Open Access

Role of four and a half LIM domain protein 1 in tumors (Review)

  • Authors:
    • Yun Tao
    • Yaxun Wu
    • Rong Shen
    • Song He
    • Xiaobing Miao
  • View Affiliations

  • Published online on: October 29, 2024     https://doi.org/10.3892/ol.2024.14783
  • Article Number: 37
  • Copyright: © Tao et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

As a cytoskeletal protein, the four and a half LIM domain protein 1 (FHL1) is widely expressed in various cells, particularly skeletal and cardiac muscle cells. FHL1 is involved in the development of the skeletal muscle and myocardium, regulations of gene transcription and thyroid function, and other physiological processes. Its expression is closely related to numerous diseases, such as skeletal muscle disease and viral infections. With the advances in research, the role of FHL1 in the development of tumors is also being revealed. The mechanism of FHL1 in the regulation of tumor growth is complex and is becoming a research focus. It is also expected to become a potential target for tumor therapy. Therefore, the present article reviewed the progress in research on the role of FHL1 in cancer.

Introduction

The four and a half LIM domain protein 1 (FHL1) belongs to the FHL family and is located in Xq26.3. It is also known as SLIM-1 or KYO-T (1,2).

As a cytoskeletal protein, FHL1 is widely expressed in humans. Relevant studies have shown that it is mainly expressed in the skeletal muscle and myocardium. FHL1 regulates cell proliferation, differentiation, apoptosis, adhesion, migration, transcription and other cellular processes, and plays an important role in cell growth (3).

FHL1 is composed of four and a half LIM domains and three subtypes have been identified. The LIM domain of FHL1 is responsible for mediating interactions between proteins. Each domain of FHL1 has a different regulatory role in protein function. Mutations in LIM2 and LIM4 cause a loss of function in FHL1, and the function of FHL1 depends on the integrity of its various domains (4).

FHL1 also plays an important regulatory role in the growth of numerous tumors, in addition to affecting the occurrence and development of skeletal muscle and myocardial diseases. In different tumors, the expression of FHL1 is upregulated or downregulated and plays a role in promoting or inhibiting tumor development. Owing to the wide expression of FHL1 in numerous tumors and its different regulatory roles in tumor development, FHL1 has become a hot topic in the field of tumor research. The present article reviewed the role of FHL1 in tumors.

Domain architectures of FHL1

A total of 8 exons were found in FHL1 gene and exons 1 and 2 are non-coding. The structure of FHL1 is characterized by the structure of the LIM domain. The N-terminus is a half LIM domain, followed by four complete LIM domains. The LIM domain was first identified during the isolation and identification of the LIN1 gene in Caenorhabditis elegans, the ISL-1 gene in rats and the MEC-1 gene in C. elegans (5). The consensus amino acid sequence of the LIM domains has been defined as Cys-X2-Cys-X16-23-His-X2-Cys-X2-Cys-X2-Cys-X16-21-Cys-X2-Cys/His/Asp, where X represents any amino acid.

The LIM domain is an important sequence mediating interactions between proteins. The structure of the LIM domain is highly conserved. It mediates the interaction between FHL1 and transcriptional regulatory factors, kinases and structural proteins such as receptor-interacting protein140 (RIP140), cytosolic tyrosine kinase Src and α-tubulin.

The alternative splicing results in the existence of three subtypes of FHL1: FHL1A is a complete FHL1 protein; FHL1B lacks the last LIM domain, which includes nuclear localization, output signal and RBP-J binding domain; and FHL1C lacks the last two LIM domains, which includes the RBP-J binding domain, and has no nuclear location signal. The RBP-J binding domain binds to RBP-J and thus inhibits the binding of RBP-J to DNA and further inhibits its transcriptional activity. The structures of FHL1A, FHL1B and FHL1C are shown in Fig. 1. Studies have indicated that FHL1A is mainly expressed in the skeletal muscle, myocardium and fibroblasts, and FHL1B/FHL1C are mainly expressed in the muscle, brain and testis (6).

The LIM domain, which is composed of ~55 amino acid residues, binds to zinc and is rich in cysteine. It also plays an important role in protein transcription and signal transduction (7). Its cysteine-rich double zinc fingers contain the consensus sequence X2:CX2CX3I–X11-15WHX2CFXCX2CX3(I/L)X4(F/Y)X8CX2C (8). The consensus sequence facilitates protein-protein interactions. The highly conserved cysteine and histidine residues mediate Zn2+ binding, thereby stabilizing the folding and structure of the LIM domain (9,10). LIM domain is able to form homo- or heterodimers via interacting with other domains. It also binds to motifs such as PDZ domains and helix-loop-helix domains and performs its function. Multiple mutations in FHL1 affect its function and are associated with various skeletal and cardiac diseases. Relevant studies have shown that FHL1 mutations mostly occur in the second and fourth LIM domains (11).

The expression of FHL1 is also affected by a variety of post-transcriptional modulations; for example, methylation silences the expression of FHL1. The deubiquitinating enzyme 15 (USP15) stabilizes FHL1 expression by regulating the ubiquitin proteasome (12).

As an important cytoskeletal protein, the structure of FHL1 and the physiological functions of each domain require further study.

FHL1 in normal physiology

FHL1 performs its physiological functions by interacting with a variety of molecules. The proteins that interact with FHL1 include structural proteins, signal transduction proteins, transcription regulators, receptors and channels, such as ERK and non-structural protein 3 (Nsp3) (13).

FHL1 plays a role in the regulation of gene transcription, viral infection, thyroid function, blood glucose levels, myoblast differentiation and other physiological processes. Its abnormal expression is closely related to numerous diseases of the skeletal muscle and myocardium (1417).

FHL1 is closely associated with gene transcription. For instance, FHL1A interacts with Gβγ- and Gαs-dependent guanine nucleotide exchange factor pleckstrin homology and RhoGEF domain containing G2 to enhance its induced serum response element (Ser)-dependent gene transcription (14).

FHL1 interacts with Pol1 transcription factor HMO1 to regulate ribosome synthesis. Interacts with Forkhead 1 (IFH1) is a weak multicopy suppressor with FHL1 deletion, and there is an interaction between FHL1 and the coactivator IFH1. TOR promotes this interaction and inhibits the interaction between FHL1 and the corepressor CRF1 to regulate the expression of ribosomal protein genes (18).

The expression of FHL1 is also associated with viral infections. For instance, FHL1 promotes the development of chikungunya and cashmere virus infection (19). The interaction between Nsp3 (HVD) and FHL1 is important for the proviral function of FHL1 (20). In addition, FHL1 interacts with the viral protein Nsp3 to regulate viral RNA replication. It is also highly expressed in chikungunya virus (CHIKV) target muscle cells and fibroblasts, and plays an important role in CHIKV gene expansion. FHL1 knockdown confers resistance to viral infections (21).

Mutations in FHL1 are associated with skeletal muscle and myocardial diseases. For instance, c.370–375del leads to reductive myopathy and c.763T>c,p.Cys255Arg is associated with hypertrophic cardiomyopathy (22,23).

Role of FHL1 in tumors

FHL1 is abnormally expressed in tumors and the expression of FHL1 is not consistent among different tumors. It promotes or inhibits the tumor development by upregulating or downregulating its expression. Simultaneously, the expression of FHL1 in tumors is regulated by epigenetic modifications. Therefore, FHL1 is a potential target in cancer research.

