Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Oncology Letters
      • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Biomedical Reports
      • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • Information for Authors
    • Information for Reviewers
    • Information for Librarians
    • Information for Advertisers
    • Conferences
  • Language Editing
Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • For Authors
    • For Reviewers
    • For Librarians
    • For Advertisers
    • Conferences
  • Language Editing
Login Register Submit
  • This site uses cookies
  • You can change your cookie settings at any time by following the instructions in our Cookie Policy. To find out more, you may read our Privacy Policy.

    I agree
Search articles by DOI, keyword, author or affiliation
Search
Advanced Search
presentation
International Journal of Molecular Medicine
Join Editorial Board Propose a Special Issue
Print ISSN: 1107-3756 Online ISSN: 1791-244X
Journal Cover
December-2025 Volume 56 Issue 6

Full Size Image

Sign up for eToc alerts
Recommend to Library

Journals

International Journal of Molecular Medicine

International Journal of Molecular Medicine

International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.

International Journal of Oncology

International Journal of Oncology

International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.

Molecular Medicine Reports

Molecular Medicine Reports

Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.

Oncology Reports

Oncology Reports

Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.

Oncology Letters

Oncology Letters

Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.

Biomedical Reports

Biomedical Reports

Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.

Molecular and Clinical Oncology

Molecular and Clinical Oncology

International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.

World Academy of Sciences Journal

World Academy of Sciences Journal

Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.

International Journal of Functional Nutrition

International Journal of Functional Nutrition

Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.

International Journal of Epigenetics

International Journal of Epigenetics

Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.

Medicine International

Medicine International

An International Open Access Journal Devoted to General Medicine.

Journal Cover
December-2025 Volume 56 Issue 6

Full Size Image

Sign up for eToc alerts
Recommend to Library

  • Article
  • Citations
    • Cite This Article
    • Download Citation
    • Create Citation Alert
    • Remove Citation Alert
    • Cited By
  • Similar Articles
    • Related Articles (in Spandidos Publications)
    • Similar Articles (Google Scholar)
    • Similar Articles (PubMed)
  • Download PDF
  • Download XML
  • View XML
Review Open Access

Emerging role of long non‑coding RNAs in the Rho/ROCK signaling pathway in tumor metastasis (Review)

  • Authors:
    • Hui Nie
    • Xuejie Yang
    • Qingning Chen
    • Xiaoyun He
    • Yingying Han
    • Qiuyan He
    • Chunlin Ou
  • View Affiliations / Copyright

    Affiliations: Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
    Copyright: © Nie et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
  • Article Number: 197
    |
    Published online on: September 12, 2025
       https://doi.org/10.3892/ijmm.2025.5638
  • Expand metrics +
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Metrics: Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )
Cited By (CrossRef): 0 citations Loading Articles...

This article is mentioned in:



Abstract

Metastasis is a critical feature of malignant tumors and a major cause of treatment failure and poor prognosis. This complex process primarily involves the rearrangement of the cytoskeleton. Among several cytoskeleton‑associated signaling pathways, the ras homolog family (Rho) and Rho‑associated coiled‑coil containing protein kinase (ROCK) signaling pathway serves a key role in cytoskeleton regulation as it controls intracellular actin dynamics and cytoskeletal remodeling to mediate cell movement. Long non‑coding RNAs (lncRNAs), known to serve important roles in tumorigenesis and metastasis, have been shown to regulate the Rho/ROCK pathway. lncRNAs modulate the Rho/ROCK pathway by sponging microRNAs or directly binding to proteins, thereby mediating tumor metastasis. Additionally, lncRNAs can be encapsulated in exosomes and transferred from donor cells to recipient cells, where they regulate components of the Rho/ROCK signaling pathway and further promote the metastatic potential. Based on their functional significance, Rho/ROCK signaling pathway‑associated lncRNAs may serve as novel tumor biomarkers and therapeutic targets. The present review summarizes the role and mechanism of Rho/ROCK signaling pathway‑associated lncRNAs in tumor metastasis, which may offer novel avenues for the diagnosis and treatment of metastatic cancer.

Introduction

Tumor metastasis is a hallmark of tumor development and progression. Tumor metastasis involves the detachment of tumor cells from the primary site through a series of biological processes, including local infiltration, entry into the vascular system, circulation, colonization of secondary tissues or organs, and the formation of secondary tumors (1-4). This complex process of tumor invasion and metastasis involves multiple factors, including changes in the tumor microenvironment, epithelial-mesenchymal transformation (EMT), tumor hypoxia, neovascularization, decreased intercellular adhesion, cytoskeleton remodeling, extracellular matrix degradation, and the formation of cellular pseudopods and protuberances (5,6). Although advances in existing treatments have improved survival rates for some patients with cancer, distant metastasis remains a major challenge (7,8). Once metastasis occurs, long-term survival markedly decreases, and numerous patients succumb to the disease shortly thereafter (9). Tumor metastasis is one of the most important contributors to cancer-related mortality and is a major obstacle in effective clinical management (10). Therefore, there is an urgent need to identify metastasis-specific biomarkers and to explore the underlying molecular mechanisms.

Among numerous pathways, the ras homolog family (Rho) and Rho-associated coiled-coil containing protein kinase (ROCK) signaling pathway serves a key role in tumor metastasis. This signaling pathway regulates cytoskeletal remodeling by modulating myosin and actin activity, establishing cell polarity, and organizing intermediate filaments. This pathway also contributes to tumor cell proliferation, contraction, migration, adhesion and cell-matrix interactions (11). The most widely studied Rho GTP enzymes include Rho, Ras-related C3 botulinum toxin substrate (Rac) and cell division cycle 42 (Cdc42) (12,13). These receptor proteins initiate cytoskeleton reorganization and regulate downstream effectors that affect tumor cell migration (14). Furthermore, the Rho/ROCK pathway can regulate HIPPO/yes-associated protein (YAP) signaling by modulating the actinomycin skeleton, with YAP acting as a mechanical sensor that alters gene expression in response to cytoskeleton dynamics (15). The cytoskeleton, a dynamic network of protein fibers in eukaryotic cells, serves an important role in cell division, intracellular transport, motility and shape maintenance (16,17). The cytoskeleton comprises three main components: Microfilaments, microtubules and intermediate filaments (18). Microfilaments are primarily involved in cytokinesis, cell motility and the formation of intracellular stress fibers. Microtubules maintain cell morphology, resist compression and facilitate intracellular trafficking. Intermediate filaments provide mechanical stability and coordinate cell migration. Together, these three cytoskeletal components coordinate cellular integrity and ensure the smooth progression of several biological functions, such as maintaining cell morphology (16), material transport (19) and cell movement (20). Microtubules serve a crucial role in cancer progression and metastasis (21,22). Microtubule-targeting agents have emerged as promising tools to disrupt cancer cell activity and inhibit metastasis (23,24).

Some antibody-drug conjugates, especially those combined with microtubule-targeting compounds, have yielded encouraging effectiveness in reducing tumor burden at metastatic sites (25,26). Nestin, an intermediate filament protein, is upregulated in several tumors, including pancreatic and prostate cancer, malignant melanoma, and glioma, and is associated with tumor aggressiveness, metastasis and poor prognosis (27,28).

Long non-coding RNAs (lncRNAs) were first identified in the nucleus and cytoplasm of eukaryotic cells (29). Most lncRNAs lack a fully functional open reading frame; however, previous studies have shown that a few lncRNAs can encode a small number of functional short peptides (30-32). Based on their location in the genome, lncRNAs have been classified into five categories: Sense, antisense, bidirectional, intergenic and intronic lncRNAs (33,34). Functionally, lncRNAs can be classified into four types: Signaling, decoy, guide and scaffold lncRNAs (35). Signaling lncRNAs can be used as marker signals in different temporal spaces, developmental stages and gene expression regulation (36). For example, there are three common signaling lncRNAs: Air, Kcnq1ot1 and Xist. They mediate the transcriptional silencing of multiple genes by interacting with chromatin and recruiting chromatin modification mechanisms (37). Guide lncRNAs can guide the localization of ribonucleoprotein complexes to specific targets. For example, the lncRNA COLDAIR can guide the polycomb repressive complex 2 (PRC2) complex to serve a key biological role in the chromatin of the floral repressor, flowering locus c (38). Traditionally, proteins have been important players in various types of scaffold complexes (39). However, it has been shown that lncRNAs can also serve as central platforms for the assembly of relevant molecular components in several biological signaling processes, and this precisely controlled property is essential for precise control of the specificity and dynamics of intermolecular interactions and signaling events (40). lncRNAs have been confirmed to be associated with various cancer types, including colorectal, gastric, hepatocellular and esophageal cancer (41-44). For example, lncRNA CCAT1 and HOTAIR in the plasma of patients with colorectal cancer (CRC) have been shown to serve as biological markers in CRC screening (41). Aberrant expression of three lncRNAs (POU3F3, SPRY4-IT1 and HNF1A-AS1) has been detected in the sera of patients with oesophageal cancer. Notably, the combined use of the three could effectively detect the early development of esophageal cancer (44). lncRNAs also serve key roles in tumor metastasis. lncRNA HIF1A-AS2 can promote the proliferation and invasion of triple-negative breast cancer (TNBC) cells (45). Compared with those in normal lung tissue, lncRNA MALAT1 expression levels are elevated in lung cancer, and abnormal MALAT1 expression can induce EMT to further promote brain metastasis in lung cancer (46). With the continuous development of high-throughput sequencing, numerous lncRNAs have been identified due to their specific expression in tumor metastasis, and as molecular biomarkers for tumor metastasis and potential therapeutic targets for several tumors (47-49). For instance, Zhang et al (50) developed a novel computational framework employing six machine learning algorithms to comprehensively analyze transcriptomic data from purified immune cells, glioblastoma cell lines and bulk glioblastoma tissue. This framework was utilized to screen for tumor-infiltrating immune cell-associated lncRNAs predictive of prognosis and response to immunotherapy in patients with glioblastoma.

In summary, among the multiple complex mechanisms of tumor metastasis, lncRNAs are emerging as important modulators of cytoskeleton dynamics, and likely affect tumor cell metastasis by directly binding to cytoskeleton-related proteins or by indirectly regulating key molecules in the Rho/ROCK signaling pathway. The present review aims to explore the molecular mechanisms through which lncRNAs mediate the Rho/ROCK signaling pathway during tumor metastasis. An improved understanding of these interactions between lncRNAs and the Rho/ROCK signaling pathway may provide novel strategies for the early diagnosis and targeted treatment of metastatic tumors.

Definition, mechanism and functions of lncRNAs

Definition and biological characteristics of lncRNAs

The majority of the human genome (76-97%) is transcribed into RNAs that are not translated into proteins, and are referred to as non-coding RNAs (ncRNAs) (51). Taking 200 nt as the threshold, ncRNAs are classified into two categories: Short ncRNAs (<200 nt) and lncRNAs (>200 nt) (52). According to their genomic proximity to neighboring transcripts, lncRNAs can be classified into four categories: i) Intergenic lncRNAs, which are located between two protein-coding transcripts; ii) intronic lncRNAs, which are present in the introns of coding transcripts; iii) sense/antisense strand lncRNAs, which have overlapping parts with introns and exons of different coding transcripts; and iv) bidirectional lncRNAs, which share a promoter and are transcribed from both sense and antisense directions of transcription start areas (53). lncRNAs are transcribed by polymerase II (54). Similar to mRNA, most lncRNAs undergo 5' end capping, polyadenylation and splicing. They are different in that the number of lncRNAs in exons is lower than that in mRNA; therefore, lncRNAs are evolutionarily conserved to a certain extent. Secondary and advanced structures are the most important features of lncRNAs. lncRNAs form secondary structures, including double helices, hairpin loops, protrusions and pseudoknots, as well as tertiary structures with non-Watson-Crick base pairs and more advanced structures. These structures form the basis for the biological functions of lncRNAs. lncRNAs are unevenly distributed in the nucleus and cytoplasm (55); however, the specific localization of lncRNAs in the cytoplasm or nucleus, and the factors determining such localization have not been fully confirmed (56).

Biological functions of lncRNAs

In the whole genome, 50-70% of genomic DNA can be transcribed; however, <2% of this is ultimately translated into proteins (57), and the vast majority of the remaining transcripts are transcribed into non-coding ncRNAs. Although not translated, previous studies have shown that ncRNAs are widely involved in various biological processes of organisms (34,58-60). Compared with mRNAs, lncRNAs have higher spatial and temporal specificity and lower interspecies conservation, and they serve an essential role in regulating chromatin dynamics (61,62), gene expression (63), cell proliferation (64), cell differentiation (65) and development of organisms (63). One of the most important functions of lncRNAs is the regulator of transcription (66,67). Among the identified action modes, signaling, decoy, scaffolding and guidance are the most frequent modes of action of lncRNAs in the cell (68). The main function of signaling type lncRNAs is to regulate transcription as a molecular signal in response to various stimuli, while the chromatin state of regulatory elements can be further inferred from the expression of related lncRNAs (36). In addition, cellular stimulation can affect the transcriptional expression of lncRNAs, which can perform regulatory functions to affect the biological processes of organisms (69). Therefore, their production and presence can be used as an indicator of transcriptional activity. For example, DNA damage can induce the transcription of lncRNA PANDA in a p53-dependent manner, which acts to limit the expression of pro-apoptotic genes and arrest the cell cycle after interacting with transcription factors (70). In an experiment involving the induction of somatic cell reprogramming in pluripotent stem cells, Loewer et al (71) demonstrated that a large number of lncRNAs were aberrantly expressed during this process. Among them, lncRNA RoR could directly regulate the pluripotency factors OCT4, SOX2 and Nanog, and served a role as a signaling molecule in the pluripotency and reprogramming of stem cells. Decoy lncRNAs can directly bind regulatory molecules such as transcription factors or RNA-binding proteins, thereby sequestering them and blocking their activity and downstream signaling pathways (72,73). For example, lncRNA PANDA inhibits the expression of apoptotic genes by directly binding to nuclear transcription factor Y subunit α, thus arresting the cell cycle (74). Scaffold lncRNAs can serve as molecular scaffolds that guide the assembly of protein complexes onto target genes (75). For example, HOTAIR serves as a molecular scaffold that links and targets the PRC2 and the lysine specific demethylase 1 histone-modifying complexes, thereby coordinating H3K27 methylation and H3K4 demethylation to reprogram chromatin states (76). Guide lncRNAs perform a 'guide-like' function, interacting with ribonucleoprotein particles and taking them to specific target genes, and serve the roles of cis-guides and trans-guides (77). In addition, some studies have shown that a small number of lncRNAs can encode short peptides that further function in the organism (78-80). With the widespread application of next-generation sequencing technologies in different tumors, the mystery of lncRNAs has been gradually unraveled. Thousands of lncRNAs are aberrantly expressed in various tumors, suggesting that they may serve an important role in the disease process of tumors (81). The validated mechanisms of lncRNAs in tumors are acting as competing endogenous RNAs (ceRNAs), participating in chromatin remodeling, regulating chromatin interactions and acting as natural antisense transcripts (82). lncRNA HOTAIR is dysregulated in a variety of cancer types (76,83,84), such as oral squamous cell carcinoma (OSCC), hepatocellular carcinoma (HCC) and breast cancer. Wu et al (76) found that HOTAIR binds with EZH2 and H3K27me3 to form a complex, which can bind to the promoter region of E-cadherin to regulate its expression, thereby affecting OSCC metastasis. Recent studies have revealed that lncRNAs can be encapsulated in exosomes, and transferred from donor cells to recipient cells in the tumor microenvironment by the fluid circulation, thereby regulating several phenotypes of tumor processes (85,86), including malignant proliferation and invasive metastasis of tumor cells, radiotherapy resistance of tumor cells, formation of the tumor microenvironment and internal angiogenesis. For example, Zhang et al (87) reported that exosomal lncRNA MALAT1 was differentially expressed in non-small cell lung cancer (NSCLC), high exosomal MALAT1 expression could promote metastasis in NSCLC and the expression levels of MALAT1 were closely associated with lymph node metastasis in patients with NSCLC. In addition, exosomal lncRNAs released into the tumor microenvironment have the potential to become tumor markers due to their specificity and sensitivity (88-90).

