Open Access

The AMPK‑mTOR axis requires increased MALAT1 expression for promoting granulosa cell proliferation in endometriosis

Retraction in: /10.3892/etm.2024.12528

  • Authors:
    • Xuejie Liu
    • Ping Zhang
    • Yanmin Li
    • Na Zhao
    • Haiyan Han
  • View Affiliations

  • Published online on: November 6, 2020     https://doi.org/10.3892/etm.2020.9453
  • Article Number: 21
  • Copyright: © Liu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

Endometriosis is a common reproductive disorder in women, with a global prevalence of 10‑15%. Long noncoding RNAs (lncRNAs) are critical to gene transcription, cell cycle modulation and immune response. The lncRNA metastasis‑associated lung adenocarcinoma transcript 1 (MALAT1) reportedly mediates autophagy of endometrial stromal cells in endometriosis. The present study aimed to evaluate the role and mechanism of MALAT1 in granulosa cells (GCs) in endometriosis. Consequently, MALAT1 expression was upregulated in GCs obtained from patients with endometriosis and in the steroidogenic human granulosa‑like tumor cell line KGN. However, MALAT1 knockdown consequently decreased the proliferation and viability of these cells, as determined by MTT and 5‑ethynyl‑2'‑deoxyuridine staining assays. Both Annexin V‑fluorescein isothiocyanate/propidium iodide flow cytometry and western blotting performed to detect proapoptotic factors indicated that MALAT1 depletion might promote KGN cell apoptosis. Furthermore, MALAT1 knockdown increased GC autophagy, as evidenced by microtubule‑associated protein 1A/1B‑light chain 3 (LC3) cleavage upregulation and p62 degradation. In addition, although 5'-AMP‑activated protein kinase (AMPK) mRNA expression and protein levels decreased in GCs obtained from patients with endometriosis and KGN cells, MALAT1 knockdown restored AMPK levels. However, addition of BML‑275 (MALAT1 inhibitor) to MALAT1‑knockdown KGN cells recovered their viability and proliferative capacity and simultaneously reduced their apoptotic and autophagic capacity. Therefore, MALAT1 may regulate GC proliferation via AMPK‑mTOR‑mediated cell apoptosis and autophagy.

Introduction

Endometriosis is prevalent in reproductive women, with a global prevalence of 10-15% (1). This disorder is responsible for various gynecological conditions, such as dysmenorrhea, vulvodynia and chronic pelvic pain (1). Furthermore, 25-50% of infertile women reportedly suffer from endometriosis (2). Despite the underlying interaction between endometriosis and infertility, this interaction has not been completely elucidated. Reportedly, endometriosis-involved infertility may be due to various factors including distorted pelvic anatomy (3). Based on laparoscopic findings, endometriosis is categorized into four different stages (3). In stage I/II (1-5 or 6-15 points), endometriosis is considered ‘minimal or mild’. Endometrial implants are small, few in number and shallow. These two stages do not correspond to pain and discomfort levels. In stage III/IV (16-40 or >40 points), endometriosis is considered ‘moderate or severe’ (3). In addition to a number of deep endometrial implants, there are large cysts in at least one ovary and numerous dense adhesions throughout the pelvic region (4). Regarding the management of endometriosis, surgical treatment is frequently adopted, but it typically results in ovary removal, thereby permanently terminating oocyte development (5). The ovarian follicular microenvironment and maternal signals are involved in folliculogenesis and oocyte development (5). Therefore, the characteristics of granulosa cells (GCs) are first investigated to elucidate their role in the pathogenesis of endometriosis-involved infertility.

An increasing number of studies have focused on the role of noncoding RNAs (6-8). Long noncoding RNAs (lncRNAs) can be classified into lncRNAs measuring >200 nucleotides in length (6). LncRNAs are critical to biological events, including gene transcription (7,8). Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is a type of lncRNA located in the human chromosome 11q13.1(9); and is upregulated in several tumors, such as non-small cell lung cancer, liver cancer and hepatocellular cancer (10-12). Moreover, MALAT1 downregulation resulted in reduced cell proliferation in human osteosarcoma and a decrease in proliferating cell nuclear antigen expression (13). Furthermore, abnormal changes in MALAT1 expression are associated with bladder cancer (14). Recent research has indicated that MALAT1 expression significantly increased in the ectopic endometrium of patients with endometriosis (15). MALAT1 also mediated hypoxia-induced prosurvival autophagy of endometrial stromal cells in endometriosis (16,17). However, another study suggested that MALAT1 levels was significantly lower in GCs obtained from infertile women with advanced endometriosis, compared to healthy controls. MALAT1 knockdown inhibited proliferation and cell cycle progression in KGN cells by inducing an increase in p21, p53, phosphorylated (p)-ERK1/2 and p-c-Jun N-terminal protein kinase protein levels, and a decrease in cyclin-dependent kinase 2, cyclin D1 and p-P38 mitogen-activated protein kinase protein levels (18). However, the role and mechanism of MALAT1 in endometriosis remain unclear.