Dysregulation of FHL1 in tumors
Low expression of FHL1 in tumors

The expression of FHL1 is downregulated in cancer types including lung, prostate, breast, ovarian, colon, thyroid, brain, kidney, liver and oral cancers, as well as melanoma (24,25).

In non-small cell lung cancer, a decrease in long intergenic noncoding RNA (LINC)00261, which has a tumor suppressor function, leads to an increased expression of miR-105, which leads to a decrease in FHL1 expression (26,27). The expression level of FHL1 correlates with that of numerous immune cells. For instance, its expression level is positively correlated with the levels of monocytes, eosinophils and neutrophils, and negatively correlated with the level of M0 macrophages (28). Increased FHL1 expression in lung adenocarcinoma predicts a good prognosis (29). In lung adenocarcinoma, the expression of FHL1 is lower than that in normal lung tissue, and patients with relatively high expression have longer overall survival (30). In papillary thyroid microcarcinoma (PTMC), the expression of FHL1 in PTMC is lower than that in normal tissues, and the expression of FHL1 is significantly lower in PTC than in PTMC (31). In gastrointestinal tumors, the FHL1 promoter region is methylated, which silences the expression of FHL1. The expression of FHL1 in gastric cancer tissues is decreased compared to that in normal mucosa. In addition, more invasion and metastasis are found in these patients (24). In liver and colorectal tumors, miR-410 is highly expressed and downregulates the expression of FHL1 to promote tumor growth (27,32,33). In breast cancer, miR-183-5p downregulates the expression of FHL1, resulting in enhanced activity of breast cancer cells (32). In head and neck and esophageal squamous cell carcinomas, FHL1 expression is downregulated, and the prognosis of these patients is poor (2,34). In pediatric acute myelocytic leukemia (AML) with FLT3-ITD mutations, high expression of FHL1 predicts poor prognosis (35).

High expression of FHL1 in tumors

The expression of FHL1 is increased in laryngeal carcinomas (36). Increased FHL1 expression has been associated with gastric signet ring cell carcinoma (37). In addition, the expression of FHL1 in patients with acute promyelocytic leukemia and AML without FLT3 mutations or PML/RAR fusion is positively correlated with the expression of Rho-related BTB domain 2, which is an indicator of poor prognosis (38). High FHL1 expression promotes glioblastoma development (39).

FHL1 as a prognostic marker

Significant differences were observed in the expressions of FHL1 between different tumors. Its expression is closely related to the prognosis of patients with various tumors. Via causing the activation of TGF-β and other signaling pathways, promoting or inhibiting the expression of tumor proliferation-related proteins such as putative specific protein 1 (SP1), FHL1 influences the proliferation and migration of tumor cells, thus resembling a prognostic marker in tumors. FHL1 is also regulated by posttranslational modifications, such as phosphorylation and methylation, thus influencing the prognosis of bladder and other tumors (40). FHL1 acts through different pathways in different tumors, thus predicting different prognoses in different tumors. Increased FHL1 expression in lung adenocarcinoma, gastric cancer, colorectal tumors, head and neck squamous cell carcinoma, glioma, liver, breast and papillary thyroid cancers (PTCs), and esophageal squamous cell carcinoma, predicts a good prognosis. A high FHL1 expression in glioblastoma and pediatric AML with FLT3-ITD mutations predicts a poor prognosis. To conclude, the expression of FHL1 is closely related to the prognoses and clinical indicators of different tumors and the role of FHL1 in different tumors is summarized in Table I.

Table I.

FHL1 as a prognostic marker in tumors.

Table I.

FHL1 as a prognostic marker in tumors.

Tumor typeFHL1 expressionTargetMechanismPrognosis(Refs.)
GlioblastomaUpregulatedSP1Promoting cell proliferationPoor(46)
Acute myelocytic leukemiaUpregulatedCD14 CD11bPromoting cell proliferationPoor(39)
Gastric cancerDownregulatedCCDC43Inhibiting cell proliferationGood(51)
Colorectal tumorDownregulatedGSK3βInhibiting cell proliferationGood(3)
GliomaDownregulatedAKTInhibiting cell proliferationGood(5)
Head and neck squamous cell carcinomaDownregulatedCyclin D1 Cyclin E p27Inhibiting cell proliferationGood(2)
Liver cancerDownregulatedSmad2Inhibiting cell proliferationGood(58)
Breast cancerDownregulatedVEGF N-cadherin vimentinInhibiting migration and invasionGood(32)
Lung cancerDownregulatedRhoGDIβInhibiting migration and invasionGood(48)
Papillary thyroid cancerDownregulatedWnt/β-cateninInhibiting cell proliferationGood(59)

[i] FHL1, four and a half LIM domain protein 1; SP1, putative specific protein 1; CCDC43, coiled-coil domain-containing protein 43; GSK3β, glycogen synthase kinase-3β; VEGF, vascular endothelial growth factor; RhoGDIβ, Rho GDP-dissociation inhibitor β.

Regulation of FHL1 signaling

The expression of FHL1 is influenced by several factors. Its expression is regulated by miRNA, Src and other proteins, as well as by methylation, ubiquitination and other post-translational modifications.

Interaction with RIP140

In breast cancer, FHL1 interacts with RIP140, in which process all of the domains of FHL1 are required. Overexpression of FHL1 has a synergistic effect on the inhibition of RIP140 in estrogen signal transduction (41).

Mediation by immortalization-upregulated protein (IMUP)

IMUP is highly expressed in pancreatic ductal adenocarcinoma and knockdown of IMUP prevents tumor cell growth. IMUP and nucleophosmin (NPMI) are co-localized in the cell nucleus. IMUP inhibits FHL1 transcription via NPMI-induced promoter methylation. Downregulation of IMUP decreases the expression of CyclinA2, cyclinE1 and cyclin-dependent kinase (CDK)2 by upregulating FHL1 expression. FHL1 regulates CyclinA2, cyclinE1 and CDK2 by promoting the degradation of CDC25A through phosphorylation (40).

Mediation by Src

Src is a membrane-bound tyrosine kinase. Src phosphorylates Crk-associated substrate and thus reduces FHL1 expression. In breast, kidney and prostate cancers, the expression of FHL1 is inhibited, possibly owing to gene methylation. FHL1 induces the expression of serum deprivation response factor in Src-transformed cells, whereas the expression of both molecules is inhibited in tumors (2,42).

Silencing by miR-410, enhancer of zeste homolog 2 (EZH2) and methylation

FHL1 may be silenced by miR-410 or EZH2 (30). Methylation of the FHL1 promoter region has been observed in gastrointestinal tumors and oral squamous cell carcinoma-derived cells, which silences the expression of FHL1 (43,44).

FHL1 signaling in cell proliferation and cell cycle

The role of FHL1 in tumor growth is complex and this effect varies among different tumors (Fig. 2). For instance, FHL1 can inhibit the progression of lung, prostate, ovarian, colon, thyroid, brain, kidney, liver and stomach cancers and promote the progression of glioblastoma. High expression of FHL1 is a risk factor for acute glioma and patients with breast cancer undergoing radiotherapy; however, it can inhibit the progression of colorectal cancer through the Wnt/β-catenin signaling pathway (45).

