.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Tsuchida K, Nakatani M, Yamakawa N, Hashimoto O, Hasegawa Y, Sugino H. Activin isoforms signal through type I receptor serine/threonine kinase ALK7. Mol Cell Endocrinol. 2004;220:59-65.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Dore JJ Jr, Yao D, Edens M, Garamszegi N, Sholl EL, Leof EB. Mechanisms of transforming growth factor-beta receptor endocytosis and intracellular sorting differ between fibroblasts and epithelial cells. Mol Biol Cell. 2001;12:675-684.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Zhou Y, Scolavino S, Funderburk SF, Ficociello LF, Zhang X, Klibanski A. Receptor internalization-independent activation of Smad2 in activin signaling. Mol Endocrinol. 2004;18:1818-1826.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Lee NY, Ray B, How T, Blobe GC. Endoglin promotes transforming growth factor beta-mediated Smad 1/5/8 signaling and inhibits endothelial cell migration through its association with GIPC. J Biol Chem. 2008;283:32527-32533.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Harrison CA, Gray PC, Fischer WH, Donaldson C, Choe S, Vale W. An activin mutant with disrupted ALK4 binding blocks signaling via type II receptors. J Biol Chem. 2004;279:28036-28044.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Murase Y, Okahashi N, Koseki T, Itoh K, Udagawa N, Hashimoto O, Sugino H, Noguchi T, Nishihara T. Possible involvement of protein kinases and Smad2 signaling pathways on osteoclast differentiation enhanced by activin A. J Cell Physiol. 2001;188:236-242.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Dupont J, McNeilly J, Vaiman A, Canepa S, Combarnous Y, Taragnat C. Activin signaling pathways in ovine pituitary and LbetaT2 gonadotrope cells. Biol Reprod. 2003;68:1877-1887.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Zhang L, Deng M, Parthasarathy R, Wang L, Mongan M, Molkentin JD, Zheng Y, Xia Y. MEKK1 transduces activin signals in keratinocytes to induce actin stress fiber formation and migration. Mol Cell Biol. 2005;25:60-65.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  McDowell N, Gurdon JB. Activin as a morphogen in Xenopus mesoderm induction. Semin Cell Dev Biol. 1999;10:311-317.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Beattie GM, Lopez AD, Bucay N, Hinton A, Firpo MT, King CC, Hayek A. Activin A maintains pluripotency of human embryonic stem cells in the absence of feeder layers. Stem Cells. 2005;23:489-495.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  de Kretser DM, Hedger MP, Loveland KL, Phillips DJ. Inhibins, activins and follistatin in reproduction. Hum Reprod Update. 2002;8:529-541.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Maguer-Satta V, Bartholin L, Jeanpierre S, Ffrench M, Martel S, Magaud JP, Rimokh R. Regulation of human erythropoiesis by activin A, BMP2, and BMP4, members of the TGFbeta family. Exp Cell Res. 2003;282:110-120.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Jones KL, de Kretser DM, Patella S, Phillips DJ. Activin A and follistatin in systemic inflammation. Mol Cell Endocrinol. 2004;225:119-125.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Chen YG, Wang Q, Lin SL, Chang CD, Chuang J, Ying SY. Activin signaling and its role in regulation of cell proliferation, apoptosis, and carcinogenesis. Exp Biol Med (Maywood). 2006;231:534-544.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Munz B, Smola H, Engelhardt F, Bleuel K, Brauchle M, Lein I, Evans LW, Huylebroeck D, Balling R, Werner S. Overexpression of activin A in the skin of transgenic mice reveals new activities of activin in epidermal morphogenesis, dermal fibrosis and wound repair. EMBO J. 1999;18:5205-5215.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Werner S, Alzheimer C. Roles of activin in tissue repair, fibrosis, and inflammatory disease. Cytokine Growth Factor Rev. 2006;17:157-171.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Matzuk MM, Kumar TR, Vassalli A, Bickenbach JR, Roop DR, Jaenisch R, Bradley A. Functional analysis of activins during mammalian development. Nature. 1995;374:354-356.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Hully JR, Chang L, Schwall RH, Widmer HR, Terrell TG, Gillett NA. Induction of apoptosis in the murine liver with recombinant human activin A. Hepatology. 