Autophagy is a highly conservative pathway that protects eukaryotic cells from harmful products (19). In addition to the critical roles of autophagy in aging, neurodegenerative diseases and cancer, the two-sided role of autophagy in cancer has gained considerable attention (20-22). Regarding tumor cells, autophagy can alter their phenotype to enhance mobility and provide essential nutrition to cells (23). By modulating autophagy, MALAT1 affects a series of biological characteristics in pancreatic cancer (24). Autophagy is promoted by 5'-AMP-activated protein kinase (AMPK), which is a key energy sensor that regulates cell metabolism to maintain energy homeostasis (25). Conversely, autophagy is inhibited by mammalian target of rapamycin (mTOR), a central cell growth regulator that integrates growth factor and nutrient signals (25). The mechanism by which MALAT1 affects endometriosis by regulating GC apoptosis and autophagy is poorly investigated. The aim of the present study was to demonstrate the role and mechanism of action of MALAT1 in the properties of GC obtained from patients with endometriosis.

Materials and methods

Ethical approval and consent

The study protocol was approved by the Ethics Committee of Affiliated Hospital of Weifang Medical University (approval no. NCT02115879). Oral informed consent was obtained from the participants of the study.

Patient samples and inclusion criteria

GC samples were collected from patients (31.8-43.3 years old, mean 37.5 years old) (with or without endometriosis) who underwent in vitro fertilization (IVF) or intracytoplasmic sperm injection (ICSI) at the Affiliated Hospital of Weifang Medical University between April 2013 and December 2018. Endometriosis was diagnosed according to laparoscopic or laparotomy findings. Patients with adenomyosis or malignant neoplasm were excluded. Endometriosis stage was assessed in accordance with the revised American Society for Reproductive Medicine criteria (26). Participants that conformed to the following inclusion criteria were included: Basal follicle-stimulating hormone levels of <10 IU/ml, basal estradiol levels of <50 pg/ml, antral follicle count (AFC) of >5 and regular menstrual cycle. At 34-36 h prior to oocyte collection, controlled ovarian stimulation was adopted to perform IVF or ICSI. Basal serum sex hormone levels in patients on days 2-3 of the menstrual cycle was measured using the human follicle stimulating hormone ELISA kit (cat. no. ab108641; Abcam) and estradiol ELISA kit (cat. no. ab108667; Abcam).

GC collection

After collection, samples were purified as described previously (27). Under transvaginal ultrasound guidance, samples were punctured to collect follicular fluid, and follicles with a diameter of >10 mm were aspirated, followed by centrifugation at 400 x g for 10 min at 4˚C. Using cell pellets, a cell suspension was prepared in a DMEM medium (10569010, Thermo Fisher Scientific, Inc.) and layered over 50% Percoll (P1644; Sigma-Aldrich; Merck KGaA), followed by centrifugation at 400 x g for 20 min at 4˚C. GCs located on the interface were collected and washed in PBS. The resultant pellet was used to perform following analysis.

Cell line and culture

The KGN cells, derived from human ovarian granulosa-like tumour, were cultured as previously described (28). KGN cells were regularly cultured (37˚C, 5% CO2) in DMEM (10569010; Thermo Fisher Scientific, Inc.) supplemented with 10% FBS (HyClone; Cytiva). For BML-275 treatment, KGN cells were incubated with 5 nM BML-275 for 8 h

Transfection

MALAT1 knockdown was demonstrated using MALAT1 short hairpin RNA (100 pmol/l, shRNA; sequence: 5'-AATGGAATATGTGTCTGGAGG-3'). The sequence of negative control shRNA is 5'-ACGACGTCAGCTGGTGCATGT-3' (100 pmol/l). At 48 h post transfection, cells were applied for the following experiments. Briefly, GCs were inoculated on six-well plates (4x104 cells/ml) and directly incubated with the MALAT1 or NC shRNA construct. After 2-3 weeks of culture in puromycin, cells with stable MALAT1 knockdown were validated using western blotting.

MTT assay

Cell survival was evaluated using an MTT assay. In brief, after MTT treatment (0.5 mg/ml) at 20 µl, cells were further incubated with 150 µl DMSO until the formazan dye was dissolved. Formazan absorbance was determined at a wavelength of 490 nm using a microplate reader (Tecan Group, Ltd.).

5-ethynyl-2'-deoxyuridine (EdU) staining assay

Following inoculation in a 24-well plate (1x104 cells/ml), cells were incubated with an EdU stock solution (Invitrogen; Thermo Fisher Scientific, Inc.) for 2 h at 48 h after transfection. Incubation was terminated by washing the cells in PBS, and cells were fixed in 4% paraformaldehyde for 20 min at 25˚C, permeabilized in 0.3% Triton X-100 for 10 min at 25˚C and blocked in 10% FBS (HyClone; GE Healthcare Life Sciences) for 60 min. Cells were sequentially probed with rabbit anti-EdU primary antibody (cat. no. ab6326; Abcam) at a dilution of 1:200 at 4˚C overnight and with secondary Cy3-conjugated antibody (1:500; cat. no. ab98416; Abcam) for 2 h. Incorporated EdU was detected using a Click-iT® EdU Kit (Thermo Fisher Scientific, Inc.). Fluorescent signals were detected under a fluorescent microscope (Olympus Corporation). Image analysis was performed using Image-Pro Plus software v6.0 (number of fields=10; magnification, x40) (Media Cybernetics, Inc.).