Promotion of tumor proliferation

FHL1 promotes the proliferation of tumor cells via interacting with SP1, Src and other proteins.

Interacting with the transcription factor SP1

FHL1 and SP1 are co-located in the cell and they form a complex. FHL1 interacts with the transcription factor SP1 to upregulate EGFR expression and activate the downstream signaling pathways, including Src, Akt, ERK1/2 and STAT3, thereby causing glioblastoma. When FHL1 is phosphorylated by Src at Y149 and Y272, it is transformed into an oncogene (46).

Mediated by Src and Kindlin-2

Src interacts with FHL1 through its kinase domain and phosphorylates FHL1 via Y149 and Y272 of FHL1. The LIM4 domain is also involved in this interaction. This process makes it a pro-cancer factor that promotes tumor proliferation. Phosphorylated FHL1 is more likely to enter the nucleus and bind to the transcription factor Bcl2 associated transcription factor 1 (BCLAF1) to promote tumor growth. Kindlin2 also interacts with FHL1 via its FERM domain to inhibit cell growth. The last LIM domain is involved in this interaction. Kindlin2 competes with Src to bind FHL1 and inhibits the SRC-mediated phosphorylation of FHL1 (4648).

Inhibition of tumor proliferation

FHL1 inhibits the growth of colon, bladder and liver tumor cells via mechanisms including blocking hypoxia-inducible factor (HIF)-α-HIF-β dimerization, interacting with coiled-coil domain-containing protein 43 (CCDC43) and regulating the Wnt/β-catenin signaling pathway. For instance, FHL1 knockdown promoted the growth of HeLa and HepG2 cells. Increased FHL1 methylation in bladder and gastrointestinal tumors was observed to lead to decreased FHL1 expression, which promoted tumor cell growth and metastasis (49).

Mediation of HIF-α-HIF-β dimerization

Kruppel-like factor 17 increases the expression of FHL1 by binding to FHL1 promoter and promoting its transcription. This process inhibits the proliferation, invasion and migration of colon tumor cells via influencing the expression of E-cadherin and N-cadherin. In colon cancer, the expression of FHL1 is downregulated, which leads to the proliferation, invasion and migration of tumor cells. As a tumor suppressor gene, FHL1 regulates tumor angiogenesis by blocking HIF-α-HIF-β dimerization and reducing VEGF expression (50).

Interaction with CCDC43

In gastric cancer, CCDC43 and FHL1 are co-localized in perinuclear and nuclear regions and the expression of CCDC43 is negatively correlated with the expression of FHL1. CCDC43 is highly expressed, whereas FHL1 is weakly expressed. There is an interaction between the two molecules. Knocking down CCDC43 improves the stability of FHL1, inhibits the growth and metastasis of gastric cancer cells and promotes apoptosis (51).

Interaction with Smad2, Smad3 and Smad4

FHL1 interacts with Smad2, Smad3 and Smad4 to activate the transcription of p21, inhibit c-Myc transcription and inhibit the growth of liver cancer cells. FHL1 and the tumor suppressor gene Smad4 synergistically inhibit VEGF promoter activity. Furthermore, a reduction in Smad4 abolishes the inhibitory effect of FHL1 on VEGF promoter activity, suggesting that FHL1 inhibits VEGF promoter activity in a Smad4-dependent way (52,53).

Interaction with estrogen receptor (ER)α and ERβ

FHL1 inhibits the growth of breast cancer cells. FHL1 interacts with ERα and ERβ in a 17β-estradiol-independent manner to reduce the transcriptional activity of ERα and ERβ, which is necessary to inhibit estrogen response transcription, resulting in lower expressions of estrogen response genes PS2 and cathepsin D. LIM1, LIM2 and LIM3 of FHL1 are required for this interaction (44,54).

Interaction with OTU domain containing 1 (OTUD1)

The ATR inhibitor VE-822 upregulates the expression level of the deubiquitinating enzyme OTUD1 to stabilize FHL1 expression through its OUT domain and inhibits the progression of lung adenocarcinoma. In this process, the deubiquitinase activity of OTUD1 is necessary. The expression of OTUD1 is downregulated in lung cancer. Similarly, the expression of FHL1 is downregulated and OTUD1 has lost its ability to inhibit lung cancer (55)

Interaction with G protein signal regulatory factor 2 (RGS2)

In tumors, the expression of RGS2 is lower in cancer than in paracancer cells. High expression of RGS2 can inhibit the invasion and proliferation of tumors. FHL1 competes with damage-specific DNA binding protein 1 to bind to RGS2, thereby reducing its ubiquitination level, stabilizing its expression and playing a role in inhibiting tumor growth.

Inhibition of VEGF promoter activity

FHL1 inhibits the VEGF promoter activity, thereby inhibiting VEGF expression. FHL1 interacts with HIF1α vis its LIM domain. The interaction between FHL1 and HIF1α can inhibit the interaction between HIF1α and HIF1β, thereby inhibiting the dimerization of HIF1α and HIF1β. This can further inhibit the binding of HIF1α and VEGF promoter, thereby inhibiting the expression of HIF1α-dependent VEGF (56).

Taking part in the transcription process

Phosphorylated FHL1 binds to the nuclear transcription factor BCLAF1 and takes part in the DNA-binding transcriptional activator activity and transcription factor binding. FHL1 is involved in an IL-15-mediated signaling pathway that activates JAK and STAT family proteins. Its expression is positively correlated with the levels of B, CD8 and CD4 cells and macrophages, and is also correlated with the expression of immune-related genes, such as CD48 and CD80 (57).

Mediation of the Wnt/β-catenin signaling pathway

The expression of FHL1 is downregulated in colorectal tumors and is associated with poor prognosis. FHL1 reduces the phosphorylation of glycogen synthase kinase 3β and activation of β-catenin, thereby negatively regulating the Wnt/β-catenin signaling pathway, causing the downregulation of cyclin D1 and CDK4 and thus inhibiting the proliferation of colon tumor cells (3).

Mediation by miR-410

MiR-410 is highly expressed in liver and colorectal tumors. MiR-410 targets FHL1 via the 3′-UTR of FHL1 and promotes FHL1 methylation, thus negatively regulating FHL1 to promote tumor growth (27,32).

Mediation of Cyclin D1, Cyclin E and p27

In head and neck squamous cell carcinoma, FHL1 expression is downregulated and it promotes tumor cell growth through dysregulation of Cyclin D1, Cyclin E and p27. Decreased FHL1 expression is closely related to poor differentiation. Downregulation of FHL1 expression in head and neck squamous cell carcinoma is usually caused by hypermethylation of its DNA promoter region and EZH2-mediated regulation of histone methylation (2).

Activation of the TGF-β signaling pathway

FHL1 and Smad2 interact in liver cancer cells by phosphorylating Smad2 at Ser465 and Ser467. FHL1 activates the TGF-β signaling pathway in a TGF-β-independent manner, thereby inhibiting tumor growth (58).

Mediation of the cell circle

FHL1 mediates cell cycle in AML and other tumors to influence the development of tumor. The expression of cell cycle-related proteins such as p21 and p27 is regulated in this process.