Rho/ROCK signaling pathway

Composition of the Rho/ROCK signaling pathway

The Rho/ROCK signaling pathway consists of the Rho-GTPase family and its downstream effector ROCK (91-93) (Fig. 1). Rho GTPases are members of the Ras superfamily of small GTP-binding proteins and contain >20 Rho proteins that can be broadly classified into the following five groups based on their primary sequence and functional type: Rho, Rac, Cdc42, Rho Family GTPase (Rnd) and Rho-related BTB domain-containing subfamilies (92). Among these, the Rho, Rac and Cdc42 subfamilies are the most intensively investigated and best characterized functionally. The Rho subfamily includes RhoA, RhoB and RhoC, which are very similar in sequence, and because they share the same set of effectors, the mode of action of the three is presumed to be similar (94). RhoA, RhoB and RhoC have two different states in organisms (inactive GDP-bound and active GTP-bound forms); the molecules cycle between the two states. The transition from the inactive GDP-bound form to the active GTP-bound form is catalyzed by the diffuse B-cell lymphoma family of RhoGTP guanine nucleotide exchange factors (95). By contrast, the transition from the active to the inactive state is mediated by intrinsic GTPase hydrolysis stimulated by Rho GTPase-activating proteins (96). In the active state, Rho proteins act on >60 downstream targets, including ROCK, mammalian homolog of Drosophila (mDia), Par6B, p21 (RAC1) activated kinase 4 and Wiskott-Aldrich syndrome proteins (97). Activation of RhoGTPase ultimately leads to the remodeling of the cytoskeleton and changes in other fundamental processes, such as cell division, adhesion and migration (98,99). The ROCK family consists of two structurally similar isoforms, ROCK1 and ROCK2. Both kinases share >30 direct downstream substrates, including myosin phosphatase-targeting subunit 1, myosin light chain (MLC) and LIM domain kinase, in addition to containing an N-terminal catalytic kinase structural domain, a central coiled-coil structural domain and a C-terminal PH structural domain (100). In Rho/ROCK signaling, Rho (RhoA/RhoC) activates Rho-related kinases (ROCK1/ROCK2) by binding to their C-terminal. ROCK regulates key proteins in the cytoskeleton by mediating the phosphorylation of MLC, and activating Lin-11, Isl1 and MEC-3 (LIM) structural domain kinases, which are widely involved in basic cellular functions, including adhesion, migration, contraction, proliferation and apoptosis (101).

Members of the Rho family. The main
members of the Rho family include the Rnd, Rac, Rho, Cdc42 and
RhoBTB subfamilies. The members of the Rho subfamily include RhoA,
RhoB and RhoC. The members of the Rho subfamily regulate the
cytoskeleton by modulating the ROCK/MLCP and ROCK/LIMK pathways.
Cdc42, cell division cycle 42; LIMK, LIM domain kinase; MLCP,
myosin light chain phosphatase; Rac, Ras-related C3 botulinum toxin
substrate; Rho, Rho family GTPases; RhoBTB, Rho-related BTB
domain-containing; Rnd, Rho family GTPase; ROCK, Rho-associated
coiled-coil containing protein kinase.

Figure 1

Members of the Rho family. The main members of the Rho family include the Rnd, Rac, Rho, Cdc42 and RhoBTB subfamilies. The members of the Rho subfamily include RhoA, RhoB and RhoC. The members of the Rho subfamily regulate the cytoskeleton by modulating the ROCK/MLCP and ROCK/LIMK pathways. Cdc42, cell division cycle 42; LIMK, LIM domain kinase; MLCP, myosin light chain phosphatase; Rac, Ras-related C3 botulinum toxin substrate; Rho, Rho family GTPases; RhoBTB, Rho-related BTB domain-containing; Rnd, Rho family GTPase; ROCK, Rho-associated coiled-coil containing protein kinase.

The vast majority of cellular activities are directly or indirectly influenced by Rho family proteins. Rho family proteins regulate the actin cytoskeleton to control cell morphology, and serve an important role in cell polarity, endocytosis, vesicle trafficking, adhesion and migration (13). RhoA interacts with effectors at the cell membrane (102). RhoA mediates the formation of myosin bundles and stress fibers (103), and serves an important role in membrane folding and lamellipodia formation (104). RhoB is present on the outer membranes of multivesicular bodies and in the plasma membrane and serves a role in vesicle transport (105). RhoC is mainly located in the plasma membrane or cytoplasmic matrix, and mainly regulates the activity of actin and myosin and limits lamellipodial broadening (106). The Rac subfamily primarily stimulates the formation of lamellar pseudopods, membrane folds and invasive foot types (107). Rac1 is commonly distributed in tissues, and mainly regulates the formation of scaffolding proteins, membrane ruffles and lamellipodia (108). Rac2 is confined to haematopoietic tissues, and regulates cell adhesion and participates in cellular immune synapse formation (109). Rac3 is a neuron-specific protein that is important for regulating autophagy, inducing the formation of invadopodia and degrading extracellular matrix (110,111) The Cdc42 subfamily mediates the formation of a third actin-based structure, the filopodia, by binding to Wiskott-Aldrich syndrome protein or neural Wiskott-Aldrich syndrome protein (112). Additionally, Cdc42 serves an important role in cell polarization (113). Rho downstream effectors include ROCK and p140mDia (114). The ROCK family belongs to the AGC family of serine/threonine protein kinases. ROCK1 serves a key role in the formation of actin bundles, actin contractility and stress fibers by mediating the cross-linking of myosin, while ROCK2 is required for phagocytosis and cell contraction and is important in stabilizing the cytoskeleton (115). The Rho/ROCK signaling pathway serves a key role in central nervous system disorders, pulmonary hypertension and other diseases (116,117).

Rho/ROCK signaling pathway and tumor metastasis

Metastasis, the spread of malignant cells to distant organs, represents the ultimate stage of cancer progression and remains a leading cause of cancer-related deaths (107). The majority of cancer deaths result not from the primary tumor but from the secondary lesions in vital organs. This complex process includes a series of sequential events: Primary tumor cells gradually acquire invasive abilities, spread through blood or lymphatic vessels (or directly invade adjacent tissues), and finally colonize and proliferate in distant organs (118). Although different cancer types and molecular subtypes exhibit marked differences in metastasis-driving genes, microenvironmental cues and anatomical dissemination routes, several core cellular processes are shared (119). Among these, cell migration is a core process that is universally present across various tumor types (120). Therefore, targeting the signaling networks that regulate cell motility could offer an effective strategy for the management of metastasis in multiple cancer types (121-123).

The Rho GTPase family functions as a core molecular switch regulating cytoskeletal dynamics and cell movement (124). Growing evidence has highlighted the critical role of this family in tumor invasion and metastasis (123,125). For example, Zhang et al (126) demonstrated that inhibition of RhoA suppressed proliferation in gastric cancer cells. In breast cancer, altered Rho GTPase signaling disrupts cytoskeleton architecture in cancer cells, and serves a critical role in cell motility, migration and invasion (127). In cholangiocarcinoma, tumor cells recruit cancer-associated fibroblasts by secreting PDGF-D, which stimulates fibroblast migration by upregulating platelet-derived growth factor receptor β and Rho GTPase, and activating the JNK pathway (128). With the emergence of pharmacological agents targeting the Rho/ROCK signaling axis, particularly selective ROCK inhibitors, this pathway has become a promising target for the development of antimetastatic treatments (123).

Regulatory mechanisms of Rho/ROCK signaling pathway-associated lncRNAs in tumor metastasis

lncRNAs regulate tumor metastasis by sponging miRNAs

The subcellular localization of lncRNAs is closely related to their biological functions (129). Unlike the relatively discrete cytoplasmic distribution of mRNAs, most lncRNAs are found in both the nucleus and cytoplasm, with the cytoplasm often being their primary site of localization (130,131). Structurally, lncRNAs contain intronic and other non-coding elements that provide potential binding sites for miRNAs. Through sequence complementarity, lncRNAs can bind intracellular miRNAs and weaken or block their ability to suppress target genes (24). This competitive binding between lncRNAs and miRNAs forms ceRNA networks, indirectly affecting components of the Rho/ROCK signaling pathway (132).

Several studies have demonstrated that lncRNAs influence components of the Rho/ROCK signaling pathway via ceRNA networks, ultimately modulating tumor cell proliferation, invasion, migration and apoptosis (38,133-181) (Table I). For example, the expression of lncRNA DAPK1 is reduced in pancreatic cancer tissues, where it modulates invasion and migration by sponging miR-182 to regulate ROCK1 expression (133). Similarly, Liu et al (134) demonstrated that lncRNA ZFAS1 promoted pancreatic cancer metastasis by acting as a sponge for miR-3924 and subsequently regulated the RhoA/ROCK2 pathway to serve a pro-metastatic role in pancreatic cancer. Furthermore, lncRNA NORAD facilitates EMT in pancreatic cancer by binding miR-125a-3p, which regulates the expression of the downstream effector molecule RhoA (135). Liu et al (136) reported that lncRNA NEAT1 acted as a ceRNA by sponging miR-382-3p and upregulating ROCK1 in ovarian cancer (OC), and thus, enhanced the metastatic potential. NEAT1 also inhibits the progression of lung adenocarcinoma by sponging miR-490-3p, leading to suppression of the RhoA/ROCK pathway (137). Furthermore, Zhang et al (138) found that lncRNA LINC01087 promoted lung adenocarcinoma progression by regulating the miR-514a-3p/CEP55/RhoA/ROCK1 axis.

Table I

Relationship between Rho/ROCK signaling pathway-associated lncRNAs and miRNAs in tumor metastasis.

Table I

Relationship between Rho/ROCK signaling pathway-associated lncRNAs and miRNAs in tumor metastasis.

Authors, yearCancer typeslncRNAsmiRNAsTarget genesExpression alterationFunctions(Refs.)
Li et al, 2020Papillary thyroid carcinomaDLEU1miR-421ROCK1↑Promotes proliferation and invasion(145)
Lian et al, 2018Nasopharyngeal carcinomaAFAP1-AS1miR-423-5pRAB11B/LASP1↑Promotes invasion and metastasis(146)
Feng et al, 2020EGFR-AS1miR-145ROCK1↑Promotes invasion and migration(147)
Zhou et al, 2020Non-small cell lung cancerZFAS1miR-590-3pCdc42↑Promotes proliferation(148)
Song et al, 2022BCYRN1miR-30b-3pROCK1↑Promotes proliferation, migration and invasion(149)
Yang et al, 2019Lung adenocarcinomaLCAT1miR-4715-5pRAC1↑Promotes proliferation and invasion(150)
Xiao et al, 2019MALAT1miR-429RhoA↑Promotes proliferation, metastasis and invasion(151)
Zhao et al, 2023NEAT1miR-490-3pRhoA↑Promotes proliferation and migration(137)
Zhang et al, 2024LINC01087miR-514a-3pCEP55/RhoA↑Promotes proliferation, migration and invasion(138)
Chen et al, 2018Hepatocellular carcinomaCDKN2BASmiR-153-5pARHGAP18↑Promotes migration(143)
Chen and Zhang, 2019LINC00339miR-152ROCK1↑Promotes proliferation and migration(152)
Wang et al, 2021LINC00491miR-324-5pROCK1↑Promotes invasion and metastasis(153)
You et al, 2021LINC00161miR-590-3pROCK2↑Promotes angiogenesis and metastasis(154)
Dong et al, 2023LINC00607miR-584-3pROCK1↑Promotes proliferation, migration and invasion(144)
Zhou et al, 2019GliomaH19miR-15bCdc42↑Promotes proliferation, migration and invasion(155)
Guo et al, 2019DANCRmiR-27a-3pROCK1↑Promotes proliferation and metastasis(156)
Ma et al, 2017SNHG15miR-153Cdc42↑Promotes vascular endothelial cell proliferation, migration and tube formation(157)
Li et al, 2020GlioblastomaXISTmiR-448ROCK1↑Promotes proliferation, migration and invasion(158)
Chen et al, 2020LINC00346miR-340-5pROCK1↑Promotes proliferation, migration and invasion(159)
Wang et al, 2021DLGAP1-AS1miR-515-5pROCK1↑Promotes proliferation(160)
Liu et al, 2019Acute myeloid leukemiaLINC00662miR-340-5pROCK1↑Promotes proliferation and inhibits apoptosis(161)
Bai et al, 2023Cervical cancerLOXL1-AS1miR-21RHOB↓Inhibits proliferation, migration and invasion(162)
Chen et al, 2020LINC02381miR-133bRhoA↑Promotes proliferation and migration(163)
Wang et al, 2018OsteosarcomaDANCRmiR-335-5pROCK1↑Promotes proliferation, metastasis, invasion and EMT(139)
Shao et al, 2020ZNF281miR-144ROCK1↑Promotes metastasis and invasion(140)
Deng et al, 2019SNHG1miR-101-3pROCK1↑Promotes proliferation, migration and invasion(141)
Wang et al, 2018SNHG5miR-26aROCK1↑Promotes proliferation, metastasis and invasion(142)
Yao et al, 2020GAS5miR-663aRHOB↓Inhibits proliferation, migration and invasion(164)
Wang et al, 2017Colorectal cancerTUG1miR-335-5pROCK1↑Promotes proliferation, migration and invasion(165)
Cui et al, 2017HOXA11-ASmiR-124-3pROCK1↑Promotes proliferation, metastasis and invasion(166)
Yu et al, 2019XISTmiR-133a-3pRhoA↑Promotes EMT and infiltration of macrophages(167)
Kong et al, 2021MCF2L-AS1miR-105-5pRAB22A↑Promotes proliferation, metastasis, invasion and EMT(168)
Horita et al, 2019UCA1 miR-18a/miR-182Cdc42↑Enhances sensitivity to oncolytic vaccinia virus(169)
Tian et al, 2021Oral squamous cell carcinomaLINC00974miR-122RhoA↑Promotes proliferation, migration and invasion(170)
Yang et al, 2020Ovarian cancerLINC00452miR-501-3pROCK1↑Promotes proliferation, migration and invasion(171)
Yang et al, 2021SNHG20miR-148aROCK1↑Promotes proliferation, migration and invasion(172)
Chen et al, 2018TDRG1miR-93RhoC↑Promotes proliferation, migration and invasion(173)
Liu et al, 2017PCA3miR-106bRhoC↑Promotes proliferation, migration and invasion(174)
Liu et al, 2018NEAT1miR-382-3pROCK1↑Promotes metastasis(136)
Li et al, 2022Breast cancerPVT1miR-148a-3pROCK1↑Promotes migration and invasion(175)
Chou et al, 2016MALAT1miR-1Cdc42↑Promotes migration and invasion(176)
Chen et al, 2021Esophageal carcinomaSNHG1miR-195Cdc42↑Promotes proliferation, migration and invasion(177)
Wang et al, 2020LIN00080miR-498Cdc42↑Promotes proliferation and invasion(178)
Zong et al, 2020Gastric cancerCTC-497E21.4miR-22RhoA↑Promotes proliferation and invasion(179)
Chen et al, 2021DSCR8miR-137Cdc42↑Promotes proliferation and invasion(180)
Liu et al, 2020Pancreatic adenocarcinomaZFAS1miR-3924ROCK2↑Promotes metastasis(134)
Li et al, 2017NORADmiR-125a-3pRhoA↑Promotes EMT(135)
Xu et al, 2017DAPK1miR-182ROCK1/RhoA↓Inhibits proliferation, migration and invasion(133)
Wang et al, 2021LINC00941miR-335-5pROCK1↑Promotes proliferation, migration, invasion and EMT(181)

[i] Cdc42, cell division cycle 42; CEP55, centrosomal protein 55; EMT, epithelial-mesenchymal transformation; LASP1, LIM and SH3 protein 1; lncRNA, long non-coding RNA; miRNA/miR, microRNA; RAB11B, Ras-related protein Rab-11B; RAB22A, Ras-related protein Rab-22A; RAC1, Rac family small GTPase 1; Rho, Rho family GTPases; RHOB, Ras homolog family member B; ROCK, Rho-associated coiled-coil containing protein kinase.

