Next Article in Journal
Advances in Biomarkers and Endogenous Regulation of Breast Cancer Stem Cells
Next Article in Special Issue
MEG3 Expression Indicates Lymph Node Metastasis and Presence of Cancer-Associated Fibroblasts in Papillary Thyroid Cancer
Previous Article in Journal
Locus-Specific Enrichment Analysis of 5-Hydroxymethylcytosine Reveals Novel Genes Associated with Breast Carcinogenesis
Previous Article in Special Issue
MiR-22 Deficiency Fosters Hepatocellular Carcinoma Development in Fatty Liver
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Long Noncoding RNAs and Circular RNAs Regulate AKT and Its Effectors to Control Cell Functions of Cancer Cells

1
School of Post-Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
2
Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaoshiung Medical University, Kaohsiung 80708, Taiwan
3
Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
4
Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
5
Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
6
Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
7
Institute of Biomedical and Genetic Engineering (IBGE), Islamabad 54000, Pakistan
8
Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Cells 2022, 11(19), 2940; https://doi.org/10.3390/cells11192940
Submission received: 10 July 2022 / Revised: 6 September 2022 / Accepted: 17 September 2022 / Published: 20 September 2022
(This article belongs to the Special Issue Regulatory Roles of Non-coding RNAs in Cancer)

Abstract

:
AKT serine-threonine kinase (AKT) and its effectors are essential for maintaining cell proliferation, apoptosis, autophagy, endoplasmic reticulum (ER) stress, mitochondrial morphogenesis (fission/fusion), ferroptosis, necroptosis, DNA damage response (damage and repair), senescence, and migration of cancer cells. Several lncRNAs and circRNAs also regulate the expression of these functions by numerous pathways. However, the impact on cell functions by lncRNAs and circRNAs regulating AKT and its effectors is poorly understood. This review provides comprehensive information about the relationship of lncRNAs and circRNAs with AKT on the cell functions of cancer cells. the roles of several lncRNAs and circRNAs acting on AKT effectors, such as FOXO, mTORC1/2, S6K1/2, 4EBP1, SREBP, and HIF are explored. To further validate the relationship between AKT, AKT effectors, lncRNAs, and circRNAs, more predicted AKT- and AKT effector-targeting lncRNAs and circRNAs were retrieved from the LncTarD and circBase databases. Consistently, using an in-depth literature survey, these AKT- and AKT effector-targeting database lncRNAs and circRNAs were related to cell functions. Therefore, some lncRNAs and circRNAs can regulate several cell functions through modulating AKT and AKT effectors. This review provides insights into a comprehensive network of AKT and AKT effectors connecting to lncRNAs and circRNAs in the regulation of cancer cell functions.

1. Introduction

AKT serine/threonine kinase (AKT) shows activation or overexpression in several cancers [1]. AKT signaling is vital for diverse regulations to modulate several cell functions [2,3,4], such as survival, proliferation, metabolism, and angiogenesis. Additionally, several AKT signaling-associated cell functions include apoptosis, autophagy, endoplasmic reticulum (ER) stress, mitochondrial morphogenesis (fission/fusion), ferroptosis, necroptosis, and DNA damage response (damage and repair), senescence, and migration (Figure 1). AKT mutation occurs in several cancer types, such as leukemia [5], breast [6], and others [7]. However, AKT mutation rates seem low (3–5%) for all cancers [7,8], and this issue was not included in this review.
AKT signaling can modulate several downstream AKT effectors, such as forkhead box transcription factors (FOXO), c-Myc, mechanistic target of rapamycin (mTOR) complex 1/2 (mTORC1/2), mTOR substrate S6 kinase 1/2 (S6K1/2; RPS6KB1/2), eukaryotic translation initiation factor 4E-binding protein 1 (4EBP1; EIF4EBP1), sterol regulatory element-binding protein 1 (SREBP1; SREBF1), and hypoxia-inducible factor (HIF) [3,9,10,11] (Figure 1). However, the complex functions of AKT effects on these cell functions need further investigation.
Emerging evidence has shown the impacts of noncoding RNA (ncRNAs), such as long noncoding RNAs (lncRNAs) and circular RNAs (circRNAs), on regulating cell progression, especially relevant in cancer cells [12]. LncRNAs are a group of ncRNA molecules containing more than 200 nucleotides. LncRNAs exhibit complex interactions with microRNAs (miRNAs), mRNAs, and proteins to regulate cell functions [13]. LncRNAs are essential modulators for regulating gene expression and affect diverse cell functions [14]. CircRNAs are other ncRNAs formed by splicing and sequentially connecting between splice donor and acceptor sites, i.e., backsplicing [15]. CircRNAs may function as protein and RNA scaffolds to bind miRNA and regulate transcription or translation in a miRNA and RNA-binding protein sponge manner [16,17].
Both lncRNAs [18,19,20] and circRNAs [21,22] are reported as potential tumor markers by regulating numerous pathways and controlling cell functions. Mounting evidence shows the interaction between ncRNAs and AKT signaling in several cancers [23,24,25]. As mentioned above, this review focuses on understanding the relationship between AKT and AKT effectors in lncRNA- and circRNA-modulating cell functions. However, the network between AKT, AKT signaling, lncRNAs, and circRNAs lacks systemic integration. In previous reports, possible cell functions were not connected to this AKT–AKT effectors–lncRNAs–circRNAs network. This warrants a detailed organization for understanding their relationships.
LncRNAs [26] and circRNAs [27] can function as miRNA sponges, modulating their target mRNA expressions. However, the miRNA information was not under the scope of this review and is not discussed. Additionally, lncRNAs can interact with DNA, RNA, and proteins for gene regulation [28]. Several DNA and proteins targeted by lncRNAs were summarized in regulating transcription, posttranscription, cellular organelles, structural functions, and genomic integrity [28]. For example, lncRNAs can control chromatin regulation through the recruitment of chromatin modifiers, decoy of chromatin modifiers, and the direct cis or trans interaction with chromatin [28]. LncRNAs also control transcription regulation through target-gene inhibition, gene activation, and multiple lncRNAs acting on the same locus. Consequently, the detailed mechanisms for lncRNAs and circRNAs to control gene expressions are complex and display distinct regulations in different cases. Therefore, the complicated mechanisms of the interactions between AKT/AKT effectors and their respective lncRNAs and circRNAs are not included in this review. Alternatively, we focused on potential target genes such as AKT and AKT effectors regulated by lncRNAs and circRNAs that were predicted by databases, as mentioned later.
This review provides an overview of AKT, AKT effectors, lncRNAs, and circRNAs in regulating cell functions (Figure 1). Firstly, it summarizes detailed information on AKT and AKT signaling-modulated functions relating lncRNAs and circRNAs to several cell functions, especially for cancer cells, including apoptosis, autophagy, ER stress, mitochondrial morphogenesis, ferroptosis, necroptosis, DNA damage response, senescence, and migration. Detailed mechanisms for most lncRNA- and circRNA-associated regulation of AKT and AKT effectors lack in-depth connection. To fill the gap, we next chose databases for lncRNAs and circRNAs that provide the predicted targets to AKT and AKT effectors (Section 2 and Section 3). Subsequently, these predicted targets (AKT and AKT effectors) of lncRNAs and circRNAs are connected to cell functions by a literature survey. Finally, AKT and AKT effectors that regulate cell functions appear to be well organized and connected to lncRNAs and circRNAs.

2. Connecting AKT/AKT Effectors and LncRNAs to Cell Functions

The human AKT family contains AKT1, AKT2, and AKT3 [29,30,31], located at chromosomes 14, 19, and 1. These AKT family members share several conserved structures containing the pleckstrin homology (PH) domain at the N-terminal, kinase domain at the middle region, and the hydrophobic regulating domain at C-terminal [32]. AKT1 expresses in ubiquitous tissues, while AKT2 and AKT3 are mainly expressed in skeletal muscle and liver [33] and in brain and testis [34], respectively.
Notably, some lncRNAs were reported to modulate the expressions of AKT1 [35], AKT2 [36], and AKT3 [37]. However, their connection to cell function was not investigated, especially for cancer cells. Hence, the relationship that connects AKT and lncRNAs to their modulating cell functions (Figure 1) was evaluated by literature retrieval (Section 2.1). However, their potential mechanisms still warrant a detailed exploration, particularly for the possible targeting to AKT by lncRNAs. Subsequently, the potential targeting to AKT1, AKT2, and AKT3 by lncRNAs and their associated cell functions are discussed (Section 2.2). By choosing the lncRNA database (LncTarD [38]), the target information of respective lncRNAs was predicted, and their impacts on cell functions were evaluated, as described later.
Some lncRNAs also were reported to modulate the expressions of AKT effectors [38]. However, their connection to cell functions has never been investigated, especially for cancer cells. Hence, the evidence that connects the AKT effectors and lncRNAs to their modulating cell functions (Figure 1) was evaluated by literature retrieval (Section 2.3). However, their potential mechanisms still warrant a detailed assessment, particularly for the possible targeting to AKT effectors by lncRNAs. Subsequently, the potential targeting to AKT effectors by lncRNAs and their associated cell functions are discussed (Section 2.4). By choosing the lncRNA database LncTarD [38], the target information of respective lncRNAs was predicted and their impacts on cell functions evaluated as described later.

2.1. Connecting AKT and LncRNAs to Cell Functions

Phosphoinositide 3-kinase (PI3K)/AKT/mTOR signaling and lncRNAs have a cross-relationship regulating carcinogenesis [24,39,40]. They are essential in regulating apoptosis, autophagy, ER stress, mitochondrial morphogenesis, ferroptosis, necroptosis, DNA damage response, senescence, and migration. These AKT-lncRNA--regulating cell functions are discussed in Section 2.1.1, Section 2.1.2, Section 2.1.3, Section 2.1.4, Section 2.1.5, Section 2.1.6 and Section 2.1.7, especially for cancer cells.

2.1.1. Apoptosis by AKT-Regulating LncRNAs

Apoptosis-modulating effects of lncRNAs involving AKT have been reported. Some lncRNA studies reported apoptosis-promoting effects in various cancer cell types connecting to AKT. Phosphatase and tension homolog deleted on chromosome ten (PTEN) is a negative modulator of AKT signaling [41]. LncRNA FER1L4 causes apoptosis of lung cancer cells by upregulating PTEN expression and dephosphorylating AKT [42]. LncRNA LINC00619 is downregulated in osteosarcoma cells, associated with AKT mRNA overexpression and its protein phosphorylation [43]. LINC00619 overexpression promotes apoptosis of osteosarcoma cells by targeting hepatocyte growth factor (HGF) and downregulating AKT mRNA expressions and its protein phosphorylation [43]. Consequently, various lncRNAs may be overexpressed in several cancers to modulate AKT for controlling apoptosis induction.
In contrast, some lncRNA studies reported apoptosis-suppressing effects of several cancer cell types connecting to AKT. LncRNA HOTAIR inhibits apoptosis of retinoblastoma cells by upregulating ribonucleotide reductase regulatory subunit M2 (RRM2) for phosphorylating AKT, reverted by HOTAIR knockdown [44]. LncRNA RP11-301G19.1 downregulation triggers apoptosis of myeloma cancer cells by dephosphorylating AKT [45]. SNHG20 silencing triggers apoptosis of lung cancer cells by dephosphorylating AKT [46]. ROR1-AS1 is overexpressed in lung cancer tissues. ROR1-AS1 inhibition triggers apoptosis in lung cancer cells by dephosphorylating AKT [47]. LINC01410 and lncRNA PITPNA-AS1 are overexpressed in glioblastoma cells [48,49]. LINC01410 knockdown induces temozolomide-induced apoptosis of glioblastoma cells by upregulating PTEN expression and dephosphorylating AKT [48]. LncRNA PITPNA-AS1 inhibits apoptosis of glioblastoma cells by upregulating epidermal growth factor receptor (EGFR) expression and phosphorylating AKT [49].
Accordingly, different lncRNAs may promote or suppress apoptosis by phosphorylating or dephosphorylating AKT to regulate its activity. As mentioned above, some tumors are overexpressed or downregulated by AKT-regulating lncRNAs. A strategy to overexpress or downregulate these specific AKT-regulating lncRNAs can improve the apoptosis-inducible effects for cancer treatment.

2.1.2. Autophagy by AKT-Regulating LncRNAs

Autophagy modulating effects of lncRNAs involving AKT have been reported. The lncRNA MEG3 overexpression blocks the phosphorylation of PI3K/AKT/mTOR to promote autophagy in tumor necrosis factor α (TNF-α)-treated keratinocytes [50]. Some lncRNAs exhibit bifunctional effects to regulate apoptosis and autophagy. LncRNA ADAMTS9-AS1 upregulation blocks apoptosis and autophagy of bladder cancer cells by phosphorylating AKT, reverted by downregulating ADAMTS9-AS1 [51]. Accordingly, different lncRNAs may promote or suppress autophagy by phosphorylating or dephosphorylating AKT to regulate its activity.

2.1.3. ER Stress by AKT-Regulating LncRNAs

Drug-induced ER stress effects of lncRNA involving AKT have been reported. 5-Fluorouracil induces ER stress and glucose-regulated protein 78 (GRP78; BiP) expression of breast cancer cells to cause 5-fluorouracil resistance, accompanied by upregulating myocardial infarction-associated transcript (MIAT lncRNA) and AKT protein expression [52]. This warrants surveying more lncRNAs that regulate other ER stress components in the future.

2.1.4. DNA Damage Response by AKT-Regulating LncRNAs

DNA repair-suppressing effects of lncRNAs involving AKT have been reported. Linc00312 directly targets the DNA-dependent protein kinase, catalytic subunit (DNA-PKcs), blocks the interaction between DNA-PKcs and Ku80, and inactivates AKT by dephosphorylation, suppressing nonhomologous end joining (NHEJ) repair in nasopharyngeal cancer cells [53]. This warrants surveying more lncRNAs that regulate other DNA damage and repair in the future.

2.1.5. Senescence by AKT-Regulating LncRNAs

Senescence-inducible effects of lncRNAs involving AKT have been reported. GAS5 silencing induces proliferation and suppresses the senescence of endothelial progenitor cells by dephosphorylating PI3K/AKT [54]. This warrants a detailed survey and examination of senescence effects of AKT-targeting lncRNAs on cancer cells in the future.

2.1.6. Migration by AKT-Regulating LncRNAs

Drug-induced migration-modulating effects of lncRNAs involving AKT have been reported. Some lncRNA studies investigated migration-promoting effects. PYCR2 knockdown suppresses the migration of colon cancer cells by downregulating matrix metalloproteinase (MMP) 2/9 and dephosphorylating AKT [55]. LINC00963 promotes metastasis of lung cancer cells by phosphorylating AKT [56]. SOX2 overlapping transcript (SOX2-OT) promotes the phosphorylation of PI3K/AKT and induces breast cancer cell metastasis [57]. MIR205HG [58] and AC099850.3 [59] enable the migration of liver cancer cells by phosphorylating AKT.
In contrast, some lncRNA studies investigated migration-suppressing effects. Platelet-derived growth factor BB (PDGF-BB) inhibits RP5-857K21.7 expression of airway smooth muscle cells (ASMCs). RP5-857K21.7 overexpression inhibits the migration of PDGF-BB-treated ASMCs through dephosphorylating AKT [60]. Accordingly, different lncRNAs may promote or suppress migration by phosphorylating or dephosphorylating AKT. This warrants a detailed survey and examination of migration effects of AKT-targeting lncRNAs on cancer cells in the future.

2.1.7. Potential Future Directions

As described above, several lncRNAs were mentioned to regulate AKT phosphorylation or dephosphorylation for its activation and inactivation and, in turn, control cell functions. Some AKT-regulating lncRNAs are abundant or scarce in various cancers. Overexpressing or downregulating these AKT-regulating lncRNAs may reverse the status of cancer cell functions to improve anticancer effects.
However, the cell function mechanism for the modulating effects of lncRNAs on AKT remains unclear, particularly for assessing the potential targeting to AKT by lncRNAs. More experiments are warranted to improve the connection between AKT-lncRNAs regulating cancer cell functions.

2.2. Connecting AKT1/AKT2/AKT3 and Database-LncRNAs to Cell Functions

To further validate the relationship between AKT and lncRNAs to cell functions, more potential AKT-targeting lncRNAs are required. By choosing lncRNA databases, such as LncTarD [38], more AKT-targeting lncRNA candidates are generated. LncTarD is a comprehensive lncRNA database, including disease-associated lncRNA-target regulations with experiment supports, associations, and targets to biological functions, as well as TCGA pan-cancer data. By individual input target genes such as “AKT1, AKT2, and AKT3,” their respective predicted lncRNAs are generated and exported. LncTarD also provides the evidence description for each predicted lncRNA. However, these LncTarD-predicted lncRNAs for AKT1, AKT2, and AKT3 did not provide potential cell functions. Subsequently, these candidates were used for a literature survey (Google Scholar and PubMed) to check their possible cell functions. Finally, the connection between these AKT-targeting database lncRNA candidates and cell functions was established (Figure 2).
Since AKT1, AKT2, and AKT3 are encoded by different genes, their related modulating lncRNAs are different as well. This lncRNA target information for AKT1, AKT2, and AKT3 was retrieved from the LncTarD database [38] and summarized in Table 1). Several lncRNAs (ENST00113, MALAT1, CDKN2B-AS1, HULC, LUCAT1, AFAP1-AS1, LINC00462, LOXL1-AS1, AB073614, H19, and SPRY4-IT1) upregulate AKT1, while some lncRNAs (GAS5, RP11-708H21.4, FOXD2-AS1, and LINC00312) downregulate AKT1. LncRNA (lncRNA-p3134) upregulates AKT2, and lncRNA (FEZF1-AS1) upregulates AKT3.
Interestingly, the lncRNA targets for AKT1, AKT2, and AKT3 are not overlapping. Notably, the investigation of AKTs should be concerned with transcriptional regulation regarding their respective lncRNAs. The relationship between AKT1, AKT2, and AKT3 connecting to database lncRNAs in regulating cell functions will be explored further below.

