Open Access

Role of LMO7 in cancer (Review)

  • Authors:
    • Qun Zeng
    • Tingting Jiang
    • Jing Wang
  • View Affiliations

  • Published online on: July 11, 2024     https://doi.org/10.3892/or.2024.8776
  • Article Number: 117
  • Copyright: © Zeng 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

Cancer constitutes a multifaceted ailment characterized by the dysregulation of numerous genes and pathways. Among these, LIM domain only 7 (LMO7) has emerged as a significant player in various cancer types, garnering substantial attention for its involvement in tumorigenesis and cancer progression. This review endeavors to furnish a comprehensive discourse on the functional intricacies and mechanisms of LMO7 in cancer, with a particular emphasis on its potential as both a therapeutic target and prognostic indicator. It delves into the molecular attributes of LMO7, its implications in cancer etiology and the underlying mechanisms propelling its oncogenic properties. Furthermore, it underscores the extant challenges and forthcoming prospects in targeting LMO7 for combating cancer.

Introduction

Cancer is a complex group of diseases characterized by the uncontrolled growth and spread of abnormal cells in the body. With the capacity to afflict any anatomical site and propagate to adjacent tissues or distant organs, cancer poses a formidable global health challenge (1,2). The World Health Organization estimates that cancer accounted for nearly 10 million deaths worldwide in 2020. Notably, the incidence of several cancer types continues to increase, imposing an onerous burden on healthcare systems and affected individuals (3). Research efforts have been instrumental in advancing our understanding of cancer biology, thereby facilitating the development of targeted therapeutic modalities and innovative treatment paradigms. Breakthroughs in precision medicine, immunotherapy and genetic manipulation have shown promising results in improving patient outcomes and prolonging survival rates.

Early detection and screening programs play a crucial role in cancer management by enabling the timely identification of cancerous cells through regular screenings, such as mammograms, colonoscopies and pap smears, ultimately leading to better treatment outcomes (3).

Given the increasing prevalence of cancer and its impact on individuals, families and societies, the importance of current cancer research and treatment cannot be overstated. Continued investments in scientific advancements and access to quality care are essential to drive further progress and improve the lives of cancer patients worldwide. Overall, the importance of cancer lies in its severe impact on physical, emotional and socioeconomic aspects of individuals and society, emphasizing the need for sustained research, prevention efforts and improved access to quality care (4).

Oncogenes, tumor suppressor gene and DNA repair genes play pivotal roles in tumorigenesis, which is the process wherein normal cells undergo malignant transformation (57). Oncogenes, when activated or mutated, harbor the potential to instigate cancer by fostering uncontrolled cellular proliferation and aberrant signaling cascades (5). Conversely, tumor suppressor genes act as guardians of cellular homeostasis, curtailing aberrant growth and facilitating DNA repair mechanisms (6). DNA repair genes, as the name suggests, are responsible for repairing DNA damage that occurs naturally or due to external factors such as exposure to radiation or harmful chemicals. The intricate interplay among these genes and their functions is contingent upon the specific cancer type and implicated genetic aberrations (7). Understanding these mechanisms is indispensable for identifying potential therapeutic targets and devising efficacious cancer interventions (7,8).

The present review centers on elucidating the pivotal role of LIM domain only 7 (LMO7) in tumorigenesis, encompassing its molecular attributes, oncogenic functions, clinical relevance and therapeutic implications. Of note, LMO7, a multifunctional protein encoded by the LMO7 gene, has garnered attention for its involvement in diverse physiological processes, including neuronal development, cardiovascular health (9,10), and notably, cancer pathogenesis (11,12). Dysregulation of LMO7 has been implicated in various malignancies, underscoring its potential as a prognostic marker and therapeutic target. This review delineates the intricate molecular characteristics and biological functions of LMO7, elucidates its implications in cancer development across different organ systems, and elaborates on the mechanistic underpinnings of its oncogenic effects. Furthermore, it delineates the challenges inherent in targeting LMO7 for therapeutic intervention and suggests avenues for future research aimed at unraveling the complexities of LMO7-mediated carcinogenesis.

Molecular characteristics and biological functions of LMO7

The LMO7 gene encodes a protein called LMO7, which is found in humans and is located on chromosome 13q14.11. The LMO7 protein is predominantly expressed in cardiac and skeletal muscle tissues (13). LMO7 protein encompasses multiple LIM domains, serving as protein-protein interaction motifs implicated in cell growth, differentiation and cytoskeletal organization (1416). In addition, LMO7 contains PDZ domains, facilitating interaction modules that typically bind to specific protein sequences, and which are involved in protein localization, signal transduction and the assembly of protein complexes within cells (17,18). LMO7 protein also contains CH domains, an actin-binding motif, which may exist as a single copy or in tandem repeats, either functioning autonomously or serving a regulatory role. CH domains are found in cytoskeletal and signal transduction proteins, including actin-binding proteins like spectrin, α-actinin, dystrophin, utrophin and fimbrin, as well as proteins essential for the regulation of cell shape (cortexillins) and signaling proteins (Vav), and are crucial for regulating cell shape and signaling (19,20). It has one CH domain at its N-terminus, a PDZ domain in the middle region and two DUF4757 domains of unknown function (Fig. 1A). The human LMO7 protein shares a high degree of homology with LMO7 proteins in other species, particularly in the CH, PDZ and LIM domains (Fig. 1B).

It has been observed that LMO7 gathers at cell-cell adhesion sites subsequent to the assembly of nectin-afadin and E-cadherin-catenin complexes, thereby influencing cell migration, intercellular communication and tissue organization (21). In Drosophila, the vertebrate homolog of LMO7, Smash, interacts with Bazooka/Par-3, canoe/afadin, and the tyrosine kinase Src42A, localizing to the zonula adherens in a plane-polarized manner. Deletion of smash leads to severe defects in the morphogenesis of embryonic epithelial tissues and organs (22). In muscle cells, LMO7 functions as a transcription factor, regulating the expression of various muscle genes, including paired box gene 3 (Pax3), Pax7, myogenic differentiation antigen (MyoD) and myogenic factor 5, thereby controlling myogenesis (1113). LMO7-null mice exhibit growth retardation, reduced fiber size, and impaired skeletal muscle and cardiac function (11). Knockdown of chicken LMO7 diminishes the number and width of myotubes and the number of MyoD-positive myoblasts, a phenotype rescued by Wnt/β-catenin activation, suggesting a role for LMO7 in the initial events of chick skeletal muscle formation, particularly in myoblast survival (12).

As a critical adapter and scaffolding protein, LMO7 interacts directly or indirectly with ~20 proteins through its three domains, thereby having vital biological functions (Fig. 1C). Using the Search Tool for the Retrieval of Interacting Genes and Proteins (STRING) database (string-db.org) to predict protein interactions of LMO7 revealed its association with various proteins, such as enamel matrix derivatives, Afadin (AFDN), actinin α 1/4 and COMM domain-containing protein 6/9, among others, to exert its functions in different tumors (Table I). With these predicted interactions, future investigations could explore the specific proteins with which LMO7 interacts to regulate tumorigenesis in various tumor tissues.

Table I.

Proteins that interact with LIM domain only 7.

Table I.

Proteins that interact with LIM domain only 7.