In osteosarcoma, the lncRNA DANCR sponges miR-335-5p and miR-1972, leading to an increase in ROCK1 expression, which promotes metastasis (139). Similarly, Shao et al (140) demonstrated that, in osteosarcoma, lncRNA ZNF281 suppressed invasion by upregulating miR-144 expression, thus suppressing ROCK1 expression. Furthermore, in osteosarcoma, lncRNA SNHG1 is overexpressed and promotes cell metastasis by altering apoptosis and the cell cycle; functional experiments have shown that SNHG1 regulated ROCK1 expression by sponging miRNA-101-3p (141). Additionally, Wang et al (142) reported that SNHG5 functioned as a ceRNA for miR-26a and activated the ROCK signaling pathway through the miR-26a/ROCK1 axis to promote osteosarcoma cell malignancy. In HCC, lncRNA CDKN2BAS is upregulated in metastatic tissues and sponges miR-153-5p to increase the expression of Rho GTPase activating protein 18 (ARHGAP18), thus enhancing cell migration (143). Furthermore, lncRNA LINC00607 promotes HCC cell proliferation, migration and invasion through the miR-584-3p/ROCK1 axis (144). Thus, cytoplasmic lncRNAs regulate tumor metastasis by forming ceRNA networks with miRNAs that target Rho/ROCK signaling components.

lncRNAs regulate tumor metastasis by directly binding proteins

In addition to sponging miRNAs by acting as ceRNAs, lncRNAs can also directly bind to specific proteins, including those involved in the Rho/ROCK signaling pathway, and regulate tumor metastasis (182) (Fig. 2). In bladder cancer, lncRNA lnc00892 is downregulated and is positively associated with the prognosis of patients with bladder cancer. Mechanistically, lnc00892 binds to c-JUN, a component of the activating protein-1 transcriptional complex, and reduces nucleolin transcription and affects the stability of RhoA/RhoC mRNAs, which suppresses bladder tumor metastasis (183). In NSCLC, lncRNA NORAD inhibits metastasis by binding to and interfering with CXC chemokine receptor 4, which in turn suppresses the RhoA/ROCK signaling pathway (184). Similarly, in bladder cancer, overexpressed lncRNA KTN1-AS1 recruits EP300, leading to histone H3 lysine 27 acetylation at the KTN1 promoter region. This epigenetic regulation ultimately affects bladder cancer growth and metastasis by mediating the Rho GTPase signaling pathway through Rac1, RhoA and Cdc42 (185). In HCC, overexpression of lncRNA SchLAH (also referred to as BC035072) inhibits distant metastasis. Functional experiments have shown that SchLAH interacts with DNA/RNA-binding protein-fusion sarcoma, regulating the mRNA expression of downstream effector molecules RhoA and Rac1 (186). Dai et al (187) used mass spectrometry and reported that a total of 93 upregulated and 352 downregulated lncRNAs were identified in patients with bladder cancer; downregulated lncRNA MUC20-9 bound directly to ROCK1 and inhibited tumor growth, migration and invasion. MUC20-9 was considered to be a potential therapeutic target for bladder cancer. In OC, lncRNA ABHD11-AS1 is upregulated and facilitates metastasis by directly binding to RhoC (188). Similarly, Wu et al (189) identified a novel lncRNA, MER52A, which was highly expressed in HCC tissues. Mechanistically, MER52A promoted HCC cell invasion and metastasis by stabilizing P120-catenin, thus activating Rac1 and Cdc42, key downstream Rho-GTPases. Furthermore, high MER52A expression was strongly associated with advanced TNM stage, poor differentiation and reduced overall survival of patients with HCC. In osteosarcoma, AFAP1-AS1 is also highly expressed and directly interacts with RhoC and activates the RhoC/ROCK1/p38MAPK signaling pathway, which in turn exerts oncogenic effects (190).

lncRNAs promote tumor invasion and
metastasis by regulating the Rho/ROCK signaling pathway. (A) lncRNA
lnc00892 inhibits metastasis of bladder cancer by binding to c-Jun,
a protein in the activator protein-1 transcription complex, which
reduces transcription of the nucleolin gene and the stability of
RhoA/RhoC mRNA in the Rho/ROCK signaling pathway. (B) lncRNA NORAD
inhibits the RhoA/ROCK signaling pathway by interfering with CXCR4,
which inhibits the proliferation, invasion and migration of
non-small cell lung cancer cells. (C) lncRNA ABHD11-AS1 binds to
RhoC and promotes the proliferation, invasion and migration of
ovarian cancer cells. (D) lncRNA MER52A promotes the invasion and
metastasis of hepatocellular carcinoma cells by stabilizing
P120-catenin and activating the downstream target molecules Rac1
and Cdc42. Cdc42, cell division cycle 42; CXCR, CXC chemokine
receptor; LIMK, LIM domain kinase; lncRNA/lnc, long non-coding RNA;
NCL, nucleolin; Rac, Ras-related C3 botulinum toxin substrate; Rho,
Rho family GTPases; ROCK, Rho-associated coiled-coil containing
protein kinase.

Figure 2

lncRNAs promote tumor invasion and metastasis by regulating the Rho/ROCK signaling pathway. (A) lncRNA lnc00892 inhibits metastasis of bladder cancer by binding to c-Jun, a protein in the activator protein-1 transcription complex, which reduces transcription of the nucleolin gene and the stability of RhoA/RhoC mRNA in the Rho/ROCK signaling pathway. (B) lncRNA NORAD inhibits the RhoA/ROCK signaling pathway by interfering with CXCR4, which inhibits the proliferation, invasion and migration of non-small cell lung cancer cells. (C) lncRNA ABHD11-AS1 binds to RhoC and promotes the proliferation, invasion and migration of ovarian cancer cells. (D) lncRNA MER52A promotes the invasion and metastasis of hepatocellular carcinoma cells by stabilizing P120-catenin and activating the downstream target molecules Rac1 and Cdc42. Cdc42, cell division cycle 42; CXCR, CXC chemokine receptor; LIMK, LIM domain kinase; lncRNA/lnc, long non-coding RNA; NCL, nucleolin; Rac, Ras-related C3 botulinum toxin substrate; Rho, Rho family GTPases; ROCK, Rho-associated coiled-coil containing protein kinase.

Rho/ROCK signaling pathway-associated lncRNAs as biomarkers and therapeutic targets in tumor metastasis

lncRNAs can serve as novel biomarkers for tumor metastasis

Tumor metastasis remains a major contributor to treatment failure and cancer-related mortality (191). Numerous patients with cancer are diagnosed at intermediate or advanced stages, when clinical symptoms appear; consequently, their prognosis is poorer than that of patients diagnosed earlier (192). Epidemiological data indicate that ~90% of cancer-related deaths are due to tumor metastasis (193). Therefore, identifying novel and reliable biomarkers and therapeutic targets associated with the metastatic potential is critical to improve early detection and intervention strategies.

Rho/ROCK signaling pathway-associated lncRNAs have increasingly been explored and defined. These lncRNAs not only serve an important role in tumor metastasis but also have a close relationship with the clinical characteristics of patients with tumors (133,135,136,140,145,151,153,159,165, 166, 171,173,179,180,188,189,194-204) (Table II). A previous study has demonstrated that lncRNA AFAP1-AS1 was upregulated in patients with HCC, and was associated with pathological stage and lymphovascular interstitial infiltration (194). Multifactorial analysis revealed that AFAP1-AS1 was an independent predictor for overall survival, supporting the hypothesis that AFAP1-AS1 is a potential therapeutic target for HCC. Similarly, lncRNA LOC284454, upregulated in nasopharyngeal carcinoma (NPC), promotes migration and invasion by regulating the cytoskeleton-related Rho/Rac signaling pathway, and is strongly associated with poor prognosis in patients with NPC (195). Hu et al (196) demonstrated that ARHGAP42, another pro-metastatic lncRNA in NPC, was associated with shorter metastasis-free survival. In vitro experiments demonstrated that ARHGAP42 promoted the metastasis and invasion of NPC cells, suggesting that it may serve as a tumor migration marker, a prognostic factor or a therapeutic target for patients with NPC. In gliomas, lncRNA LINC00346 is upregulated and is strongly associated with both disease-free and overall survival, suggesting its usage as a potential therapeutic target and therapeutic candidate (159). lncRNA MAGI2-AS3 expression is downregulated in serum samples from patients with HCC with distant metastases compared with in those from non-metastatic patients. Overexpression of MAGI2-AS3 suppresses cell invasion and migration by modulating ROCK2, while its downregulation is associated with distant recurrence following surgical resection (197). In papillary thyroid carcinoma (PTC), lncRNA DLEU1 expression is elevated and associated with lymph node metastasis and clinical stage (145). Follow-up bioinformatics and dual luciferase reporter gene analysis revealed that DLEU1 affects PTC proliferation, survival and metastasis by regulating miR-421 binding to the 3' untranslated region of ROCK1 in TPC-1 cells. Furthermore, lncRNA CDKN2B-AS1 expression is elevated in laryngeal squamous cell carcinoma (LSCC) tissues, and its high expression is positively associated with overall survival, lymph node metastasis and the clinical stage of patients with LSCC (198).

Table II

Rho family GTPases/Rho-associated coiled-coil containing protein kinase signaling pathway-associated lncRNAs as novel tumor biomarkers.

Table II

Rho family GTPases/Rho-associated coiled-coil containing protein kinase signaling pathway-associated lncRNAs as novel tumor biomarkers.

Authors, yearTypes of tumorlncRNAsExpressionClinicopathological features(Refs.)
Li et al, 2020Papillary thyroid carcinomaDLEU1↑TNM stage and lymph node metastasis(145)
Fan et al, 2019Nasopharyngeal carcinomaLOC284454↑TNM stages and cancer metastasis(195)
Hu et al, 2018ARHGAP42↑Cancer metastasis and metastasis-free survival(196)
Xiao et al, 2019Lung adenocarcinomaMALAT1↑TNM stage, tumor size and lymphatic metastasis(151)
Ma et al, 2018LINC00707↑TNM stage, tumor size and lymphatic metastasis(199)
Fang et al, 2020Hepatocellular carcinomaMAGI2-AS↓Tumor size, differentiation and cancer metastasis(197)
Zhang et al, 2016AFAP1-AS1↑Pathological stage and lymphovascular invasion(194)
Wang et al, 2021LINC00491↑TNM stage and lymph node metastasis(153)
Wu et al, 2020lncMER52A↑TNM stage, differentiation and overall prognosis(189)
Song et al, 2020Cervical cancerOIP5-AS1↑Tumor size, differentiation, clinical stage and lymph node metastasis(200)
Shao et al, 2020OsteosarcomaZNF281↑Clinical stage(140)
Wang et al, 2017TUG1↑TNM stage and distant metastasis(165)
Cui et al, 2017HOXA11-AS↑Clinical stage, distant metastasis and overall prognosis(166)
Liu et al, 2020Laryngeal squamous cell cancerCDKN2B-AS1↑Overall prognosis, clinical stage and lymph node metastasis(198)
Chen et al, 2020GliomaLINC00346↑Disease-free survival and overall prognosis(159)
Pan et al, 2019Ovarian cancerLINC00339↑FIGO stage and differentiation(201)
Chen et al, 2018PCGEM1↑ Differentiation(202)
Chen et al, 2018TDRG1↑ Differentiation(173)
Wu et al, 2017ABHD11-AS 1↑Tumor stage(188)
Liu et al, 2018NEAT1↑FIGO stage, tumor size, peritoneal metastasis and differentiation(136)
Yang et al, 2020LINC00452↑Recurrence-free survival(171)
Yuan et al, 2020Bladder cancerEGFR-AS1↑Overall prognosis(203)
Shang et al, 2018RetinoblastomaBDNF-AS↓Clinical stage and differentiation(204)
Zong et al, 2020Gastric cancerCTC-497E21.4↑T stage, lymph node metastasis and perineural invasion(179)
Chen et al, 2021DSCR8↑Tumor size, cancer metastasis and tumor stage(180)
Li et al, 2017Pancreatic cancerNORAD↑Overall prognosis(135)
Xu et al, 2017DAPK1↓Pathological stage and lymph node metastasis(133)

[i] When the lncRNA was upregulated in the tumor tissue, high expression of the lncRNA was positively associated with adverse clinicopathological features in patients with cancer. When the lncRNA was downregulated in the tumor tissue, low expression of the lncRNA was positively associated with adverse clinicopathological features in patients with cancer. FIGO, International Federation of Gynecology and Obstetrics; lncRNA, long non-coding RNA.

Rho/ROCK signaling pathway-associated lncRNAs may also serve as liquid biopsy biomarkers. Liquid biopsy, an innovative technology, has opened novel pathways for the early diagnosis and prognostic evaluation of cancer. Compared with traditional tissue biopsies, liquid biopsy enables real-time monitoring of disease progression and therapeutic responses (205). Liquid biopsy platforms can simultaneously detect multiple circulating biomarkers, including circulating tumor cells (CTCs), circulating tumor DNA, exosomes, tumor-educated platelets and circulating free RNA. Among these, exosomes, nanosized extracellular vesicles (30-150 nm), serve crucial roles in intercellular communication and carry active molecules such as proteins and nucleic acids (DNA and RNA, including lncRNAs) (206,207). Previous studies have shown that several lncRNAs can be encapsulated in exosomes and transferred from donor cells to recipient cells, where they regulate components of the Rho/ROCK signaling pathway, thereby regulating cancer progression and metastasis (154,208,209). For example, You et al (154) demonstrated that exosome-derived lncRNA LINC00161 sponged miR-590-3p to promote HCC metastasis by activating the ROCK signaling pathway, suggesting that LINC00161 may serve as a novel prognostic marker for HCC. Furthermore, Ding et al (209) reported that CRC cell-derived exosomal lncRNA BANCR promoted M2 macrophage polarization and enhanced CRC cell proliferation and invasion via the RhoA/ROCK pathway, with insulin like growth factor 2 mRNA binding protein 2 acting as a mediator. Together, these findings suggest that Rho/ROCK pathway-associated lncRNAs, particularly those in exosomes, are emerging as valuable molecular biomarkers with potential applications in liquid biopsy-based diagnostics.

lncRNAs can serve as potential therapeutic targets for tumor metastasis

In addition to their diagnostic utility, Rho/ROCK signaling pathway-associated lncRNAs represent potential therapeutic targets for metastatic cancer (154,210) (Fig. 3). As shown in Fig. 3, You et al (154) established a xenograft model in which tail-vein inoculation of HCC cells produced widespread metastases; notably, tail-vein injection of HCC cells with stable LINC00161 knockdown markedly attenuated both tumor initiation and metastatic dissemination in vivo, indicating that LINC00161 silencing suppressed HCC tumorigenesis and metastasis. Horita et al (210) reported that lncRNA UCA1 is upregulated in OC and enhances oncolytic poxvirus dissemination by modulating the Cdc42 signaling pathway, a key regulator of cytoskeletal reorganization (210). Similarly, UCA1 sponges miR-18a/miR-182 to regulate Cdc42/filopodia, thereby increasing sensitivity to poxvirus-based virotherapy in CRC (169). Clinically, glucocorticoids, such as dexamethasone and prednisolone, can be used in combination with other chemotherapeutic agents for the treatment of hematologic malignancies. In acute myeloid leukemia, lncRNA HOTAIRM1 promotes glucocorticoid resistance by binding to the transcriptional repressor region of ARHGAP18, which in turn activates the RhoA/ROCK1 signaling pathway. This finding has implications for the optimization of glucocorticoid-based leukemia treatment strategies (211). In oral squamous cell cancer, low expression levels of lncRNA LOC441178 are associated with longer postoperative survival, suggesting a potential role as a tumor suppressor and prognostic marker (212). In osteosarcoma, lncRNA CCHE1 expression is higher in patients with distant recurrence after surgery and associated with ROCK1 expression, suggesting that CCHE1 may help guide subsequent chemotherapy or radiotherapy (213). Rho-GTPases and their downstream signaling molecules have been shown to serve a key role in regulating tumor angiogenesis, invasion, metastasis and EMT (214-216). Targeting EMT is considered a relatively promising strategy to inhibit metastasis and improve the survival rate of patients with cancer (191). lncRNA AFAP1-AS1 expression is upregulated in osteosarcoma, and is involved in the growth and metastasis of osteosarcoma by interacting with RhoC and regulating EMT. Therefore, AFAP1-AS1 inhibitors may serve as therapeutic agents in osteosarcoma (190). Furthermore, targeting lncRNAs involved in drug resistance has also shown promise. In cholangiocarcinoma, lncRNA CCAT1 modulates EMT via ROCK2. CCAT1 knockdown increased the susceptibility of an erlotinib-resistant cholangiocarcinoma cell line xenograft to erlotinib in vivo, suggesting that targeting the miR-181a-5p/ROCK2 signaling axis could overcome resistance (217). Using a small-molecule microarray, Abulwerdi et al (218) identified several chemotypes that bind the MALAT1 element for nuclear expression triplex-binding chemotypes (referred to as MALAT1-IN-1). Notably, compounds 5 and 16 lowered MALAT1 RNA levels and suppressed branching morphogenesis in mammary tumor organoids, establishing triplex-targeting scaffolds as preclinical leads for anticancer therapeutics and molecular probes.