1994;20:854-862.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Schwall RH, Robbins K, Jardieu P, Chang L, Lai C, Terrell TG. Activin induces cell death in hepatocytes in vivo and in vitro. Hepatology. 1993;18:347-356.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Yasuda H, Mine T, Shibata H, Eto Y, Hasegawa Y, Takeuchi T, Asano S, Kojima I. Activin A: an autocrine inhibitor of initiation of DNA synthesis in rat hepatocytes. J Clin Invest. 1993;92:1491-1496.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Takabe K, Lebrun JJ, Nagashima Y, Ichikawa Y, Mitsuhashi M, Momiyama N, Ishikawa T, Shimada H, Vale WW. Interruption of activin A autocrine regulation by antisense oligodeoxynucleotides accelerates liver tumor cell proliferation. Endocrinology. 1999;140:3125-3132.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Gold EJ, Zhang X, Wheatley AM, Mellor SL, Cranfield M, Risbridger GP, Groome NP, Fleming JS. betaA- and betaC-activin, follistatin, activin receptor mRNA and betaC-activin peptide expression during rat liver regeneration. J Mol Endocrinol. 2005;34:505-515.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Date M, Matsuzaki K, Matsushita M, Tahashi Y, Sakitani K, Inoue K. Differential regulation of activin A for hepatocyte growth and fibronectin synthesis in rat liver injury. J Hepatol. 2000;32:251-260.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Zhang YQ, Shibata H, Schrewe H, Kojima I. Reciprocal expression of mRNA for inhibin betaC and betaA subunits in hepatocytes. Endocr J. 1997;44:759-764.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Wada W, Kuwano H, Hasegawa Y, Kojima I. The dependence of transforming growth factor-beta-induced collagen production on autocrine factor activin A in hepatic stellate cells. Endocrinology. 2004;145:2753-2759.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Endo D, Kogure K, Hasegawa Y, Maku-uchi M, Kojima I. Activin A augments vascular endothelial growth factor activity in promoting branching tubulogenesis in hepatic sinusoidal endothelial cells. J Hepatol. 2004;40:399-404.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  De Bleser PJ, Niki T, Xu G, Rogiers V, Geerts A. Localization and cellular sources of activins in normal and fibrotic rat liver. Hepatology. 1997;26:905-912.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Gold EJ, Francis RJ, Zimmermann A, Mellor SL, Cranfield M, Risbridger GP, Groome NP, Wheatley AM, Fleming JS. Changes in activin and activin receptor subunit expression in rat liver during the development of CCl4-induced cirrhosis. Mol Cell Endocrinol. 2003;201:143-153.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Huang X, Li DG, Wang ZR, Wei HS, Cheng JL, Zhan YT, Zhou X, Xu QF, Li X, Lu HM. Expression changes of activin A in the development of hepatic fibrosis. World J Gastroenterol. 2001;7:37-41.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Sugiyama M, Ichida T, Sato T, Ishikawa T, Matsuda Y, Asakura H. Expression of activin A is increased in cirrhotic and fibrotic rat livers. Gastroenterology. 1998;114:550-558.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Gressner OA, Lahme B, Siluschek M, Rehbein K, Weiskirchen R, Gressner AM. Intracrine signalling of activin A in hepatocytes upregulates connective tissue growth factor (CTGF/CCN2) expression. Liver Int. 2008;28:1207-1216.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Patella S, Phillips DJ, de Kretser DM, Evans LW, Groome NP, Sievert W. Characterization of serum activin-A and follistatin and their relation to virological and histological determinants in chronic viral hepatitis. J Hepatol. 2001;34:576-583.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Yuen MF, Norris S, Evans LW, Langley PG, Hughes RD. Transforming growth factor-beta 1, activin and follistatin in patients with hepatocellular carcinoma and patients with alcoholic cirrhosis. Scand J Gastroenterol. 2002;37:233-238.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Pirisi M, Fabris C, Luisi S, Santuz M, Toniutto P, Vitulli D, Federico E, Del Forno M, Mattiuzzo M, Branca B. Evaluation of circulating activin-A as a serum marker of hepatocellular carcinoma. Cancer Detect Prev. 2000;24:150-155.