2.2.1. AKT1-, AKT2-, and AKT3-Targeting Database LncRNAs and Cell Functions

Although the respective AKT1-, AKT2-, and AKT3-targeting lncRNAs have been reported before (Table 1), the cell functions were not connected to these AKT1-, AKT2-, and AKT3-targeting database lncRNAs. Here, we summarize and integrate available information from our in-depth literature search on Google Scholar and PubMed (Figure 2) and provide novel information about the networking of the AKT1-, AKT2-, and AKT3-targeting lncRNAs and cell functions. Twelve AKT-targeting database lncRNAs are connected to cell functions, as summarized, especially for cancer cells (Table 2).
(1)
AKT1-Targeting ENST00113 and Cell Functions
LncRNA ENST00113 modulating cell functions are summarized in Table 2). LncRNA ENST00113 enhances atherosclerosis development [62]. ENST00113 enhances proliferation and migration, but inhibits apoptosis of human umbilical vein endothelial cells by phosphorylating PI3K/AKT/mTOR [62]. AKT inactivation by inhibitor or siRNA suppresses atherosclerosis by upregulating the autophagy of macrophages [63]. Accordingly, this warrants a detailed assessment of the impact of ENST00113 on modulating autophagy and careful examination of cell functions due to ENST00113 on cancer cells in the future.
(2)
AKT1-Targeting MALAT1 and Cell Functions
MALAT1 modulating cell functions are summarized in Table 2. MALAT1 enhances the proliferation and autophagy of glioma cells [65]. MALAT1 inhibition suppresses oxygen-glucose deprivation/reoxygenation-triggered apoptosis, and ER stress [66]. Upregulation of mitochondrial fusion protein mitofusin 1 (MFN1) reverses microvascular dysfunction and cardiac microvascular endothelial cell damage enhanced by MALAT1 knockdown by suppressing mitochondrial fission and apoptosis [64]. MALAT1 enhances proliferation and suppresses the senescence of gallbladder cancer cells [67]. MALAT1 improves the proliferation and migration of colon cancer cells [68]. This deserves a careful examination of cell functions due to MALAT1 to provide more evidence for impacts on cancer cells in the future.
(3)
AKT1-Targeting GAS5 and Cell Functions
LncRNA GAS5 modulating cell functions are summarized in Table 2. GAS5 upregulation inhibits proliferation and promotes apoptosis of pituitary neuroendocrine [69] and liver [70] cancer cells. GAS5 induces autophagy of colon [71] and breast [72] cancer cells. GAS5 blocks high glucose-induced ER stress and apoptosis of retinal epithelial cells [73]. GAS5 pathways impact ferroptosis-associated gene expressions in heart-failure tissue [74]. GAS5 knockdown increases cell viability in the hypoxia-reoxygenation model by reducing the expression of necrosis markers, such as lactate dehydrogenase [75]. GAS5 upregulation suppresses DNA repair to ionizing radiation [76]. GAS5 overexpression blocks the senescence of vascular smooth muscle cells [77]. GAS5 upregulation blocks propofol-induced migration of glioma cells [78]. Accordingly, some cell functions of GAS5 were not investigated in cancer cells. Careful examination of some cell functions due to GAS5 acting on cancer cells is needed.
(4)
AKT1-Targeting CDKN2B-AS1 and Cell Functions
CDKN2B-AS1 modulating cell functions are summarized in Table 2. CDKN2B-AS1 inhibits apoptosis and senescence of cervical cancer, which can be reverted by CDKN2B-AS1 silencing [79]. CDKN2B-AS1 upregulation induces autophagy of idiopathic pulmonary fibrosis [80]. Additionally, stress-associated endoplasmic reticulum protein 1 (SERP1) downregulates CDKN2B-AS1 and ER stress of oxygen deprivation-induced injury in cardiomyocytes [81]. CDKN2B-AS1 modifies senescence and apoptosis, involving cell cycle disturbance and DNA damage [82]. CDKN2B-AS1 improves the proliferation and metastasis of liver cancer cells [83]. Careful examination of cell functions and CDKN2B-AS1 is needed to provide more evidence for impacts on cancer cells in the future.
(5)
AKT1-Targeting HULC and Cell Functions
HULC modulating cell functions are summarized in Table 2. HULC suppresses apoptosis of osteosarcoma [84] and lung cancer cells [85]. HULC induces autophagy of liver cancer cells [86]. HULC enhances the DNA repair of liver cancer stem cells [87]. Additionally, hepatitis B virus X protein upregulates HULC and downregulates senescence protein p18 expressions, suggesting that HULC can modulate cellular senescence [88]. HULC enhances the migration of pancreatic [89] and liver [90] cancer cells. This warrants a detailed examination of cell functions influenced by HULC to provide more evidence for their impacts on cancer cells in the future.
(6)
AKT1-Targeting LUCAT1 and Cell Functions
LUCAT1 modulating cell functions are summarized in Table 2. LUCAT1 overexpression induces autophagy and metastasis, but suppresses apoptosis of lung cancer cells and promotes its cisplatin resistance [91]. LUCAT1 is reported to be the ferroptosis-related lncRNA correlated with renal cancer survival [92]. LUCAT1 is reported to be the necroptosis-related lncRNA in liver tumors [93]. LUCAT1 suppresses DNA damage and apoptosis of colon cancer cells [94]. A detailed examination of cell functions and LUCAT1 is warranted to provide more evidence for their impacts on cancer cells in the future.
(7)
AKT1-Targeting RP11-708H21.4, AFAP1-AS1, LINC00462, and Cell Functions
RP11-708H21.4, AFAP1-AS1, and LINC00462 modulating cell functions are summarized in Table 2. RP11-708H21.4 has low expression in colon cancer cells. RP11-708H21.4 overexpression decreases proliferation and migration and triggers apoptosis of colon cancer cells by dephosphorylating AKT and mTOR [95]. Additionally, AFAP1-AS1 silencing decreases proliferation and migration and induces apoptosis of lung cancer cells [96]. LINC00462 silencing suppresses high glucose-triggered apoptosis of renal tubular epithelial cells [97]. LINC00462 improves the invasion of pancreatic cancer cells [98]. A detailed assessment of cell functions influenced by RP11-708H21.4, AFAP1-AS1, and LINC00462 is warranted to provide more evidence for their impacts on cancer cells in the future.
(8)
AKT1-Targeting LOXL1-AS1, FOXD2-AS1, AB073614, and Cell Functions
LOXL1-AS1, FOXD2-AS1, and AB073614 modulating cell functions are summarized in Table 2. LOXL1-AS1 suppresses proliferation and migration and enhances apoptosis of breast cancer cells [99]. The lncRNA FOXD2-AS1 knockdown decreases proliferation and migration but triggers apoptosis of glioma cells [100]. AB073614 enhances proliferation and triggers apoptosis of cervical cancer cells [101]. AB073614 improves metastasis of gastric cancer cells [102]. This warrants a detailed assessment of cell functions influenced by LOXL1-AS1, FOXD2-AS1, and AB073614 to provide more evidence for their impacts on cancer cells in the future.
(9)
AKT1-Targeting H19 and Cell Functions
H19 modulating cell functions are summarized in Table 2. H19 silencing blocks proliferation and triggers apoptosis of vascular smooth muscle cells [103]. H19 upregulation enhances invasion and autophagy of trophoblast cells [104]. H19 knockdown promotes resveratrol-induced ER stress and necroptosis of gastric cancer cells by increasing GRP78, receptor-interacting serine/threonine-protein kinase 1 (RIPK1), and mixed lineage kinase domain-like (MLKL) expressions [105]. H19 silencing enhances the ferroptosis of lung cancer cells [107]. Additionally, lncRNA H19 downregulates mitochondrial fusion expression of the MFN2 gene in renal tissues of diabetic rats [106]. H19 silencing suppresses ionizing radiation-induced DNA damage of lung cancer cells, but enhances DNA repair [108]. H19 triggers the senescence of cardiomyocytes [109]. Accordingly, some cell functions of H19 have not been investigated in cancer cells. Careful examination of some cell functions influenced by H19 is needed to provide more evidence for their impacts on cancer cells in the future.
(10)
AKT1-Targeting SPRY4-IT1, LINC00312, and Cell Functions
SPRY4-IT1 and LINC00312 modulating cell functions are summarized in Table 2. SPRY4-IT1 downregulation improves apoptosis of pancreatic cancer cells [110]. SPRY4-IT1-expressing primary human melanocytes show gene expression changes along with apoptosis and DNA damage responses [111]. SPRY4-IT1 enhances metastasis in nasopharyngeal cancer cells [112]. LINC00312 suppresses proliferation and triggers apoptosis of lung cancer cells [113]. LINC00312 suppresses DNA repair of nasopharyngeal cancer cells by targeting DNA-PKcs [53]. LINC00312 suppresses the migration of bladder cancer cells [114]. A detailed assessment of cell functions influenced by SPRY4-IT1 and LINC00312 is warranted to provide more evidence for their impacts on cancer cells in the future.
(11)
AKT2-Targeting LncRNA-p3134 and Cell Functions
lncRNA-p3134 modulating cell functions are summarized in Table 2. For AKT2, lncRNA-p3134 upregulation suppresses the β-cell apoptosis of pancreatic β-cells [115]. According to our literature survey, other cell functions related to AKT2 have not been reported.
(12)
AKT3-Targeting FEZF1-AS1 and Cell Functions
FEZF1-AS1 modulating cell functions are summarized in Table 2. For AKT3, FEZF1-AS1 exhibits higher expression in ovarian cancer tissues and cells than normal controls [116]. Ovarian cancer patients with high FEZF1-AS1 show a poor prognosis. FEZF1-AS1 silencing inhibits proliferation and induces apoptosis of ovarian cancer cells [116]. Similarly, FEZF1-AS1 is overexpressed in gastric tumors. FEZF1-AS1 overexpression improves proliferation and autophagy of gastric cancer cells, reverted by ATG5 silencing [117]. A detailed assessment of cell functions influenced by FEZF1-AS1 is warranted to provide more evidence for their impacts on cancer cells in the future.

2.2.2. Potential Future Directions

As described above, a literature survey connected AKT1-, AKT2-, and AKT3-targeting database lncRNAs to several cell functions. However, most information was derived from AKT1 in our survey (Table 2). AKT2 and AKT3 were rarely investigated. This warrants a detailed assessment of the role of AKT2 and AKT3 targeting by lncRNAs in regulating cancer cell functions in the future. Some lncRNAs reported in some cell functions but not others are based on a literature survey. Their possible contributions to unreported cell functions are not excluded and need further inspection.

2.3. Connecting AKT Effectors and LncRNAs to Cell Functions

AKT controls the expressions of several downstream effectors. In turn, AKT effectors exert comprehensive cell functions [3,9,10,11]. Since AKT had a cross-relationship to lncRNAs as described above, lncRNAs may exhibit the impact on most AKT effectors (FOXO, c-Myc, mTORC1, SREBP1, and HIF) (Table 3). The connection between lncRNAs to other AKT effectors (S6K1, S6K2, and 4EBP1) was rarely reported. In the following, we summarize evidence connecting some AKT effectors (FOXO, c-Myc, mTORC1, SREBP1, and HIF) and lncRNAs to cancer cell functions (Section 2.3.1, Section 2.3.2, Section 2.3.3, Section 2.3.4, Section 2.3.5 and Section 2.3.6).

2.3.1. AKT Effector (FOXO)-Regulating LncRNAs and Cell Functions

The relationship between FOXO, lncRNA, and cell functions such as autophagy, ER stress, necroptosis, DNA damage response, and senescence were rarely reported. Other functions, such as apoptosis, ferroptosis, and migration were mentioned, as follows (Table 3).
(1)
Apoptosis by FOXO-Regulating LncRNAs
Several lncRNA studies investigated apoptosis modulating effects involving FOXO (Table 3). Under energy stress, FOXO upregulates FOXO-induced lncRNA 1 (FILNC1) to suppress proliferation and induce apoptosis of renal cancer cells [118]. siRNA may induce or suppress apoptosis involving FOXO. In contrast, LINC00899 silencing downregulates FOXO expression and induces apoptosis of spinal ependymoma cells [119]. A detailed assessment of apoptosis influenced by more FOXO-regulating lncRNAs is warranted to provide more evidence for their impacts on cancer cells in the future.
(2)
Ferroptosis by FOXO-Regulating LncRNAs
Several lncRNA studies investigated ferroptosis modulating effects involving FOXO (Table 3). Seventeen ferroptosis-related lncRNAs were associated with gastric cancer [120] and upregulated FOXO3. Some lncRNAs are risk for gastric cancer, such as VCAN-AS1, OVAAL, PCDH10-DT, ENSG00000240661.1, RPH3AL-AS1, ITGB1-DT, LINC02915, FLJ42969, NDST1-AS1, ENSG00000247134.5, and ENSG00000248362.1). Other lncRNAs are protective for gastric cancer, such as FAM239A, LINC01210, ENSG00000265334.1, LINC01775, ENSG00000273293.1, and ENSG00000230107.1) [120]. A detailed assessment of ferroptosis influenced by more FOXO-regulating lncRNAs is warranted to provide more evidence for their impacts on cancer cells in the future.
(3)
Migration by FOXO-Regulating LncRNAs
Several lncRNA studies investigated migration-modulating effects involving FOXO (Table 3). LINC00899 knockdown inhibits FOXO expression and migration of spinal ependymoma cells [119]. Oncogene E26 transformation-specific or E-twenty-six (ETS)-related gene (ERG), an oncogenic transcription factor, upregulates LINC00920 to promote the proliferation and migration of prostate cancer cells by downregulating FOXO expression [121]. A detailed assessment of migration influenced by more FOXO-regulating lncRNAs is warranted to provide more evidence for their impacts on cancer cells in the future.

2.3.2. AKT Effector (c-Myc)-Regulating LncRNAs and Cell Functions

There is little information about the relationship between c-Myc, lncRNA, and cell functions, such as mitochondrial morphogenesis. Other functions, such as apoptosis, autophagy, ER stress, ferroptosis, necroptosis, DNA damage response, senescence, and migration were mentioned as follows (Table 3).
(1)
Apoptosis by c-Myc-Regulating LncRNAs
Several lncRNA studies investigated apoptosis-modulating effects involving c-Myc (Table 3). In some cases, lncRNAs may regulate c-Myc by direct targeting. Lnc-EPIC1 silencing triggers apoptosis of colon cancer cells by directly binding to c-Myc and downregulating c-Myc downstream effectors [122]. However, most of the c-Myc-regulating lncRNAs did not investigate their targeting potential. Inhibition of lncRNA MIR22HG suppresses proliferation and induces apoptosis of esophageal cancer cells via downregulating c-Myc expression [123]. LncRNA KCNQ1OT1 silencing causes apoptosis of acute myeloid leukemia by decreasing c-Myc expression [124]. LINC01503 is downregulated by c-Myc silencing to induce apoptosis of lung cancer cells [125], suggesting that c-Myc may upregulate LINC01503 to inhibit the apoptosis of lung cancer cells [125]. A detailed assessment of apoptosis influenced by more c-Myc-regulating lncRNAs is warranted to provide more evidence for their impacts on cancer cells in the future.
(2)
Autophagy by c-Myc-Regulating LncRNAs
Several lncRNA studies investigated autophagy-modulating effects involving c-Myc (Table 3). LncRNA may induce or suppress autophagy connected to c-Myc. c-Myc-induced lncRNA MEG3 activates mitophagy to alleviate kidney ischemia–reperfusion injury [126]. In contrast, MIR7-3HG, an Myc-dependent lncRNA, blocks the autophagy of cervical cancer cells [127]. LncRNA NFYC-AS1 silencing activates autophagy of lung cancer cells by downregulating c-Myc [128].A detailed assessment of autophagy influenced by more c-Myc-regulating lncRNAs is warranted to provide more evidence for their impacts on cancer cells in the future.
(3)
ER Stress by c-Myc-Regulating LncRNAs
Several lncRNA studies investigated ER stress-modulating effects involving c-Myc (Table 3). c-Myc improves adaptive ER stress [129]. Metformin upregulates the expressions of lncRNA MALAT1 and ER stress genes, while MALAT1 knockdown in metformin-treated breast cancer cells shows reduced phosphorylation of c-Myc [130]. Accordingly, MALAT1 is a potential upstream regulator to c-Myc for triggering ER stress. IA detailed assessment of ER stress influenced by more c-Myc-regulating lncRNAs is warranted to provide more evidence for their impacts on cancer cells in the future.
(4)
Ferroptosis by c-Myc-Regulating LncRNAs
Ferroptosis-modulating effects of lncRNAs involving c-Myc were reported (Table 3). Transcription factor AP-2 gamma (TFAP2C) transcriptionally activates lncRNA PCAT1 to suppress ferroptosis of prostate cancer cells by interacting with c-Myc [131]. A detailed assessment of ferroptosis influenced by more c-Myc-regulating lncRNAs is warranted to provide more evidence for their impacts on cancer cells in the future.
(5)
Necroptosis by c-Myc-Regulating LncRNAs
Necroptosis-modulating effects of lncRNAs involving c-Myc were reported (Table 3). Linc00176 is highly expressed in liver cancer cells, which is activated by c-Myc. Linc00176 knockdown promotes necroptosis of liver cancer cells [132]. Accordingly, c-Myc may modulate linc00176 expression to control necroptosis. A detailed assessment of necroptosis influenced by more c-Myc-regulating lncRNAs is warranted to provide more evidence for their impacts on cancer cells in the future.
(6)
DNA Damage Response by c-Myc-Regulating LncRNAs
Several lncRNA studies investigated DNA damage response-modulating effects involving c-Myc (Table 3). LncRNA may induce or suppress DNA repair connecting to c-Myc. LncRNA PVT1 improves DNA repair and suppresses cell apoptosis of nasopharyngeal cancer cells [133]. p53 activates PVT1b to reduce c-Myc transcription and suppress carcinogenesis [134]. A detailed investigation of the interaction between PVT1b and Myc in modulating DNA repair is particularly needed here. Similarly, noncoding RNA activated by DNA damage (NORAD) knockdown in neuroblastoma cells upregulates the poly [ADP-ribose] polymerase 1 (PARP1), a DNA damage sensor for DNA repair [135]. In contrast, in gene set enrichment analysis (GSEA), head neck cancer patients with low lncRNA NEAT1 expression exhibit upregulation of c-Myc and DNA repair signaling [136]. A detailed assessment of DNA damage response influenced by more c-Myc-regulating lncRNAs is warranted to provide more evidence for their impacts on cancer cells in the future.
(7)
Senescence by c-Myc-Regulating LncRNAs
Several lncRNA studies investigated senescence-modulating effects involving c-Myc (Table 3). Several lncRNA studies reported senescence-suppressing results connecting to c-Myc. LncRNA PARROT, an upstream modulator of c-Myc, is downregulated in the senescence of human mammary epithelial cells [137]. c-Myc may transcriptionally activate some lncRNAs, such as USP2-AS1, to inhibit senescence and improve the proliferation of lung cancer cells [138]. C1RL-AS1 knockdown promotes the senescence of gastric cancer cells by decreasing c-Myc expression [139]. c-Myc upregulates ovarian adenocarcinoma-amplified lncRNA (OVAAL) transcription to promote tumor growth and inhibit senescence [140]. A detailed assessment of senescence influenced by more c-Myc-regulating lncRNAs is warranted to provide more evidence for their impacts on cancer cells in the future.
(8)
Migration by c-Myc-Regulating LncRNAs
Several lncRNA studies investigated migration-modulating effects involving c-Myc (Table 3). Several lncRNA studies reported migration-promoting results connecting to c-Myc. LINC00665 promotes c-Myc transcriptional activity to enhance the migration of lung cancer cells [141]. LncRNA AFAP1-AS1 [142] and MIR210HG [143] strengthen the migration of lung and gastric cancer cells by upregulating c-Myc, respectively. c-Myc can bind to the LINC01050 promoter to improve transcription of LINC01050 and enhances metastasis of gastric cancer cells [144]. A detailed assessment of migration influenced by more c-Myc-regulating lncRNAs is warranted to provide more evidence for their impacts on cancer cells in the future.

2.3.3. AKT Effector (mTORC1)-Regulating LncRNAs and Cell Functions

There is little information about the relationship between mTORC1, lncRNA, and cell functions. Other functions, such as apoptosis, autophagy, and migration, were mentioned as follows (Table 3).
(1)
Apoptosis by mTORC1-Regulating LncRNAs
There are studies on apoptosis modulating the effects of lncRNA involving mTORC1 (Table 3). LncRNA H19 suppresses mTORC1 expression of pituitary tumors [145]. Additionally, the apoptosis-promoting effects of lncRNA were reported. LINC00998 enhances mTORC2 decay and apoptosis to suppress carcinogenesis, reverted by mTORC2 overexpression [146]. A detailed assessment of apoptosis influenced by more mTORC1-regulating lncRNAs is warranted to provide more evidence for their impacts on cancer cells in the future.
(2)
Autophagy by mTORC1-Regulating LncRNAs
Several lncRNA studies investigated autophagy-modulating effects involving mTORC1 (Table 3). Autophagy-inducing or -suppressing lncRNAs connecting to mTORC1 were reported. LncRNA ZNNT1 promotes autophagy of uveal melanoma cells by mTORC1 inhibitor [147]. In contrast, HAGLR opposite strand lncRNA (HAGLROS) binds to mTORC1 components and activates mTORC1 signaling by mTOR phosphorylation to inhibit autophagy, contributing to gastric carcinogenesis [148]. A detailed assessment of autophagy influenced by more mTORC1-regulating lncRNAs is warranted to provide more evidence for their impacts on cancer cells in the future.
(3)
Senescence by mTORC1-Regulating LncRNAs
Senescence-modulating effects of lncRNAs involving mTORC1 were reported (Table 3). Senescence-promoting effects of lncRNA connecting to mTORC1 were demonstrated. In non-TGF-β-treated cells, silencing of the metastasis-associated in lung adenocarcinoma transcript 1 (MALAT1) activates mTORC1 [149], associated with cell senescence in chronic obstructive pulmonary disease (COPD). Accordingly, the senescence effects of mTORC1-regulating lncRNAs were not well investigated in cancer cells. A careful examination for senescence influenced by mTORC1-regulating lncRNAs on cancer cells is warranted.
(4)
Migration by mTORC1-Regulating LncRNAs
Several lncRNA studies investigated migration-modulating effects involving mTORC1 (Table 3). In particular, migration-promoting effects of lncRNA connecting to mTORC1 were reported. RHPN1-AS1 silencing blocks the migration of nasopharyngeal cancer cells by decreasing MMP 2/9 expression [150]. LINC00958 activates the mTORC1 to promote the epithelial-mesenchymal transition (EMT) and migration of liver cancer cells [151]. A detailed assessment of migration influenced by more mTORC1-regulating lncRNAs is warranted to provide more evidence for their impacts on cancer cells in the future.

2.3.4. AKT Effector (SREBP1)-Regulating LncRNAs and Cell Functions

As mentioned above, the relationship between SREBP1, lncRNA, and cell functions was rarely reported. Other functions, such as apoptosis and autophagy, were mentioned as follows (Table 3).
(1)
Apoptosis by SREBP1-Regulating LncRNAs
Several lncRNA studies investigated apoptosis modulating effects involving SREBP1 (Table 3). The apoptosis-promoting and -suppressing effects of lncRNA connecting to SREBP1 were reported. SREBP1, SREBP2, and lncRNA ENST00000416361 were upregulated in coronary artery disease patients, accompanied by apoptosis. Inhibition of lncRNA ENST00000416361 downregulates SREBP1 and SREBP2 [152]. In contrast, free fatty acid triggers apoptosis of liver LO2 cells associated with downregulating AC012668. Overexpression of AC012668, a lncRNA, downregulates SREBP1 expression [153]. Accordingly, the relationship between SREBP1 and apoptosis warrants a detailed investigation, especially for cancer cells.
(2)
Autophagy by SREBP1-Regulating LncRNAs
Autophagy-modulating effects of lncRNAs involving SREBP1 were reported (Table 3). HAGLROS knockdown downregulates SREBP1 and induces autophagy to reduce intrahepatic cholangiocarcinoma cell proliferation [154]. A detailed assessment of autophagy influenced by more SREBP1-regulating lncRNAs is warranted to provide more evidence for their impacts on cancer cells in the future.

2.3.5. AKT Effector (HIF)-Regulating LncRNAs and Cell Functions

The role of HIF in regulating lncRNA-associated ER stress, necroptosis, DNA damage response, and senescence was rarely reported. Other functions involving HIF and lncRNA are summarized in Table 3.
(1)
Apoptosis by HIF-Regulating LncRNAs
Several lncRNA studies investigated apoptosis-modulating effects involving HIF (Table 3). The apoptosis-promoting and -suppressing effects of lncRNA connecting to HIF were reported. LncRNA TSLNC8 triggers apoptosis of lung cancer cells by regulating HIF-1α (HIF1A) signaling [155]. lincRNA-p21 is a target of p53 and HIF1A mRNA [156]. UVB upregulates lincRNA-p21 expression to induce apoptosis in keratinocytes [156]. It raises the possibility that lincRNA-p21 triggers apoptosis by regulating HIF1A. LncRNA nuclear factor of activated T cells (NFAT) silencing suppresses hypoxia-triggered apoptosis of cardiomyocytes by enhancing HIF1A expression [157].
In contrast, JPX overexpression inhibits apoptosis of nucleus pulposus cells by upregulating HIF1A [158]. Similarly, UCA1 overexpression blocks apoptosis of breast cancer cells by HIF1A inhibitor [159]. A detailed assessment of apoptosis influenced by more HIF-regulating lncRNAs is warranted to provide more evidence for their impacts on cancer cells in the future.
(2)
Autophagy by HIF-Regulating LncRNAs
Several lncRNA studies reported autophagy-modulating effects involving HIF (Table 3). The autophagy-promoting effects of lncRNA connecting to HIF were reported. Hypoxia upregulates lncRNA-MALAT1 and induces autophagy of endometrial stromal cells by upregulating HIF1A expression [160]. Hypoxia upregulates MALAT1 to trigger autophagy of vascular endothelial cell injury by downregulating HIF1A [161]. PVT1 lncRNA knockdown suppresses autophagy by downregulating HIF1A in pancreatic cancer cells [162]. A detailed assessment of autophagy influenced by more HIF-regulating lncRNAs is warranted to provide more evidence for their impacts on cancer cells in the future.
(3)
Ferroptosis by HIF-Regulating LncRNAs
Ferroptosis-modulating effects of lncRNAs involving HIF were reported (Table 3). The ferroptosis-suppressing effects of lncRNA connecting to HIF were reported. Hypoxia-upregulated HIF1A/lncRNA-PMAN suppressed ferroptosis of gastric cancer cells [163]. A detailed assessment of ferroptosis influenced by more HIF-regulating lncRNAs is warranted to provide more evidence for their impacts on cancer cells in the future.
(4)
DNA Damage Response by HIF-Regulating LncRNAs
DNA repair-modulating effects of lncRNA involving HIF were reported (Table 3). The DNA repair-suppressing effects of lncRNA connecting to HIF were reported. LncRNA HITT (HIF1A inhibitor at translation level) directly interacts with ataxia-telangiectasia mutated (ATM) and suppresses homologous recombination repair in human colon cancer tissues [164]. A detailed assessment of DNA damage response influenced by more HIF-regulating lncRNAs is warranted to provide more evidence for their impacts on cancer cells in the future.
(5)
Migration by HIF-Regulating LncRNAs
Several lncRNA studies investigated migration-modulating effects involving HIF (Table 3). The migration-promoting and -suppressing effects of lncRNA connecting to HIF were reported. HIF1A and HIF-2α can transcriptionally activate hypoxia-responsive lncRNA MALAT1 to enhance the migration of breast cancer cells [165]. LncRNA ZFPM2-AS1 enhances the migration of liver cancer cells by upregulating HIF1A [166]. LINC00649 enhances metastasis of breast cancer cells by increasing HIF1A stability [167]. LncRNA MIR17HG improves the migration of retinoblastoma cells by increasing HIF1A expression [168]. HIF1A upregulates TM4SF1-AS1 expression to enhance the migration of liver cancer cells [169]. LncRNA FAM83A-AS1 enhances the migration of lung cancer cells by upregulating HIF1A [170]. In contrast, lncRNA TSLNC8 suppresses migration effects on lung cancer cells by regulating HIF1A signaling [155]. A detailed assessment of migration influenced by more HIF-regulating lncRNAs is warranted to provide more evidence for their impacts on cancer cells in the future.