AML

In AML, FHL1 is highly expressed and promotes the proliferation of AML cells. High FHL1 expression can lead to a decrease in the proportion of cells in G1 phase and a decrease in CDK4. FHL1 knockdown leads to an increase in the proportion of cells in G1 phase. Loss of FHL1 also leads to increased expression of CD11b and CD14. Overexpression of FHL1 prevents all-trans retinoic acid-induced differentiation of HL-60 cells. Consumption of FHL1 inhibits cell proliferation and induces cell differentiation (39).

Lung cancer

The expression of FHL1 in lung tumor tissues is lower than that in normal tissues. FHL1 inhibits the growth of tumor cells by inducing G1 and G2/M phase cell cycle arrest and decreasing the number of cells in S-phase. Overexpression of FHL1 increases the expression of p21 to increase the proportion of cells in G1 and G2/M phases, increases the expression of p27 to promote G1 phase arrest, and decreases the expression of CyclinD1, CyclinB1 and CyclinA (53).

Glioma

FHL1 inhibits the growth of glioma cells and its expression level is negatively associated with prognosis. FHL1 maintains cells in the G0/G1 phase and inhibits tumor growth. When FHL1 is knocked down, cells enter the S phase, which promotes cell growth. FHL1 interacts with AKT and inhibits the binding of AKT and PI3K, thereby inhibiting glioma growth (5).

In conclusion, FHL1 mediates various cellular pathways in tumor progression and mediates tumor growth. FHL1 mainly functions through interacting with proteins, particularly transcription factors such as SP1, to activate downstream signaling pathways. It also affects the transcriptional activity of proteins such as p21 to regulate the growth of tumor cells. Cyclins are also regulated by FHL1 to mediate tumor growth.

FHL1 signaling in tumor invasion and migration

In bladder, breast, lung, papillary thyroid and other cancers, FHL1 inhibits tumor invasion and metastasis by inhibiting the Wnt/β-catenin signaling pathway, and is a good potential therapeutic target. In bladder tumors, the expression of FHL1 is decreased and its methylation level is increased, leading to tumor invasion and migration. Given the complexity of its regulatory mechanisms, the different mechanisms by which FHL1 mediates tumor invasion and migration need to be further explored (49).

Mediation by miR-183-5p

In breast cancer, miR-183-5p downregulates the expression of FHL1 and mediates the upregulation of VEGF, N-cadherin and vimentin, and the downregulation of p53 and E-cadherin, resulting in enhanced activity of breast cancer cells and enhanced invasion and migration abilities (32).

Increase in the expression of RhoGDIβ

The expression of FHL1 is inhibited in lung cancer, thus reducing the expression of RhoGDIβ and leading to tumor invasion (48).

Mediation by miR-96-5p

MiR-96-5p inhibits the expression of FHL1 and promotes the proliferation, invasion and migration of lung adenocarcinomas, resulting in a decrease in the overall survival of patients (27).

Inhibition of the Wnt/β-catenin signaling pathway

FHL1 expression is downregulated in PTC and its expression is negatively associated with T/N staging. Its expression is also related to the progression-free and disease-free intervals. FHL1 inhibits the Wnt/β-catenin signaling pathway, thereby inhibiting the proliferation, invasion and migration of PTC cells (59).

FHL1 signaling in tumor therapy resistance

The comprehensive application of various treatment methods can effectively improve the curative effects in patients with tumors. However, treatment resistance remains an important reason for poor prognosis. FHL1 is abnormally expressed in various tumors and is closely associated with drug resistance during tumor therapy. The mechanism by which it mediates tumor resistance also differs among different tumors.

Induction of radiation resistance

In breast cancer, ionizing radiation can induce the expression of FHL1, thereby inhibiting the activity of CDC25C through its interaction with checkpoint kinase 2 and CDC25, and thus causes radiation resistance (46,60).

Increase of sensitivity to paclitaxel

In hepatocellular carcinoma, knockdown of FHL1 can increase the sensitivity to paclitaxel by inducing the activation of caspase-3 and caspase-9 (46).

Induction of resistance to cytarabin-based induction chemotherapy

High expression of FHL1 is associated with poor prognosis in AML and is an independent prognostic factor. Src phosphorylation of FHL1 promotes cell proliferation. High expression of FHL1 is associated with drug resistance in AML. In patients with AML receiving cytarabin-based induction chemotherapy, high FHL1 predicts a poor prognosis. High FHL1 levels lead to increased expression of ATP-binding cassette, sub-family C (CFTR/MRP), member 1 (ABCC1) and ABCC4, which are associated with the transport of chemotherapy drugs, and which are subsequently excreted into the extracellular system. Simultaneously, the expression of SLC29A1 decreases and the entry of cytarabine into the cell, resulting in drug resistance (61).

Increasing sensitivity to olaparib

In patients with ovarian cancer, tabersonine increases the sensitivity of ovarian cancer cells to the poly ADP-ribose polymerase inhibitor olaparib. In addition, glycyrrhizin increases FHL expression. They act synergistically to decrease epithelial-mesenchymal transformation, thereby inhibiting the invasion and migration of ovarian cancer cells. The combination of tabersonine and olaparib increases the expression of E-cadherin, decreases the expression of N-cadherin and vimentin, and effectively inhibits tumor growth (62).

Inducing resistance to platinum and paclitaxel

In patients with ovarian cancer, low FHL1 expression predicts higher sensitivity to platinum and paclitaxel and a better prognosis. Relevant studies have shown that FHL1 is co-expressed with Filamin C, γ (FLNC), Caveolin-1 and FLNA, and is related to the abundance of macrophages, thus affecting the sensitivity of patients to chemotherapy (45).

In brief, the expression of FHL1 in tumors is complex and the role of FHL1 varies in different tumors, which deserves further study. At the same time, FHL1 has become a potential target in tumor treatment. In different tumors, tumor growth could be inhibited by inducing or inhibiting the expression level of FHL1 or by affecting its post-translational modifications, such as phosphorylation, ubiquitination and methylation (40,44). The expression level of FHL1 could also be used as a reference for estimating the sensitivity to chemoradiotherapy treatment in certain tumors.

However, research on the role of FHL1 in tumor progression and treatment resistance is still limited. The mechanisms of FHL1 affecting tumor cell proliferation and migration have mainly been obtained through the study of cell lines and need to be further verified in clinical samples. There is also a lack of multicenter clinical trials to further increase the reliability of the findings. Furthermore, research methods such as bioinformatics have been rarely used in this field.

Conclusions

FHL1 is an important cytoskeletal protein that is expressed in various cells (6366). It plays a regulatory role in the development of myocardial and skeletal muscle diseases, such as reducing body myopathy and cardiac hypertrophy (6772). It is also widely expressed in tumor cells, including lung, ovarian and liver cancers. In different tumors, FHL1 functions differently to influence the fate of the tumor (73,74). In numerous tumors such as colon cancer and esophageal cancer, FHL1 functions as a tumor suppressor gene that inhibits tumor growth, while it is also reported that FHL1 promotes the development of glioblastoma and other tumors (7577). In the process of tumor occurrence and development, it is regulated by numerous signaling pathways, and it promotes or inhibits tumor occurrence and development by regulating the EGFR and Wnt/β-catenin signaling pathways. Post-transcriptional modifications also affect the function of FHL1. Its expression can be silenced via methylation and it can also be stabilized by deubiquitinating enzymes such as USP15. The regulatory role of FHL1 in tumors is complex and relevant studies suggest that the phosphorylation of FHL1 by Src may play an important regulatory role in the function of FHL1 in tumor growth. When phosphorylated, FHL1 is more likely to be located in the nucleus and facilitates tumor growth.

