International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Cancer is a multifactorial disease due to various causes and impacts, such as environmental factors, infectious agents, genetic alterations and epigenetic shifts (1,2). Advances in genetic research have not only elucidated the pathogenesis of several cancer types, such as acute myeloid leukemia (AML), non-small cell lung cancer (NSCLC) and prostate cancer (PCa), but have also directly contributed to the development of treatments (3–6). For example, imatinib is an effective treatment for leukemia caused by mutations in the breakpoint cluster region-Abelson, offering the possibility of long-term disease remission. Furthermore, <2% of the entire genome encode proteins, while the rest of the genetic material is composed of non-coding genes, which are key contributors to tumorigenesis among multiple cancer types, such as PCa and colorectal cancer (CRC) (7–9).
Transcripts >200 nucleotides in length and which have little or no ability to code for proteins are termed long non-coding RNAs (lncRNAs) (10,11). Previous studies identified that several dysregulated lncRNAs contribute to the development and progression of cancer types, working as oncogenes or tumor suppressors (12–15). HOTAIRM1 is a recently identified lncRNA located in the HOXA gene cluster on the human chromosome 7p15.2 (Fig. 1) (16). HOTAIRM1 was first identified as involved in the differentiation of granulocytes in the NB4 promyelocytic leukemia model (17). Since its identification, HOTAIRM1 has gained notable attention in cancer research (18–20).
The relevant studies in the present review were collected using PubMed (https://pubmed.ncbi.nlm.nih.gov) with a combination of the following terms: ‘HOTAIRM1’, ‘cancer’, ‘tumor’ and ‘disease’. English-language publications from the past 10 years were selected. Two reviewers (YJ and XL) independently performed an initial screening of titles and abstracts. The reference lists of potentially eligible articles were cross-checked to ensure extensive coverage of the literature. A total of 103 articles meeting the predefined literature searching criteria were identified by May 2025.
Therefore, the present review provides a comprehensive overview of the current understanding of the expression, roles and molecular mechanisms of HOTAIRM1 to regulate cancer.
Several previous studies have documented the abnormal expression of lncRNAs in different diseases, particularly in cancer (21–23). The dysregulation of lncRNAs contributes to the development of tumors through the promotion, proliferation, invasion and metastasis of cancer cells (24–26). The lncRNA HOTAIRM1 is upregulated in different types of cancer such as glioma (27–35), AML (36–38), osteosarcoma (OS) (39), endometrial cancer (EC) (40), thyroid cancer (TC) (41,42), NSCLC (43,44), oral squamous cell carcinoma (OSCC) (45), PCa (46), pancreatic ductal adenocarcinoma (PDAC) (47,48) and ovarian cancer (OC) (49,50). The involvement of HOTAIRM1 in governing tumor characteristics, including proliferation, invasion and metastasis, has been demonstrated. However, HOTAIRM1 expression is downregulated in papillary TC (PTC) (51), head and neck tumor (HNT) (52), hepatocellular carcinoma (HCC) (53), lung adenocarcinoma (ADC) (54), gastric cancer (GC) (55,56) and CRC (Table I) (57,58). These collective findings position HOTAIRM1 as a key tumor-related lncRNA, whose dysregulation could drive oncogenic pathways and offers a promising target for future cancer interventions.
Glioma is the most common primary central nervous system tumor (59). Several previous studies have reported a notable increase in HOTAIRM1 expression in glioblastoma tissues and cell lines when compared with their normal counterparts (27–32). Glioblastoma tissues and cells exhibit an abnormal HOTAIRM1 upregulation, which promotes glioma malignancy by enhancing cell proliferation, migration, invasion and VM formation. The high HOTAIRM1 expression is mediated by METTL3-dependent m6A modification (33). Furthermore, Snail family transcriptional repressor 2 transcriptionally activated HOTAIRM1 and a strong association was observed between increased HOTAIRM1 expression and worse prognosis in glioma (34). HOTAIRM1 is also strongly associated with decreased survival rates of patients diagnosed with glioblastoma (35).
AML is a heterogeneous disease characterized by genetic irregularities (including mutations in genes like FLT3, NPM1 and RUNX1) and epigenetic alterations (such as mutations in regulators of DNA methylation like DNMT3A, TET2 and IDH1/2) (60,61). Jing et al (36) observed that HOTAIRM1 expression was increased in 14 AML samples with nucleophosmin 1 (NPM1) mutations when compared with AML samples without NPM1 mutations. This finding suggested a potential functional link between HOTAIRM1 and this specific genetic irregularity, implying that HOTAIRM1 upregulation may be part of the oncogenic machinery in NPM1-mutated AML. Furthermore, Kaplan-Meier survival analysis on patients with AML demonstrated a notably reduced survival time in the group with high HOTAIRM1 expression. A different investigation demonstrated that the treatment of acute promyelocytic leukemia cells with all-trans retinoic acid led to an increase in HOTAIRM1 expression, which is key to myeloid differentiation. Notably, the transcription factor PU.1 binds to a specific DNA site (+1,100) in the promoter of the HOTAIRM1 gene, therefore activating it and increasing its expression (37). Furthermore, Hu et al (38) demonstrated that HOTAIRM1 expression is increased in patients with AML compared with individuals without the disease. The functional analysis indicated that HOTAIRM1 downregulation inhibits the growth and triggers cell death in AML cells, indicating its tumorigenic function in AML.
OS is a prevalent and aggressive form of primary bone cancer that mostly affects children and teenagers (62). HOTAIRM1 expression is markedly increased in both OS samples and cell lines compared with their non-cancerous counterparts. The association between HOTAIRM1 expression and the clinicopathological features of patients with OS revealed that individuals with high HOTAIRM1 expression are prone to having an advanced TNM stage (as defined by the American Joint Committee on Cancer, 8th edition) (63). HOTAIRM1 upregulation stimulates the growth and movement of cells while inhibiting cell death. These findings suggest that HOTAIRM1, a cancer-causing gene in OS, potentially holds promise in the identification and management of this disease (39).
In the female reproductive system, EC ranks as one of the top three prevalent malignancies (64). Li et al (40) identified markedly higher HOTAIRM1 expression in type I EC tissues compared with normal endometrium tissues and showed it is associated with the clinicopathological features of affected patients. The increased expression of HOTAIRM1 is also strongly associated with advanced International Federation of Gynecology and Obstetrics (FIGO) stage (according to the FIGO staging system, 2023 edition) (65) and the presence of lymph node metastasis. The suppression of HOTAIRM1 inhibits the growth, movement, infiltration of type I EC cells, epithelial-mesenchymal transition (EMT) and tumor growth in vivo (40).
TC is divided into three main histological groups: i) Differentiated TC, which includes papillary, follicular and oncocytic carcinomas; ii) medullary TC, often associated with multiple endocrine neoplasia type 2 syndrome; and iii) anaplastic TC, an aggressive malignancy that often arises from pre-existing differentiated lesions and is characterized by a high mortality rate (66,67). Zhang et al (41) reported that HOTAIRM1 gene is amplified and its expression is increased in anaplastic TC compared with PTC and normal thyroid tissue. Furthermore, patients with anaplastic TC indicating higher HOTAIRM1 copy number and expression have worse survival outcomes. The role of HOTAIRM1 in TC appears to be complex and potentially context-dependent. While one study by Li et al (42) reported that elevated HOTAIRM1 expression in TC cells and tissues was associated with advanced TNM stage and lymph node metastasis, another investigation found contrary evidence, demonstrating that HOTAIRM1 expression was notably reduced in PTC tissues and that lower levels correlated with lymph node metastasis and more advanced disease (51). This discrepancy highlights the need for further research to clarify the precise function of HOTAIRM1 in TC progression.