ProteinFull nameFunctions(Refs.)
EMDEnamel matrix derivativesStabilizes and promotes the formation of a nuclear actin cortical network(70,71)
AFDNAfadinPlays a role in the organization of homotypic, interneuronal and heterotypic cell-cell adherens junctions(7274)
ACTN1Actinin α 1Anchors actin to a variety of intracellular structures(75,76)
ACTN4Actinin α 4Involved in vesicular trafficking via its association with the CART complex(77,78)
COMMD6COMM domain-containing protein 6Modulates activity of cullin-RING E3 ubiquitin ligase complexes. Inhibits TNF-induced NF-κB1 activation(79,80)
COMMD9COMM domain-containing protein 9Modulates Na(+) transport; downregulates activation of NF-κB(81,82)
Nectin-1Nectin cell adhesion molecule 1Promotes cell-cell contacts by forming homophilic or heterophilic trans-dimers(83,84)
Nectin-2Nectin cell adhesion molecule 2Modulator of T-cell signaling(85,86)
EDNRBEndothelin receptor type BMediates its action by association with G proteins that activate a phosphatidylinositol-calcium second messenger system(87,88)
UCHL1Ubiquitin carboxyl-terminal hydrolase isozyme L1Has ATP-independent ubiquitin ligase activity(89,90)
UCHL3Ubiquitin carboxyl-terminal hydrolase isozyme L3Controls levels of cellular ubiquitin through processing of ubiquitin precursors and ubiquitinated protein(91,92)
SKP1S-phase kinase associated protein 1Mediates the ubiquitination of proteins involved in cell cycle progression, signal transduction and transcription(93,94)
SKP2S-phase kinase associated protein 2Specifically recognizes phosphorylated CDKN1B/p27kip and is involved in the regulation of G1/S transition(95,96)
RBX1Ring box protein 1Mediates ubiquitination of target proteins(97,98)
VCLVinculinInvolved in cell-matrix adhesion and cell-cell adhesion(99,100)
NFKBIANuclear factor of κ light polypeptide gene enhancer in B-cells inhibitor, αInhibits the activity of dimeric NF-κB/REL complexes(101,102)
FBXO4F-box protein 4Promotes ubiquitination of cyclin D1 and its subsequent proteasomal degradation(103,104)
FBXW11F-box and WD-40 domain protein 11Mediates the ubiquitination of phosphorylated β-catenin and participates in Wnt signaling(105,106)
FBXL15F-box/LRR-repeat protein 15Acts as a positive regulator of the bone morphogenetic protein signaling pathway(107,108)
CDK2Cyclin-dependent kinase 2Control of the cell cycle; essential for meiosis(109,110)

LMO7 gene in cancer development

LMO7 is a protein that has a role in cell adhesion, cytoskeletal organization and cellular signaling pathways (2325). While extensive research is still needed to fully understand the precise role of LMO7 in cancer, LMO7 has been implicated as an oncogene in several types of cancer (11,12).

In breast cancer, elevated LMO7 levels correlate with aggressive phenotypes characterized by enhanced cell migration and invasiveness and are positively regulated by CUGBP Elav-like family member 1 (26,27). Higher levels of LMO7 expression have been associated with more aggressive breast cancer phenotypes, including increased cell migration and invasion (26,28). Serum response factor (SRF) regulates specific functions such as muscle development and breast cancer metastasis. LMO7 orchestrates the myocardin-related transcription factors (MRTFs) as coactivators promoting cytoskeletal rearrangements conducive to cancer cell motility (26,29). LMO7 is a specific regulator of MRTFs and plays a vital role in breast cancer cell migration (26,30). LMO7 activates MRTFs by relieving actin-mediated inhibition in cooperation with Rho GTPase. Disruption of actin-RPEL interactions eliminates Rho dependency, allowing strong Rho-independent activation of LMO7 (30,31). LMO7 reduces the G-actin/F-actin ratio by colocalizing with F-actin. Knockdown of LMO7 compromises MRTF activities and impairs breast cancer cell migration (26,31). LMO7 is upregulated in invasive breast carcinoma and correlates with increased expression of SRF target genes regulating muscle and actin cytoskeleton functions (23,32), suggesting a cell-specific mechanism regulating Rho-MRTF-SRF signaling and breast cancer cell migration (26).

A novel fusion gene, LMO7-BRAF, was identified in thyroid cancer (33). Enhanced expression of the LMO7-BRAF fusion protein stimulates endogenous ERK1/2 phosphorylation and promotes anchorage-independent cell growth. The LMO7-BRAF fusion is a recurrent somatic alteration that occurred in 2% of thyroid cancer and presented as an oncogenic alteration (33).

Transcription factor krüppel-like factor 4 binds to methylated CpGs at the enhancer regions of LMO7 and activates LMO7 expression via 3D chromatin loop formation with its promoter regions, influencing cellular functions in human glioblastoma cells (34).

LMO7 has been suggested to act as a tumor suppressor for murine lung adenocarcinoma. LMO7-deficient mice develop irregular and prominent epithelial lesions in terminal and respiratory bronchioles at a young age, whereas these mice tend to develop lung adenocarcinoma at an old age (28,35). Leucine-rich repeats and immunoglobulin-like domains proteins 3 (LRIG3) interacts with LMO7 and LIM and calponin homology domains 1 (LIMCH1), with co-localization and co-immunoprecipitation observed between LRIG1/3 and LMO7/LIMCH1 (36). LMO7 and LIMCH1 are highly expressed in normal lung tissue but reduced in malignant tissue. LMO7 immunoreactivity predicts poor prognosis in LRIG1-positive tumors (36). MicroRNA (miR)-96, as a serum biomarker for lung cancer, inhibits the expression of LMO7 by binding to its 3′-UTR, ultimately regulating lung carcinogenesis through the miR-96-LMO7 axis (37).

LMO7 has been found to be overexpressed in pancreatic cancer (PC), with recent studies identifying LMO7 as a potential prognostic marker for PC (38,39). Its overexpression is associated with tumor progression and poor patient survival. Studies have shown that knockdown or knockout of LMO7 in mouse PC cells leads to PC cell cycle arrest and apoptosis, significantly inhibiting PC cell proliferation, anchorage-free colony formation, migration in vitro, and slowing down the growth and metastasis of pancreatic carcinoma in vivo (28,40).

In inflammatory hepatocellular adenomas (IHCA), among the five IHCA cases with fyn related Src family tyrosine kinase (FRK) gene rearrangements, LMO7 was identified to be fused to the exon 3–8 region of FRK (39). In tumor cells, human genes use alternative transcription start sites (TSS) to control mRNA levels and expand transcriptional output, thereby promoting carcinogenesis (41,42). A study analyzing 108 colorectal cancer samples using exome arrays identified multiple genes, including LMO7, relative to normal mucosa, showing tumor-specific alternative TSS use in both adenoma and cancer samples (28,43).

LMO7 is upregulated in human malignancies, including colorectal cancer, with no transcriptional upregulation of the LMO7 observed in adenomas compared with normal mucosa (28,44,45). However, upregulation of LMO7 transcription was observed in cancer. LMO7 expression in primary tumors with p53 mutation was significantly higher than that in tumors without p53 mutation (45,46).

While LMO7 is not commonly discussed as a well-established oncogene, emerging evidence suggests its potential oncogenic role in certain cancers; LMO7 was upregulated in most tumors, but downregulated in certain other tumors (Fig. 2) (28); data were from GEPIA 2 (http://gepia2.cancer-pku.cn/). It is important to note that more research is needed to fully understand the molecular mechanisms by which LMO7 functions as an oncogene in cancer. This will help determine its potential as a therapeutic target and develop strategies to inhibit its activity for the treatment of specific cancers.

Figure 2.

Gene expression profile across all T and N samples. Each dot represents the expression in a sample. The red font indicates that the TPM of LMO7 is significantly upregulated in this tumor type, the green font indicates that the TPM of LMO7 is significantly downregulated in this tumor type and the black font indicates that there is no significant difference between T and N samples. The data were obtained from GEPIA 2. LMO7, LIM domain only 7; T, tumor; N, paired normal tissues; ACC, adrenocortical carcinoma; BLCA, bladder urothelial carcinoma; BRCA, breast invasive carcinoma; CESC, cervical squamous cell carcinoma and endocervical adenocarcinoma; CHOL, cholangiocarcinoma; COAD, colon adenocarcinoma; DLBC, lymphoid neoplasm diffuse large B-cell lymphoma; ESCA, esophageal cancer; GBM, glioblastoma multiforme; HNSC, head and neck squamous cell carcinoma; KICH, kidney chromophobe; KIRC, kidney renal clear cell carcinoma; KIRP, kidney renal papillary cell carcinoma; LAML, acute myeloid leukemia; LGG, brain lower grade glioma; LIHC, liver hepatocellular carcinoma; LUAD, lung adenocarcinoma; LUSC, lung squamous cell carcinoma; MESO, mesothelioma; OV, ovarian serous cystadenocarcinoma; PAAD, pancreatic adenocarcinoma; PCPG, pheochromocytoma and paraganglioma; PRAD, prostate adenocarcinoma; READ, rectum adenocarcinoma; SARC, sarcoma; SKCM, skin cutaneous melanoma; STAD, stomach adenocarcinoma; TGCT, testicular germ cell tumors; THCA, thyroid carcinoma; THYM, thymoma; UCEC, uterine corpus endometrial carcinoma; UCS, uterine carcinosarcoma; UVM, uveal melanoma.

