Skip to main content
Erschienen in: Inflammation 1/2024

Open Access 29.10.2023 | RESEARCH

lncRNA HOTAIRM1 Activated by HOXA4 Drives HUVEC Proliferation Through Direct Interaction with Protein Partner HSPA5

verfasst von: Yu Zhou, Qiang Wu, Xiangshu Long, Youfu He, Jing Huang

Erschienen in: Inflammation | Ausgabe 1/2024

Abstract

Despite the substantial progress in deciphering the pathogenesis of atherosclerosis (AS), cardiovascular mortality is still increasing. Therefore, atherosclerotic cardiovascular disease remains a sweeping epidemic that jeopardizes human health. Disentangling the molecular underpinnings of AS is imperative in the molecular cardiology field. Overwhelming evidence has indicated that the recognition of a fascinating class of players, known as long non-coding RNAs (lncRNAs), provides causality for coordinating AS. However, the function and mechanism of HOTAIRM1 are still poorly understood in human umbilical vein endothelial cells (HUVECs) and AS. Herein, we primarily underscored that lncRNA HOTAIRM1 is potentially responsible for AS; as such, it was dramatically up-regulated in HUVECs upon ox-LDL stimulation. Functionally, HOTAIRM1 knockdown attenuated HUVEC proliferation and potentiated apoptosis in the absence and presence of ox‐LDL. Furthermore, HOTAIRM1 was preferentially located in the nuclei of HUVECs. Mechanistically, HOXA4 is directly bound to the HOTAIRM1 promoter and activated its transcription. Of note, a positive feedback signaling between HOXA4 and HOTAIRM1 was determined. Intriguingly, the interplay between HOTAIRM1 and HSPA5 occurred in an RNA-binding protein pattern and a transcription-dependent regulatory manner. In addition, HSPA5 overexpression partially antagonized HUVEC proliferation inhibition of HOTAIRM1 depletion. Taken together, our findings delineate a pivotal functional interaction among HOXA4, HOTAIRM1, and HSPA5 as a novel regulatory circuit for modulating HUVEC proliferation. An in-depth investigation of the HOXA4-HOTAIRM1-HSPA5 axis promises to yield significant breakthroughs in identifying the molecular mechanisms governing AS and developing therapeutic avenues for AS.

Graphical Abstract

Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1007/​s10753-023-01919-x.

Highlights

• HOTAIRM1 was up-regulated in HUVECs upon the presence of ox-LDL
• In vitro HOTAIRM1 depletion resulted in impaired HUVEC proliferation and increased apoptosis
• HOXA4 directly bound to the HOTAIRM1 promoter and initiated its transcriptional activation
• A core binding site with the base sequence “GTGATTAA” as the HOXA4 response element in the HOTAIRM1 promoter was identified
• A positive feedback signaling code existed between HOTAIRM1 and HOXA4 in HUVECs
• HOTAIRM1 bound and modulated HSPA5 in RNA-binding protein fashion and a transcription-dependent regulatory manner

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

INTRODUCTION

Atherosclerosis (AS) is a complex and chronic inflammatory disease characterized by lipid deposition in the arterial wall that is responsible for diverse cardiovascular events [1, 2]. The aberrant vascular endothelial cell (VEC) proliferation is a hallmark of early AS and vascular injury progression [3]. Endothelial cell injury or dysfunction is the initial step in AS [4]. VECs are one of the primary cellular constituents of the blood vessel wall, and the destruction of their integrity is a pivotal factor in AS pathogenesis [5]. The dynamic equilibrium between VEC proliferation and apoptosis is the key cytological basis for maintaining VEC integrity and ensuring normal vascular function [6]. Thus, interrogating novel molecules affecting the proliferation and apoptosis of VEC is important in AS. In this study, a cellular model induced by ox-LDL was used to elucidate the mechanism underlying AS.
Long non-coding RNAs (lncRNAs) represent a broad class of non-coding RNAs that are over 200 nucleotides in length [7, 8]. Burgeoning findings have shown that lncRNAs are versatile regulators of multilevel gene expression, including transcription, post-transcriptional processes, alternative splicing, protein translation and modification, protein transport, and localization [913]. lncRNAs are pivotal in shaping cellular activity as functional units in various cancers, such as cell proliferation, migration, and apoptosis [14]. There is a burgeoning interest in lncRNAs because of their roles in AS development and progression [15]. For instance, the lncRNA HOXA11-AS promotes vascular endothelial cell injury in AS by regulating the miR-515-5p/ROCK1 axis [16]; lncRNA DANCR accelerates AS development by regulating the miR-214-5p/COX20 signaling pathway [17]; and lncRNA PVT1 knockdown reduces AS by inactivating the MAPK/NF-κB pathway [18]. Although no such role has yet been delineated regarding HOXA transcript antisense RNA myeloid-specific 1 (HOTAIRM1) in the cardiology field and less is known about its functional annotation and molecular underpinnings in human umbilical vein endothelial cell (HUVEC), it appears that lncRNAs can still have an impact on AS onset and progression. Therefore, deciphering the functional significance and molecular machinery of HOTAIRM1 is crucially important in this field; however, it is increasingly challenging.
HOXA transcript antisense RNA myeloid-specific 1 (HOTAIRM1), located at the 5′ end of homeobox A (HOXA) gene cluster with a length of 1052 bp, is a natural antisense transcript of HOXA1 gene exclusively expressed in the myeloid lineage [19] and is induced during neuronal differentiation [20]. HOTAIRM1 was initially discovered and investigated in NB4 promyelocytic leukemia cells, and its knockdown halted all-trans retinoid acid (ATRA)-induced granulocytic differentiation [21, 22]. More recently, it was shown to be important in multiple cancers, such as breast cancer [23], lung cancer [24], and glioblastoma multiforme [25]. Notwithstanding, the role of HOTAIRM1 in diseases has largely highlighted its implication in tumors, as to whether it is of biological significance, and whether its new perspective in HUVECs remains to be elucidated functionally and mechanistically. The primary signaling pathway regarding HOTAIRM1 refers to the NF-κB [2628] and Wnt signaling pathways [29, 30] in various biological regulations to date. Robust data indicate that these pathways are strongly associated with AS. Therefore, we speculate that HOTAIRM1 is likely to play an important role in AS onset and progression. Further exploration is imperiously warranted, and insights into the mechanistic detail of HOTAIRM1 may shed light on a new direction in therapeutic strategies for AS.
In this study, an unprecedented role of HOTAIRM1 was corroborated in HUVECs; we determined the influence of ox-LDL on HOTAIRM1 expression in HUVECs and provided mechanistic insights into the HUVEC proliferation regulation of HOTAIRM1 by the interplay between HOXA4 and its downstream associated protein partner, HSPA5.

MATERIALS AND METHODS

This study was approved by the Ethics Committee of Guizhou Provincial People’s Hospital. The animal experimental protocol was approved by the Animal Ethics Committee of Guizhou Provincial People’s Hospital (License number: 2022–038).

AS Mouse Model

Male ApoE mice in the AS group were fed a high-fat diet (21% fat and 0.15% cholesterol) at 4 weeks of age. Male ApoE mice in the control group were fed a normal diet (4% fat and 0% cholesterol). After 12 weeks, the mice were sacrificed, and the aortas were removed for further experiments.

Cell Culture, Treatment, and Transfection

HUVECs were obtained from the American Type Culture Collection (ATCC, Manassas, VA, USA, AC337632). The cells were cultured in an F-12 K medium (Invitrogen, Carlsbad, CA, USA, 21127022) containing 10% FBS (Gibco) and 1% penicillin–streptomycin (Thermo Fisher Scientific, 15140122). All cells were grown at 37 ℃ in a humidified atmosphere containing 5% CO2. For ox-LDL induction experiments, HUVECs were induced with ox-LDL (100 μg/mL, UnionBiol, Beijing, China, jk-002) for 24 h when the cells reached 70–80% confluence. HOXA4 and HSPA5 in HUVECs were knocked down using small interfering RNAs (siRNAs), and HOTAIRM1 was silenced using antisense oligonucleotides (ASOs). The ASOs and siRNAs were synthesized by GenePharma (Shanghai, China). The pcDNA3.1 vectors from Invitrogen were used to construct pcDNA3.1/HOXA4, pcDNA3.1/HOTAIRM1, and pcDNA3.1/HSPA5, with the empty vector as the negative control. Lipofectamine 3000 (Invitrogen, L3000015) was used for the transfection experiments.

