Open Access

Physiological and pathological roles of Hic‑5 in several organs (Review)

  • Authors:
    • Shun Yao
    • Zhen Tu
    • Xingyue Yang
    • Li Zhang
    • Yuling Zhong
    • Liming Zheng
    • Hui Wang
    • Zhiqiang Yi
    • Jiaxing An
    • Hai Jin
    • Guorong Wen
    • Biguang Tuo
  • View Affiliations

  • Published online on: October 10, 2022     https://doi.org/10.3892/ijmm.2022.5194
  • Article Number: 138
  • Copyright: © Yao et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY_NC 4.0].

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Integrins allow cells to adhere to the extracellular matrix and promote the recruitment of other integrins, resulting in the formation of focal adhesion sites at the binding sites. Focal adhesion sites play essential roles in the assembly of the cytoskeleton and are vital in shaping the structure of cells. They also play other regulatory roles by influencing numerous biological functions, such as cell proliferation and apoptosis. Hydrogen peroxide‑inducible clone 5 (Hic‑5) is a member of the Paxillin family of proteins and is an adhesive plaque scaffolding protein. Its expression can be detected in both vascular and smooth muscle cells. Thus, it plays an essential role in vascular remodeling, as well as in fibrotic diseases. Hic‑5 functions as a coactivator of steroid receptors, thus playing a role in steroid hormone‑dependent diseases. It also plays a vital role in the invasive metastasis of various types of cancer. Moreover, several studies have demonstrated that Hic‑5 plays a critical role in transcriptional regulation, as well as in numerous signaling pathways. Therefore, the inhibition of the functions of Hic‑5 may prevent the development or halt the progression of several diseases. Its use as a therapeutic target in future investigations may thus aid in the treatment of several diseases, including various types of cancer. The present review article focused on the expression and functions of Hic‑5 in different organs, with the aim of highlighting novel possibilities for future research.

1. Introduction

Hydrogen peroxide-inducible clone 5 (Hic-5) was isolated from a transforming growth factor-β (TGF-β) or hydrogen peroxide-inducible gene clone by ablative hybridization in 1994 by Shibanuma et al (1). Subsequently, after studying Hic-5 at the molecular and cellular level, it was found that Hic-5 is a homolog of Paxillin and is currently considered a member of the Paxillin protein family (2). The Paxillin family includes Paxillin, Leupaxin and Hic-5, and they function as molecular adapters that deliver signals when the cellular adhesion environment changes (2).

Hic-5 has a high degree of homology with Paxillin and its intracellular localization is similar (Table I) (2-19). It is primarily restricted to focal adhesion sites, and sites of adhesion with the extracellular matrix (ECM) via integrins (2). In contrast to the abnormal development of extra-embryonic tissues, and the segmentation of the heart and body in mouse fetuses following the knockout of Paxillin in mice (20), no effects on homeostasis and development were observed in Hic-5 knockout mice (13). This indicates that Hic-5 is not required for development, and suggests that Paxillin and Hic-5 have different physiological functions. Hic-5 expression has been detected in smooth muscle cells (SMCs) of various tissues (21), and a higher level of expression has been observed in the lungs and spleen (1). Hic-5 expression has been assessed and shown to vary in several cell lines (2). It is highly expressed in mesenchymal cell lines, such as fibroblasts, whilst it exhibits lower levels of expression in epithelial cell lines (2). The fact that Hic-5 expression can be detected in different tissues and cells also implies that it plays a central role in the pathophysiology of different organs.

Table I

Characterization and expression of Paxillin and Hic-5.

Table I

Characterization and expression of Paxillin and Hic-5.

ProteinMolecular weight (kDa)ChromosomeDomain structureExpressing tissues or cellsFunctionsRelated disease(Refs.)
Hic-55516p11 (human) 7 (mouse)Four LD motifs Four LIM domainsWidespread high level of expression in SMCs, platelets, and fibroblasts, etc.Regulation of the expression of multiple genes through transcriptional regulation. Interacts with multiple structural and signaling moleculesVascular injury, fibrosis, cancer, etc.(2-14)
Paxillin6812q24.2 (human) 5 (mouse)Five LD motifs Four LIM domainsUniversally expressedEmbryogenesis, involved in the adhesion and metastasis of tumor cells, regulates signaling pathways, etc.Cancer, inflammation, etc.(15-19)

[i] Hic-5, hydrogen peroxide-inducible clone 5.

However, the physiological and pathological roles of Hic-5 have not yet been systematically clarified. Therefore, the present review summarized the expression of Hic-5 in different organs and its role in various diseases.

2. Structure and function of Hic-5

Hic-5 has a molecular weight of 55 kDa and consists of 444 amino acids; its gene is located in chromosome 16p11 in humans and in chromosome 7 mice (3). A long intron can be found in the genetic structure of Hic-5 between the N-terminal and C-terminal structural domains (22). Hic-5 consists of four Leu- and Asp-rich LD domains in the N-terminal region; LD1 is missing in one isoform; and there are four LiM domains with two zinc fingers in the C-terminal region (2). Hic-5 also includes multiple phosphorylation sites; tyrosine phosphorylation can occur in response to stimuli, such as osmotic pressure, serum factors, lysophosphatidic acid (LPA) and endothelin (8,23), which further regulate signaling associated with lamellar pseudopodia formation to influence cell motility (24). The structural domains involved in protein interactions include the LD and LiM structural domains, and these allow Hic-5 to function as a junction molecule and thus facilitate protein-protein interactions. It can also serve as a docking site for signaling proteins, such as vinculin and focal adhesion kinase (FAK) (25,26). Additionally, Hic-5 can function as a linker molecule in the integrin substrate complex and can modulate integrin signaling through interactions with binding molecules (2). Under normal culture conditions, the majority of cellular behaviors are almost unaffected by Hic-5 expression (2). When healthy adhesion maintenance is compromised under conditions of stress that affect focal adhesion sites, Hic-5 may inhibit adhesion and excessive changes in cytoskeletal structure by antagonizing Paxillin (21,27).

There is a nuclear export signal (NES) that overlaps with the amino-terminal LD structural domain, and this allows Hic-5 to enter the nucleus from the cytoplasm via an oxidation-sensitive NES (28). Hic-5 is transferred from a focal adhesion site to actin stress fibers in the presence of organic stress, thereby regulating cell contractility (21). In various pathophysiological processes, Hic-5 functions primarily through two different pathways. First, it can affect the transcriptional levels of several nuclear receptors (29), including the glucocorticoid receptor (GR) (4,30), the androgen receptor (AR) (31) and the progesterone receptor (13). Hic-5 may affect the genomic occupancy of GR, as well as the assembly of transcriptional complexes and may thus function as a glucocorticoid regulatory switch for several genes (30). After entering the nucleus, Hic-5 can regulate the expression of several genes through transcriptional regulation (4-6). For example, it can repress Lef/Tcf-driven transcription. Multiple sequences upstream of c-fos (such as GC/Sp1 and Ets, amongst others) can activate Hic-5 (Fig. 1) (5). The P21 promoter region can also respond to Hic-5 in the nucleus, which can bind to Sp1, Smad3, and p300 to create a transcriptional complex (6). Hic-5 also interacts with glucocorticoid response promoters (such as TIF-2 and p300, among others) and coactivators, and thus functions as a steroid receptor coactivator (6,32-34). In addition, Hic-5 acts in conjunction with various structural and signaling molecules (such as FAK, vinculin and PtP-PEST, amongst others) to form a scaffold for integrin signaling (Fig. 2) (8-10,14). Hic-5 has been shown to be essential to the adhesion formation of the three-dimensional (3D) structure of the ECM (35). Moreover, when Hic-5 expression is low, fibroblasts exhibit higher migratory capacity, and when its expression is high, the migratory capacity of fibroblasts is reduced and the contractile capacity is increased, increasing the contraction and matrix remodeling of the ECM (36). This allows Hic-5 to function as a key factor in various types of fibrotic diseases (7,11,37).

3. Hic-5 and the regulation of signaling pathways

A main mechanism through which Hic-5 exerts its effects is via the activation of various signaling pathways (Fig. 3). It can induce the formation of abdominal aortic aneurysm (AAA) by promoting the activity of the JNK pathway (38). The TGF-β/Smad pathway is a crucial factor in fibrotic diseases (39,40), and TGF-β can promote Hic-5 expression (1). Previous studies have also demonstrated that Hic-5 can promote the development of liver fibrosis and pancreatic fibrosis via the activation of Smad2/Smad3 and inhibition of Smad7 (7,41). It also inhibits the apoptosis of prostate cancer cells by binding and interacting with Smad1 (42). Hic-5 inhibits β-catenin function upon interaction with Smad4, thereby promoting the proliferation and inhibiting apoptosis of osteosarcoma (OS) cells (43). Hic-5 can also promote inflammation by activating the activity of the NF-κB pathway (44) and can promote cell apoptosis through the Toll-like receptor 4 (TLR4)/Fas-associated protein with death domain (FADD) signaling pathway (45). Hic-5 can promote hepatocarcinogenesis by activating the reactive oxygen species (ROS)/JNK pathway (46), while in a variety of tumors, Hic-5 can induce epithelial-mesenchymal transition (EMT) by activating RhoA/ROCK I signaling, thus promoting invasion and metastasis (12,47,48). Hic-5 may play a critical role in other signaling pathways; however, current research is limited, and further research is required to elucidate the roles of Hic-5 in other signaling pathways.