NSCLC is responsible for ~85% of cancer-related mortality globally, making it the primary contributor to lung cancer mortalities worldwide (68). Chen et al (43) observed that HOTAIRM1 expression is markedly increased in NSCLC tissues compared with tissues of a control group. A different study demonstrated that HOTAIRM1 expression is associated with tumor histological differentiation, tumor size, TNM stage and Ki-67 expression in patients with NSCLC. Furthermore, HOTAIRM1 expression is increased in NSCLC compared with that in adjacent non-cancerous tissues. Patients with NSCLC with low HOTAIRM1 expression have a markedly longer overall survival compared with those with high expression (44).
OSCC is a malignant tumor with the highest occurrence rate among tumors affecting mouth and face. OSCC is well known for its tendency to recur and spread to other parts of the body (69,70). Yu et al (45) reported that HOTAIRM1 expression is increased in OSCC and it is closely associated with poor prognosis. Systematic bioinformatics analyses revealed that HOTAIRM1 is associated with tumor stage, overall survival, genomic instability, tumor cell stemness, tumor microenvironment activity and immunocyte infiltration.
PCa is a major contributor to cancer-associated mortality among men, particularly in Western countries, with Africa and Asia having the lowest incidence rates (71,72). Wang et al (46) reported that HOTAIRM1 expression is increased in PCa cells. The inhibition of HOTAIRM1 expression prevents tumor cell proliferation while inducing programmed cell death through the modulation of proteins associated with apoptosis. The inhibition of HOTAIRM1 expression limits the activity of the Wnt pathway in PCa cells, thereby suppressing the malignant characteristics of tumor cells.
PDAC is the most common form of PCa arising from the epithelial lining of the pancreatic duct (73). Samples of 47 PDAC tissues and 5 cell lines exhibit an abnormal increase of HOTAIRM1 expression compared with its expression in a control group (47). Similarly, Zhou et al (48) reported that HOTAIRM1 expression is increased in 12 PDAC tissue samples when compared with the corresponding non-tumor samples.
OC is the eighth most common type of cancer among women worldwide and is the third most frequent gynecological cancer after cervical cancer and EC (74). Ye et al (49) reported an overexpression of HOTAIRM1 in the human ovarian cancer cell line SKOV3. Inhibition of HOTAIRM1 expression reduces cell proliferation and increases cell death. Chao et al (50) observed that HOTAIRM1 expression is reduced in both ovarian cancer tissues and cells and advanced FIGO stage and lymphatic metastasis are associated with reduced HOTAIRM1 expression. HOTAIRM1 overexpression inhibits the growth and invasion of OC cells, while enhancing apoptosis. Furthermore, HOTAIRM1 slows OC tumor growth in vivo.
Head and neck cancer is a frequently diagnosed form of cancer globally, with an annual incidence of >600,000 new cases (75,76). Zheng et al (52) reported that HOTAIRM1 expression is decreased in 43 head carcinoma tissues and 41 neck carcinoma tissues when compared with the corresponding adjacent normal tissues. Furthermore, no association was identified between HOTAIRM1 expression and age, sex or tumor location. However, patients with increased HOTAIRM1 expression have a higher probability to develop an advanced TNM stage, suggesting that although HOTAIRM1 is generally downregulated in head and neck cancer, tumors that maintain a relatively higher expression (although still lower compared with normal tissues) are associated with increased malignancy and progression.
HCC is classified as the sixth most prevalent tumor and the third main reason of cancer fatality (77). Zhang et al (53) reported that HOTAIRM1 expression is lower in HCC tissues compared with that in the adjacent non-cancerous tissues. Furthermore, the receiver operating characteristic curve demonstrated that HOTAIRM1 expression so a notable level of sensitivity and specificity in detecting HCC. The absence of disease progression in patients with HCC is associated with tumor size and HOTAIRM1 expression. However, no association was observed with age, sex, γ-glutamyl transferase levels, α-fetoprotein levels, Child-Pugh grade (as defined by the American Association for the Study of Liver Diseases practice guidelines) (78), hepatitis B surface antigen status, presence of cirrhosis, number of tumors, micro-vessel metastasis, tumor differentiation and TNM stage of HCC.
Among NSCLCs, ADC is one of the key histological subtypes with high incidence and mortality (79,80). Thus, current studies on HOTAIRM1 primarily focus on ADC rather than other NSCLC subtypes, such as squamous cell carcinoma or large cell carcinoma. Chen et al (54) reported a notable decrease in HOTAIRM1 expression in ADC tissues compared with that in normal lung tissues. A clear association exists between the decrease of HOTAIRM1 expression and clinical stage, metastasis to lymph nodes and tumor size. Furthermore, HOTAIRM1 inhibition is associated with poor overall survival in patients with ADC, as demonstrated by the Kaplan-Meier analysis.
GC is a prevalent malignancy worldwide. GC ranks as the second most prevalent form of cancer in China and ranks as the third leading cause of cancer-associated mortality (81). Xu et al (55) identified a notable decrease of HOTAIRM1 expression in GC tissues is associated with a low survival rate among patients with GC. These findings are consistent with those reported by Lu et al (56) suggesting a marked decrease in HOTAIRM1 expression in 20 gastric cancer tissues and cell lines. Furthermore, HOTAIRM1 expression is associated with the clinicopathological features of patients with GC and a strong association exists between decreased HOTAIRM1 expression and advanced TNM stage as well as lymph node metastasis.
CRC is one of the most prevalent malignancies worldwide, with particularly high incidence in Western countries (82,83). Wan et al (57) reported that HOTAIRM1 expression is reduced in CRC tissues compared with that in normal tissues. Furthermore, in the matched cohort, plasma HOTAIRM1 levels were markedly lower in patients with CRC compared with those in healthy controls. Ren et al (58) identified a similar scenario where HOTAIRM1 expression is downregulated in both CRC tissues and cell lines and is even lower in 5-fluorouracil (FU)-resistant CRC tissues and cell lines. This progressive downregulation in resistant tissues and cell lines strongly implied that the loss of HOTAIRM1 is a key event in the acquisition of chemoresistance, consistent with its established role in suppressing cancer progression through mechanisms such as the miR-17-5p/BTG3 axis (58).
LncRNAs interact with DNA, RNA or proteins as molecular absorbers, frameworks and stimulators, exerting regulatory functions in different biological processes, including gene regulation, cellular differentiation and human diseases, particularly cancer (84–88). HOTAIRM1 is involved in normal and abnormal biological processes. Several molecular functions have been identified after years of research and they are categorized into three primary pathways: i) Interaction with DNA; ii) interaction with RNA; and iii) interaction with proteins.
HOTAIRM1 is involved in the methylation alteration of several genes that are associated with tumors and in the modification of histones (Fig. 2). HOTAIRM1 controls gene expression through its interaction with polycomb repressive complex 2 (PRC2), which consists of enhancer of zeste homolog 2 (EZH2), suppressor of zeste 12 homolog and embryonic ectoderm development protein. HOTAIRM1 catalyzes the dimethylation and trimethylation of the histone 3 lysine residue 27 (H3K27me3), thus controlling the expression of its gene. Li et al (89) identified that HOTAIRM1 triggers the transcription of the homeobox A1 (HOXA1) gene by reducing the levels of histone H3 lysine 9 (H3K9) dimethylation, H3K27me3 and DNA methylation, which are markers associated with the suppression of gene expression. HOTAIRM1 prevents the recruitment of histone H3K9 methyltransferase, EZH2 and DNA methyltransferases to the HOXA1 promoter during its interaction with them, thereby reducing their local abundance at this site (89). Kim et al (90) reported that HOTAIRM1 directly interacts with EZH2, the histone methyltransferases responsible for H3K27me3 trimethylation, thereby preventing the deposition of H3K27me3 marks at the putative HOXA1 promoter. Therefore, HOXA1 expression is increased in ER+ breast cancer cells. Furthermore, HOTAIRM1 interacts with the histone demethylases PRC2 and ubiquitously transcribed tetratricopeptide repeat on chromosome X/mixed lineage leukemia to control chromatin structure and subsequently impacts the transcriptional activity of the HOXA gene cluster (91).