2.3.6. Potential Future Directions

As described above, several lncRNAs were mentioned to regulate AKT effectors and, in turn, control cell functions. Overexpressing or downregulating these AKT effector-regulating lncRNAs may reverse the status of cancer cell functions to improve the anticancer effects. However, the cell function mechanism for the modulating impact of lncRNAs on AKT effectors remains unclear, particularly for assessing the potential targeting to AKT effectors by lncRNAs. More experiments are warranted to improve understanding of the connection between AKT effectors and lncRNAs regulating cancer cell functions.

2.4. Connecting AKT Effectors and Database LncRNAs to Cell Functions

To further validate the relationship of AKT effectors and lncRNAs to cell functions, more AKT effector-targeting lncRNAs are required. By choosing an lncRNA database such as the LncTarD database [38], more AKT effector-targeting lncRNA candidates are generated. By individual input target genes such as “FOXO, c-Myc, mTOR, RPTOR, MLST8, AKT1S1, DEPTOR, RPS6KB1, RPS6KB2, 4EBP1, SREBF1, and HIF1A,” their respective predicted lncRNAs are generated and exported. However, these LncTarD-predicted lncRNAs for AKT effectors did not provide potential cell functions. Subsequently, these candidates were used for literature searches (Google Scholar and PubMed) to establish the connection between AKT effector-targeting lncRNAs and cell functions (Figure 2).
In addition to Table 3, several database lncRNAs also target AKT effectors, but their relationships to cell functions are not reported. Some lncRNA target information related to AKT effectors was retrieved from the LncTarD database [38] and summarized in Table 4). c-Myc upregulates several lncRNAs (PVT1, HOTTIP, CRNDE, CCAT2, HNF1A-AS1, SNHG1, NEAT1H19, CERNA2, TUG1, PCAT1, LINC-ROR, FILNC1, and THORLNC) and downregulates some lncRNAs (HULC, PCAT1, lncRNA-BCAT1, and PCAT6). S6K1 upregulates several lncRNAs (HOTAIR and PCGEM1) and downregulates some lncRNAs (RP11-708H21.4). SREBP1 upregulates lncRNA (LNCARSR). HIF upregulates several lncRNAs (HOTAIR, RAB4B-EGLN2, MEG3, and RPL13AP23) and downregulates some lncRNA (CPS1-IT1, MIR31HG, and MALAT1) [38].
mTORC1 consists of mTOR, regulatory-associated protein of mTOR (raptor; RPTOR), mammalian lethal with SEC13 protein 8 (MLST8), proline-rich AKT substrate of 40 kDa (PRAS40; AKT1S1), and DEP domain-containing mTOR-interacting protein (DEPTOR). After retrieval from the LncTarD database, other AKT effectors, such as RPTOR, MLST8, AKT1S1, DEPTOR, and S6K2, targeted by lncRNAs were not available and not shown (Table 4).
Interestingly, most AKT effector-targeting lncRNAs do not overlap, but some AKT effector-targeting lncRNAs overlap. The latter holds for PVT1, which can target the AKT effectors c-Myc and mTOR (Table 4 and Table 5). HOTAIR can target the AKT effectors (mTOR, S6K1, and HIF1A). Additionally, some lncRNAs may provide dual functions for different cancer cells. For example, HOTAIR is upregulated in cervical cancer cells but downregulated in oral cancer cells [38]. UCA1 is upregulated in bladder cancer cells but downregulated in colon cancer cells. Although the respective lncRNAs of these AKT effectors were reported, the cell functions were not connected to these AKT effector-associated lncRNAs.
Here, we summarize the literature search (Google Scholar and PubMed) and provide novel information for networking these AKT effector-associated lncRNAs and cell functions (Table 5) (Section 2.4.1, Section 2.4.2, Section 2.4.3, Section 2.4.4, Section 2.4.5 and Section 2.4.6). AKT effectors such as c-Myc, mTOR, S6K1, SREBP1, and HIF were included. c-Myc was the target for lncRNAs (PVT1, HOTTIP, CRNDE, HULC, CCAT2, HNF1A-AS1, PCAT1, SNHG1, lncRNA-BCAT1, NEAT1, H19, CERNA2, PCAT6, TUG1, LINC-ROR, FILNC1, and THORLNC). Their respective cell functions are listed in Table 5. Some AKT effector-targeting lncRNAs (Table 5), such as HULC, H19, MALAT1, ENST00113, RP11-708H21.4, GAS5, and lncRNA-p3134, are not described here because they are the same as AKT-targeting lncRNAs (Table 2). Therefore, detailed information on cell functions for lncRNAs targeting mTOR, S6K1, SREBP1, and HIF are shown, especially for cancer cells (Table 5).

2.4.1. AKT Effector (c-Myc)-Targeting LncRNAs and Cell Functions

Several c-Myc-targeting lncRNAs and their respective cell functions (Table 5) were mentioned in detail, as follows.
(1)
c-Myc-Targeting PVT1 and Cell Functions
PVT1 modulating cell functions are summarized in Table 5. PVT1 inhibits apoptosis of colon [171] and thyroid [172] cancer cells. PVT1 promotes the autophagy of liver cancer cells [173]. PVT1 upregulation suppresses inflammation-induced mitochondrial fission and enhances mitochondrial fusion of myoblasts [252]. PVT1 downregulation promotes the ferroptosis of live cancer cells [174]. Additionally, PVT1 was reported as a necroptosis-associated lncRNA of gastric cancer [175]. PVT1b, the p53-dependent PVT1 isoform, is a modulator of senescence [176]. PVT1 silencing triggers apoptosis and suppresses the radioresistance of nasopharyngeal cancer cells by inhibiting DNA repair [133]. PVT1 promotes the invasion of bladder cancer cells [177]. A careful examination of some cell functions influenced by c-Myc-targeting PVT1 is warranted to provide more evidence for their impacts on cancer cells in the future.
(2)
c-Myc-Targeting HOTTIP and Cell Functions
HOTTIP modulating cell functions are summarized in Table 5. HOTTIP silencing triggers apoptosis of human retinoblastoma cells, while HOTTIP overexpression suppresses apoptosis [178]. HOTTIP knockdown suppresses proliferation and migration but causes autophagy of renal cancer cells, reverted by autophagy inhibitor [179]. HOTTIP promotes DNA repair of UV-irradiated spermatogenic cells by upregulating γH2AX and p53 expression [180]. HOTTIP is involved in regulating senescence [181]. HOTTIP enhances the proliferation and migration of osteosarcoma cells [182]. A careful examination of some cell functions influenced by c-Myc-targeting HOTTIP is warranted to provide more evidence for their impacts on cancer cells in the future.
(3)
c-Myc-Targeting CRNDE and Cell Functions
CRNDE modulating cell functions are summarized in Table 5. CRNDE knockdown enhances apoptosis of colon cancer cells [183]. CRNDE enhances ATG4B-dependent autophagy of liver cancer cells [184]. Additionally, CRNDE silencing suppresses ER stress and the migration of endothelial cells [185]. Inhibition of CRNDE with oxaliplatin treatment enhances DNA damage and apoptosis of colon cancer cells, reverted by upregulating CRNDE with OXA oxaliplatin [186]. A careful examination of some cell functions influenced by c-Myc-targeting CRNDE is warranted to provide more evidence for their impacts on cancer cells in the future.
(4)
c-Myc-Targeting CCAT2 and HNF1A-AS1 and Cell Functions
CCAT2 and HNF1A-AS1 modulating cell functions are summarized in Table 5. CCAT2 inhibits apoptosis of colorectal cancer cells [187]. CCAT2 induces autophagy and migration of liver cancer cells [188]. HNF1A-AS1 inhibits apoptosis of bladder cancer cells [189]. HNF1A-AS1 promotes the autophagy of liver cancer cells [190]. HNF1A-AS1 enhances the invasion of lung cancer cells [191]. A careful examination of some cell functions influenced by c-Myc-targeting CCAT2 and HNF1A-AS1 is warranted to provide more evidence for their impacts on cancer cells in the future.
(5)
c-Myc-Targeting PCAT1 and Cell Functions
PCAT1 modulating cell functions are summarized in Table 5. PCAT1 knockdown triggers apoptosis of head and neck cancer cells [192]. Transcription factor AP-2 gamma (TFAP2C)-dependent PCAT1 suppresses ferroptosis of prostate cancer cells [131]. PCAT1 silencing promotes radiation-induced DNA damage [193]. PCAT1 improves the migration of laryngeal cancer cells [194]. A careful examination of some cell functions influenced by c-Myc-targeting PCAT1 is warranted to provide more evidence for their impacts on cancer cells in the future.
(6)
c-Myc-Targeting SNHG1 and LncRNA-BCAT1 and Cell Functions
SNHG1 and lncRNA-BCAT1 modulating cell functions are summarized in Table 5. SNHG1 silencing triggers apoptosis and blocks the migration of liver cancer cells [195]. SNHG1 induces autophagy and invasion of bladder cancer cells [196]. Downregulation of nonsense-mediated mRNA decay (NMD) effectors (SMG1 and SMG7) upregulate SNHG1 gene expression during ER stress [197]. lncRNA-BCAT1 upregulation decreases the proliferation and invasion of colon cancer cells [198]. A careful examination of some cell functions influenced by c-Myc-targeting SNHG1 and lncRNA-BCAT1 is warranted to provide more evidence for their impacts on cancer cells in the future.
(7)
c-Myc-Targeting NEAT1 and Cell Functions
NEAT1 modulating cell functions are summarized in Table 5. NEAT1 inhibits proliferation and migration and induces apoptosis of cervical cancer cells [199]. NEAT1 promotes autophagy of liver cancer cells to induce radioresistance [253]. NEAT1 overexpression inhibits ER stress and migration and promotes apoptosis in gastric cancer cells [200]. Additionally, NEAT1 blocks the homologous recombination of the DNA repair pathway to inhibit the proliferation of multiple myeloma [202]. NEAT1 inhibits the doxorubicin-triggered senescence of cardiomyocytes [203]. Exosome-derived NEAT1 enhances ferroptosis to promote sepsis-induced encephalopathy [201]. A careful examination of some cell functions influenced by c-Myc-targeting NEAT1 is warranted to provide more evidence for their impacts on cancer cells in the future.
(8)
c-Myc-Targeting CERNA2 and PCAT6 and Cell Functions
CERNA2 and PCAT6 modulating cell functions are summarized in Table 5. CERNA2 downregulation suppresses proliferation and triggers apoptosis of gastric cancer cells [204]. CERNA2 silencing suppresses the migration of cervical cancer cells [205]. PCAT6 suppresses apoptosis of colon cancer cells [206]. Additionally, PCAT6 induces autophagy and improves the malignancy of colon cancer cells [207]. PCAT6 was reported to be a ferroptosis-associated lncRNA for diagnosing liver cancer cells [208]. High PCAT6 levels were linked to the worse overall survival of colon cancer, accompanied by changing base excision repair and senescence [209]. PCAT6 silencing blocks the proliferation and invasion of lung cancer cells [210]. A careful examination of some cell functions influenced by c-Myc-targeting CERNA2 and PCAT6 is warranted to provide more evidence for their impacts on cancer cells in the future.
(9)
c-Myc-Targeting TUG1 and Cell Functions
TUG1 modulating cell functions are summarized in Table 5. TUG1 suppresses apoptosis of cervical cancer cells [211]. TUG1 suppresses ER stress and apoptosis of renal tubular epithelial cells [213]. TUG1 inhibits ferroptosis of hypoxia/reoxygenation treated proximal tubular epithelial cells [214]. Additionally, TUG1 silencing suppresses bupivacaine-induced DNA damage for neurotoxicity [215]. TUG1 upregulation improves the senescence of lung cancer cells [216]. TUG1 improves the autophagy of colorectal cancer cells to enhance cisplatin resistance [212]. TUG1 enhances the proliferation and invasion of osteosarcoma cells [217]. A careful examination of some cell functions influenced by c-Myc-targeting TUG1 is warranted to provide more evidence for their impacts on cancer cells in the future.
(10)
c-Myc-Targeting LINC-ROR and FILNC1 and Cell Functions
LINC-ROR and FILNC1 modulating cell functions are summarized in Table 5. Breast cancer cells highly express LINC-ROR, suppressing gemcitabine-induced autophagy and apoptosis [218]. Arsenite enhances LINC-ROR expression involved in DNA repair [219]. LINC-ROR enhances the migration of pancreatic cancer cells [220]. FILNC1 knockdown suppresses apoptosis of renal cancer cells [118]. A careful examination of some cell functions influenced by c-Myc-targeting LINC-ROR and FILNC1 is warranted to provide more evidence for their impacts on cancer cells in the future.

2.4.2. AKT Effector (mTOR)-Targeting LncRNAs and Cell Functions

Several mTOR-targeting lncRNAs and their respective cell functions (Table 5) were mentioned in detail.
(1)
mTOR-Targeting HOTAIR and Cell Functions
HOTAIR modulating cell functions are summarized in Table 5. Propofol suppresses HOTAIR to trigger apoptosis of cervical cancer cells [221]. HOTAIR promotes the autophagy of gastrointestinal stromal cancer cells to enhance their resistance to imatinib [222]. HOTAIR upregulation suppresses the paeonol-inhibiting ferroptosis of neuronal cells [224]. Additionally, DNA damage promotes HOTAIR expression in ovarian cancer cells. HOTAIR overexpression enhances DNA damage response [225]. HOTAIR improves interleukin 6 secretion after DNA damage associated with senescence [254]. Furthermore, HOTAIR knockdown inhibits autophagy and migration of cervical cancer cells [223]. A careful examination of some cell functions influenced by mTOR-targeting HOTAIR is warranted to provide more evidence for their impacts on cancer cells in the future.
(2)
mTOR-Targeting UCA1 and Cell Functions
UCA1 modulating cell functions are summarized in Table 5. Curcumin suppresses proliferation and promotes apoptosis of lung cancer cells by inhibiting UCA1 [226]. UCA1 induces autophagy of leukemia cells [227]. UCA1 inhibits ER stress to suppress ischemia/reperfusion-triggered apoptosis of cardiomyocytes [228]. UCA1 silencing upregulates dynamin-related protein 1 (DRP1) and FIS1 expression leading to mitochondria fission of pancreatic cancer cells [229]. Additionally, UCA1 silencing promotes temozolomide-induced apoptosis and DNA damage to glioma cells [230]. Coactivators of activator protein 1 (AP1) and estrogen receptor α (CAPERα) cooperate with UCA1 to induce senescence of human foreskin fibroblasts [231]. UCA1 knockdown suppresses EMT expression and migration of pulmonary fibrosis [232]. A careful examination of some cell functions influenced by mTOR-targeting UCA1 is warranted to provide more evidence for their impacts on cancer cells in the future.

2.4.3. AKT Effector (S6K1/2)-Targeting LncRNAs and Cell Functions

Several S6K1/2-targeting lncRNAs, such as RP11-708H21.4 and PCGEM1, and their respective cell functions (Table 5) were mentioned in detail. RP11-708H21.4 upregulation decreases proliferation and migration and induces apoptosis of colon cancer cells [95]. Additionally, exosomal PCGEM1 enhances interleukin-1β-induced apoptosis of chondrocytes [233]. LV3-shRNA-PCGEM1 promotes baicalein-induced autophagy of prostate cancer cells [234]. PCGEM1 enhances the proliferation and migration of cervical cancer cells [235]. A careful examination of some cell functions influenced by S6K1/2-targeting RP11-708H21.4 and PCGEM1 is warranted to provide more evidence for their impacts on cancer cells in the future.

2.4.4. AKT Effector (SREBP1)-Targeting LncRNAs and Cell Functions

The SREBP1-targeting lncRNA LNCARSR and its respective cell functions (Table 5) were mentioned in detail. LNCARSR silencing triggers apoptosis of osteosarcoma cells [236]. LNCARSR improves the proliferation and invasion of ovarian cancer cells [237]. A careful examination of some cell functions influenced by SREBP1-targeting LNCARSR is warranted to provide more evidence for their impacts on cancer cells in the future.

2.4.5. AKT Effector (HIF)-Targeting LncRNAs and Cell Functions

Several HIF-targeting lncRNAs and their respective cell functions (Table 5) were mentioned in detail.
(1)
HIF1A-Targeting CPS1-IT1 and Cell Functions
CPS1-IT1 modulating cell functions are summarized in Table 5. CPS1-IT1 overexpression triggers apoptosis of colon cancer cells, reverted by CPS1-IT1 silencing [238]. LncRNA CPS1-IT1 inhibits EMT and migration of colon cancer cells by downregulating hypoxia-induced autophagy [239]. CPS1-IT1 overexpression inhibits the proliferation and migration of glioma cells [240]. A careful examination of some cell functions influenced by HIF-targeting CPS1-IT1 is warranted to provide more evidence for their impacts on cancer cells in the future.
(2)
HIF1A-Targeting MIR31HG and Cell Functions
MIR31HG modulating cell functions are summarized in Table 5. MIR31HG improves proliferation and suppresses head and neck cancer cell apoptosis [241]. Additionally, MIR31HG silencing improves the senescence phenotype of fibroblasts [242]. MIR31HG silencing suppresses the migration of neuroblastoma cells [243]. A careful examination of some cell functions influenced by HIF1A-targeting MIR31HG is warranted to provide more evidence for their impacts on cancer cells in the future.
(3)
HIF1A-Targeting MEG3 and Cell Functions
MEG3 modulating cell functions are summarized in Table 5. MEG3 upregulation promotes ER stress-associated protein expressions and triggers apoptosis of esophageal cancer cells [244]. MEG3 overexpression triggers autophagy of ovarian cancer cells [245]. MEG3 silencing inhibits DRP1 expression and mitochondrial fission of podocytes, reverted by MEG3 overexpression [246]. Additionally, MEG3 silencing suppresses the ferroptosis of rat brain microvascular endothelial cells [247]. MEG3 triggers necroptosis of neuron cells [248]. MEG3 maintains endothelial function by modulating the DNA damage response [249]. MEG3 suppresses the senescence of vascular endothelial cells [250]. MEG3 decreases the proliferation and invasion of colon cancer cells [251]. A careful examination of more cell functions influenced by HIF1A-targeting MEG3 on cancer cells is warranted.

2.4.6. Relationship between AKT- and AKT Effector-Targeting LncRNAs

Some AKT- and AKT effector-targeting lncRNAs do not overlap, but some overlap (Table 2 and Table 4). ENST00113 and GAS5 target AKT1 and mTOR. MALAT1 targets AKT1, mTOR, and HIFA. HULC and H19 target AKT1, c-Myc, and mTOR. RP11-708H21.4 targets AKT1, mTOR, and S6K1. LncRNA-p3134 can target AKT2 and mTOR. These results provide indirect evidence that these lncRNAs may modulate AKT to regulate some AKT effectors, such as mTOR, c-Myc, and S6K1.
Some studies provide direct evidence that these lncRNAs may modulate AKT to regulate some AKT effectors. AKT and mTOR induce macrophage autophagy, as evidenced by their inhibitors [63]. ENST00113 silencing blocks the migration of HUVEC cells, accompanied by dephosphorylation of AKT and mTOR [62]. H19 upregulation improves AKT and mTOR phosphorylation to induce invasion and autophagy of trophoblast cells, reverted by H19 knockdown [104]. Consequently, some lncRNAs can modulate AKT and AKT effectors to regulate cell functions, as shown in Figure 1.
As described above, we provided comprehensive information for connecting AKT/AKT effectors with lncRNAs regulating cell functions. AKT1, AKT2, and AKT3 can control several AKT effectors. AKT stimulates mTORC1 through mTOR phosphorylation [255] and, in turn, suppresses 4EBP1 expression, a c-Myc negative regulator [256]. mTOR also phosphorylates and activates S6K1/2 [255] to upregulate SREBP1 expression [4]. mTOR upregulates HIF1A expression. Meanwhile, AKT inhibits FOXO expression [4,257]. However, the information for AKT- and its effector-targeting lncRNAs were arranged in different sections and tables, lacking a schematic summary. Therefore, we provide a schematic overview (Figure 3), including the AKT, its effectors, and all database lncRNAs mentioned, and show the points of the AKT pathway that they are involved in.

2.4.7. Potential Future Directions

As described above, a literature survey connected AKT effector-targeting database lncRNAs to several cell functions. A careful inspection is still needed before performing more experiments to validate the targeting because they are the predicted candidates. It A deeper assessment for exploring the role of AKT effectors targeted by lncRNAs in regulating cancer cell functions is warranted.

3. Connecting AKT/AKT Effectors and CircRNAs to Cell Functions

In the following, the literature survey evidence to connect AKT and circRNAs to AKT signal-modulating cell functions (Figure 1) is described later (Section 3.1).
Notably, some circRNAs were reported to modulate the expressions of AKT1 [258], AKT2 [259], and AKT3 [260]. However, their potential mechanisms still warrant a detailed exploration, particularly for the possible targeting to AKT by circRNAs. Subsequently, the potential targeting to AKT1, AKT2, and AKT3 by circRNAs and their associated cell functions are discussed in Section 3.2. By choosing the circRNA database (circBase [61]), the target information of respective circRNAs was predicted, and their impacts on cell functions were evaluated, as described later.
Some circRNAs were reported to modulate the expressions of AKT effectors [61]. However, their connection to cell functions has never been investigated, especially for cancer cells. Hence, the evidence that connects the AKT effectors and circRNAs to their modulating cell functions (Figure 1) was evaluated by literature retrieval (Section 3.3). However, their potential mechanisms still warrant a detailed exploration, particularly for the possible targeting to AKT effectors by circRNAs. Subsequently, the potential targeting to AKT effectors by circRNAs and their associated cell functions are discussed in Section 3.4. By choosing the circRNA database (circBase [61]), the target information of respective circRNAs was predicted, and their impacts on cell functions were evaluated, as described later.