Mechanisms underlying the oncogenic effects of LMO7

LMO7 has been found to play a role in promoting cell proliferation (37). Studies have elucidated LMO7′s role in promoting cell proliferation by fostering dysregulated cell cycle progression and mitigating apoptotic signaling pathways (11,2631). Furthermore, LMO7 facilitates cancer cell migration and invasion by modulating the dynamics of actin cytoskeleton and focal adhesion complexes, thereby augmenting cellular motility and tissue invasiveness (3439) (Table I). LMO7 may affect the dynamics of actin filaments and focal adhesions, thereby enhancing cell motility and invasion into surrounding tissues (3439).

LMO7 has been implicated in promoting epithelial-mesenchymal transition (EMT) (47), potentially regulating EMT-related genes and signaling pathways, leading to the loss of cell-cell adhesion and the acquisition of invasive properties by cancer cells (Fig. 3). LMO7 may modulate the activity of these signaling pathways, consequently promoting oncogenic effects such as enhanced cell survival, proliferation and metastasis (37). Specifically, LMO7 has been shown to promote EMT by regulating the activity of transcription factors such as snail, slug and ZEB1, which have pivotal roles in EMT (Fig. 3) (48). Through activation of these transcription factors, LMO7 facilitates the reprogramming of target genes involved in the EMT process, leading to the loss of epithelial characteristics and the acquisition of mesenchymal traits (47,48), facilitating cell invasion and migration, crucial for processes such as tissue repair and wound healing.

LMO7 interacts with AKT, a downstream effector of PI3K, potentially contributing to the activation of AKT signaling (49). This interaction suggests that LMO7 may promote cell survival and growth by enhancing the activity of the PI3K/AKT/mTOR pathway (49). Furthermore, LMO7 may modulate Rho-GTPase activity, impacting cellular processes involved in cancer progression, such as cell migration and invasion (Fig. 3) (50). In addition, LMO7 has been shown to bind to and modulate the activity of MEK1, a kinase upstream of ERK, potentially influencing cell proliferation and survival through the regulation of the ERK/MAPK pathway (Fig. 3) (49). Further research is necessary to fully understand the precise mechanisms and consequences of LMO7′s interactions with these pathways in cancer development and progression.

Epigenetic modifications play a critical role in regulating gene expression and can contribute to cancer development and progression (51). Evidence suggests that epigenetic alterations may influence LMO7 expression in different cancer types (29). For instance, in breast cancer, LMO7 has been found to undergo hypermethylation, resulting in reduced gene expression (47). Hypermethylation of the LMO7 promoter region has been associated with tumor progression and poor patient prognosis (52). Further investigation into LMO7-associated epigenetic alterations may offer insight into potential diagnostic, prognostic and therapeutic strategies for cancer patients.

Furthermore, LMO7 has been identified as a TGF-β1 target gene in hepatoma cells, functioning in vascular physiology and fibrosis (53). It is triggered by injury and TGF-β in vascular smooth muscle cells in vitro (53). Loss of LMO7 enhanced TGF-β signaling by upregulating TGF-β1 mRNA, TGF-β protein, integrin-αvβ3, latent TGF-β activation, downstream effectors Smad3 phosphorylation and connective tissue growth factor (53). Mechanistically, LMO7′s LIM domain interacts with activator protein 1 transcription factors c-Fos and c-Jun, promoting their degradation and interrupting activator protein 1-dependent TGF-β autoinduction (53). A study has shown that the increase in LMO7 mRNA expression synchronizes with TGF-β1-induced invasion, with higher LMO7 expression in high metastatic cells compared to low metastatic cells (36). Induced by TGF-β, LMO7 may limit vascular fibrosis by negative feedback regulation of the TGF-β pathway (Fig. 3), suggesting implications for intimal hyperplasia, wound healing and fibrotic diseases and potentially impacting tumor angiogenesis.

Researchers have found that the curcumin analogue CA-5f {(3E,5E)-3-(3,4-dimethoxybenzylidene)-5-[(1H-indol-3-yl)methylene]-1-methylpiperidin-4-one}, as an anticancer therapeutic agent (54), reduced LMO7 protein levels, which induced the accumulation of microtubule-associated protein 1 light chain 3β and sequestosome 1, increased mitochondrial reactive oxygen species levels and then inhibited autophagosome-lysosome fusion and induced cell death (55). It may thus be suggested that LMO7 may be the target of CA-5f, and related antisense RNA can be designed from LMO7 to interfere with the expression of LMO7 and inhibit the growth of tumor cells.

LMO7 also has a role in inflammation. In the mouse model of autoimmune hepatitis (AIH), injection of bone marrow mesenchymal stem cells (BMSCs) upregulated the levels of LMO7 and downregulated the levels of AP-1 and TGF-β, while the expression of AP-1 and TGF-β was upregulated in the LMO7 interference group. BMSCs can significantly reduce liver injury in the mouse AIH model by regulating the LMO7/AP-1/TGF-β signaling pathway to alleviate liver fibrosis of autoimmune hepatitis (56). The expression of the LMO7 gene was found to be upregulated in gastric epithelial AGS cells (a gastric cancer cell line) infected with Helicobacter pylori, which is involved in the adhesion, invasion and possibly proliferation of gastric epithelial cells (57). The expression of LMO7 in H. pylori may be involved in the carcinogenesis or differentiation of gastric epithelial cells through direct interactions with other proteins. The proteins that bind to LMO7 and the functional role of protein interactions in cell adhesion should be investigated in gastric epithelial cells stimulated by H. pylori. AFDN, vinculin and other proteins that were mentioned in Table I play a role in cell-cell adherens junctions (Table I).

Prognostic relevance of LMO7 in cancer

The clinical relevance of LMO7 in cancer extends beyond its prognostic value to encompass its potential as a therapeutic target for precision medicine approaches. Of note, elevated LMO7 expression correlates with adverse clinical outcomes and poor patient prognosis across various cancer types, thereby underscoring its utility as a prognostic biomarker (37,47). However, it is important to note that the prognostic relevance of LMO7 may vary across different cancer types and stages (Fig. 4) (28); data were from GEPIA 2.

Compared with human papillomavirus (HPV)-dependent disease, patients with HPV-independent vulvar squamous cell carcinoma (VSCC) have a survival deficit and data suggest that leucine rich repeats and immunoglobulin like domains 2 (LRIG2) and LMO7 are positive prognostic factors in HPV-independent cases; LMO7 is a positive prognostic factor in the most advanced tumors. Therefore, these markers may provide tools for individual treatment strategy selection in patients with VSCC. However, more studies are needed to further elucidate the functional and prognostic value of the molecular markers studied in VSCC (58). High expression of LMO7 antisense RNA 1 was associated with poor survival in kidney cancer (59).

LMO7, along with aminopeptidase like 1, von Willebrand factor, aldehyde dehydrogenase 2 family member, NUAK family kinase 1 and tumor protein, translationally-controlled 1, named the castration-resistant PC-derived prognosis signature, is considered an important molecular signature for predicting progression-free survival (PFS) of patients with castration-resistant PC for therapeutic decision-making (60).

In breast cancer, elevated LMO7 expression correlates with an unfavorable patient prognosis (46,47). Similarly, in colorectal cancer, LMO7 expression serves as a potential prognostic marker, with higher levels associated with adverse patient outcomes, including reduced overall survival and disease-free survival (46,47,50). In addition, increased LMO7 expression correlates with advanced tumor stages, lymph node metastasis and poor tumor differentiation in colorectal cancer cases (44,45). In lung cancer, multiple studies consistently link increased LMO7 expression with unfavorable patient outcomes, including diminished overall survival and disease-free survival rates (Fig. 4) (28,3436). However, contradictory evidence suggests that elevated LMO7 levels may paradoxically be associated with improved overall survival (Fig. 4) (28). The association between LMO7 expression and prognosis in other cancer types remains to be elucidated. Further research, considering additional factors, is essential to establish LMO7 as a reliable and independent prognostic biomarker in specific cancer contexts.