HOTAIRM1 also interacts with RNA to mediate several molecular functions, including the promotion of cell proliferation (33,36), invasion and metastasis (40,58). The relevance of competitive endogenous RNA (ceRNA) networks in cancer initiation and progression has become apparent in recent years (92,93). MicroRNAs (miR/miRNA) are involved in the ceRNA network, exerting a negative influence on mRNA expression. The canonical mechanism involves miRNAs binding to the 3′-untranslated regions of target mRNAs, which leads to their demethylation and subsequent destabilization (94,95). HOTAIRM1 competitively binds to specific miRNAs through the ceRNA mechanism, thereby relieving the repression of their target mRNAs and modulating tumor progression (Fig. 3). HOTAIRM1 competitively binds to miR-152-3p, functioning as a ceRNA that inhibits miR-152-3p activity and therefore upregulates its target Unc-51 like kinase 3 (ULK3), thereby modulating various leukemic cell processes, including autophagy, proliferation and apoptosis (36). Yu et al (39) identified that HOTAIRM1 functions as a molecular sponge for miR-664b-3p, thereby upregulating Ras homolog enriched in brain (Rheb) and activating the mTOR pathway to promote the Warburg effect in OS. Furthermore, it enhances E26 transformation-specific 1 (ETS1) mRNA expression and facilitates the osteogenic differentiation of mesenchymal stem cells derived from human bone marrow (96). Similarly, HOTAIRM1 promotes PH domain leucine-rich repeat protein phosphatase 1 upregulation in GC cells by sponging miR-29b-1-5p, thus demonstrating a classic ceRNA mechanism (55). Wang et al (97) reported that the HOTAIRM1/miR-519a-3p axis is markedly involved in the proliferation, apoptosis, inflammation and oxidative stress of neuroblastoma cells when exposed to the toxic metabolite 1-methyl-4-phenylpyridinium. HOTAIRM1 directly regulates mRNA stability. Specifically, its binding to insulin-like growth factor binding protein 2 (IGFBP2) mRNA increases IGFBP2 expression, thus promoting glioma cell proliferation, migration, invasion and vasculogenic mimicry formation (27).
Several lncRNAs including HOTAIRM1 are involved in the molecular regulation of proteins by directly binding to them (Fig. 4). For example, HOTAIRM1 interacts with heat shock protein family A (Hsp70) member 5 (HSPA5) and transcriptionally regulates its expression, with the resulting effects on proliferation being partly dependent on this chaperone (98). Han et al (98) demonstrated that HOTAIRM1 forms a complex with polypyrimidine tract-binding protein 1 and insulin-like growth factor 2 mRNA-binding protein 2, strengthening their interaction and facilitating their recruitment to the serine hydroxymethyltransferase 2 (SHMT2) mRNA. Therefore, the stability of SHMT2 mRNA has improved, leading to an increase in SHMT2 protein expression. This ultimately induces mitochondrial activity and promotes the malignant advancement of glioma. Liu et al (28) reported that HOTAIRM1 binds to the RNA-binding protein fused in sarcoma (FUS), thereby regulating E2F transcription factor 7 expression, promoting the proliferation, migration and invasion of glioma stem cell-transformed mesenchymal stem cells. In addition, Chen et al (99) demonstrated that the regulation of programmed cell death-ligand 1 expression in lung alveolar epithelial cells is controlled by HOTAIRM1 through its interaction with the key transcription factor HOXA1, having alleviated lung injury and improved survival of mice. Jing et al (36) demonstrated that the interaction of HOTAIRM1 with early growth response 1 (EGR1) and murine double minute 2 homolog (MDM2) suggests that it serves as a scaffold facilitating MDM2 recruitment to EGR1, thereby promoting autophagy and proliferation in leukemia cells.
Cancer is complex disease characterized by multiple genetic abnormalities, including epigenetic alterations, chromosomal translocations and gene deletions or amplifications. The human genome encodes several lncRNAs (such as PVT1, HOTAIR and MALAT1), most of which are not translated into proteins. Despite their lack of coding potential, lncRNAs exert key regulatory functions among different cellular processes (11,100). Among them, HOTAIRM1 modulates multiple signaling pathways (Wnt, Caspase signaling pathways) and is associated with hallmark cancer traits such as cell proliferation, apoptosis, invasion, metastasis, metabolic reprogramming and angiogenesis (Fig. 5).
Dysregulated cell proliferation resulting in uncontrolled growth represents a fundamental hallmark of tumorigenesis (101). Aberrant expression of HOTAIRM1 has been reported in multiple tumor types and is closely associated with enhanced tumor cell proliferation. For example, Wu et al (33) identified an increase in HOTAIRM1 expression in both glioma tissues and cells. Furthermore, HOTAIRM1 overexpression increases the proliferation of glioma cells. Zhou et al (102) identified a key functional connection among HOXA4, HOTAIRM1 and HSPA5, forming a novel regulatory circuit that governs HUVEC proliferation. HOTAIRM1 promotes OS cell proliferation by sponging miR-664b-3p, thereby activating the mTOR pathway (39). On the other hand, HOTAIRM1 also suppresses the proliferative ability of granulosa cells (103). Furthermore, Jing et al (36) demonstrated that HOTAIRM1 markedly promotes the proliferation of AML cells harboring NPM1 mutations.
Apoptosis is a programmed form of cell death that maintains cellular balance. The induction of apoptosis in cancer cells is a key strategy in clinical cancer therapy. Dahariya et al (104) reported that HOTAIRM1 functions as a molecular decoy for miR-125b regulating apoptosis to facilitate the terminal differentiation of megakaryocytes, thereby regulating key processes including apoptosis, cyclin D1-dependent cell cycling, and reactive oxygen species production. HOTAIRM1 also induces Jurkat cell apoptosis through the KIT/AKT signaling pathway (105). Liu et al (106) demonstrated that the loss of HOTAIRM1 activity causes an abnormal increase in chondrocyte apoptosis. Similarly, Ye et al (49) reported that HOTAIRM1 suppression increases the expression of pro-apoptotic factors, including Bad and Bax, while it reduces the expression of BH3 interacting-domain death agonist and Bcl-2 (anti-apoptotic factors) in ovarian cancer cells.
Autophagy is a self-degradative process in which cytoplasmic proteins and organelles are delivered to lysosomes to maintain cellular metabolism and homeostasis. A previous study has reported that HOTAIRM1 induces autophagy by upregulating ULK3 expression, leading to an increased expression of autophagy-related proteins including microtubule-associated protein LC3 II and a concomitant decrease in the autophagy substrate p62, thereby promoting cell proliferation and exerting a pro-tumorigenic effect (36).
Metastasis is a complex, multistep process involving EMT, invasion, intravasation, cell survival in the bloodstream, extravasation and the formation of secondary tumor colonies. The initial step in cancer metastasis is the EMT, during which epithelial cells lose their polarity and intercellular adhesion, acquire mesenchymal characteristics and gain enhanced migratory and invasive abilities. The upregulation of HOXA1 expression by HOTAIRM1 enhances the ability of type I EC cells to undergo EMT and metastasis, resulting in a decrease in the epithelial marker E-cadherin expression and an increase in the mesenchymal marker N-cadherin (40). Ren et al (58) examined the impact of HOTAIRM1 dysregulation in 5-FU-resistant CRC cells and identified that the increased expression of HOTAIRM1 reduces cell invasion and migration according to EMT assay.