Evaluation of early apoptosis

Apoptosis was assessed using the Annexin V-fluorescein isothiocyanate (FITC)/PI detection kit (cat. no. ab14085; Abcam). The experiment was performed according to the manufacturer's protocol. Briefly, cells (1x104) were sequentially incubated with 20 µl PI binding buffer (50 µg/ml) and 10 µl Annexin V-FITC (10 µg/ml) in the dark. Subsequently, apoptosis was assessed using BD FACSCalibur™ flow cytometer (BD Biosciences) equipped with FACS Diva version 6.0 software (BD Biosciences) to determine early apoptosis cells.

Western blotting

Cell lysate was prepared with RIPA buffer (R0278; Sigma-Aldrich; Merck KGaA). Subsequently, protein concentration was measured using the Bicinchoninic Acid Protein Quantitation kit (cat. no. ab102536; Abcam). A total of 20 µg protein were separated on 10% SDS-PAGE (Bio-Rad Laboratories, Inc.) and electrically transferred onto PVDF membranes (EMD Millipore), followed by blocking in 5% BSA (A4161; Sigma-Aldrich; Merck KGaA) for 1 h at 25˚C. Proteins on PVDF membranes were sequentially probed with primary antibody overnight at 4˚C, followed by secondary antibody incubation for 1 h at 25˚C. The information for antibodies are as follows: Primary antibodies, Bax (1:1,000; cat. no. ab216494; Abcam), cleaved caspase-3 (1:1,000; cat. no. ab2302; Abcam), caspase-3 (1:1,000; cat. no. ab4051; Abcam), β-actin antibody (1:5,000; cat. no. ab8227; Abcam), LC3B (1:500; cat. no. ab51520; Abcam), p62 (1:1,000; cat. no. ab91526; Abcam), AMPK (1:1,000; cat. no. ab3759; Abcam), phosphor-AMPK (1:200; cat. no. ab23875; Abcam) and mTOR (1:1,000; cat. no. ab2732; Abcam); Secondary antibodies, HRP-conjugated rabbit or mouse antibodies (1:1,000; cat. nos. ab6721 and ab6728, respectively; Abcam). β-actin served as the internal reference. Protein bands were developed on the membrane. Protein bands were visualized using an ECL Western Blotting kit (Thermo Fisher Scientific, Inc.), and quantified using the C-DiGit Blot Scanner (LI-COR Biosciences).

RNA isolation and quantitative PCR (qPCR)

RNA from the transfected cells or tissues was extracted using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. Total RNA (1 µg) was reverse transcribed into cDNA using the miScript Reverse Transcription kit for incubation at 55˚C for 10 min (Qiagen, Inc.), according to the manufacturer's protocol. mRNAs were quantified using SYBR-Green (Roche Applied Science), with GADPH as the internal reference. qPCR was performed on a 20-µl system using the SYBR-Green PCR Master Mix (Thermo Fisher Scientific, Inc.). The following thermocycling conditions were used for the qPCR: Initial denaturation at 95˚C for 5 min; 40 cycles of 95˚C for 10 sec, 57˚C for 30 sec and 72˚C for 10 sec. Target values were calculated using the 2-ΔΔCq method (mean of control samples) (29) and the values were normalized to the negative control groups. The primers used were as follows: MALAT1, forward, 5'-GAATTGCGTCATTTAAAGCCTAGTT-3', and reverse, 5'-GTTTCATCCTACCACTCCCAATTAAT-3'; AMPK, forward, 5'-AGAGCCCCATCTGTCCTCTC-3' and reverse, 5'-ACTGGTAGTCTGCAAAACCAAA-3'; and GAPDH, forward, 5'-GGAGCGAGATCCCTCCAAAAT-3' and reverse, 5'-GGCTGTTGTCATACTTCTCATGG-3'.

Immunofluorescence assay (IFA)

Regarding the microtubule-associated proteins 1A/1B light chain 3B (LC3B) puncta assay, the KGN cells in a 24-well plate (1x104 cells/ml), were transfected with green fluorescent protein (GFP)-LC3B expression constructs (Addgene, Inc.). Images were captured using a fluorescent microscope (Olympus Corporation; magnification, x400) (30). Moreover, the GFP puncta in 10 different microscopic fields was assessed.

AMPK activity

Following the aforementioned transfections, AMPK activity was measured using a kinase assay buffer (V9101; Promega Corporation) with the AMP-[γ-32P] ATP mixture and SAMS peptide (ab120182; Abcam). The reaction was terminated using a phosphocellulose paper, and AMPK activity was measured. Briefly, samples were incubated with SAMS peptide at 30˚C for 20 min and then spotted with 15 µl of samples on a disc of P81 Whatman phosphocellulose paper and allowed to absorb (1-3 sec). Subsequently, the discs were washed in a stirring bath containing 200 ml 1% phosphoric acid, acetone and petroleum ether. The AMPK activity was counted using Cerenkov counting (31).