3.1. Connecting AKT and CircRNAs to Cell Functions

AKT signaling and circRNAs have a cross-relationship regulating carcinogenesis [261,262,263]. AKT-regulating circRNAs are essential in controlling apoptosis, autophagy, ER stress, senescence, and migration. The connections between mitochondrial morphogenesis, ferroptosis, necroptosis, DNA damage response, and circRNAs are rarely reported. Therefore, these cell functions with regard to apoptosis, autophagy, ER stress, senescence, and migration connecting to circRNAs are described in Section 3.1.1, Section 3.1.2, Section 3.1.3, Section 3.1.4, Section 3.1.5 and Section 3.1.6, especially for cancer cells.

3.1.1. Apoptosis by AKT-Regulating CircRNAs

Apoptosis-modulating effects of circRNAs involving AKT have been reported. Several AKT-regulating circRNAs can regulate the apoptosis of cancer cells. Circ_AKT3 knockdown induces apoptosis of gastric cancer cells [264]. CircPIP5K1A overexpression suppresses apoptosis of glioma cells by phosphorylating PI3K/AKT [265]. Baicalein causes apoptosis and upregulates circHIAT1 of cervical cancer cells by dephosphorylating AKT/mTOR [266]. CircRNA_100395 overexpression induces apoptosis of gastric cancer cells by downregulating PI3K/AKT [267]. CircHIPK3 overexpression suppresses apoptosis of oral cancer cells [268]. CircRNA_0001400 knockdown triggers apoptosis of cervical cancer cells [269]. Similarly, AKT-regulating circRNAs also regulate apoptosis of noncancerous cells. CircRNA_0040414 knockdown blocks apoptosis of cardiomyocytes by downregulating PTEN and upregulating AKT [270]. Notably, some circRNA studies reported the bifunctional effects on cell functions. Circ_PRKDC knockdown induces apoptosis and autophagy in leukemia cells via dephosphorylating PI3K/AKT/mTOR [271]. Surveying more AKT-regulating circRNAs that regulate apoptosis is warranted.

3.1.2. Autophagy by AKT-Regulating CircRNAs

Some circRNAs promoting and suppressing autophagy involving AKT have been reported. Several AKT-regulating circRNAs regulate the autophagy of cancer cells. CircCDR1as induces autophagy of oral cancer cells via phosphorylating AKT and ERK1/2 [272]. RNA-binding protein FUS, overexpressed in pancreatic cancer cells, upregulates circRHOBTB3 and induces autophagy by dephosphorylating AKT [273]. Similarly, AKT-regulating circRNAs also regulate the autophagy of noncancerous cells. CircRNA_103124 overexpression in Crohn’s disease triggers autophagy by dephosphorylating AKT2 [274]. In contrast, ciRS-7 suppresses starvation-triggered autophagy of esophageal cancer cells by phosphorylating AKT [275]. CircPARD3 suppresses the autophagy of laryngeal cancer cells by phosphorylating AKT [276]. Surveying more AKT-regulating circRNAs that regulate autophagy is warranted.

3.1.3. ER Stress by AKT-Regulating CircRNAs

ER stress-modulating effects of circRNAs involving AKT have been reported in cancer cells. Overexpression of circCDR1as causes ER stress of oral cancer cells under a hypoxic microenvironment, accompanied by phosphorylating AKT [272]. A careful examination of ER stress influenced by more AKT-regulating circRNAs on cancer cells is warranted.

3.1.4. Senescence by AKT-Regulating CircRNAs

Senescence-modulating effects of circRNAs involving AKT have been reported. AKT-regulating circRNAs may regulate the senescence of noncancerous cells. Circ_FOXO3 overexpression induces cardiac senescence, and circ_FOXO3 knockdown suppresses senescence of mouse embryonic fibroblasts [277]. By contrast, the role of AKT-regulating circRNAs in regulating the senescence of cancer cells was rarely reported. Notably, AKT phosphorylating downregulates FOXO3 during cancer development [278], and therefore the role of FOXO3 in the regulation of senescence involving AKT-regulating circ_FOXO3 needs to be examined in cancer cells. This warrants a detailed assessment of senescence influenced by more AKT-regulating circRNAs on cancer cells in the future.

3.1.5. Migration by AKT-Regulating CircRNAs

Migration-modulating effects of circRNAs involving AKT have been reported in cancer cells. The migration-promoting and -suppressing effects of circRNAs connecting to AKT have been investigated. S100A4 promotes the migration of esophageal cancer cells by phosphorylating AKT [279]. ZNF139/circZNF139 enhances the migration of bladder cancer cells by phosphorylating AKT [280]. Circ_0010882 stimulates the migration of gastric cancer cells by phosphorylating AKT [261]. Similarly, circ_0002984 promotes the migration of vascular smooth muscle cells by phosphorylating AKT [281]. In contrast, circ_100395 overexpression reduced the migration of papillary thyroid cancer cells by dephosphorylating AKT [282]. This warrants surveying more AKT-regulating circRNAs that regulate migration in the future.

3.1.6. Potential Future Directions

As described above, several circRNAs were mentioned to regulate AKT phosphorylation or dephosphorylation for its activation and inactivation, thereby regulating cell functions. Overexpressing or downregulating these AKT-regulating circRNAs may reverse the status of cancer cell functions to improve the anticancer effects.
However, the cell function mechanism for the modulating effects of circRNAs on AKT remains unclear, particularly for the assessment of the potential targeting to AKT by circRNAs. More experiments are warranted to improve the connection between AKT-circRNAs regulating cancer cell functions.

3.2. Connecting AKT1/AKT2/AKT3 and Database CircRNAs to Cell Functions

Similarly to the database for lncRNA strategy (Figure 2), AKT-targeting circRNAs were retrieved from circBase [61] by individual input target genes, such as AKT1, AKT2, and AKT3, and processed through literature search by Google Scholar and PubMed to connect their respective cell functions. Since AKT1, AKT2, and AKT3 are encoded by different genes, their related modulating circRNAs are different (Table 6). The human circRNA target information for AKT1, AKT2, and AKT3 was retrieved from circBase [61].
Interestingly, the predicted human circRNA targets for AKT1, AKT2, and AKT3 are not overlapping based on circBase retrieval (Table 6). Notably, the investigation for AKT should be concerned with transcriptional regulation regarding their respective circRNAs.
Although respective circRNAs of AKT1, AKT2, and AKT3 genes were reported, the cell functions were not connected to the AKT1-, AKT2-, or AKT3-associated circRNAs in circBase [61]. Accordingly, we searched the literature via Google Scholar and PubMed and found novel information for networking the AKT1-, AKT2-, and AKT3-associated circRNAs and cell functions, especially for cancer cells.
Although many circRNAs were shown to target AKT1, AKT2, and AKT3, only some were capable of modulating cell functions (apoptosis and migration) in cancer cells based on the literature search. Several cell functions were not reported in AKT1, AKT2, and AKT3, such as autophagy, ER stress, mitochondrial morphogenesis, ferroptosis, necroptosis, DNA damage response, and senescence.
Several kinds of AKT1-, AKT2-, and AKT3-regulating circRNAs are overexpressed in many cancer cells. For example, circAKT1 (circ_0033550) is highly expressed in cervical cancer cells (Table 6) [283]. Circ_0033550 enhances cervical tumor growth. Transforming growth factor beta (TGF-β) can upregulate circ_0033550 to promote AKT1 and EMT expression in cervical cancer cells [283]. Accordingly, circAKT1 is a potential target to slow the progression of cervical cancer development.
In addition, circAKT2 (circ_0051079) is overexpressed in osteosarcoma tissues and cell lines and enhances their proliferation and metastasis (Table 6) [284]. In contrast, circAKT2 knockdown inhibits tumor growth of osteosarcoma.
Several kinds of circAKT3 show an impact on cell functions (Table 6). CircAKT3 (circ_0017252) upregulation suppresses tumor growth and metastasis of renal cancer cells by inhibiting E-cadherin degradation [285]. CircAKT3 (circ_0017247) enhances migration and invasion of melanoma cells, reverted by circ_0017247 knockdown [286]. Similarly, circ_0017247 enhances the migration of lung cancer cells by upregulating EMT [287]. CircAKT3 (circ_0000199) upregulation increases proliferation and blocks apoptosis of oral cancer cells, reverted by silencing circ_0000199 [288].
Although the database provides many AKT-targeting circRNA candidates, most of them have rarely been investigated. A careful examination of more cell functions influenced by more AKT1-, AKT2-, and AKT3-targeting circRNAs on cancer cells is warranted. Overexpressing or downregulating these AKT-regulating database circRNAs may reverse the status of cancer cell functions to modulate their anticancer effects.

3.3. Connecting AKT Effectors and CircRNAs to Cell Functions

Since AKT had a cross-relationship to circRNAs as described above, the circRNAs may exhibit the impact on AKT effectors. In the following, we summarize the evidence connecting AKT effectors to circRNAs. Only some circRNAs were reported to regulate some AKT effectors (c-Myc, mTORC1, and HIF), and other AKT effectors (FOXO, S6K1, S6K2, 4EBP1, and SREBP1) were not reported. According to our literature survey (Google Scholar and PubMed), only some circRNAs could modulate cell functions (apoptosis, autophagy, and migration) (Section 3.3.1, Section 3.3.2, Section 3.3.3 and Section 3.3.4), especially for cancer cells. Several cell functions were not reported in AKT effectors, such as ER stress, mitochondrial morphogenesis, ferroptosis, necroptosis, DNA damage response, and senescence, which were not listed.

3.3.1. Apoptosis and Migration by AKT Effector (c-Myc)-Regulating CircRNAs

As mentioned above, the relationship between c-Myc, circRNAs, and cell functions was rarely reported, except for apoptosis and migration. Some circRNAs can regulate apoptosis of cancer cells involving c-Myc. CircPVT1 silencing enhances apoptosis of acute lymphoblastic leukemia cells by downregulating c-Myc [289]. CircRHOT1 knockdown triggers apoptosis of lung cancer cells by decreasing c-Myc expression [290].
Additionally, some circRNAs can regulate migration involving c-Myc. The migration-promoting and -suppressing effects of circRNA connecting to c-Myc were reported in cancer cells. Several kinds of c-Myc-regulating circRNAs are overexpressed in many cancer cells. Modulating circRNAs may improve the anticancer effects by suppressing migration. For example, circZFR is overexpressed in liver cancer cells [291]. CircZFR knockdown inhibits the migration of liver cancer cells by downregulating c-Myc expression [291]. Similarly, circRNA_010763 is highly expressed in lung cancer cells. CircRNA_010763 improves the invasion of lung cancer cells by upregulating c-Myc expression [292]. Circ_NOTCH1 is overexpressed in nasopharyngeal [293] and gastric [294] cancer cells. Circ_NOTCH1 silencing inhibits the migration of nasopharyngeal cancer cells, where c-Myc can bind to the NOTCH1 promoter to transcriptionally activate circ_NOTCH1 [293]. Similarly, circ_NOTCH1 enhances metastasis of gastric cancer cells [294]. CircCCDC66 silencing decreases the migration of gastric cancer cells by downregulating c-Myc expression [295]. In contrast, circCDYL overexpression by plasmids blocks the migration of bladder cancer cells by reducing c-Myc expression [296]. Surveying more c-Myc-regulating circRNAs that control apoptosis and migration is warranted.

3.3.2. Apoptosis, Autophagy, and Migration by AKT Effector (mTORC1)-Regulating CircRNAs

As mentioned above, the relationship between mTORC1, circRNAs, and cell functions was rarely reported, except for apoptosis, autophagy, and migration. Several kinds of mTORC1-regulating circRNAs are overexpressed in many cancer cells. Some circRNAs can regulate apoptosis involving mTORC1. Circ_ZNF512 knockdown inhibits apoptosis to reduce myocardial tissue injury by downregulating mTORC1 [297].
Some circRNAs can regulate autophagy involving mTORC1 [297,298]. The autophagy-promoting and -suppressing effects of circRNAs connecting to mTORC1 were reported. Circ_FOXO3 enhances autophagy of brain microvascular endothelial cells by inhibiting mTORC1 [298]. In contrast, circ_ZNF512 knockdown enhances autophagy of cardiomyocytes by downregulating mTORC1 expression [297].
Some circRNAs can regulate migration involving mTORC1. LDLRAD3 silencing decreases the migration of lung cancer cells by dephosphorylating mTOR for mTORC1 inactivation [299]. A connection between migration, mTORC2, and cicrRNAs has not been published as yet. Surveying more mTORC1-regulating circRNAs that control apoptosis is warranted.

3.3.3. Apoptosis and Migration by AKT Effector (HIF)-Regulating CircRNAs

Some circRNAs can regulate apoptosis involving HIF. The apoptosis-promoting and -suppressing effects of circRNA connecting to HIF were reported. CircVEGFC improves high glucose-promoted apoptosis of vascular endothelial cells by downregulating HIF1A [300]. Circ_0010729 suppresses apoptosis of vascular endothelial cells by upregulating HIF1A [301]. In contrast, circRNA_100859 is overexpressed in colon cancer tissues and suppresses apoptosis by downregulating HIF1A [302].
Some circRNAs can regulate migration involving HIF. The migration-promoting and suppressing effects of circRNA connecting to HIF were reported. Several kinds of HIF-regulating circRNAs, such as circAGFG1, circASXL1, circ-0046600, and circPVT1, are overexpressed in many cancer cells [303,304,305,306]. Modulating HIF1A can regulate the levels of certain circRNAs. For example, circAGFG1 is highly expressed in lung cancer cells. CircAGFG1 enhances the migration of lung cancer cells by upregulating HIF1A [303]. Inhibition of circASXL1 blocks migration and HIF1A expression of lung cancer cells [304]. Circ-0046600 knockdown suppresses the migration of liver cancer cells by upregulating HIF1A [305]. Overexpressed circPVT1 enhances the migration of breast cancer cells by overexpressing HIF1A [306]. In contrast, HIF1A-regulating circRNA such as circ_EPHB4 is downregulated in cancer cells. Overexpression of circ_EPHB4, exhibiting low levels in liver cancer cells, suppressed the migration by downregulating HIF1A [307]. Surveying more HIF-regulating circRNAs that control apoptosis is warranted.

3.3.4. Potential Future Directions

As described above, several circRNAs were mentioned to regulate AKT effectors and, in turn, control cell functions. Overexpressing or downregulating these AKT effector-regulating circRNAs may reverse the status of cancer cell functions to improve anticancer effects.
However, the cell function mechanism for the modulating effects of circRNAs on AKT effectors remains unclear, particularly for the assessment of the potential targeting to AKT effectors by circRNAs. More experiments are warranted to improve the connection between AKT effectors and circRNAs regulating cancer cell functions.

3.4. Connecting AKT Effectors and Database-CircRNAs to Cell Functions

Similarly to the database lncRNA strategy (Figure 2), AKT effector-targeting circRNAs were retrieved from circBase [61]. For the input target genes FOXO, c-Myc, mTOR, RPTOR, MLST8, AKT1S1, DEPTOR, RPS6KB1, RPS6KB2, 4EBP1, SREBF1, and HIF1A, their respective predicted circRNAs were generated and exported. Subsequently, they were processed through the literature search (Google Scholar and PubMed) to connect their respective cell functions.
In addition to Section 3.3, several circRNAs also target AKT effectors, but their relationship to cell function is not reported. Some circRNA target information to AKT effectors was retrieved from circBase [61] (Table 7).
Interestingly, the AKT effector-targeting circRNAs do not overlap (Table 7). Although the respective circRNAs of these AKT effectors were reported, the cell functions were not connected to these AKT effector-associated circRNAs.
Here, we summarize the literature search on Google Scholar and PubMed, which provided novel information for networking these AKT effector-associated circRNAs and cell functions (Table 8). Several cell functions were not reported in AKT effectors, such as autophagy, ER stress, mitochondrial morphogenesis, ferroptosis, necroptosis, DNA damage response, and senescence, which were not listed (Table 8).
For example, circMYC (circ_0085533) is more expressed in melanoma tissues than in normal tissues [308]. CircMYC knockdown suppresses cell proliferation and apoptosis of melanoma cells, reverted by circMYC overexpression [308] (Table 8).
circmTOR (circ_0009805) is overexpressed in severe preeclamptic placentas [309]. CircmTOR (circ_0009792) is upregulated in the proliferation of vascular smooth muscle cells [310] (Table 8). Other cell functions were rarely reported for this circmTOR. This warrants a detailed investigation of more cell functions involving circmTOR.
circAKT1S1 (circ_0000950) is highly expressed in the cell models of Alzheimer’s disease. CircAKT1S1 induces apoptosis of neuron cells [311]. CircAKT1S1 silencing promotes proliferation and suppresses apoptosis of neurons [312] (Table 8). Accordingly, it is a potential target to slow down the progression of Alzheimer’s disease.
circHIF1A (circ_0032138) upregulation enhances proliferation and metastasis of breast cancer cells and tissue, reverted by circHIF1A silencing [313] (Table 8). CircHIF1A (circ_0006393) is downregulated in glucocorticoid-induced osteoporosis [314]. In contrast, circ_0006393 overexpression upregulates osteogenesis-associated gene expression [314].
Although the database provides many AKT-targeting circRNA candidates, most of them were rarely investigated. Only a few AKT effector-targeting circRNAs were reported to connect to some functions of cancer and noncancer cells. This warrants a detailed evaluation of more cancer cell functions in AKT effector-targeting circRNAs in the future. Overexpressing or downregulating these AKT effector-regulating database circRNAs may reverse the status of cancer cell function to improve anticancer effects.

4. Conclusions

Several lncRNAs and circRNAs may regulate numerous pathways and control diverse cell functions, which are not unique to AKT and AKT effectors. For the sake of their critical regulations, as mentioned above, this review focused on AKT and AKT effectors modulating by lncRNAs and circRNAs; however, the impact of lncRNAs and circRNAs on AKT and AKT effectors in modulating cell function remains unclear. This systematic review aimed to organize the current knowledge for connecting AKT and AKT effectors to lncRNAs and circRNAs. The collected literature herein suggests that these AKT-lncRNA, AKT-effector lncRNA, AKT circRNA, and AKT-effector circRNA connections are responsible for regulating several cancer cell functions.
Databases for lncRNAs and circRNAs, such as LincTarD and circBase, provide comprehensive AKT- and AKT effector-targeting candidates for lncRNAs and circRNAs. However, their impacts on cell functions were not provided in these databases. Accordingly, the potential regulation of cell functions for more AKT- and AKT effector-targeting lncRNAs and circRNAs warrants a detailed investigation. Our literature survey shows that these AKT- and AKT effector-targeting database lncRNAs and circRNAs are organized and connected to cancer cell functions. Notably, database-predicted AKT- and AKT effector-targeting lncRNAs and circRNAs may be derived from the literature on some cancer cell lines. Since the genetic profiles of different cancer cells are different, the database-predicted lncRNAs and circRNAs candidates for AKT and AKT effectors may be limited to some cancer cell types but not others. Similarly, the organized cell functions for the AKT- and AKT effector-regulating or -targeting lncRNAs and circRNAs were also reported from different cancer cells or specific environments. Careful assessment is still required where the targeting mechanisms are concerned.
Two gaps are still present in the present review. Several lncRNAs that regulate AKT and AKT effectors were surveyed and provided a reliable connection between each other. Although the emerging evidence was collected to provide updated information, these literature-survey lncRNAs still lack the potential targeting information. Another gap is the systemic update for the databases. It is possible that some new findings or data were not immediately updated in LncTarD or circBase. Hence, more experiments to provide validated information for mechanisms regulating AKT, AKT effectors, lncRNAs, and circRNAs are required in order to fill these gaps. Consequently, the validated information can provide the resource for updating the databases for lncRNAs and circRNAs.
In conclusion, this review provides relevant information for relating lncRNAs and circRNAs to AKT and its effectors in modulating several cancer cell functions. With the help of bioinformatics and a literature survey, the detailed mechanism of targeting information to AKT and AKT effectors was well connected to lncRNAs and circRNAs and organized to regulate cell functions. This work also sheds light on AKT-signaling studies investigating potential impacts on lncRNAs and circRNAs for regulating cancer cell functions.

Author Contributions

Conceptualization, J.-Y.T., Y.-T.C., A.A.F., H.-W.C.; methodology, Y.-T.C., J.-P.S., K.-H.Y., F.-R.C., M.-F.H.; supervision, A.A.F., H.-W.C.; writing—original draft, J.-Y.T., Y.-T.C., H.-W.C.; writing—review and editing, A.A.F., H.-W.C. All authors have read and agreed to the published version of the manuscript.

Funding

This study was partly supported by funds from the Ministry of Science and Technology (MOST 111-2320-B-037-015-MY3 and MOST 110-2314-B-037-074-MY3), Kaohsiung Medical University (KMU-DK(A)111008), and Kaohsiung Medical University Research Center (KMU-TC108A04).

Acknowledgments

The authors thank our colleague Hans-Uwe Dahms for editing the manuscript.

Conflicts of Interest

The authors declare that there are no conflicts of interest.

Abbreviations

AKT: AKT serine/threonine kinase; ASMCs: airway smooth muscle cells; AP1: activator protein 1; ATM: ataxia-telangiectasia mutated; GRP78 (BiP): glucose-regulated protein 78; CAPERα: estrogen receptor α; circRNAs: circular RNAs; COPD: chronic obstructive pulmonary disease; DEPTOR: DEP domain-containing mTOR-interacting protein; DNA-PKcs: DNA-dependent protein kinase, catalytic subunit; DRP1: dynamin-related protein 1; 4EBP1 (EIF4EBP1): eukaryotic translation initiation factor 4E-binding protein 1; EMT: epithelial–mesenchymal transition; ER: endoplasmic reticulum; HAGLROS: HAGLR opposite strand lncRNA; HIF: hypoxia-inducible factor; HIF1A: HIF-1α; FILNC1: FOXO-induced lncRNA 1; HITT: HIF1A inhibitor at translation level; FOXO: forkhead box transcription factors; lncRNAs: long noncoding RNAs; MALAT1: metastasis-associated in lung adenocarcinoma transcript 1; MFN1: mitofusin 1; MLKL: mixed lineage kinase domain-like; MLST8: mammalian lethal with SEC13 protein 8; mTOR (raptor, RPTOR): mechanistic target of rapamycin; mTORC1/2: mechanistic target of rapamycin complex 1/2; NFAT: nuclear factor of activated T cells; ncRNAs: noncoding RNAs; NORAD: noncoding RNA activated by DNA damage; NHEJ: nonhomologous end joining; NMD: mRNA decay; PDGF-BB: platelet-derived growth factor BB; PARP1: poly(ADP-ribose) polymerase 1; PI3K: phosphoinositide 3 kinase; PRAS40 (AKT1S1): proline-rich AKT substrate of 40 kDa; RIPK1: receptor-interacting serine/threonine-protein kinase 1; S6K1/2 (RPS6KB1/2): mTOR substrate S6 kinase 1/2; SERP1: stress-associated endoplasmic reticulum protein 1; SOX2-OT: SOX2 overlapping transcript; SREBP1 (SREBF1): sterol regulatory element-binding protein 1; TFAP2C: transcription factor AP-2 gamma; TGF-β: transforming growth factor beta.