However, owing to the complexity of its regulatory mechanisms, the role of FHL1 in different tumors has remained to be fully elucidated (7880). The results of the present study suggest that FHL1 can regulate tumor growth by regulating the cell cycle of tumor cells, as well as other signaling pathways such as Wnt/β-catenin and TGF-β. The different roles of FHL1 in different tumors may be caused by factors including regional differences, tumor differences and small sample size. At the same time, the location of FHL1 and its regulatory mechanism is rarely reported. FHL1 was mainly located in the cytoplasm and whether FHL1 has different functions in the nucleus and cytoplasm, and its regulatory mechanism, still warrant further study. The relationship between the location of FHL1 and its role in tumor progression are also a potential research field.

In addition, the subtype of FHL1 expressed in various tumors deserves further study, and the relationship between the expression of various subtypes and tumor occurrence and development has been rarely reported. This could also be a potential target for treatments against FHL1.

Of note, the abnormal expression of FHL1 is also closely related to tumor resistance to radiotherapy and chemotherapy. Therefore, FHL1 is a promising target for cancer research and may serve as an effective therapeutic target. However, in view of its complex roles in various tumors, its mechanism of action in different tumors requires further study. The relationship between the expression level of FHL1 and sensitivity to chemoradiotherapy has only been studied in a small number of tumors. Whether there is a relationship between FHL1 expression and therapeutic resistance in other tumors deserves further investigation.

Acknowledgements

Not applicable.

Funding

This study was supported by the Natural Science Foundation of Nantong (grant no. JC2023038).

Availability of data and materials

Not applicable.

Authors' contributions

XM and SH contributed to the conceptualization of this study and provided constructive guidance. YT and YW collected relevant studies and wrote the manuscript. RS participated in the study design and completion. All authors 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.

References

1 

Han S, Cui C, He H, Shen X, Chen Y, Wang Y, Li D, Zhu Q and Yin H: FHL1 regulates myoblast differentiation and autophagy through its interaction with LC3. J Cell Physiol. 235:4667–4678. 2019. View Article : Google Scholar : PubMed/NCBI

2 

Cao W, Liu J, Xia R, Lin L, Wang X, Xiao M, Zhang C, Li J, Ji T and Chen W: X-linked FHL1 as a novel therapeutic target for head and neck squamous cell carcinoma. Oncotarget. 7:14537–14550. 2016. View Article : Google Scholar : PubMed/NCBI

3 

Liu Y, Wang C, Cheng P, Zhang S, Zhou W, Xu Y, Xu H and Ji G: FHL1 inhibits the progression of colorectal cancer by regulating the Wnt/β-catenin signaling pathway. J Cancer. 12:5345–5354. 2021. View Article : Google Scholar : PubMed/NCBI

4 

Keßler M, Kieltsch A, Kayvanpour E, Katus HA, Schoser B, Schessl J, Just S and Rottbauer W: A zebrafish model for FHL1-opathy reveals Loss-of-function effects of human FHL1 mutations. Neuromuscul Disord. 28:521–531. 2018. View Article : Google Scholar : PubMed/NCBI

5 

Li SZ, Hu YY, Zhao JL, Zang J, Fei Z, Han H and Qin HY: Downregulation of FHL1 protein in glioma inhibits tumor growth through PI3K/AKT signaling. Oncol Lett. 19:3781–3788. 2020.PubMed/NCBI

6 

Ding J, Cong Y, Li F, Liu B, Wu D, Miao J and Wang L: Muscle death participates in myofibrillar abnormalities in FHL1 knockout mice. Biochem Biophys Res Commun. 523:105–111. 2020. View Article : Google Scholar : PubMed/NCBI

7 

Weng J, Liao M, Zou S, Bao J, Zhou J, Qu L, Feng R, Feng X, Zhao Z and Jing Z: Downregulation of FHL1 expression in thoracic aortic dissection: Implications in aortic wall remodeling and pathogenesis of thoracic aortic dissection. Ann Vasc Surg. 25:240–247. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Lee SMY, Tsui SKW, Chan KK, Garcia-Barcelo M, Waye MM, Fung KP, Liew CC and Lee CY: Chromosomal mapping, tissue distribution and cDNA sequence of Four-and-a-half LIM domain protein 1 (FHL1). Gene. 216:163–170. 1998. View Article : Google Scholar : PubMed/NCBI

9 

Wilding BR, McGrath MJ, Bonne G and Mitchell CA: FHL1 mutations that cause clinically distinct human myopathies form protein aggregates and impair myoblast differentiation. J Cell Sci. 127:2269–2281. 2014.PubMed/NCBI

10 

Wei X and Zhang H: Four and a half LIM domains protein 1 can be as a double-edged sword in cancer progression. Cancer Biol Med. 17:270–281. 2020. View Article : Google Scholar : PubMed/NCBI

11 

Selcen D, Bromberg MB, Chin SS and Engel AG: Reducing bodies and myofibrillar myopathy features in FHL1 muscular dystrophy. Neurology. 77:1951–1959. 2011. View Article : Google Scholar : PubMed/NCBI

12 

Isumi Y, Hirata T, Saitoh H, Miyakawa T, Murakami K, Kudoh G, Doi H, Ishibashi K and Nakajima H: Transgenic overexpression of USP15 in the heart induces cardiac remodeling in mice. Biochem Biophys Res Commun. 405:216–221. 2011. View Article : Google Scholar : PubMed/NCBI

13 

Shathasivam T, Kislinger T and Gramolini AO: Genes, proteins and complexes: The multifaceted nature of FHL family proteins in diverse tissues. J Cell Mol Med. 14:2702–2720. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Nishikawa M, Sato K, Nakano S, Yamakawa H, Nagase T and Ueda H: Specific activation of PLEKHG2-induced serum response element-dependent gene transcription by four-and-a-half LIM domains (FHL) 1, but not FHL2 or FHL3. Small GTPases. 10:361–366. 2017. View Article : Google Scholar : PubMed/NCBI

15 

Zabalegui F, Castañeda SL, Amin G, Belli C, Miriuka SG and Moro LN: Derivation of two human induced pluripotent stem cell lines carrying a missense mutation in FHL1 (c.377G > A, p.C126Y) linked to familial muscular dystrophy. Stem Cell Res. 75:1033072024. View Article : Google Scholar : PubMed/NCBI

16 

López Blázquez M, Fernández Ávila AI, Álvarez García-Rovés R, Centeno Jiménez M, Gómez González C and Espinosa Castro MÁ: Description of a novel variant in the FHL1 gene associated with hypertrophic cardiomyopathy with early and aggressive presentation. Rev Esp Cardiol (Engl Ed). 75:968–970. 2022.PubMed/NCBI