Genomic alteration types

LMO7 mutations have been identified within tumor tissues, implicating their role in cancer development and progression. These mutations encompass various types, broadly categorized as mutation, structural variant, amplification, deep deletion and multiple alterations (Fig. 5) (61,62); data were from cBioPortal for Cancer Genomics (http://www.cbioportal.org). An analysis in cBioPortal revealed the presence of two missense mutations (P928Q and S1259R) in LMO7 among cancer patients. In addition, LMO7 demonstrated high-level amplification in lung adenocarcinoma, leading to LMO7-ITGBL1 fusion and a Q1334* nonsense mutation (61,62). Deep deletion of LMO7 has been observed in prostate adenocarcinoma and lung squamous cell carcinoma genomes, resulting in TPTE2P5-LMO7 fusion and an I774V missense mutation, respectively (62). It is important to note that the specific consequences of LMO7 mutations in tumor tissues can vary based on the mutation and cancer context. Further research is imperative to comprehensively understand the impact of LMO7 mutations on tumorigenesis and potentially identify novel therapeutic targets.

Challenges and future perspectives

Although the expression pattern of LMO7 has been explored across various cancer types, its precise molecular functions in tumorigenesis and cancer progression remain largely elusive. Further research is imperative to identify the specific mechanisms through which LMO7 promotes cancer development and metastasis. Numerous studies investigating LMO7 and cancer have relied on relatively small sample sizes, potentially constraining the generalizability and statistical power of their findings (61,62). Robust larger-scale studies are necessary to corroborate the significance of LMO7 across different cancer types (6365). The heterogeneity observed within cancer samples, including variations in tumor stage, grade and molecular subtypes, complicates the interpretation of the role of LMO7. Investigating LMO7 expression and its correlation with patient outcomes across large, well-characterized cohorts is necessary to better understand its clinical relevance. Standardized methodologies for studying LMO7 in cancer research are lacking, with studies employing diverse experimental approaches like immunohistochemistry, gene expression profiling and functional assays, posing challenges for comparison and integration of findings across studies. The functional characterization of LMO7 in cancer has mainly relied on in vitro cell culture experiments and xenograft models (2830). The development of more sophisticated preclinical models is crucial for validating its role in cancer progression, as these models fail to fully replicate the complex tumor microenvironment and organismal interactions.

Targeting LMO7 for cancer treatment shows promise due to its involvement in various cellular processes contributing to tumorigenesis (2631,66,67). However, several challenges need to be addressed before LMO7-based therapies can be effectively developed. These include the complexity of LMO7 function, its context-dependent roles in different cancer types and the absence of specific inhibitors or modulators targeting LMO7. The diverse functions of LMO7 in cell adhesion, cytoskeletal organization, signal transduction and gene regulation make it difficult to selectively target its activity without interfering with important cellular processes (2325). Interfering with LMO7 function may lead to unintended consequences, undesirable side effects or the limitation of therapeutic efficacy. The role and expression levels of LMO7 may vary among different cancer types and even among subtypes of the same cancer, posing challenges for targeted therapies. Further studies are needed to understand the precise mechanism by which LMO7 is involved in the genesis of different cancer types and to identify reliable biomarkers for patient stratification. Currently, there is a lack of specific inhibitors or modulators selectively targeting LMO7. High-throughput screening and rational drug design methods can be utilized to identify small molecules or peptides interfering with LMO7 interactions or functional domains.

To overcome the challenges in understanding the role of LMO7 in various cancer types, continued research is essential. This includes conducting comprehensive studies to investigate the molecular mechanisms by which LMO7 promotes tumor progression and metastasis. Additional experiments using cellular and animal models can provide valuable insight into its specific contributions to cancer development.

In PC, prediction of PFS and castration resistance molecular characteristics is crucial for making treatment decisions, but current methods lack reliability. Researchers have applied the Robust Rank Aggregation method to analyze the transcriptome profile of PC to identify 287 differentially expressed genes, including LMO7 (68,69). However, this work has yet to be validated and clinically applied.

Further research is needed to elucidate the role of LMO7 as a diagnostic and prognostic biomarker. If LMO7 is confirmed as an oncogene, strategies for therapeutic targeting can be explored, such as developing small-molecule inhibitors specific to LMO7 or gene therapy approaches to suppress its expression. In-depth molecular characterization of LMO7 and its associated signaling pathways can help identify potential drug targets and combination therapies to overcome challenges such as drug resistance and metastasis. Analyzing LMO7 expression levels in patient samples through techniques such as immunohistochemistry can establish correlations between LMO7 expression and clinical outcomes, including patient survival and response to therapy. Longitudinal studies can also provide information about dynamic changes in LMO7 expression during disease progression and its potential as a predictive biomarker or therapeutic target. In addition, the development of reliable biomarkers associated with LMO7 may aid in monitoring treatment response and disease progression.

The present review delved into the intricate role of LMO7 in cancer development, elucidating the molecular mechanisms underlying its oncogenic effects. With its involvement across various cancer types and potential as a prognostic marker and therapeutic target, understanding the function of LMO7 offers valuable insight into cancer biology and paves the way for precision medicine. However, further research is imperative to overcome current limitations and fully harness the potential of targeting LMO7 in cancer therapy. By unraveling the mysteries surrounding LMO7, we can advance towards personalized and effective cancer treatments.

Acknowledgements

Not applicable.

Funding

This work was supported by the Natural Science Foundation of Hunan Province (grant no. 2024JJ5325) and the Excellent Youth Project of Hunan Provincial Education Department (grant no. 22B0411).

Availability of data and materials

The experimental data and the simulation results that support the findings of this review are available from the STRING database (string-db.org), GEPIA 2 (http://gepia2.cancer-pku.cn/) and cBioPortal (http://www.cbioportal.org).

Authors' contributions

QZ and TJ: Substantial contributions to conception and design of the work, acquisition, analysis and interpretation of data for the study; drafting the manuscript or reviewing it critically for important intellectual content. JW: Conception and design, revising the manuscript, final approval of the version to be published, agreement to be accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved. 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.

References

1 

de Visser KE and Joyce JA: The evolving tumor microenvironment: From cancer initiation to metastatic outgrowth. Cancer Cell. 41:374–403. 2023. View Article : Google Scholar : PubMed/NCBI

2 

Feinberg AP and Levchenko A: Epigenetics as a mediator of plasticity in cancer. Science. 379:eaaw38352023. View Article : Google Scholar : PubMed/NCBI

3 

Wang X, Chen G, Zhang Y, Ghareeb WM, Yu Q, Zhu H, Lu X, Huang Y, Huang S, Hou D and Chi P: The impact of circumferential tumour location on the clinical outcome of rectal cancer patients managed with neoadjuvant chemoradiotherapy followed by total mesorectal excision. Eur J Surg Oncol. 46:1118–1123. 2020. View Article : Google Scholar : PubMed/NCBI

4 

Diori Karidio I and Sanlier SH: Reviewing cancer's biology: An eclectic approach. J Egypt Natl Canc Inst. 33:322021. View Article : Google Scholar : PubMed/NCBI

5 

Zeng Q and Jiang T: The role of FHL1 in tumors. Gene. 11:1483472024. View Article : Google Scholar : PubMed/NCBI

6 

Dietlein F, Wang AB, Fagre C, Tang A, Besselink NJM, Cuppen E, Li C, Sunyaev SR, Neal JT and Van Allen EM: Genome-wide analysis of somatic noncoding mutation patterns in cancer. Science. 376:eabg56012022. View Article : Google Scholar : PubMed/NCBI

7 

Lou Z, Gong YQ, Zhou X and Hu GH: Low expression of miR-199 in hepatocellular carcinoma contributes to tumor cell hyper-proliferation by negatively suppressing XBP1. Oncol Lett. 16:6531–6539. 2018.PubMed/NCBI