4. Role of Hic-5 in the cardiovascular system

A high expression of Hic-5 has been detected in both vascular and visceral SMCs in mice and humans (49); under physiological conditions, Hic-5 expression has been shown to play a very limited role in intravascular homeostasis (13). Moreover, it has been demonstrated Hic-5−/− mice do not exhibit any notable abnormalities compared with the wild-type mice in terms of arterial structure and function (3). However, other researchers have found a reduced endothelial cell (EC) expansion in capillaries and an impaired structural organization of cells on basement membrane extracts following the knockdown of Hic-5 (50). Hic-5 has also been found to be expressed in mouse ECs, which are involved in both EC spreading and migration (3). In ECs, the knockdown of Hic-5 has been found to reduce EC sprouting and lumen formation, whereas the sprouting deficiency is restored upon the re-expression of Hic-5. Hic-5 can interact with membrane type-1 matrix metalloproteinase (MT1-MMPs) under the stimulation of pro-angiogenic factors (51). During EC sprouting, the expression of MT1-MMPs and Hic-5 complexes increases, and this promotes the formation of MT1-MMPs and FAK complexes, thus playing a role in EC matrix proteolysis and cell motility (51). All types of vessels contain ECs and vascular SMCs (VSMCs); this suggests that Hic-5 functions as an essential factor in all types of vascular diseases.

Hic-5 and vascular injury

A previous study on Hic-5 found that it carried out a regulatory function in the activation of integrin αIIbβ3 and platelet aggregation in mice (52). However, a subsequent study found that the knockdown of Hic-5 did not affect hemostasis and experimental thrombosis (53), possibly as the function affected by Hic-5 absence was compensated for by Paxillin and Leupaxin, which are also expressed in mouse platelets (54).

In a previous study, in a rat model of wire-mediated femoral artery injury, the expression of Hic-5 was significantly downregulated after 4 days, followed by a gradual recovery to normal levels after 2 weeks, suggesting that Hic-5 expression is downregulated during the acute phase of vascular injury (13). By contrast, in Hic-5−/− mice, a significant reduction in the arterial mesangial area with vascular injury was observed along with the accelerated formation of neointima and promotion of chronic apoptosis of VSMCs (13). Similarly, in another study, in the mouse SM SV30 cell line, the expression of Hic-5 was notably increased after differentiation above undifferentiated levels, and furthermore, in a rat model of acute vascular injury following the local delivery of Hic-5 using an adenoviral vector, neointima formation was inhibited, primarily by reducing the migration of vascular SM cells (55). However, further investigations are required to explore the specific underlying mechanisms.

Hic-5 and atherosclerosis

The adhesion of monocytes and ECs is a key factor in the development of atherosclerotic plaques (56,57). The APOE−/− model is currently the most well-recognized and widely used model for studying atherosclerosis. Arita-Okubo et al (58) extracted the aortas of APOE−/− and LDLR−/− mice and found that Hic-5 deficiency inhibited the adhesion of THP-1 monocytes to the aorta typically stimulated with TNF-α or oxidized low-density lipoprotein. Electron microscopic analysis of aortas extracted from APOE−/− mice revealed that Hic-5 deletion significantly inhibited TNF-α-induced microvilli-like structures. Further experiments revealed that the knockdown of Hic-5 significantly inhibited atherosclerotic changes in mice. The same reduction in the number of surface microvillus-like structures was found in human umbilical vein ECs (HUVECs) in which Hic-5 was knocked down, while the number of surface microvillus-like structures was significantly increased in HUVECs after overexpression of Hic-5 (58). These findings indicate that Hic-5 in ECs not only promotes the formation of microvillous-like structures, but also promotes the recruitment of monocytes, which ultimately leads to atherosclerosis (58). However, the exact role of Hic-5 has not yet been determined, thus the mechanisms through which Hic-5 affects atherosclerosis warrant further investigations in future studies.

Hic-5 and AAA

In a previous study in which models of AAA were constructed by the administration of angiotensin II in APOE−/− and APOE−/− Hic-5−/− mice, it was found that Hic-5−/− mice exhibited a significantly lower degree of AAA and had smaller maximum aortic diameters (38). Abdominal artery rupture and mass hemorrhaging were observed in the APOE−/− mice, but not in the APOE−/− Hic-5−/− mice, indicating that the knockdown of Hic-5 prevented AAA formation and rupture (38). Further mechanistic analyses revealed that Hic-5 promoted the activation of the JNK pathway, subsequently increasing MMP expression and promoting the development of AAAs (38).

Hic-5 and hypertrophic heart disease

Hic-5 has also been shown to be expressed in neonatal rat ventricular myocytes (NRVMs) (59). In NRVMs, epinephrine induced the expression of Hic-5, and Hic-5 overexpression significantly increased the number of cells with organized cytoskeleton (59). Mechanistically, Hic-5 exerted its effects via regulation of the ERK1/2 signaling pathway (59). Increased levels of Hic-5 were found in a mouse model of hypertrophic heart disease established using thoracic aortic constriction (59). However, the elevated of Hic-5 in the model alone did not suggest that it played a role in the development of hypertrophic heart disease; thus, future investigations are required to determine its role in hypertrophic heart disease.

5. Role of Hic-5 in the digestive system

Hic-5 and the liver
Hic-5 and liver fibrosis

Previous studies have demonstrated that Hic-5 is not expressed in quiescent hepatic stellate cells (HSCs), although its expression is significantly increased following HSC activation (7,60). The significant upregulation of HIC-5 expression has also been found in liver tissues from patients with fibrosis, or in mice subjected to bile duct ligation or mice with CCl4-induced mouse liver fibrosis, as well as in activated HSCs (7). It has been revealed that HSCs from Hic-5−/− mice exhibit a significantly reduced activation when cultured (7). Additionally, Hic-5 has been found to promote fibrosis by activating the TGF-β/Smad2 pathway, while inhibiting Smad7 (7).

Hic-5 and hepatic ischemia-reperfusion injury (HIRI)

In a previous study, Hic-5 expression was shown to be increased in models of HIRI, and the expression of inflammatory chemokines was found to be decreased in a Hic-5−/− HIRI mouse model, while the extent of liver injury was found to be reduced compared with wild-type mice (45). Further experiments revealed that Hic-5 activated the NF-κB signaling pathway to enhance the inflammatory response, and activated the TLR4/FADD pathway to increase hepatocyte apoptosis, which ultimately led to the exacerbation of the lesions in mice (45). This suggests that Hic-5 targeted therapy may be beneficial for liver transplantation-induced HIRI. Similarly, Hic-5 may play a role in ischemia-reperfusion injury in other organs (such as the lungs); however, further studies are required to confirm this.

Hic-5 and chronic pancreatitis (CP)

Similar to liver fibrosis, pancreatic fibrosis is also primarily caused by the activation of pancreatic stellate cells (PSCs) (61). A high expression of Hic-5 was likewise found in pancreatic tissues of patients with CP and was primarily expressed in activated PSCs (41). In a previous study, by culturing pancreatic primary PSCs, it was found that the knockdown of HIC-5 inhibited PSC activation. The same study subsequently found a significant attenuation of pancreatic fibrosis in Hic-5−/− mice in a model of caerulein-induced CP, suggesting that Hic-5 can promote the development of CP (41). By culturing primary PSCs, Hic-5 was shown to increase the activation of PSCs by increasing the phosphorylation of Smad2, ultimately promoting the development of CP (41). Another study also found that the knockdown of Hic-5 inhibited NF-κB/p65 expression (44). This also suggested that Hic-5 can be used as a diagnostic marker and a potential therapeutic target in CP.

6. Hic-5 and the urinary system

Hic-5 and the kidneys

In rat kidneys, Hic-5 expression has been detected in mesenchymal cells, mesangial cells and ECs (62). This suggests that Hic-5 may be involved in the development of glomerulosclerosis (11). Another study also demonstrated that the increased expression of Hic-5 in glomerular mesangial cells was not dependent on the TGF-β-induced pro-sclerotic phenotype (63); however, that study only illustrated the increased expression of Hic-5 in glomerulosclerosis, which does not yet indicate a role in the development of glomerulosclerosis, and therefore further studies are required to determine this.

Hic-5 in the prostate

Previous research detected Hic-5 expression in the normal prostate mesenchyme and exhibited a lesser reduction in the tumor-associated mesenchyme (31). Hic-5 expression was subsequently found in both the normal and tumor prostate epithelium, as well as in prostate cancer cells and prostate cancer tissues (42,64,65). Hic-5 regulates the transcription of AR (31). When steroid ligands are absent, Hic-5 can interact with nuclear receptor co-blockers to inhibit transcription (34,66). In prostate myofibroblasts, Hic-5 rapidly translocates to the nucleus in response to the action of androgens, consistent with the increased phosphorylation of adherent spot kinase. Hic-5 acts as a co-regulator and AR can lead to androgen-induced transcriptional enhancement through interaction with the coactivator, Hic-5/ARA55, thereby affecting androgenic effects on growth, cell adhesion, motility and invasion regulation (67). It was similarly found that the mRNA levels of Hic-5/ARA55 in normal or benign prostate hypertrophy tissues were reduced in hormone-independent prostate cancer tissues (12,64); in normal prostate tissues, it was primarily expressed in the interstitium (31,68), and in prostate cancer tissues it was also expressed in the interstitium. Hic-5/ARA55 was also observed in the cytoplasm and focal adhesion sites of the prostate mesenchymal cell line, WPMY-1 (31). Hic-5/ARA55 activates AR activity by interacting with the endogenous androgen response promoter (31). CYP24A1 encodes the 1,25D3 metabolizing enzyme 24-hydroxylase (69). Hic-5 is a coactivator of the vitamin D3 receptor (VDR) and can limit the negative feedback circuit of VDR activity by participating in VDR-mediated transcription of CYP24A1 (69).

7. Hic-5 and tumors

Tumor cells exhibit two interchangeable patterns of cell movement, mesenchymal or amoeboid; during invasion, tumor cells can migrate to different 3D microenvironments through these two switchable motility modes, and this allows them to invade the tumor stroma and circulatory system (35,70). Gulvady et al (71) found that the expression levels of Hic-5 varied greatly by analyzing the expression of Paxillin and Hic-5 in a variety of tumor cells, while the levels of Paxillin were relatively stable. It was also found that cancer cell lines with low Hic-5:Paxillin ratios lacked the ability to efficiently convert to a mesenchymal phenotype, while cell lines with a high Hic-5:Paxillin ratios were able to adequately switch from an amoeboidal to a mesenchymal phenotype. In a variety of tumors, Hic-5 has been shown to induce EMT by activating RhoA/ROCK I signaling, thus promoting invasion and metastasis (12,47,48), which highlights its potential as a biomarker for metastasis in several tumors and as a therapeutic target.