17 

Albrecht I, Wick C, Hallgren Å, Tjärnlund A, Nagaraju K, Andrade F, Thompson K, Coley W, Phadke A, Diaz-Gallo LM, et al: Development of autoantibodies against muscle-specific FHL1 in severe inflammatory myopathies. J Clin Invest. 125:4612–4624. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Martin DE, Soulard A and Hall MN: TOR regulates ribosomal protein gene expression via PKA and the forkhead transcription factor FHL1. Cell. 119:969–979. 2004. View Article : Google Scholar : PubMed/NCBI

19 

Ng WH, Liu X, Ling ZL, Santos CNO, Magalhães LS, Kueh AJ, Herold MJ, Taylor A, Freitas JR, Koit S, et al: FHL1 promotes chikungunya and o'nyong-nyong virus infection and pathogenesis with implications for alphavirus vaccine design. Nat Commun. 14:66052023. View Article : Google Scholar : PubMed/NCBI

20 

Meertens L, Hafirassou ML, Couderc T, Bonnet-Madin L, Kril V, Kümmerer BM, Labeau A, Brugier A, Simon-Loriere E, Burlaud-Gaillard J, et al: FHL1 is a major host factor for chikungunya virus infection. Nature. 574:259–263. 2019. View Article : Google Scholar : PubMed/NCBI

21 

Meertens L, Hafirassou ML, Couderc T, Bonnet-Madin L, Kril V, Kümmerer BM, Labeau A, Brugier A, Simon-Loriere E, Burlaud-Gaillard J, et al: FHL1 is a key player of chikungunya virus tropism and pathogenesis. C R Biol. 343:79–89. 2021. View Article : Google Scholar : PubMed/NCBI

22 

Mota IA, Correia CdC, Fontana PN and Carvalho AAS: Reducing body myopathy-A new pathogenic FHL1 variant and literature review. Neuromuscul Disord. 31:847–853. 2021. View Article : Google Scholar : PubMed/NCBI

23 

Binder MS, Brown E, Aversano T, Wagner KR, Calkins H and Barth AS: Novel FHL1 mutation associated with hypertrophic cardiomyopathy, sudden cardiac death, and myopathy. JACC Case Rep. 2:372–377. 2020. View Article : Google Scholar : PubMed/NCBI

24 

Sakashita K, Mimori K, Tanaka F, Kamohara Y, Inoue H, Sawada T, Hirakawa K and Mori M: Clinical significance of loss of Fhl1 expression in human gastric cancer. Ann Surg Oncol. 15:2293–2300. 2008. View Article : Google Scholar : PubMed/NCBI

25 

Pereira CM, de Carvalho AC, da Silva FR, Melendez ME, Lessa RC, Andrade VCC, Kowalski LP, Vettore AL and Carvalho AL: In vitro and in silico validation of CA3 and FHL1 downregulation in oral cancer. BMC Cancer. 18:1932018. View Article : Google Scholar : PubMed/NCBI

26 

Wang Z, Zhang J, Yang B, Li R, Jin L, Wang Z, Yu H, Liu C, Mao Y and You Q: Long intergenic noncoding RNA 00261 acts as a tumor suppressor in non-small cell lung cancer via regulating miR-105/FHL1 axis. J Cancer. 10:6414–6421. 2019. View Article : Google Scholar : PubMed/NCBI

27 

Zhou F, Qian C, Chen T, Zang X and Huang T: MiR-96-5p facilitates lung adenocarcinoma cell phenotypes by inhibiting FHL1. Comput Math Methods Med. 2022:78912222022. View Article : Google Scholar : PubMed/NCBI

28 

Wang F, Su Q and Li C: Identidication of novel biomarkers in Non-small cell lung cancer using machine learning. Sci Rep. 12:166932022. View Article : Google Scholar : PubMed/NCBI

29 

Chen H, Lai X, Zhu Y, Huang H, Zeng L and Zhang L: Quantitative proteomics identified circulating biomarkers in lung adenocarcinoma diagnosis. Clin Proteomics. 19:442022. View Article : Google Scholar : PubMed/NCBI

30 

Song J, Liang K, Wei T, Li L, Huang Z, Chen G, Mao N and Yang J: Expression and predictive significance of FHL1 and SLIT3 in surgically resected lung adenocarcinoma. Comb Chem High Throughput Screen. 26:2226–2237. 2023. View Article : Google Scholar : PubMed/NCBI

31 

Yang F, Lian M, Ma H, Feng L, Shen X, Chen J and Fang J: Identification of key genes associated with papillary thyroid microcarcinoma characteristics by integrating transcriptome sequencing and weighted gene co-expression network analysis. Gene. 811:1460862022. View Article : Google Scholar : PubMed/NCBI

32 

Li Y, Zeng Qa, Qiu J, Pang T, Ye F, Huang L and Zhang X: MiR-183-5p promotes proliferation, metastasis and angiogenesis in breast cancer cells through negatively regulating four and a Half LIM protein 1. J Breast Cancer. 23:355–372. 2020. View Article : Google Scholar : PubMed/NCBI

33 

Wang Y, Fu J, Jiang M, Zhang X, Cheng L, Xu X, Fan Z, Zhang J, Ye Q and Song H: MiR-410 is overexpressed in liver and colorectal tumors and enhances tumor cell growth by silencing FHL1 via a Direct/Indirect mechanism. PLoS One. 9:e1087082014. View Article : Google Scholar : PubMed/NCBI

34 

Ji C, Liu H, Xiang M, Liu J, Yue F, Wang W and Chu X: Deregulation of decorin and FHL1 are associated with esophageal squamous cell carcinoma progression and poor prognosis. Int J Clin Exp Med. 8:20965–20970. 2015.PubMed/NCBI

35 

Eshibona N, Giwa A, Rossouw SC, Gamieldien J, Christoffels A and Bendou H: Upregulation of FHL1, SPNS3, and MPZL2 predicts poor prognosis in pediatric acute myeloid leukemia patients with FLT3-ITD mutation. Leuk Lymphoma. 63:1897–1906. 2022. View Article : Google Scholar : PubMed/NCBI

36 

Zhang J, Hu X, Liu S, He Y, Lyu L and Jiang L: Clinical value screening, prognostic significance, and key gene identification of TrkB in laryngeal carcinoma. Dis Markers. 2022:13540052022.PubMed/NCBI

37 

Das K, Chan XB, Epstein D, Te Teh B, Kim KM, Kim ST, Park SH, Kang WK, Rozen S, Lee J and Tan P: NanoString expression profiling identifies candidate biomarkers of RAD001 response in metastatic gastric cancer. ESMO Open. 1:e0000092016. View Article : Google Scholar : PubMed/NCBI

38 

Liu1 P, Ma Q, Chen H, Zhang L and Zhang X: Identification of RHOBTB2 aberration as an independent prognostic indicator in acute myeloid leukemia. Aging (Albany NY). 13:15269–15284. 2021. View Article : Google Scholar : PubMed/NCBI

39 

Liu Y, Li H, Zhao Y, Li D, Zhang Q, Fu J and Fan S: Targeting FHL1 impairs cell proliferation and differentiation of acute myeloid leukemia cells. Biochem Cell Biol. 100:301–308. 2022. View Article : Google Scholar : PubMed/NCBI