References

  1. Revathidevi, S.; Munirajan, A.K. Akt in cancer: Mediator and more. Semin. Cancer Biol. 2019, 59, 80–91. [Google Scholar] [CrossRef]
  2. Manning, B.D.; Toker, A. AKT/PKB signaling: Navigating the network. Cell 2017, 169, 381–405. [Google Scholar] [CrossRef]
  3. Tang, J.Y.; Cheng, Y.B.; Chuang, Y.T.; Yang, K.H.; Chang, F.R.; Liu, W.; Chang, H.W. Oxidative stress and AKT-associated angiogenesis in a zebrafish model and its potential application for withanolides. Cells 2022, 11, 961. [Google Scholar] [CrossRef]
  4. Shiau, J.P.; Chuang, Y.T.; Cheng, Y.B.; Tang, J.Y.; Hou, M.F.; Yen, C.Y.; Chang, H.W. Impacts of oxidative stress and PI3K/AKT/mTOR on metabolism and the future direction of investigating fucoidan-modulated metabolism. Antioxidants 2022, 11, 911. [Google Scholar] [CrossRef]
  5. Kim, M.S.; Jeong, E.G.; Yoo, N.J.; Lee, S.H. Mutational analysis of oncogenic AKT E17K mutation in common solid cancers and acute leukaemias. Br. J. Cancer 2008, 98, 1533–1535. [Google Scholar] [CrossRef]
  6. Stemke-Hale, K.; Gonzalez-Angulo, A.M.; Lluch, A.; Neve, R.M.; Kuo, W.L.; Davies, M.; Carey, M.; Hu, Z.; Guan, Y.; Sahin, A.; et al. An integrative genomic and proteomic analysis of PIK3CA, PTEN, and AKT mutations in breast cancer. Cancer Res 2008, 68, 6084–6091. [Google Scholar] [CrossRef]
  7. Mundi, P.S.; Sachdev, J.; McCourt, C.; Kalinsky, K. AKT in cancer: New molecular insights and advances in drug development. Br. J. Clin. Pharm. 2016, 82, 943–956. [Google Scholar] [CrossRef]
  8. Yi, K.H.; Lauring, J. Recurrent AKT mutations in human cancers: Functional consequences and effects on drug sensitivity. Oncotarget 2016, 7, 4241–4251. [Google Scholar] [CrossRef]
  9. Khan, M.A.; Jain, V.K.; Rizwanullah, M.; Ahmad, J.; Jain, K. PI3K/AKT/mTOR pathway inhibitors in triple-negative breast cancer: A review on drug discovery and future challenges. Drug Discov. Today 2019, 24, 2181–2191. [Google Scholar] [CrossRef]
  10. Hoxhaj, G.; Manning, B.D. The PI3K-AKT network at the interface of oncogenic signalling and cancer metabolism. Nat. Rev. Cancer 2020, 20, 74–88. [Google Scholar] [CrossRef]
  11. Shiau, J.P.; Chuang, Y.T.; Yang, K.H.; Chang, F.R.; Sheu, J.H.; Hou, M.F.; Jeng, J.H.; Tang, J.Y.; Chang, H.W. Brown algae-derived fucoidan exerts oxidative stress-dependent antiproliferation on oral cancer cells. Antioxidants 2022, 11, 841. [Google Scholar] [CrossRef]
  12. Anastasiadou, E.; Jacob, L.S.; Slack, F.J. Non-coding RNA networks in cancer. Nat. Rev. Cancer 2018, 18, 5–18. [Google Scholar] [CrossRef]
  13. Yang, G.; Lu, X.; Yuan, L. LncRNA: A link between RNA and cancer. Biochim. Biophys. Acta 2014, 1839, 1097–1109. [Google Scholar] [CrossRef]
  14. Peng, W.X.; Koirala, P.; Mo, Y.Y. LncRNA-mediated regulation of cell signaling in cancer. Oncogene 2017, 36, 5661–5667. [Google Scholar] [CrossRef]
  15. Patop, I.L.; Wüst, S.; Kadener, S. Past, present, and future of circ RNA s. EMBO J. 2019, 38, e100836. [Google Scholar] [CrossRef]
  16. Zhou, W.Y.; Cai, Z.R.; Liu, J.; Wang, D.S.; Ju, H.Q.; Xu, R.H. Circular RNA: Metabolism, functions and interactions with proteins. Mol. Cancer 2020, 19, 172. [Google Scholar] [CrossRef]
  17. Lee, Y.C.; Wang, W.Y.; Lin, H.H.; Huang, Y.R.; Lin, Y.C.; Hsiao, K.Y. The functional roles and regulation of circular RNAs during cellular stresses. Non-Coding RNA 2022, 8, 38. [Google Scholar] [CrossRef]
  18. Quan, J.; Pan, X.; Zhao, L.; Li, Z.; Dai, K.; Yan, F.; Liu, S.; Ma, H.; Lai, Y. LncRNA as a diagnostic and prognostic biomarker in bladder cancer: A systematic review and meta-analysis. OncoTargets Ther. 2018, 11, 6415–6424. [Google Scholar] [CrossRef]
  19. Guglas, K.; Bogaczynska, M.; Kolenda, T.; Rys, M.; Teresiak, A.; Blizniak, R.; Lasinska, I.; Mackiewicz, J.; Lamperska, K. lncRNA in HNSCC: Challenges and potential. Contemp. Oncol. 2017, 21, 259–266. [Google Scholar] [CrossRef]
  20. Chang, Y.S.; Lee, Y.T.; Yen, J.C.; Chang, Y.C.; Lin, L.L.; Chan, W.L.; Chang, W.C.; Lin, S.Y.; Chang, J.G. Long noncoding RNA NTT context-dependently regulates MYB by interacting with activated complex in hepatocellular carcinoma cells. Front. Oncol. 2021, 11, 592045. [Google Scholar] [CrossRef]
  21. Meng, S.; Zhou, H.; Feng, Z.; Xu, Z.; Tang, Y.; Li, P.; Wu, M. CircRNA: Functions and properties of a novel potential biomarker for cancer. Mol. Cancer 2017, 16, 94. [Google Scholar] [CrossRef]
  22. Wang, H.Y.; Wang, Y.P.; Zeng, X.; Zheng, Y.; Guo, Q.H.; Ji, R.; Zhou, Y.N. Circular RNA is a popular molecule in tumors of the digestive system (Review). Int. J. Oncol. 2020, 57, 21–42. [Google Scholar] [CrossRef]
  23. Fattahi, S.; Amjadi-Moheb, F.; Tabaripour, R.; Ashrafi, G.H.; Akhavan-Niaki, H. PI3K/AKT/mTOR signaling in gastric cancer: Epigenetics and beyond. Life Sci. 2020, 262, 118513. [Google Scholar] [CrossRef]
  24. Moafian, Z.; Maghrouni, A.; Soltani, A.; Hashemy, S.I. Cross-talk between non-coding RNAs and PI3K/AKT/mTOR pathway in colorectal cancer. Mol. Biol. Rep. 2021, 48, 4797–4811. [Google Scholar] [CrossRef]
  25. Sanaei, M.J.; Baghery Saghchy Khorasani, A.; Pourbagheri-Sigaroodi, A.; Shahrokh, S.; Zali, M.R.; Bashash, D. The PI3K/Akt/mTOR axis in colorectal cancer: Oncogenic alterations, non-coding RNAs, therapeutic opportunities, and the emerging role of nanoparticles. J. Cell. Physiol. 2022, 237, 1720–1752. [Google Scholar] [CrossRef]
  26. Paraskevopoulou, M.D.; Hatzigeorgiou, A.G. Analyzing miRNA-lncRNA interactions. Methods Mol. Biol. 2016, 1402, 271–286. [Google Scholar] [CrossRef]
  27. Sakshi, S.; Jayasuriya, R.; Ganesan, K.; Xu, B.; Ramkumar, K.M. Role of circRNA-miRNA-mRNA interaction network in diabetes and its associated complications. Mol. Ther. Nucleic Acids 2021, 26, 1291–1302. [Google Scholar] [CrossRef]
  28. Statello, L.; Guo, C.J.; Chen, L.L.; Huarte, M. Gene regulation by long non-coding RNAs and its biological functions. Nat. Rev. Mol. Cell Biol. 2021, 22, 96–118. [Google Scholar] [CrossRef]
  29. Koseoglu, S.; Lu, Z.; Kumar, C.; Kirschmeier, P.; Zou, J. AKT1, AKT2 and AKT3-dependent cell survival is cell line-specific and knockdown of all three isoforms selectively induces apoptosis in 20 human tumor cell lines. Cancer Biol. Ther. 2007, 6, 755–762. [Google Scholar] [CrossRef]
  30. Brand, Y.; Levano, S.; Radojevic, V.; Naldi, A.M.; Setz, C.; Ryan, A.F.; Pak, K.; Hemmings, B.A.; Bodmer, D. All Akt isoforms (Akt1, Akt2, Akt3) are involved in normal hearing, but only Akt2 and Akt3 are involved in auditory hair cell survival in the mammalian inner ear. PLoS ONE 2015, 10, e0121599. [Google Scholar] [CrossRef]
  31. Du, Z.; Yang, D.; Zhang, Y.; Xuan, X.; Li, H.; Hu, L.; Ruan, C.; Li, L.; Chen, A.; Deng, L.; et al. AKT2 deficiency impairs formation of the BCR signalosome. Cell Commun. Signal. 2020, 18, 56. [Google Scholar] [CrossRef]
  32. Cohen, M.M., Jr. The AKT genes and their roles in various disorders. Am. J. Med. Genet. Part A 2013, 161A, 2931–2937. [Google Scholar] [CrossRef]
  33. Jaiswal, N.; Gavin, M.G.; Quinn, W.J., 3rd; Luongo, T.S.; Gelfer, R.G.; Baur, J.A.; Titchenell, P.M. The role of skeletal muscle Akt in the regulation of muscle mass and glucose homeostasis. Mol. Metab. 2019, 28, 1–13. [Google Scholar] [CrossRef]
  34. Yang, Z.Z.; Tschopp, O.; Hemmings-Mieszczak, M.; Feng, J.; Brodbeck, D.; Perentes, E.; Hemmings, B.A. Protein kinase B alpha/Akt1 regulates placental development and fetal growth. J. Biol. Chem. 2003, 278, 32124–32131. [Google Scholar] [CrossRef]
  35. Yong, H.; Wu, G.; Chen, J.; Liu, X.; Bai, Y.; Tang, N.; Liu, L.; Wei, J. lncRNA MALAT1 accelerates skeletal muscle cell apoptosis and inflammatory response in sepsis by decreasing BRCA1 expression by recruiting EZH2. Mol. Ther. Nucleic Acids 2020, 19, 97–108. [Google Scholar] [CrossRef]
  36. Qu, F.; Cao, P. Long noncoding RNA SOX2OT contributes to gastric cancer progression by sponging miR-194-5p from AKT2. Exp. Cell Res. 2018, 369, 187–196. [Google Scholar] [CrossRef]
  37. Lu, Z.; Luo, T.; Pang, T.; Du, Z.; Yin, X.; Cui, H.; Fang, G.; Xue, X. MALAT1 promotes gastric adenocarcinoma through the MALAT1/miR-181a-5p/AKT3 axis. Open Biol. 2019, 9, 190095. [Google Scholar] [CrossRef]
  38. Zhao, H.; Shi, J.; Zhang, Y.; Xie, A.; Yu, L.; Zhang, C.; Lei, J.; Xu, H.; Leng, Z.; Li, T.; et al. LncTarD: A manually-curated database of experimentally-supported functional lncRNA-target regulations in human diseases. Nucleic Acids Res. 2020, 48, D118–D126. [Google Scholar] [CrossRef]
  39. Wu, Y.; Zhang, Y.; Qin, X.; Geng, H.; Zuo, D.; Zhao, Q. PI3K/AKT/mTOR pathway-related long non-coding RNAs: Roles and mechanisms in hepatocellular carcinoma. Pharmacol. Res. Off. J. Ital. Pharmacol. Soc. 2020, 160, 105195. [Google Scholar] [CrossRef]
  40. Ghafouri-Fard, S.; Abak, A.; Tondro Anamag, F.; Shoorei, H.; Majidpoor, J.; Taheri, M. The emerging role of non-coding RNAs in the regulation of PI3K/AKT pathway in the carcinogenesis process. Biomed. Pharmacother. Biomed. Pharmacother. 2021, 137, 111279. [Google Scholar] [CrossRef]
  41. Tsai, C.Y.; Wu, J.C.C.; Fang, C.; Chang, A.Y.W. PTEN, a negative regulator of PI3K/Akt signaling, sustains brain stem cardiovascular regulation during mevinphos intoxication. Neuropharmacology 2017, 123, 175–185. [Google Scholar] [CrossRef] [PubMed]
  42. Ouyang, L.; Yang, M.; Wang, X.; Fan, J.; Liu, X.; Zhang, Y.; Shu, Y. Long noncoding RNA FER1L4 inhibits cell proliferation and promotes cell apoptosis via the PTEN/AKT/p53 signaling pathway in lung cancer. Oncol. Rep. 2021, 45, 359–367. [Google Scholar] [CrossRef] [PubMed]
  43. Zi, X.; Zhang, G.; Qiu, S. Up-regulation of LINC00619 promotes apoptosis and inhibits proliferation, migration and invasion while promoting apoptosis of osteosarcoma cells through inactivation of the HGF-mediated PI3K-Akt signalling pathway. Epigenetics Off. J. DNA Methylation Soc. 2022, 17, 147–160. [Google Scholar] [CrossRef] [PubMed]
  44. Fu, K.; Zhang, K.; Zhang, X. LncRNA HOTAIR facilitates proliferation and represses apoptosis of retinoblastoma cells through the miR-20b-5p/RRM2/PI3K/AKT axis. Orphanet J. Rare Dis. 2022, 17, 119. [Google Scholar] [CrossRef]
  45. Wang, F.; Luo, Y.; Zhang, L.; Younis, M.; Yuan, L. The LncRNA RP11-301G19.1/miR-582-5p/HMGB2 axis modulates the proliferation and apoptosis of multiple myeloma cancer cells via the PI3K/AKT signalling pathway. Cancer Gene Ther. 2022, 29, 292–303. [Google Scholar] [CrossRef]
  46. Chen, H.; Tan, X.; Ding, Y. Knockdown SNHG20 suppresses nonsmall cell lung cancer development by repressing proliferation, migration and invasion, and inducing apoptosis by regulating miR-2467-3p/E2F3. Cancer Biother. Radiopharm. 2021, 36, 360–370. [Google Scholar] [CrossRef]
  47. Li, F.; Gu, F.; Li, Q.; Zhai, C.; Gong, R.; Zhu, X. ROR1-AS1 knockdown inhibits growth and invasion and promotes apoptosis in NSCLC cells by suppression of the PI3K/Akt/mTOR pathway. J. Biochem. Mol. Toxicol. 2021, 35, e22726. [Google Scholar] [CrossRef]
  48. Fu, T.; Yang, Y.; Mu, Z.; Sun, R.; Li, X.; Dong, J. Silencing lncRNA LINC01410 suppresses cell viability yet promotes apoptosis and sensitivity to temozolomide in glioblastoma cells by inactivating PTEN/AKT pathway via targeting miR-370-3p. Immunopharmacol. Immunotoxicol. 2021, 43, 680–692. [Google Scholar] [CrossRef]
  49. Geng, S.; Tu, S.; Fu, W.; Wang, J.; Bai, Z. LncRNA PITPNA-AS1 stimulates cell proliferation and suppresses cell apoptosis in glioblastoma via targeting miR-223-3p/EGFR axis and activating PI3K/AKT signaling pathway. Cell Cycle 2021, 20, 1988–1998. [Google Scholar] [CrossRef]
  50. Tang, Z.L.; Zhang, K.; Lv, S.C.; Xu, G.W.; Zhang, J.F.; Jia, H.Y. LncRNA MEG3 suppresses PI3K/AKT/mTOR signalling pathway to enhance autophagy and inhibit inflammation in TNF-alpha-treated keratinocytes and psoriatic mice. Cytokine 2021, 148, 155657. [Google Scholar] [CrossRef]
  51. Yang, G.; Li, Z.; Dong, L.; Zhou, F. lncRNA ADAMTS9-AS1 promotes bladder cancer cell invasion, migration, and inhibits apoptosis and autophagy through PI3K/AKT/mTOR signaling pathway. Int. J. Biochem. Cell Biol. 2021, 140, 106069. [Google Scholar] [CrossRef] [PubMed]
  52. Yao, X.; Tu, Y.; Xu, Y.; Guo, Y.; Yao, F.; Zhang, X. Endoplasmic reticulum stress confers 5-fluorouracil resistance in breast cancer cell via the GRP78/OCT4/lncRNA MIAT/AKT pathway. Am. J. Cancer Res. 2020, 10, 838–855. [Google Scholar] [PubMed]
  53. Guo, Z.; Wang, Y.H.; Xu, H.; Yuan, C.S.; Zhou, H.H.; Huang, W.H.; Wang, H.; Zhang, W. LncRNA linc00312 suppresses radiotherapy resistance by targeting DNA-PKcs and impairing DNA damage repair in nasopharyngeal carcinoma. Cell Death Dis. 2021, 12, 69. [Google Scholar] [CrossRef] [PubMed]
  54. Yao, J.; Shi, Z.; Ma, X.; Xu, D.; Ming, G. lncRNA GAS5/miR-223/NAMPT axis modulates the cell proliferation and senescence of endothelial progenitor cells through PI3K/AKT signaling. J. Cell. Biochem. 2019, 120, 14518–14530. [Google Scholar] [CrossRef] [PubMed]
  55. Yin, F.; Huang, X.; Xuan, Y. Pyrroline-5-carboxylate reductase-2 promotes colorectal cancer progression via activating PI3K/AKT/mTOR pathway. Dis. Markers 2021, 2021, 9950663. [Google Scholar] [CrossRef]
  56. Xie, Z.; Zhong, C.; Shen, J.; Jia, Y.; Duan, S. LINC00963: A potential cancer diagnostic and therapeutic target. Biomed. Pharmacother. Biomed. Pharmacother. 2022, 150, 113019. [Google Scholar] [CrossRef]
  57. Zhang, W.; Yang, S.; Chen, D.; Yuwen, D.; Zhang, J.; Wei, X.; Han, X.; Guan, X. SOX2-OT induced by PAI-1 promotes triple-negative breast cancer cells metastasis by sponging miR-942-5p and activating PI3K/Akt signaling. Cell. Mol. Life Sci. 2022, 79, 59. [Google Scholar] [CrossRef]
  58. Zhang, W.; Liang, F.; Li, Q.; Sun, H.; Li, F.; Jiao, Z.; Lei, J. LncRNA MIR205HG accelerates cell proliferation, migration and invasion in hepatoblastoma through the activation of MAPK signaling pathway and PI3K/AKT signaling pathway. Biol Direct 2022, 17, 2. [Google Scholar] [CrossRef]
  59. Zhong, F.; Liu, S.; Hu, D.; Chen, L. LncRNA AC099850.3 promotes hepatocellular carcinoma proliferation and invasion through PRR11/PI3K/AKT axis and is associated with patients prognosis. J. Cancer 2022, 13, 1048–1060. [Google Scholar] [CrossRef]
  60. Wang, X.; Xu, L.; Yu, Y.; Fu, Y. LncRNA RP5-857K21.7 inhibits PDGF-BB-induced proliferation and migration of airway smooth muscle cells through the miR-508-3p/PI3K/AKT/mTOR axis. Autoimmunity 2022, 55, 65–73. [Google Scholar] [CrossRef]
  61. Glazar, P.; Papavasileiou, P.; Rajewsky, N. circBase: A database for circular RNAs. RNA 2014, 20, 1666–1670. [Google Scholar] [CrossRef] [PubMed]
  62. Yao, X.; Yan, C.; Zhang, L.; Li, Y.; Wan, Q. LncRNA ENST00113 promotes proliferation, survival, and migration by activating PI3K/Akt/mTOR signaling pathway in atherosclerosis. Medicine 2018, 97, e0473. [Google Scholar] [CrossRef] [PubMed]
  63. Zhai, C.; Cheng, J.; Mujahid, H.; Wang, H.; Kong, J.; Yin, Y.; Li, J.; Zhang, Y.; Ji, X.; Chen, W. Selective inhibition of PI3K/Akt/mTOR signaling pathway regulates autophagy of macrophage and vulnerability of atherosclerotic plaque. PLoS ONE 2014, 9, e90563. [Google Scholar] [CrossRef] [PubMed]
  64. Chen, Y.; Li, S.; Zhang, Y.; Wang, M.; Li, X.; Liu, S.; Xu, D.; Bao, Y.; Jia, P.; Wu, N.; et al. The lncRNA Malat1 regulates microvascular function after myocardial infarction in mice via miR-26b-5p/Mfn1 axis-mediated mitochondrial dynamics. Redox Biol. 2021, 41, 101910. [Google Scholar] [CrossRef]
  65. Fu, Z.; Luo, W.; Wang, J.; Peng, T.; Sun, G.; Shi, J.; Li, Z.; Zhang, B. Malat1 activates autophagy and promotes cell proliferation by sponging miR-101 and upregulating STMN1, RAB5A and ATG4D expression in glioma. Biochem. Biophys. Res. Commun. 2017, 492, 480–486. [Google Scholar] [CrossRef]
  66. Jia, Y.; Yi, L.; Li, Q.; Liu, T.; Yang, S. LncRNA MALAT1 aggravates oxygen-glucose deprivation/reoxygenation-induced neuronal endoplasmic reticulum stress and apoptosis via the miR-195a-5p/HMGA1 axis. Biol. Res. 2021, 54, 8. [Google Scholar] [CrossRef]
  67. Lin, N.; Yao, Z.; Xu, M.; Chen, J.; Lu, Y.; Yuan, L.; Zhou, S.; Zou, X.; Xu, R. Long noncoding RNA MALAT1 potentiates growth and inhibits senescence by antagonizing ABI3BP in gallbladder cancer cells. J. Exp. Clin. Cancer Res. 2019, 38, 244. [Google Scholar] [CrossRef]
  68. Yang, M.H.; Hu, Z.Y.; Xu, C.; Xie, L.Y.; Wang, X.Y.; Chen, S.Y.; Li, Z.G. MALAT1 promotes colorectal cancer cell proliferation/migration/invasion via PRKA kinase anchor protein 9. Biochim. Biophys. Acta 2015, 1852, 166–174. [Google Scholar] [CrossRef]