Cell differentiation is the process through which cells acquire specialized function and form distinct tissues. By contrast, carcinogenesis represents a breakdown of normal differentiation, resulting in the uncontrolled proliferation of immature or aberrant cells. Tollis et al (107) used a model framework of spinal motor neurons to demonstrate that neuronal HOTAIRM1, a specific isoform of HOTAIRM1, influences cell fate between motor neurons and interneurons by promoting motor neuron differentiation. HOTAIRM1 contributes to proper progression during the early stages of neuronal differentiation. It also influences the core pluripotency network composed of NANOG, POU class 5 homeobox 1 and Sox2, thereby helping to maintain cells in an undifferentiated state (108). Wang et al (73) identified HOTAIRM1 as a novel regulator of the osteogenic differentiation of bone marrow-derived mesenchymal stem cells. This regulation occurs through the miR-152-3p/ETS1 axis, suggesting that HOTAIRM1 may represent a potential therapeutic target for osteoporosis. Furthermore, Dahariya et al (104) demonstrated that HOTAIRM1 regulates the p53-mediated control of cyclin D1 expression during megakaryocytopoiesis. The function of HOTAIRM1 is to promote megakaryocyte maturation by acting as a molecular sponge for miR-125b.
Tumor cells undergo notable metabolic reprogramming to support their rapid proliferation. Unlike regular cells, they preferentially use aerobic glycolysis, an energetically inefficient pathway with a considerably higher turnover rate, even under oxygen-sufficient conditions, a phenomenon known as the Warburg effect. The miR-664b-3p/Rheb/mTOR axis markedly improves the Warburg effect in OS cells, due to the notable enhancement by HOTAIRM1 (39). HOTAIRM1 knockdown attenuates the Warburg effect in NSCLC by reducing glucose uptake and lactate production. This metabolic suppression is associated with a marked decrease in hexokinase 2 protein expression, a key glycolytic enzyme that phosphorylates glucose to initiate glycolysis and sustain cancer cell energy metabolism (43). Han et al (98) reported that HOTAIRM1 increases SHMT2 protein expression by improving the lifespan of its mRNA, leading to the stimulation of mitochondrial activity through oxidative phosphorylation and serine metabolism.
Despite notable advances in antitumor therapies, resistance to chemotherapy, radiotherapy, targeted therapy and immunotherapy still represents a major obstacle to an effective cancer treatment (109). HOTAIRM1 is involved in the response to therapy by cancer cells. According to previous research, HOTAIRM1 interacts with the inhibitory region of ρ GTPase-activating protein 18 and suppresses its expression, resulting in the activation of the Ras homolog family member A/ρ-associated coiled-coil containing protein kinase 1 signaling pathway and enhancing leukemia cell resistance to glucocorticoid treatment (110). Gu et al (111) reported that HOTAIRM1 upregulation leads to lenvatinib resistance by reducing miR-34a expression and increasing Beclin-1 expression in HCC cancer cells. In addition, Chen et al (112) revealed that HOTAIRM1 suppression increases the effectiveness of cytarabine in killing cells by controlling the Wnt/β-catenin/platelet-type isoform of phosphofructokinase signaling pathway. In vitro and in vivo, HOTAIRM1 functions as a tumor suppressor in 5-FU-resistant CRC cells by reducing the activity of the miR-17-5p/B-cell translocation gene 3 (BTG3) pathway and preventing the development of multi-drug resistance (58).
Cancer remains a major global threat to human health. Although recent advances have slightly reduced the overall mortality rates, notable obstacles persist in the early diagnosis and effective management. Numerous patients are still diagnosed at advanced stages of the disease, which notably worsens their prognosis (113,114). Hence, it is key to identify novel biomarkers and examine the different molecular pathways involved in cancer for its timely detection and management.
Several studies have indicated the abnormal expression of lncRNAs in several diseases and their ability to function as tumor suppressors or oncogenes (115,116). HOTAIRM1 exhibits an abnormal expression in both tumor tissues and cells of different types of cancer and it serves as a standalone indicator for unfavorable prognosis in a wide range of carcinomas. HOTAIRM1 upregulation promotes tumor cell proliferation in several neoplasms, including glioma, AML, OS, EC, TC, NSCLC, OSCC, PCa, PDAC and OC. However, HOTAIRM1 upregulation suppresses cancer cell proliferation in PTC, OC, HNT, HCC, ADC, GC and CRC. Furthermore, a strong association exists between atypical HOTAIRM1 expression and several clinical and pathological characteristics of tumors, including age, tumor size, invasion of blood vessels, metastasis, overall survival and recurrence. These findings support the idea that the cellular function of HOTAIRM1 is not static but context-dependent.
Initially, HOTAIRM1 triggers H3K27me3 by interacting with EZH2/PRC2 and also participates in the alteration of chromatin structure by directing the recruitment of chromatin-modifying enzymes to a specific gene location. Furthermore, HOTAIRM1 acts as a molecular sponge for miRNAs, exerting its regulatory effects through the HOTAIRM1-miRNA-mRNA pathway. In addition, HOTAIRM1 binds to functional proteins within both the nucleus and cytoplasm, thereby modulating their expression and markedly influencing tumor progression. Other potential molecular mechanisms underlying the biological functions of HOTAIRM1 warrant further investigation.
HOTAIRM1 potentially regulates tumors and impacts multiple aspects of cancer, such as cell proliferation, death, invasion, spread, cellular differentiation, metabolism and resistance to chemotherapy. Nevertheless, in certain contexts, such as drug resistance, the exact function of HOTAIRM1 remains to be elucidated, as current studies report conflicting results that require further detailed investigation. Liang et al (110) revealed that HOTAIRM1 promotes GC resistance by inhibiting apoptosis in leukemia cells. Notably, Ren et al (58) reported that HOTAIRM1 suppresses the miR-17-5p/BTG3 pathway, leading to the inhibition of multi-drug resistance. Currently, further in-depth research is required to explore the potential ability of HOTAIRM1 to control additional characteristics of cancer, including the evasion of immune surveillance, genome instability and mutation, non-mutational epigenetic reprogramming, unlocking phenotypic plasticity and polymorphic microbiomes.
Not applicable.
The present study was supported by Natural Science Foundation of Beijing Municipal (grant no. 7254399) and supported by the Fundamental Research Funds for the Central Universities (grant no. APL24100310010301071027).
Not applicable.