40 

Luo Q, Pan Y, Fu Q, Zhang X, Zhou S, Yu P, Tian H, Liu P, Chen S, Zhang H and Qin T: Immortalization-upregulated protein promotes pancreatic cancer progression by regulating NPM1/FHL1-mediated cell-cycle-checkpoint protein activity. Cell Biol Toxicol. 39:2069–2087. 2022. View Article : Google Scholar : PubMed/NCBI

41 

Lin J, Ding L, Jin R, Zhang H, Cheng L, Qin X, Chai J and Ye Q: Four and a half LIM domains 1 (FHL1) and receptor interacting protein of 140 kDa (RIP140) interact and cooperate in estrogen signaling. Int J Biochem Cell Biol. 41:1613–1618. 2009. View Article : Google Scholar : PubMed/NCBI

42 

Li X, Jia Z, Shen Y, Ichikawa H, Jarvik J, Nagele RG and Goldberg GS: Coordinate suppression of Sdpr and Fhl1 expression in tumors of the breast, kidney, and prostate. Cancer Sci. 99:1326–1333. 2008. View Article : Google Scholar : PubMed/NCBI

43 

Koike K, Kasamatsu A, Iyoda M, Saito Y, Kouzu Y, Koike H, Sakamoto Y, Ogawara K, Tanzawa H and Uzawa K: High prevalence of epigenetic inactivation of the human four and a half LIM domains 1 gene in human oral cancer. Int J Oncol. 42:141–150. 2013. View Article : Google Scholar : PubMed/NCBI

44 

Asada K, Ando T, Niwa T, Nanjo S, Watanabe N, Okochi-Takada E, Yoshida T, Miyamoto K, Enomoto S, Ichinose M, et al: FHL1 on chromosome X is a single-hit gastrointestinal tumor-suppressor gene and contributes to the formation of an epigenetic field defect. Oncogene. 32:2140–2149. 2012. View Article : Google Scholar : PubMed/NCBI

45 

Chen X, Yu Y, Su Y, Shi L, Xie S, Hong Y, Liu X and Yin F: Low FHL1 expression indicates a good prognosis and drug sensitivity in ovarian cancer. Funct Integr Genomics. 24:252024. View Article : Google Scholar : PubMed/NCBI

46 

Sun L, Chen L, Zhu H, Li Y, Chen CC and Li M: FHL1 promotes glioblastoma aggressiveness through regulating EGFR expression. FEBS Lett. 595:85–98. 2020. View Article : Google Scholar : PubMed/NCBI

47 

Wang X, Wei X, Yuan Y, Sun Q, Zhan J, Zhang J, Tang Y, Li F, Ding L, Ye Q and Zhang H: Src-mediated phosphorylation converts FHL1 from tumor suppressor to tumor promoter. J Cell Biol. 217:1335–1351. 2018. View Article : Google Scholar : PubMed/NCBI

48 

Shi MK, Xuan YL and He XF: FHL1 overexpression as a inhibitor of lung cancer cell invasion via increasing RhoGDIß mRNA expression. Cell J. 24:239–244. 2022.PubMed/NCBI

49 

Matsumoto M, Kawakami K, Enokida H, Toki K, Matsuda R, Chiyomaru T, Nishiyama K, Kawahara K, Seki N and Nakagawa M: CpG hypermethylation of human four-and-a-half LIM domains 1 contributes to migration and invasion activity of human bladder cancer. Int J Mol Med. 26:241–247. 2010.PubMed/NCBI

50 

Yi S, Luo M, Peng Y, Chen Y and Yu D: Anti-oncogenic mechanism of KLF17 in colon cancer by repressing cell migration and invasion via FHL1 upregulation. Chi J Physiol. 66:534–545. 2023. View Article : Google Scholar : PubMed/NCBI

51 

Chen Y, Pei M, Li J, Wang Z, Liu S, Xiang L, Zhang J, Hong L, Lin J, Dai W, et al: Disruption of the CCDC43-FHL1 interaction triggers apoptosis in gastric cancer cells. Exp Cell Res. 415:1131072022. View Article : Google Scholar : PubMed/NCBI

52 

Zhou Z, Lu J, Dou J, Lv Z, Qin XI and Lin J: FHL1 and Smad4 synergistically inhibit vascular endothelial growth factor expression. Mol Med Rep. 7:649–653. 2013. View Article : Google Scholar : PubMed/NCBI

53 

Niu C, Liang C, Guo J, Cheng L, Zhang H, Qin X, Zhang Q, Ding L, Yuan B, Xu X, et al: Downregulation and growth inhibitory role of FHL1 in lung cancer. Int J Cancer. 130:2549–2556. 2011. View Article : Google Scholar : PubMed/NCBI

54 

Ding L, Niu C, Zheng Y, Xiong Z, Liu Y, Lin J, Sun H, Huang K, Yang W, Li X and Ye Q: FHL1 interacts with oestrogen receptors and regulates breast cancer cell growth. J Cell Mol Med. 15:72–85. 2011. View Article : Google Scholar : PubMed/NCBI

55 

Zhang Q, Li J, Chen Z, Jiang K, Yang K, Huang F, Huang A, Zhang X, Zhang J and Wang H: VE-822 upregulates the deubiquitinase OTUD1 to stabilize FHL1 to inhibit the progression of lung adenocarcinoma. Cell Oncol. 46:1001–1014. 2023. View Article : Google Scholar : PubMed/NCBI

56 

Lin J, Qin X, Zhu Z, Mu J, Zhu L, Wu K, Jiao H, Xu X and Ye Q: FHL family members suppress vascular endothelial growth factor expression through blockade of dimerization of HIF1α and HIF1β. IUBMB Life. 64:921–930. 2012. View Article : Google Scholar : PubMed/NCBI

57 

Zhang J, Li H, Guo M, Zhang J, Zhang G, Sun N, Feng Y, Cui W and Xu F: FHL1 as a novel prognostic biomarker and correlation with immune infiltration levels in lung adenocarcinoma. Immunotherapy. 15:235–252. 2023. View Article : Google Scholar : PubMed/NCBI

58 

Ding L, Wang Z, Yan J, Yang X, Liu A, Qiu W, Zhu J, Han J, Zhang H, Lin J, et al: Human four-and-a-half LIM family members suppress tumor cell growth through a TGF-β-like signaling pathway. J Clin Invest. 119:349–361. 2009.PubMed/NCBI

59 

Chen J, Zeng C, Jin J, Zhang P, Zhang Y, Zhang H, Li Y and Guan H: Overexpression of FHL1 suppresses papillary thyroid cancer proliferation and progression via inhibiting Wnt/β-catenin pathway. Endocrine. 85:238–249. 2024. View Article : Google Scholar : PubMed/NCBI

60 

Xu X, Fan Z, Liang C, Li L, Wang L, Liang Y, Wu J, Chang S, Yan Z, Lv Z, et al: A signature motif in LIM proteins mediates binding to checkpoint proteins and increases tumour radiosensitivity. Nat Commun. 8:140592017. View Article : Google Scholar : PubMed/NCBI

61 

Fu Y, Xu M, Cui Z, Yang Z, Zhang Z, Yin X, Huang X, Zhou M, Wang X and Chen C: Genome-wide identification of FHL1 as a powerful prognostic candidate and potential therapeutic target in acute myeloid leukaemia. EBioMedicine. 52:1026642020. View Article : Google Scholar : PubMed/NCBI