8 

Li L, Wang S and Zhou W: Balance cell apoptosis and pyroptosis of caspase-3-activating chemotherapy for better antitumor therapy. Cancers (Basel). 15:262022. View Article : Google Scholar : PubMed/NCBI

9 

Du TT, Dewey JB, Wagner EL, Cui R, Heo J, Park JJ, Francis SP, Perez-Reyes E, Guillot SJ, Sherman NE, et al: LMO7 deficiency reveals the significance of the cuticular plate for hearing function. Nat Commun. 10:11172019. View Article : Google Scholar : PubMed/NCBI

10 

Huang W, Xu Q, Su J, Tang L, Hao ZZ, Xu C, Liu R, Shen Y, Sang X, Xu N, et al: Linking transcriptomes with morphological and functional phenotypes in mammalian retinal ganglion cells. Cell Rep. 40:1113222022. View Article : Google Scholar : PubMed/NCBI

11 

Mull A, Kim G and Holaska JM: LMO7-null mice exhibit phenotypes consistent with emery-dreifuss muscular dystrophy. Muscle Nerve. 51:222–228. 2015. View Article : Google Scholar : PubMed/NCBI

12 

Possidonio AC, Soares CP, Fontenele M, Morris ER, Mouly V, Costa ML and Mermelstein C: Knockdown of Lmo7 inhibits chick myogenesis. FEBS Lett. 590:317–329. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Gomes G, do Amaral MJ, Bagri KM, Vasconcellos LM, Almeida MDS, Alvares LE and Mermelstein C: New findings on LMO7 transcripts, proteins and regulatory regions in human and vertebrate model organisms and the intracellular distribution in skeletal muscle cells. Int J Mol Sci. 22:128852021. View Article : Google Scholar : PubMed/NCBI

14 

She M, Tang M, Jiang T and Zeng Q: The roles of the LIM domain proteins in Drosophila cardiac and hematopoietic morphogenesis. Front Cardiovasc Med. 8:6168512021. View Article : Google Scholar : PubMed/NCBI

15 

She M, Zhang J, Jiang T, Zhang Y, Liu Y, Tang M and Zeng Q: The function of Lmpt in Drosophila heart tissue. Biochem Biophys Res Commun. 612:15–21. 2022. View Article : Google Scholar : PubMed/NCBI

16 

Zhang J, She M, Dai Y, Nie X, Tang M and Zeng Q: Lmpt regulates the function of Drosophila muscle by acting as a repressor of Wnt signaling. Gene. 876:1475142023. View Article : Google Scholar : PubMed/NCBI

17 

Sánta A, Czajlik A, Batta G, Péterfia B and Gáspári Z: Resonance assignment of the Shank1 PDZ domain. Biomol NMR Assign. 16:121–127. 2022. View Article : Google Scholar : PubMed/NCBI

18 

Mieszczanek J, Strutt H, Rutherford TJ, Strutt D, Bienz M and Gammons MV: Selective function of the PDZ domain of Dishevelled in noncanonical Wnt signalling. J Cell Sci. 135:jcs2595472022. View Article : Google Scholar : PubMed/NCBI

19 

Palani S, Ghosh S, Ivorra-Molla E, Clarke S, Suchenko A, Balasubramanian MK and Köster DV: Calponin-homology domain mediated bending of membrane-associated actin filaments. Elife. 10:e610782021. View Article : Google Scholar : PubMed/NCBI

20 

Mei L, Reynolds MJ, Garbett D, Gong R, Meyer T and Alushin GM: Structural mechanism for bidirectional actin cross-linking by T-plastin. Proc Natl Acad Sci USA. 119:e22053701192022. View Article : Google Scholar : PubMed/NCBI

21 

Ecke M, Prassler J and Gerisch G: Expanding ring-shaped cleavage furrows in multinucleate cells. Mol Biol Cell. 34:ar272023. View Article : Google Scholar : PubMed/NCBI

22 

Cuadrado M and Robles-Valero J: VAV proteins as double agents in cancer: Oncogenes with tumor suppressor roles. Biology (Basel). 10:8882021.PubMed/NCBI

23 

Guérin A, Roy NH, Kugler EM, Berry L, Burkhardt JK, Shin JB and Striepen B: Cryptosporidium rhoptry effector protein ROP1 injected during invasion targets the host cytoskeletal modulator LMO7. Cell Host Microbe. 29:1407–1420.e5. 2021. View Article : Google Scholar : PubMed/NCBI

24 

Li M, An Z, Tang Q, Ma Y, Yan J, Chen S and Wang Y: Mixed responses to first-line alectinib in non-small cell lung cancer patients with rare ALK gene fusions: A case series and literature review. J Cell Mol Med. 25:9476–9481. 2021. View Article : Google Scholar : PubMed/NCBI

25 

Hariyanto NI, Yo EC and Wanandi SI: Regulation and signaling of TGF-β autoinduction. Int J Mol Cell Med. 10:234–247. 2021.PubMed/NCBI

26 

Perou CM, Sørlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA, Pollack JR, Ross DT, Johnsen H, Akslen LA, et al: Molecular portraits of human breast tumours. Nature. 406:747–752. 2000. View Article : Google Scholar : PubMed/NCBI

27 

Xia H, Chen D, Wu Q, Wu G, Zhou Y, Zhang Y and Zhang L: CELF1 preferentially binds to exon-intron boundary and regulates alternative splicing in HeLa cells. Biochim Biophys Acta Gene Regul Mech. 1860:911–921. 2017. View Article : Google Scholar : PubMed/NCBI

28 

Tang Z, Li C, Kang B, Gao G, Li C and Zhang Z: GEPIA: A web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res. 45((W1)): W98–W102. 2017. View Article : Google Scholar : PubMed/NCBI

29 

Miano JM, Long X and Fujiwara K: Serum response factor: Master regulator of the actin cytoskeleton and contractile apparatus. Am J Physiol Cell Physiol. 292:C70–C81. 2007. View Article : Google Scholar : PubMed/NCBI

30 

Medjkane S, Perez-Sanchez C, Gaggioli C, Sahai E and Treisman R: Myocardin-related transcription factors and SRF are required for cytoskeletal dynamics and experimental metastasis. Nat Cell Biol. 11:257–268. 2009. View Article : Google Scholar : PubMed/NCBI

31 

Pomiès P, Pashmforoush M, Vegezzi C, Chien KR, Auffray C and Beckerle MC: The cytoskeleton-associated PDZ-LIM protein, ALP, acts on serum response factor activity to regulate muscle differentiation. Mol Biol Cell. 18:1723–1733. 2007. View Article : Google Scholar : PubMed/NCBI

32 

Kim D, Jung SH and Chung YJ: Development of an RNA sequencing panel to detect gene fusions in thyroid cancer. Genomics Inform. 19:e412021. View Article : Google Scholar : PubMed/NCBI

33 

He H, Li W, Yan P, Bundschuh R, Killian JA, Labanowska J, Brock P, Shen R, Heerema NA and de la Chapelle A: Identification of a recurrent LMO7-BRAF fusion in papillary thyroid carcinoma. Thyroid. 28:748–754. 2018. View Article : Google Scholar : PubMed/NCBI

34 

Oyinlade O, Wei S, Kammers K, Liu S, Wang S, Ma D, Huang ZY, Qian J, Zhu H, Wan J and Xia S: Analysis of KLF4 regulated genes in cancer cells reveals a role of DNA methylation in promoter-enhancer interactions. Epigenetics. 13:751–768. 2018. View Article : Google Scholar : PubMed/NCBI

35 

Tanaka-Okamoto M, Hori K, Ishizaki H, Hosoi A, Itoh Y, Wei M, Wanibuchi H, Mizoguchi A, Nakamura H and Miyoshi J: Increased susceptibility to spontaneous lung cancer in mice lacking LIM-domain only 7. Cancer Sci. 100:608–616. 2009. View Article : Google Scholar : PubMed/NCBI

36 

Nakamura H, Hori K, Tanaka-Okamoto M, Higashiyama M, Itoh Y, Inoue M, Morinaka S and Miyoshi J: Decreased expression of LMO7 and its clinicopathological significance in human lung adenocarcinoma. Exp Ther Med. 2:1053–1057. 2011. View Article : Google Scholar : PubMed/NCBI