Effect of long non-coding RNAs on
tumor metastasis in vivo. (A) A mouse xenograft model was
established. HCC cells with stable expression of shLINC00161 were
injected into the tail vein, and tumor growth and metastasis in
mice were observed (154). The
study aimed to demonstrate the effect of lncRNAs on HCC metastasis
in an in vivo model. (B) KFTX OC cells with high expression
of UCA1 were intraperitoneally injected into mice, then
OVV-VGFΔ/O1LΔ was intraperitoneally injected into mice, and the
tumor growth was observed (210). The study aimed to demonstrate
the effect of lncRNAs on OC metastasis in an in vivo model.
Cdc42, cell division cycle 42; HCC, hepatocellular carcinoma;
lncRNA, long non-coding RNA; OC, ovarian cancer; OVV-VGFΔ/O1LΔ,
oncolytic vaccinia virus-VGF deleted/O1L deletion; PTX, paclitaxel;
sh, short hairpin RNA; UCA1, urothelial carcinoma-associated 1.

Figure 3

Effect of long non-coding RNAs on tumor metastasis in vivo. (A) A mouse xenograft model was established. HCC cells with stable expression of shLINC00161 were injected into the tail vein, and tumor growth and metastasis in mice were observed (154). The study aimed to demonstrate the effect of lncRNAs on HCC metastasis in an in vivo model. (B) KFTX OC cells with high expression of UCA1 were intraperitoneally injected into mice, then OVV-VGFΔ/O1LΔ was intraperitoneally injected into mice, and the tumor growth was observed (210). The study aimed to demonstrate the effect of lncRNAs on OC metastasis in an in vivo model. Cdc42, cell division cycle 42; HCC, hepatocellular carcinoma; lncRNA, long non-coding RNA; OC, ovarian cancer; OVV-VGFΔ/O1LΔ, oncolytic vaccinia virus-VGF deleted/O1L deletion; PTX, paclitaxel; sh, short hairpin RNA; UCA1, urothelial carcinoma-associated 1.

Inhibitors targeting the Rho/ROCK signaling pathway have gained attention in drug research for tumor-targeted therapy (91). These inhibitors of the Rho/ROCK pathway are divided into three categories: Inhibitors of ROCK, geranylgeranyltransferase-1 and 3-hydroxy-3-methyl-glutaryl-CoA reductase (219-222). MBQ-167, a newly developed Rac and Cdc42 inhibitor, has been shown to inhibit p21 activated protein kinase signaling, metastatic cell migration and mammosphere growth in TNBC. Its short half-life and low toxicity make this inhibitor a promising candidate for future TNBC therapy (219). NecroX-5 has demonstrated anti-metastatic effects in lung cancer, breast cancer and melanoma models by suppressing the expression of Cdc42, Rac1 and RhoA (220). NSC23766, the first Rac1-specific inhibitor, blocks Rac1 activation by targeting the guanine nucleotide exchange factors. NSC23766 inhibits the invasion and migration of human HCC by interfering with the Rac1/JNK or LIM and cysteine-rich domains 1-Rac1 pathways (221). Inhibition of Rac1 by NSC23766 affects the proliferation and migration of NSCLC (222). Overall, Rho/ROCK pathway inhibitors represent a promising strategy to overcome drug resistance and prevent tumor metastasis. Therefore, the development of Rho/ROCK pathway inhibitors remains a key clinical strategy in cancer therapy.

Conclusions and future perspectives

Tumorigenesis is a complex process that involves multiple malignant phenotypes, among which metastasis is a leading contributor to cancer-related mortality. Tumor metastasis involves a series of complex pathophysiological changes, with cytoskeletal reorganization serving a critical role in cancer cell invasion and migration. Among the classical signaling pathways implicated in cytoskeletal reorganization, the Rho/ROCK signaling pathway has attracted considerable attention. Accumulating evidence has also demonstrated that lncRNAs regulate tumorigenic processes, including proliferation, migration, metastasis and resistance to radiotherapy (198,223). With the development of high-throughput sequencing and related technologies, an increasing number of lncRNAs have been identified that interact, either directly or indirectly, with key cytoskeletal regulators such as ROCK1 and ROCK2, mediating tumor metastasis by altering the three-dimensional structure of cancer cells and regulating the reconstruction of the cytoskeleton. As research continues to uncover the specific mechanisms and clinical implications of Rho/ROCK signaling pathway-associated lncRNAs, several promising applications have emerged. First, Rho/ROCK signaling pathway-associated lncRNAs may serve as pathological and liquid biopsy markers for patients with tumors. Unlike protein-based pathological tissue biomarkers, which often reflect downstream changes, the aberrant expression of lncRNAs often occurs earlier than changes at the protein level, providing greater tissue specificity and expression sensitivity for early cancer diagnosis (224,225). Their tissue specificity, dynamic expression and distinct subcellular localization make them particularly suitable for precise classification and prognosis (226). While tissue biopsy remains the gold standard for diagnosing tumor subtypes and grades, its invasiveness limits its applications for dynamic monitoring of disease progression. Liquid biopsy, on the other hand, offers a minimally invasive monitoring strategy by analyzing circulating tumor components in bodily fluids such as blood and urine (227). In addition, circulating lncRNAs exhibit high stability and resistance to nuclease degradation, offering advantages over other biomarkers such as cell-free DNA (including circulating tumor DNA), CTCs and exosomal RNA (228). Several lncRNAs, including those detectable in liver, colorectal, gastric and prostate cancer, have already shown promise as reliable diagnostic and prognostic markers (229-231). Second, next-generation sequencing, combined with artificial intelligence (AI) and machine learning, has accelerated the collaborative development of basic cancer research and clinical oncology (232-234). Tumor genomic databases constructed from high-throughput sequencing systematically analyze disease-specific expression profiles and tumor microenvironment characteristics. Researchers can leverage advanced machine learning algorithms to deeply mine sequencing data, successfully identifying functional lncRNAs that can serve as biomarkers for personalized treatment decisions or prognostic predictions. Deep learning algorithms enable the precise identification of Rho/ROCK signaling pathway-associated lncRNAs, as well as the prediction of their interacting proteins and binding miRNAs, providing a novel strategy for the development of cancer-specific diagnostic and prognostic biomarkers. Third, despite the potential of RNA-based therapeutics, clinical translation is challenged by issues such as target specificity, low delivery efficiency and in vivo stability (235). Notably, while a few miRNA drugs have entered phase II/III clinical trials, the development of inhibitors targeting lncRNAs remains in the preclinical stage (236-238). However, nanotechnology provides a promising strategy to overcome these limitations. Nanomaterials can enable precise targeting at the tissue and even subcellular levels, delivering complex therapeutic molecules to key metastatic sites (such as lymph nodes, liver and lungs) and specific subcellular regions (239). Advances in translational medicine have focused on co-delivering lncRNA inhibitors with small molecule drugs or immune checkpoint inhibitors via nanoparticle systems, synergistically enhancing antitumor efficacy (240-243). Based on this research, the future development of nanoparticle carriers encapsulating Rho/ROCK signaling pathway-associated lncRNA inhibitors may improve the prognosis of patients with metastatic cancer.

Nevertheless, existing research on Rho/ROCK signaling pathway-associated lncRNAs still has several limitations. First, although high-throughput studies have identified numerous dysregulated lncRNAs in tumor tissues (244,245), the specific molecular mechanisms by which Rho/ROCK signaling pathway-associated lncRNAs influence metastasis require further elucidation. Second, Rho/ROCK signaling pathway-associated lncRNAs may regulate gene expression through multiple miRNA sponging interactions or binding proteins, but the specific target axis may not be limited to one or two axes. Third, some lncRNAs implicated in the Rho/ROCK signaling pathway may also regulate other signaling cascades. In addition, the Rho/ROCK signaling pathway mainly promotes tumor metastasis; however, a study has reported that the Rho/ROCK signaling pathway may induce apoptosis (246). Street et al (247) demonstrated that the ROCK inhibitor Y27632 could enhance the survival of neuroblastoma cells. Therefore, future studies must validate pathway specificity when proposing drug targets. Fourth, whether Rho/ROCK signaling pathway-associated lncRNAs can be used as therapeutic targets still requires further research at present. Existing studies are only at the stage of animal experiments (135,146,150,153,154), and there is still a long way to go from basic research to clinical application. Therefore, the relationship between Rho/ROCK signaling pathway-associated lncRNAs and tumor metastasis needs to be further clarified. Fifth, although the drug delivery systems of nanoparticle-based lncRNA inhibitors show potential (239-241), further clinical validation is needed to ensure their efficacy, safety and long-term effects of the delivery of nucleic acids in humans.

In summary, lncRNAs associated with the Rho/ROCK signaling pathway are emerging as key regulators of cytoskeletal remodeling and tumor metastasis. Through interactions with miRNAs, proteins and exosomal transport mechanisms, these lncRNAs regulate key processes involved in cancer cell migration, invasion and metastasis. They hold promise not only as novel biomarkers for early cancer diagnosis but also as therapeutic targets to prevent metastasis. Advances in high-throughput sequencing, AI-driven bioinformatics and nanomedicine provide novel opportunities to leverage lncRNAs in personalized treatment strategies and improve prognostic predictions. However, further research is needed to fully elucidate the mechanisms by which lncRNAs regulate the Rho/ROCK pathway and to confirm their clinical applicability. The present review highlights the growing potential of Rho/ROCK signaling pathway-associated lncRNAs as promising diagnostic and therapeutic targets, with the potential to improve the management and prognosis of metastatic cancer.

Availability of data and materials

Not applicable.

Authors' contributions

HN and XY performed the literature search. HN and XY prepared the first draft of the manuscript. QC, XH and YH wrote and edited the manuscript. HN and YH drew the figures. QH and CO prepared the tables and were responsible for revising the manuscript. CO obtained funding support. Data authentication is not applicable. All authors reviewed the manuscript, and have read and approved of the final version of the manuscript.

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.

Abbreviations:

Rho

ras homolog family

ROCK

Rho-associated coiled-coil containing protein kinase

Rac

Ras-related C3 botulinum toxin substrate

lncRNA

long non-coding RNA

ceRNA

competing endogenous RNA

EMT

epithelial-mesenchymal transformation

Cdc42

cell division cycle 42

mDia

mammalian homolog to Drosophila

ARHGAP18

Rho GTPase activating protein 18

NSCLC

non-small cell lung cancer

NPC

nasopharyngeal carcinoma

PTC

papillary thyroid carcinoma

TNBC

triple-negative breast cancer

CRC

colorectal cancer

HCC

hepatocellular carcinoma

Rnd

Rho family GTPase

Acknowledgements

Not applicable.

Funding

The present study was supported by the National Natural Science Foundation of China (grant no. 82373062) and the Central South University Innovation-Driven Research Programme (grant no. 2023CXQD075).

References

1 

Valastyan S and Weinberg RA: Tumor metastasis: Molecular insights and evolving paradigms. Cell. 147:275–292. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Han Y, Wang D, Peng L, Huang T, He X, Wang J and Ou C: Single-cell sequencing: A promising approach for uncovering the mechanisms of tumor metastasis. J Hematol Oncol. 15:592022. View Article : Google Scholar : PubMed/NCBI

3 

Zhou H, He X, He Y, Ou C and Cao P: Exosomal circRNAs: Emerging players in tumor metastasis. Front Cell Dev Biol. 9:7862242021. View Article : Google Scholar : PubMed/NCBI

4 

Liao Z, Nie H, Wang Y, Luo J, Zhou J and Ou C: The emerging landscape of long non-coding RNAs in colorectal cancer metastasis. Front Oncol. 11:6413432021. View Article : Google Scholar : PubMed/NCBI

5 

Hanahan D and Weinberg RA: Hallmarks of cancer: The next generation. Cell. 144:646–674. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Li Y, Liu F, Cai Q, Deng L, Ouyang Q, Zhang XH and Zheng J: Invasion and metastasis in cancer: Molecular insights and therapeutic targets. Signal Transduct Target Ther. 10:572025. View Article : Google Scholar : PubMed/NCBI

7 

Francoeur AA, Monk BJ and Tewari KS: Treatment advances across the cervical cancer spectrum. Nat Rev Clin Oncol. 22:182–199. 2025. View Article : Google Scholar : PubMed/NCBI

8 

Kaur R, Bhardwaj A and Gupta S: Cancer treatment therapies: Traditional to modern approaches to combat cancers. Mol Biol Rep. 50:9663–9676. 2023. View Article : Google Scholar : PubMed/NCBI

9 

Klein CA: Cancer progression and the invisible phase of metastatic colonization. Nat Rev Cancer. 20:681–694. 2020. View Article : Google Scholar : PubMed/NCBI

10 

Gerstberger S, Jiang Q and Ganesh K: Metastasis. Cell. 186:1564–1579. 2023. View Article : Google Scholar : PubMed/NCBI

11 

Johan MZ and Samuel MS: Rho-ROCK signaling regulates tumor-microenvironment interactions. Biochem Soc Trans. 47:101–108. 2019. View Article : Google Scholar

12 

Hall A: Rho family GTPases. Biochem Soc Trans. 40:1378–1382. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Etienne-Manneville S and Hall A: Rho GTPases in cell biology. Nature. 420:629–635. 2002. View Article : Google Scholar : PubMed/NCBI

14 

Liu C, Chen S, Zhang Y, Zhou X, Wang H, Wang Q and Lan X: Mechanisms of Rho GTPases in regulating tumor proliferation, migration and invasion. Cytokine Growth Factor Rev. 80:168–174. 2024. View Article : Google Scholar : PubMed/NCBI

15 

Alarcon VB and Marikawa Y: Trophectoderm formation: Regulation of morphogenesis and gene expressions by RHO, ROCK, cell polarity, and HIPPO signaling. Reproduction. 164:R75–R86. 2022. View Article : Google Scholar : PubMed/NCBI

16 

Agrawal A, Scott ZC and Koslover EF: Morphology and transport in eukaryotic cells. Annu Rev Biophys. 51:247–266. 2022. View Article : Google Scholar : PubMed/NCBI

17 

Ando D, Korabel N, Huang KC and Gopinathan A: Cytoskeletal network morphology regulates intracellular transport dynamics. Biophys J. 109:1574–1582. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Fletcher DA and Mullins RD: Cell mechanics and the cytoskeleton. Nature. 463:485–492. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Park J, Wu Y, Kim JS, Byun J, Lee J and Oh YK: Cytoskeleton-modulating nanomaterials and their therapeutic potentials. Adv Drug Deliv Rev. 211:1153622024. View Article : Google Scholar : PubMed/NCBI

20 

Leduc C and Etienne-Manneville S: Intermediate filaments in cell migration and invasion: The unusual suspects. Curr Opin Cell Biol. 32:102–112. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Li M, Peng L, Wang Z, Liu L, Cao M, Cui J, Wu F and Yang J: Roles of the cytoskeleton in human diseases. Mol Biol Rep. 50:2847–2856. 2023. View Article : Google Scholar : PubMed/NCBI

22 

Thapa N, Wen T, Cryns VL and Anderson RA: Regulation of cell adhesion and migration via microtubule cytoskeleton organization, cell polarity, and phosphoinositide signaling. Biomolecules. 13:14302023. View Article : Google Scholar : PubMed/NCBI

23 

Mangaonkar S, Nath S and Chatterji BP: Microtubule dynamics in cancer metastasis: Harnessing the underappreciated potential for therapeutic interventions. Pharmacol Ther. 263:1087262024. View Article : Google Scholar : PubMed/NCBI

24 

Fanale D, Bronte G, Passiglia F, Calò V, Castiglia M, Di Piazza F, Barraco N, Cangemi A, Catarella MT, Insalaco L, et al: Stabilizing versus destabilizing the microtubules: A double-edge sword for an effective cancer treatment option? Anal Cell Pathol (Amst). 2015:6909162015.PubMed/NCBI

25 

Dumontet C, Reichert JM, Senter PD, Lambert JM and Beck A: Antibody-drug conjugates come of age in oncology. Nat Rev Drug Discov. 22:641–661. 2023. View Article : Google Scholar : PubMed/NCBI

26 

Cai J, He M, Wang Y, Zhang H, Xu Y, Wang Y, You C and Gao H: Discovery of a novel microtubule destabilizing agent targeting the colchicine site based on molecular docking. Biochem Pharmacol. 234:1168042025. View Article : Google Scholar : PubMed/NCBI

27 

Castaldo V, Minopoli M, Di Modugno F, Sacconi A, Liguoro D, Frigerio R, Ortolano A, Di Martile M, Gesualdi L, Madonna G, et al: Upregulated expression of miR-4443 and miR-4488 in drug resistant melanomas promotes migratory and invasive phenotypes through downregulation of intermediate filament nestin. J Exp Clin Cancer Res. 42:3172023. View Article : Google Scholar : PubMed/NCBI

28 

Ishiwata T, Matsuda Y and Naito Z: Nestin in gastrointestinal and other cancers: Effects on cells and tumor angiogenesis. World J Gastroenterol. 17:409–418. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Atianand MK, Caffrey DR and Fitzgerald KA: Immunobiology of long noncoding RNAs. Annu Rev Immunol. 35:177–198. 2017. View Article : Google Scholar : PubMed/NCBI