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Elsammak MY, Amin GM, Khalil GM, Ragab WS, Abaza MM. Possible contribution of serum activin A and IGF-1 in the development of hepatocellular carcinoma in Egyptian patients suffering from combined hepatitis C virus infection and hepatic schistosomiasis. Clin Biochem. 2006;39:623-629.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Hughes RD, Evans LW. Activin A and follistatin in acute liver failure. Eur J Gastroenterol Hepatol. 2003;15:127-131.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Lin SD, Kawakami T, Ushio A, Sato A, Sato S, Iwai M, Endo R, Takikawa Y, Suzuki K. Ratio of circulating follistatin and activin A reflects the severity of acute liver injury and prognosis in patients with acute liver failure. J Gastroenterol Hepatol. 2006;21:374-380.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Yndestad A, Haukeland JW, Dahl TB, Bjoro K, Gladhaug IP, Berge C, Damas JK, Haaland T, Loberg EM, Linnestad P. A complex role of activin A in non-alcoholic fatty liver disease. Am J Gastroenterol. 2009;104:2196-2205.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Grusch M, Drucker C, Peter-Vorosmarty B, Erlach N, Lackner A, Losert A, Macheiner D, Schneider WJ, Hermann M, Groome NP. Deregulation of the activin/follistatin system in hepatocarcinogenesis. J Hepatol. 2006;45:673-680.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Wagner K, Peters M, Scholz A, Benckert C, Ruderisch HS, Wiedenmann B, Rosewicz S. Activin A stimulates vascular endothelial growth factor gene transcription in human hepatocellular carcinoma cells. Gastroenterology. 2004;126:1828-1843.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Clotman F, Jacquemin P, Plumb-Rudewiez N, Pierreux CE, Van der Smissen P, Dietz HC, Courtoy PJ, Rousseau GG, Lemaigre FP. Control of liver cell fate decision by a gradient of TGF beta signaling modulated by Onecut transcription factors. Genes Dev. 2005;19:1849-1854.  [PubMed]  [DOI]  [Cited in This Article: ]
62.  Basma H, Soto-Gutierrez A, Yannam GR, Liu L, Ito R, Yamamoto T, Ellis E, Carson SD, Sato S, Chen Y. Differentiation and transplantation of human embryonic stem cell-derived hepatocytes. Gastroenterology. 2009;136:990-999.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Cai J, Zhao Y, Liu Y, Ye F, Song Z, Qin H, Meng S, Chen Y, Zhou R, Song X. Directed differentiation of human embryonic stem cells into functional hepatic cells. Hepatology. 2007;45:1229-1239.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Hay DC, Fletcher J, Payne C, Terrace JD, Gallagher RC, Snoeys J, Black JR, Wojtacha D, Samuel K, Hannoun Z. Highly efficient differentiation of hESCs to functional hepatic endoderm requires ActivinA and Wnt3a signaling. Proc Natl Acad Sci USA. 2008;105:12301-1236.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Hay DC, Zhao D, Fletcher J, Hewitt ZA, McLean D, Urruticoechea-Uriguen A, Black JR, Elcombe C, Ross JA, Wolf R. Efficient differentiation of hepatocytes from human embryonic stem cells exhibiting markers recapitulating liver development in vivo. Stem Cells. 2008;26:894-902.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Tuuri T, Eramaa M, Hilden K, Ritvos O. The tissue distribution of activin beta A- and beta B-subunit and follistatin messenger ribonucleic acids suggests multiple sites of action for the activin-follistatin system during human development. J Clin Endocrinol Metab. 1994;78:1521-1524.  [PubMed]  [DOI]  [Cited in This Article: ]
67.  Vassalli A, Matzuk MM, Gardner HA, Lee KF, Jaenisch R. Activin/inhibin beta B subunit gene disruption leads to defects in eyelid development and female reproduction. Genes Dev. 1994;8:414-427.  [PubMed]  [DOI]  [Cited in This Article: ]
68.  Brown CW, Houston-Hawkins DE, Woodruff TK, Matzuk MM. Insertion of Inhbb into the Inhba locus rescues the Inhba-null phenotype and reveals new activin functions. Nat Genet. 2000;25:453-457.  [PubMed]  [DOI]  [Cited in This Article: ]