  69. Wang, H.; Wu, B.; Wang, H.; Jiang, C.; Liu, Z. LncRNA growth arrest specific transcript 5 inhibits the growth of pituitary neuroendocrine tumors via miR-27a-5p/cylindromatosis axis. Bioengineered 2022, 13, 10274–10286. [Google Scholar] [CrossRef]
  70. Zhang, W.Y.; Zhan, H.L.; Li, M.K.; Wu, G.D.; Liu, Z.; Wu, L.F. Long noncoding RNA Gas5 induces cell apoptosis and inhibits tumor growth via activating the CHOP-dependent endoplasmic reticulum stress pathway in human hepatoblastoma HepG2 cells. J. Cell. Biochem. 2022, 123, 231–247. [Google Scholar] [CrossRef]
  71. Liu, L.; Wang, H.J.; Meng, T.; Lei, C.; Yang, X.H.; Wang, Q.S.; Jin, B.; Zhu, J.F. lncRNA GAS5 inhibits cell migration and invasion and promotes autophagy by targeting miR-222-3p via the GAS5/PTEN-signaling pathway in CRC. Mol. Ther. Nucleic Acids 2019, 17, 644–656. [Google Scholar] [CrossRef]
  72. Li, G.; Qian, L.; Tang, X.; Chen, Y.; Zhao, Z.; Zhang, C. Long noncoding RNA growth arrestspecific 5 (GAS5) acts as a tumor suppressor by promoting autophagy in breast cancer. Mol. Med. Rep. 2020, 22, 2460–2468. [Google Scholar] [CrossRef] [PubMed]
  73. Jiang, L.; Wang, C.; Shen, X. LncRNA GAS5 suppresses ER stress induced apoptosis and inflammation by regulating SERCA2b in HG treated retinal epithelial cell. Mol. Med. Rep. 2020, 22, 1072–1080. [Google Scholar] [CrossRef] [PubMed]
  74. Zheng, Y.; Zhang, Y.; Zhang, X.; Dang, Y.; Cheng, Y.; Hua, W.; Teng, M.; Wang, S.; Lu, X. Novel lncRNA-miRNA-mRNA competing endogenous RNA triple networks associated programmed cell death in heart failure. Front. Cardiovasc. Med. 2021, 8, 747449. [Google Scholar] [CrossRef] [PubMed]
  75. Han, Y.; Wu, N.; Xia, F.; Liu, S.; Jia, D. Long noncoding RNA GAS5 regulates myocardial ischemiareperfusion injury through the PI3K/AKT apoptosis pathway by sponging miR5325p. Int. J. Mol. Med. 2020, 45, 858–872. [Google Scholar] [CrossRef] [PubMed]
  76. Ma, Y.; Yu, L.; Yan, W.; Qiu, L.; Zhang, J.; Jia, X. lncRNA GAS5 sensitizes breast cancer cells to ionizing radiation by inhibiting DNA repair. BioMed Res. Int. 2022, 2022, 1987519. [Google Scholar] [CrossRef]
  77. Chen, T.; Liang, Q.; Xu, J.; Zhang, Y.; Zhang, Y.; Mo, L.; Zhang, L. MiR-665 regulates vascular smooth muscle cell senescence by interacting With LncRNA GAS5/SDC1. Front. Cell Dev. Biol. 2021, 9, 700006. [Google Scholar] [CrossRef]
  78. Cheng, Y.; Zheng, L.; Yang, C.; Zhang, W.; Wang, H. Propofol inhibits proliferation and migration of glioma cells by up-regulating lncRNA GAS5. Toxicol. In Vitro Int. J. Publ. Assoc. BIBRA 2022, 80, 105321. [Google Scholar] [CrossRef]
  79. Zhu, L.; Zhang, Q.; Li, S.; Jiang, S.; Cui, J.; Dang, G. Interference of the long noncoding RNA CDKN2B-AS1 upregulates miR-181a-5p/TGFbetaI axis to restrain the metastasis and promote apoptosis and senescence of cervical cancer cells. Cancer Med. 2019, 8, 1721–1730. [Google Scholar] [CrossRef]
  80. Yang, M.; Yin, E.; Xu, Y.; Liu, Y.; Li, T.; Dong, Z.; Tai, W. CDKN2B antisense RNA 1 expression alleviates idiopathic pulmonary fibrosis by functioning as a competing endogenouse RNA through the miR-199a-5p/Sestrin-2 axis. Bioengineered 2022, 13, 7746–7759. [Google Scholar] [CrossRef]
  81. LIU, Y.; ZHANG, X.; REN, C.; ZHU, W.; DAI, J.; LAI, Y. Protective effect of stress-associated endoplasmic reticulum protein 1 on glucose and oxygen deprivation-induced injury in cardiomyocytes. Chin. J. Geriatr. 2019, 12, 678–682. [Google Scholar]
  82. Puvvula, P.K. LncRNAs regulatory networks in cellular senescence. Int. J. Mol. Sci. 2019, 20, 2615. [Google Scholar] [CrossRef] [PubMed]
  83. Huang, Y.; Xiang, B.; Liu, Y.; Wang, Y.; Kan, H. LncRNA CDKN2B-AS1 promotes tumor growth and metastasis of human hepatocellular carcinoma by targeting let-7c-5p/NAP1L1 axis. Cancer Lett. 2018, 437, 56–66. [Google Scholar] [CrossRef]
  84. Kong, D.; Wang, Y. Knockdown of lncRNA HULC inhibits proliferation, migration, invasion, and promotes apoptosis by sponging miR-122 in osteosarcoma. J. Cell. Biochem. 2018, 119, 1050–1061. [Google Scholar] [CrossRef]
  85. Liu, L.; Zhou, X.Y.; Zhang, J.Q.; Wang, G.G.; He, J.; Chen, Y.Y.; Huang, C.; Li, L.; Li, S.Q. LncRNA HULC promotes non-small cell lung cancer cell proliferation and inhibits the apoptosis by up-regulating sphingosine kinase 1 (SPHK1) and its downstream PI3K/Akt pathway. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 8722–8730. [Google Scholar] [CrossRef] [PubMed]
  86. Xiong, H.; Ni, Z.; He, J.; Jiang, S.; Li, X.; He, J.; Gong, W.; Zheng, L.; Chen, S.; Li, B.; et al. LncRNA HULC triggers autophagy via stabilizing Sirt1 and attenuates the chemosensitivity of HCC cells. Oncogene 2017, 36, 3528–3540. [Google Scholar] [CrossRef]
  87. Wang, L.; Li, X.; Qin, R.; Lu, Y.; Chen, Y.; Xie, S.; Jiang, X.; Lu, D. MiR-26a-1 promotes DNA damage repair by inhibiting Sirt1 and KDM5A in human liver cancer stem cells. Res. Sq. 2021; preprint. [Google Scholar] [CrossRef]
  88. Grammatikakis, I.; Panda, A.C.; Abdelmohsen, K.; Gorospe, M. Long noncoding RNAs(lncRNAs) and the molecular hallmarks of aging. Aging 2014, 6, 992–1009. [Google Scholar] [CrossRef]
  89. Feng, H.; Wei, B.; Zhang, Y. Long non-coding RNA HULC promotes proliferation, migration and invasion of pancreatic cancer cells by down-regulating microRNA-15a. Int. J. Biol. Macromol. 2019, 126, 891–898. [Google Scholar] [CrossRef]
  90. Yan, C.; Wei, S.; Han, D.; Wu, L.; Tan, L.; Wang, H.; Dong, Y.; Hua, J.; Yang, W. LncRNA HULC shRNA disinhibits miR-377-5p to suppress the growth and invasion of hepatocellular carcinoma in vitro and hepatocarcinogenesis in vivo. Ann. Transl. Med. 2020, 8, 1294. [Google Scholar] [CrossRef]
  91. Shen, Q.; Xu, Z.; Xu, S. Long noncoding RNA LUCAT1 contributes to cisplatin resistance by regulating the miR514a3p/ULK1 axis in human nonsmall cell lung cancer. Int. J. Oncol. 2020, 57, 967–979. [Google Scholar] [CrossRef]
  92. Xing, X.L.; Yao, Z.Y.; Ou, J.; Xing, C.; Li, F. Development and validation of ferroptosis-related lncRNAs prognosis signatures in kidney renal clear cell carcinoma. Cancer Cell Int. 2021, 21, 591. [Google Scholar] [CrossRef] [PubMed]
  93. Ning, J.; Sun, K.; Fan, X.; Jia, K.; Wang, X.; Ma, C.; Wei, L. Necroptosis-related lncRNAs and hepatocellular carcinoma undoubtedly secret. Res. Sq. 2022. [Google Scholar] [CrossRef]
  94. Huan, L.; Guo, T.; Wu, Y.; Xu, L.; Huang, S.; Xu, Y.; Liang, L.; He, X. Hypoxia induced LUCAT1/PTBP1 axis modulates cancer cell viability and chemotherapy response. Mol. Cancer 2020, 19, 11. [Google Scholar] [CrossRef]
  95. Sun, L.; Jiang, C.; Xu, C.; Xue, H.; Zhou, H.; Gu, L.; Liu, Y.; Xu, Q. Down-regulation of long non-coding RNA RP11-708H21.4 is associated with poor prognosis for colorectal cancer and promotes tumorigenesis through regulating AKT/mTOR pathway. Oncotarget 2017, 8, 27929–27942. [Google Scholar] [CrossRef]
  96. Sun, J.; Min, H.; Yu, L.; Yu, G.; Shi, Y.; Sun, J. The knockdown of LncRNA AFAP1-AS1 suppressed cell proliferation, migration, and invasion, and promoted apoptosis by regulating miR-545-3p/hepatoma-derived growth factor axis in lung cancer. Anti-Cancer Drugs 2021, 32, 11–21. [Google Scholar] [CrossRef]
  97. Wang, R.; Yan, Y.; Li, C. LINC00462 is involved in high glucose-induced apoptosis of renal tubular epithelial cells via AKT pathway. Cell Biol. Int. 2019. ahead of print. [Google Scholar] [CrossRef]
  98. Zhou, B.; Guo, W.; Sun, C.; Zhang, B.; Zheng, F. Linc00462 promotes pancreatic cancer invasiveness through the miR-665/TGFBR1-TGFBR2/SMAD2/3 pathway. Cell Death Dis. 2018, 9, 706. [Google Scholar] [CrossRef]
  99. Li, G.H.; Yu, J.H.; Yang, B.; Gong, F.C.; Zhang, K.W. LncRNA LOXL1-AS1 inhibited cell proliferation, migration and invasion as well as induced apoptosis in breast cancer via regulating miR-143-3p. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 10400–10409. [Google Scholar] [CrossRef]
  100. Gu, N.; Wang, X.; Di, Z.; Xiong, J.; Ma, Y.; Yan, Y.; Qian, Y.; Zhang, Q.; Yu, J. Silencing lncRNA FOXD2-AS1 inhibits proliferation, migration, invasion and drug resistance of drug-resistant glioma cells and promotes their apoptosis via microRNA-98-5p/CPEB4 axis. Aging 2019, 11, 10266–10283. [Google Scholar] [CrossRef]
  101. Guo, L.Y.; Qin, C.F.; Zou, H.X.; Song, M.Y.; Gong, M.L.; Chen, C. LncRNA AB073614 promotes the proliferation and inhibits apoptosis of cervical cancer cells by repressing RBM5. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 7201. [Google Scholar] [CrossRef]
  102. Wu, X.Y.; Zhou, H.Y.; Yao, X.M.; Chen, X.D.; Wu, J.; Lu, X.C. Long non-coding RNA AB073614 promotes metastasis of gastric cancer cells by upregulating IGF-2. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 7207. [Google Scholar] [CrossRef]
  103. Zhang, L.; Cheng, H.; Yue, Y.; Li, S.; Zhang, D.; He, R. H19 knockdown suppresses proliferation and induces apoptosis by regulating miR-148b/WNT/beta-catenin in ox-LDL-stimulated vascular smooth muscle cells. J. Biomed. Sci. 2018, 25, 11. [Google Scholar] [CrossRef]
  104. Xu, J.; Xia, Y.; Zhang, H.; Guo, H.; Feng, K.; Zhang, C. Overexpression of long non-coding RNA H19 promotes invasion and autophagy via the PI3K/AKT/mTOR pathways in trophoblast cells. Biomed. Pharmacother. 2018, 101, 691–697. [Google Scholar] [CrossRef]
  105. Li, T.; Zhang, X.; Cheng, L.; Li, C.; Wu, Z.; Luo, Y.; Zhou, K.; Li, Y.; Zhao, Q.; Huang, Y. Modulation of lncRNA H19 enhances resveratrol-inhibited cancer cell proliferation and migration by regulating endoplasmic reticulum stress. J. Cell. Mol. Med. 2022, 26, 2205–2217. [Google Scholar] [CrossRef]
  106. Sultan, H.K.; El-Ayat, W.M.; AbouGhalia, A.H.; Lasheen, N.N.; Moustafa, A.S. Study of long non-coding RNA and mitochondrial dysfunction in diabetic rats. Tissue Cell 2021, 71, 101516. [Google Scholar] [CrossRef]
  107. Zhang, R.; Pan, T.; Xiang, Y.; Zhang, M.; Xie, H.; Liang, Z.; Chen, B.; Xu, C.; Wang, J.; Huang, X.; et al. Curcumenol triggered ferroptosis in lung cancer cells via lncRNA H19/miR-19b-3p/FTH1 axis. Bioact. Mater. 2022, 13, 23–36. [Google Scholar] [CrossRef]
  108. Wang, D.; Sun, Y.; Lin, L.; Sang, Y.; Yang, F.; Zhang, J.; Jia, L.; Xu, Z.; Zhang, W. Long non-coding RNA H19 and the underlying epigenetic function in response to DNA damage of lung cancer cells. Am. J. Transl. Res. 2021, 13, 5835–5850. [Google Scholar]
  109. Zhuang, Y.; Li, T.; Xiao, H.; Wu, J.; Su, S.; Dong, X.; Hu, X.; Hua, Q.; Liu, J.; Shang, W.; et al. LncRNA-H19 drives cardiomyocyte senescence by targeting miR-19a/socs1/p53 axis. Front. Pharmacol. 2021, 12, 631835. [Google Scholar] [CrossRef]
  110. Yao, Y.; Gao, P.; Chen, L.; Wang, W.; Zhang, J.; Li, Q.; Xu, Y. Upregulated long non-coding RNA SPRY4-IT1 predicts dismal prognosis for pancreatic ductal adenocarcinoma and regulates cell proliferation and apoptosis. Gene 2018, 659, 52–58. [Google Scholar] [CrossRef]
  111. Zhao, W.; Mazar, J.; Lee, B.; Sawada, J.; Li, J.L.; Shelley, J.; Govindarajan, S.; Towler, D.; Mattick, J.S.; Komatsu, M.; et al. The long noncoding RNA SPRIGHTLY regulates cell proliferation in primary human melanocytes. J. Investig. Dermatol. 2016, 136, 819–828. [Google Scholar] [CrossRef]
  112. Li, Y.; Liao, Z.; Wang, R.; Liang, Z.; Lin, Z.; Deng, S.; Chen, L.; Liu, Z.; Feng, S. Long non-coding RNA SPRY4-IT1 promotes proliferation and metastasis in nasopharyngeal carcinoma cell. PeerJ 2022, 10, e13221. [Google Scholar] [CrossRef] [PubMed]
  113. Zhu, Q.; Lv, T.; Wu, Y.; Shi, X.; Liu, H.; Song, Y. Long non-coding RNA 00312 regulated by HOXA5 inhibits tumour proliferation and promotes apoptosis in non-small cell lung cancer. J. Cell. Mol. Med. 2017, 21, 2184–2198. [Google Scholar] [CrossRef] [PubMed]
  114. Wang, Y.Y.; Wu, Z.Y.; Wang, G.C.; Liu, K.; Niu, X.B.; Gu, S.; Meng, J.S. LINC00312 inhibits the migration and invasion of bladder cancer cells by targeting miR-197-3p. Tumour Biol. 2016, 37, 14553–14563. [Google Scholar] [CrossRef]
  115. Ruan, Y.; Lin, N.; Ma, Q.; Chen, R.; Zhang, Z.; Wen, W.; Chen, H.; Sun, J. Circulating LncRNAs analysis in patients with type 2 diabetes reveals novel genes influencing glucose metabolism and islet beta-cell function. Cell. Physiol. Biochem. 2018, 46, 335–350. [Google Scholar] [CrossRef] [PubMed]
  116. Zhao, X.; Cheng, Z.; Wang, J. Long Noncoding RNA FEZF1-AS1 promotes proliferation and inhibits apoptosis in ovarian cancer by activation of JAK-STAT3 pathway. Med. Sci. Monit. 2018, 24, 8088–8095. [Google Scholar] [CrossRef] [PubMed]
  117. Gui, Z.; Zhao, Z.; Sun, Q.; Shao, G.; Huang, J.; Zhao, W.; Kuang, Y. LncRNA FEZF1-AS1 promotes multi-drug resistance of gastric cancer cells via upregulating ATG5. Front. Cell Dev. Biol. 2021, 9, 749129. [Google Scholar] [CrossRef] [PubMed]
  118. Xiao, Z.D.; Han, L.; Lee, H.; Zhuang, L.; Zhang, Y.; Baddour, J.; Nagrath, D.; Wood, C.G.; Gu, J.; Wu, X.; et al. Energy stress-induced lncRNA FILNC1 represses c-Myc-mediated energy metabolism and inhibits renal tumor development. Nat. Commun. 2017, 8, 783. [Google Scholar] [CrossRef]
  119. Chen, Q.B.; Li, Z.H.; Fu, Y.; Lv, N.N.; Tian, N.; Han, L.; Tian, Y. Downregulated long non-coding RNA LINC00899 inhibits invasion and migration of spinal ependymoma cells via RBL2-dependent FoxO pathway. Cell Cycle 2019, 18, 2566–2579. [Google Scholar] [CrossRef]
  120. Pan, J.; Zhang, X.; Fang, X.; Xin, Z. Construction on of a ferroptosis-related lncRNA-based model to improve the prognostic evaluation of gastric cancer patients based on bioinformatics. Front. Genet. 2021, 12, 739470. [Google Scholar] [CrossRef]
  121. Angeles, A.K.; Heckmann, D.; Flosdorf, N.; Duensing, S.; Sultmann, H. The ERG-regulated LINC00920 promotes prostate cancer cell survival via the 14-3-3epsilon-FOXO pathway. Mol. Cancer Res. 2020, 18, 1545–1559. [Google Scholar] [CrossRef] [PubMed]
  122. Wu, Q.; Wei, J.; Zhao, C.; Xiang, S.; Shi, M.; Wang, Y. Targeting LncRNA EPIC1 to inhibit human colon cancer cell progression. Aging 2020, 12. ahead of print. [Google Scholar] [CrossRef]
  123. Su, W.; Guo, C.; Wang, L.; Wang, Z.; Yang, X.; Niu, F.; Tzou, D.; Yang, X.; Huang, X.; Wu, J.; et al. LncRNA MIR22HG abrogation inhibits proliferation and induces apoptosis in esophageal adenocarcinoma cells via activation of the STAT3/c-Myc/FAK signaling. Aging 2019, 11, 4587–4596. [Google Scholar] [CrossRef] [PubMed]
  124. Cheng, P.; Lu, P.; Guan, J.; Zhou, Y.; Zou, L.; Yi, X.; Cheng, H. LncRNA KCNQ1OT1 controls cell proliferation, differentiation and apoptosis by sponging miR-326 to regulate c-Myc expression in acute myeloid leukemia. Neoplasma 2020, 67, 238–248. [Google Scholar] [CrossRef] [PubMed]
  125. Zhang, M.L.; Zhao, T.T.; Du, W.W.; Yang, Z.F.; Peng, W.; Cui, Z.J. C-MYC-induced upregulation of LINC01503 promotes progression of non-small cell lung cancer. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 11120–11127. [Google Scholar] [CrossRef] [PubMed]
  126. Liu, D.; Liu, Y.; Zheng, X.; Liu, N. c-MYC-induced long noncoding RNA MEG3 aggravates kidney ischemia-reperfusion injury through activating mitophagy by upregulation of RTKN to trigger the Wnt/beta-catenin pathway. Cell Death Dis. 2021, 12, 191. [Google Scholar] [CrossRef]
  127. Capizzi, M.; Strappazzon, F.; Cianfanelli, V.; Papaleo, E.; Cecconi, F. MIR7-3HG, a MYC-dependent modulator of cell proliferation, inhibits autophagy by a regulatory loop involving AMBRA1. Autophagy 2017, 13, 554–566. [Google Scholar] [CrossRef]
  128. Song, Y.; Du, J.; Lu, P.; Zou, Q.; Zeng, S.; Liu, M.; Hu, X.; Ma, W.; Lin, H.; Liu, X.; et al. LncRNA NFYC-AS1 promotes the development of lung adenocarcinomas through autophagy, apoptosis, and MET/c-Myc oncogenic proteins. Ann. Transl. Med. 2021, 9, 1621. [Google Scholar] [CrossRef]
  129. Zhang, T.; Li, N.; Sun, C.; Jin, Y.; Sheng, X. MYC and the unfolded protein response in cancer: Synthetic lethal partners in crime? EMBO Mol. Med. 2020, 12, e11845. [Google Scholar] [CrossRef]
  130. Huang, Y.; Zhou, Z.; Zhang, J.; Hao, Z.; He, Y.; Wu, Z.; Song, Y.; Yuan, K.; Zheng, S.; Zhao, Q.; et al. lncRNA MALAT1 participates in metformin inhibiting the proliferation of breast cancer cell. J. Cell. Mol. Med. 2021, 25, 7135–7145. [Google Scholar] [CrossRef]
  131. Jiang, X.; Guo, S.; Xu, M.; Ma, B.; Liu, R.; Xu, Y.; Zhang, Y. TFAP2C-mediated lncRNA PCAT1 inhibits ferroptosis in docetaxel-resistant prostate cancer through c-Myc/miR-25-3p/SLC7A11 signaling. Front. Oncol. 2022, 12, 862015. [Google Scholar] [CrossRef] [PubMed]
  132. Tran, D.D.H.; Kessler, C.; Niehus, S.E.; Mahnkopf, M.; Koch, A.; Tamura, T. Myc target gene, long intergenic noncoding RNA, Linc00176 in hepatocellular carcinoma regulates cell cycle and cell survival by titrating tumor suppressor microRNAs. Oncogene 2018, 37, 75–85. [Google Scholar] [CrossRef] [PubMed]