Hic-5 and hepatocellular carcinoma (HCC)

Hic-5 is also involved in HCC progression. The upregulation of proline-rich tyrosine kinase 2 (Pyk2) is expressed in HCC liver tissues and is associated with a worse prognosis (72). Other studies have demonstrated that Pyk2 upregulates the activation and localization of Hic-5 (73). In another study, Wu et al (46) found that Hic-5 expression was upregulated in HCC cell lines that originated from high motility cancers, but not in HCC cells originated from low motility cancers when they analyzed liver tissues from patients with HCC, suggesting that its overexpression was closely associated with the metastasis of HCC. This suggests that Hic-5 may be used as a biomarker for metastasis in with HCC. The same authors also found that the knockdown of Hic-5 in high motility-derived HCC cells significantly reduced the invasive ability of HCC cells (46). Further analyses revealed that Hic-5 affected HCC progression through the ROS/JNK pathway (46). However, that study was limited to assessing the progression of HCC in vitro, and further studies are thus required to confirm its role in vivo and to elucidate the specific underlying mechanisms.

Hic-5 and pancreatic cancer (PC)

In a previous study, the upregulated expression of Hic-5 was detected in PC, and the knockdown of Hic-5 revealed that PC cell (PCC) proliferation was suppressed and apoptosis was increased, while PCC invasion and migration were reduced (74). The same study also found that patients with a high expression of Hic-5 in PC had lower survival rates (74). However, there are fewer studies investigating the role of Hic-5 in PC; thus, further cellular and animal models are required to examine the mechanisms through which Hic-5 specifically affects PC and to identify novel strategies for the management of PC.

Hic-5 and esophageal cancer

The tumor microenvironment plays an essential role in tumor development and metastasis (75); cancer-associated fibroblasts (CAFs) are an important component of the tumor microenvironment (76) that release cytokines into the ECM, thereby promoting circulating tumor metastasis (77,78). In a previous study, the high expression of Hic-5 was detected in the CAFs of esophageal squamous cell carcinoma (ESCC) (79). That study further demonstrated that the invasion and migration of cells was inhibited following the knockdown of Hic-5 in KYSE150/TE1 esophageal cancer cells co-cultured with CAFs (79). RNA-seq revealed that Hic-5 in CAFs may promote tumor progression by regulating cytokines (such as CCL2) and altering the ECM (79). The same study also found an association with esophageal cancer lymph node metastasis through survival analysis (79). This suggests that the levels of Hic-5 may be used as a marker of lymph node metastasis in ESCC. The effect of Hic-5 on the biological behavior of esophageal cancer cells has only been demonstrated in vitro; thus, in vivo models are warranted to explore the role of Hic-5 and its specific mechanisms of action in esophageal cancer.

Hic-5 and colon cancer

PPARγ is a major regulator of adipocyte differentiation (80). Hic-5 has been shown to bind to and activate PPARγ, and both Hic-5 and PPARγ are expressed in normal and malignant intestines (33). In a mouse colon cancer model, the expression of both PPARγ and Hic-5 has been found to be notably decreased (33). Hic-5 has also been found to enhance PPARγ-mediated epithelial gene induction in colon cancer cells (33).

The high expression of Hic-5 was also previously detected in CAFs of tumor tissues from patients with colon cancer (81). The addition of CAF supernatant to normal fibroblasts induced the expression of Hic-5 (81). Following the establishment of a mouse colon tumor model by azoxymethane induction, no tumors were found in Hic-5−/− mice compared with wild-type mice, where tumors were found in 55% of mice after 20 weeks; proliferative polyps were found in only one location after 24 weeks, and advanced adenocarcinomas was only observed in wild-type mice (81). Further analyses revealed that HIC-5 induced lysyl oxidase (LOX) expression following nucleation, and LOX promoted the cross-linking of collagen fibers and thus increased the stiffness of ECM, which eventually promoted tumor progression (81). This suggests that Hic-5 may serve as a therapeutic target for the management of colon cancer.

Hic-5 and prostate cancer

Previous research has shown that Hic-5 can interact with Smad1 to inhibit the apoptosis of prostate cancer cells (42). LNCaP is a prostate epithelial cell line that does not express Hic-5. Hic-5 overexpression using lentiviral transfection and subsequent administration in mice has been shown to result in the inhibition of tumor growth (64). Further mechanistic research has revealed that Hic-5/ARA55 can inhibit c-Myc expression in an androgen-dependent manner (65). The expression of c-Myc is increased when deprived of androgens, and Hic-5/ARA55 also inhibits c-Myc expression by suppressing it in a TCF4-dependent and non-dependent manner (64).

Hic-5 could interact with 1,25D3 to inhibit prostate cancer cell proliferation. Following androgen deprivation, Hic-5 induces different responses in prostate tumors to 1,25D3, particularly during androgen deprivation therapy (69). Hic-5 can play a differential role in the adjuvant treatment of VDR activity in prostate cancer. This suggests that patients with prostate cancer with a downregulated expression of Hic-5 may benefit more from treatment with VDR ligands (69).

Hic-5 and breast cancer

Gulvady et al (71) found that the expression of Hic-5 in breast cancer cells was upregulated. They further constructed a mouse polyoma middle T-antigen breast cancer model and found that CAFs in Hic-5−/− mice could not effectively form fibrillar adhesion in both 2D and 3D (82). Using bioinformatics analysis, it was found that the high expression of Hic-5 in patients with breast cancer was negatively associated with the distant metastasis-free survival (DMFS) of patients (83).

Hic-5 and ovarian cancer

The high expression of Hic-5 was previously found in advanced ovarian cancer, and EMT independent of TGFβ-1 was induced by the overexpression of Hic-5 in the ovarian cancer cell line, A2780 (which is typically morphologically epithelial), accompanied with enhanced cell proliferation and migratory/invasive ability, as well as increased resistance to chemotherapeutic agents (48). In addition, in epithelial ovarian cancer cells, the knockdown of Hic-5 inhibited its proliferation, migration/invasion and induced mesenchymal-epithelial transition, and it was further shown that it may promote EMT in ovarian cancer via the regulation of the RhoA/ROCK signaling pathway (48). However, further in vivo studies are required to investigate the role of Hic-5 in ovarian cancer.

Hic-5 and osteosarcoma (OS)

The high expression of Hic-5 was previously detected in both OS tissues and cells, and a higher percentage of low-grade OS tissues exhibited an upregulated expression of Hic-5 (46.7%), while its expression was reduced in the majority of high-grade OS patient tissues (53.3%) (43). The knockdown of Hic-5 suppressed the proliferation of OS cells, whereas it promoted apoptosis. Additionally, it was found that Hic-5 expression in exosomes was similarly reduced following the knockdown of Hic-5, and it was further found that Hic-5 interacted with Smad4 through the exosomal pathway to promote the activation of β-catenin, thus promoting the proliferation of OS cells (43).

Hic-5 and melanoma

Hic-5 expression was previously detected in both human melanoma cells and murine B16-F1 cells (84). The knockdown of Hic-5 in murine melanoma B16-F1 cells was shown to result in reduced cell proliferation and migration, and significantly inhibited tumor growth and lung metastasis in further subcutaneous tumorigenesis assays in mice (84). It was also found an increase in RhoA activity upon the knockdown of Hic-5, accompanied by an altered amoeboid-like phenotype, leading to a loss of cell plasticity, which may also be responsible for its effect on cell motility (84). This suggests that Hic-5 may influence melanoma development via modulation of the RhoA/ROCK signaling pathway.

8. Hic-5 and other diseases

Hic-5 and Alzheimer's disease (AD)

In a previous study using a model AD, an increased expression of Hic-5 was detected in pyramidal neurons in the CA1 region of the hippo-campus (85). It is possible that Hic-5-mediated intracellular signaling in the ECM is involved in the pathogenesis of AD. However, to the best of our knowledge, no further studies have been performed to date to confirm whether Hic-5 is involved in AD development, and thus further research is required to determine this.

Hic-5 and osteoarthritis

A mouse model of osteoarthritis of the knee was previously established by surgical induction, and a significantly lower degree of cartilage degradation was observed in Hic-5−/− mice than in wild-type mice after 8 weeks (86). Furthermore, by extracting primary chondrocyte cultures, it was found that Hic-5 promoted the development of osteoarthritis primarily by promoting the expression of inflammatory cytokines, or mechanical load-induced MMP13, and a disintegrin and metalloproteinase with thrombospondin type 1 motif 5 (86).

Hic-5 and skin

Hic-5 modulates androgen sensitivity in hair follicle papillae (87), while in scar-forming myofibroblasts, autocrine TGF-β upregulates Hic-5 expression, further downregulating the autocrine loop of Hic-5 and collagen synthesis (88). The overexpression of Hic-5 in keloid-derived fibroblasts was previously found to increase collagen expression in keloid scars (89). Similarly, Hic-5 was confirmed to be expressed in keloid specimens (8/15) using immunohistochemistry, and was not observed in normal tissues; its expression was shown to be primarily localized in the nucleus where it stimulated Smad2/3 expression via other pathways not dependent on Smad7 thus promoting scar formation (89). Hic-5 expression was found to be increased in patients with systemic sclerosis (SSc) and SSc dermal fibroblasts. In addition, in SSc fibroblasts, the knockdown of Hic-5 reduced the production of type I collagen by >50%. This suggests that Hic-5 can promote the formation of fibrosis in SSc, indicating that it may be a therapeutic target for the management of SSc fibrosis (90). However, that study had a small clinical sample; thus, further clinical samples are warranted to confirm these findings and the underlying molecular mechanism need to be assessed.