Quantitative Real-time Polymerase Chain Reaction

Total RNA was extracted from the arterial tissue and HUVECs using TRIzol reagent according to the manufacturer’s protocol (Thermo Fisher Scientific, 12183555), and cDNA was acquired following reverse transcription. Quantitative real-time polymerase chain reaction (RT-qPCR) was performed using the SYBR Green Master Mix (Takara, Dalian, China, RR036A). The results were normalized to the expression of β-actin, and data were analyzed based on the 2 − ΔΔCt method. Supplementary File 1 lists the forward and reverse primers.

Western Blotting

Total cellular protein was extracted using RIPA lysis buffer (Beyotime, Shanghai, China, P0013B) and subjected to sodium dodecyl sulfate–polyacrylamide gel electrophoresis. Separated proteins were transferred to PVDF membranes, treated with 5% skim milk at room temperature for 2 h, and then incubated with corresponding primary antibodies against HOXA4 (Abcam, 131049), HSPA5 (Abcam, ab108615), PCNA (CST, 13110), cyclin D1 (Abcam, ab16663), CKD4 (Abcam, ab108357), caspase-3 (Abcam, ab32351), caspase-9 (Abcam, ab32539), Bax (Abcam, ab32503), Bcl-2 (Abcam, ab185002), and GAPDH (Abcam, ab181602) overnight at 4 °C, followed by conjugation with horseradish peroxidase at 37 °C for 2 h. Band density was visualized using an ECL kit (Invitrogen, Carlsbad, CA, USA, WP20005).

Oil Red O Staining and H&E Staining

Tissues were stained with filtered Oil Red O solution (Sigma). Oil red staining was visualized under a microscope. The root and aortic arch bifurcation vessels were immediately fixed in 4% neutral formaldehyde solution, dehydrated by soaking in 75% distilled water, and immersed in formaldehyde solution for H&E staining.

Cell Proliferation Assays and Cell Cycle Analysis

For the Cell Counting Kit-8 (CCK-8) assay, to detect the viability of HUVECs, transfected-HUVECs were added to CCK-8 solution (Dojindo, KMJ, Japan, CK04) at the indicated time and incubated for 2 h at 37 °C; the absorbance was determined at the wavelength of 450 nm. For the EdU assay, HUVECs were incubated in a 24-well plate for transfection for 72 h and subsequently added to 100 µL of EdU (RIBOBIO, Shanghai, China, C10310-1). A fluorescence microscope was used to visualize the cells (Olympus, Tokyo, Japan). For cell cycle assay, after transfection for 72 h, cells were digested and washed with PBS and then fixed in 70% ice-cold ethanol at 4 °C overnight. The cells were treated with 500 µL of RNase A, stained with 50 μg/mL propidium iodide (PI) for 15 min, and examined by flow cytometry.

Cell Apoptosis Assay

For apoptosis analysis, cells were double-stained with 7-AAD and PE per the manufacturer’s instructions (559763; PE Annexin V Apoptosis Detection Kit I, BD, USA, 559763). GFP-positive cells were assessed by flow cytometry (FACSCantoII, 338960; BD Biosciences, San Jose, CA, USA, 338960). Terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) staining was carried out to test cell apoptosis. Briefly, the cells were fixed using 4% formaldehyde, followed by staining with the TUNEL kit according to the manufacturer’s instructions (Vazyme, TUNEL Bright-Red Apoptosis Detection Kit, A113). The TUNEL-positive cells were counted using a fluorescence microscope.

Chromatin Immunoprecipitation Assay

Chromatin immunoprecipitation (ChIP) analysis was conducted using a Millipore ChIP Assay Kit (Millipore, MA, USA, 17–295) according to the manufacturer’s instructions. HUVECs were cross-linked with formaldehyde and sonicated at 200–1000 bp. Sonicated lysates were cleared and incubated overnight at 4 °C with magnetic beads coupled with HOXA4 antibody (Santa Cruz, sc-398426). RT-qPCR was used to analyze the precipitated chromatin DNA.

Dual-Luciferase Reporter Assay

To dissect the transcriptional regulation of HOXA4 on HOTAIRM1, the sequence of the wild-type or indicated site-mutated HOTAIRM1 promoter (WT, Site 1-MUT, Site 2-MUT, Site 3-MUT, Site 4-MUT, and MUT) was synthesized and cloned into the pGL3 vector to construct the corresponding reporter plasmids. Thereafter, the indicated recombinant reporter was co-transfected with an empty vector or a HOXA4 plasmid into HUVECs. After 48 h, the luciferase activity in each group was measured using a Dual-Luciferase Reporter Assay System (Promega, Madison, WI, USA).

Fluorescence In Situ Hybridization Assay

A fluorescent in situ Hybridization (FISH) Kit was used to analyze HOTAIRM1 subcellular location (GenePharma, Suzhou, China). Cy3-labeled HOTAIRM1 probe was also synthesized. Images were acquired using a confocal microscope (Olympus). GAPDH and U6 served as controls for cytoplasmic and nuclear RNA, respectively.

RNA Pull-Down Assay and Mass Spectrometry

Biotin-labeled sense and antisense HOTAIRM1 sequences were obtained using the Transcript Aid T7 High-Yield Transcription Kit (Thermo Scientific, K0441). A MEGAclearTM Kit (Thermo Scientific) was used to recycle the sequences per the manufacturer’s instructions. The sequences were incubated with cell lysates for 4 h at 4 °C, and then the biotin-labeled RNAs and their binding protein partner were pulled down by streptavidin magnetic beads (Thermo, USA, 20164) at 4 °C overnight. Proteins were separated by electrophoresis and visualized using a Coomassie Blue Staining Kit (Beyotime, China, P0017A). Different bands between sense and antisense HOTAIRM1 were identified using mass spectrometry (Thermo Scientific, Bremen, Germany, 24600).

RNA Immunoprecipitation Assay

An RNA immunoprecipitation (RIP) assay was used to determine whether HOTAIRM1 interacted with HOXA4/HSPA5 using the Magna RIP RNA-binding protein immunoprecipitation kit (Millipore, Billerica, MA, USA, 17–700). Cells were lysed with RIP buffer, and the cell extract was mixed with agarose beads carrying anti-HOXA4/HSPA5 or anti-lgG antibodies. Enrichment of the coprecipitated RNAs was detected by RT-qPCR.

Statistical Analysis

Statistical data from each group were analyzed using GraphPad 6.0. Data are expressed as mean ± standard deviation (SD). Intergroup differences were assessed using Student’s t-test, and multivariate statistical analysis was performed using analysis of variance (ANOVA). Statistical significance was considered at p < 0.05.

RESULTS

HOTAIRM1 was Up-Regulated in the Presence of Ox-LDL

As a first step in elucidating the biological function of HOTAIRM1 in AS, ox-LDL was used to simulate a cell model of AS. To screen the appropriate ox-LDL concentration, the viability and apoptosis were measured in HUVECs stimulated with different ox-LDL concentrations (0, 25, 50, 75, 100, and 125 μg/mL). The results indicated that the introduction of ox-LDL impaired HUVEC viability and contributed to apoptosis compared to the untreated group (Fig. 1a, b). Ultimately, 100 μg/mL was selected for subsequent cell experiments. RT-qPCR was performed to examine whether the presence of ox-LDL affected HOTAIRM1 expression. Ox-LDL enhanced HOTAIRM1 expression in HUVECs in a dose- and time-dependent manner (Fig. 1c, d). In addition, Hotairm1 expression was up-regulated in the aorta of HFD-fed ApoE − / − mice (Fig. 1e). Oil red O and H&E staining represented that the area of arterial tissue damage in the ApoE − / − mice fed with high-fat-diet (HFD) was increased compared with that in the ApoE − / − mice fed with normal diet (ND) (Fig. 1f, g). These findings suggest that HOTAIRM1 is possibly a regulator in the biological regulation of HUVECs.