  133. He, Y.; Jing, Y.; Wei, F.; Tang, Y.; Yang, L.; Luo, J.; Yang, P.; Ni, Q.; Pang, J.; Liao, Q.; et al. Long non-coding RNA PVT1 predicts poor prognosis and induces radioresistance by regulating DNA repair and cell apoptosis in nasopharyngeal carcinoma. Cell Death Dis. 2018, 9, 235. [Google Scholar] [CrossRef] [PubMed]
  134. Olivero, C.E.; Martinez-Terroba, E.; Zimmer, J.; Liao, C.; Tesfaye, E.; Hooshdaran, N.; Schofield, J.A.; Bendor, J.; Fang, D.; Simon, M.D.; et al. p53 activates the long noncoding RNA Pvt1b to inhibit Myc and suppress tumorigenesis. Mol. Cell 2020, 77, 761–774.e768. [Google Scholar] [CrossRef] [PubMed]
  135. Yu, Y.; Chen, F.; Jin, Y.; Yang, Y.; Wang, S.; Zhang, J.; Chen, C.; Zeng, Q.; Han, W.; Wang, H.; et al. Downregulated NORAD in neuroblastoma promotes cell proliferation via chromosomal instability and predicts poor prognosis. Acta Biochim. Pol. 2020, 67, 595–603. [Google Scholar] [CrossRef] [PubMed]
  136. Kozlowska, J.; Koziol, K.; Stasiak, M.; Obacz, J.; Guglas, K.; Poter, P.; Mackiewicz, A.; Kolenda, T. The role of NEAT1 lncRNA in squamous cell carcinoma of the head and neck is still difficult to define. Contemp. Oncol. 2020, 24, 96–105. [Google Scholar] [CrossRef]
  137. Vucicevic, D.; Gehre, M.; Dhamija, S.; Friis-Hansen, L.; Meierhofer, D.; Sauer, S.; Orom, U.A. The long non-coding RNA PARROT is an upstream regulator of c-Myc and affects proliferation and translation. Oncotarget 2016, 7, 33934–33947. [Google Scholar] [CrossRef]
  138. Li, B.; Zhang, G.; Wang, Z.; Yang, Y.; Wang, C.; Fang, D.; Liu, K.; Wang, F.; Mei, Y. c-Myc-activated USP2-AS1 suppresses senescence and promotes tumor progression via stabilization of E2F1 mRNA. Cell Death Dis. 2021, 12, 1006. [Google Scholar] [CrossRef]
  139. Zhen-Hua, W.; Yi-Wei, G.; Li-Qin, Z.; Jie-Yun, Z.; Zhe, G.; Wei-Jian, G. Silencing of LncRNA C1RL-AS1 suppresses the malignant phenotype in gastric cancer cells via the AKT/beta-Catenin/c-Myc pathway. Front. Oncol. 2020, 10, 1508. [Google Scholar] [CrossRef]
  140. Sang, B.; Zhang, Y.Y.; Guo, S.T.; Kong, L.F.; Cheng, Q.; Liu, G.Z.; Thorne, R.F.; Zhang, X.D.; Jin, L.; Wu, M. Dual functions for OVAAL in initiation of RAF/MEK/ERK prosurvival signals and evasion of p27-mediated cellular senescence. Proc. Natl. Acad. Sci. USA 2018, 115, E11661–E11670. [Google Scholar] [CrossRef]
  141. Wang, A.; Zhang, T.; Wei, W.; Wang, H.; Zhang, Z.; Yang, W.; Xia, W.; Mao, Q.; Xu, L.; Jiang, F.; et al. The long noncoding RNA LINC00665 facilitates c-Myc transcriptional activity via the miR-195-5p MYCBP axis to promote progression of lung adenocarcinoma. Front. Oncol. 2021, 11, 666551. [Google Scholar] [CrossRef]
  142. Zhong, Y.; Yang, L.; Xiong, F.; He, Y.; Tang, Y.; Shi, L.; Fan, S.; Li, Z.; Zhang, S.; Gong, Z.; et al. Long non-coding RNA AFAP1-AS1 accelerates lung cancer cells migration and invasion by interacting with SNIP1 to upregulate c-Myc. Signal Transduct. Target. Ther. 2021, 6, 240. [Google Scholar] [CrossRef] [PubMed]
  143. Li, Z.Y.; Xie, Y.; Deng, M.; Zhu, L.; Wu, X.; Li, G.; Shi, N.X.; Wen, C.; Huang, W.; Duan, Y.; et al. c-Myc-activated intronic miR-210 and lncRNA MIR210HG synergistically promote the metastasis of gastric cancer. Cancer Lett. 2022, 526, 322–334. [Google Scholar] [CrossRef] [PubMed]
  144. Ji, Z.; Tang, T.; Chen, M.; Dong, B.; Sun, W.; Wu, N.; Chen, H.; Feng, Q.; Yang, X.; Jin, R.; et al. C-Myc-activated long non-coding RNA LINC01050 promotes gastric cancer growth and metastasis by sponging miR-7161-3p to regulate SPZ1 expression. J. Exp. Clin. Cancer Res. 2021, 40, 351. [Google Scholar] [CrossRef] [PubMed]
  145. Wu, Z.R.; Yan, L.; Liu, Y.T.; Cao, L.; Guo, Y.H.; Zhang, Y.; Yao, H.; Cai, L.; Shang, H.B.; Rui, W.W.; et al. Inhibition of mTORC1 by lncRNA H19 via disrupting 4E-BP1/Raptor interaction in pituitary tumours. Nat. Commun. 2018, 9, 4624. [Google Scholar] [CrossRef]
  146. Fang, X.; Pan, X.; Mai, H.; Yuan, X.; Liu, S.; Wen, F. LINC00998 functions as a novel tumor suppressor in acute myeloid leukemia via regulating the ZFP36 ring finger protein/mammalian target of rapamycin complex 2 axis. Bioengineered 2021, 12, 10363–10372. [Google Scholar] [CrossRef]
  147. Li, P.; He, J.; Yang, Z.; Ge, S.; Zhang, H.; Zhong, Q.; Fan, X. ZNNT1 long noncoding RNA induces autophagy to inhibit tumorigenesis of uveal melanoma by regulating key autophagy gene expression. Autophagy 2020, 16, 1186–1199. [Google Scholar] [CrossRef]
  148. Chen, J.F.; Wu, P.; Xia, R.; Yang, J.; Huo, X.Y.; Gu, D.Y.; Tang, C.J.; De, W.; Yang, F. STAT3-induced lncRNA HAGLROS overexpression contributes to the malignant progression of gastric cancer cells via mTOR signal-mediated inhibition of autophagy. Mol. Cancer 2018, 17, 6. [Google Scholar] [CrossRef]
  149. Hu, T.J.; Huang, H.B.; Shen, H.B.; Chen, W.; Yang, Z.H. Role of long non-coding RNA MALAT1 in chronic obstructive pulmonary disease. Exp. Ther. Med. 2020, 20, 2691–2697. [Google Scholar] [CrossRef]
  150. Zhao, Y.; Zhou, H.; Dong, W. LncRNA RHPN1-AS1 promotes the progression of nasopharyngeal carcinoma by targeting CELF2 expression. Exp. Mol. Pathol. 2021, 122, 104671. [Google Scholar] [CrossRef]
  151. Lan, C.; Wang, Y.; Su, X.; Lu, J.; Ma, S. LncRNA LINC00958 activates mTORC1/P70S6K signalling pathway to promote epithelial-mesenchymal transition process in the hepatocellular carcinoma. Cancer Investig. 2021, 39, 539–549. [Google Scholar] [CrossRef]
  152. Li, P.; Yan, X.; Xu, G.; Pang, Z.; Weng, J.; Yin, J.; Li, M.; Yu, L.; Chen, Q.; Sun, K. A novel plasma lncRNA ENST00000416361 is upregulated in coronary artery disease and is related to inflammation and lipid metabolism. Mol. Med. Rep. 2020, 21, 2375–2384. [Google Scholar] [CrossRef] [Green Version]
  153. Chen, X.; Ma, H.; Gao, Y.; Jin, Y.; Ning, W.; Hou, Y.; Su, J. Long non-coding RNA AC012668 suppresses non-alcoholic fatty liver disease by competing for microRNA miR-380-5p with lipoprotein-related protein LRP2. Bioengineered 2021, 12, 6738–6747. [Google Scholar] [CrossRef]
  154. Ma, J.; Feng, J.; Zhou, X. Long non-coding RNA HAGLROS regulates lipid metabolism reprogramming in intrahepatic cholangiocarcinoma via the mTOR signaling pathway. Exp. Mol. Pathol. 2020, 115, 104466. [Google Scholar] [CrossRef]
  155. Fan, H.; Li, J.; Wang, J.; Hu, Z. Long non-coding RNAs (lncRNAs) tumor-suppressive role of lncRNA on chromosome 8p12 (TSLNC8) inhibits tumor metastasis and promotes apoptosis by regulating interleukin 6 (IL-6)/signal transducer and activator of transcription 3 (STAT3)/hypoxia-inducible factor 1-alpha (HIF-1alpha) signaling pathway in non-small cell lung cancer. Med. Sci. Monit. 2019, 25, 7624–7633. [Google Scholar] [CrossRef]
  156. Hall, J.R.; Messenger, Z.J.; Tam, H.W.; Phillips, S.L.; Recio, L.; Smart, R.C. Long noncoding RNA lincRNA-p21 is the major mediator of UVB-induced and p53-dependent apoptosis in keratinocytes. Cell Death Dis. 2015, 6, e1700. [Google Scholar] [CrossRef]
  157. Deng, X.; Liu, Y.; Xu, Z.; Wang, Z. lncRNA nuclear factor of activated T cells knockdown alleviates hypoxia/reoxygenation-induced cardiomyocyte apoptosis by upregulating HIF-1alpha expression. J. Cardiovasc. Pharmacol. 2022, 79, 479–488. [Google Scholar] [CrossRef]
  158. Yang, H.; Wang, G.; Liu, J.; Lin, M.; Chen, J.; Fang, Y.; Li, Y.; Cai, W.; Zhan, D. LncRNA JPX regulates proliferation and apoptosis of nucleus pulposus cells by targeting the miR-18a-5p/HIF-1alpha/Hippo-YAP pathway. Biochem. Biophys. Res. Commun. 2021, 566, 16–23. [Google Scholar] [CrossRef]
  159. Choudhry, H. UCA1 overexpression promotes hypoxic breast cancer cell proliferation and inhibits apoptosis via HIF-1alpha activation. J. Oncol. 2021, 2021, 5512156. [Google Scholar] [CrossRef]
  160. Liu, H.; Zhang, Z.; Xiong, W.; Zhang, L.; Du, Y.; Liu, Y.; Xiong, X. Long non-coding RNA MALAT1 mediates hypoxia-induced pro-survival autophagy of endometrial stromal cells in endometriosis. J. Cell. Mol. Med. 2019, 23, 439–452. [Google Scholar] [CrossRef]
  161. Liu, H.; Shi, C.; Deng, Y. MALAT1 affects hypoxia-induced vascular endothelial cell injury and autophagy by regulating miR-19b-3p/HIF-1alpha axis. Mol. Cell. Biochem. 2020, 466, 25–34. [Google Scholar] [CrossRef] [PubMed]
  162. Liu, Y.F.; Luo, D.; Li, X.; Li, Z.Q.; Yu, X.; Zhu, H.W. PVT1 knockdown inhibits autophagy and improves gemcitabine sensitivity by regulating the MiR-143/HIF-1alpha/VMP1 axis in pancreatic cancer. Pancreas 2021, 50, 227–234. [Google Scholar] [CrossRef] [PubMed]
  163. Lin, Z.; Song, J.; Gao, Y.; Huang, S.; Dou, R.; Zhong, P.; Huang, G.; Han, L.; Zheng, J.; Zhang, X.; et al. Hypoxia-induced HIF-1alpha/lncRNA-PMAN inhibits ferroptosis by promoting the cytoplasmic translocation of ELAVL1 in peritoneal dissemination from gastric cancer. Redox Biol. 2022, 52, 102312. [Google Scholar] [CrossRef] [PubMed]
  164. Zhao, K.; Wang, X.; Xue, X.; Li, L.; Hu, Y. A long noncoding RNA sensitizes genotoxic treatment by attenuating ATM activation and homologous recombination repair in cancers. PLoS Biol. 2020, 18, e3000666. [Google Scholar] [CrossRef] [PubMed]
  165. Shih, C.H.; Chuang, L.L.; Tsai, M.H.; Chen, L.H.; Chuang, E.Y.; Lu, T.P.; Lai, L.C. Hypoxia-induced MALAT1 promotes the proliferation and migration of breast cancer cells by sponging MiR-3064-5p. Front. Oncol. 2021, 11, 658151. [Google Scholar] [CrossRef]
  166. Song, Y.; Jin, X.; Liu, Y.; Wang, S.; Bian, F.; Zhao, Q.; Shi, H.; Gao, Z. Long noncoding RNA ZFPM2-AS1 promotes the proliferation, migration, and invasion of hepatocellular carcinoma cells by regulating the miR-576-3p/HIF-1alpha axis. Anti-Cancer Drugs 2021, 32, 812–821. [Google Scholar] [CrossRef]
  167. Zhang, J.; Du, C.; Zhang, L.; Wang, Y.; Zhang, Y.; Li, J. LncRNA LINC00649 promotes the growth and metastasis of triple-negative breast cancer by maintaining the stability of HIF-1alpha through the NF90/NF45 complex. Cell Cycle 2022, 21, 1034–1047. [Google Scholar] [CrossRef]
  168. Yan, J.; Deng, Y.X.; Cai, Y.L.; Cong, W.D. LncRNA MIR17HG promotes the proliferation, migration, and invasion of retinoblastoma cells by up-regulating HIF-1alpha expression via sponging miR-155-5p. Kaohsiung J. Med. Sci. 2022, 38, 554–564. [Google Scholar] [CrossRef]
  169. Zeng, Z.; Shi, Z.; Liu, Y.; Zhao, J.; Lu, Q.; Guo, J.; Liu, X.; Huang, D.; Xu, Q. HIF-1alpha-activated TM4SF1-AS1 promotes the proliferation, migration, and invasion of hepatocellular carcinoma cells by enhancing TM4SF1 expression. Biochem. Biophys. Res. Commun. 2021, 566, 80–86. [Google Scholar] [CrossRef]
  170. Chen, Z.; Hu, Z.; Sui, Q.; Huang, Y.; Zhao, M.; Li, M.; Liang, J.; Lu, T.; Zhan, C.; Lin, Z.; et al. LncRNA FAM83A-AS1 facilitates tumor proliferation and the migration via the HIF-1alpha/glycolysis axis in lung adenocarcinoma. Int. J. Biol. Sci. 2022, 18, 522–535. [Google Scholar] [CrossRef]
  171. Liu, Y.; Wu, Y.; Zhu, Z.; Gong, J.; Dou, W. Knockdown of lncRNA PVT1 inhibits the proliferation and accelerates the apoptosis of colorectal cancer cells via the miR761/MAPK1 axis. Mol. Med. Rep. 2021, 24, 794. [Google Scholar] [CrossRef] [PubMed]
  172. Ibrahiem, A.T.; Makhdoom, A.K.; Alanazi, K.S.; Alanazi, A.M.; Mukhlef, A.M.; Elshafey, S.H.; Toraih, E.A.; Fawzy, M.S. Analysis of anti-apoptotic PVT1 oncogene and apoptosis-related proteins (p53, Bcl2, PD-1, and PD-L1) expression in thyroid carcinoma. J. Clin. Lab. Anal. 2022, 36, e24390. [Google Scholar] [CrossRef] [PubMed]
  173. Yang, L.; Peng, X.; Jin, H.; Liu, J. Long non-coding RNA PVT1 promotes autophagy as ceRNA to target ATG3 by sponging microRNA-365 in hepatocellular carcinoma. Gene 2019, 697, 94–102. [Google Scholar] [CrossRef]
  174. He, G.N.; Bao, N.R.; Wang, S.; Xi, M.; Zhang, T.H.; Chen, F.S. Ketamine induces ferroptosis of liver cancer cells by targeting lncRNA PVT1/miR-214-3p/GPX4. Drug Des. Dev. Ther. 2021, 15, 3965–3978. [Google Scholar] [CrossRef] [PubMed]
  175. Luo, L.; Li, L.; Liu, L.; Feng, Z.; Zeng, Q.; Shu, X.; Cao, Y.; Li, Z. A necroptosis-related lncRNA-based signature to predict prognosis and probe molecular characteristics of stomach adenocarcinoma. Front. Genet. 2022, 13, 833928. [Google Scholar] [CrossRef]
  176. Tesfaye, E.; Martinez-Terroba, E.; Bendor, J.; Winkler, L.; Olivero, C.; Chen, K.; Feldser, D.M.; Zamudio, J.R.; Dimitrova, N. The p53 transcriptional response across tumor types reveals core and senescence-specific signatures modulated by long noncoding RNAs. Proc. Natl. Acad. Sci. USA 2021, 118, e2025539118. [Google Scholar] [CrossRef]
  177. Jiang, X.; Li, H.; Fang, Y.; Xu, C. LncRNA PVT1 contributes to invasion and doxorubicin resistance of bladder cancer cells through promoting MDM2 expression and AURKB-mediated p53 ubiquitination. Environ. Toxicol. 2022, 37, 1495–1508. [Google Scholar] [CrossRef]
  178. Yuan, X.; Sun, Z.; Cui, C. Knockdown of lncRNA HOTTIP inhibits retinoblastoma progression by modulating the miR-101-3p/STC1 axis. Technol. Cancer Res. Treat. 2021, 20, 1533033821997831. [Google Scholar] [CrossRef]
  179. Su, Y.; Lu, J.; Chen, X.; Liang, C.; Luo, P.; Qin, C.; Zhang, J. Long non-coding RNA HOTTIP affects renal cell carcinoma progression by regulating autophagy via the PI3K/Akt/Atg13 signaling pathway. J. Cancer Res. Clin. Oncol. 2019, 145, 573–588. [Google Scholar] [CrossRef]
  180. Liang, M.; Hu, K. Involvement of lncRNA-HOTTIP in the repair of ultraviolet light-induced DNA damage in spermatogenic cells. Mol. Cells 2019, 42, 794–803. [Google Scholar] [CrossRef]
  181. Zhang, X.; Zhang, W.; Jiang, Y.; Liu, K.; Ran, L.; Song, F. Identification of functional lncRNAs in gastric cancer by integrative analysis of GEO and TCGA data. J. Cell. Biochem. 2019, 120, 17898–17911. [Google Scholar] [CrossRef] [PubMed]
  182. Yao, X.Y.; Liu, J.F.; Luo, Y.; Xu, X.Z.; Bu, J. LncRNA HOTTIP facilitates cell proliferation, invasion, and migration in osteosarcoma by interaction with PTBP1 to promote KHSRP level. Cell Cycle 2021, 20, 283–297. [Google Scholar] [CrossRef] [PubMed]
  183. Yang, W.; Wang, Y.; Tao, C.; Li, Y.; Cao, S.; Yang, X. CRNDE silencing promotes apoptosis and enhances cisplatin sensitivity of colorectal carcinoma cells by inhibiting the Akt/mTORC1-mediated Warburg effect. Oncol. Lett. 2022, 23, 70. [Google Scholar] [CrossRef] [PubMed]
  184. Chen, L.; Sun, L.; Dai, X.; Li, T.; Yan, X.; Zhang, Y.; Xiao, H.; Shen, X.; Huang, G.; Xiang, W.; et al. LncRNA CRNDE promotes ATG4B-mediated autophagy and alleviates the sensitivity of sorafenib in hepatocellular carcinoma cells. Front. Cell Dev. Biol. 2021, 9, 687524. [Google Scholar] [CrossRef]
  185. Moran, M.; Cheng, X.; Shihabudeen Haider Ali, M.S.; Wase, N.; Nguyen, N.; Yang, W.; Zhang, C.; DiRusso, C.; Sun, X. Transcriptome analysis-identified long noncoding RNA CRNDE in maintaining endothelial cell proliferation, migration, and tube formation. Sci. Rep. 2019, 9, 19548. [Google Scholar] [CrossRef]
  186. Gao, H.; Song, X.; Kang, T.; Yan, B.; Feng, L.; Gao, L.; Ai, L.; Liu, X.; Yu, J.; Li, H. Long noncoding RNA CRNDE functions as a competing endogenous RNA to promote metastasis and oxaliplatin resistance by sponging miR-136 in colorectal cancer. OncoTargets Ther. 2017, 10, 205–216. [Google Scholar] [CrossRef]
  187. Gao, P.; Sun, D.; Guo, H.; Wu, Z.; Chen, J. LncRNA CCAT2 promotes proliferation and suppresses apoptosis of colorectal cancer cells. J. BUON 2020, 25, 1840–1846. [Google Scholar]
  188. Shi, J.; Guo, C.; Ma, J. CCAT2 enhances autophagy-related invasion and metastasis via regulating miR-4496 and ELAVL1 in hepatocellular carcinoma. J. Cell. Mol. Med. 2021, 25, 8985–8996. [Google Scholar] [CrossRef]
  189. Zhan, Y.; Li, Y.; Guan, B.; Wang, Z.; Peng, D.; Chen, Z.; He, A.; He, S.; Gong, Y.; Li, X.; et al. Long non-coding RNA HNF1A-AS1 promotes proliferation and suppresses apoptosis of bladder cancer cells through upregulating Bcl-2. Oncotarget 2017, 8, 76656–76665. [Google Scholar] [CrossRef]
  190. Liu, Z.; Wei, X.; Zhang, A.; Li, C.; Bai, J.; Dong, J. Long non-coding RNA HNF1A-AS1 functioned as an oncogene and autophagy promoter in hepatocellular carcinoma through sponging hsa-miR-30b-5p. Biochem. Biophys. Res. Commun. 2016, 473, 1268–1275. [Google Scholar] [CrossRef]
  191. Zhang, G.; An, X.; Zhao, H.; Zhang, Q.; Zhao, H. Long non-coding RNA HNF1A-AS1 promotes cell proliferation and invasion via regulating miR-17-5p in non-small cell lung cancer. Biomed. Pharmacother. 2018, 98, 594–599. [Google Scholar] [CrossRef]