Hic-5 and the eyes

Aqueous humor (AH) homeostasis is critical for maintaining normal intraocular pressure (91). By contrast, increased obstruction or resistance to the atrial fluid flow through the trabecular meshwork (TM) causes increased intraocular pressure and is a major risk element for primary intraocular pressure elevation (92). In a previous study, in human TM (HTM) cells, Hic-5 expression was limited to the focal adhesion of HTM cells, as well as throughout the TM and AH efflux pathways of the Schlemm's canal; the administration of recombinant Hic-5 was found to result in the redistribution of actin stress fibers, focal adhesion and αv integrins in the focal adhesion, along with an increase in a-SMA, and increased collagen-1 expression, whereas the knockdown of Hic-5 reversed these effects. In the presence of dexamethasone, Hic-5 in TM increased resistance to AH efflux via the trabecular pathway, which suggests that Hic-5 plays a critical role in regulating eye AH outflow through the TM (93).

9. Perspectives of Hic-5 in clinical applications

TGF-β plays a key role in the progression of various diseases. Hic-5 expression can be induced by TGF-β, plays a key role in various diseases (Table II) (7,11,13,31,38,41-46,48,55,58-60, 64,71,74,79,81,83-86,90,94-122) and the negative effects of its knockdown are limited and not severe; thus, targeting Hic-5 may be a suitable approach for the management of several diseases. Hic-5 expression was previously found to be significantly elevated in liver fibrosis, pancreatic fibrosis, HIRI, glomerulosclerosis and SSc, while disease pathology was reduced following the knockdown of Hic-5 (7,11,41,44,45,88,90), which also suggests its potential use as a biomarker for the diagnosis of fibrotic diseases and as a potential therapeutic target. The role of Hic-5 in other fibrotic diseases likely promotes fibrosis via the activation of the TGF-β/Smad pathway.

Table II

Role of Hic-5 and TGF-β in different diseases.

Table II

Role of Hic-5 and TGF-β in different diseases.

DiseaseHic-5TGF-β(Refs.)
Vascular injury• Expression was downregulated during the acute phase of vascular injury. Hic-5−/− mice exhibited a significant reduction in the arterial mesangial area as well as accelerated neonatal intima formation and increased levels of chronic apoptosis of vascular SMCs.• Increased expression after vascular injury.
• Promotes the proliferation of neoplastic endothelium.
(13,55,94,95)
Atherosclerosis• Plays an essential role in the formation of microvillous structures of endothelial cells.
• Promotes atherosclerosis by recruiting monocytes and interacting with monocytes.
• Decreased TGF-β levels in patients with advanced atherosclerosis.
• Promotes the development of atherosclerosis.
(58,95,96)
Abdominal aortic aneurysm• Activates the JNK pathway to enhance MMP expression in VSMCs and promote AAA formation.• Decrease aneurysm formation and progression.
• TGF-β1 can decrease pseudoaneurysm formation and progression.
(38,96,97)
Hypertrophic heart disease• Overexpression of Hic-5 increases the number of cells in the cytoskeletal tissue.• Not reported(59)
Liver fibrosis• Expression is upregulated in patients with liver fibrosis and in mouse models.
• Exerts its effect by activating the TGF-β/Smad2 pathway while suppressing Smad7.
• Activation of hepatic stellate cells through the
• TGF-β/Smad signaling pathway, thereby promoting liver fibrosis
(7,60,98,99)
Hepatic ischemia-reperfusion injury (HIRI)• Highly expressed in HIRI models.
• Promotes hepatocyte apoptosis through the TLR4-FADD pathway and inflammation through the NF-κB pathway.
• Serum TGF-β1 levels are upregulated in the HIRI mouse model.(45,100)
Hepatocellular carcinoma (HCC)• Highly expressed in HCC cells originating from cells with a high degree of motility.
• Promotes HCC development through the ROS-JNK signaling pathway.
• Limiting hepatocyte proliferation under normal conditions, but enabling chemically induced HCC in the heterozygous state.
• Promotes HCC progression and immune escape.
(46,99,101-103)
Chronic pancreatitis• Highly expressed in patients with CP.
• Exerts its effect by activating the TGF-β/Smad2 signaling pathway and NF-κB signaling pathway.
• Highly expressed in a mouse model of CP.
• Promotes CP progression.
(41,44,104)
Pancreatic cancer• Highly expressed in pancreatic cancer.
• Promotes the proliferation of PCCs, reduces apoptosis and promotes the invasion and migration of PCCs.
• Increased expression in patients with pancreatic ductal adenocarcinoma (PDAC).
• Inhibition of TGF-β receptor inhibits tumor growth and metastasis.
(74,99,105)
Esophageal cancer• Highly expressed in ESCC.
• Promotes invasion and migration of esophageal squamous carcinoma cells. Could be used as a marker of lymph node metastasis in ESCC.
• Associated with the overall survival time in esophageal cancer.(79,99,106)
Colorectal cancer• Highly expressed in the CAFs of patients with CRC.
• Promotes the expression of LOX, promotes the cross-linking of collagen fibers, and thus increases the stiffness of the ECM, ultimately promoting tumor progression.
• Highly expressed in CRC.
• Inhibition of TGF-β signaling inhibits CRC metastasis
(81,99,107,108)
Glomerulosclerosis• Increased expression in a rat model of glomerulosclerosis.
• Kidney fibrosis can be reduced by targeting TGF-β downstream molecules.
• Highly expressed in various kidney diseases associated with fibrosis.(11,109,110)
Prostate cancer• Expressed in tumor prostate epithelium, AR-deficient prostate cancer cells, and in tissue from untreated stage IV prostate cancer.
• Interacts with Smad1 to inhibit apoptosis.
• Inhibits proliferation in early stages of prostate cancer, promotes proliferation and metastasis in advanced stages.(31,42,64, 111-113)
Breast cancer• Increased expression in breast cancer cells.
• Negatively associated with the DMFS of breast cancer patients.
• TGF-β1 activated cancer-associated fibroblasts to promote breast cancer invasion, metastasis and EMT(71,83,114)
Ovarian cancer• Highly expressed in ovarian cancer.
• Promotes EMT in ovarian cancer via RhoA/ROCK signaling.
• Induces metastasis or EMT in advanced ovarian cancer(48,115)
Osteosarcoma• Highly expressed in the osteosarcoma.
• Activates β-catenin by interacting with smad4 to promote osteosarcoma cell proliferation and inhibit osteosarcoma apoptosis.
• Serum levels are increased in patients with osteosarcoma.(43,116,117)
Melanoma• Expressed in human melanoma cells.
• Promotes proliferation and migration.
• Promotes invasion and metastasis.
• Promotes the invasion and metastasis of melanoma cells.
(84,118)
Alzheimer's disease• Increased expression in a rat model of AD• Serum levels are increased in patients with AD.
• Reduce amyloid β-protein (Aβ) pathology of brain parenchyma.
• May be a useful biological marker for patients with AD.
(85,119,120)
Osteoarthritis• Promotes the development of osteoarthritis by increasing the expression of MMP13 and ADAMTS5• Serum levels are increased in patients with osteoarthritis.
• Promotes osteoarthritis progression.
(86,121)
Systemic sclerosis• Increased expression in systemic sclerosis skin.• Elevated serum levels in patients with systemic sclerosis.(90,122)

[i] Hic-5, hydrogen peroxide-inducible clone 5; SMCs, smooth muscle cells; VSMCS, vascular smooth muscle cells; AAA, abdominal aortic aneurysm; EMT, epithelial-mesenchymal transition; TLR4, Toll-like receptor 4; FADD, Fas-associated protein with death domain; PCCs, pancreatic cancer cells; ESCC, esophageal squamous cell carcinoma; CRC, colorectal cancer; DMFS, distant metastasis-free survival; AD, Alzheimer's disease.

In tumors, the high expression of Hic-5 was found to promote the transformation of tumor cells with an amoeboid phenotype to a mesenchymal phenotype and promote EMT through the RhoA/ROCK pathway, thus promoting tumor cell plasticity and invasiveness (12,47,48), which also suggests that Hic-5 may be used as a biomarker for tumor metastasis. Related studies have demonstrated that Hic-5 expression is upregulated in HCC with a low migratory capacity and is also associated with lymph node metastasis in patients with esophageal cancer (46,79). Therefore, it may be used as a biomarker for the detection of metastases in patients with HCC, and lymph node metastases in patients with esophageal cancer. It is also negatively associated with the survival of with PC and the DMFS of patients with breast cancer and may serve as a biomarker for predicting a poor prognosis of patients with PC and breast cancer. The knockdown of Hic-5 in HCC cells, PC cells, esophageal cancer cells, ovarian cancer, melanoma and cholangiocarcinoma cells led to the inhibition of tumor cell invasion and migration (43,46,48,74,79,84,123), and the knockdown of Hic-5 in HuCCT1 cholangiocarcinoma cells was also found to inhibit cholangiocarcinoma migration in a concentration-dependent manner (123). This suggests that it may be possible to inhibit tumor metastasis in these tumors by targeting Hic-5. However, the effects of the knockdown of Hic-5 were only verified at the cellular level in these tumors, and further in vivo experiments are thus required to confirm these results. In addition, the knockdown of Hic-5 was previously found to suppress tumorigenesis in mouse models of colon and breast cancer (81,82). In patients with PC, those with a downregulated HIC-5 expression were found to benefit more from VDR ligand therapy (69). Furthermore, the majority of research has focused on the effects of Hic-5 on tumor metastasis, and thus further studies are required to explore the effects of Hic-5 on tumorigenesis.