HOTAIRM1 Depletion Blocked Proliferation and Expedited Apoptosis of HUVECs in Either the Absence or Presence of Ox-LDL

Next, we sought to elucidate the biological role of HOTAIRM1 in HUVECs. Due to ox-LDL can induce HOTAIRM1 expression, ASOs that mediate HOTAIRM1 knockdown were screened; therefore, three individual ASOs were used to knock down their expression, and the silencing efficacy was evaluated by qPCR (Fig. 2a). In order to explore the effects of HOTAIRM1 on the proliferation and apoptosis of HUVEC, firstly, EdU and CCK-8 assays revealed that HOTAIRM1 knockdown remarkably reduced HUVEC proliferation irrespective of ox‐LDL treatment (Fig. 2b, c), and later, flow cytometry assay was performed to determine whether HOTAIRM1 depletion dampened cell proliferation by altering cell cycle progression. The results showed that the cell cycle was dramatically stalled at the G1-G0 phase in HOTAIRM1-silenced HUVECs (Fig. 2d), indicating that HOTAIRM1 deficiency impaired HUVEC proliferation, partly by regulating cell cycle progression. Furthermore, the silencing of HOTAIRM1 strikingly expedited HUVEC apoptosis (Fig. 2e, g). Finally, western blotting results revealed that the expression of cell cycle- and proliferation-related proteins, such as cyclin D1, CKD4, and PCNA, dramatically reduced upon HOTAIRM1 knockdown with a concurrent increase in apoptosis markers, including caspase-3, caspase-9, and Bax (Fig. 2F, H). Thus, HOTAIRM1 may play an important role in modulating HUVEC proliferation and apoptosis.

HOTAIRM1 was Transcriptionally Activated by HOXA4

Mounting evidence suggests that the transcription of several lncRNAs can be regulated by transcription factors (TFs) via direct binding. To tease out potential upstream mediators exquisitely governing HOTAIRM1 in HUVECs, we conducted a motif analysis in the HOTAIRM1 promoter region using JASPAR (http://​jaspar.​binf.​ku.​dk/​). The result implicated that HOXA4 might be a potential TF with probable binding motif “ATTA” (Fig. 3a), indicating that HOTAIRM1 may be modulated by HOXA4 at the transcriptional level. Because HOXA4 appeared to harbor numerous seemingly binding sites predicted by JASPAR in the HOTAIRM1 promoter region (Supplementary File 2), we constructed different HOTAIRM1 promoter truncations to identify the core promoter binding region. The HOTAIRM1 promoter region containing fragments of nucleotides − 2000 to 0 bp, − 1500 to 0 bp, − 1000 to 0 bp, and − 500 to 0 bp were cloned into a PGL3 luciferase reporter construct. When the reporter was co-transfected with a HOXA4 overexpression construct into HUVECs, it displayed remarkable activation by HOXA4 within the promoter region of HOTAIRM1 ranging from − 2000 to 0 bp, − 1500 to 0 bp, and − 1000 to 0 bp. Furthermore, compared to the control group, multiple luciferase activities were enhanced in the HOTAIRM1 promoter regions. Luciferase activity from − 2000 to 0 bp was akin to that from − 1500 to 0 bp, while luciferase activity from − 1500 to 0 bp was higher than that from − 500 to 0 bp. However, there was no influence on the promoter activity of HOTAIRM1 in the region from − 500 to 0 bp (Fig. 3b). A thorough analysis of the aforementioned results showed that HOXA4 promotes HOTAIRM1 transcription. Of note, the core binding region located between − 1500 and − 500 bp was defined, and the JASPAR website again predicted four possible binding sites for HOXA4 in this region, namely, binding site 1 located between − 1130 and − 1123 bp (ACCATTAG), site 2 between − 1109 and − 1102 bp (GTGATTAA), site 3 between − 916 and − 909 bp (ATCATTAT), and site 4 between − 869 and − 862 bp (ATAATCAA) of the HOTAIRM1 promoter region (Fig. 3c). To identify the specific binding site of HOXA4 responsible for its binding to HOTAIRM1, a series of small-scale deletion mutants were generated. As shown in Fig. 3d, HOXA4 overexpression led to a substantial increase in the luciferase activity of HOTAIRM1 promoter with wild-type or sites 1/3/4 mutations in HUVECs, with barely altered luciferase activity of HOTAIRM1 promoter with site 2 or all four sites mutations, thereby revealing that the site 2 with − 1109 to − 1102 bp was important for HOXA4-mediated HOTAIRM1 activation. ChIP-qPCR assay was used to further examine the role of HOXA4 in the HOTAIRM1 promoter region. Interestingly, the occupation of HOXA4 in the HOTAIRM1 promoter region was substantially increased compared to that in the IgG control (Fig. 3e). Collectively, the investigation of transcriptional regulation revealed that HOXA4 directly binds to the HOTAIRM1 promoter and facilitates its transcription.
The regulatory role of HOXA4 on HOTAIRM1 has not been addressed thus far. The siRNAs specifically targeting HOXA4 were generated, and silencing efficiency was evaluated by qPCR and western blotting (Fig. 3f, g). Indeed, knocking down or overexpressing HOXA4 led to decreased or increased HOTAIRM1 expression, respectively (Fig. 3h). Functionally, CCK-8 and EdU assays showed that HOXA4 knockdown dampened the proliferation of HUVECs (Fig. 3i, j). Whether HOTAIRM1 is involved in the contribution of HOXA4 to cell proliferation regulation remains unknown. Rescue experiments showed that HOTAIRM1 overexpression partially counteracted the HOXA4 depletion-mediated proliferation impairment (Fig. 3k, l). Cumulatively, the effect may account for the fact that HOXA4 functions, at least in part, by being contingent upon HOTAIRM1 and indicates that HOXA4 can bind and activate the HOTAIRM1 promoter.

Positive Feedback Signaling Between HOTAIRM1 and HOXA4 in HUVECs

The introduction of ox-LDL led to a dramatic increase in the protein level of HOXA4 in HUVECs (Fig. 4a). To gain insight into how HOTAIRM1 affects cell proliferation, we first examined the localization of HOTAIRM1 through NCBI (https://​www.​ncbi.​nlm.​nih.​gov/​). HOTAIRM1 and HOXA4 are located adjacent to the same chromosome and in opposite directions of transcription (Fig. 4b). Existing data have demonstrated that lncRNAs can function in cis to regulate the expression of adjacent protein-coding genes [31], indicating that HOTAIRM1 may modulate HOXA4 expression, which has yet to be imprinted in HUVECs. HOTAIRM1 knockdown substantially diminished, whereas its overexpression markedly augmented HOXA4 expression at the mRNA and protein levels (Fig. 4c, d). To further elucidate how HOTAIRM1 influences HOXA4 expression, the possible interaction between HOTAIRM1 and HOXA4 was assessed using bioinformatics analyses. Using the RNA–protein interaction prediction (RPISeq) website (http://​pridb.​gdcb.​iastate.​edu/​RPISeq/​), HOXA4 was shown to interact with HOTAIRM1 using the random forest (RF) classifier (= 0.90) and support vector machine (SVM) classifier (= 0.96) (Fig. 4e), suggesting that HOTAIRM1 could interact with HOXA4. Inspired by these observations, follow-up RIP, RNA pull-down, and western blotting experiments were performed to determine whether HOTAIRM1 could interact with HOXA4 in an RBP manner. Unfortunately, the experimental results described here yielded no evidence of the corresponding RBP patterns (Fig. 4f, g). Overall, these discoveries, coupled with our existing knowledge presented above on the transcription promotion of HOTAIRM1 initiated by HOXA4, support the notion that HOTAIRM1 tuned the expression of HOXA4 in a non-RBP manner. Of course, this raises a major intriguing problem in the HOTAIRM1-mediated regulatory axis, which is a candidate for downstream HOTAIRM1-associated protein partners.