Statistical analysis

Data was presented as the mean ± standard deviation. Comparison between multiple groups or two groups was performed using one-way ANOVA with Tukey's post-hoc test or Student's t-test, respectively. P<0.05 was considered to indicate a statistically significant difference.

Results

MALAT1 is expressed in GCs in endometriosis

Compared with normal GCs, MALAT1 expression significantly increased in GCs in endometriosis (Fig. 1A). MALAT1 expression obtained from patients with stage III/IV endometriosis was significantly higher compared with patients with stage I/II endometriosis (Fig. 1B). In addition, as a GC tumor-derived cell line, KGN is commonly used to study the molecular mechanism underlying endometriosis (18,32,33); thus, MALAT1 expression was compared between KGN cells and normal GCs. MALAT1 expression was significantly upregulated in KGN cells compared with normal GCs (Fig. 1C); therefore, MALAT1 may be involved in GC activity in endometriosis.

MALAT1 knockdown inhibits GC proliferation in endometriosis

Following transfection with MALAT1, MALAT1 expression significantly decreased compared with the NC group (Fig. 2A). MTT assay indicated that at 24-96 h after transfection, KGN cell proliferation significantly decreased compared with the NC group (Fig. 2B). EdU staining assay demonstrated that MALAT1 knockdown significantly reduced EdU-positive cell levels compared with that the NC group (Fig. 2C). Therefore, MALAT1 may play a role in GC cell proliferation. In addition, flow cytometry revealed that MALAT1 knockdown led to an increase in the apoptotic cell number, levels of Bax, total caspase-3, and cleaved caspase-3, which are biomarkers of apoptosis (Fig. 3A and B). Additionally, whether MALAT1 knockdown caused the autophagy of KGN cells was also investigated. The location of LC3B protein following MALAT1 knockdown was traced using IFA. LC3B (green) accumulated after MALAT1 shRNA transfection (Fig. 4A). MALAT1 knockdown enhanced the autophagy rate of KGN cells, as indicated by increased LC3B biosynthesis and processing (reduced LC3B I and II levels) and p62 degradation (Fig. 4B). Therefore, MALAT1 may inhibit autophagy of KGN cells, which is partially responsible for its facilitative effect on GC proliferation.

MALAT1 reduces AMPK1 expression in GCs

MALAT1 targets the AMPK-mTOR pathway (18,34); thus, the AMPK levels in GCs obtained from patients with endometriosis and in KGN cells were determined. AMPK mRNA levels were significantly decreased in GCs and KGN cells compared with normal GCs (Fig. 5A and B). In addition, the effect of MALAT1 knockdown on AMPK1 expression in KGN cells was analysed using western blotting. Compared with the NC group, the expression levels of total and phosphorylated AMPK1 were significantly upregulated by MALAT1 knockdown, but its downstream target mTOR was significantly downregulated (Fig. 5C). In addition, MALAT1 knockdown significantly increased AMPK1 activity (Fig. 5D). Therefore, AMPK1 expression increased following MALAT1 knockdown.

Inhibition of AMPK1 attenuates the effect of MALAT1 knockdown on KGN cell viability

To determine whether AMPK1 deactivation suppresses the influence of MALAT1 on KGN cell viability, apoptosis and autophagy, AMPK1 activity in KGN cells was inhibited by BML-275 treatment. AMPK1 activity detection assay was conducted to confirm AMPK1 deactivation (Fig. 6A). Western blotting demonstrated that compared with the knockdown (KD)/vehicle (0.1% DMSO) group, phosphorylated AMPK1 levels significantly downregulated, however, mTOR expression significantly upregulated due to BML-275 treatment of MALAT1-knockdown KGN cells (Fig. 6B). In addition, with the inhibition of AMPK1, KGN cell proliferation (which was prevented by MALAT1 knockdown) recovered, as demonstrated by EdU staining assay (Fig. 6C).

To evaluate the role of AMPK1 in apoptosis, KGN apoptosis was assessed by flow cytometry. Compared with the KD/Vehicle group, BML-275 treatment significantly decreased the number of apoptotic KGN cells that underwent transfection with MALAT1 knockdown (Fig. 6D). In addition, BML-275 treatment downregulated autophagy, as evidenced by the attenuated LC3B expression (Fig. 6E). Therefore, AMPK1 deactivation may restore proliferation, which was inhibited by MALAT1 knockdown via apoptosis and autophagy.

Discussion

The results of the present study demonstrated that MALAT1 is upregulated in GCs obtained from patients with endometriosis and serves a positive role in GC viability by mediating AMPK-mTOR pathway activation. In addition, MALAT1 knockdown facilitated GC autophagy and apoptosis, thereby reducing GC proliferation. Additionally, MALAT1 expression was negatively associated with AMPK mRNA expression. AMPK upregulation and mTOR downregulation by MALAT1 knockdown may be caused by the direct or indirect interaction between AMPK and MALAT1; however, this notion requires further investigation.