In future studies, drugs targeting Hic-5 need to be investigated, which may provide novel options for the treatment of tumors. Moreover, Hic-5 promotes CAF protofiber adhesion by direct interaction with Tensin1, and the interaction is through a phosphorylation-dependent mechanism (83). Therefore, Hic-5 plays a key role in CAFs, and future research on drugs targeting the tumor microenvironment using Hic-5 as a target may provide novel avenues for the management and treatment of several types of cancer.

10. Conclusions

As an important component of focal adhesion sites, Hic-5 plays an essential role in cell proliferation, migration, differentiation and apoptosis. It also plays a key role in transcriptional regulation, as well as in the regulation of signaling pathways, and in the suppression of its functions can prevent the development of several diseases. Thus, Hic-5 may serve as a diagnostic biomarker, as well as a therapeutic target for a wide range of diseases, including tumors. Several studies have confirmed the expression of Hic-5 in various organs and its pathological roles in different systems; however, the exact physiological and pathological roles of Hic-5 in several other organs remain unclear and additional studies are required to explore its functions. The present review summarized the role of Hic-5 in a variety of organs and their associated diseases with the aim of providing an up-to-date background on the current body of literature from which future studies may be designed.

Availability of data and materials

Not applicable.

Authors' contributions

SY and ZT drafted the manuscript and contributed equally. XY, LiZhang, YZ, LZheng and HW participated in the literature search and analysis of the data to be included in the review. ZY, JA and HJ were involved in the design of the study and assisted in the preparation of the figures and tables. GW and BT edited and revised the manuscript. All authors have read and approved the final version of the manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

Funding

The present study was supported by grants from the National Natural Science Foundation of China (grant nos. 81960507, 82073087 and 82160112), and the Collaborative Innovation Center of Chinese Ministry of Education (2020-39), the Science and Technology Bureau fund of Zunyi City [grant no. ZUN SHI KE HE HZ ZI (2019)93-Hao] and the Science and Technology Plan Project of Guizhou Province [grant nos. QIAN KE HE JI CHU-ZK (2021) YI BAN451 and QIAN KE HE LH ZI (2017)7095 HAO].

References

1 

Shibanuma M, Mashimo J, Kuroki T and Nose K: Characterization of the TGF beta 1-inducible hic-5 gene that encodes a putative novel zinc finger protein and its possible involvement in cellular senescence. J Biol Chem. 269:26767–26774. 1994. View Article : Google Scholar : PubMed/NCBI

2 

Shibanuma M, Mori K and Nose K: HIC-5: A mobile molecular scaffold regulating the anchorage dependence of cell growth. Int J Cell Biol. 2012:4261382012. View Article : Google Scholar

3 

Kim-Kaneyama JR, Lei XF, Arita S, Miyauchi A, Miyazaki T and Miyazaki A: Hydrogen peroxide-inducible clone 5 (Hic-5) as a potential therapeutic target for vascular and other disorders. J Atheroscler Thromb. 19:601–607. 2012. View Article : Google Scholar : PubMed/NCBI

4 

Yang L, Guerrero J, Hong H, DeFranco DB and Stallcup MR: Interaction of the tau2 transcriptional activation domain of glucocorticoid receptor with a novel steroid receptor coactivator, Hic-5, which localizes to both focal adhesions and the nuclear matrix. Mol Biol Cell. 11:2007–2018. 2000. View Article : Google Scholar : PubMed/NCBI

5 

Kim-Kaneyama JR, Shibanuma M and Nose K: Transcriptional activation of the c-fos gene by a LIM protein, Hic-5. Biochem Biophys Res Commun. 299:360–365. 2002. View Article : Google Scholar : PubMed/NCBI

6 

Shibanuma M, Kim-Kaneyama JR, Sato S and Nose K: A LIM protein, Hic-5, functions as a potential coactivator for Sp1. J Cell Biochem. 91:633–645. 2004. View Article : Google Scholar : PubMed/NCBI

7 

Lei XF, Fu W, Kim-Kaneyama JR, Omoto T, Miyazaki T, Li B and Miyazaki A: Hic-5 deficiency attenuates the activation of hepatic stellate cells and liver fibrosis through upregulation of Smad7 in mice. J Hepatol. 64:110–117. 2016. View Article : Google Scholar

8 

Matsuya M, Sasaki H, Aoto H, Mitaka T, Nagura K, Ohba T, Ishino M, Takahashi S, Suzuki R and Sasaki T: Cell adhesion kinase beta forms a complex with a new member, Hic-5, of proteins localized at focal adhesions. J Biol Chem. 273:1003–1014. 1998. View Article : Google Scholar : PubMed/NCBI

9 

Nishiya N, Shirai T, Suzuki W and Nose K: Hic-5 interacts with GIT1 with a different binding mode from paxillin. J Biochem. 132:279–289. 2002. View Article : Google Scholar : PubMed/NCBI

10 

Fujita H, Kamiguchi K, Cho D, Shibanuma M, Morimoto C and Tachibana K: Interaction of Hic-5, A senescence-related protein, with focal adhesion kinase. J Biol Chem. 273:26516–26521. 1998. View Article : Google Scholar : PubMed/NCBI

11 

Hornigold N, Craven RA, Keen JN, Johnson T, Banks RE and Mooney AF: Upregulation of Hic-5 in glomerulosclerosis and its regulation of mesangial cell apoptosis. Kidney Int. 77:329–338. 2010. View Article : Google Scholar

12 

Mestayer C, Blanchère M, Jaubert F, Dufour B and Mowszowicz I: Expression of androgen receptor coactivators in normal and cancer prostate tissues and cultured cell lines. Prostate. 56:192–200. 2003. View Article : Google Scholar : PubMed/NCBI

13 

Kim-Kaneyama JR, Takeda N, Sasai A, Miyazaki A, Sata M, Hirabayashi T, Shibanuma M, Yamada G and Nose K: Hic-5 deficiency enhances mechanosensitive apoptosis and modulates vascular remodeling. J Mol Cell Cardiol. 50:77–86. 2011. View Article : Google Scholar

14 

Nishiya N, Iwabuchi Y, Shibanuma M, Côté JF, Tremblay ML and Nose K: Hic-5, a paxillin homologue, binds to the protein-tyrosine phosphatase PEST (PTP-PEST) through its LIM 3 domain. J Biol Chem. 274:9847–9853. 1999. View Article : Google Scholar : PubMed/NCBI

15 

López-Colomé AM, Lee-Rivera I, Benavides-Hidalgo R and López E: Paxillin: A crossroad in pathological cell migration. J Hematol Oncol. 10:502017. View Article : Google Scholar : PubMed/NCBI

16 

Ma X and Hammes SR: Paxillin actions in the nucleus. Steroids. 133:87–92. 2018. View Article : Google Scholar :

17 

Xu W, Alpha KM, Zehrbach NM and Turner CE: Paxillin promotes breast tumor collective cell invasion through maintenance of adherens junction integrity. Mol Biol Cell. 33:ar142022. View Article : Google Scholar :

18 

Tanaka N, Minemura C, Asai S, Kikkawa N, Kinoshita T, Oshima S, Koma A, Kasamatsu A, Hanazawa T, Uzawa K and Seki N: Identification of miR-199-5p and miR-199-3p target genes: Paxillin facilities cancer cell aggressiveness in head and neck squamous cell carcinoma. Genes (Basel). 12:19102021. View Article : Google Scholar

19 

Ripamonti M, Wehrle-Haller B and de Curtis I: Paxillin: A hub for mechano-transduction from the β3 integrin-talin-kindlin axis. Front Cell Dev Biol. 10:8520162022. View Article : Google Scholar

20 

Hagel M, George EL, Kim A, Tamimi R, Opitz SL, Turner CE, Imamoto A and Thomas SM: The adaptor protein paxillin is essential for normal development in the mouse and is a critical transducer of fibronectin signaling. Mol Cell Biol. 22:901–915. 2002. View Article : Google Scholar : PubMed/NCBI

21 

Kim-Kaneyama JR, Suzuki W, Ichikawa K, Ohki T, Kohno Y, Sata M, Nose K and Shibanuma M: Uni-axial stretching regulates intracellular localization of Hic-5 expressed in smooth-muscle cells in vivo. J Cell Sci. 118:937–949. 2005. View Article : Google Scholar : PubMed/NCBI

22 

Mashimo J, Shibanuma M, Satoh H, Chida K and Nose K: Genomic structure and chromosomal mapping of the mouse hic-5 gene that encodes a focal adhesion protein. Gene. 249:99–103. 2000. View Article : Google Scholar : PubMed/NCBI

23 

Panetti TS, Hannah DF, Avraamides C, Gaughan JP, Marcinkiewicz C, Huttenlocher A and Mosher DF: Extracellular matrix molecules regulate endothelial cell migration stimulated by lysophosphatidic acid. J Thromb Haemost. 2:1645–1656. 2004. View Article : Google Scholar : PubMed/NCBI

24 

Hetey SE, Lalonde DP and Turner CE: Tyrosine-phosphorylated Hic-5 inhibits epidermal growth factor-induced lamellipodia formation. Exp Cell Res. 311:147–156. 2005. View Article : Google Scholar : PubMed/NCBI

25 

Tumbarello DA, Brown MC and Turner CE: The paxillin LD motifs. FEBS Lett. 513:114–118. 2002. View Article : Google Scholar : PubMed/NCBI

26 

Brown MC and Turner CE: Paxillin: Adapting to change. Physiol Rev. 84:1315–1339. 2004. View Article : Google Scholar : PubMed/NCBI

27 

Nishiya N, Tachibana K, Shibanuma M, Mashimo JI and Nose K: Hic-5-reduced cell spreading on fibronectin: Competitive effects between paxillin and Hic-5 through interaction with focal adhesion kinase. Mol Cell Biol. 21:5332–5345. 2001. View Article : Google Scholar : PubMed/NCBI