30 

Wang D, Han Y, Peng L, Huang T, He X, Wang J and Ou C: Crosstalk between N6-methyladenosine (m6A) modification and noncoding RNA in tumor microenvironment. Int J Biol Sci. 19:2198–2219. 2023. View Article : Google Scholar : PubMed/NCBI

31 

Ou C, Sun Z, He X, Li X, Fan S, Zheng X, Peng Q, Li G, Li X and Ma J: Targeting YAP1/LINC00152/FSCN1 signaling axis prevents the progression of colorectal cancer. Adv Sci (Weinh). 7:19013802020. View Article : Google Scholar : PubMed/NCBI

32 

He X, Yu B, Kuang G, Wu Y, Zhang M, Cao P and Ou C: Long noncoding RNA DLEU2 affects the proliferative and invasive ability of colorectal cancer cells. J Cancer. 12:428–437. 2021. View Article : Google Scholar : PubMed/NCBI

33 

Chan JJ and Tay Y: Noncoding RNA:RNA regulatory networks in cancer. Int J Mol Sci. 19:13102018. View Article : Google Scholar : PubMed/NCBI

34 

Yan H and Bu P: Non-coding RNA in cancer. Essays Biochem. 65:625–639. 2021. View Article : Google Scholar : PubMed/NCBI

35 

Chen LL and Kim VN: Small and long non-coding RNAs: Past, present, and future. Cell. 187:6451–6485. 2024. View Article : Google Scholar : PubMed/NCBI

36 

Nadhan R, Isidoro C, Song YS and Dhanasekaran DN: Signaling by LncRNAs: Structure, cellular homeostasis, and disease pathology. Cells. 11:25172022. View Article : Google Scholar : PubMed/NCBI

37 

Mohammad F, Mondal T and Kanduri C: Epigenetics of imprinted long noncoding RNAs. Epigenetics. 4:277–286. 2009. View Article : Google Scholar : PubMed/NCBI

38 

Heo JB and Sung S: Vernalization-mediated epigenetic silencing by a long intronic noncoding RNA. Science. 331:76–79. 2011. View Article : Google Scholar

39 

Good MC, Zalatan JG and Lim WA: Scaffold proteins: Hubs for controlling the flow of cellular information. Science. 332:680–686. 2011. View Article : Google Scholar : PubMed/NCBI

40 

Spitale RC, Tsai MC and Chang HY: RNA templating the epigenome: Long noncoding RNAs as molecular scaffolds. Epigenetics. 6:539–543. 2011. View Article : Google Scholar : PubMed/NCBI

41 

Zhao W, Song M, Zhang J, Kuerban M and Wang H: Combined identification of long non-coding RNA CCAT1 and HOTAIR in serum as an effective screening for colorectal carcinoma. Int J Clin Exp Pathol. 8:14131–14140. 2015.

42 

Shao Y, Ye M, Jiang X, Sun W, Ding X, Liu Z, Ye G, Zhang X, Xiao B and Guo J: Gastric juice long noncoding RNA used as a tumor marker for screening gastric cancer. Cancer. 120:3320–3328. 2014. View Article : Google Scholar : PubMed/NCBI

43 

Li J, Wang X, Tang J, Jiang R, Zhang W, Ji J and Sun B: HULC and Linc00152 act as novel biomarkers in predicting diagnosis of hepatocellular carcinoma. Cell Physiol Biochem. 37:687–696. 2015. View Article : Google Scholar : PubMed/NCBI

44 

Tong YS, Wang XW, Zhou XL, Liu ZH, Yang TX, Shi WH, Xie HW, Lv J, Wu QQ and Cao XF: Identification of the long non-coding RNA POU3F3 in plasma as a novel biomarker for diagnosis of esophageal squamous cell carcinoma. Mol Cancer. 14:32015. View Article : Google Scholar : PubMed/NCBI

45 

Jiang YZ, Liu YR, Xu XE, Jin X, Hu X, Yu KD and Shao ZM: Transcriptome analysis of triple-negative breast cancer reveals an integrated mRNA-lncRNA signature with predictive and prognostic value. Cancer Res. 76:2105–2114. 2016. View Article : Google Scholar : PubMed/NCBI

46 

Shen L, Chen L, Wang Y, Jiang X, Xia H and Zhuang Z: Long noncoding RNA MALAT1 promotes brain metastasis by inducing epithelial-mesenchymal transition in lung cancer. J Neurooncol. 121:101–108. 2015. View Article : Google Scholar

47 

Yu W, Ma Y, Hou W, Wang F, Cheng W, Qiu F, Wu P and Zhang G: Identification of immune-related lncRNA prognostic signature and molecular subtypes for glioblastoma. Front Immunol. 12:7069362021. View Article : Google Scholar : PubMed/NCBI

48 

Kong X, Qi J, Yan Y, Chen L, Zhao Y, Fang Z, Fan J, Liu M and Liu Y: Comprehensive analysis of differentially expressed profiles of lncRNAs, mRNAs, and miRNAs in laryngeal squamous cell carcinoma in order to construct a ceRNA network and identify potential biomarkers. J Cell Biochem. 120:17963–17974. 2019. View Article : Google Scholar : PubMed/NCBI

49 

Jing Z, Guo S, Zhang P and Liang Z: LncRNA-Associated ceRNA network reveals novel potential biomarkers of laryngeal squamous cell carcinoma. Technol Cancer Res Treat. 19:15330338209857872020. View Article : Google Scholar : PubMed/NCBI

50 

Zhang H, Zhang N, Wu W, Zhou R, Li S, Wang Z, Dai Z, Zhang L, Liu Z, Zhang J, et al: Machine learning-based tumor-infiltrating immune cell-associated lncRNAs for predicting prognosis and immunotherapy response in patients with glioblastoma. Brief Bioinform. 23:bbac3862022. View Article : Google Scholar : PubMed/NCBI

51 

Nemeth K, Bayraktar R, Ferracin M and Calin GA: Non-coding RNAs in disease: From mechanisms to therapeutics. Nat Rev Genet. 25:211–232. 2024. View Article : Google Scholar

52 

Fatica A and Bozzoni I: Long non-coding RNAs: New players in cell differentiation and development. Nat Rev Genet. 15:7–21. 2014. View Article : Google Scholar

53 

Katayama S, Tomaru Y, Kasukawa T, Waki K, Nakanishi M, Nakamura M, Nishida H, Yap CC, Suzuki M, Kawai J, et al: Antisense transcription in the mammalian transcriptome. Science. 309:1564–1566. 2005. View Article : Google Scholar : PubMed/NCBI

54 

Bunch H, Lawney BP, Burkholder A, Ma D, Zheng X, Motola S, Fargo DC, Levine SS, Wang YE and Hu G: RNA polymerase II promoter-proximal pausing in mammalian long non-coding genes. Genomics. 108:64–77. 2016. View Article : Google Scholar : PubMed/NCBI

55 

Nojima T and Proudfoot NJ: Mechanisms of lncRNA biogenesis as revealed by nascent transcriptomics. Nat Rev Mol Cell Biol. 23:389–406. 2022. View Article : Google Scholar : PubMed/NCBI

56 

Ulitsky I and Bartel DP: lincRNAs: Genomics, evolution, and mechanisms. Cell. 154:26–46. 2013. View Article : Google Scholar : PubMed/NCBI

57 

Collins FS, Morgan M and Patrinos A: The human genome project: Lessons from large-scale biology. Science. 300:286–290. 2003. View Article : Google Scholar : PubMed/NCBI

58 

Green ED, Watson JD and Collins FS: Human genome project: Twenty-five years of big biology. Nature. 526:29–31. 2015. View Article : Google Scholar : PubMed/NCBI

59 

Hombach S and Kretz M: Non-coding RNAs: Classification, biology and functioning. Adv Exp Med Biol. 937:3–17. 2016. View Article : Google Scholar : PubMed/NCBI

60 

Koffler-Brill T, Noy Y and Avraham KB: The long and short: Non-coding RNAs in the mammalian inner ear. Hear Res. 428:1086662023. View Article : Google Scholar :

61 

Zhao J, Sun BK, Erwin JA, Song JJ and Lee JT: Polycomb proteins targeted by a short repeat RNA to the mouse X chromosome. Science. 322:750–756. 2008. View Article : Google Scholar : PubMed/NCBI

62 

Yang F, Deng X, Ma W, Berletch JB, Rabaia N, Wei G, Moore JM, Filippova GN, Xu J, Liu Y, et al: The lncRNA Firre anchors the inactive X chromosome to the nucleolus by binding CTCF and maintains H3K27me3 methylation. Genome Biol. 16:522015. View Article : Google Scholar : PubMed/NCBI

63 

Martianov I, Ramadass A, Barros AS, Chow N and Akoulitchev A: Repression of the human dihydrofolate reductase gene by a non-coding interfering transcript. Nature. 445:666–670. 2007. View Article : Google Scholar : PubMed/NCBI

64 

Bailey C, Pich O, Thol K, Watkins TBK, Luebeck J, Rowan A, Stavrou G, Weiser NE, Dameracharla B, Bentham R, et al: Origins and impact of extrachromosomal DNA. Nature. 635:193–200. 2024. View Article : Google Scholar : PubMed/NCBI

65 

Lopez-Pajares V, Qu K, Zhang J, Webster DE, Barajas BC, Siprashvili Z, Zarnegar BJ, Boxer LD, Rios EJ, Tao S, et al: A LncRNA-MAF:MAFB transcription factor network regulates epidermal differentiation. Dev Cell. 32:693–706. 2015. View Article : Google Scholar : PubMed/NCBI

66 

Cesana M, Cacchiarelli D, Legnini I, Santini T, Sthandier O, Chinappi M, Tramontano A and Bozzoni I: A long noncoding RNA controls muscle differentiation by functioning as a competing endogenous RNA. Cell. 147:358–369. 2011. View Article : Google Scholar : PubMed/NCBI

67 

Wang Y, Xu Z, Jiang J, Xu C, Kang J, Xiao L, Wu M, Xiong J, Guo X and Liu H: Endogenous miRNA sponge lincRNA-RoR regulates Oct4, Nanog, and Sox2 in human embryonic stem cell self-renewal. Dev Cell. 25:69–80. 2013. View Article : Google Scholar : PubMed/NCBI

68 

Li X, Wu Z, Fu X and Han W: lncRNAs: Insights into their function and mechanics in underlying disorders. Mutat Res Rev Mutat Res. 762:1–21. 2014. View Article : Google Scholar : PubMed/NCBI

69 

Rapicavoli NA, Qu K, Zhang J, Mikhail M, Laberge RM and Chang HY: A mammalian pseudogene lncRNA at the interface of inflammation and anti-inflammatory therapeutics. Elife. 2:e007622013. View Article : Google Scholar : PubMed/NCBI

70 

Hung T, Wang Y, Lin MF, Koegel AK, Kotake Y, Grant GD, Horlings HM, Shah N, Umbricht C, Wang P, et al: Extensive and coordinated transcription of noncoding RNAs within cell-cycle promoters. Nat Genet. 43:621–629. 2011. View Article : Google Scholar : PubMed/NCBI

71 

Loewer S, Cabili MN, Guttman M, Loh YH, Thomas K, Park IH, Garber M, Curran M, Onder T, Agarwal S, et al: Large intergenic non-coding RNA-RoR modulates reprogramming of human induced pluripotent stem cells. Nat Genet. 42:1113–1117. 2010. View Article : Google Scholar : PubMed/NCBI

72 

Jiang M, Zhang S, Yang Z, Lin H, Zhu J, Liu L, Wang W, Liu S, Liu W, Ma Y, et al: Self-Recognition of an inducible host lncRNA by RIG-I feedback restricts innate immune response. Cell. 173:906–919. 2018. View Article : Google Scholar : PubMed/NCBI

73 

Ferrer J and Dimitrova N: Transcription regulation by long non-coding RNAs: Mechanisms and disease relevance. Nat Rev Mol Cell Biol. 25:396–415. 2024. View Article : Google Scholar : PubMed/NCBI

74 

Kallen AN, Zhou XB, Xu J, Qiao C, Ma J, Yan L, Lu L, Liu C, Yi JS, Zhang H, et al: The imprinted H19 lncRNA antagonizes let-7 microRNAs. Mol Cell. 52:101–112. 2013. View Article : Google Scholar : PubMed/NCBI

75 

Yang L, Froberg JE and Lee JT: Long noncoding RNAs: Fresh perspectives into the RNA world. Trends Biochem Sci. 39:35–43. 2014. View Article : Google Scholar :

76 

Wu Y, Zhang L, Zhang L, Wang Y, Li H, Ren X, Wei F, Yu W, Liu T, Wang X, et al: Long non-coding RNA HOTAIR promotes tumor cell invasion and metastasis by recruiting EZH2 and repressing E-cadherin in oral squamous cell carcinoma. Int J Oncol. 46:2586–2594. 2015. View Article : Google Scholar : PubMed/NCBI

77 

Bonasio R, Tu S and Reinberg D: Molecular signals of epigenetic states. Science. 330:612–616. 2010. View Article : Google Scholar : PubMed/NCBI

78 

Ingolia NT, Lareau LF and Weissman JS: Ribosome profiling of mouse embryonic stem cells reveals the complexity and dynamics of mammalian proteomes. Cell. 147:789–802. 2011. View Article : Google Scholar : PubMed/NCBI

79 

Zhao S, Meng J and Luan Y: LncRNA-Encoded short peptides identification using feature subset recombination and ensemble learning. Interdiscip Sci. 14:101–112. 2022. View Article : Google Scholar

80 

Zhao S, Meng J, Kang Q and Luan Y: Identifying LncRNA-encoded short peptides using optimized hybrid features and ensemble learning. IEEE/ACM Trans Comput Biol Bioinform. 19:2873–2881. 2022. View Article : Google Scholar

81 

Jiang W, Zhang X, Xu Z, Cheng Q, Li X, Zhu Y, Lu F, Dong L, Zeng L, Zhong W, et al: High-throughput single-nucleus RNA profiling of minimal puncture FFPE samples reveals spatiotemporal heterogeneity of cancer. Adv Sci (Weinh). 12:e24107132025. View Article : Google Scholar

82 

Tsai MC, Manor O, Wan Y, Mosammaparast N, Wang JK, Lan F, Shi Y, Segal E and Chang H: Long noncoding RNA as modular scaffold of histone modification complexes. Science. 329:689–693. 2010. View Article : Google Scholar : PubMed/NCBI

83 

Gupta RA, Shah N, Wang KC, Kim J, Horlings HM, Wong DJ, Tsai MC, Hung T, Argani P, Rinn JL, et al: Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature. 464:1071–1076. 2010. View Article : Google Scholar : PubMed/NCBI

84 

Wang BR, Chu DX, Cheng MY, Jin Y, Luo HG and Li N: Progress of HOTAIR-microRNA in hepatocellular carcinoma. Hered Cancer Clin Pract. 20:42022. View Article : Google Scholar : PubMed/NCBI

85 

Zhang J, Luo Q, Li X, Guo J, Zhu Q, Lu X, Wei L, Xiang Z, Peng M, Ou C and Zou Y: Novel role of immune-related non-coding RNAs as potential biomarkers regulating tumour immunoresponse via MICA/NKG2D pathway. Biomark Res. 11:862023. View Article : Google Scholar : PubMed/NCBI

86 

Zhou M, He X, Mei C and Ou C: Exosome derived from tumor-associated macrophages: Biogenesis, functions, and therapeutic implications in human cancers. Biomark Res. 11:1002023. View Article : Google Scholar : PubMed/NCBI

87 

Zhang R, Xia Y, Wang Z, Zheng J, Chen Y, Li X, Wang Y and Ming H: Serum long non coding RNA MALAT-1 protected by exosomes is up-regulated and promotes cell proliferation and migration in non-small cell lung cancer. Biochem Biophys Res Commun. 490:406–414. 2017. View Article : Google Scholar : PubMed/NCBI

88 

Zhang W, Wang Q, Yang Y, Zhou S, Zhang P and Feng T: The role of exosomal lncRNAs in cancer biology and clinical management. Exp Mol Med. 53:1669–1673. 2021. View Article : Google Scholar : PubMed/NCBI

89 

Tao Y, Tang Y, Yang Z, Wu F, Wang L, Yang L, Lei L, Jing Y, Jiang X, Jin H, et al: Exploration of serum exosomal LncRNA TBILA and AGAP2-AS1 as promising biomarkers for diagnosis of non-small cell lung cancer. Int J Biol Sci. 16:471–482. 2020. View Article : Google Scholar : PubMed/NCBI