62 

Chen X, Yan Y, Liu Y, Yi Q and Xu Z: Tabersonine enhances olaparib sensitivity through FHL1-Mediated Epithelial-mesenchymal transition in an ovarian tumor. J Nat Prod. 87:837–848. 2024. View Article : Google Scholar : PubMed/NCBI

63 

Liu M, Meng J, Chen X, Wang F and Han Z: Long non-coding RNA Small Nucleolar RNA Host Gene 4 ameliorates cigarette Smoke-induced proliferation, apoptosis, inflammation, and airway remodeling in alveolar epithelial cells through the modulation of the mitogen-activated protein kinase signaling pathway via the microRNA-409-3p/Four and a Half LIM Domains 1 axis. Eur J Med Res. 29:3092024. View Article : Google Scholar : PubMed/NCBI

64 

Ding J, Cong YF and Liu B: Aberrant protein turn-over associated with myofibrillar disorganization in FHL1 knockout mice. Front Genet. 9:2732018. View Article : Google Scholar : PubMed/NCBI

65 

Wong CH, Fung YW, Ng EK, Lee SM, Waye MM and Tsui SK: LIM domain protein FHL1B interacts with PP2A catalytic β subunit-A novel cell cycle regulatory pathway. FEBS Lett. 584:4511–4516. 2010. View Article : Google Scholar : PubMed/NCBI

66 

Wu J, Zhao K, Du Z, Chen Y, Zhang F, Jiang W, Zheng J, Wu X, Shen C and Xiao X: Systemic effect of FHL1 on neuromuscular junction and myotube formation via insulin-like growth factor and myostatin signaling pathways. Biochem Biophys Res Commun. 537:125–131. 2021. View Article : Google Scholar : PubMed/NCBI

67 

Schreckenbach T, Henn W, Kress W, Roos A, Maschke M, Feiden W, Dillmann U, Schulz JB, Weis J and Claeys KG: Novel FHL1 mutation in a family with reducing body myopathy. Muscle Nerve. 47:127–134. 2012. View Article : Google Scholar : PubMed/NCBI

68 

Peters S: Electrocardiographic analysis in unclassifiable arrhythmic cardiomyopathy associated with Emery-Dreifuss caused by a mutation in FHL1. Int J Cardiol. 214:1362016. View Article : Google Scholar : PubMed/NCBI

69 

Luo YB, Peng Y, Lu Y, Li Q, Duan H, Bi F and Yang H: Expanding the Clinico-genetic spectrum of myofibrillar myopathy: Experience from a Chinese neuromuscular center. Front Neurol. 11:10142020. View Article : Google Scholar : PubMed/NCBI

70 

Hu Z, Zhu Y, Liu X, Zhang W, Liu J, Wu S, Xiao J, Yuan Y and Wang Z: FHL1-related clinical, muscle MRI and genetic features in six Chinese patients with reducing body myopathy. J Hum Genet. 64:919–926. 2019. View Article : Google Scholar : PubMed/NCBI

71 

Sepp R, Hategan L, Csányi B, Borbás J, Tringer A, Pálinkás ED, Nagy V, Takács H, Latinovics D, Nyolczas N, et al: The genetic architecture of hypertrophic cardiomyopathy in hungary: Analysis of 242 patients with a panel of 98 genes. Diagnostics (Basel). 12:11322022. View Article : Google Scholar : PubMed/NCBI

72 

Lin Y, Ban R, Qiao L, Chen J, Liu M, Liu J and Shi Q: Identification of novel FHL1 mutations associated with X-linked scapuloperoneal myopathy in unrelated Chinese patients. J Hum Genet. 68:477–484. 2023. View Article : Google Scholar : PubMed/NCBI

73 

Liu E, Jelinek J, Pastore YD, Guan Y, Prchal JF and Prchal JT: Discrimination of polycythemias and thrombocytoses by novel, simple, accurate clonality assays and comparison with PRV-1 expression and BFU-E response to erythropoietin. Blood. 101:3294–3301. 2003. View Article : Google Scholar : PubMed/NCBI

74 

Zhang F, Feng F, Yang P, Li Z, You J, Xie W, Gao X and Yang J: Four-and-a-half-LIM protein 1 down-regulates estrogen receptor α activity through repression of AKT phosphorylation in human breast cancer cell. Int J Biochem Cell Biol. 44:320–326. 2012. View Article : Google Scholar : PubMed/NCBI

75 

Zeng Y, Zeng D, Qi X, Wang H, Wang X, Dai X and Qu L: FHL1: A novel diagnostic marker for papillary thyroid carcinoma. Pathol Int. 74:520–529. 2024. View Article : Google Scholar : PubMed/NCBI

76 

Balraj A, Muthamilselvan S, Raja R and Palaniappan A: PRADclass: Hybrid Gleason grade-informed computational strategy identifies consensus biomarker features predictive of aggressive prostate adenocarcinoma. Technol Cancer Res Treat. 23:153303382312223892024. View Article : Google Scholar : PubMed/NCBI

77 

Cai Y, Xia L, Zhu H, Cheng H, Tian Y, Sun L, Wang J, Lu N, Wang J and Chen Y: MiR-3682-3p promotes esophageal cancer progression by targeting FHL1 and activating the Wnt/β-catenin signaling pathway. Cell Signal. 119:1111552024. View Article : Google Scholar : PubMed/NCBI

78 

Zhao JL, Liang SQ, Fu W, Zhu BK, Li SZ, Han H and Qin HY: The LIM domain protein FHL1C interacts with tight junction protein ZO-1 contributing to the epithelial-mesenchymal transition (EMT) of a breast adenocarcinoma cell line. Gene. 542:182–189. 2014. View Article : Google Scholar : PubMed/NCBI

79 

Dai J, Yu X, Han Y, Chai L, Liao Y, Zhong P, Xie R, Sun X, Huang Q and Wang J: TMT-labeling proteomics of papillary thyroid carcinoma reveal invasive biomarkers. J Cancer. 11:6122–6132. 2020. View Article : Google Scholar : PubMed/NCBI

80 

Fu W, Wang K, Zhao JL, Yu HC, Li SZ, Lin Y, Liang L, Huang SY, Liang YM, Han H and Qin HY: FHL1C induces apoptosis in notch1-dependent T-ALL cells through an interaction with RBP-J. BMC Cancer. 14:4632014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2025
Volume 29 Issue 1

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Tao Y, Wu Y, Shen R, He S and Miao X: Role of four and a half LIM domain protein 1 in tumors (Review). Oncol Lett 29: 37, 2025.
APA
Tao, Y., Wu, Y., Shen, R., He, S., & Miao, X. (2025). Role of four and a half LIM domain protein 1 in tumors (Review). Oncology Letters, 29, 37. https://doi.org/10.3892/ol.2024.14783
MLA
Tao, Y., Wu, Y., Shen, R., He, S., Miao, X."Role of four and a half LIM domain protein 1 in tumors (Review)". Oncology Letters 29.1 (2025): 37.
Chicago
Tao, Y., Wu, Y., Shen, R., He, S., Miao, X."Role of four and a half LIM domain protein 1 in tumors (Review)". Oncology Letters 29, no. 1 (2025): 37. https://doi.org/10.3892/ol.2024.14783