Identification of HSPA5 as a Binding Partner for HOTAIRM1 in HUVECs

To delineate the RNA-binding protein landscape in the HOTAIRM1-mediated signaling network, we aimed to identify HOTAIRM1-associated proteins responsible for modulating cell proliferation. The subcellular localization of lncRNAs determines their modes of action [32]. Hence, the subcellular location of HOTAIRM1 was predicted using lncATLAS (https://​lncatlas.​crg.​eu/​), and the results indicated that it was preferentially localized in the nuclei of HUVECs (Fig. 5a). To unequivocally visualize HOTAIRM1 distribution as described by the RNA-FISH assay, it was also principally located in the nucleus of HUVECs (Fig. 5b), which enabled HOTAIRM1 to play an important role at the transcriptional level. Overwhelming evidence has conferred multiple regulatory functions to lncRNAs via interactions with diverse RNA-binding protein partners in the nucleus. Toward this end, HOTAIRM1-interacted proteins were identified and characterized by RNA pull-down assay, followed by mass spectrometry in HUVECs (Fig. 5c). Consequently, 233 proteins associated with HOTAIRM1 were identified using mass spectrometry (Supplementary File 3), and HSPA5 was identified as a prominent HOTAIRM1-binding protein among the top ten hit candidate proteins (Fig. 5d).
To further confirm the HOTAIRM1-HSPA5 interaction, the RNA–protein interaction prediction (RPISeq) website (http://​pridb.​gdcb.​iastate.​edu/​RPISeq/​) was used to predict whether HOTAIRM1 could bind to the HSPA5 protein. The scores for the random forest (RF) classifier and support vector machine (SVM) classifier are 0.7 and 0.85 (both over 0.5) (Fig. 5e), respectively, raising the likelihood that HOTAIRM1 could interact with HSPA5. A follow-up RIP experiment was carried out for the RNA-HSPA5 complex utilizing the HSPA5 antibody to corroborate this speculation. Notably, upon comparison with the IgG-control group, HSPA5 caused a robust HOTAIRM1 enrichment in HUVECs (Fig. 5f). Overall, these data indicate that HOTAIRM1 physically interacts with HSPA5 in HUVECs in an RBP manner.

HOTAIRM1 Regulated HSPA5 Expression in a Transcription-Dependent Manner and its Effect on Proliferation was HSPA5 Partially Dependent

Based on these results, we further investigated whether HOTAIRM1 could regulate HSPA5 expression in HUVECs and found that the protein level of HSPA5 was markedly up-regulated in HUVECs (Fig. 6a), which was in accordance with HOTAIRM1 expression in the presence of ox-LDL. The gain-and-loss-of-function experiments were performed to elucidate the regulatory role of HOTAIRM1 in HSPA5 expression. HOTAIRM1 knockdown mitigated this effect; however, its overexpression increased HSPA5 protein expression (Fig. 6b, c). Down-regulation or up-regulation of HOTAIRM1 remarkably decreased or increased HSPA5 expression at the mRNA level, respectively (Fig. 6d), indicating that HOTAIRM1 modulates the expression of HSPA5 in a transcription-dependent manner. To determine whether HSPA5 is responsible for the role of HOTAIRM1 in HUVEC proliferation, siRNA and overexpression plasmids specifically targeting HSPA5 were utilized to evaluate the protein expression level in HUVECs (Fig. 6e). EdU and CCK-8 assays demonstrated that HSPA5 knockdown markedly retarded HUVEC proliferation (Fig. 6f, g). We then transfected HOTAIRM1-silenced HUVECs with an HSPA5-overexpressing plasmid or empty vector and found that HSPA5 overexpression partly countered the effect of HOTAIRM1 depletion on HUVEC proliferation suppression (Fig. 6h, i), thereby showing that HOTAIRM1-mediated proliferation modulation was dependent on HSPA5. Cell fate determination was tightly orchestrated by a series of molecular signals to unveil the regulatory network of the HOXA4-HOTAIRM1-HSPA5 axis, and the effect of HSPA5 on HOXA4 expression was monitored. HSPA5 knockdown blunted, whereas its overexpression potentiated HOXA4 expression at the mRNA and protein levels in HUVECs (Fig. 6j, k).