90 

Sun W, Jiang C, Liu Q, Wang N, Huang R, Jiang G and Yang Y: Exosomal noncoding RNAs: Decoding their role in thyroid cancer progression. Front Endocrinol (Lausanne). 15:13372262024. View Article : Google Scholar : PubMed/NCBI

91 

Hanifa M, Singh M, Randhawa PK, Jaggi AS and Bali A: A focus on Rho/ROCK signaling pathway: An emerging therapeutic target in depression. Eur J Pharmacol. 946:1756482023. View Article : Google Scholar : PubMed/NCBI

92 

Narumiya S, Tanji M and Ishizaki T: Rho signaling, ROCK and mDia1, in transformation, metastasis and invasion. Cancer Metastasis Rev. 28:65–76. 2009. View Article : Google Scholar : PubMed/NCBI

93 

Chin VT, Nagrial AM, Chou A, Biankin AV, Gill AJ, Timpson P and Pajic M: Rho-associated kinase signalling and the cancer microenvironment: novel biological implications and therapeutic opportunities. Expert Rev Mol Med. 17:e172015. View Article : Google Scholar : PubMed/NCBI

94 

Burridge K and Wennerberg K: Rho and Rac take center stage. Cell. 116:167–179. 2004. View Article : Google Scholar : PubMed/NCBI

95 

Zheng Y: Dbl family guanine nucleotide exchange factors. Trends Biochem Sci. 26:724–732. 2001. View Article : Google Scholar : PubMed/NCBI

96 

Moon SY and Zheng Y: Rho GTPase-activating proteins in cell regulation. Trends Cell Biol. 13:13–22. 2003. View Article : Google Scholar

97 

Jin D, Durgan J and Hall A: Functional cross-talk between Cdc42 and two downstream targets, Par6B and PAK4. Biochem J. 467:293–302. 2015. View Article : Google Scholar : PubMed/NCBI

98 

Seetharaman S and Etienne-Manneville S: Cytoskeletal crosstalk in cell migration. Trends Cell Biol. 30:720–735. 2020. View Article : Google Scholar : PubMed/NCBI

99 

Navarro-Lérida I, Sánchez-Álvarez M and Del Pozo M: Post-Translational modification and subcellular compartmentalization: Emerging concepts on the regulation and physiopathological relevance of RhoGTPases. Cells. 10:19902021. View Article : Google Scholar : PubMed/NCBI

100 

Fukata Y, Amano M and Kaibuchi K: Rho-Rho-kinase pathway in smooth muscle contraction and cytoskeletal reorganization of non-muscle cells. Trends Pharmacol Sci. 22:32–39. 2001. View Article : Google Scholar : PubMed/NCBI

101 

Hall A: Rho GTPases and the control of cell behaviour. Biochem Soc Trans. 33:891–895. 2005. View Article : Google Scholar : PubMed/NCBI

102 

Bishop AL and Hall A: Rho GTPases and their effector proteins. Biochem J. 348:241–255. 2000. View Article : Google Scholar : PubMed/NCBI

103 

Ridley AJ, Paterson HF, Johnston CL, Diekmann D and Hall A: The small GTP-binding protein rac regulates growth factor-induced membrane ruffling. Cell. 70:401–410. 1992. View Article : Google Scholar : PubMed/NCBI

104 

Patel S, McKeon D, Sao K, Yang C, Naranjo NM, Svitkina TM and Petrie RJ: Myosin II and Arp2/3 cross-talk governs intracellular hydraulic pressure and lamellipodia formation. Mol Biol Cell. 32:579–589. 2021. View Article : Google Scholar : PubMed/NCBI

105 

Eckenstaler R, Hauke M and Benndorf RA: A current overview of RhoA, RhoB, and RhoC functions in vascular biology and pathology. Biochem Pharmacol. 206:1153212022. View Article : Google Scholar : PubMed/NCBI

106 

Lou Y, Jiang Y, Liang Z, Liu B, Li T and Zhang D: Role of RhoC in cancer cell migration. Cancer Cell Int. 21:5272021. View Article : Google Scholar : PubMed/NCBI

107 

Steffen A, Koestler SA and Rottner K: Requirements for and consequences of Rac-dependent protrusion. Eur J Cell Biol. 93:184–193. 2014. View Article : Google Scholar : PubMed/NCBI

108 

Acevedo A and González-Billault C: Crosstalk between Rac1-mediated actin regulation and ROS production. Free Radic Biol Med. 116:101–113. 2018. View Article : Google Scholar : PubMed/NCBI

109 

Donkó A, Sharapova SO, Kabat J, Ganesan S, Hauck FH, Bergerson JRE, Marois L, Abbott J, Moshous D, Williams KW, et al: Clinical and functional spectrum of RAC2-related immunodeficiency. Blood. 143:1476–1487. 2024. View Article : Google Scholar : PubMed/NCBI

110 

Donnelly SK, Cabrera R, Mao SPH, Christin JR, Wu B, Guo W, Bravo-Cordero JJ, Condeelis JS, Segall JE and Hodgson L: Rac3 regulates breast cancer invasion and metastasis by controlling adhesion and matrix degradation. J Cell Biol. 216:4331–4349. 2017. View Article : Google Scholar : PubMed/NCBI

111 

He D, Xu L, Wu Y, Yuan Y, Wang Y, Liu Z, Zhang C, Xie W, Zhang L, Geng Z, et al: Rac3, but not Rac1, promotes ox-LDL induced endothelial dysfunction by downregulating autophagy. J Cell Physiol. 235:1531–1542. 2020. View Article : Google Scholar

112 

Nobes CD and Hall A: Rho, rac, and cdc42 GTPases regulate the assembly of multimolecular focal complexes associated with actin stress fibers, lamellipodia, and filopodia. Cell. 81:53–62. 1995. View Article : Google Scholar : PubMed/NCBI

113 

Nobes CD and Hall A: Rho GTPases control polarity, protrusion, and adhesion during cell movement. J Cell Biol. 144:1235–1244. 1999. View Article : Google Scholar : PubMed/NCBI

114 

Watanabe N, Madaule P, Reid T, Ishizaki T, Watanabe G, Kakizuka A, Saito Y, Nakao K, Jockusch BM and Narumiya S: p140mDia, a mammalian homolog of Drosophila diaphanous, is a target protein for Rho small GTPase and is a ligand for profilin. EMBO J. 16:3044–3056. 1997. View Article : Google Scholar : PubMed/NCBI

115 

Jerrell RJ and Parekh A: Matrix rigidity differentially regulates invadopodia activity through ROCK1 and ROCK2. Biomaterials. 84:119–129. 2016. View Article : Google Scholar : PubMed/NCBI

116 

Kubo T, Yamaguchi A, Iwata N and Yamashita T: The therapeutic effects of Rho-ROCK inhibitors on CNS disorders. Ther Clin Risk Manag. 4:605–615. 2008. View Article : Google Scholar : PubMed/NCBI

117 

Oka M, Fagan KA, Jones PL and McMurtry IF: Therapeutic potential of RhoA/Rho kinase inhibitors in pulmonary hypertension. Br J Pharmacol. 155:444–454. 2008. View Article : Google Scholar : PubMed/NCBI

118 

Naxerova K: Evolutionary paths towards metastasis. Nat Rev Cancer. 25:545–560. 2025. View Article : Google Scholar : PubMed/NCBI

119 

Majidpoor J and Mortezaee K: Steps in metastasis: An updated review. Med Oncol. 38:32021. View Article : Google Scholar : PubMed/NCBI

120 

Steeg PS: Tumor metastasis: Mechanistic insights and clinical challenges. Nat Med. 12:895–904. 2006. View Article : Google Scholar : PubMed/NCBI

121 

Bakir B, Chiarella AM, Pitarresi JR and Rustgi AK: EMT, MET, plasticity, and tumor metastasis. Trends Cell Biol. 30:764–776. 2020. View Article : Google Scholar : PubMed/NCBI

122 

Guo S, Huang J, Li G, Chen W, Li Z and Lei J: The role of extracellular vesicles in circulating tumor cell-mediated distant metastasis. Mol Cancer. 22:1932023. View Article : Google Scholar : PubMed/NCBI

123 

Matsuoka T and Yashiro M: Rho/ROCK signaling in motility and metastasis of gastric cancer. World J Gastroenterol. 20:13756–13766. 2014. View Article : Google Scholar : PubMed/NCBI

124 

Crosas-Molist E, Samain R, Kohlhammer L, Orgaz JL, George SL, Maiques O, Barcelo J and Sanz-Moreno V: Rho GTPase signaling in cancer progression and dissemination. Physiol Rev. 102:455–510. 2022. View Article : Google Scholar

125 

Itoh K, Yoshioka K, Akedo H, Uehata M, Ishizaki T and Narumiya S: An essential part for Rho-associated kinase in the transcellular invasion of tumor cells. Nat Med. 5:221–225. 1999. View Article : Google Scholar : PubMed/NCBI

126 

Zhang S, Tang Q, Xu F, Xue Y, Zhen Z, Deng Y, Liu M, Chen J, Liu S, Qiu M, et al: RhoA regulates G1-S progression of gastric cancer cells by modulation of multiple INK4 family tumor suppressors. Mol Cancer Res. 7:570–580. 2009. View Article : Google Scholar : PubMed/NCBI

127 

Kazmi N, Robinson T, Zheng J, Kar S, Martin RM and Ridley AJ: Rho GTPase gene expression and breast cancer risk: A mendelian randomization analysis. Sci Rep. 12:14632022. View Article : Google Scholar : PubMed/NCBI

128 

Cadamuro M, Nardo G, Indraccolo S, Dall'olmo L, Sambado L, Moserle L, Franceschet I, Colledan M, Massani M, Stecca T, et al: Platelet-derived growth factor-D and Rho GTPases regulate recruitment of cancer-associated fibroblasts in cholangiocarcinoma. Hepatology. 58:1042–1053. 2013. View Article : Google Scholar : PubMed/NCBI

129 

Bridges MC, Daulagala AC and Kourtidis A: LNCcation: lncRNA localization and function. J Cell Biol. 220:e2020090452021. View Article : Google Scholar : PubMed/NCBI

130 

Nie H, Liao Z, Wang Y, Zhou J, He X and Ou C: Exosomal long non-coding RNAs: Emerging players in cancer metastasis and potential diagnostic biomarkers for personalized oncology. Genes Dis. 8:769–780. 2021. View Article : Google Scholar : PubMed/NCBI

131 

Wang Y, Nie H, He X, Liao Z, Zhou Y, Zhou J and Ou C: The emerging role of super enhancer-derived noncoding RNAs in human cancer. Theranostics. 10:11049–11062. 2020. View Article : Google Scholar : PubMed/NCBI

132 

Kimura T, Horikoshi Y, Kuriyagawa C and Niiyama Y: Rho/ROCK pathway and noncoding RNAs: Implications in ischemic stroke and spinal cord injury. Int J Mol Sci. 22:115732021. View Article : Google Scholar : PubMed/NCBI

133 

Xu X, Wang X, Geng C, Nie X and Bai C: Long-chain non-coding RNA DAPK1 targeting miR-182 regulates pancreatic cancer invasion and metastasis through ROCK-1/rhoa signaling pathway. Int J Clin Exp Pathol. 10:9273–9283. 2017.PubMed/NCBI

134 

Liu J, Zhu Y and Ge C: LncRNA ZFAS1 promotes pancreatic adenocarcinoma metastasis via the RHOA/ROCK2 pathway by sponging miR-3924. Cancer Cell Int. 20:2492020. View Article : Google Scholar : PubMed/NCBI

135 

Li H, Wang X, Wen C, Huo Z, Wang W, Zhan Q, Cheng D, Chen H, Deng X, Peng C and Shen B: Long noncoding RNA NORAD, a novel competing endogenous RNA, enhances the hypoxia-induced epithelial-mesenchymal transition to promote metastasis in pancreatic cancer. Mol Cancer. 16:1692017. View Article : Google Scholar : PubMed/NCBI

136 

Liu Y, Wang Y, Fu X and Lu Z: Long non-coding RNA NEAT1 promoted ovarian cancer cells' metastasis through regulation of miR-382-3p/ROCK1 axial. Cancer Sci. 109:2188–2198. 2018. View Article : Google Scholar : PubMed/NCBI

137 

Zhao L, Ren C, Fang Y, Li P, Zhang H, Zuo L, Zhang J and Zhao J: MiR-490-3p sponged by lncRNA NEAT1 can attenuate lung adenocarcinoma progression by suppressing the RhoA/ROCK signaling pathway. Ann Clin Lab Sci. 53:42–51. 2023.PubMed/NCBI

138 

Zhang X, Wang DJ, Jia L and Zhang W: N6-methyladenosine-mediated LINC01087 promotes lung adenocarcinoma progression by regulating miR-514a-3p to upregulate centrosome protein 55. Kaohsiung J Med Sci. 40:801–818. 2024. View Article : Google Scholar : PubMed/NCBI

139 

Wang Y, Zeng X, Wang N, Zhao W, Zhang X, Teng S, Zhang Y and Lu Z: Long noncoding RNA DANCR, working as a competitive endogenous RNA, promotes ROCK1-mediated proliferation and metastasis via decoying of miR-335-5p and miR-1972 in osteosarcoma. Mol Cancer. 17:892018. View Article : Google Scholar : PubMed/NCBI

140 

Shao Y, Tong Z, Wei J and Yang T: LncRNA-zinc finger protein 281 downregulates rho-associated coiled-coil containing protein kinase 1 by upregulating miR-144 in osteosarcoma. Oncol Lett. 20:792020. View Article : Google Scholar : PubMed/NCBI

141 

Deng R, Zhang J and Chen J: lncRNA SNHG1 negatively regulates miRNA-101-3p to enhance the expression of ROCK1 and promote cell proliferation, migration and invasion in osteosarcoma. Int J Mol Med. 43:1157–1166. 2019.

142 

Wang Z, Wang Z, Liu J and Yang H: Long non-coding RNA SNHG5 sponges miR-26a to promote the tumorigenesis of osteosarcoma by targeting ROCK1. Biomed Pharmacother. 107:598–605. 2018. View Article : Google Scholar : PubMed/NCBI

143 

Chen J, Huang X, Wang W, Xie H, Li J, Hu Z, Zheng Z, Li H and Teng L: LncRNA CDKN2BAS predicts poor prognosis in patients with hepatocellular carcinoma and promotes metastasis via the miR-153-5p/ARHGAP18 signaling axis. Aging (Albany NY). 10:3371–3381. 2018. View Article : Google Scholar : PubMed/NCBI

144 

Dong S, Wang W, Liao Z, Fan Y, Wang Q and Zhang L: MYC-activated LINC00607 promotes hepatocellular carcinoma progression by regulating the miR-584-3p/ROCK1 axis. J Gene Med. 25:e34772023. View Article : Google Scholar : PubMed/NCBI

145 

Li R, Wan T, Qu J, Yu Y and Zheng R: Long non-coding RNA DLEUI promotes papillary thyroid carcinoma progression by sponging miR-421 and increasing ROCK1 expression. Aging (Albany NY). 12:20127–20138. 2020.PubMed/NCBI

146 

Lian Y, Xiong F, Yang L, Bo H, Gong Z, Wang Y, Wei F, Tang Y, Li X, Liao Q, et al: Long noncoding RNA AFAP1-AS1 acts as a competing endogenous RNA of miR-423-5p to facilitate nasopharyngeal carcinoma metastasis through regulating the Rho/Rac pathway. J Exp Clin Cancer Res. 37:2532018. View Article : Google Scholar : PubMed/NCBI

147 

Feng Z, Li X, Qiu M, Luo R, Lin J and Liu B: LncRNA EGFR-AS1 upregulates ROCK1 by sponging miR-145 to promote esophageal squamous cell carcinoma cell invasion and migration. Cancer Biother Radiopharm. 35:66–71. 2020.

148 

Zhou Y, Si L, Liu Z, Shi Y and Agula B: Long noncoding RNA zfas1 promotes progression of NSCLC via regulating of miR-590-3p. Cell Transplant. 29:9636897209194352020. View Article : Google Scholar : PubMed/NCBI

149 

Song S, Bian WG, Qin Z, Zeng D, Xu JJ and Tang HC: LncRNA BCYRN1 promotes cell migration and invasion of non-small cell lung cancer via the miR-30b-3p/ROCK1 axis. Neoplasma. 69:583–593. 2022. View Article : Google Scholar : PubMed/NCBI

150 

Yang J, Qiu Q, Qian X, Yi J, Jiao Y, Yu M, Li X, Li J, Mi C, Zhang J, et al: Long noncoding RNA LCAT1 functions as a ceRNA to regulate RAC1 function by sponging miR-4715-5p in lung cancer. Mol Cancer. 18:1712019. View Article : Google Scholar : PubMed/NCBI

151 

Xiao H, Zhu Q and Zhou J: Long non-coding RNA MALAT1 interaction with miR-429 regulates the proliferation and EMT of lung adenocarcinoma cells through RhoA. Int J Clin Exp Pathol. 12:419–430. 2019.