Endometriosis typically results in infertility caused by genetic anomaly (2). LncRNAs, initially considered as spurious transcriptional noise, serve key roles in biological events, such as apoptosis and inflammation (35,36). MALAT1 was first discovered in lung cancer (37), and may be involved in cancer development by altering the characteristics of tumor cells (10-12). In addition, abnormal MALAT1 expression is one of the prognostic factors for cancer (38). The present study showed that MALAT1 expression was upregulated in endometriosis samples and cell lines, which is consistent with previous findings (18). A previous study also evidenced that MALAT1 knockdown in KGN cells inhibited cell proliferation and cell cycle progression which mainly focused on cell death and cell cycle-associated signal pathways, such as P53, phosphorylated ERK1/2, and phosphorylated c-JNK protein levels, as well as CDK2, cyclin D1 and phosphorylated P38 MAPK protein levels (18), whereas the present study mainly focused on apoptosis- and autophagy-related signal pathway-associated genes, such as caspase-3, mTOR and LC3 signals.

Autophagy is crucial to pathophysiological processes (39), and cancer cells often block apoptotic pathways (40). The sensitivity of cells to apoptosis or autophagy may differ from the status of different lncRNAs. For instance, in response to high glucose levels, the expression of the lncRNA H19 decreased, thereby initiating autophagy by repressing the PI3K/AKT/mTOR pathway (41). The lncRNA growth arrest-specific 5 (GAS5) can modulate the sensitivity of non-small-cell lung cancer cells to cisplatin treatment, thereby repressing autophagy (42). Specifically, GAS5 is upregulated in osteoarthritis to trigger apoptosis (43). AMPK is activated by mTOR inhibition or serine/threonine-protein kinase ULK1 phosphorylation (44), whereas, the mTOR complex modulates various intracellular pathways; therefore, it participates in several biological events, such as autophagy (45). However, mTOR is inhibited by the regulatory-associated protein of mTOR or raptor phosphorylation, which is attained by AMPK activation (44,45). Upon activation, AMPK mediated a proapoptotic effect to inhibit mTOR complex 1expression or increase proapoptotic protein levels (45). In the present study, MALAT1 knockdown increased GC autophagy and apoptosis in KGN cells; hence, MALAT1 may have a protective role in cell survival. Meanwhile, AMPK upregulation by MALAT1 depletion promoted GC autophagy and apoptosis, whereas its inhibition by BML-275 treatment restored cell survival; these results are consistent with those of previous studies (44,45). Rapamycin was widely reported to induce autophagy in different cell lines (45). A number of studies have demonstrated that rapamycin inhibition of mTOR resulted in both reduced granulosa cell proliferation and reduced follicle growth in vitro via mediating mTOR, Akt and cyclin D2 expression, and ERK phosphorylation (46-53). The lack of rapamycin experiments is a limitation of the present study and will be fully researched in future studies.

In conclusion, MALAT1 expression is significantly upregulated in endometriosis GCs associated with AFC. In addition, MALAT1 knockdown facilitated GC autophagy and apoptosis, thereby reducing their proliferation. Additionally, lncRNA MALAT1 expression is negatively associated with AMPK mRNA expression. MALAT1 alteration in GCs may reflect the status of endometriosis, thereby providing novel insights into the pathogenesis of endometriosis.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

XL and HH conceived the study and designed the experiments. PZ, YL and NZ contributed to data collection, performed data analysis and interpreted the results. XL wrote the manuscript. HH contributed to critical revision of article. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The protocol was approved by the Ethics Committee of Affiliated Hospital of Weifang Medical University (approval no. NCT02115879). Informed consent was obtained from all participants included in the study.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Olive DL and Pritts EA: Treatment of endometriosis. N Engl J Med. 345:266–275. 2001.PubMed/NCBI View Article : Google Scholar

2 

Missmer SA, Hankinson SE, Spiegelman D, Barbieri RL, Marshall LM and Hunter DJ: Incidence of laparoscopically confirmed endometriosis by demographic, anthropometric, and lifestyle factors. Am J Epidemiol. 160:784–796. 2004.PubMed/NCBI View Article : Google Scholar

3 

De Ziegler D, Borghese B and Chapron C: Endometriosis and infertility: Pathophysiology and management. Lancet. 376:730–738. 2010.PubMed/NCBI View Article : Google Scholar

4 

Medicine PCotASfR: Endometriosis and infertility. Fertility Sterility. 82:40–45. 2004.PubMed/NCBI View Article : Google Scholar

5 

Coticchio G, Dal Canto M, Mignini Renzini M, Guglielmo MC, Brambillasca F, Turchi D, Novara PV and Fadini R: Oocyte maturation: Gamete-somatic cells interactions, meiotic resumption, cytoskeletal dynamics and cytoplasmic reorganization. Hum Reprod Update. 21:427–454. 2015.PubMed/NCBI View Article : Google Scholar