69.  Kobayashi T, Niimi S, Fukuoka M, Hayakawa T. Regulation of inhibin beta chains and follistatin mRNA levels during rat hepatocyte growth induced by the peroxisome proliferator di-n-butyl phthalate. Biol Pharm Bull. 2002;25:1214-1216.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  Niimi S, Horikawa M, Seki T, Ariga T, Kobayashi T, Hayakawa T. Effect of activins AB and B on DNA synthesis stimulated by epidermal growth factor in primary cultured rat hepatocytes. Biol Pharm Bull. 2002;25:437-440.  [PubMed]  [DOI]  [Cited in This Article: ]
71.  Kim BC, van Gelder H, Kim TA, Lee HJ, Baik KG, Chun HH, Lee DA, Choi KS, Kim SJ. Activin receptor-like kinase-7 induces apoptosis through activation of MAPKs in a Smad3-dependent mechanism in hepatoma cells. J Biol Chem. 2004;279:28458-28465.  [PubMed]  [DOI]  [Cited in This Article: ]
72.  Bertolino P, Holmberg R, Reissmann E, Andersson O, Berggren PO, Ibanez CF. Activin B receptor ALK7 is a negative regulator of pancreatic beta-cell function. Proc Natl Acad Sci USA. 2008;105:7246-7251.  [PubMed]  [DOI]  [Cited in This Article: ]
73.  Carlsson LM, Jacobson P, Walley A, Froguel P, Sjostrom L, Svensson PA, Sjoholm K. ALK7 expression is specific for adipose tissue, reduced in obesity and correlates to factors implicated in metabolic disease. Biochem Biophys Res Commun. 2009;382:309-314.  [PubMed]  [DOI]  [Cited in This Article: ]
74.  Sjoholm K, Palming J, Lystig TC, Jennische E, Woodruff TK, Carlsson B, Carlsson LM. The expression of inhibin beta B is high in human adipocytes, reduced by weight loss, and correlates to factors implicated in metabolic disease. Biochem Biophys Res Commun. 2006;344:1308-1314.  [PubMed]  [DOI]  [Cited in This Article: ]
75.  Hoetten G, Neidhardt H, Schneider C, Pohl J. Cloning of a new member of the TGF-beta family: a putative new activin beta C chain. Biochem Biophys Res Commun. 1995;206:608-613.  [PubMed]  [DOI]  [Cited in This Article: ]
76.  Schmitt J, Hotten G, Jenkins NA, Gilbert DJ, Copeland NG, Pohl J, Schrewe H. Structure, chromosomal localization, and expression analysis of the mouse inhibin/activin beta C (Inhbc) gene. Genomics. 1996;32:358-366.  [PubMed]  [DOI]  [Cited in This Article: ]
77.  Gold EJ, O’Bryan MK, Mellor SL, Cranfield M, Risbridger GP, Groome NP, Fleming JS. Cell-specific expression of betaC-activin in the rat reproductive tract, adrenal and liver. Mol Cell Endocrinol. 2004;222:61-69.  [PubMed]  [DOI]  [Cited in This Article: ]
78.  Esquela AF, Zimmers TA, Koniaris LG, Sitzmann JV, Lee SJ. Transient down-regulation of inhibin-betaC expression following partial hepatectomy. Biochem Biophys Res Commun. 1997;235:553-556.  [PubMed]  [DOI]  [Cited in This Article: ]
79.  Takamura K, Tsuchida K, Miyake H, Tashiro S, Sugino H. Activin and activin receptor expression changes in liver regeneration in rat. J Surg Res. 2005;126:3-11.  [PubMed]  [DOI]  [Cited in This Article: ]
80.  Vejda S, Erlach N, Peter B, Drucker C, Rossmanith W, Pohl J, Schulte-Hermann R, Grusch M. Expression of activins C and E induces apoptosis in human and rat hepatoma cells. Carcinogenesis. 2003;24:1801-1809.  [PubMed]  [DOI]  [Cited in This Article: ]
81.  Huang X, Li D, Lu H, Wang Z, Wei H, Wang Y, Zhang J, Xu Q. [Expression of activins, follistatin mRNA in the development of hepatic fibrosis]. Zhonghua Ganzangbing Zazhi. 2002;10:85-88.  [PubMed]  [DOI]  [Cited in This Article: ]
82.  Lau AL, Kumar TR, Nishimori K, Bonadio J, Matzuk MM. Activin betaC and betaE genes are not essential for mouse liver growth, differentiation, and regeneration. Mol Cell Biol. 2000;20:6127-6137.  [PubMed]  [DOI]  [Cited in This Article: ]
83.  Chabicovsky M, Herkner K, Rossmanith W. Overexpression of activin beta(C) or activin beta(E) in the mouse liver inhibits regenerative deoxyribonucleic acid synthesis of hepatic cells. Endocrinology. 2003;144:3497-3504.  [PubMed]  [DOI]  [Cited in This Article: ]
84.  Wada W, Maeshima A, Zhang YQ, Hasegawa Y, Kuwano H, Kojima I. Assessment of the function of the betaC-subunit of activin in cultured hepatocytes. Am J Physiol Endocrinol Metab. 2004;287:E247-E54.  [PubMed]  [DOI]  [Cited in This Article: ]
85.  Wada W, Medina J, Hasegawa Y, Kuwano H, Kojima I. Adenovirus-mediated overexpression of the activin betaC subunit accelerates liver regeneration in partially hepatectomized rats. J Hepatol. 2005;43:823-828.  [PubMed]  [DOI]  [Cited in This Article: ]
86.  Butler CM, Gold EJ, Risbridger GP. Should activin betaC be more than a fading snapshot in the activin/TGFbeta family album? Cytokine Growth Factor Rev. 2005;16:377-385.  [PubMed]  [DOI]  [Cited in This Article: ]