152 

Chen K and Zhang L: LINC00339 regulates ROCK1 by miR-152 to promote cell proliferation and migration in hepatocellular carcinoma. J Cell Biochem. 120:14431–14443. 2019. View Article : Google Scholar : PubMed/NCBI

153 

Wang W, Yang T, Li D, Huang Y, Bai G and Li Q: LINC00491 promotes cell growth and metastasis through miR-324-5p/ROCK1 in liver cancer. J Transl Med. 19:5042021. View Article : Google Scholar : PubMed/NCBI

154 

You LN, Tai QW, Xu L, Hao Y, Guo WJ, Zhang Q, Tong Q, Zhang H and Huang WK: Exosomal LINC00161 promotes angiogenesis and metastasis via regulating miR-590-3p/ROCK axis in hepatocellular carcinoma. Cancer Gene Ther. 28:719–736. 2021. View Article : Google Scholar : PubMed/NCBI

155 

Zhou Y, Fan RG, Qin CL, Jia J, Wu XD and Zha WZ: LncRNA-H19 activates CDC42/PAK1 pathway to promote cell proliferation, migration and invasion by targeting miR-15b in hepatocellular carcinoma. Genomics. 111:1862–1872. 2019. View Article : Google Scholar

156 

Guo D, Li Y, Chen Y, Zhang D, Wang X, Lu G, Ren M, Lu X and He S: DANCR promotes HCC progression and regulates EMT by sponging miR-27a-3p via ROCK1/LIMK1/COFILIN1 pathway. Cell Prolif. 52:e126282019. View Article : Google Scholar : PubMed/NCBI

157 

Ma Y, Xue Y, Liu X, Qu C, Cai H, Wang P, Li Z, Li Z and Liu Y: SNHG15 affects the growth of glioma microvascular endothelial cells by negatively regulating miR-153. Oncol Rep. 38:3265–3277. 2017. View Article : Google Scholar : PubMed/NCBI

158 

Li HL, Han PH, Pan DQ, Chen G, Lu XH and Li J: LncRNA XIST regulates cell proliferation, migration and invasion of glioblastoma via regulating miR-448 and ROCK1. J Biol Regul Homeost Agents. 34:2049–2058. 2020.PubMed/NCBI

159 

Chen X, Li D, Chen L, Hao B, Gao Y, Li L, Zhou C, He X and Cao Y: Long noncoding RNA LINC00346 promotes glioma cell migration, invasion and proliferation by up-regulating ROCK1. J Cell Mol Med. 24:13010–13019. 2020. View Article : Google Scholar : PubMed/NCBI

160 

Wang Z, Han Y, Li Q, Wang B and Ma J: LncRNA DLGAP1-AS1 accelerates glioblastoma cell proliferation through targeting miR-515-5p/ROCK1/NFE2L1 axis and activating Wnt signaling pathway. Brain Behav. 11:e23212021. View Article : Google Scholar : PubMed/NCBI

161 

Liu Y, Gao X and Tian X: High expression of long intergenic non-coding RNA LINC00662 contributes to malignant growth of acute myeloid leukemia cells by upregulating ROCK1 via sponging microRNA-340-5p. Eur J Pharmacol. 859:1725352019. View Article : Google Scholar : PubMed/NCBI

162 

Bai H, Li X and Wu S: Up-regulation of long non-coding RNA LOXL1-AS1 functions as an oncogene in cervical squamous cell carcinoma by sponging miR-21. Arch Physiol Biochem. 129:143–147. 2023. View Article : Google Scholar

163 

Chen X, Zhang Z, Ma Y, Su H, Xie P and Ran J: LINC02381 promoted cell viability and migration via targeting miR-133b in cervical cancer cells. Cancer Manag Res. 12:3971–3979. 2020. View Article : Google Scholar : PubMed/NCBI

164 

Yao X, Li X, Luo Y, Xu X, Liu J and Bu J: LncRNA GAS5 regulates osteosarcoma cell proliferation, migration, and invasion by regulating RHOB via sponging miR-663a. Cancer Manag Res. 12:8253–8261. 2020. View Article : Google Scholar : PubMed/NCBI

165 

Wang Y, Yang T, Zhang Z, Lu M, Zhao W, Zeng X and Zhang W: Long non-coding RNA TUG1 promotes migration and invasion by acting as a ceRNA of miR-335-5p in osteosarcoma cells. Cancer Sci. 108:859–867. 2017. View Article : Google Scholar : PubMed/NCBI

166 

Cui M, Wang J, Li Q, Zhang J, Jia J and Zhan X: Long non-coding RNA HOXA11-AS functions as a competing endogenous RNA to regulate ROCK1 expression by sponging miR-124-3p in osteosarcoma. Biomed Pharmacother. 92:437–444. 2017. View Article : Google Scholar : PubMed/NCBI

167 

Yu X, Wang D, Wang X, Sun S, Zhang Y, Wang S, Miao R, Xu X and Qu X: CXCL12/CXCR4 promotes inflammation-driven colorectal cancer progression through activation of RhoA signaling by sponging miR-133a-3p. J Exp Clin Cancer Res. 38:322019. View Article : Google Scholar : PubMed/NCBI

168 

Kong W, Li H, Xie L, Cui G, Gu W, Zhang H, Ma W and Zhou Y: LncRNA MCF2L-AS1 aggravates the malignant development of colorectal cancer via targeting miR-105-5p/RAB22A axis. BMC Cancer. 21:10692021. View Article : Google Scholar : PubMed/NCBI

169 

Horita K, Kurosaki H, Nakatake M, Ito M, Kono H and Nakamura T: Long noncoding RNA UCA1 enhances sensitivity to oncolytic vaccinia virus by sponging miR-18a/miR-182 and modulating the Cdc42/filopodia axis in colorectal cancer. Biochem Biophys Res Commun. 516:831–838. 2019. View Article : Google Scholar : PubMed/NCBI

170 

Tian Y, Zhong L, Gao S, Yu Y, Sun D, Liu X, Ji J, Yao Y, Liu Y and Jiang Z: LncRNA LINC00974 downregulates miR-122 to upregulate RhoA in oral squamous cell carcinoma. Cancer Biother Radiopharm. 36:18–22. 2021.

171 

Yang J, Wang WG and Zhang KQ: LINC00452 promotes ovarian carcinogenesis through increasing ROCK1 by sponging miR-501-3p and suppressing ubiquitin-mediated degradation. Aging (Albany NY). 12:21129–21146. 2020. View Article : Google Scholar : PubMed/NCBI

172 

Yang Q and Dong YJ: LncRNA SNHG20 promotes migration and invasion of ovarian cancer via modulating the microRNA-148a/ROCK1 axis. J Ovarian Res. 14:1682021. View Article : Google Scholar : PubMed/NCBI

173 

Chen S, Wang LL, Sun KX, Xiu YL, Zong ZH, Chen X and Zhao Y: The role of the long non-coding RNA TDRG1 in epithelial ovarian carcinoma tumorigenesis and progression through miR-93/RhoC pathway. Mol Carcinog. 57:225–234. 2018. View Article : Google Scholar

174 

Liu Y, Zong ZH, Guan X, Wang LL and Zhao Y: The role of long non-coding RNA PCA3 in epithelial ovarian carcinoma tumorigenesis and progression. Gene. 633:42–47. 2017. View Article : Google Scholar : PubMed/NCBI

175 

Li HL, Wang CP, Zhang Y, Du JX, Han LH and Yang HY: Long non-coding RNA PVT1 facilitates cell migration and invasion by regulating miR-148a-3p and ROCK1 in breast cancer. Clin Transl Oncol. 24:882–891. 2022. View Article : Google Scholar

176 

Chou J, Wang B, Zheng T, Li X, Zheng L, Hu J, Zhang Y, Xing Y and Xi T: MALAT1 induced migration and invasion of human breast cancer cells by competitively binding miR-1 with cdc42. Biochem Biophys Res Commun. 472:262–269. 2016. View Article : Google Scholar : PubMed/NCBI

177 

Chen Y, Sheng HG, Deng FM and Cai LL: Downregulation of the long noncoding RNA SNHG1 inhibits tumor cell migration and invasion by sponging miR-195 through targeting Cdc42 in oesophageal cancer. Kaohsiung J Med Sci. 37:181–191. 2021. View Article : Google Scholar

178 

Wang Z, Liu J, Wang R, Wang Q, Liang R and Tang J: Long non-coding RNA taurine upregulated gene 1 (TUG1) downregulation constrains cell proliferation and invasion through regulating cell division cycle 42 (CDC42) expression Via MiR-498 in esophageal squamous cell carcinoma cells. Med Sci Monit. 26:e9197142020.PubMed/NCBI

179 

Zong W, Feng W, Jiang Y, Cao Y, Ke Y, Shi X, Ju S, Cong H, Wang X, Cui M and Jing R: LncRNA CTC-497E21.4 promotes the progression of gastric cancer via modulating miR-22/NET1 axis through RhoA signaling pathway. Gastric Cancer. 23:228–240. 2020. View Article : Google Scholar

180 

Chen Z, Xu C, Pan X, Cheng G, Liu M, Li J and Mei Y: lncRNA DSCR8 mediates miR-137/Cdc42 to regulate gastric cancer cell proliferation, invasion, and cell cycle as a competitive endogenous RNA. Mol Ther Oncolytics. 22:468–482. 2021. View Article : Google Scholar : PubMed/NCBI

181 

Wang J, He Z, Xu J, Chen P and Jiang J: Long noncoding RNA LINC00941 promotes pancreatic cancer progression by competitively binding miR-335-5p to regulate ROCK1-mediated LIMK1/Cofilin-1 signaling. Cell Death Dis. 12:362021. View Article : Google Scholar : PubMed/NCBI

182 

Ferrè F, Colantoni A and Helmer-Citterich M: Revealing protein-lncRNA interaction. Brief Bioinform. 17:106–116. 2016. View Article : Google Scholar

183 

Ren S, Zhang N, Shen L, Lu Y, Chang Y, Lin Z, Sun N, Zhang Y, Xu J, Huang H and Jin H: Lnc00892 competes with c-Jun to block NCL transcription, reducing the stability of RhoA/RhoC mRNA and impairing bladder cancer invasion. Oncogene. 40:6579–6589. 2021. View Article : Google Scholar : PubMed/NCBI

184 

Wu Y, Shen QW, Niu YX, Chen XY, Liu HW and Shen XY: LncNORAD interference inhibits tumor growth and lung cancer cell proliferation, invasion and migration by down-regulating CXCR4 to suppress RhoA/ROCK signaling pathway. Eur Rev Med Pharmacol Sci. 24:5446–5455. 2020.PubMed/NCBI

185 

Hu X, Xiang L, He D, Zhu R, Fang J, Wang Z and Cao K: The long noncoding RNA KTN1-AS1 promotes bladder cancer tumorigenesis via KTN1 cis-activation and the consequent initiation of Rho GTPase-mediated signaling. Clin Sci (Lond). 135:555–574. 2021. View Article : Google Scholar : PubMed/NCBI

186 

Ge Z, Cheng Z, Yang X, Huo X, Wang N, Wang H, Wang C, Gu D, Zhao F, Yao M, et al: Long noncoding RNA SchLAH suppresses metastasis of hepatocellular carcinoma through interacting with fused in sarcoma. Cancer Sci. 108:653–662. 2017. View Article : Google Scholar : PubMed/NCBI

187 

Dai R, Zhou Y, Chen Z, Zou Z, Pan Z, Liu P and Gao X: Lnc-MUC20-9 binds to ROCK1 and functions as a tumor suppressor in bladder cancer. J Cell Biochem. 121:4214–4225. 2020. View Article : Google Scholar : PubMed/NCBI

188 

Wu DD, Chen X, Sun KX, Wang LL, Chen S and Zhao Y: Role of the lncRNA ABHD11-AS(1) in the tumorigenesis and progression of epithelial ovarian cancer through targeted regulation of RhoC. Mol Cancer. 16:1382017. View Article : Google Scholar : PubMed/NCBI

189 

Wu Y, Zhao Y, Huan L, Zhao J, Zhou Y, Xu L, Hu Z, Liu Y, Chen Z, Wang L, et al: An LTR retrotransposon-derived long noncoding RNA lncMER52A promotes hepatocellular carcinoma progression by binding p120-catenin. Cancer Res. 80:976–987. 2020. View Article : Google Scholar

190 

Shi D, Wu F, Mu S, Hu B, Zhong B, Gao F, Qing X, Liu J, Zhang Z and Shao Z: LncRNA AFAP1-AS1 promotes tumorigenesis and epithelial-mesenchymal transition of osteosarcoma through RhoC/ROCK1/p38MAPK/Twist1 signaling pathway. J Exp Clin Cancer Res. 38:3752019. View Article : Google Scholar : PubMed/NCBI

191 

Huang Y, Hong W and Wei X: The molecular mechanisms and therapeutic strategies of EMT in tumor progression and metastasis. J Hematol Oncol. 15:1292022. View Article : Google Scholar : PubMed/NCBI

192 

Kunc M, Skrzypkowska P, Pęksa R and Biernat W: Tumor-to-tumor metastases: Systematic review and meta-analysis of 685 reported cases. Clin Exp Metastasis. 42:142025. View Article : Google Scholar : PubMed/NCBI

193 

Peng L, Wang D, Han Y, Huang T, He X, Wang J and Ou C: Emerging role of cancer-associated fibroblasts-derived exosomes in tumorigenesis. Front Immunol. 12:7953722021. View Article : Google Scholar

194 

Zhang JY, Weng MZ, Song FB, Xu YG, Liu Q, Wu JY, Qin J, Jin T and Xu J: Long noncoding RNA AFAP1-AS1 indicates a poor prognosis of hepatocellular carcinoma and promotes cell proliferation and invasion via upregulation of the RhoA/Rac2 signaling. Int J Oncol. 48:1590–1598. 2016. View Article : Google Scholar : PubMed/NCBI

195 

Fan C, Tang Y, Wang J, Wang Y, Xiong F, Zhang S, Li X, Xiang B, Wu X, Guo C, et al: Long non-coding RNA LOC284454 promotes migration and invasion of nasopharyngeal carcinoma via modulating the Rho/Rac signaling pathway. Carcinogenesis. 40:380–391. 2019. View Article : Google Scholar

196 

Hu Q, Lin X, Ding L, Zeng Y, Pang D, Ouyang N, Xiang Y and Yao H: ARHGAP42 promotes cell migration and invasion involving PI3K/Akt signaling pathway in nasopharyngeal carcinoma. Cancer Med. 7:3862–3874. 2018. View Article : Google Scholar : PubMed/NCBI

197 

Fang G, Wang J, Sun X, Xu R, Zhao X, Shao L, Sun C and Wang Y: LncRNA MAGI2-AS3 is downregulated in the distant recurrence of hepatocellular carcinoma after surgical resection and affects migration and invasion via ROCK2. Ann Hepatol. 19:535–540. 2020. View Article : Google Scholar : PubMed/NCBI

198 

Liu F, Xiao Y, Ma L and Wang J: Regulating of cell cycle progression by the lncRNA CDKN2B-AS1/miR-324-5p/ROCK1 axis in laryngeal squamous cell cancer. Int J Biol Markers. 35:47–56. 2020. View Article : Google Scholar : PubMed/NCBI

199 

Ma T, Ma H, Zou Z, He X, Liu Y, Shuai Y, Xie M and Zhang Z: The long intergenic noncoding RNA 00707 promotes lung adenocarcinoma cell proliferation and migration by regulating Cdc42. Cell Physiol Biochem. 45:1566–1580. 2018. View Article : Google Scholar : PubMed/NCBI

200 

Song L, Wang L, Pan X and Yang C: lncRNA OIP5-AS1 targets ROCK1 to promote cell proliferation and inhibit cell apoptosis through a mechanism involving miR-143-3p in cervical cancer. Braz J Med Biol Res. 53:e88832020. View Article : Google Scholar : PubMed/NCBI

201 

Pan L, Meng Q, Li H, Liang K and Li B: LINC00339 promotes cell proliferation, migration, and invasion of ovarian cancer cells via miR-148a-3p/ROCK1 axes. Biomed Pharmacother. 120:1094232019. View Article : Google Scholar : PubMed/NCBI