DISCUSSION

The discovery of lncRNAs has opened up a new chapter and revolutionized the molecular and cellular biology fields. In recent years, studies on lncRNA biology have shown that they represent a diverse and prevalent group of RNAs and have emerged as appealing orchestrators at the forefront of AS and associated cardiovascular events. AS is a ubiquitous phenomenon that underlies the initiation and progression of several cardiovascular disorders. Aberrant endothelial cell proliferation is closely related to various cardiovascular diseases, including AS; however, whether lncRNA HOTAIRM1 is also involved in this process remains unclear. Ox-LDL has long been associated with AS [33, 34]. For this purpose, the detailed molecular underpinning of HOTAIRM1 in the ox-LDL milieu is important for a deeper understanding of AS. A HUVEC model exposed to ox-LDL was initially used to investigate AS. As such, it is unclear whether HOTAIRM1 is responsive to ox-LDL or whether HOTAIRM1 yields profound insights into its function in response to ox-LDL stimuli in HUVECs. To clarify these issues, we quantified the expression of HOTAIRM1 and demonstrated its effects on the related phenotypes in the absence and presence of ox‐LDL. In this study, a blueprint of functional and mechanistic associations was shaped; ox-LDL-driven HOTAIRM1 was activated by HOXA4 in HUVECs via direct association with its promoter. A barrage of subsequent functional analyses of HOTAIRM1 revealed a reduction of its knockdown on proliferation and noteworthy promotion of apoptosis in HUVECs with or without ox-LDL, suggesting that HOTAIRM1 may be an extraordinarily vital factor in the regulation of proliferation in HUVECs. These findings may open the intriguing perspective of HOTAIRM1 as a plausible therapeutic target for stemming AS.
Little is known about the upstream signals responsible for triggering HOTAIRM1 expression, although HOTAIRM1 is involved in the biological behavior of cells by modulating downstream target genes. Uncovering why HOTAIRM1 is highly expressed in ox-LDL-treated HUVECs is crucial for probing the relevant regulatory circuits. Therefore, we describe HOXA4 as a novel upstream transcription factor that activates HOTAIRM1. HOXA4, a member of the HOX gene family, functions as a transcription factor that regulates the expression of downstream genes to build segments or structures within the body [35]. Mechanistically, the mode of action is mainly via transcriptional activation or repression of target genes [36]. Previous reports have shown that HOXA4 acts as a tumor suppressor in various cancers [37]. A recent report uncovered that HOXA4 affected lung cancer progression by inhibiting the Wnt-b-catenin signaling through GSK3B up-regulation [38]. For the implication of HOXA4 in tumorigenesis, multiple studies have been conducted to elaborate the transcription landscape. For the most part, these findings largely underscored protein-coding genes, delving deeply into their regulatory network, especially at the lncRNA layer, which will be beneficial for unraveling relevant regulatory mechanisms. More specifically, the mechanism by which HOXA4 modulates cell proliferation by regulating the transcription of lncRNAs remains unknown. Therefore, HOXA4 occupies the promoter region of HOTAIRM1 as a transcriptional activator, at least to some extent, which may be partially responsible for HOTAIRM1 up-regulation in HUVECs exposed to ox-LDL. Foremost among these was a key binding site with the base sequence “GTGATTAA” as the HOXA4 response element in the HOTAIRM1 promoter that mediated its transcription activation. Structurally, HOXA4, one of the homeobox family genes, comprises two exons and one intron. Its function depends on an evolutionarily conserved 60 amino-acid homeodomains critical for DNA binding at specifically identified sites covering only four base-pair sequences (TAAT/ATTA/TTAT/ATAA), indicating low functional specificity [39, 40]. The mainly identified binding site base sequence herein is “GTGATTAA,” including the consensus sequence “ATTA,” which is consistent with previous investigation. Functionally, in vitro HOTAIRM1 impairment blunted HUVEC proliferation. Inhibition of proliferation elicited by HOXA4 depletion was partially abrogated by HOTAIRM1 overexpression in HUVECs. In summary, upon HOXA4 reduction, the restored proliferation rates of HUVECs could be attributed to enhanced HOTAIRM1 levels, indicating that HOTAIRM1, a downstream target gene of HOXA4, performs its function through its involvement in HOXA4-mediated proliferation modulation.
A plethora of lncRNAs has been proposed to regulate gene expression in a cis- or trans-manner [41], as exemplified by DEANR1-mediated DE differentiation by regulating its adjoining gene FOXA2 [42]. More than 60% of the endoderm-expressed lncRNAs exhibit coordinated expression changes with nearby protein-coding genes [43]. Thus, we investigated whether HOTAIRM1 facilitated HUVEC proliferation by regulating HOXA4 expression. As mentioned previously, an intriguing discovery was the interplay between HOTAIRM1 and its adjacent protein-coding gene HOXA4 in HUVECs. Scilicet, HOXA4-activated HOTAIRM1, in turn, regulates HOXA4 expression, eventually forming a positive feedback signal to ensure HOTAIRM1 expression. Therefore, we further investigated the possible interaction between HOTAIRM1 and HOXA4 protein in a RBP manner. Notably, the interrogation was at odds with aforehand prediction work, a failure to observe the corresponding regulation between HOTAIRM1 and HOXA4. Accordingly, alternative or additional patterns of HOTAIRM1-mediated HOXA4 regulation cannot be excluded. A previous report showed that HOTAIRM1 recruits EZH2 and SUZ12 to the promoter of the target gene HOXA1, leading to the trimethylation of histone H3K27 and epigenetic silencing of HOXA1 [44]. Therefore, we postulated that HOTAIRM1 might regulate the expression through HOXA4 epigenetic regulation. Besides the probable epigenetic surveillance mechanism, other regulatory mechanisms may exist that are yet to be documented. Although we have thus far failed to unveil convincing evidence, further substantiation may potentially unmask a sparkling aspect of the regulatory networks afforded by HOTAIRM1 at the epigenetic level.
As previously shown, lncRNAs regulate gene expression at the pre-transcriptional, transcriptional, and post-transcriptional levels [45]. lncRNA modulation of cellular processes hinges partly on cellular localization; nuclear lncRNAs are abundant for functionality implicated in chromatin interactions, transcriptional regulation, and RNA processing, whereas cytoplasmic lncRNAs can modulate mRNA stability or translation and affect cellular signaling cascades [46]. The interaction of lncRNAs with functional partners, particularly proteins, remains largely unknown and has recently become an emerging research hotspot. Much effort is being devoted to identifying these interacting partners as a strategy for gaining insight into the molecular mechanisms. lncRNA regulation of genes is in the spotlight because of their enrichment in the nucleus, where they perform various cellular biological processes, often through their ability to directly interact with proteins [4749]. Herein, an initial analysis of the subcellular localization of HOTAIRM1 using online lncATLAS prediction and fluorescent in situ hybridization showed an overwhelming enrichment in the nucleus of HUVECs, suggesting that HOTAIRM1 may regulate gene expression at the transcriptional level. RNA pull-down and liquid chromatography-mass spectrometry (LC–MS) were used to screen HOTAIRM1-associated proteins. These data allowed us to identify HSPA5 as an important binding partner of HOTAIRM1 in HUVECs. RIP assay revealed the binding of HOTAIRM1 to HSPA5. However, an additional aspect related to this binding has emerged: which region of HOTAIRM1 interacts with HSPA5. Evidence of specific binding region is currently lacking, and follow-up deletion mapping experiments will be undertaken to dissect the structural determinant elements for such reciprocal interactions in the future. Beyond mutual interactions, HOTAIRM1 down-regulation or up-regulation also resulted in decreased or increased HSPA5 levels in HUVECs. Similar to HOTAIRM1 expression, HSPA5 was also up-regulated in HUVECs exposed to ox-LDL. Functionally, the silencing of HSPA5 led to impaired HUVEC proliferation, comparable to that acquired by HOTAIRM1 knockdown.
We articulate whether HOTAIRM1-mediated proliferation is required for HSPA5 involvement. HSPA5 overexpression can largely lead to the reverse of the proliferation phenotype observed upon HOTAIRM1’s loss of function, suggesting that HSPA5 is a major functional target of HOTAIRM1. Collectively, HOTAIRM1, a crucial factor in HUVEC proliferation regulation, functions by directly interacting with and modulating HSPA5. Our discovery of such interactions expanded the view of the role of HOTAIRM1, including its functional involvement as both a lncRNA itself and a player that exerts its effect through RNA–protein interaction in HUVECs.
Ultimately, there were several emphases described herein that were worth reiterating: HOTAIRM1, a functional lncRNA identified in this report, was up-regulated in the presence of ox-LDL and was required for HUVEC proliferation. We emphasized that HOTAIRM1 in HUVECs, transcriptionally activated by HOXA4, coupled with HOTAIRM regulation exhibited a positive feedback signaling pattern. Moreover, HOTAIRM1 played its action in HUVEC proliferation modulation, in part, by interacting with and reclining on HSPA5. In conclusion, our findings identified HOTAIRM1 as an orchestrator of HUVEC proliferation and revealed a heretofore uncharacterized regulatory mechanism by which HUVEC proliferation is tightly governed by HOXA4-HOTAIRM1-HSPA5-driven axis. A summary of these mechanisms is presented in Fig. 7. As shown previously and substantiated herein, thorough mechanistic studies of the fundamental actions of HOTAIRM1 and the identification of its associated protein partners are critical for the exploitation and fulfillment of appealing therapeutic targets for AS. This study may pave the way for addressing the momentous question in HOTAIRM1-mediated regulation of HUVEC proliferation and AS. Although very promising, this study still has limitation exists because it principally focused on in vitro culture experiments with little functional data derived from animal evidence. Consequently, visualization of the therapeutic effects of targeting HOTAIRM1 or pertinent molecules could be performed using an animal model of AS, which is certainly subject to in-depth investigation. Thus, trailblazing research remains to be conducted in the field in the future. Although unprecedented advances have been made, many questions and challenges associated with HOTAIRM1 remain. Further thorough investigations may lead to ground-breaking results regarding AS development.

Declarations

Competing interests

The authors declare no competing interests.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Jetzt e.Med zum Sonderpreis bestellen!

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Jetzt bestellen und 100 € sparen!

e.Dent – Das Online-Abo der Zahnmedizin

Online-Abonnement

Mit e.Dent erhalten Sie Zugang zu allen zahnmedizinischen Fortbildungen und unseren zahnmedizinischen und ausgesuchten medizinischen Zeitschriften.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Jetzt bestellen und 100 € sparen!