87.  Gold E, Jetly N, O’Bryan MK, Meachem S, Srinivasan D, Behuria S, Sanchez-Partida LG, Woodruff T, Hedwards S, Wang H. Activin C antagonizes activin A in vitro and overexpression leads to pathologies in vivo. Am J Pathol. 2009;174:184-195.  [PubMed]  [DOI]  [Cited in This Article: ]
88.  Hashimoto O, Tsuchida K, Ushiro Y, Hosoi Y, Hoshi N, Sugino H, Hasegawa Y. cDNA cloning and expression of human activin betaE subunit. Mol Cell Endocrinol. 2002;194:117-122.  [PubMed]  [DOI]  [Cited in This Article: ]
89.  O’Bryan MK, Sebire KL, Gerdprasert O, Hedger MP, Hearn MT, de Kretser DM. Cloning and regulation of the rat activin betaE subunit. J Mol Endocrinol. 2000;24:409-418.  [PubMed]  [DOI]  [Cited in This Article: ]
90.  Hashimoto O, Ushiro Y, Sekiyama K, Yamaguchi O, Yoshioka K, Mutoh K, Hasegawa Y. Impaired growth of pancreatic exocrine cells in transgenic mice expressing human activin betaE subunit. Biochem Biophys Res Commun. 2006;341:416-424.  [PubMed]  [DOI]  [Cited in This Article: ]
91.  Atienzar F, Gerets H, Dufrane S, Tilmant K, Cornet M, Dhalluin S, Ruty B, Rose G, Canning M. Determination of phospholipidosis potential based on gene expression analysis in HepG2 cells. Toxicol Sci. 2007;96:101-114.  [PubMed]  [DOI]  [Cited in This Article: ]
92.  Sawada H, Takami K, Asahi S. A toxicogenomic approach to drug-induced phospholipidosis: analysis of its induction mechanism and establishment of a novel in vitro screening system. Toxicol Sci. 2005;83:282-292.  [PubMed]  [DOI]  [Cited in This Article: ]
93.  Rosendahl A, Checchin D, Fehniger TE, ten Dijke P, Heldin CH, Sideras P. Activation of the TGF-beta/activin-Smad2 pathway during allergic airway inflammation. Am J Respir Cell Mol Biol. 2001;25:60-68.  [PubMed]  [DOI]  [Cited in This Article: ]
94.  Nishino Y, Ooishi R, Kurokawa S, Fujino K, Murakami M, Madarame H, Hashimoto O, Sugiyama K, Funaba M. Gene expression of the TGF-beta family in rat brain infected with Borna disease virus. Microbes Infect. 2009;11:737-743.  [PubMed]  [DOI]  [Cited in This Article: ]
95.  Sekiyama K, Hashimoto O, Ushiro Y, Adachi C, Kikusui T, Tanemura K, Hasegawa Y. Abnormalities in aggression and anxiety in transgenic mice overexpressing activin E. Biochem Biophys Res Commun. 2009;385:319-323.  [PubMed]  [DOI]  [Cited in This Article: ]
96.  Chow LS, Lam CW, Chan SY, Tsao SW, To KF, Tong SF, Hung WK, Dammann R, Huang DP, Lo KW. Identification of RASSF1A modulated genes in nasopharyngeal carcinoma. Oncogene. 2006;25:310-316.  [PubMed]  [DOI]  [Cited in This Article: ]
97.  Fisher TS, Etages SD, Hayes L, Crimin K, Li B. Analysis of ARD1 function in hypoxia response using retroviral RNA interference. J Biol Chem. 2005;280:17749-17757.  [PubMed]  [DOI]  [Cited in This Article: ]
98.  Lasorella A, Uo T, Iavarone A. Id proteins at the cross-road of development and cancer. Oncogene. 2001;20:8326-8333.  [PubMed]  [DOI]  [Cited in This Article: ]
99.  Sjoblom T, Jones S, Wood LD, Parsons DW, Lin J, Barber TD, Mandelker D, Leary RJ, Ptak J, Silliman N. The consensus coding sequences of human breast and colorectal cancers. Science. 2006;314:268-274.  [PubMed]  [DOI]  [Cited in This Article: ]
100.  Purdue MP, Graubard BI, Chanock SJ, Rubertone MV, Erickson RL, McGlynn KA. Genetic variation in the inhibin pathway and risk of testicular germ cell tumors. Cancer Res. 2008;68:3043-3048.  [PubMed]  [DOI]  [Cited in This Article: ]
101.  Ueno N, Ling N, Ying SY, Esch F, Shimasaki S, Guillemin R. Isolation and partial characterization of follistatin: a single-chain Mr 35,000 monomeric protein that inhibits the release of follicle-stimulating hormone. Proc Natl Acad Sci USA. 1987;84:8282-8286.  [PubMed]  [DOI]  [Cited in This Article: ]
102.  Esch FS, Shimasaki S, Mercado M, Cooksey K, Ling N, Ying S, Ueno N, Guillemin R. Structural characterization of follistatin: a novel follicle-stimulating hormone release-inhibiting polypeptide from the gonad. Mol Endocrinol. 1987;1:849-855.  [PubMed]  [DOI]  [Cited in This Article: ]
103.  Phillips DJ, de Kretser DM. Follistatin: a multifunctional regulatory protein. Front Neuroendocrinol. 1998;19:287-322.  [PubMed]  [DOI]  [Cited in This Article: ]