28 

Shibanuma M, Kim-Kaneyama JR, Ishino K, Sakamoto N, Hishiki T, Yamaguchi K, Mori K, Mashimo J and Nose K: Hic-5 communicates between focal adhesions and the nucleus through oxidant-sensitive nuclear export signal. Mol Biol Cell. 14:1158–1171. 2003. View Article : Google Scholar : PubMed/NCBI

29 

Heitzer MD and DeFranco DB: Hic-5, an adaptor-like nuclear receptor coactivator. Nucl Recept Signal. 4:e0192006. View Article : Google Scholar : PubMed/NCBI

30 

Chodankar R, Wu DY, Schiller BJ, Yamamoto KR and Stallcup MR: Hic-5 is a transcription coregulator that acts before and/or after glucocorticoid receptor genome occupancy in a gene-selective manner. Proc Natl Acad Sci USA. 111:4007–4012. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Heitzer MD and DeFranco DB: Hic-5/ARA55, a LIM domain-containing nuclear receptor coactivator expressed in prostate stromal cells. Cancer Res. 66:7326–7333. 2006. View Article : Google Scholar : PubMed/NCBI

32 

Ghogomu SM, van Venrooy S, Ritthaler M, Wedlich D and Gradl D: HIC-5 is a novel repressor of lymphoid enhancer factor/T-cell factor-driven transcription. J Biol Chem. 281:1755–1764. 2006. View Article : Google Scholar

33 

Drori S, Girnun GD, Tou L, Szwaya JD, Mueller E, Xia K, Shivdasani RA and Spiegelman BM: Hic-5 regulates an epithelial program mediated by PPARgamma. Genes Dev. 19:362–375. 2005. View Article : Google Scholar : PubMed/NCBI

34 

Heitzer MD and DeFranco DB: Mechanism of action of Hic-5/androgen receptor activator 55, a LIM domain-containing nuclear receptor coactivator. Mol Endocrinol. 20:56–64. 2006. View Article : Google Scholar

35 

Deakin NO and Turner CE: Distinct roles for paxillin and Hic-5 in regulating breast cancer cell morphology, invasion, and metastasis. Mol Biol Cell. 22:327–341. 2011. View Article : Google Scholar :

36 

Vohnoutka RB, Gulvady AC, Goreczny G, Alpha K, Handelman SK, Sexton JZ and Turner CE: The focal adhesion scaffold protein Hic-5 regulates vimentin organization in fibroblasts. Mol Biol Cell. 30:3037–3056. 2019. View Article : Google Scholar : PubMed/NCBI

37 

Paul J, Singh AK, Kathania M, Elviche TL, Zeng M, Basrur V, Theiss AL and Venuprasad K: IL-17-driven intestinal fibrosis is inhibited by Itch-mediated ubiquitination of HIC-5. Mucosal Immunol. 11:427–436. 2018. View Article : Google Scholar

38 

Lei XF, Kim-Kaneyama JR, Arita-Okubo S, Offermanns S, Itabe H, Miyazaki T and Miyazaki A: Identification of Hic-5 as a novel scaffold for the MKK4/p54 JNK pathway in the development of abdominal aortic aneurysms. J Am Heart Assoc. 3:e0007472014. View Article : Google Scholar : PubMed/NCBI

39 

Frangogiannis N: Transforming growth factor-β in tissue fibrosis. J Exp Med. 217:e201901032020. View Article : Google Scholar

40 

Peng D, Fu M, Wang M, Wei Y and Wei X: Targeting TGF-β signal transduction for fibrosis and cancer therapy. Mol Cancer. 21:1042022. View Article : Google Scholar

41 

Gao L, Lei XF, Miyauchi A, Noguchi M, Omoto T, Haraguchi S, Miyazaki T, Miyazaki A and Kim-Kaneyama JR: Hic-5 is required for activation of pancreatic stellate cells and development of pancreatic fibrosis in chronic pancreatitis. Sci Rep. 10:191052020. View Article : Google Scholar : PubMed/NCBI

42 

Shola DT, Wang H, Wahdan-Alaswad R and Danielpour D: Hic-5 controls BMP4 responses in prostate cancer cells through inter-acting with Smads 1,5 and 8. Oncogene. 31:2480–2490. 2012. View Article : Google Scholar

43 

Sha L, Ma D and Chen C: Exosome-mediated Hic-5 regulates proliferation and apoptosis of osteosarcoma via Wnt/β-catenin signal pathway. Aging (Albany NY). 12:23598–23608. 2020. View Article : Google Scholar

44 

Chen H, Tan P, Qian B, Du Y, Wang A, Shi H, Huang Z, Huang S, Liang T and Fu W: Hic-5 deficiency protects cerulein-induced chronic pancreatitis via down-regulation of the NF-κB (p65)/IL-6 signalling pathway. J Cell Mol Med. 24:1488–1503. 2020. View Article : Google Scholar

45 

Gao L, Qian B, Chen H, Wang A, Li Q, Li J, Tan P, Xia X, Du Y and Fu W: Hic-5 deficiency attenuates hepatic ischemia reperfusion injury through TLR4/NF-κB signaling pathways. Life Sci. 249:1175172020. View Article : Google Scholar

46 

Wu JR, Hu CT, You RI, Pan SM, Cheng CC, Lee MC, Wu CC, Chang YJ, Lin SC, Chen CS, et al: Hydrogen peroxide inducible clone-5 mediates reactive oxygen species signaling for hepatocellular carcinoma progression. Oncotarget. 6:32526–32544. 2015. View Article : Google Scholar : PubMed/NCBI

47 

Tumbarello DA and Turner CE: Hic-5 contributes to epithelial-mesenchymal transformation through a RhoA/ROCK-dependent pathway. J Cell Physiol. 211:736–747. 2007. View Article : Google Scholar : PubMed/NCBI

48 

Sheta R, Wang ZQ, Bachvarova M, Plante M, Gregoire J, Renaud MC, Sebastianelli A, Gobeil S, Morin C, Macdonald E, et al: Hic-5 regulates epithelial to mesenchymal transition in ovarian cancer cells in a TGFβ1-independent manner. Oncotarget. 8:82506–82530. 2017. View Article : Google Scholar : PubMed/NCBI

49 

Yuminamochi T, Yatomi Y, Osada M, Ohmori T, Ishii Y, Nakazawa K, Hosogaya S and Ozaki Y: Expression of the LIM proteins paxillin and Hic-5 in human tissues. J Histochem Cytochem. 51:513–521. 2003. View Article : Google Scholar : PubMed/NCBI

50 

Komorowsky C, Samarin J, Rehm M, Guidolin D and Goppelt-Struebe M: Hic-5 as a regulator of endothelial cell morphology and connective tissue growth factor gene expression. J Mol Med (Berl). 88:623–631. 2010. View Article : Google Scholar

51 

Dave JM, Abbey CA, Duran CL, Seo H, Johnson GA and Bayless KJ: Hic-5 mediates the initiation of endothelial sprouting by regulating a key surface metalloproteinase. J Cell Sci. 129:743–756. 2016.PubMed/NCBI

52 

Kim-Kaneyama JR, Miyauchi A, Lei XF, Arita S, Mino T, Takeda N, Kou K, Eto K, Yoshida T, Miyazaki T, et al: Identification of Hic-5 as a novel regulatory factor for integrin αIIbβ3 activation and platelet aggregation in mice. J Thromb Haemost. 10:1867–1874. 2012. View Article : Google Scholar : PubMed/NCBI

53 

Popp M, Thielmann I, Nieswandt B and Stegner D: Normal platelet integrin function in mice lacking hydrogen peroxide-induced clone-5 (Hic-5). PLoS One. 10:e01334292015. View Article : Google Scholar : PubMed/NCBI

54 

Gao J, Huang M, Lai J, Mao K, Sun P, Cao Z, Hu Y, Zhang Y, Schulte ML, Jin C, et al: Kindlin supports platelet integrin αIIbβ3 activation by interacting with paxillin. J Cell Sci. 130:3764–3775. 2017.PubMed/NCBI

55 

Kim-Kaneyama JR, Wachi N, Sata M, Enomoto S, Fukabori K, Koh K, Shibanuma M and Nose K: Hic-5, an adaptor protein expressed in vascular smooth muscle cells, modulates the arterial response to injury in vivo. Biochem Biophys Res Commun. 376:682–687. 2008. View Article : Google Scholar : PubMed/NCBI

56 

Vergallo R and Crea F: Atherosclerotic plaque healing. N Engl J Med. 383:846–857. 2020. View Article : Google Scholar : PubMed/NCBI

57 

Almeida SO and Budoff M: Effect of statins on atherosclerotic plaque. Trends Cardiovasc Med. 29:451–455. 2019. View Article : Google Scholar : PubMed/NCBI

58 

Arita-Okubo S, Kim-Kaneyama JR, Lei XF, Fu WG, Ohnishi K, Takeya M, Miyauchi A, Honda H, Itabe H, Miyazaki T and Miyazaki A: Role of Hic-5 in the formation of microvilli-like structures and the monocyte-endothelial interaction that accelerates atherosclerosis. Cardiovasc Res. 105:361–371. 2015. View Article : Google Scholar : PubMed/NCBI

59 

Yund EE, Hill JA and Keller RS: Hic-5 is required for fetal gene expression and cytoskeletal organization of neonatal cardiac myocytes. J Mol Cell Cardiol. 47:520–527. 2009. View Article : Google Scholar : PubMed/NCBI

60 

Ji J, Yu F, Ji Q, Li Z, Wang K, Zhang J, Lu J, Chen L, E Q, Zeng Y and Ji Y: Comparative proteomic analysis of rat hepatic stellate cell activation: A comprehensive view and suppressed immune response. Hepatology. 56:332–349. 2012. View Article : Google Scholar : PubMed/NCBI

61 

Vege SS and Chari ST: Chronic pancreatitis. N Engl J Med. 386:869–878. 2022. View Article : Google Scholar : PubMed/NCBI