YJ and YG conceptualized the present review. YJ and XL prepared the original draft. YJ and YG reviewed and edited the manuscript. YJ and YG provided supervision and project administration. YJ obtained funding for the present review. All authors read and approved the final manuscript. Data authentication is not applicable.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
|
lncRNAs |
long non-coding RNAs |
|
HOTAIRM1 |
HOXA transcript antisense RNA myeloid-specific 1 |
|
AML |
acute myeloid leukemia |
|
OS |
osteosarcoma |
|
EC |
endometrial cancer |
|
EMT |
epithelial-mesenchymal transition |
|
TC |
thyroid cancer |
|
NSCLC |
non-small cell lung cancer |
|
OSCC |
oral squamous cell carcinoma |
|
PCa |
prostate cancer |
|
PDAC |
pancreatic ductal adenocarcinoma |
|
HCC |
hepatocellular carcinoma |
|
ADC |
lung adenocarcinoma |
|
GC |
gastric cancer |
|
CRC |
colorectal cancer |
|
PRC2 |
polycomb repressive complex 2 |
|
H3K27me3 |
lysine residue 27 of histone 3 |
|
Vaghari-Tabari M, Ferns GA, Qujeq D, Andevari AN, Sabahi Z and Moein ZS: Signaling, metabolism, and cancer: An important relationship for therapeutic intervention. J Cell Physiol. 236:5512–5532. 2021. View Article : Google Scholar | |
|
Giunta S: Decoding human cancer with whole genome sequencing: A review of PCAWG Project studies published in February 2020. Cancer Metastasis Rev. 40:909–924. 2021. View Article : Google Scholar | |
|
Chi Y, Wang D, Wang J, Yu W and Yang J: Long non-coding RNA in the pathogenesis of cancers. Cells. 8:10152019. View Article : Google Scholar | |
|
Ramezankhani R, Solhi R, Es HA, Vosough M and Hassan M: Novel molecular targets in gastric adenocarcinoma. Pharmacol Ther. 220:1077142021. View Article : Google Scholar | |
|
Guo N, Liu JB, Li W, Ma YS and Fu D: The power and the promise of CRISPR/Cas9 genome editing for clinical application with gene therapy. J Adv Res. 40:135–152. 2022. View Article : Google Scholar | |
|
Traba J, Sack MN, Waldmann TA and Anton OM: Immunometabolism at the nexus of cancer therapeutic efficacy and resistance. Front Immunol. 12:6572932021. View Article : Google Scholar | |
|
Ghafouri-Fard S, Khoshbakht T, Taheri M and Hajiesmaeili M: Long intergenic non-protein coding RNA 460: Review of its role in carcinogenesis. Pathol Res Pract. 225:1535562021. View Article : Google Scholar | |
|
Hovhannisyan H and Gabaldón T: The long non-coding RNA landscape of Candida yeast pathogens. Nat Commun. 12:73172021. View Article : Google Scholar | |
|
Kovalenko TF, Yadav B, Anufrieva KS, Rubtsov YP, Zatsepin TS, Shcherbinina EY, Solyus EM, Staroverov DB, Larionova TD, Latyshev YA, et al: Functions of long non-coding RNA ROR in patient-derived glioblastoma cells. Biochimie. 200:131–139. 2022. View Article : Google Scholar | |
|
Statello L, Guo CJ, Chen LL and Huarte M: Gene regulation by long non-coding RNAs and its biological functions. Nat Rev Mol Cell Biol. 22:96–118. 2021. View Article : Google Scholar | |
|
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 | |
|
Sun KK, Zu C, Wu XY, Wang QH, Hua P, Zhang YF, Shen XJ and Wu YY: Identification of lncRNA and mRNA regulatory networks associated with gastric cancer progression. Front Oncol. 13:11404602023. View Article : Google Scholar | |
|
Pierce JB, Zhou H, Simion V and Feinberg MW: Long noncoding RNAs as therapeutic targets. Adv Exp Med Biol. 1363:161–175. 2022. View Article : Google Scholar | |
|
Sun W, Xu J, Wang L, Jiang Y, Cui J, Su X, Yang F, Tian L, Si Z and Xing Y: Non-coding RNAs in cancer therapy-induced cardiotoxicity: Mechanisms, biomarkers, and treatments. Front Cardiovasc Med. 9:9461372022. View Article : Google Scholar | |
|
Zhu Y, Ren J, Wu X, Zhang Y, Wang Y, Xu J, Tan Q, Jiang Y and Li Y: lncRNA ENST00000422059 promotes cell proliferation and inhibits cell apoptosis in breast cancer by regulating the miR-145-5p/KLF5 axis. Acta Biochim Biophys Sin (Shanghai). 55:1892–1901. 2023. | |
|
Bah I, Youssef D, Yao ZQ, McCall CE and El Gazzar M: Hotairm1 controls S100A9 protein phosphorylation in myeloid-derived suppressor cells during sepsis. J Clin Cell Immunol. 14:10006912023. | |
|
Zhang X, Weissman SM and Newburger PE: Long intergenic non-coding RNA HOTAIRM1 regulates cell cycle progression during myeloid maturation in NB4 human promyelocytic leukemia cells. RNA Biol. 11:777–787. 2014. View Article : Google Scholar | |
|
Basyegit H, Tatar BG, Kose S, Gunduz C, Ozmen Yelken B and Yilmaz Susluer S: Exploring the role of long non-coding RNAs in predicting outcomes for hepatitis B patients. Asian Pac J Cancer Prev. 25:4313–4321. 2024. View Article : Google Scholar | |
|
Bagheri-Mohammadi S, Karamivandishi A, Mahdavi SA and Siahposht-Khachaki A: New sights on long non-coding RNAs in glioblastoma: A review of molecular mechanism. Heliyon. 10:e397442024. View Article : Google Scholar | |
|
Nekoeian S, Rostami T, Norouzy A, Hussein S, Tavoosidana G, Chahardouli B, Rostami S, Asgari Y and Azizi Z: Identification of lncRNAs associated with the progression of acute lymphoblastic leukemia using a competing endogenous RNAs network. Oncol Res. 30:259–268. 2023. View Article : Google Scholar | |
|
Chen R, Zhou D, Chen Y, Chen M and Shuai Z: Understanding the role of exosomal lncRNAs in rheumatic diseases: A review. PeerJ. 11:e164342023. View Article : Google Scholar | |
|
Ramos TAR, Urquiza-Zurich S, Kim SY, Gillette TG, Hill JA, Lavandero S, Rêgo TG and Maracaja-Coutinho V: Single-cell transcriptional landscape of long non-coding RNAs orchestrating mouse heart development. Cell Death Dis. 14:8412023. View Article : Google Scholar | |
|
Hu C, Dai Q, Zhang R, Yang H, Wang M, Gu K, Yang J, Meng W, Chen P and Xu M: Case report: Identification of a novel LYN::LINC01900 transcript with promyelocytic phenotype and TP53 mutation in acute myeloid leukemia. Front Oncol. 13:13224032023. View Article : Google Scholar | |
|
Wang D, Zhao X, Li S, Guo H, Li S and Yu D: The impact of LncRNA-SOX2-OT/let-7c-3p/SKP2 Axis on head and neck squamous cell carcinoma progression: Insights from bioinformatics analysis and experimental validation. Cell Signal. 115:1110182024. View Article : Google Scholar | |
|
Yu M, He X, Liu T and Li J: lncRNA GPRC5D-AS1 as a ceRNA inhibits skeletal muscle aging by regulating miR-520d-5p. Aging (Albany NY). 15:13980–13997. 2023. View Article : Google Scholar | |
|
Zhang X, Zhong Y, Liu L, Jia C, Cai H, Yang J, Wu B and Lv Z: Fasting regulates mitochondrial function through lncRNA PRKCQ-AS1-mediated IGF2BPs in papillary thyroid carcinoma. Cell Death Dis. 14:8272023. View Article : Google Scholar | |
|