37 

Karlsson T, Kvarnbrink S, Holmlund C, Botling J, Micke P, Henriksson R, Johansson M and Hedman H: LMO7 and LIMCH1 interact with LRIG proteins in lung cancer, with prognostic implications for early-stage disease. Lung Cancer. 125:174–184. 2018. View Article : Google Scholar : PubMed/NCBI

38 

Wu H, Zhou J, Mei S, Wu D, Mu Z, Chen B, Xie Y, Ye Y and Liu J: Circulating exosomal microRNA-96 promotes cell proliferation, migration and drug resistance by targeting LMO7. J Cell Mol Med. 21:1228–1236. 2017. View Article : Google Scholar : PubMed/NCBI

39 

Ren B, Cui M, Yang G, Wang H, Feng M, You L and Zhao Y: Tumor microenvironment participates in metastasis of pancreatic cancer. Mol Cancer. 17:1082018. View Article : Google Scholar : PubMed/NCBI

40 

Bayard Q, Caruso S, Couchy G, Rebouissou S, Bioulac Sage P, Balabaud C, Paradis V, Sturm N, de Muret A, Guettier C, et al: Recurrent chromosomal rearrangements of ROS1, FRK and IL6 activating JAK/STAT pathway in inflammatory hepatocellular adenomas. Gut. 69:1667–1676. 2020. View Article : Google Scholar : PubMed/NCBI

41 

Liu X, Yuan H, Zhou J, Wang Q, Qi X, Bernal C, Avella D, Kaifi JT, Kimchi ET, Timothy P, et al: LMO7 as an unrecognized factor promoting pancreatic cancer progression and metastasis. Front Cell Dev Biol. 9:6473872021. View Article : Google Scholar : PubMed/NCBI

42 

Davuluri RV, Suzuki Y, Sugano S, Plass C and Huang TH: The functional consequences of alternative promoter use in mammalian genomes. Trends Genet. 24:167–177. 2008. View Article : Google Scholar : PubMed/NCBI

43 

Inchingolo MA, Diman A, Adamczewski M, Humphreys T, Jaquier-Gubler P and Curran JA: TP53BP1, a dual-coding gene, uses promoter switching and translational reinitiation to express a smORF protein. iScience. 26:1067572023. View Article : Google Scholar : PubMed/NCBI

44 

Thorsen K, Schepeler T, Øster B, Rasmussen MH, Vang S, Wang K, Hansen KQ, Lamy P, Pedersen JS, Eller A, et al: Tumor-specific usage of alternative transcription start sites in colorectal cancer identified by genome-wide exon array analysis. BMC Genomics. 12:5052011. View Article : Google Scholar : PubMed/NCBI

45 

Furuya M, Tsuji N, Endoh T, Moriai R, Kobayashi D, Yagihashi A and Watanabe N: A novel gene containing PDZ and LIM domains, PCD1, is overexpressed in human colorectal cancer. Anticancer Res. 22:4183–4186. 2002.PubMed/NCBI

46 

Kang S, Xu H, Duan X, Liu JJ, He Z, Yu F, Zhou S, Meng XQ, Cao M and Kennedy GC: PCD1, a novel gene containing PDZ and LIM domains, is overexpressed in several human cancers. Cancer Res. 60:5296–5302. 2000.PubMed/NCBI

47 

Jiang ZR, Yang LH, Jin LZ, Yi LM, Bing PP, Zhou J and Yang JS: Identification of novel cuproptosis-related lncRNA signatures to predict the prognosis and immune microenvironment of breast cancer patients. Front Oncol. 12:9886802022. View Article : Google Scholar : PubMed/NCBI

48 

Bao Z, Zeng W, Zhang D, Wang L, Deng X, Lai J, Li J, Gong J and Xiang G: SNAIL induces EMT and lung metastasis of tumours secreting CXCL2 to promote the invasion of M2-type immunosuppressed macrophages in colorectal cancer. Int J Biol Sci. 18:2867–2881. 2022. View Article : Google Scholar : PubMed/NCBI

49 

Chen J, Chen L, Hua J and Song W: Long-term dynamic compression enhancement TGF-β3-induced chondrogenesis in bovine stem cells: A gene expression analysis. BMC Genom Data. 22:132021. View Article : Google Scholar : PubMed/NCBI

50 

Hu Q, Guo C, Li Y, Aronow BJ and Zhang J: LMO7 mediates cell-specific activation of the Rho-myocardin-related transcription factor-serum response factor pathway and plays an important role in breast cancer cell migration. Mol Cell Biol. 31:3223–3240. 2011. View Article : Google Scholar : PubMed/NCBI

51 

Sun L, Zhang H and Gao P: Metabolic reprogramming and epigenetic modifications on the path to cancer. Protein Cell. 13:877–919. 2022. View Article : Google Scholar : PubMed/NCBI

52 

He B, Dai C, Lang J, Bing P, Tian G, Wang B and Yang J: A machine learning framework to trace tumor tissue-of-origin of 13 types of cancer based on DNA somatic mutation. Biochim Biophys Acta Mol Basis Dis. 1866:1659162020. View Article : Google Scholar : PubMed/NCBI

53 

Xie Y, Ostriker AC, Jin Y, Hu H, Sizer AJ, Peng G, Morris AH, Ryu C, Herzog EL, Kyriakides T, et al: LMO7 is a negative feedback regulator of transforming growth factor β signaling and fibrosis. Circulation. 139:679–693. 2019. View Article : Google Scholar : PubMed/NCBI

54 

Kim M and Moon A: A curcumin analog CA-5f inhibits urokinase-type plasminogen activator and invasive phenotype of triple-negative breast cancer cells. Toxicol Res. 38:19–26. 2021. View Article : Google Scholar : PubMed/NCBI

55 

Zhang L, Qiang P, Yu J, Miao Y, Chen Z, Qu J, Zhao Q, Chen Z, Liu Y, Yao X, et al: Identification of compound CA-5f as a novel late-stage autophagy inhibitor with potent anti-tumor effect against non-small cell lung cancer. Autophagy. 15:391–406. 2019. View Article : Google Scholar : PubMed/NCBI

56 

Chen ZK, Chen DZ, Cai C, Jin LL, Xu J, Tu YL, Huang XZ, Xu JL, Chen MZ, Xue FB, et al: BMSCs attenuate hepatic fibrosis in autoimmune hepatitis through regulation of LMO7-AP1-TGFβ signaling pathway. Eur Rev Med Pharmacol Sci. 25:1600–1611. 2021.PubMed/NCBI

57 

Lim JW, Kim H and Kim KH: Cell adhesion-related gene expression by Helicobacter pylori in gastric epithelial AGS cells. Int J Biochem Cell Biol. 35:1284–1296. 2003. View Article : Google Scholar : PubMed/NCBI

58 

Zhang Y, Liu Q, Cui M, Wang M, Hua S, Gao J and Liao Q: Comprehensive analysis of expression, prognostic value, and immune infiltration for ubiquitination-related FBXOs in pancreatic ductal adenocarcinoma. Front Immunol. 12:7744352022. View Article : Google Scholar : PubMed/NCBI

59 

Stefansson K, Oda H, Öfverman C, Lundin E, Hedman H and Lindquist D: LRIG1-2 and LMO7 immunoreactivity in vulvar squamous cell carcinoma: Association with prognosis in relation to HPV-DNA and p16INK4a status. Oncol Rep. 42:142–150. 2019.PubMed/NCBI

60 

Zheng H, Li BH, Liu C, Jia L and Liu FT: Comprehensive analysis of lncRNA-mediated ceRNA crosstalk and identification of prognostic biomarkers in Wilms' tumor. Biomed Res Int. 2020:49516922020.PubMed/NCBI

61 

A J, Zhang B, Zhang Z, Hu H and Dong JT: Novel gene signatures predictive of patient recurrence-free survival and castration resistance in prostate cancer. Cancers (Basel). 13:9172021. View Article : Google Scholar : PubMed/NCBI