Literatur
1.
Zurück zum Zitat Libby, P., P.M. Ridker, and A. Maseri. 2002. Inflammation and atherosclerosis. Circulation 105: 1135–1143.CrossRefPubMed Libby, P., P.M. Ridker, and A. Maseri. 2002. Inflammation and atherosclerosis. Circulation 105: 1135–1143.CrossRefPubMed
2.
Zurück zum Zitat Gallino, A., V. Aboyans, C. Diehm, et al. 2014. European Society of Cardiology Working Group on Peripheral Circulation. Non-coronary atherosclerosis. Eur Heart J. 35: 1112–1119.PubMed Gallino, A., V. Aboyans, C. Diehm, et al. 2014. European Society of Cardiology Working Group on Peripheral Circulation. Non-coronary atherosclerosis. Eur Heart J. 35: 1112–1119.PubMed
3.
Zurück zum Zitat Gao, F., X-C. Wang, Z-D. Luo, et al. 2022. LncRNA HOXA11-AS promotes vascular endothelial cell injury in atherosclerosis by regulating the miR-515–5p/ROCK1 axis. J ESC Heart Fail 9: 2259–2271. Gao, F., X-C. Wang, Z-D. Luo, et al. 2022. LncRNA HOXA11-AS promotes vascular endothelial cell injury in atherosclerosis by regulating the miR-515–5p/ROCK1 axis. J ESC Heart Fail 9: 2259–2271.
4.
Zurück zum Zitat Qin, B., Y. Shu, L. Long, H. Li, X. Men, L. Feng, H. Yang, and Z. Lu. 2018. MicroRNA-142-3p Induces atherosclerosis-associated endothelial cell apoptosis by directly targeting Rictor. Cellular Physiology and Biochemistry 47: 1589–1603.CrossRefPubMed Qin, B., Y. Shu, L. Long, H. Li, X. Men, L. Feng, H. Yang, and Z. Lu. 2018. MicroRNA-142-3p Induces atherosclerosis-associated endothelial cell apoptosis by directly targeting Rictor. Cellular Physiology and Biochemistry 47: 1589–1603.CrossRefPubMed
5.
Zurück zum Zitat Li, J., Y. Chen, J. Gao, Y. Chen, C. Zhou, and X. Lin. 2020. Eva1a ameliorates atherosclerosis by promoting re-endothelialization of injured arteries via Rac1/Cdc42/Arpc1b. Cardiovascular Research 117: 450–461.CrossRef Li, J., Y. Chen, J. Gao, Y. Chen, C. Zhou, and X. Lin. 2020. Eva1a ameliorates atherosclerosis by promoting re-endothelialization of injured arteries via Rac1/Cdc42/Arpc1b. Cardiovascular Research 117: 450–461.CrossRef
6.
Zurück zum Zitat Long, X., G. You, Q. Wu, Y. Zhou, Y. Xiao, F. Yu, et al. 2020. HomeoboxC6 affects the apoptosis of human vascular endothelial cells and is involved in atherosclerosis. Journal of Cellular Physiology 236: 1913–1925.CrossRefPubMed Long, X., G. You, Q. Wu, Y. Zhou, Y. Xiao, F. Yu, et al. 2020. HomeoboxC6 affects the apoptosis of human vascular endothelial cells and is involved in atherosclerosis. Journal of Cellular Physiology 236: 1913–1925.CrossRefPubMed
7.
Zurück zum Zitat Evans, J.R., F.Y. Feng, and A.M. Chinnaiyan. 2016. The bright side of dark matter: LncRNAs in cancer. The Journal of Clinical Investigation 126: 2775–2782.CrossRefPubMedPubMedCentral Evans, J.R., F.Y. Feng, and A.M. Chinnaiyan. 2016. The bright side of dark matter: LncRNAs in cancer. The Journal of Clinical Investigation 126: 2775–2782.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Quinn, J.J., and H.Y. Chang. 2016. Unique features of long non-coding RNA biogenesis and function. Nature Reviews Genetics 17: 47–62.CrossRefPubMed Quinn, J.J., and H.Y. Chang. 2016. Unique features of long non-coding RNA biogenesis and function. Nature Reviews Genetics 17: 47–62.CrossRefPubMed
10.
Zurück zum Zitat Lee, S., F. Kopp, T.C. Chang, A. Sataluri, B. Chen, S. Sivakumar, et al. 2016. Noncoding RNA NORAD regulates genomic stability by sequestering PUMILIO proteins. Cell 164: 69–80.CrossRefPubMed Lee, S., F. Kopp, T.C. Chang, A. Sataluri, B. Chen, S. Sivakumar, et al. 2016. Noncoding RNA NORAD regulates genomic stability by sequestering PUMILIO proteins. Cell 164: 69–80.CrossRefPubMed
11.
Zurück zum Zitat Wilusz, J.E., H. Sunwoo, and D.L. Spector. 2009. Long noncoding RNAs: Functional surprises from the RNA world. Genes & Development 23: 1494–1504.CrossRef Wilusz, J.E., H. Sunwoo, and D.L. Spector. 2009. Long noncoding RNAs: Functional surprises from the RNA world. Genes & Development 23: 1494–1504.CrossRef
12.
Zurück zum Zitat Huang, X., X. Xie, P. Liu, L. Yang, B. Chen, C. Song, et al. 2018. Adam12 and lnc015192 act as ceRNAs in breast cancer by regulating miR-34a. Oncogene 37: 6316–6326.CrossRefPubMed Huang, X., X. Xie, P. Liu, L. Yang, B. Chen, C. Song, et al. 2018. Adam12 and lnc015192 act as ceRNAs in breast cancer by regulating miR-34a. Oncogene 37: 6316–6326.CrossRefPubMed
13.
Zurück zum Zitat Wang, P., Y. Xue, Y. Han, L. Lin, C. Wu, S. Xu, et al. 2014. The STAT3-binding long noncoding RNA lnc-DC controls human dendritic cell differentiation. Science 344: 310–313.CrossRefPubMed Wang, P., Y. Xue, Y. Han, L. Lin, C. Wu, S. Xu, et al. 2014. The STAT3-binding long noncoding RNA lnc-DC controls human dendritic cell differentiation. Science 344: 310–313.CrossRefPubMed
14.
Zurück zum Zitat Song, X., Z. Liu, and Z. Yu. 2019. LncRNA NEF is downregulated in triple negative breast cancer and correlated with poor prognosis. Acta Biochim. et. Biophys. Sin. 51: 386–392.CrossRef Song, X., Z. Liu, and Z. Yu. 2019. LncRNA NEF is downregulated in triple negative breast cancer and correlated with poor prognosis. Acta Biochim. et. Biophys. Sin. 51: 386–392.CrossRef
15.
Zurück zum Zitat Chen, L., et al. 2017. Exosomal lncRNA GAS5 regulates the apoptosis of macrophages and vascular endothelial cells in atherosclerosis. PLoS ONE 12: e0185406.CrossRefPubMedPubMedCentral Chen, L., et al. 2017. Exosomal lncRNA GAS5 regulates the apoptosis of macrophages and vascular endothelial cells in atherosclerosis. PLoS ONE 12: e0185406.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Gao. F., X-C. Wang, Z-D. Luo, et al. 2022. LncRNA HOXA11-AS promotes vascular endothelial cell injury in atherosclerosis by regulating the miR-515–5p/ROCK1 axis.[J]. ESC Heart Fail 9: 2259–2271. Gao. F., X-C. Wang, Z-D. Luo, et al. 2022. LncRNA HOXA11-AS promotes vascular endothelial cell injury in atherosclerosis by regulating the miR-515–5p/ROCK1 axis.[J]. ESC Heart Fail 9: 2259–2271.
17.
Zurück zum Zitat Zhang, R., Y. Hao, and J. Zhang. 2022. The lncRNA DANCR promotes development of atherosclerosis by regulating the miR-214-5p/COX20 signaling pathway. Cellular & Molecular Biology Letters 27: 15.CrossRef Zhang, R., Y. Hao, and J. Zhang. 2022. The lncRNA DANCR promotes development of atherosclerosis by regulating the miR-214-5p/COX20 signaling pathway. Cellular & Molecular Biology Letters 27: 15.CrossRef
18.