Wu Z, Lin Y and Wei N: N6-methyladenosine-modified HOTAIRM1 promotes vasculogenic mimicry formation in glioma. Cancer Sci. 114:129–141. 2023. View Article : Google Scholar | |
|
Liu L, Zhou Y, Dong X, Li S, Cheng S, Li H, Li Y, Yuan J, Wang L and Dong J: HOTAIRM1 maintained the malignant phenotype of tMSCs transformed by GSCs via E2F7 by binding to FUS. J Oncol. 2022:77344132022. | |
|
Shi T, Guo D, Xu H, Su G, Chen J, Zhao Z, Shi J, Wedemeyer M, Attenello F, Zhang L and Lu W: HOTAIRM1, an enhancer lncRNA, promotes glioma proliferation by regulating long-range chromatin interactions within HOXA cluster genes. Mol Biol Rep. 47:2723–2733. 2020. View Article : Google Scholar | |
|
Wang H, Li H, Jiang Q, Dong X, Li S, Cheng S, Shi J, Liu L, Qian Z and Dong J: HOTAIRM1 promotes malignant progression of transformed fibroblasts in glioma stem-like cells remodeled microenvironment via regulating miR-133b-3p/TGFβ Axis. Front Oncol. 11:6031282021. View Article : Google Scholar | |
|
Xia H, Liu Y, Wang Z, Zhang W, Qi M, Qi B and Jiang X: Long noncoding RNA HOTAIRM1 maintains tumorigenicity of glioblastoma stem-like cells through regulation of HOX gene expression. Neurotherapeutics. 17:754–764. 2020. View Article : Google Scholar | |
|
Hao Y, Li X, Chen H, Huo H, Liu Z and Chai E: Over-expression of long noncoding RNA HOTAIRM1 promotes cell proliferation and invasion in human glioblastoma by up-regulating SP1 via sponging miR-137. Neuroreport. 31:109–117. 2020. View Article : Google Scholar | |
|
Wu Z and Wei N: METTL3-mediated HOTAIRM1 promotes vasculogenic mimicry icontributionsn glioma via regulating IGFBP2 expression. J Transl Med. 21:8552023. View Article : Google Scholar | |
|
Xie P, Li X, Chen R, Liu Y, Liu DC, Liu W, Cui G and Xu J: Upregulation of HOTAIRM1 increases migration and invasion by glioblastoma cells. Aging (Albany NY). 13:2348–2364. 2020. View Article : Google Scholar | |
|
Ahmadov U, Picard D, Bartl J, Silginer M, Trajkovic-Arsic M, Qin N, Blümel L, Wolter M, Lim JKM, Pauck D, et al: The long non-coding RNA HOTAIRM1 promotes tumor aggressiveness and radiotherapy resistance in glioblastoma. Cell Death Dis. 12:8852021. View Article : Google Scholar | |
|
Jing Y, Jiang X, Lei L, Peng M, Ren J, Xiao Q, Tao Y, Tao Y, Huang J, Wang L, et al: Mutant NPM1-regulated lncRNA HOTAIRM1 promotes leukemia cell autophagy and proliferation by targeting EGR1 and ULK3. J Exp Clin Cancer Res. 40:3122021. View Article : Google Scholar | |
|
Wei S, Zhao M, Wang X, Li Y and Wang K: PU.1 controls the expression of long noncoding RNA HOTAIRM1 during granulocytic differentiation. J Hematol Oncol. 9:442016. View Article : Google Scholar | |
|
Hu N, Chen L, Li Q and Zhao H: LncRNA HOTAIRM1 is involved in the progression of acute myeloid leukemia through targeting miR-148b. RSC Adv. 9:10352–10359. 2019. View Article : Google Scholar | |
|
Yu X, Duan W, Wu F, Yang D, Wang X, Wu J, Zhou D and Shen Y: LncRNA-HOTAIRM1 promotes aerobic glycolysis and proliferation in osteosarcoma via the miR-664b-3p/Rheb/mTOR pathway. Cancer Sci. 114:3537–3552. 2023. View Article : Google Scholar | |
|
Li X, Pang L, Yang Z, Liu J, Li W and Wang D: LncRNA HOTAIRM1/HOXA1 axis promotes cell proliferation, migration and invasion in endometrial cancer. Onco Targets Ther. 12:10997–11015. 2019. View Article : Google Scholar | |
|
Zhang L, Zhang J, Li S, Zhang Y, Liu Y, Dong J, Zhao W, Yu B, Wang H and Liu J: Genomic amplification of long noncoding RNA HOTAIRM1 drives anaplastic thyroid cancer progression via repressing miR-144 biogenesis. RNA Biol. 18:547–562. 2021. View Article : Google Scholar | |
|
Li C, Chen X, Liu T and Chen G: lncRNA HOTAIRM1 regulates cell proliferation and the metastasis of thyroid cancer by targeting Wnt10b. Oncol Rep. 45:1083–1093. 2021. View Article : Google Scholar | |
|
Chen D, Li Y, Wang Y and Xu J: LncRNA HOTAIRM1 knockdown inhibits cell glycolysis metabolism and tumor progression by miR-498/ABCE1 axis in non-small cell lung cancer. Genes Genomics. 43:183–194. 2021. View Article : Google Scholar | |
|
Xiong F, Yin H, Zhang H, Zhu C, Zhang B, Chen S, Ling C and Chen X: Clinicopathologic features and the prognostic implications of long noncoding RNA HOTAIRM1 in non-small cell lung cancer. Genet Test Mol Biomarkers. 24:47–53. 2020. View Article : Google Scholar | |
|
Yu Y, Niu J, Zhang X, Wang X, Song H, Liu Y, Jiao X and Chen F: Identification and validation of HOTAIRM1 as a novel biomarker for oral squamous cell carcinoma. Front Bioeng Biotechnol. 9:7985842022. View Article : Google Scholar | |
|
Wang L, Wang L, Wang Q, Yosefi B, Wei S, Wang X and Shen D: The function of long noncoding RNA HOTAIRM1 in the progression of prostate cancer cells. Andrologia. 53:e138972021. View Article : Google Scholar | |
|
Luo Y, He Y, Ye X, Song J, Wang Q, Li Y and Xie X: High expression of long noncoding RNA HOTAIRM1 is associated with the proliferation and migration in pancreatic ductal adenocarcinoma. Pathol Oncol Res. 25:1567–1577. 2019. View Article : Google Scholar | |
|
Zhou Y, Gong B, Jiang ZL, Zhong S, Liu XC, Dong K, Wu HS, Yang HJ and Zhu SK: Microarray expression profile analysis of long non-coding RNAs in pancreatic ductal adenocarcinoma. Int J Oncol. 48:670–680. 2016. View Article : Google Scholar | |
|
Ye L, Meng X, Xiang R, Li W and Wang J: Investigating function of long noncoding RNA of HOTAIRM1 in progression of SKOV3 ovarian cancer cells. Drug Dev Res. 82:1162–1168. 2021. View Article : Google Scholar | |
|
Chao H, Zhang M, Hou H, Zhang Z and Li N: HOTAIRM1 suppresses cell proliferation and invasion in ovarian cancer through facilitating ARHGAP24 expression by sponging miR-106a-5p. Life Sci. 243:1172962020. View Article : Google Scholar | |
|
Li D, Chai L, Yu X, Song Y, Zhu X, Fan S, Jiang W, Qiao T, Tong J, Liu S, et al: The HOTAIRM1/miR-107/TDG axis regulates papillary thyroid cancer cell proliferation and invasion. Cell Death Dis. 11:2272020. View Article : Google Scholar | |
|
Zheng M, Liu X, Zhou Q and Liu G: HOTAIRM1 competed endogenously with miR-148a to regulate DLGAP1 in head and neck tumor cells. Cancer Med. 7:3143–3156. 2018. View Article : Google Scholar | |
|
Zhang Y, Mi L, Xuan Y, Gao C, Wang YH, Ming HX and Liu J: LncRNA HOTAIRM1 inhibits the progression of hepatocellular carcinoma by inhibiting the Wnt signaling pathway. Eur Rev Med Pharmacol Sci. 22:4861–4868. 2018. | |
|
Chen TJ, Gao F, Yang T, Li H, Li Y, Ren H and Chen MW: LncRNA HOTAIRM1 inhibits the proliferation and invasion of lung adenocarcinoma cells via the miR-498/WWOX Axis. Cancer Manag Res. 12:4379–4390. 2020. View Article : Google Scholar | |
|