6 

Bertone P, Stolc V, Royce TE, Rozowsky JS, Urban AE, Zhu X, Rinn JL, Tongprasit W, Samanta M, Weissman S, et al: Global identification of human transcribed sequences with genome tiling arrays. Science. 306:2242–2246. 2004.PubMed/NCBI View Article : Google Scholar

7 

Ørom UA, Derrien T, Beringer M, Gumireddy K, Gardini A, Bussotti G, Lai F, Zytnicki M, Notredame C, Huang Q, et al: Long noncoding RNAs with enhancer-like function in human cells. Cell. 143:46–58. 2010.PubMed/NCBI View Article : Google Scholar

8 

Szymanski M, Barciszewska MZ, Erdmann VA and Barciszewski J: A new frontier for molecular medicine: Noncoding RNAs. Biochim Biophys Acta. 1756:65–75. 2005.PubMed/NCBI View Article : Google Scholar

9 

Ji P, Diederichs S, Wang W, Böing S, Metzger R, Schneider PM, Tidow N, Brandt B, Buerger H, Bulk E, et al: MALAT-1, a novel noncoding RNA, and thymosin β4 predict metastasis and survival in early-stage non-small cell lung cancer. Oncogene. 22:8031–8041. 2003.PubMed/NCBI View Article : Google Scholar

10 

Schmidt LH, Spieker T, Koschmieder S, Schäffers S, Humberg J, Jungen D, Bulk E, Hascher A, Wittmer D, Marra A, et al: The long noncoding MALAT-1 RNA indicates a poor prognosis in non-small cell lung cancer and induces migration and tumor growth. J Thorac Oncol. 6:1984–1992. 2011.PubMed/NCBI View Article : Google Scholar

11 

Guerrieri F: Long non-coding RNAs era in liver cancer. World J Hepatol. 7(1971)2015.PubMed/NCBI View Article : Google Scholar

12 

Lai MC, Yang Z, Zhou L, Zhu QQ, Xie HY, Zhang F, Wu LM, Chen LM and Zheng SS: Long non-coding RNA MALAT-1 overexpression predicts tumor recurrence of hepatocellular carcinoma after liver transplantation. Med Oncol. 29:1810–1816. 2012.PubMed/NCBI View Article : Google Scholar

13 

Dong Y, Liang G, Yuan B, Yang C, Gao R and Zhou X: MALAT1 promotes the proliferation and metastasis of osteosarcoma cells by activating the PI3K/Akt pathway. Tumor Biol. 36:1477–1486. 2015.PubMed/NCBI View Article : Google Scholar

14 

Li C, Cui Y, Liu LF, Ren WB, Li QQ, Zhou X, Li YL, Li Y, Bai XY and Zu XB: High expression of long noncoding RNA MALAT1 indicates a poor prognosis and promotes clinical progression and metastasis in bladder cancer. Clin Genitourin Cancer. 15:570–576. 2017.PubMed/NCBI View Article : Google Scholar

15 

Liang Z, Chen Y, Zhao Y, Xu C, Zhang A, Zhang Q, Wang D, He J, Hua W and Duan P: miR-200c suppresses endometriosis by targeting MALAT1 in vitro and in vivo. Stem Cell Res Ther. 8(251)2017.PubMed/NCBI View Article : Google Scholar

16 

Lelli A, Nolan KA, Santambrogio S, Gonçalves AF, Schönenberger MJ, Guinot A, Frew IJ, Marti HH, Hoogewijs D and Wenger RH: Induction of long noncoding RNA MALAT1 in hypoxic mice. Hypoxia (Auckl). 3(45)2015.PubMed/NCBI View Article : Google Scholar

17 

Liu H, Zhang Z, Xiong W, Zhang L, Du Y, Liu Y and Xiong X: Long non-coding RNA MALAT 1 mediates hypoxia-induced pro-survival autophagy of endometrial stromal cells in endometriosis. J Cell Mol Med. 23:439–452. 2019.PubMed/NCBI View Article : Google Scholar

18 

Li Y, Liu YD, Chen SL, Chen X, Ye DS, Zhou XY, Zhe J and Zhang J: Down-regulation of long non-coding RNA MALAT1 inhibits granulosa cell proliferation in endometriosis by up-regulating P21 via activation of the ERK/MAPK pathway. Mol Hum Reprod. 25:17–29. 2019.PubMed/NCBI View Article : Google Scholar

19 

Galluzzi L, Pietrocola F, Levine B and Kroemer G: Metabolic control of autophagy. Cell. 159:1263–1276. 2014.PubMed/NCBI View Article : Google Scholar

20 

Martinez-Lopez N, Athonvarangkul D and Singh R: Autophagy and aging. Adv Exp Med Biol. 847:73–87. 2015.PubMed/NCBI View Article : Google Scholar

21 

Nixon RA: The role of autophagy in neurodegenerative disease. Nat Med. 19:983–997. 2013.PubMed/NCBI View Article : Google Scholar

22 

Levy JMM, Towers CG and Thorburn A: Targeting autophagy in cancer. Nat Rev Cancer. 17:528–542. 2017.PubMed/NCBI View Article : Google Scholar