104.  Ullman CG, Perkins SJ. The Factor I and follistatin domain families: the return of a prodigal son. Biochem J. 1997;326:939-941.  [PubMed]  [DOI]  [Cited in This Article: ]
105.  Patthy L, Nikolics K. Functions of agrin and agrin-related proteins. Trends Neurosci. 1993;16:76-81.  [PubMed]  [DOI]  [Cited in This Article: ]
106.  Deli A, Kreidl E, Santifaller S, Trotter B, Seir K, Berger W, Schulte-Hermann R, Rodgarkia-Dara C, Grusch M. Activins and activin antagonists in hepatocellular carcinoma. World J Gastroenterol. 2008;14:1699-1709.  [PubMed]  [DOI]  [Cited in This Article: ]
107.  Harrison CA, Gray PC, Vale WW, Robertson DM. Antagonists of activin signaling: mechanisms and potential biological applications. Trends Endocrinol Metab. 2005;16:73-78.  [PubMed]  [DOI]  [Cited in This Article: ]
108.  Michel U, Rao A, Findlay JK. Rat follistatin: ontogeny of steady-state mRNA levels in different tissues predicts organ-specific functions. Biochem Biophys Res Commun. 1991;180:223-230.  [PubMed]  [DOI]  [Cited in This Article: ]
109.  Sugino K, Kurosawa N, Nakamura T, Takio K, Shimasaki S, Ling N, Titani K, Sugino H. Molecular heterogeneity of follistatin, an activin-binding protein. Higher affinity of the carboxyl-terminal truncated forms for heparan sulfate proteoglycans on the ovarian granulosa cell. J Biol Chem. 1993;268:15579-15587.  [PubMed]  [DOI]  [Cited in This Article: ]
110.  Schneyer AL, Rzucidlo DA, Sluss PM, Crowley WF Jr. Characterization of unique binding kinetics of follistatin and activin or inhibin in serum. Endocrinology. 1994;135:667-674.  [PubMed]  [DOI]  [Cited in This Article: ]
111.  Harrington AE, Morris-Triggs SA, Ruotolo BT, Robinson CV, Ohnuma S, Hyvonen M. Structural basis for the inhibition of activin signalling by follistatin. EMBO J. 2006;25:1035-1045.  [PubMed]  [DOI]  [Cited in This Article: ]
112.  de Winter JP, ten Dijke P, de Vries CJ, van Achterberg TA, Sugino H, de Waele P, Huylebroeck D, Verschueren K, van den Eijnden-van Raaij AJ. Follistatins neutralize activin bioactivity by inhibition of activin binding to its type II receptors. Mol Cell Endocrinol. 1996;116:105-114.  [PubMed]  [DOI]  [Cited in This Article: ]
113.  Thompson TB, Lerch TF, Cook RW, Woodruff TK, Jardetzky TS. The structure of the follistatin:activin complex reveals antagonism of both type I and type II receptor binding. Dev Cell. 2005;9:535-543.  [PubMed]  [DOI]  [Cited in This Article: ]
114.  Shimasaki S, Koga M, Esch F, Cooksey K, Mercado M, Koba A, Ueno N, Ying SY, Ling N, Guillemin R. Primary structure of the human follistatin precursor and its genomic organization. Proc Natl Acad Sci USA. 1988;85:4218-4222.  [PubMed]  [DOI]  [Cited in This Article: ]
115.  Keutmann HT, Schneyer AL, Sidis Y. The role of follistatin domains in follistatin biological action. Mol Endocrinol. 2004;18:228-240.  [PubMed]  [DOI]  [Cited in This Article: ]
116.  Schneyer A, Schoen A, Quigg A, Sidis Y. Differential binding and neutralization of activins A and B by follistatin and follistatin like-3 (FSTL-3/FSRP/FLRG). Endocrinology. 2003;144:1671-1674.  [PubMed]  [DOI]  [Cited in This Article: ]
117.  Iemura S, Yamamoto TS, Takagi C, Uchiyama H, Natsume T, Shimasaki S, Sugino H, Ueno N. Direct binding of follistatin to a complex of bone-morphogenetic protein and its receptor inhibits ventral and epidermal cell fates in early Xenopus embryo. Proc Natl Acad Sci USA. 1998;95:9337-9342.  [PubMed]  [DOI]  [Cited in This Article: ]
118.  Amthor H, Nicholas G, McKinnell I, Kemp CF, Sharma M, Kambadur R, Patel K. Follistatin complexes Myostatin and antagonises Myostatin-mediated inhibition of myogenesis. Dev Biol. 2004;270:19-30.  [PubMed]  [DOI]  [Cited in This Article: ]
119.  Glister C, Kemp CF, Knight PG. Bone morphogenetic protein (BMP) ligands and receptors in bovine ovarian follicle cells: actions of BMP-4, -6 and -7 on granulosa cells and differential modulation of Smad-1 phosphorylation by follistatin. Reproduction. 2004;127:239-254.  [PubMed]  [DOI]  [Cited in This Article: ]
120.  Kogure K, Zhang YQ, Maeshima A, Suzuki K, Kuwano H, Kojima I. The role of activin and transforming growth factor-beta in the regulation of organ mass in the rat liver. Hepatology. 2000;31:916-921.  [PubMed]  [DOI]  [Cited in This Article: ]