62 

Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, Sun Y, Jacobsen A, Sinha R, Larsson E, et al: Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal. 6:pl12013. View Article : Google Scholar : PubMed/NCBI

63 

Mao X, Chen Y, Lu X, Jin S, Jiang P, Deng Z, Zhu X, Cai Q, Wu C and Kang S: Tissue resident memory T cells are enriched and dysfunctional in effusion of patients with malignant tumor. J Cancer. 14:1223–1231. 2023. View Article : Google Scholar : PubMed/NCBI

64 

Huang A and Zhou W: Mn-based cGAS-STING activation for tumor therapy. Chin J Cancer Res. 35:19–43. 2023. View Article : Google Scholar : PubMed/NCBI

65 

Feng S, Song G, Liu L, Liu W, Liang G and Song Z: Allergen-specific immunotherapy induces monocyte-derived dendritic cells but attenuates their maturation and cytokine production in the lesional skin of an atopic dermatitis mouse model. J Dermatol. 49:1310–1319. 2022. View Article : Google Scholar : PubMed/NCBI

66 

Guo Z, Wang YJ, He BS and Zhou J: Linc00312 single nucleotide polymorphism as biomarker for chemoradiotherapy induced hematotoxicity in nasopharyngeal carcinoma patients. Dis Markers. 2022:67078212022. View Article : Google Scholar : PubMed/NCBI

67 

Wang X, Yang T, Shi S, Xu C, Wang F, Dai D, Guan G, Zhang Y, Wang S, Wang J, et al: Heterogeneity-induced NGF-NGFR communication inefficiency promotes mitotic spindle disorganization in exhausted T cells through PREX1 suppression to impair the anti-tumor immunotherapy with PD-1 mAb in hepatocellular carcinoma. Cancer Med. 13:e67362024. View Article : Google Scholar : PubMed/NCBI

68 

Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, Jacobsen A, Byrne CJ, Heuer ML, Larsson E, et al: The cBio cancer genomics portal: An open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2:401–404. 2012. View Article : Google Scholar : PubMed/NCBI

69 

Fu S, Duan L, Zhong Y and Zeng Y: Comparison of surgical excision followed by adjuvant radiotherapy and laser combined with steroids for the treatment of keloids: A systematic review and meta-analysis. Int Wound J. 21:e144492023. View Article : Google Scholar : PubMed/NCBI

70 

Lee B, Lee S, Lee Y, Park Y and Shim J: Emerin represses STAT3 signaling through nuclear membrane-based spatial control. Int J Mol Sci. 22:66692021. View Article : Google Scholar : PubMed/NCBI

71 

Wu KY, Xie H, Zhang ZL, Li ZX, Shi L, Zhou W, Zeng J, Tian Z, Zhang Y, Ding YB and Shen WG: Emerin knockdown induces the migration and invasion of hepatocellular carcinoma cells by up-regulating the cytoplasmic p21. Neoplasma. 69:59–70. 2022. View Article : Google Scholar : PubMed/NCBI

72 

Awotoye W, Mossey PA, Hetmanski JB, Gowans LJJ, Eshete MA, Adeyemo WL, Alade A, Zeng E, Adamson O, James O, et al: Damaging mutations in AFDN contribute to risk of nonsyndromic cleft lip with or without cleft palate. Cleft Palate Craniofac J. 61:697–705. 2024. View Article : Google Scholar : PubMed/NCBI

73 

Berg HE, Greipp PT, Baughn LB, Falcon CP, Jackson CC and Peterson JF: Detection of a cryptic KMT2A/AFDN gene fusion [ins(6;11)(q27;q23q23)] in a pediatric patient with newly diagnosed acute myeloid leukemia. Lab Med. 53:e95–e99. 2022. View Article : Google Scholar : PubMed/NCBI

74 

Bill M, Mrózek K, Kohlschmidt J, Eisfeld AK, Walker CJ, Nicolet D, Papaioannou D, Blachly JS, Orwick S, Carroll AJ, et al: Mutational landscape and clinical outcome of patients with de novo acute myeloid leukemia and rearrangements involving 11q23/KMT2A. Proc Natl Acad Sci USA. 117:26340–26346. 2020. View Article : Google Scholar : PubMed/NCBI

75 

Chen Q, Zhou XW, Zhang AJ and He K: ACTN1 supports tumor growth by inhibiting Hippo signaling in hepatocellular carcinoma. J Exp Clin Cancer Res. 40:232021. View Article : Google Scholar : PubMed/NCBI

76 

Wang R, Gao Y and Zhang H: ACTN1 interacts with ITGA5 to promote cell proliferation, invasion and epithelial-mesenchymal transformation in head and neck squamous cell carcinoma. Iran J Basic Med Sci. 26:200–207. 2023.PubMed/NCBI

77 

Chen Q, Wang H, Li Z, Li F, Liang L, Zou Y, Shen H, Li J, Xia Y, Cheng Z, et al: Circular RNA ACTN4 promotes intrahepatic cholangiocarcinoma progression by recruiting YBX1 to initiate FZD7 transcription. J Hepatol. 76:135–147. 2022. View Article : Google Scholar : PubMed/NCBI

78 

Tentler D, Lomert E, Novitskaya K and Barlev NA: Role of ACTN4 in tumorigenesis, metastasis, and EMT. Cells. 8:14272019. View Article : Google Scholar : PubMed/NCBI

79 

Singla A, Chen Q, Suzuki K, Song J, Fedoseienko A, Wijers M, Lopez A, Billadeau DD, van de Sluis B and Burstein E: Regulation of murine copper homeostasis by members of the COMMD protein family. Dis Model Mech. 14:dmm0459632021. View Article : Google Scholar : PubMed/NCBI

80 

Iwuchukwu I, Nguyen D, Beavers M, Tran V, Sulaiman W, Fannin E, Lasseigne L, Ramsay E, Wilson J and Bazan NG: MicroRNA regulatory network as biomarkers of late seizure in patients with spontaneous intracerebral hemorrhage. Mol Neurobiol. 57:2346–2357. 2020. View Article : Google Scholar : PubMed/NCBI

81 

Neveu B, Richer C, Cassart P, Caron M, Jimenez-Cortes C, St-Onge P, Fuchs C, Garnier N, Gobeil S and Sinnett D: Identification of new ETV6 modulators through a high-throughput functional screening. iScience. 25:1038582022. View Article : Google Scholar : PubMed/NCBI

82 

da Silva AN, Ibelli AMG, Savoldi IR, Cantão ME, Zanella EL, Marques MG, da Silva MVGB, de Peixoto JO, Ledur MC, Lopes JS, et al: Whole-exome sequencing indicated new candidate genes associated with unilateral cryptorchidism in pigs. Sex Dev. 17:56–66. 2023. View Article : Google Scholar : PubMed/NCBI

83 

Barcelo J and Sanz-Moreno V: NECTIN1 is a melanoma metastasis suppressor gene. Nat Genet. 54:1776–1777. 2022. View Article : Google Scholar : PubMed/NCBI

84 

Ablain J, Al Mahi A, Rothschild H, Prasad M, Aires S, Yang S, Dokukin ME, Xu S, Dang M, Sokolov I, et al: Loss of NECTIN1 triggers melanoma dissemination upon local IGF1 depletion. Nat Genet. 54:1839–1852. 2022. View Article : Google Scholar : PubMed/NCBI

85 

Ho DW, Tsui YM, Chan LK, Sze KM, Zhang X, Cheu JW, Chiu YT, Lee JM, Chan AC, Cheung ET, et al: Single-cell RNA sequencing shows the immunosuppressive landscape and tumor heterogeneity of HBV-associated hepatocellular carcinoma. Nat Commun. 12:36842021. View Article : Google Scholar : PubMed/NCBI

86 

Zhang S, Jiang C, Su Y, Gui J, Yue Z, Jian B, He S and Ma X: Nectin2 influences cell apoptosis by regulating ANXA2 expression in neuroblastoma. Acta Biochim Biophys Sin (Shanghai). 55:356–366. 2023. View Article : Google Scholar : PubMed/NCBI