Zurück zum Zitat Du, H., H. Zhang, R. Yang, et al. 2021. Small interfering RNA-induced silencing lncRNA PVT1 inhibits atherosclerosis via inactivating the MAPK/NF-κB pathway.[J]. Aging (Albany NY). 13: 24449–24463. Du, H., H. Zhang, R. Yang, et al. 2021. Small interfering RNA-induced silencing lncRNA PVT1 inhibits atherosclerosis via inactivating the MAPK/NF-κB pathway.[J]. Aging (Albany NY). 13: 24449–24463.
19.
Zurück zum Zitat Zhang, X., Z. Lian, C. Padden, M.B. Gerstein, J. Rozowsky, M. Snyder, et al. 2009. A myelopoiesis-associated regulatory intergenic noncoding RNA transcript within the human HOXA cluster. Blood 113: 2526–2534.CrossRefPubMedPubMedCentral Zhang, X., Z. Lian, C. Padden, M.B. Gerstein, J. Rozowsky, M. Snyder, et al. 2009. A myelopoiesis-associated regulatory intergenic noncoding RNA transcript within the human HOXA cluster. Blood 113: 2526–2534.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Lin, M., E. Pedrosa, A. Shah, A. Hrabovsky, S. Maqbool, D. Zheng, et al. 2011. RNA-Seq of human neurons derived from iPS cells reveals candidate long noncoding RNAs involved in neurogenesis and neuropsychiatric disorders. PLoS ONE 6: e23356.CrossRefPubMedPubMedCentral Lin, M., E. Pedrosa, A. Shah, A. Hrabovsky, S. Maqbool, D. Zheng, et al. 2011. RNA-Seq of human neurons derived from iPS cells reveals candidate long noncoding RNAs involved in neurogenesis and neuropsychiatric disorders. PLoS ONE 6: e23356.CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Zhang, X., Z. Lian, C. Padden, et al. 2009. A myelopoiesis-associated regulatory intergenic noncoding RNA transcript within the human HOXA cluster.[J]. Blood 113: 2526–34. Zhang, X., Z. Lian, C. Padden, et al. 2009. A myelopoiesis-associated regulatory intergenic noncoding RNA transcript within the human HOXA cluster.[J]. Blood 113: 2526–34.
22.
Zurück zum Zitat Zhang, X., M. Weissman Sherman, E. Newburger Peter. 2014. Long intergenic non-coding RNA HOTAIRM1 regulates cell cycle progression during myeloid maturation in NB4 human promyelocytic leukemia cells.[J]. RNA Biology 11: 777–87. Zhang, X., M. Weissman Sherman, E. Newburger Peter. 2014. Long intergenic non-coding RNA HOTAIRM1 regulates cell cycle progression during myeloid maturation in NB4 human promyelocytic leukemia cells.[J]. RNA Biology 11: 777–87.
23.
24.
Zurück zum Zitat R. Krumlauf. 1994. Hox genes in vertebrate development. Cell 78: 191–201. R. Krumlauf. 1994. Hox genes in vertebrate development. Cell 78: 191–201.
25.
Zurück zum Zitat Li, Q., C. Dong, J. Cui, Y. Wang, and X. Hong. 2018. 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. Journal of Experimental & Clinical Cancer Research 37: 265.CrossRef Li, Q., C. Dong, J. Cui, Y. Wang, and X. Hong. 2018. 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. Journal of Experimental & Clinical Cancer Research 37: 265.CrossRef
26.
Zurück zum Zitat Farina, F.M., S. Serio, I.F. Hall, S. Zani, G.A. Cassanmagnago, M. Climent, E. Civilini, G. Condorelli, M. Quintavalle, and L. Elia. 2022. The epigenetic enzyme DOT1L orchestrates vascular smooth muscle cell-monocyte crosstalk and protects against atherosclerosis via the NF-κB pathway. European Heart Journal 43: 4562–4576.CrossRefPubMed Farina, F.M., S. Serio, I.F. Hall, S. Zani, G.A. Cassanmagnago, M. Climent, E. Civilini, G. Condorelli, M. Quintavalle, and L. Elia. 2022. The epigenetic enzyme DOT1L orchestrates vascular smooth muscle cell-monocyte crosstalk and protects against atherosclerosis via the NF-κB pathway. European Heart Journal 43: 4562–4576.CrossRefPubMed
27.
Zurück zum Zitat Wang, D., Z. Tan, J. Yang, L. Li, H. Li, H. Zhang, H. Liu, Y. Liu, L. Wang, Q. Li, H. Guo. 2023. Perfluorooctane sulfonate promotes atherosclerosis by modulating M1 polarization of macrophages through the NF-κB pathway. Ecotoxicology and Environmental Safety 249:114384. Wang, D., Z. Tan, J. Yang, L. Li, H. Li, H. Zhang, H. Liu, Y. Liu, L. Wang, Q. Li, H. Guo. 2023. Perfluorooctane sulfonate promotes atherosclerosis by modulating M1 polarization of macrophages through the NF-κB pathway. Ecotoxicology and Environmental Safety 249:114384.
28.
Zurück zum Zitat Luo, H., J. Wang, C. Qiao, N. Ma, D. Liu, and W. Zhang. 2015. Pycnogenol attenuates atherosclerosis by regulating lipid metabolism through the TLR4-NF-κB pathway. Experimental & Molecular Medicine 47: e191.CrossRef Luo, H., J. Wang, C. Qiao, N. Ma, D. Liu, and W. Zhang. 2015. Pycnogenol attenuates atherosclerosis by regulating lipid metabolism through the TLR4-NF-κB pathway. Experimental & Molecular Medicine 47: e191.CrossRef
29.
Zurück zum Zitat Krishna, S.M., S.W. Seto, R.J. Jose, J. Li, S.K. Morton, E. Biros, Y. Wang, V. Nsengiyumva, J.H. Lindeman, G.G. Loots, C.M. Rush, J.M. Craig, and J. Golledge. 2017. Wnt Signaling pathway inhibitor sclerostin inhibits angiotensin II-induced aortic aneurysm and atherosclerosis. Arteriosclerosis, Thrombosis, and Vascular Biology 37: 553–566.CrossRefPubMed Krishna, S.M., S.W. Seto, R.J. Jose, J. Li, S.K. Morton, E. Biros, Y. Wang, V. Nsengiyumva, J.H. Lindeman, G.G. Loots, C.M. Rush, J.M. Craig, and J. Golledge. 2017. Wnt Signaling pathway inhibitor sclerostin inhibits angiotensin II-induced aortic aneurysm and atherosclerosis. Arteriosclerosis, Thrombosis, and Vascular Biology 37: 553–566.CrossRefPubMed
30.
Zurück zum Zitat Ramsey Stephen, A., Y. Vengrenyuk, P. Menon. et al. 2014. Epigenome-guided analysis of the transcriptome of plaque macrophages during atherosclerosis regression reveals activation of the Wnt signaling pathway.[J]. PLoS Genetics 10: e1004828. Ramsey Stephen, A., Y. Vengrenyuk, P. Menon. et al. 2014. Epigenome-guided analysis of the transcriptome of plaque macrophages during atherosclerosis regression reveals activation of the Wnt signaling pathway.[J]. PLoS Genetics 10: e1004828.
31.
Zurück zum Zitat Winkler, L., M. Jimenez, T. Zimmer Joshua. et al. 2022. Functional elements of the cis-regulatory lincRNA-p21.[J]. Cell Reports 39: 110687. Winkler, L., M. Jimenez, T. Zimmer Joshua. et al. 2022. Functional elements of the cis-regulatory lincRNA-p21.[J]. Cell Reports 39: 110687.
33.
Zurück zum Zitat Wang, Z-C., K-M. Niu, Y-J Wu. et al. 2022. A dual Keap1 and p47 inhibitor Ginsenoside Rb1 ameliorates high glucose/ox-LDL-induced endothelial cell injury and atherosclerosis.[J]. Cell Death and Disease13: 824. Wang, Z-C., K-M. Niu, Y-J Wu. et al. 2022. A dual Keap1 and p47 inhibitor Ginsenoside Rb1 ameliorates high glucose/ox-LDL-induced endothelial cell injury and atherosclerosis.[J]. Cell Death and Disease13: 824.