121.  Takabe K, Wang L, Leal AM, Macconell LA, Wiater E, Tomiya T, Ohno A, Verma IM, Vale W. Adenovirus-mediated overexpression of follistatin enlarges intact liver of adult rats. Hepatology. 2003;38:1107-1115.  [PubMed]  [DOI]  [Cited in This Article: ]
122.  Kogure K, Omata W, Kanzaki M, Zhang YQ, Yasuda H, Mine T, Kojima I. A single intraportal administration of follistatin accelerates liver regeneration in partially hepatectomized rats. Gastroenterology. 1995;108:1136-1142.  [PubMed]  [DOI]  [Cited in This Article: ]
123.  Kogure K, Zhang YQ, Shibata H, Kojima I. Immediate onset of DNA synthesis in remnant rat liver after 90% hepatectomy by an administration of follistatin. J Hepatol. 1998;29:977-984.  [PubMed]  [DOI]  [Cited in This Article: ]
124.  Endo D, Maku-Uchi M, Kojima I. Activin or follistatin: which is more beneficial to support liver regeneration after massive hepatectomy? Endocr J. 2006;53:73-78.  [PubMed]  [DOI]  [Cited in This Article: ]
125.  Patella S, Phillips DJ, Tchongue J, de Kretser DM, Sievert W. Follistatin attenuates early liver fibrosis: effects on hepatic stellate cell activation and hepatocyte apoptosis. Am J Physiol Gastrointest Liver Physiol. 2006;290:G137-G144.  [PubMed]  [DOI]  [Cited in This Article: ]
126.  Rossmanith W, Chabicovsky M, Grasl-Kraupp B, Peter B, Schausberger E, Schulte-Hermann R. Follistatin overexpression in rodent liver tumors: a possible mechanism to overcome activin growth control. Mol Carcinog. 2002;35:1-5.  [PubMed]  [DOI]  [Cited in This Article: ]
127.  Fuwii M, Ishikawa M, Iuchi M, Tashiro S. Effect of follistatin on rat liver regeneration and tumor growth after portal occlusion. Hepatogastroenterology. 2005;52:833-838.  [PubMed]  [DOI]  [Cited in This Article: ]
128.  Mashima H, Kanzaki M, Nobusawa R, Zhang YQ, Suzuki M, Mine T, Kojima I. Derangements in the activin-follistatin system in hepatoma cells. Gastroenterology. 1995;108:834-840.  [PubMed]  [DOI]  [Cited in This Article: ]
129.  Wang F, Denison S, Lai JP, Philips LA, Montoya D, Kock N, Schule B, Klein C, Shridhar V, Roberts LR. Parkin gene alterations in hepatocellular carcinoma. Genes Chromosomes. Cancer. 2004;40:85-96.  [PubMed]  [DOI]  [Cited in This Article: ]
130.  Fujiwara M, Marusawa H, Wang HQ, Iwai A, Ikeuchi K, Imai Y, Kataoka A, Nukina N, Takahashi R, Chiba T. Parkin as a tumor suppressor gene for hepatocellular carcinoma. Oncogene. 2008;27:6002-6011.  [PubMed]  [DOI]  [Cited in This Article: ]
131.  Beale G, Chattopadhyay D, Gray J, Stewart S, Hudson M, Day C, Trerotoli P, Giannelli G, Manas D, Reeves H. AFP, PIVKAII, GP3, SCCA-1 and follisatin as surveillance biomarkers for hepatocellular cancer in non-alcoholic and alcoholic fatty liver disease. BMC Cancer. 2008;8:200.  [PubMed]  [DOI]  [Cited in This Article: ]
132.  Gao X, Hu H, Zhu J, Xu Z. Identification and characterization of follistatin as a novel angiogenin-binding protein. FEBS Lett. 2007;581:5505-5510.  [PubMed]  [DOI]  [Cited in This Article: ]
133.  Shibanuma M, Mashimo J, Mita A, Kuroki T, Nose K. Cloning from a mouse osteoblastic cell line of a set of transforming-growth-factor-beta 1-regulated genes, one of which seems to encode a follistatin-related polypeptide. Eur J Biochem. 1993;217:13-19.  [PubMed]  [DOI]  [Cited in This Article: ]
134.  Kawabata D, Tanaka M, Fujii T, Umehara H, Fujita Y, Yoshifuji H, Mimori T, Ozaki S. Ameliorative effects of follistatin-related protein/TSC-36/FSTL1 on joint inflammation in a mouse model of arthritis. Arthritis Rheum. 2004;50:660-668.  [PubMed]  [DOI]  [Cited in This Article: ]
135.  Miyamae T, Marinov AD, Sowders D, Wilson DC, Devlin J, Boudreau R, Robbins P, Hirsch R. Follistatin-like protein-1 is a novel proinflammatory molecule. J Immunol. 2006;177:4758-4762.  [PubMed]  [DOI]  [Cited in This Article: ]
136.  Oshima Y, Ouchi N, Sato K, Izumiya Y, Pimentel DR, Walsh K. Follistatin-like 1 is an Akt-regulated cardioprotective factor that is secreted by the heart. Circulation. 2008;117:3099-3108.  [PubMed]  [DOI]  [Cited in This Article: ]
137.  Mashimo J, Maniwa R, Sugino H, Nose K. Decrease in the expression of a novel TGF beta1-inducible and ras-recision gene, TSC-36, in human cancer cells. Cancer Lett. 1997;113:213-219.  [PubMed]  [DOI]  [Cited in This Article: ]