23 

White E, Mehnert JM and Chan CS: Autophagy, metabolism, and cancer. Clin Cancer Res. 21:5037–5046. 2015.PubMed/NCBI View Article : Google Scholar

24 

Yuan P, Cao W, Zang Q, Li G, Guo X and Fan J: The HIF-2α-MALAT1-miR-216b axis regulates multi-drug resistance of hepatocellular carcinoma cells via modulating autophagy. Biochem Biophys Res Commun. 478:1067–1073. 2016.PubMed/NCBI View Article : Google Scholar

25 

Kim J, Kundu M, Viollet B and Guan KL: AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat Cell Biol. 13:132–141. 2011.PubMed/NCBI View Article : Google Scholar

26 

Revised American Society for Reproductive Medicine classification of endometriosis. Fertil Steril 67: 817-821, 1997.

27 

Kaur S, Archer KJ, Devi MG, Kriplani A, Strauss JF III and Singh R: Differential gene expression in granulosa cells from polycystic ovary syndrome patients with and without insulin resistance: Identification of susceptibility gene sets through network analysis. J Clin Endocrinol Metab. 97:E2016–E2021. 2012.PubMed/NCBI View Article : Google Scholar

28 

Nishi Y, Yanase T, Mu Y, Oba K, Ichino I, Saito M, Nomura M, Mukasa C, Okabe T, Goto K, et al: Establishment and characterization of a steroidogenic human granulosa-like tumor cell line, KGN, that expresses functional follicle-stimulating hormone receptor. Endocrinology. 142:437–345. 2001.PubMed/NCBI View Article : Google Scholar

29 

Yang J, Takahashi Y, Cheng E, Liu J, Terranova PF, Zhao B, Thrasher JB, Wang HG and Li B: GSK-3beta promotes cell survival by modulating Bif-1-dependent autophagy and cell death. J Cell Sci. 123:861–870. 2010.PubMed/NCBI View Article : Google Scholar

30 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Method. 25:402–408. 2001.PubMed/NCBI View Article : Google Scholar

31 

Al-Masri MS: Cerenkov counting technique. J Radioanalytical Nuclear Chem. 207:205–213. 1996.

32 

Deura I, Harada T, Taniguchi F, Iwabe T, Izawa M and Terakawa N: Reduction of estrogen production by interleukin-6 in a human granulosa tumor cell line may have implications for endometriosis-associated infertility. Fertil Steril. 83 (Suppl 1):1086–1092. 2005.PubMed/NCBI View Article : Google Scholar

33 

Lin X, Dai Y, Tong X, Xu W, Huang Q, Jin X, Li C, Zhou F, Zhou H, Lin X, et al: Excessive oxidative stress in cumulus granulosa cells induced cell senescence contributes to endometriosis-associated infertility. Redox Biol. 30(101431)2020.PubMed/NCBI View Article : Google Scholar

34 

Zhu Y, Yang T, Duan J, Mu N and Zhang T: MALAT1/miR-15b-5p/MAPK1 mediates endothelial progenitor cells autophagy and affects coronary atherosclerotic heart disease via mTOR signaling pathway. Aging (Albany NY). 11:1089–1109. 2019.PubMed/NCBI View Article : Google Scholar

35 

Mercer TR, Dinger ME and Mattick JS: Long non-coding RNAs: Insights into functions. Nat Rev Genet. 10:155–159. 2009.PubMed/NCBI View Article : Google Scholar

36 

Wilusz JE, Sunwoo H and Spector DL: Long noncoding RNAs: Functional surprises from the RNA world. Genes Dev. 23:1494–1504. 2009.PubMed/NCBI View Article : Google Scholar

37 

Xu C, Yang M, Tian J, Wang X and Li Z: MALAT-1: A long non-coding RNA and its important 3'end functional motif in colorectal cancer metastasis. Int J Oncol. 39:169–175. 2011.PubMed/NCBI View Article : Google Scholar

38 

Tian X and Xu G: Clinical value of lncRNA MALAT1 as a prognostic marker in human cancer: Systematic review and meta-analysis. BMJ Open. 5(e008653)2015.PubMed/NCBI View Article : Google Scholar

39 

Yang L, Wang H, Shen Q, Feng L and Jin H: Long non-coding RNAs involved in autophagy regulation. Cell Death Dis. 8(e3073)2017.PubMed/NCBI View Article : Google Scholar

40 

Rossi MN and Antonangeli F: LncRNAs: New players in apoptosis control. Int J Cell Biol. 2014(473857)2014.PubMed/NCBI View Article : Google Scholar

41 

Zhuo C, Jiang R, Lin X and Shao M: LncRNA H19 inhibits autophagy by epigenetically silencing of DIRAS3 in diabetic cardiomyopathy. Oncotarget. 8:1429–1437. 2017.PubMed/NCBI View Article : Google Scholar

42 

Zhang N, Yang G, Shao X and Wei L: GAS5 modulated autophagy is a mechanism modulating cisplatin sensitivity in NSCLC cells. Eur Rev Med Pharmacol Sci. 20:2271–2277. 2016.PubMed/NCBI