87 

Bhave S, Guyer RA, Picard N, Omer M, Hotta R and Goldstein AM: Ednrb−/− mice with hirschsprung disease are missing Gad2-expressing enteric neurons in the ganglionated small intestine. Front Cell Dev Biol. 10:9172432022. View Article : Google Scholar : PubMed/NCBI

88 

Zheng Z, Gao M, Tang C, Huang L, Gong Y, Liu Y and Wang J: E. coli JM83 damages the mucosal barrier in Ednrb knockout mice to promote the development of Hirschsprung-associated enterocolitis via activation of TLR4/p-p38/NF-κB signaling. Mol Med Rep. 25:1682022. View Article : Google Scholar : PubMed/NCBI

89 

Geng B, Wang X, Park KH, Lee KE, Kim J, Chen P, Zhou X, Tan T, Yang C, Zou X, et al: UCHL1 protects against ischemic heart injury via activating HIF-1α signal pathway. Redox Biol. 52:1022952022. View Article : Google Scholar : PubMed/NCBI

90 

Mondal M, Conole D, Nautiyal J and Tate EW: UCHL1 as a novel target in breast cancer: Emerging insights from cell and chemical biology. Br J Cancer. 126:24–33. 2022. View Article : Google Scholar : PubMed/NCBI

91 

Tang J, Yang Q, Mao C, Xiao D, Liu S, Xiao L, Zhou L, Wu G and Tao Y: The deubiquitinating enzyme UCHL3 promotes anaplastic thyroid cancer progression and metastasis through Hippo signaling pathway. Cell Death Differ. 30:1247–1259. 2023. View Article : Google Scholar : PubMed/NCBI

92 

He R, Zhou Y, Liu J, Zhang X, Zhao X, An L, Li Z and Cheng F: UCHL3 plays an important role in the occurrence and development of melanoma. Oncol Lett. 22:7562021. View Article : Google Scholar : PubMed/NCBI

93 

Thompson LL, Rutherford KA, Lepage CC and McManus KJ: Aberrant SKP1 expression: Diverse mechanisms impacting genome and chromosome stability. Front Cell Dev Biol. 10:8595822022. View Article : Google Scholar : PubMed/NCBI

94 

Biryukov M, Dmitrieva A, Vavilova V, Ustyantsev K, Bazarova E, Sukhikh I, Berezikov E and Blinov A: Mlig-SKP1 gene is required for spermatogenesis in the flatworm macrostomum lignano. Int J Mol Sci. 23:151102022. View Article : Google Scholar : PubMed/NCBI

95 

Engeland K: Cell cycle regulation: p53-p21-RB signaling. Cell Death Differ. 29:946–960. 2022. View Article : Google Scholar : PubMed/NCBI

96 

Salaroglio IC, Belisario DC, Bironzo P, Ananthanarayanan P, Ricci L, Digiovanni S, Fontana S, Napoli F, Sandri A, Facolmatà C, et al: SKP2 drives the sensitivity to neddylation inhibitors and cisplatin in malignant pleural mesothelioma. J Exp Clin Cancer Res. 41:752022. View Article : Google Scholar : PubMed/NCBI

97 

Surka C, Jin L, Mbong N, Lu CC, Jang IS, Rychak E, Mendy D, Clayton T, Tindall E, Hsu C, et al: CC-90009, a novel cereblon E3 ligase modulator, targets acute myeloid leukemia blasts and leukemia stem cells. Blood. 137:661–677. 2021. View Article : Google Scholar : PubMed/NCBI

98 

Jia L and Sun Y: RBX1/ROC1-SCF E3 ubiquitin ligase is required for mouse embryogenesis and cancer cell survival. Cell Div. 4:162009. View Article : Google Scholar : PubMed/NCBI

99 

Bays JL and DeMali KA: Vinculin in cell-cell and cell-matrix adhesions. Cell Mol Life Sci. 74:2999–3009. 2017. View Article : Google Scholar : PubMed/NCBI

100 

Shih YT, Wei SY, Chen JH, Wang WL, Wu HY, Wang MC, Lin CY, Lee PL, Lin CY, Chiang HC, et al: Vinculin phosphorylation impairs vascular endothelial junctions promoting atherosclerosis. Eur Heart J. 44:304–318. 2023. View Article : Google Scholar : PubMed/NCBI

101 

Yang W, Li J, Zhang M, Yu H, Zhuang Y, Zhao L, Ren L, Gong J, Bi H, Zeng L, et al: Elevated expression of the rhythm gene NFIL3 promotes the progression of TNBC by activating NF-κB signaling through suppression of NFKBIA transcription. J Exp Clin Cancer Res. 41:672022. View Article : Google Scholar : PubMed/NCBI

102 

Sarkozy C, Hung SS, Chavez EA, Duns G, Takata K, Chong LC, Aoki T, Jiang A, Miyata-Takata T, Telenius A, et al: Mutational landscape of gray zone lymphoma. Blood. 137:1765–1776. 2021. View Article : Google Scholar : PubMed/NCBI

103 

Qie S: The E3 ubiquitin ligase fbxo4 functions as a tumor suppressor: Its biological importance and therapeutic perspectives. Cancers (Basel). 14:21332022. View Article : Google Scholar : PubMed/NCBI

104 

Mucha B, Qie S, Bajpai S, Tarallo V, Diehl JN, Tedeschi F, Zhou G, Gao Z, Flashner S, Klein-Szanto AJ, et al: Tumor suppressor mediated ubiquitylation of hnRNPK is a barrier to oncogenic translation. Nat Commun. 13:66142022. View Article : Google Scholar : PubMed/NCBI

105 

Wang L, Piao Y, Zhang D, Feng W, Wang C, Cui X, Ren Q, Zhu X and Zheng G: Fbxw11 impairs the repopulation capacity of hematopoietic stem/progenitor cells. Stem Cell Res Ther. 13:2452022. View Article : Google Scholar : PubMed/NCBI

106 

Chen C, Zhou H, Zhang X, Liu Z and Ma X: Association of FBXW11 levels with tumor development and prognosis in chondrosarcoma. Cancer Biomark. 35:429–437. 2022. View Article : Google Scholar : PubMed/NCBI

107 

Zou C, Chen Y, Smith RM, Snavely C, Li J, Coon TA, Chen BB, Zhao Y and Mallampalli RK: SCF(Fbxw15) mediates histone acetyltransferase binding to origin recognition complex (HBO1) ubiquitin-proteasomal degradation to regulate cell proliferation. J Biol Chem. 288:6306–6316. 2013. View Article : Google Scholar : PubMed/NCBI

108 

De La Chesnaye E, Méndez JP, López-Romero R, De Los Angeles Romero-Tlalolini M, Vergara MD, Salcedo M and Ojeda SR: FBXW12, a novel F box protein-encoding gene, is deleted or methylated in some cases of epithelial ovarian cancer. Int J Clin Exp Pathol. 8:10192–10203. 2015.PubMed/NCBI

109 

Zhang J, Gan Y, Li H, Yin J, He X, Lin L, Xu S, Fang Z, Kim BW, Gao L, et al: Inhibition of the CDK2 and cyclin A complex leads to autophagic degradation of CDK2 in cancer cells. Nat Commun. 13:28352022. View Article : Google Scholar : PubMed/NCBI

110 

Arora M, Moser J, Hoffman TE, Watts LP, Min M, Musteanu M, Rong Y, Ill CR, Nangia V, Schneider J, et al: Rapid adaptation to CDK2 inhibition exposes intrinsic cell-cycle plasticity. Cell. 186:2628–2643.e21. 2023. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2024
Volume 52 Issue 3

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zeng Q, Jiang T and Wang J: Role of LMO7 in cancer (Review). Oncol Rep 52: 117, 2024
APA
Zeng, Q., Jiang, T., & Wang, J. (2024). Role of LMO7 in cancer (Review). Oncology Reports, 52, 117. https://doi.org/10.3892/or.2024.8776
MLA
Zeng, Q., Jiang, T., Wang, J."Role of LMO7 in cancer (Review)". Oncology Reports 52.3 (2024): 117.
Chicago
Zeng, Q., Jiang, T., Wang, J."Role of LMO7 in cancer (Review)". Oncology Reports 52, no. 3 (2024): 117. https://doi.org/10.3892/or.2024.8776