138.  Sumitomo K, Kurisaki A, Yamakawa N, Tsuchida K, Shimizu E, Sone S, Sugino H. Expression of a TGF-beta1 inducible gene, TSC-36, causes growth inhibition in human lung cancer cell lines. Cancer Lett. 2000;155:37-46.  [PubMed]  [DOI]  [Cited in This Article: ]
139.  Hodgson G, Hager JH, Volik S, Hariono S, Wernick M, Moore D, Nowak N, Albertson DG, Pinkel D, Collins C. Genome scanning with array CGH delineates regional alterations in mouse islet carcinomas. Nat Genet. 2001;29:459-464.  [PubMed]  [DOI]  [Cited in This Article: ]
140.  Chan QK, Ngan HY, Ip PP, Liu VW, Xue WC, Cheung AN. Tumor suppressor effect of follistatin-like 1 in ovarian and endometrial carcinogenesis: a differential expression and functional analysis. Carcinogenesis. 2009;30:114-121.  [PubMed]  [DOI]  [Cited in This Article: ]
141.  Reddy SP, Britto R, Vinnakota K, Aparna H, Sreepathi HK, Thota B, Kumari A, Shilpa BM, Vrinda M, Umesh S. Novel glioblastoma markers with diagnostic and prognostic value identified through transcriptome analysis. Clin Cancer Res. 2008;14:2978-2987.  [PubMed]  [DOI]  [Cited in This Article: ]
142.  Slany A, Haudek VJ, Zwickl H, Gundacker NC, Grusch M, Weiss TS, Seir K, Rodgarkia-Dara C, Hellerbrand C, Gerner C. Cell Characterization by Proteome Profiling Applied to Primary Hepatocytes and Hepatocyte Cell Lines Hep-G2 and Hep-3B. J Proteome Res. 2009;14:Epub ahead of print.  [PubMed]  [DOI]  [Cited in This Article: ]
143.  Kato MV, Sato H, Tsukada T, Ikawa Y, Aizawa S, Nagayoshi M. A follistatin-like gene, mac25, may act as a growth suppressor of osteosarcoma cells. Oncogene. 1996;12:1361-1364.  [PubMed]  [DOI]  [Cited in This Article: ]
144.  Kato MV. A secreted tumor-suppressor, mac25, with activin-binding activity. Mol Med. 2000;6:126-135.  [PubMed]  [DOI]  [Cited in This Article: ]
145.  Hwa V, Oh Y, Rosenfeld RG. The insulin-like growth factor-binding protein (IGFBP) superfamily. Endocr Rev. 1999;20:761-787.  [PubMed]  [DOI]  [Cited in This Article: ]
146.  Komatsu S, Okazaki Y, Tateno M, Kawai J, Konno H, Kusakabe M, Yoshiki A, Muramatsu M, Held WA, Hayashizaki Y. Methylation and downregulated expression of mac25/insulin-like growth factor binding protein-7 is associated with liver tumorigenesis in SV40T/t antigen transgenic mice, screened by restriction landmark genomic scanning for methylation (RLGS-M). Biochem Biophys Res Commun. 2000;267:109-117.  [PubMed]  [DOI]  [Cited in This Article: ]
147.  Tsuchida K, Arai KY, Kuramoto Y, Yamakawa N, Hasegawa Y, Sugino H. Identification and characterization of a novel follistatin-like protein as a binding protein for the TGF-beta family. J Biol Chem. 2000;275:40788-40796.  [PubMed]  [DOI]  [Cited in This Article: ]
148.  Hayette S, Gadoux M, Martel S, Bertrand S, Tigaud I, Magaud JP, Rimokh R. FLRG (follistatin-related gene), a new target of chromosomal rearrangement in malignant blood disorders. Oncogene. 1998;16:2949-2954.  [PubMed]  [DOI]  [Cited in This Article: ]
149.  Tortoriello DV, Sidis Y, Holtzman DA, Holmes WE, Schneyer AL. Human follistatin-related protein: a structural homologue of follistatin with nuclear localization. Endocrinology. 2001;142:3426-3434.  [PubMed]  [DOI]  [Cited in This Article: ]
150.  Bartholin L, Maguer-Satta V, Hayette S, Martel S, Gadoux M, Corbo L, Magaud JP, Rimokh R. Transcription activation of FLRG and follistatin by activin A, through Smad proteins, participates in a negative feedback loop to modulate activin A function. Oncogene. 2002;21:2227-2235.  [PubMed]  [DOI]  [Cited in This Article: ]
151.  Mukherjee A, Sidis Y, Mahan A, Raher MJ, Xia Y, Rosen ED, Bloch KD, Thomas MK, Schneyer AL. FSTL3 deletion reveals roles for TGF-beta family ligands in glucose and fat homeostasis in adults. Proc Natl Acad Sci USA. 2007;104:1348-1353.  [PubMed]  [DOI]  [Cited in This Article: ]
152.  Razanajaona D, Joguet S, Ay AS, Treilleux I, Goddard-Leon S, Bartholin L, Rimokh R. Silencing of FLRG, an antagonist of activin, inhibits human breast tumor cell growth. Cancer Res. 2007;67:7223-7229.  [PubMed]  [DOI]  [Cited in This Article: ]
153.  Glusman G, Kaur A, Hood L, Rowen L. An enigmatic fourth runt domain gene in the fugu genome: ancestral gene loss versus accelerated evolution. BMC Evol Biol. 2004;4:43.  [PubMed]  [DOI]  [Cited in This Article: ]