34.
Zurück zum Zitat Chen, T., W. Huang, J. Qian. et al. 2020. Macrophage-derived myeloid differentiation protein 2 plays an essential role in ox-LDL-induced inflammation and atherosclerosis.[J]. EBioMedicine 53: 102706. Chen, T., W. Huang, J. Qian. et al. 2020. Macrophage-derived myeloid differentiation protein 2 plays an essential role in ox-LDL-induced inflammation and atherosclerosis.[J]. EBioMedicine 53: 102706.
35.
Zurück zum Zitat Tian, X., et al. 2018. Long non-coding RNA HOXA transcript antisense RNA myeloid specific 1-HOXA1 axis downregulates the immunosuppressive activity of myeloid-derived suppressor cells in lung cancer. Frontiers in Immunology 9: 473.CrossRefPubMedPubMedCentral Tian, X., et al. 2018. Long non-coding RNA HOXA transcript antisense RNA myeloid specific 1-HOXA1 axis downregulates the immunosuppressive activity of myeloid-derived suppressor cells in lung cancer. Frontiers in Immunology 9: 473.CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Krumlauf, R. 2018. Hox genes, clusters and collinearity. International Journal of Developmental Biology 62: 659–663.CrossRefPubMed Krumlauf, R. 2018. Hox genes, clusters and collinearity. International Journal of Developmental Biology 62: 659–663.CrossRefPubMed
37.
Zurück zum Zitat Klausen, C., P.C. Leung, and N. Auersperg. 2009. Cell motility and spreading are suppressed by HOXA4 in ovarian cancer cells: Possible involvement of beta1 integrin. Molecular Cancer Research 7: 1425–1437.CrossRefPubMed Klausen, C., P.C. Leung, and N. Auersperg. 2009. Cell motility and spreading are suppressed by HOXA4 in ovarian cancer cells: Possible involvement of beta1 integrin. Molecular Cancer Research 7: 1425–1437.CrossRefPubMed
38.
Zurück zum Zitat Cheng, S., F. Qian, Q. Huang, L. Wei, Y. Fu, and Y. Du. 2018. HOXA4, down-regulated in lung cancer, inhibits the growth, motility and invasion of lung cancer cells. Cell Death & Disease 9: 465.CrossRef Cheng, S., F. Qian, Q. Huang, L. Wei, Y. Fu, and Y. Du. 2018. HOXA4, down-regulated in lung cancer, inhibits the growth, motility and invasion of lung cancer cells. Cell Death & Disease 9: 465.CrossRef
39.
Zurück zum Zitat Chariot, A., J. Gielen, M.P. Merville, and V. Bours. 1999. The homeodomain-containing proteins–an update on their interacting partners. Biochemical Pharmacology 58: 1851–1857.CrossRefPubMed Chariot, A., J. Gielen, M.P. Merville, and V. Bours. 1999. The homeodomain-containing proteins–an update on their interacting partners. Biochemical Pharmacology 58: 1851–1857.CrossRefPubMed
40.
Zurück zum Zitat Banerjee-Basu, S., and A.D. Baxevanis. 2001. Molecular evolution of the homeodomain family of transcription factors. Nucleic Acids Research 29: 3258–3269.CrossRefPubMedPubMedCentral Banerjee-Basu, S., and A.D. Baxevanis. 2001. Molecular evolution of the homeodomain family of transcription factors. Nucleic Acids Research 29: 3258–3269.CrossRefPubMedPubMedCentral
41.
Zurück zum Zitat Rinn, J.L., and H.Y. Chang. 2012. Genome regulation by long noncoding RNAs. Annual Review of Biochemistry 81: 145–166.CrossRefPubMed Rinn, J.L., and H.Y. Chang. 2012. Genome regulation by long noncoding RNAs. Annual Review of Biochemistry 81: 145–166.CrossRefPubMed
42.
Zurück zum Zitat Jiang, W., Y. Liu, R. Liu. et al. 2015. The lncRNA DEANR1 facilitates human endoderm differentiation by activating FOXA2 expression.[J]. Cell Reports 11: 137–48. Jiang, W., Y. Liu, R. Liu. et al. 2015. The lncRNA DEANR1 facilitates human endoderm differentiation by activating FOXA2 expression.[J]. Cell Reports 11: 137–48.
43.
Zurück zum Zitat Sigova, A.A., A.C. Mullen, B. Molinie, et al. 2013. Divergent transcription of long noncoding RNA/mRNA gene pairs in embryonic stem cells. Proc Natl Acad Sci U S A. 110: 2876–2881.CrossRefPubMedPubMedCentral Sigova, A.A., A.C. Mullen, B. Molinie, et al. 2013. Divergent transcription of long noncoding RNA/mRNA gene pairs in embryonic stem cells. Proc Natl Acad Sci U S A. 110: 2876–2881.CrossRefPubMedPubMedCentral
44.
Zurück zum Zitat Li, F., Y. Xu, X. Xu, S. Ge, F. Zhang, H. Zhang, X. Fan. 2020. lncRNA HotairM1 depletion promotes self-renewal of cancer stem cells through HOXA1-nanog regulation loop.[J]. Molecular Therapy Nucleic Acids 22: 456–470. Li, F., Y. Xu, X. Xu, S. Ge, F. Zhang, H. Zhang, X. Fan. 2020. lncRNA HotairM1 depletion promotes self-renewal of cancer stem cells through HOXA1-nanog regulation loop.[J]. Molecular Therapy Nucleic Acids 22: 456–470.
47.
Zurück zum Zitat Wen, Z., L. Lian, H. Ding, Y. Hu, Z. Xiao, K. Xiong, et al. 2020. LncRNA ANCR promotes hepatocellular carcinoma metastasis through upregulating HNRNPA1 expression. RNA Biology 17: 381–394.CrossRefPubMedPubMedCentral Wen, Z., L. Lian, H. Ding, Y. Hu, Z. Xiao, K. Xiong, et al. 2020. LncRNA ANCR promotes hepatocellular carcinoma metastasis through upregulating HNRNPA1 expression. RNA Biology 17: 381–394.CrossRefPubMedPubMedCentral
48.
Zurück zum Zitat Ferre, F., A. Colantoni, and M. Helmer-Citterich. 2016. Revealing protein-lncRNA interaction. Bioinform. 17: 106–116. Ferre, F., A. Colantoni, and M. Helmer-Citterich. 2016. Revealing protein-lncRNA interaction. Bioinform. 17: 106–116.
49.
Zurück zum Zitat Yao, R.W., Y. Wang, and L.L. Chen. 2019. Cellular functions of long noncoding RNAs. Nature Cell Biology 21: 542–551.CrossRefPubMed Yao, R.W., Y. Wang, and L.L. Chen. 2019. Cellular functions of long noncoding RNAs. Nature Cell Biology 21: 542–551.CrossRefPubMed
Metadaten
Titel
lncRNA HOTAIRM1 Activated by HOXA4 Drives HUVEC Proliferation Through Direct Interaction with Protein Partner HSPA5
verfasst von
Yu Zhou
Qiang Wu
Xiangshu Long
Youfu He
Jing Huang
Publikationsdatum
29.10.2023
Verlag
Springer US
Erschienen in
Inflammation / Ausgabe 1/2024
Print ISSN: 0360-3997
Elektronische ISSN: 1573-2576
DOI
https://doi.org/10.1007/s10753-023-01919-x

Weitere Artikel der Ausgabe 1/2024

Inflammation 1/2024 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

Nach Herzinfarkt mit Typ-1-Diabetes schlechtere Karten als mit Typ 2?

29.05.2024 Herzinfarkt Nachrichten

Bei Menschen mit Typ-2-Diabetes sind die Chancen, einen Myokardinfarkt zu überleben, in den letzten 15 Jahren deutlich gestiegen – nicht jedoch bei Betroffenen mit Typ 1.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.