Xu F, Chen M, Chen H, Wu N, Qi Q, Jiang X, Fang D, Feng Q, Jin R and Jiang L: The curcumin analog Da0324 inhibits the proliferation of gastric cancer cells via HOTAIRM1/miR-29b-1-5p/PHLPP1 Axis. J Cancer. 13:2644–2655. 2022. View Article : Google Scholar | |
|
Lu R, Zhao G, Yang Y, Jiang Z, Cai J, Zhang Z and Hu H: Long noncoding RNA HOTAIRM1 inhibits cell progression by regulating miR-17-5p/PTEN axis in gastric cancer. J Cell Biochem. 120:4952–4965. 2019. View Article : Google Scholar | |
|
Wan L, Kong J, Tang J, Wu Y, Xu E, Lai M and Zhang H: HOTAIRM1 as a potential biomarker for diagnosis of colorectal cancer functions the role in the tumour suppressor. J Cell Mol Med. 20:2036–2044. 2016. View Article : Google Scholar | |
|
Ren T, Hou J, Liu C, Shan F, Xiong X, Qin A, Chen J and Ren W: The long non-coding RNA HOTAIRM1 suppresses cell progression via sponging endogenous miR-17-5p/B-cell translocation gene 3 (BTG3) axis in 5-fluorouracil resistant colorectal cancer cells. Biomed Pharmacother. 117:1091712019. View Article : Google Scholar | |
|
Zhang W, Dang R, Liu H, Dai L, Liu H, Adegboro AA, Zhang Y, Li W, Peng K, Hong J and Li X: Machine learning-based investigation of regulated cell death for predicting prognosis and immunotherapy response in glioma patients. Sci Rep. 14:41732024. View Article : Google Scholar | |
|
Zafar N, Ghias K and Fadoo Z: Genetic aberrations involved in relapse of pediatric acute myeloid leukemia: A literature review. Asia Pac J Clin Oncol. 17:e135–e141. 2021. View Article : Google Scholar | |
|
Hayatigolkhatmi K, Valzelli R, El Menna O and Minucci S: Epigenetic alterations in AML: Deregulated functions leading to new therapeutic options. Int Rev Cell Mol Biol. 387:27–75. 2024. View Article : Google Scholar | |
|
Ding Y and Chen Q: Wnt/β-catenin signaling pathway: An attractive potential therapeutic target in osteosarcoma. Front Oncol. 14:14569592025. View Article : Google Scholar | |
|
Amin MB, Greene FL, Edge SB, Compton CC, Gershenwald JE, Brookland RK, Meyer L, Gress DM, Byrd DR and Winchester DP: The Eighth Edition AJCC Cancer Staging Manual: Continuing to build a bridge from a population-based to a more “personalized” approach to cancer staging. CA Cancer J Clin. 67:93–99. 2017. | |
|
Ding S, Hao Y, Qi Y, Wei H, Zhang J and Li H: Molecular mechanism of tumor-infiltrating immune cells regulating endometrial carcinoma. Genes Dis. 12:1014422024. View Article : Google Scholar | |
|
Menendez-Santos M, Gonzalez-Baerga C, Taher D, Waters R, Virarkar M and Bhosale P: Endometrial cancer: 2023 Revised FIGO staging system and the role of imaging. Cancers (Basel). 16:18692024. View Article : Google Scholar | |
|
Chen DW, Lang BHH, McLeod DSA, Newbold K and Haymart MR: Thyroid cancer. Lancet. 401:1531–1544. 2023. View Article : Google Scholar | |
|
Boucai L, Zafereo M and Cabanillas ME: Thyroid cancer: A review. JAMA. 331:425–435. 2024. View Article : Google Scholar | |
|
Meyer ML, Fitzgerald BG, Paz-Ares L, Cappuzzo F, Jänne PA, Peters S and Hirsch FR: New promises and challenges in the treatment of advanced non-small-cell lung cancer. Lancet. 404:803–822. 2024. View Article : Google Scholar | |
|
Badwelan M, Muaddi H, Ahmed A, Lee KT and Tran SD: Oral squamous cell carcinoma and concomitant primary tumors, what do we know? A review of the literature. Curr Oncol. 30:3721–3734. 2023. View Article : Google Scholar | |
|
Jagadeesan D, Sathasivam KV, Fuloria NK, Balakrishnan V, Khor GH, Ravichandran M, Solyappan M, Fuloria S, Gupta G, Ahlawat A, et al: Comprehensive insights into oral squamous cell carcinoma: Diagnosis, pathogenesis, and therapeutic advances. Pathol Res Pract. 261:1554892024. View Article : Google Scholar | |
|
Almeeri MNE, Awies M and Constantinou C: Prostate cancer, pathophysiology and recent developments in management: A narrative review. Curr Oncol Rep. 26:1511–1519. 2024. View Article : Google Scholar | |
|
Wilson TK and Zishiri OT: Prostate cancer: A review of genetics, current biomarkers and personalised treatments. Cancer Rep (Hoboken). 7:e700162024. | |
|
Dhillon J and Betancourt M: Pancreatic ductal adenocarcinoma. Monogr Clin Cytol. 26:74–91. 2020. View Article : Google Scholar | |
|
Konstantinopoulos PA and Matulonis UA: Clinical and translational advances in ovarian cancer therapy. Nat Cancer. 4:1239–1257. 2023. View Article : Google Scholar | |
|
Chow LQM: Head and neck cancer. N Engl J Med. 382:60–72. 2020. View Article : Google Scholar | |
|
Kudrimoti A and Kudrimoti MR: Head and neck cancers. Prim Care. 52:139–155. 2025. View Article : Google Scholar | |
|
Da BL, Suchman KI, Lau L, Rabiee A, He AR, Shetty K, Yu H, Wong LL, Amdur RL, Crawford JM, et al: Pathogenesis to management of hepatocellular carcinoma. Genes Cancer. 13:72–87. 2022. View Article : Google Scholar | |
|
Akhras A, Beran A, Guardiola J, Bhavsar-Burke I, Reyes B and Ur Rahman A: S2808 direct oral anticoagulants versus warfarin in elderly patients with child-pugh C cirrhosis and atrial fibrillation: A real-world perspective. Am J Gastroenterol. 120((10S2)): pS6032025. | |
|
Tawfiq RK, de Camargo Correia GS, Li S, Zhao Y, Lou Y and Manochakian R: Targeting lung cancer with precision: The ADC therapeutic revolution. Curr Oncol Rep. 27:669–686. 2025. View Article : Google Scholar | |
|
Merle G, Friedlaender A, Desai A and Addeo A: Antibody drug conjugates in lung cancer. Cancer J. 28:429–435. 2022. View Article : Google Scholar | |
|
López MJ, Carbajal J, Alfaro AL, Saravia LG, Zanabria D, Araujo JM, Quispe L, Zevallos A, Buleje JL, Cho CE, et al: Characteristics of gastric cancer around the world. Crit Rev Oncol Hematol. 181:1038412023. View Article : Google Scholar | |
|
Baidoun F, Elshiwy K, Elkeraie Y, Merjaneh Z, Khoudari G, Sarmini MT, Gad M, Al-Husseini M and Saad A: Colorectal cancer epidemiology: Recent trends and impact on outcomes. Curr Drug Targets. 22:998–1009. 2021. View Article : Google Scholar | |
|
Mahmoud NN: Colorectal cancer: Preoperative evaluation and staging. Surg Oncol Clin N Am. 31:127–141. 2022. View Article : Google Scholar | |
|
Salido-Guadarrama I, Romero-Cordoba SL and Rueda-Zarazua B: Multi-Omics Mining of lncRNAs with biological and clinical relevance in cancer. Int J Mol Sci. 24:166002023. View Article : Google Scholar | |
|
Mahato RK, Bhattacharya S, Khullar N, Sidhu IS, Reddy PH, Bhatti GK and Bhatti JS: Targeting long non-coding RNAs in cancer therapy using CRISPR-Cas9 technology: A novel paradigm for precision oncology. J Biotechnol. 379:98–119. 2024. View Article : Google Scholar | |
|
Yang Q, Fu Y, Wang J, Yang H and Zhang X: Roles of lncRNA in the diagnosis and prognosis of triple-negative breast cancer. J Zhejiang Univ Sci B. 24:1123–1140. 2023.(In English, Chinese). View Article : Google Scholar | |