43 

Song J, Ahn C, Chun CH and Jin EJ: A long non-coding RNA, GAS5, plays a critical role in the regulation of miR-21 during osteoarthritis. J Orthop Res. 32:1628–1635. 2014.PubMed/NCBI View Article : Google Scholar

44 

Gwinn DM, Shackelford DB, Egan DF, Mihaylova MM, Mery A, Vasquez DS, Turk BE and Shaw RJ: AMPK phosphorylation of raptor mediates a metabolic checkpoint. Mol Cell. 30:214–226. 2008.PubMed/NCBI View Article : Google Scholar

45 

Villanueva-Paz M, Cotan D, Garrido-Maraver J, Oropesa-Ávila M, de la Mata M, Delgado-Pavón A, de Lavera I, Alcocer-Gómez E, Álvarez-Córdoba M and Sánchez-Alcázar JA: AMPK regulation of cell growth, apoptosis, autophagy, and bioenergetics. Exp Suppl. 107:45–71. 2016.PubMed/NCBI View Article : Google Scholar

46 

Yaba A, Bianchi V, Borini A and Johnson J: A putative mitotic checkpoint dependent on mTOR function controls cell proliferation and survival in ovarian granulosa cells. Reprod Sci. 15:128–138. 2008.PubMed/NCBI View Article : Google Scholar

47 

Kayampilly PP and Menon K: Follicle-stimulating hormone increases tuberin phosphorylation and mammalian target of rapamycin signaling through an extracellular signal-regulated kinase-dependent pathway in rat granulosa cells. Endocrinology. 148:3950–3957. 2007.PubMed/NCBI View Article : Google Scholar

48 

Rico C, Laguë MN, Lefèvre P, Tsoi M, Dodelet-Devillers A, Kumar V, Lapointe E, Paquet M, Nadeau MÈ and Boerboom D: Pharmacological targeting of mammalian target of rapamycin inhibits ovarian granulosa cell tumor growth. Carcinogenesis. 33:2283–2292. 2012.PubMed/NCBI View Article : Google Scholar

49 

da Rosa P, Dau A, De Cesaro M, Dos Santos J, Gasperin B, Duggavathi R, Bordignon V and Gonçalves P: Mechanistic target of rapamycin is activated in bovine granulosa cells after LH surge but is not essential for ovulation. Reprod Domest Anim. 51:766–773. 2016.PubMed/NCBI View Article : Google Scholar

50 

Zhou J, Yao W, Liu K, Wen Q, Wu W, Liu H and Li Q: MicroRNA let-7g regulates mouse granulosa cell autophagy by targeting insulin-like growth factor 1 receptor. Int J Biochem Cell Biol. 78:130–140. 2016.PubMed/NCBI View Article : Google Scholar

51 

Park Y, Maizels ET, Feiger ZJ, Alam H, Peters CA, Woodruff TK, Unterman TG, Lee EJ, Jameson JL and Hunzicker-Dunn M: Induction of cyclin D2 in rat granulosa cells requires FSH-dependent relief from FOXO1 repression coupled with positive signals from Smad. J Biol Chem. 280:9135–9148. 2005.PubMed/NCBI View Article : Google Scholar

52 

Yu J, Yaba A, Kasiman C, Thomson T and Johnson J: mTOR controls ovarian follicle growth by regulating granulosa cell proliferation. PLoS One. 6(e21415)2011.PubMed/NCBI View Article : Google Scholar

53 

Yu J, Thomson TC and Johnson J: Cross talk between estradiol and mTOR kinase in the regulation of ovarian granulosa proliferation. Reprod Sci. 19:143–151. 2012.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

January-2021
Volume 21 Issue 1

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Liu X, Zhang P, Li Y, Zhao N and Han H: The AMPK‑mTOR axis requires increased MALAT1 expression for promoting granulosa cell proliferation in endometriosis Retraction in /10.3892/etm.2024.12528. Exp Ther Med 21: 21, 2021
APA
Liu, X., Zhang, P., Li, Y., Zhao, N., & Han, H. (2021). The AMPK‑mTOR axis requires increased MALAT1 expression for promoting granulosa cell proliferation in endometriosis Retraction in /10.3892/etm.2024.12528. Experimental and Therapeutic Medicine, 21, 21. https://doi.org/10.3892/etm.2020.9453
MLA
Liu, X., Zhang, P., Li, Y., Zhao, N., Han, H."The AMPK‑mTOR axis requires increased MALAT1 expression for promoting granulosa cell proliferation in endometriosis Retraction in /10.3892/etm.2024.12528". Experimental and Therapeutic Medicine 21.1 (2021): 21.
Chicago
Liu, X., Zhang, P., Li, Y., Zhao, N., Han, H."The AMPK‑mTOR axis requires increased MALAT1 expression for promoting granulosa cell proliferation in endometriosis Retraction in /10.3892/etm.2024.12528". Experimental and Therapeutic Medicine 21, no. 1 (2021): 21. https://doi.org/10.3892/etm.2020.9453