62 

Jamba A, Kondo S, Urushihara M, Nagai T, Kim-Kaneyama JR, Miyazaki A and Kagami S: Hydrogen peroxide-inducible clone-5 regulates mesangial cell proliferation in proliferative glomerulonephritis in mice. PLoS One. 10:e01227732015. View Article : Google Scholar : PubMed/NCBI

63 

Hornigold N and Mooney A: Extracellular matrix-induced Hic-5 expression in glomerular mesangial cells leads to a prosclerotic phenotype independent of TGF-β. FASEB J. 29:4956–4967. 2015. View Article : Google Scholar : PubMed/NCBI

64 

Li X, Martinez-Ferrer M, Botta V, Uwamariya C, Banerjee J and Bhowmick NA: Epithelial Hic-5/ARA55 expression contributes to prostate tumorigenesis and castrate responsiveness. Oncogene. 30:167–177. 2011. View Article : Google Scholar :

65 

Cárdenas S, Colombero C, Panelo L, Dakarapu R, Falck JR, Costas MA and Nowicki S: GPR75 receptor mediates 20-HETE-signaling and metastatic features of androgen-insensitive prostate cancer cells. Biochim Biophys Acta Mol Cell Biol Lipids. 1865:1585732020. View Article : Google Scholar :

66 

Lee BH and Stallcup MR: Different chromatin and DNA sequence characteristics define glucocorticoid receptor binding sites that are blocked or not blocked by coregulator Hic-5. PLoS One. 13:e01969652018. View Article : Google Scholar : PubMed/NCBI

67 

Leach DA, Need EF, Trotta AP, Grubisha MJ, DeFranco DB and Buchanan G: Hic-5 influences genomic and non-genomic actions of the androgen receptor in prostate myofibroblasts. Mol Cell Endocrinol. 384:185–199. 2014. View Article : Google Scholar : PubMed/NCBI

68 

Li P, Yu X, Ge K, Melamed J, Roeder RG and Wang Z: Heterogeneous expression and functions of androgen receptor co-factors in primary prostate cancer. Am J Pathol. 161:1467–1474. 2002. View Article : Google Scholar : PubMed/NCBI

69 

Solomon JD, Heitzer MD, Liu TT, Beumer JH, Parise RA, Normolle DP, Leach DA, Buchanan G and DeFranco DB: VDR activity is differentially affected by Hic-5 in prostate cancer and stromal cells. Mol Cancer Res. 12:1166–1180. 2014. View Article : Google Scholar : PubMed/NCBI

70 

Mao X, Xu J, Wang W, Liang C, Hua J, Liu J, Zhang B, Meng C, Yu X and Shi S: Crosstalk between cancer-associated fibroblasts and immune cells in the tumor microenvironment: New findings and future perspectives. Mol Cancer. 20:1312021. View Article : Google Scholar : PubMed/NCBI

71 

Gulvady AC, Dubois F, Deakin NO, Goreczny GJ and Turner CE: Hic-5 expression is a major indicator of cancer cell morphology, migration, and plasticity in three-dimensional matrices. Mol Biol Cell. 29:1704–1717. 2018. View Article : Google Scholar : PubMed/NCBI

72 

Shen T and Guo Q: Role of Pyk2 in human cancers. Med Sci Monit. 24:8172–8182. 2018. View Article : Google Scholar : PubMed/NCBI

73 

Sun CK, Ng KT, Lim ZX, Cheng Q, Lo CM, Poon RT, Man K, Wong N and Fan ST: Proline-rich tyrosine kinase 2 (Pyk2) promotes cell motility of hepatocellular carcinoma through induction of epithelial to mesenchymal transition. PLoS One. 6:e188782011. View Article : Google Scholar : PubMed/NCBI

74 

Qian B, Wei L, Yang Z, He Q, Chen H, Wang A, Yang D, Li Q, Li J, Zheng S and Fu W: Hic-5 in pancreatic stellate cells affects proliferation, apoptosis, migration, invasion of pancreatic cancer cells and postoperative survival time of pancreatic cancer. Biomed Pharmacother. 121:1093552020. View Article : Google Scholar

75 

Hanahan D: Hallmarks of cancer: New dimensions. Cancer Discov. 12:31–46. 2022. View Article : Google Scholar : PubMed/NCBI

76 

Biffi G and Tuveson DA: Diversity and biology of cancer-associated fibroblasts. Physiol Rev. 101:147–176. 2021. View Article : Google Scholar :

77 

Sun X, He X, Zhang Y, Hosaka K, Andersson P, Wu J, Wu J, Jing X, Du Q, Hui X, et al: Inflammatory cell-derived CXCL3 promotes pancreatic cancer metastasis through a novel myofibroblast-hijacked cancer escape mechanism. Gut. 71:129–147. 2022. View Article : Google Scholar

78 

Zhang M, Liu ZZ, Aoshima K, Cai WL, Sun H, Xu T, Zhang Y, An Y, Chen JF, Chan LH, et al: CECR2 drives breast cancer metastasis by promoting NF-κB signaling and macrophage-mediated immune suppression. Sci Transl Med. 14:eabf54732022. View Article : Google Scholar

79 

Du X, Xu Q, Pan D, Xu D, Niu B, Hong W, Zhang R, Li X and Chen S: HIC-5 in cancer-associated fibroblasts contributes to esophageal squamous cell carcinoma progression. Cell Death Dis. 10:8732019. View Article : Google Scholar : PubMed/NCBI

80 

Hernandez-Quiles M, Broekema MF and Kalkhoven E: PPARgamma in metabolism, immunity, and cancer: Unified and diverse mechanisms of action. Front Endocrinol (Lausanne). 12:6241122021. View Article : Google Scholar

81 

Omoto T, Kim-Kaneyama JR, Lei XF, Orimo A, Ohnishi K, Yoshihara K, Miyauchi A, Li S, Gao L, Umemoto T, et al: The impact of stromal Hic-5 on the tumorigenesis of colorectal cancer through lysyl oxidase induction and stromal remodeling. Oncogene. 37:1205–1219. 2018. View Article : Google Scholar

82 

Goreczny GJ, Ouderkirk-Pecone JL, Olson EC, Krendel M and Turner CE: Hic-5 remodeling of the stromal matrix promotes breast tumor progression. Oncogene. 36:2693–2703. 2017. View Article : Google Scholar :

83 

Goreczny GJ, Forsythe IJ and Turner CE: Hic-5 regulates fibrillar adhesion formation to control tumor extracellular matrix remodeling through interaction with tensin1. Oncogene. 37:1699–1713. 2018. View Article : Google Scholar : PubMed/NCBI

84 

Noguchi F, Inui S, Nakajima T and Itami S: Hic-5 affects proliferation, migration and invasion of B16 murine melanoma cells. Pigment Cell Melanoma Res. 25:773–782. 2012. View Article : Google Scholar : PubMed/NCBI

85 

Caltagarone J, Hamilton RL, Murdoch G, Jing Z, DeFranco DB and Bowser R: Paxillin and hydrogen peroxide-inducible clone 5 expression and distribution in control and Alzheimer disease hippocampi. J Neuropathol Exp Neurol. 69:356–371. 2010. View Article : Google Scholar : PubMed/NCBI

86 

Miyauchi A, Kim-Kaneyama JR, Lei XF, Chang SH, Saito T, Haraguchi S, Miyazaki T and Miyazaki A: Alleviation of murine osteoarthritis by deletion of the focal adhesion mechanosensitive adapter, Hic-5. Sci Rep. 9:157702019. View Article : Google Scholar : PubMed/NCBI

87 

Inui S, Fukuzato Y, Nakajima T, Kurata S and Itami S: Androgen receptor co-activator Hic-5/ARA55 as a molecular regulator of androgen sensitivity in dermal papilla cells of human hair follicles. J Invest Dermatol. 127:2302–2306. 2007. View Article : Google Scholar : PubMed/NCBI

88 

Dabiri G, Tumbarello DA, Turner CE and Van de Water L: Hic-5 promotes the hypertrophic scar myofibroblast phenotype by regulating the TGF-beta1 autocrine loop. J Invest Dermatol. 128:2518–2525. 2008. View Article : Google Scholar : PubMed/NCBI

89 

Inui S, Shono F, Noguchi F, Nakajima T, Hosokawa K and Itami S: In vitro and in vivo evidence of pathogenic roles of Hic-5/ARA55 in keloids through Smad pathway and profibrotic transcription. J Dermatol Sci. 58:152–154. 2010. View Article : Google Scholar : PubMed/NCBI

90 

Piera-Velazquez S, Fertala J, Huaman-Vargas G, Louneva N and Jiménez SA: Increased expression of the transforming growth factor β-inducible gene HIC-5 in systemic sclerosis skin and fibroblasts: A novel antifibrotic therapeutic target. Rheumatology (Oxford). 59:3092–3098. 2020. View Article : Google Scholar

91 

Reina-Torres E, De Ieso ML, Pasquale LR, Madekurozwa M, van Batenburg-Sherwood J, Overby DR and Stamer WD: The vital role for nitric oxide in intraocular pressure homeostasis. Prog Retin Eye Res. 83:1009222021. View Article : Google Scholar :

92 

Nair KS, Srivastava C, Brown RV, Koli S, Choquet H, Kang HS, Kuo YM, Grimm SA, Sutherland C, Badea A, et al: GLIS1 regulates trabecular meshwork function and intraocular pressure and is associated with glaucoma in humans. Nat Commun. 12:48772021. View Article : Google Scholar : PubMed/NCBI

93 

Pattabiraman PP and Rao PV: Hic-5 regulates actin cytoskeletal reorganization and expression of fibrogenic markers and myocilin in trabecular meshwork cells. Invest Ophthalmol Vis Sci. 56:5656–5669. 2015. View Article : Google Scholar : PubMed/NCBI

94 

You Q, Duan L, Wang F, Du X and Xiao M: Characterization of the inhibition of vein graft intimal hyperplasia by a biodegradable vascular stent. Cell Biochem Biophys. 59:99–107. 2011. View Article : Google Scholar