|
Alharthi NS, Al-Zahrani MH, Hazazi A, Alhuthali HM, Gharib AF, Alzahrani S, Altalhi W, Almalki WH and Khan FR: Exploring the lncRNA-VEGF axis: Implications for cancer detection and therapy. Pathol Res Pract. 253:1549982023. View Article : Google Scholar | |
|
Mehmandar-Oskuie A, Jahankhani K, Rostamlou A, Mardafkan N, Karamali N, Razavi ZS and Mardi A: Molecular mechanism of lncRNAs in pathogenesis and diagnosis of auto-immune diseases, with a special focus on lncRNA-based therapeutic approaches. Life Sci. 336:1223222024. View Article : Google Scholar | |
|
Li Q, Dong C, Cui J, Wang Y and Hong X: Over-expressed lncRNA HOTAIRM1 promotes tumor growth and invasion through up-regulating HOXA1 and sequestering G9a/EZH2/Dnmts away from the HOXA1 gene in glioblastoma multiforme. J Exp Clin Cancer Res. 37:2652018. View Article : Google Scholar | |
|
Kim CY, Oh JH, Lee JY and Kim MH: The LncRNA HOTAIRM1 promotes tamoxifen resistance by mediating HOXA1 expression in ER+ breast cancer cells. J Cancer. 11:3416–3423. 2020. View Article : Google Scholar | |
|
Wang XQ and Dostie J: Reciprocal regulation of chromatin state and architecture by HOTAIRM1 contributes to temporal collinear HOXA gene activation. Nucleic Acids Res. 45:1091–1104. 2017. | |
|
Usman M, Li A, Wu D, Qinyan Y, Yi LX, He G and Lu H: The functional role of lncRNAs as ceRNAs in both ovarian processes and associated diseases. Noncoding RNA Res. 9:165–177. 2023. View Article : Google Scholar | |
|
Kim S: LncRNA-miRNA-mRNA regulatory networks in skin aging and therapeutic potentials. Front Physiol. 14:13031512023. View Article : Google Scholar | |
|
Lin Y, Wen H, Yang B, Wang C and Liang R: Integrated bioinformatics and validation to construct lncRNA-miRNA-mRNA ceRNA network in status epilepticus. Heliyon. 9:e222052023. View Article : Google Scholar | |
|
Jiang M, Wang Z, Lu T, Li X, Yang K, Zhao L, Zhang D, Li J and Wang L: Integrative analysis of long noncoding RNAs dysregulation and synapse-associated ceRNA regulatory axes in autism. Transl Psychiatry. 13:3752023. View Article : Google Scholar | |
|
Wang X, Liu Y and Lei P: LncRNA HOTAIRM1 promotes osteogenic differentiation of human bone marrow-derived mesenchymal stem cells by targeting miR-152-3p/ETS1 axis. Mol Biol Rep. 50:5597–5608. 2023. View Article : Google Scholar | |
|
Wang G, Yu Y and Wang Y: Effects of propofol on neuroblastoma cells via the HOTAIRM1/miR-519a-3p axis. Transl Neurosci. 13:57–69. 2022. View Article : Google Scholar | |
|
Han W, Wang S, Qi Y, Wu F, Tian N, Qiang B and Peng X: Targeting HOTAIRM1 ameliorates glioblastoma by disrupting mitochondrial oxidative phosphorylation and serine metabolism. iScience. 25:1048232022. View Article : Google Scholar | |
|
Chen W, Liu J, Ge F, Chen Z, Qu M, Nan K, Gu J, Jiang Y, Gao S and Liao Y: Long noncoding RNA HOTAIRM1 promotes immunosuppression in sepsis by inducing T cell exhaustion. J Immunol. 208:618–632. 2022. View Article : Google Scholar | |
|
Peng WX, Koirala P and Mo YY: LncRNA-mediated regulation of cell signaling in cancer. Oncogene. 36:5661–5667. 2017. View Article : Google Scholar | |
|
Ge T, Gu X, Jia R, Ge S, Chai P, Zhuang A and Fan X: Crosstalk between metabolic reprogramming and epigenetics in cancer: Updates on mechanisms and therapeutic opportunities. Cancer Commun (Lond). 42:1049–1082. 2022. View Article : Google Scholar | |
|
Zhou Y, Wu Q, Long X, He Y and Huang J: lncRNA HOTAIRM1 Activated by hoxa4 drives huvec proliferation through direct interaction with protein partner HSPA5. Inflammation. 47:421–437. 2024. View Article : Google Scholar | |
|
Guo H, Li T and Sun X: LncRNA HOTAIRM1, miR-433-5p and PIK3CD function as a ceRNA network to exacerbate the development of PCOS. J Ovarian Res. 14:192021. View Article : Google Scholar | |
|
Dahariya S, Raghuwanshi S, Thamodaran V, Velayudhan SR and Gutti RK: Role of long non-coding RNAs in human-induced pluripotent stem cells derived megakaryocytes: A p53, HOX antisense intergenic RNA Myeloid 1, and miR-125b interaction study. J Pharmacol Exp Ther. 384:92–101. 2023. View Article : Google Scholar | |
|
Li YR, Yan WJ, Cai LL and Deng XL: Effect of down-regulation of LncRNA-HOTAIRM1 to proliferation, apoptosis and KIT/AKT pathway of jurkat cells. Zhongguo Shi Yan Xue Ye Xue Za Zhi. 29:1123–1128. 2021.(In Chinese). | |
|
Liu WB, Li GS, Shen P, Li YN and Zhang FJ: Long non-coding RNA HOTAIRM1-1 silencing in cartilage tissue induces osteoarthritis through microRNA-125b. Exp Ther Med. 22:9332021. View Article : Google Scholar | |
|
Tollis P, Vitiello E, Migliaccio F, D'Ambra E, Rocchegiani A, Garone MG, Bozzoni I, Rosa A, Carissimo A, Laneve P and Caffarelli E: The long noncoding RNA nHOTAIRM1 is necessary for differentiation and activity of iPSC-derived spinal motor neurons. Cell Death Dis. 14:7412023. View Article : Google Scholar | |
|
Segal D, Coulombe S, Sim J and Josée Dostie J: A conserved HOTAIRM1-HOXA1 regulatory axis contributes early to neuronal differentiation. RNA Biol. 20:1523–1539. 2023. View Article : Google Scholar | |
|
Zamame Ramirez JA, Romagnoli GG and Kaneno R: Inhibiting autophagy to prevent drug resistance and improve anti-tumor therapy. Life Sci. 265:1187452021. View Article : Google Scholar | |
|
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 | |
|
Gu D, Tong M, Wang J, Zhang B, Liu J, Song G and Zhu B: Overexpression of the lncRNA HOTAIRM1 promotes lenvatinib resistance by downregulating miR-34a and activating autophagy in hepatocellular carcinoma. Discov Oncol. 14:662023. View Article : Google Scholar | |
|
Chen L, Hu N, Wang C and Zhao H: HOTAIRM1 knockdown enhances cytarabine-induced cytotoxicity by suppression of glycolysis through the Wnt/β-catenin/PFKP pathway in acute myeloid leukemia cells. Arch Biochem Biophys. 680:1082442020. View Article : Google Scholar | |
|
Gustafson MP, Ligon JA, Bersenev A, McCann CD, Shah NN and Hanley PJ: Emerging frontiers in immuno- and gene therapy for cancer. Cytotherapy. 25:20–32. 2023. View Article : Google Scholar | |
|
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 | |
|
Hashemi M, Moosavi MS, Abed HM, Dehghani M, Aalipour M, Heydari EA, Behroozaghdam M, Entezari M, Salimimoghadam S, Gunduz ES, et al: Long non-coding RNA (lncRNA) H19 in human cancer: From proliferation and metastasis to therapy. Pharmacol Res. 184:1064182022. View Article : Google Scholar | |
|
McCabe EM and Rasmussen TP: lncRNA involvement in cancer stem cell function and epithelial-mesenchymal transitions. Semin Cancer Biol. 75:38–48. 2021. View Article : Google Scholar |