202 

Chen S, Wang LL, Sun KX, Liu Y, Guan X, Zong ZH and Zhao Y: LncRNA PCGEM1 induces ovarian carcinoma tumorigenesis and progression through RhoA pathway. Cell Physiol Biochem. 47:1578–1588. 2018. View Article : Google Scholar : PubMed/NCBI

203 

Yuan S, Luan X, Chen H, Shi X and Zhang X: Long non-coding RNA EGFR-AS1 sponges micorRNA-381 to upregulate ROCK2 in bladder cancer. Oncol Lett. 19:1899–1905. 2020.PubMed/NCBI

204 

Shang W, Yang Y, Zhang J and Wu Q: Long noncoding RNA BDNF-AS is a potential biomarker and regulates cancer development in human retinoblastoma. Biochem Biophys Res Commun. 497:1142–1148. 2018. View Article : Google Scholar

205 

Nikanjam M, Kato S and Kurzrock R: Liquid biopsy: Current technology and clinical applications. J Hematol Oncol. 15:1312022. View Article : Google Scholar : PubMed/NCBI

206 

He X, Kuang G, Wu Y and Ou C: Emerging roles of exosomal miRNAs in diabetes mellitus. Clin Transl Med. 11:e4682021. View Article : Google Scholar : PubMed/NCBI

207 

Zhuang T, Wang S, Yu X, He X, Guo H and Ou C: Current status and future perspectives of platelet-derived extracellular vesicles in cancer diagnosis and treatment. Biomark Res. 12:882024. View Article : Google Scholar : PubMed/NCBI

208 

Wu S, Cui Y, Zhao H, Xiao X, Gong L, Xu H, Zhou Q, Ma D and Li X: Trophoblast exosomal UCA1 induces endothelial Injury through the PFN1-RhoA/ROCK pathway in preeclampsia: A human-specific adaptive pathogenic mechanism. Oxid Med Cell Longev. 2022:21989232022. View Article : Google Scholar : PubMed/NCBI

209 

Ding AX, Wang H, Zhang JM, Yang W and Kuang YT: lncRNA BANCR promotes the colorectal cancer metastasis through accelerating exosomes-mediated M2 macrophage polarization via regulating RhoA/ROCK signaling. Mol Cell Biochem. 479:13–27. 2024. View Article : Google Scholar

210 

Horita K, Kurosaki H, Nakatake M, Kuwano N, Oishi T, Itamochi H, Sato S, Kono H, Ito M, Hasegawa K, et al: lncRNA UCA1-mediated Cdc42 signaling promotes oncolytic vaccinia virus cell-to-cell spread in ovarian cancer. Mol Ther Oncolytics. 13:35–48. 2019. View Article : Google Scholar : PubMed/NCBI

211 

Liang L, Gu W, Li M, Gao R, Zhang X, Guo C and Mi S: The long noncoding RNA HOTAIRM1 controlled by AML1 enhances glucocorticoid resistance by activating RHOA/ROCK1 pathway through suppressing ARHGAP18. Cell Death Dis. 12:7022021. View Article : Google Scholar : PubMed/NCBI

212 

Xu K, Tian H, Zhao S, Yuan D, Jiang L, Liu X, Zou B and Zhang J: Long noncoding RNA LOC441178 reduces the invasion and migration of squamous carcinoma cells by targeting ROCK1. Biomed Res Int. 2018:43576472018. View Article : Google Scholar : PubMed/NCBI

213 

Zhang Z, Yu T and Geng W: Long non-coding RNA CCHE1 participates in postoperative distant recurrence but not local recurrence of osteosarcoma possibly by interacting with ROCK1. BMC Musculoskelet Disord. 21:4622020. View Article : Google Scholar : PubMed/NCBI

214 

Maldonado MDM and Dharmawardhane S: Targeting Rac and Cdc42 GTPases in cancer. Cancer Res. 78:3101–3111. 2018. View Article : Google Scholar : PubMed/NCBI

215 

Guo X, Stafford LJ, Bryan B, Xia C, Ma W, Wu X, Liu D, Songyang Z and Liu M: A Rac/Cdc42-specific exchange factor, GEFT, induces cell proliferation, transformation, and migration. J Biol Chem. 278:13207–13215. 2003. View Article : Google Scholar : PubMed/NCBI

216 

Hosseini K, Frenzel A and Fischer-Friedrich E: EMT induces characteristic changes of Rho GTPases and downstream effectors with a mitosis-specific twist. Phys Biol. 20:2023. View Article : Google Scholar : PubMed/NCBI

217 

Zhou W, Li X, Zhang B, Peng H, Quan C, Xiao X, Luo M, Huang Y, Xu D, Huang K, et al: The long non-coding RNA CCAT1 promotes erlotinib resistance in cholangiocarcinoma by inducing epithelial-mesenchymal transition via the miR-181a-5p/ROCK2 axis. Am J Cancer Res. 14:2852–2867. 2024. View Article : Google Scholar : PubMed/NCBI

218 

Abulwerdi FA, Xu W, Ageeli AA, Yonkunas MJ, Arun G, Nam H, Schneekloth JS Jr, Dayie TK, Spector D, Baird N and Le Grice SFJ: Selective small-molecule targeting of a triple helix encoded by the long noncoding RNA, MALAT1. ACS Chem Biol. 14:223–235. 2019. View Article : Google Scholar : PubMed/NCBI

219 

Cruz-Collazo A, Ruiz-Calderon JF, Picon H, Borrero-Garcia LD, Lopez I, Castillo-Pichardo L, Del Mar Maldonado M, Duconge J, Medina JI, Bayro MJ, et al: Efficacy of Rac and Cdc42 inhibitor MBQ-167 in triple-negative breast cancer. Mol Cancer Ther. 20:2420–2432. 2021. View Article : Google Scholar : PubMed/NCBI

220 

Moon GT, Lee JH, Jeong SH, Jin SW and Park YM: NecroX-5 can suppress melanoma metastasis by reducing the expression of Rho-family GTPases. J Clin Med. 10:27902021. View Article : Google Scholar : PubMed/NCBI

221 

Li D, Ding X, Xie M, Huang Z, Han P, Tian D and Xia L: CAMSAP2-mediated noncentrosomal microtubule acetylation drives hepatocellular carcinoma metastasis. Theranostics. 10:3749–3766. 2020. View Article : Google Scholar : PubMed/NCBI

222 

Gastonguay A, Berg T, Hauser AD, Schuld N, Lorimer E and Williams CL: The role of Rac1 in the regulation of NF-κB activity, cell proliferation, and cell migration in non-small cell lung carcinoma. Cancer Biol Ther. 13:647–656. 2012. View Article : Google Scholar : PubMed/NCBI

223 

Zhou M, He X, Zhang J, Mei C, Zhong B and Ou C: tRNA-derived small RNAs in human cancers: Roles, mechanisms, and clinical application. Mol Cancer. 23:762024. View Article : Google Scholar : PubMed/NCBI

224 

Sarfi M, Abbastabar M and Khalili E: Long noncoding RNAs biomarker-based cancer assessment. J Cell Physiol. 234:16971–16986. 2019. View Article : Google Scholar : PubMed/NCBI

225 

Mouraviev V, Lee B, Patel V, Albala D, Johansen TE, Partin A, Ross A and Perera RJ: Clinical prospects of long noncoding RNAs as novel biomarkers and therapeutic targets in prostate cancer. Prostate Cancer Prostatic Dis. 19:14–20. 2016. View Article : Google Scholar

226 

Goyal B, Yadav SRM, Awasthee N, Gupta S, Kunnumakkara AB and Gupta SC: Diagnostic, prognostic, and therapeutic significance of long non-coding RNA MALAT1 in cancer. Biochim Biophys Acta Rev Cancer. 1875:1885022021. View Article : Google Scholar : PubMed/NCBI

227 

Alix-Panabières C and Pantel K: Liquid biopsy: From discovery to clinical application. Cancer Discov. 11:858–873. 2021. View Article : Google Scholar : PubMed/NCBI

228 

Diez-Fraile A, Ceulaer J, Derpoorter C, Spaas C, Backer T, Lamoral P, Abeloos J and Lammens T: Circulating non-coding RNAs in head and neck cancer: Roles in diagnosis, prognosis, and therapy monitoring. Cells. 10:482020. View Article : Google Scholar

229 

Guo X, Peng Y, Song Q, Wei J, Wang X, Ru Y, Xu S, Cheng X, Li X, Wu D, et al: A liquid biopsy signature for the early detection of gastric cancer in patients. Gastroenterology. 165:402–413. 2023. View Article : Google Scholar : PubMed/NCBI

230 

Raza A, Khan AQ, Inchakalody VP, Mestiri S, Yoosuf ZSKM, Bedhiafi T, El-Ella DMA, Taib N, Hydrose S, Akbar S, et al: Dynamic liquid biopsy components as predictive and prognostic biomarkers in colorectal cancer. J Exp Clin Cancer Res. 41:992022. View Article : Google Scholar : PubMed/NCBI

231 

Kabzinski J, Kucharska-Lusina A and Majsterek I: RNA-based liquid biopsy in head and neck cancer. Cells. 12:19162023. View Article : Google Scholar : PubMed/NCBI

232 

Zhang N, Zhang H, Wu W, Zhou R, Li S, Wang Z, Dai Z, Zhang L, Liu F, Liu Z, et al: Machine learning-based identification of tumor-infiltrating immune cell-associated lncRNAs for improving outcomes and immunotherapy responses in patients with low-grade glioma. Theranostics. 12:5931–5948. 2022. View Article : Google Scholar : PubMed/NCBI

233 

Lu Q, Li J, Chen W, Wang Z, Wang D, Liu C, Sun Y, Jiang H, Zhang C, Chang Y, et al: NetLnc: A network-based computational framework to identify immune checkpoint-related lncRNAs for Immunotherapy response in melanoma. Int J Mol Sci. 26:45572025. View Article : Google Scholar : PubMed/NCBI

234 

Liu Y, Chen J, Yang D, Liu C, Tang C, Cai S and Huang Y: Machine learning combined with multi-omics to identify immune-related LncRNA signature as biomarkers for predicting breast cancer prognosis. Sci Rep. 15:238632025. View Article : Google Scholar : PubMed/NCBI

235 

Zhu Y, Zhu L, Wang X and Jin H: RNA-based therapeutics: An overview and prospectus. Cell Death Dis. 13:6442022. View Article : Google Scholar : PubMed/NCBI

236 

Puri B, Majumder S and Gaikwad AB: LncRNA MALAT1 as a potential diagnostic and therapeutic target in kidney diseases. Pathol Res Pract. 266:1557832025. View Article : Google Scholar

237 

Chen Y, Li Z, Chen X and Zhang S: Long non-coding RNAs: From disease code to drug role. Acta Pharm Sin B. 11:340–354. 2021. View Article : Google Scholar : PubMed/NCBI

238 

Botti G, Marra L, Malzone MG, Anniciello A, Botti C, Franco R and Cantile M: LncRNA HOTAIR as prognostic circulating Marker and potential therapeutic target in patients with tumor diseases. Curr Drug Targets. 18:27–34. 2017. View Article : Google Scholar

239 

Prabhakaran R, Thamarai R, Sivasamy S, Dhandayuthapani S, Batra J, Kamaraj C, Karthik K, Shah MA and Mallik S: Epigenetic frontiers: miRNAs, long non-coding RNAs and nanomaterials are pioneering to cancer therapy. Epigenetics Chromatin. 17:312024. View Article : Google Scholar : PubMed/NCBI

240 

Yang L, Li Z, Huang X, Huang L, Fu Y, Zhao B, Zhang Y, Ma L, Jing S, Fu L, et al: VPS9D1-AS1 antisense therapy via lipid nanoparticles reprograms cold tumors and enhances immunotherapy in colorectal cancer. J Control Release. 384:1138652025. View Article : Google Scholar : PubMed/NCBI

241 

He X, Lin F, Jia R, Xia Y, Liang Z, Xiao X, Hu Q, Deng X, Li Q and Sheng W: Coordinated modulation of long non-coding RNA ASBEL and curcumin co-delivery through multicomponent nanocomplexes for synchronous triple-negative breast cancer theranostics. J Nanobiotechnology. 21:3972023. View Article : Google Scholar : PubMed/NCBI

242 

Jia F, Li Y, Deng X, Wang X, Cui X, Lu J, Pan Z and Wu Y: Self-assembled fluorescent hybrid nanoparticles-mediated collaborative lncRNA CCAT1 silencing and curcumin delivery for synchronous colorectal cancer theranostics. J Nanobiotechnology. 19:2382021. View Article : Google Scholar : PubMed/NCBI

243 

Zhang Y, Liu F, Tan L, Li X, Dai Z, Cheng Q, Liu J, Wang Y, Huang L, Wang L and Wang Z: LncRNA-edited biomimetic nanovaccines combined with anti-TIM-3 for augmented immune checkpoint blockade immunotherapy. J Control Release. 361:671–680. 2023. View Article : Google Scholar : PubMed/NCBI

244 

Herman AB, Tsitsipatis D and Gorospe M: Integrated lncRNA function upon genomic and epigenomic regulation. Mol Cell. 82:2252–2266. 2022. View Article : Google Scholar : PubMed/NCBI

245 

Yip CW, Sivaraman DM, Prabhu AV and Shin JW: Functional annotation of lncRNA in high-throughput screening. Essays Biochem. 65:761–773. 2021. View Article : Google Scholar : PubMed/NCBI

246 

Street CA and Bryan BA: Rho kinase proteins-pleiotropic modulators of cell survival and apoptosis. Anticancer Res. 31:3645–3657. 2011.PubMed/NCBI

247 

Street CA, Routhier AA, Spencer C, Perkins AL, Masterjohn K, Hackathorn A, Montalvo J, Dennstedt EA and Bryan BA: Pharmacological inhibition of Rho-kinase (ROCK) signaling enhances cisplatin resistance in neuroblastoma cells. Int J Oncol. 37:1297–1305. 2010.PubMed/NCBI

Related Articles

  • Abstract
  • View
  • Download
  • Twitter
Copy and paste a formatted citation
Spandidos Publications style
Nie H, Yang X, Chen Q, He X, Han Y, He Q and Ou C: Emerging role of long non‑coding RNAs in the Rho/ROCK signaling pathway in tumor metastasis (Review). Int J Mol Med 56: 197, 2025.
APA
Nie, H., Yang, X., Chen, Q., He, X., Han, Y., He, Q., & Ou, C. (2025). Emerging role of long non‑coding RNAs in the Rho/ROCK signaling pathway in tumor metastasis (Review). International Journal of Molecular Medicine, 56, 197. https://doi.org/10.3892/ijmm.2025.5638
MLA
Nie, H., Yang, X., Chen, Q., He, X., Han, Y., He, Q., Ou, C."Emerging role of long non‑coding RNAs in the Rho/ROCK signaling pathway in tumor metastasis (Review)". International Journal of Molecular Medicine 56.6 (2025): 197.
Chicago
Nie, H., Yang, X., Chen, Q., He, X., Han, Y., He, Q., Ou, C."Emerging role of long non‑coding RNAs in the Rho/ROCK signaling pathway in tumor metastasis (Review)". International Journal of Molecular Medicine 56, no. 6 (2025): 197. https://doi.org/10.3892/ijmm.2025.5638
Copy and paste a formatted citation
x
Spandidos Publications style
Nie H, Yang X, Chen Q, He X, Han Y, He Q and Ou C: Emerging role of long non‑coding RNAs in the Rho/ROCK signaling pathway in tumor metastasis (Review). Int J Mol Med 56: 197, 2025.
APA
Nie, H., Yang, X., Chen, Q., He, X., Han, Y., He, Q., & Ou, C. (2025). Emerging role of long non‑coding RNAs in the Rho/ROCK signaling pathway in tumor metastasis (Review). International Journal of Molecular Medicine, 56, 197. https://doi.org/10.3892/ijmm.2025.5638
MLA
Nie, H., Yang, X., Chen, Q., He, X., Han, Y., He, Q., Ou, C."Emerging role of long non‑coding RNAs in the Rho/ROCK signaling pathway in tumor metastasis (Review)". International Journal of Molecular Medicine 56.6 (2025): 197.
Chicago
Nie, H., Yang, X., Chen, Q., He, X., Han, Y., He, Q., Ou, C."Emerging role of long non‑coding RNAs in the Rho/ROCK signaling pathway in tumor metastasis (Review)". International Journal of Molecular Medicine 56, no. 6 (2025): 197. https://doi.org/10.3892/ijmm.2025.5638
Follow us
  • Twitter
  • LinkedIn
  • Facebook
About
  • Spandidos Publications
  • Careers
  • Cookie Policy
  • Privacy Policy
How can we help?
  • Help
  • Live Chat
  • Contact
  • Email to our Support Team