95 

Low EL, Baker AH and Bradshaw AC: TGFβ, smooth muscle cells and coronary artery disease: A review. Cell Signal. 53:90–101. 2019. View Article : Google Scholar :

96 

Bai H, Lee JS, Hu H, Wang T, Isaji T, Liu S, Guo J, Liu H, Wolf K, Ono S, et al: Transforming growth factor-β1 inhibits pseudoaneurysm formation after aortic patch angioplasty. Arterioscler Thromb Vasc Biol. 38:195–205. 2018. View Article : Google Scholar

97 

Goumans MJ and Ten Dijke P: TGF-β signaling in control of cardiovascular function. Cold Spring Harb Perspect Biol. 10:a0222102018. View Article : Google Scholar

98 

Boers W, Aarrass S, Linthorst C, Pinzani M, Elferink RO and Bosma P: Transcriptional profiling reveals novel markers of liver fibrogenesis: Gremlin and insulin-like growth factor-binding proteins. J Biol Chem. 281:16289–16295. 2006. View Article : Google Scholar : PubMed/NCBI

99 

Gough NR, Xiang X and Mishra L: TGF-β signaling in liver, pancreas, and gastrointestinal diseases and cancer. Gastroenterology. 161:434–452.e15. 2021. View Article : Google Scholar

100 

Nogueira MA, Coelho AM, Sampietre SN, Patzina RA, Pinheiro da Silva F, D'Albuquerque LA and Machado MC: Beneficial effects of adenosine triphosphate-sensitive K+ channel opener on liver ischemia/reperfusion injury. World J Gastroenterol. 20:15319–15326. 2014. View Article : Google Scholar : PubMed/NCBI

101 

Ma C, Kesarwala AH, Eggert T, Medina-Echeverz J, Kleiner DE, Jin P, Stroncek DF, Terabe M, Kapoor V, ElGindi M, et al: NAFLD causes selective CD4(+) T lymphocyte loss and promotes hepatocarcinogenesis. Nature. 531:253–257. 2016. View Article : Google Scholar : PubMed/NCBI

102 

Shen Y, Wei Y, Wang Z, Jing Y, He H, Yuan J, Li R, Zhao Q, Wei L, Yang T and Lu J: TGF-β regulates hepatocellular carcinoma progression by inducing Treg cell polarization. Cell Physiol Biochem. 35:1623–1632. 2015. View Article : Google Scholar

103 

Caja L, Dituri F, Mancarella S, Caballero-Diaz D, Moustakas A, Giannelli G and Fabregat I: TGF-β and the tissue microenvironment: Relevance in fibrosis and cancer. Int J Mol Sci. 19:12942018. View Article : Google Scholar

104 

Bansod S, Doijad N and Godugu C: Berberine attenuates severity of chronic pancreatitis and fibrosis via AMPK-mediated inhibition of TGF-β1/Smad signaling and M2 polarization. Toxicol Appl Pharmacol. 403:1151622020. View Article : Google Scholar

105 

Gore J, Imasuen-Williams IE, Conteh AM, Craven KE, Cheng M and Korc M: Combined targeting of TGF-β, EGFR and HER2 suppresses lymphangiogenesis and metastasis in a pancreatic cancer model. Cancer Lett. 379:143–153. 2016. View Article : Google Scholar : PubMed/NCBI

106 

Song W, Dai WJ, Zhang MH, Wang H and Yang XZ: Comprehensive analysis of the expression of TGF-β signaling regulators and prognosis in human esophageal cancer. Comput Math Methods Med. 2021:18122272021. View Article : Google Scholar

107 

Calon A, Lonardo E, Berenguer-Llergo A, Espinet E, Hernando-Momblona X, Iglesias M, Sevillano M, Palomo-Ponce S, Tauriello DV, Byrom D, et al: Stromal gene expression defines poor-prognosis subtypes in colorectal cancer. Nat Genet. 47:320–329. 2015. View Article : Google Scholar : PubMed/NCBI

108 

Tauriello DVF, Palomo-Ponce S, Stork D, Berenguer-Llergo A, Badia-Ramentol J, Iglesias M, Sevillano M, Ibiza S, Cañellas A, Hernando-Momblona X, et al: TGFβ drives immune evasion in genetically reconstituted colon cancer metastasis. Nature. 554:538–543. 2018. View Article : Google Scholar : PubMed/NCBI

109 

Meng XM: Inflammatory mediators and renal fibrosis. Adv Exp Med Biol. 1165:381–406. 2019. View Article : Google Scholar : PubMed/NCBI

110 

Gu YY, Liu XS, Huang XR, Yu XQ and Lan HY: Diverse role of TGF-β in kidney disease. Front Cell Dev Biol. 8:1232020. View Article : Google Scholar

111 

Wang H, Song K, Krebs TL, Yang J and Danielpour D: Smad7 is inactivated through a direct physical interaction with the LIM protein Hic-5/ARA55. Oncogene. 27:6791–6805. 2008. View Article : Google Scholar : PubMed/NCBI

112 

Thompson-Elliott B, Johnson R and Khan SA: Alterations in TGFβ signaling during prostate cancer progression. Am J Clin Exp Urol. 9:318–328. 2021.

113 

Mirzaei S, Paskeh MDA, Saghari Y, Zarrabi A, Hamblin MR, Entezari M, Hashemi M, Aref AR, Hushmandi K, Kumar AP, et al: Transforming growth factor-beta (TGF-β) in prostate cancer: A dual function mediator? Int J Biol Macromol. 206:435–452. 2022. View Article : Google Scholar : PubMed/NCBI

114 

Huang M, Fu M, Wang J, Xia C, Zhang H, Xiong Y, He J, Liu J, Liu B, Pan S and Liu F: TGF-β1-activated cancer-associated fibroblasts promote breast cancer invasion, metastasis and epithelial-mesenchymal transition by autophagy or overexpression of FAP-α. Biochem Pharmacol. 188:1145272021. View Article : Google Scholar

115 

Roane BM, Arend RC and Birrer MJ: Review: Targeting the transforming growth factor-beta pathway in ovarian cancer. Cancers (Basel). 11:6682019. View Article : Google Scholar

116 

Lamora A, Talbot J, Bougras G, Amiaud J, Leduc M, Chesneau J, Taurelle J, Stresing V, Le Deley MC, Heymann MF, et al: Overexpression of smad7 blocks primary tumor growth and lung metastasis development in osteosarcoma. Clin Cancer Res. 20:5097–5112. 2014. View Article : Google Scholar : PubMed/NCBI

117 

Xu S, Yang S, Sun G, Huang W and Zhang Y: Transforming growth factor-beta polymorphisms and serum level in the development of osteosarcoma. DNA Cell Biol. 33:802–806. 2014. View Article : Google Scholar : PubMed/NCBI

118 

Bu MT, Chandrasekhar P, Ding L and Hugo W: The roles of TGF-β and VEGF pathways in the suppression of antitumor immunity in melanoma and other solid tumors. Pharmacol Ther. 240:1082112022.Epub ahead of print. View Article : Google Scholar

119 

Zheng C, Zhou XW and Wang JZ: The dual roles of cytokines in Alzheimer's disease: Update on interleukins, TNF-α, TGF-β and IFN-γ. Transl Neurodegener. 5:72016. View Article : Google Scholar

120 

Park JK, Lee KJ, Kim JY and Kim H: The association of blood-based inflammatory factors IL-1β, TGF-β and CRP with cognitive function in Alzheimer's disease and mild cognitive impairment. Psychiatry Investig. 18:11–18. 2021. View Article : Google Scholar : PubMed/NCBI

121 

van der Kraan PM: The changing role of TGFβ in healthy, ageing and osteoarthritic joints. Nat Rev Rheumatol. 13:155–163. 2017. View Article : Google Scholar : PubMed/NCBI

122 

Kuźnik-Trocha K, Winsz-Szczotka K, Komosińska-Vassev K, Jura-Półtorak A, Kotulska-Kucharz A, Kucharz EJ, Kotyla P and Olczyk K: Plasma glycosaminoglycan profiles in systemic sclerosis: Associations with MMP-3, MMP-10, TIMP-1, TIMP-2, and TGF-beta. Biomed Res Int. 2020:64165142020. View Article : Google Scholar

123 

Wu WS, Ling CH, Lee MC, Cheng CC, Chen RF, Lin CF, You RI and Chen YC: Targeting Src-Hic-5 signal cascade for preventing migration of cholangiocarcinoma cell HuCCT1. Biomedicines. 10:10222022. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2022
Volume 50 Issue 5

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yao S, Tu Z, Yang X, Zhang L, Zhong Y, Zheng L, Wang H, Yi Z, An J, Jin H, Jin H, et al: Physiological and pathological roles of Hic‑5 in several organs (Review). Int J Mol Med 50: 138, 2022
APA
Yao, S., Tu, Z., Yang, X., Zhang, L., Zhong, Y., Zheng, L. ... Tuo, B. (2022). Physiological and pathological roles of Hic‑5 in several organs (Review). International Journal of Molecular Medicine, 50, 138. https://doi.org/10.3892/ijmm.2022.5194
MLA
Yao, S., Tu, Z., Yang, X., Zhang, L., Zhong, Y., Zheng, L., Wang, H., Yi, Z., An, J., Jin, H., Wen, G., Tuo, B."Physiological and pathological roles of Hic‑5 in several organs (Review)". International Journal of Molecular Medicine 50.5 (2022): 138.
Chicago
Yao, S., Tu, Z., Yang, X., Zhang, L., Zhong, Y., Zheng, L., Wang, H., Yi, Z., An, J., Jin, H., Wen, G., Tuo, B."Physiological and pathological roles of Hic‑5 in several organs (Review)". International Journal of Molecular Medicine 50, no. 5 (2022): 138. https://doi.org/10.3892/ijmm.2022.5194