  192. Sur, S.; Nakanishi, H.; Steele, R.; Ray, R.B. Depletion of PCAT-1 in head and neck cancer cells inhibits tumor growth and induces apoptosis by modulating c-Myc-AKT1-p38 MAPK signalling pathways. BMC Cancer 2019, 19, 354. [Google Scholar] [CrossRef] [PubMed]
  193. Zhang, P.; Liu, Y.; Fu, C.; Wang, C.; Duan, X.; Zou, W.; Zhao, T. Knockdown of long non-coding RNA PCAT1 in glioma stem cells promotes radiation sensitivity. Med. Mol. Morphol. 2019, 52, 114–122. [Google Scholar] [CrossRef] [PubMed]
  194. Hu, W.; Dong, N.; Huang, J.; Ye, B. Long non-coding RNA PCAT1 promotes cell migration and invasion in human laryngeal cancer by sponging miR-210-3p. J. BUON 2019, 24, 2429–2434. [Google Scholar]
  195. Meng, F.; Liu, J.; Lu, T.; Zang, L.; Wang, J.; He, Q.; Zhou, A. SNHG1 knockdown upregulates miR-376a and downregulates FOXK1/Snail axis to prevent tumor growth and metastasis in HCC. Mol. Ther. Oncolytics 2021, 21, 264–277. [Google Scholar] [CrossRef] [PubMed]
  196. Guo, C.; Li, X.; Xie, J.; Liu, D.; Geng, J.; Ye, L.; Yan, Y.; Yao, X.; Luo, M. Long noncoding RNA SNHG1 activates autophagy and promotes cell invasion in bladder cancer. Front. Oncol. 2021, 11, 660551. [Google Scholar] [CrossRef] [PubMed]
  197. Usuki, F.; Fujimura, M.; Yamashita, A. Endoplasmic reticulum stress preconditioning modifies intracellular mercury content by upregulating membrane transporters. Sci. Rep. 2017, 7, 12390. [Google Scholar] [CrossRef]
  198. Xie, F.; Xiang, X.; Huang, Q.; Ran, P.; Yuan, Y.; Li, Q.; Qi, G.; Guo, X.; Xiao, C.; Zheng, S. Reciprocal control of lncRNA-BCAT1 and beta-catenin pathway reveals lncRNA-BCAT1 long non-coding RNA acts as a tumor suppressor in colorectal cancer. Oncotarget 2017, 8, 23628–23637. [Google Scholar] [CrossRef]
  199. Geng, F.; Jia, W.C.; Li, T.; Li, N.; Wei, W. Knockdown of lncRNA NEAT1 suppresses proliferation and migration, and induces apoptosis of cervical cancer cells by regulating the miR377/FGFR1 axis. Mol. Med. Rep. 2022, 25, 10. [Google Scholar] [CrossRef]
  200. Zhou, Y.; Sha, Z.; Yang, Y.; Wu, S.; Chen, H. lncRNA NEAT1 regulates gastric carcinoma cell proliferation, invasion and apoptosis via the miR500a3p/XBP1 axis. Mol. Med. Rep. 2021, 24, 503. [Google Scholar] [CrossRef]
  201. Wei, X.B.; Jiang, W.Q.; Zeng, J.H.; Huang, L.Q.; Ding, H.G.; Jing, Y.W.; Han, Y.L.; Li, Y.C.; Chen, S.L. Exosome-derived lncRNA NEAT1 exacerbates sepsis-associated encephalopathy by promoting ferroptosis through regulating miR-9-5p/TFRC and GOT1 axis. Mol. Neurobiol. 2022, 59, 1954–1969. [Google Scholar] [CrossRef] [PubMed]
  202. Taiana, E.; Favasuli, V.; Ronchetti, D.; Todoerti, K.; Pelizzoni, F.; Manzoni, M.; Barbieri, M.; Fabris, S.; Silvestris, I.; Gallo Cantafio, M.E.; et al. Long non-coding RNA NEAT1 targeting impairs the DNA repair machinery and triggers anti-tumor activity in multiple myeloma. Leukemia 2020, 34, 234–244. [Google Scholar] [CrossRef] [PubMed]
  203. Zhuang, L.; Xia, W.; Chen, D.; Ye, Y.; Hu, T.; Li, S.; Hou, M. Exosomal LncRNA-NEAT1 derived from MIF-treated mesenchymal stem cells protected against doxorubicin-induced cardiac senescence through sponging miR-221-3p. J. Nanobiotechnol. 2020, 18, 157. [Google Scholar] [CrossRef] [PubMed]
  204. Zhang, J.; Han, X.; Jiang, L.; Han, Z.; Wang, Z. LncRNA CERNA2 is an independent predictor for clinical prognosis and is related to tumor development in gastric cancer. Int. J. Clin. Exp. Pathol. 2018, 11, 5783–5791. [Google Scholar]
  205. Wang, M.; Ouyang, J.; Li, H. CERNA2: A predictor for clinical progression and poor prognosis in cervical carcinoma. J. Cell. Biochem. 2019, 120, 11216–11221. [Google Scholar] [CrossRef]
  206. Huang, W.; Su, G.; Huang, X.; Zou, A.; Wu, J.; Yang, Y.; Zhu, Y.; Liang, S.; Li, D.; Ma, F.; et al. Long noncoding RNA PCAT6 inhibits colon cancer cell apoptosis by regulating anti-apoptotic protein ARC expression via EZH2. Cell Cycle 2019, 18, 69–83. [Google Scholar] [CrossRef]
  207. Xu, G.; Yang, M.; Wang, Q.; Zhao, L.; Zhu, S.; Zhu, L.; Xu, T.; Cao, R.; Li, C.; Liu, Q.; et al. A novel prognostic prediction model for colorectal cancer based on nine autophagy-related long noncoding RNAs. Front. Oncol. 2021, 11, 613949. [Google Scholar] [CrossRef]
  208. Fang, C.; Liu, S.; Feng, K.; Huang, C.; Zhang, Y.; Wang, J.; Lin, H.; Wang, J.; Zhong, C. Ferroptosis-related lncRNA signature predicts the prognosis and immune microenvironment of hepatocellular carcinoma. Sci. Rep. 2022, 12, 6642. [Google Scholar] [CrossRef]
  209. Han, L.; Sun, Y.; Sun, D. LncRNA PCAT6 as a predictor of poor colorectal cancer patient prognosis: A TCGA dataset analysis. Res. Sq. 2021; preprint. [Google Scholar]
  210. Wan, L.; Zhang, L.; Fan, K.; Cheng, Z.X.; Sun, Q.C.; Wang, J.J. Knockdown of long noncoding RNA PCAT6 inhibits proliferation and invasion in lung cancer cells. Oncol. Res. 2016, 24, 161–170. [Google Scholar] [CrossRef]
  211. Liu, J.; Wu, D.; Lin, X.; Hong, Y.; Wang, X.; Zheng, C.; Wu, Z.; Hong, Y.; Lv, Y. Long non-coding RNA TUG1 sponges microRNA-381-3p to facilitate cell viability and attenuate apoptosis in cervical cancer by elevating MDM2 expression. Life Sci. 2021, 267, 118902. [Google Scholar] [CrossRef]
  212. Xia, C.; Li, Q.; Cheng, X.; Wu, T.; Gao, P.; Gu, Y. Insulin-like growth factor 2 mRNA-binding protein 2-stabilized long non-coding RNA Taurine up-regulated gene 1 (TUG1) promotes cisplatin-resistance of colorectal cancer via modulating autophagy. Bioengineered 2022, 13, 2450–2469. [Google Scholar] [CrossRef] [PubMed]
  213. Meng, D.; Wu, L.; Li, Z.; Ma, X.; Zhao, S.; Zhao, D.; Qin, G. LncRNA TUG1 ameliorates diabetic nephropathy via inhibition of PU.1/RTN1 signaling pathway. J. Leukoc. Biol. 2022, 111, 553–562. [Google Scholar] [CrossRef] [PubMed]
  214. Sun, Z.; Wu, J.; Bi, Q.; Wang, W. Exosomal lncRNA TUG1 derived from human urine-derived stem cells attenuates renal ischemia/reperfusion injury by interacting with SRSF1 to regulate ASCL4 mediated ferroptosis. Res. Sq. 2022; preprint. [Google Scholar] [CrossRef] [PubMed]
  215. Lai, L.; Wang, Y.; Peng, S.; Guo, W.; Li, F.; Xu, S. P53 and taurine upregulated gene 1 promotes the repair of the deoxyribonucleic acid damage induced by bupivacaine in murine primary sensory neurons. Bioengineered 2022, 13, 7439–7456. [Google Scholar] [CrossRef]
  216. Guo, S.; Zhang, L.; Zhang, Y.; Wu, Z.; He, D.; Li, X.; Wang, Z. Long non-coding RNA TUG1 enhances chemosensitivity in non-small cell lung cancer by impairing microRNA-221-dependent PTEN inhibition. Aging 2019, 11, 7553–7569. [Google Scholar] [CrossRef]
  217. Yao, Q.; Li, Y.; Pei, Y.; Xie, B. Long non-coding RNA taurine up regulated 1 promotes osteosarcoma cell proliferation and invasion through upregulating Ezrin expression as a competing endogenous RNA of micro RNA-377-3p. Bioengineered 2022, 13, 1767–1778. [Google Scholar] [CrossRef] [PubMed]
  218. Chen, Y.M.; Liu, Y.; Wei, H.Y.; Lv, K.Z.; Fu, P.F. Large intergenic non-coding RNA-ROR reverses gemcitabine-induced autophagy and apoptosis in breast cancer cells. Oncotarget 2016, 7, 59604–59617. [Google Scholar] [CrossRef]
  219. Li, X.; Zuo, C.; Sun, D.; Zhao, T.; Zhang, Z. Arsenite increases linc-ROR in human bronchial epithelial cells that can be inhibited by antioxidant factors. Biol. Trace Elem. Res. 2020, 198, 131–141. [Google Scholar] [CrossRef]
  220. Chen, W.; Wang, H.; Liu, Y.; Xu, W.; Ling, C.; Li, Y.; Liu, J.; Chen, M.; Zhang, Y.; Chen, B.; et al. Linc-RoR promotes proliferation, migration, and invasion via the Hippo/YAP pathway in pancreatic cancer cells. J. Cell. Biochem. 2020, 121, 632–641. [Google Scholar] [CrossRef]
  221. Zhang, D.; Zhou, X.H.; Zhang, J.; Zhou, Y.X.; Ying, J.; Wu, G.Q.; Qian, J.H. Propofol promotes cell apoptosis via inhibiting HOTAIR mediated mTOR pathway in cervical cancer. Biochem. Biophys. Res. Commun. 2015, 468, 561–567. [Google Scholar] [CrossRef]
  222. Zhang, J.; Chen, K.; Tang, Y.; Luan, X.; Zheng, X.; Lu, X.; Mao, J.; Hu, L.; Zhang, S.; Zhang, X.; et al. LncRNA-HOTAIR activates autophagy and promotes the imatinib resistance of gastrointestinal stromal tumor cells through a mechanism involving the miR-130a/ATG2B pathway. Cell Death Dis. 2021, 12, 367. [Google Scholar] [CrossRef] [PubMed]
  223. Guo, X.; Xiao, H.; Guo, S.; Li, J.; Wang, Y.; Chen, J.; Lou, G. Long noncoding RNA HOTAIR knockdown inhibits autophagy and epithelial-mesenchymal transition through the Wnt signaling pathway in radioresistant human cervical cancer HeLa cells. J. Cell. Physiol. 2019, 234, 3478–3489. [Google Scholar] [CrossRef] [PubMed]
  224. Jin, Z.L.; Gao, W.Y.; Liao, S.J.; Yu, T.; Shi, Q.; Yu, S.Z.; Cai, Y.F. Paeonol inhibits the progression of intracerebral haemorrhage by mediating the HOTAIR/UPF1/ACSL4 axis. ASN Neuro 2021, 13, 17590914211010647. [Google Scholar] [CrossRef] [PubMed]
  225. Ozes, A.R.; Miller, D.F.; Ozes, O.N.; Fang, F.; Liu, Y.; Matei, D.; Huang, T.; Nephew, K.P. NF-kappaB-HOTAIR axis links DNA damage response, chemoresistance and cellular senescence in ovarian cancer. Oncogene 2016, 35, 5350–5361. [Google Scholar] [CrossRef] [PubMed]
  226. Wang, W.H.; Chen, J.; Zhang, B.R.; Lu, S.J.; Wang, F.; Peng, L.; Dai, J.H.; Sun, Y.Z. Curcumin inhibits proliferation and enhances apoptosis in A549 cells by downregulating lncRNA UCA1. Die Pharm. 2018, 73, 402–407. [Google Scholar] [CrossRef]
  227. Li, J.J.; Chen, X.F.; Wang, M.; Zhang, P.P.; Zhang, F.; Zhang, J.J. Long non-coding RNA UCA1 promotes autophagy by targeting miR-96-5p in acute myeloid leukaemia. Clin. Exp. Pharmacol. Physiol. 2020, 47, 877–885. [Google Scholar] [CrossRef]
  228. Chen, J.; Hu, Q.; Zhang, B.F.; Liu, X.P.; Yang, S.; Jiang, H. Long noncoding RNA UCA1 inhibits ischaemia/reperfusion injury induced cardiomyocytes apoptosis via suppression of endoplasmic reticulum stress. Genes Genom. 2019, 41, 803–810. [Google Scholar] [CrossRef]
  229. Teng, B.; Feng, T.; Li, W.; Wang, Z. Abnormal expression of lncRNA UCA1 disturbed cell apoptosis through mediating mitochondrial dynamics in PDAC. Neoplasma 2021, 68, 334–341. [Google Scholar] [CrossRef]
  230. Cheng, M.; Wang, Q.; Chen, L.; Zhao, D.; Tang, J.; Xu, J.; He, Z. LncRNA UCA1/miR-182-5p/MGMT axis modulates glioma cell sensitivity to temozolomide through MGMT-related DNA damage pathways. Hum. Pathol. 2022, 123, 59–73. [Google Scholar] [CrossRef]
  231. Kumar, P.P.; Emechebe, U.; Smith, R.; Franklin, S.; Moore, B.; Yandell, M.; Lessnick, S.L.; Moon, A.M. Coordinated control of senescence by lncRNA and a novel T-box3 co-repressor complex. eLife 2014, 3, e02805. [Google Scholar] [CrossRef]
  232. Yang, G.; Tian, Y.; Li, C.; Xia, J.; Qi, Y.; Yao, W.; Hao, C. LncRNA UCA1 regulates silicosis-related lung epithelial cell-to-mesenchymal transition through competitive adsorption of miR-204-5p. Toxicol. Appl. Pharmacol. 2022, 441, 115977. [Google Scholar] [CrossRef] [PubMed]
  233. Zeng, G.; Deng, G.; Xiao, S.; Li, F. Fibroblast-like synoviocytes-derived exosomal PCGEM1 accelerates il-1beta-induced apoptosis and cartilage matrix degradation by miR-142-5p/RUNX2 in chondrocytes. Immunol. Investig. 2021, 51, 1–18. [Google Scholar] [CrossRef]
  234. Han, Z.; He, J.; Zou, M.; Chen, W.; Lv, Y.; Li, Y. Small interfering RNA target for long noncoding RNA PCGEM1 increases the sensitivity of LNCaP cells to baicalein. Anat. Rec. 2020, 303, 2077–2085. [Google Scholar] [CrossRef] [PubMed]
  235. Zhang, Q.; Zheng, J.; Liu, L. The long noncoding RNA PCGEM1 promotes cell proliferation, migration and invasion via targeting the miR-182/FBXW11 axis in cervical cancer. Cancer Cell Int. 2019, 19, 304. [Google Scholar] [CrossRef]
  236. Shen, P.; Cheng, Y. Long noncoding RNA lncARSR confers resistance to Adriamycin and promotes osteosarcoma progression. Cell Death Dis. 2020, 11, 362. [Google Scholar] [CrossRef]
  237. Shu, C.; Yan, D.; Mo, Y.; Gu, J.; Shah, N.; He, J. Long noncoding RNA lncARSR promotes epithelial ovarian cancer cell proliferation and invasion by association with HuR and miR-200 family. Am. J. Cancer Res. 2018, 8, 981–992. [Google Scholar]
  238. Zhang, W.; Yuan, W.; Song, J.; Wang, S.; Gu, X. LncRna CPS1-IT1 suppresses cell proliferation, invasion and metastasis in colorectal cancer. Cell. Physiol. Biochem. 2017, 44, 567–580. [Google Scholar] [CrossRef]
  239. Zhang, W.; Yuan, W.; Song, J.; Wang, S.; Gu, X. LncRNA CPS1-IT1 suppresses EMT and metastasis of colorectal cancer by inhibiting hypoxia-induced autophagy through inactivation of HIF-1alpha. Biochimie 2018, 144, 21–27. [Google Scholar] [CrossRef]
  240. Chen, H.; Li, Q.; Liang, J.; Jin, M.; Lu, A. LncRNA CPS1-IT1 serves as anti-oncogenic role in glioma. Biomed. Pharmacother. 2019, 118, 109277. [Google Scholar] [CrossRef]
  241. Wang, R.; Ma, Z.; Feng, L.; Yang, Y.; Tan, C.; Shi, Q.; Lian, M.; He, S.; Ma, H.; Fang, J. LncRNA MIR31HG targets HIF1A and P21 to facilitate head and neck cancer cell proliferation and tumorigenesis by promoting cell-cycle progression. Mol. Cancer 2018, 17, 162. [Google Scholar] [CrossRef]
  242. Montes, M.; Nielsen, M.M.; Maglieri, G.; Jacobsen, A.; Hojfeldt, J.; Agrawal-Singh, S.; Hansen, K.; Helin, K.; van de Werken, H.J.G.; Pedersen, J.S.; et al. The lncRNA MIR31HG regulates p16(INK4A) expression to modulate senescence. Nat. Commun. 2015, 6, 6967. [Google Scholar] [CrossRef] [PubMed]
  243. Cai, P.; Li, H.; Huo, W.; Zhu, H.; Xu, C.; Zang, R.; Lv, W.; Xia, Y.; Tang, W. Aberrant expression of LncRNA-MIR31HG regulates cell migration and proliferation by affecting miR-31 and miR-31* in Hirschsprung’s disease. J. Cell. Biochem. 2018, 119, 8195–8203. [Google Scholar] [CrossRef] [PubMed]
  244. Huang, Z.-L.; Chen, R.-P.; Zhou, X.-T.; Zhan, H.-L.; Hu, M.-M.; Liu, B.; Wu, G.-D.; Wu, L.-F. Long non-coding RNA MEG3 induces cell apoptosis in esophageal cancer through endoplasmic reticulum stress. Oncol. Rep. 2017, 37, 3093–3099. [Google Scholar] [CrossRef]
  245. Xiu, Y.L.; Sun, K.X.; Chen, X.; Chen, S.; Zhao, Y.; Guo, Q.G.; Zong, Z.H. Upregulation of the lncRNA Meg3 induces autophagy to inhibit tumorigenesis and progression of epithelial ovarian carcinoma by regulating activity of ATG3. Oncotarget 2017, 8, 31714–31725. [Google Scholar] [CrossRef]
  246. Deng, Q.; Wen, R.; Liu, S.; Chen, X.; Song, S.; Li, X.; Su, Z.; Wang, C. Increased long noncoding RNA maternally expressed gene 3 contributes to podocyte injury induced by high glucose through regulation of mitochondrial fission. Cell Death Dis. 2020, 11, 814. [Google Scholar] [CrossRef]
  247. Chen, C.; Huang, Y.; Xia, P.; Zhang, F.; Li, L.; Wang, E.; Guo, Q.; Ye, Z. Long noncoding RNA Meg3 mediates ferroptosis induced by oxygen and glucose deprivation combined with hyperglycemia in rat brain microvascular endothelial cells, through modulating the p53/GPX4 axis. Eur. J. Histochem. 2021, 65, 3224. [Google Scholar] [CrossRef] [PubMed]
  248. Balusu, S.; Horré, K.; Thrupp, N.; Snellinx, A.; Serneels, L.; Chrysidou, I.; Arranz, A.M.; Sierksma, A.; Simrén, J.; Karikari, T.K. Long noncoding RNA MEG3 activates neuronal necroptosis in Alzheimer’s disease. BioRxiv, 2022; preprint. [Google Scholar] [CrossRef]
  249. Shihabudeen Haider Ali, M.S.; Cheng, X.; Moran, M.; Haemmig, S.; Naldrett, M.J.; Alvarez, S.; Feinberg, M.W.; Sun, X. LncRNA Meg3 protects endothelial function by regulating the DNA damage response. Nucleic Acids Res. 2019, 47, 1505–1522. [Google Scholar] [CrossRef]
  250. Lan, Y.; Li, Y.J.; Li, D.J.; Li, P.; Wang, J.Y.; Diao, Y.P.; Ye, G.D.; Li, Y.F. Long noncoding RNA MEG3 prevents vascular endothelial cell senescence by impairing miR-128-dependent Girdin downregulation. Am. J. Physiol. Cell Physiol. 2019, 316, C830–C843. [Google Scholar] [CrossRef]
  251. Wang, G.; Ye, Q.; Ning, S.; Yang, Z.; Chen, Y.; Zhang, L.; Huang, Y.; Xie, F.; Cheng, X.; Chi, J.; et al. LncRNA MEG3 promotes endoplasmic reticulum stress and suppresses proliferation and invasion of colorectal carcinoma cells through the MEG3/miR-103a-3p/PDHB ceRNA pathway. Neoplasma 2021, 68, 362–374. [Google Scholar] [CrossRef]
  252. Li, F.; Liu, J.; Tang, S.; Yan, J.; Chen, H.; Li, D.; Yan, X. Quercetin regulates inflammation, oxidative stress, apoptosis, and mitochondrial structure and function in H9C2 cells by promoting PVT1 expression. Acta Histochem. 2021, 123, 151819. [Google Scholar] [CrossRef] [PubMed]
  253. Sakaguchi, H.; Tsuchiya, H.; Kitagawa, Y.; Tanino, T.; Yoshida, K.; Uchida, N.; Shiota, G. NEAT1 confers radioresistance to hepatocellular carcinoma cells by inducing autophagy through GABARAP. Int. J. Mol. Sci. 2022, 23, 711. [Google Scholar] [CrossRef] [PubMed]
  254. De Visser, K.E.; Eichten, A.; Coussens, L.M. Paradoxical roles of the immune system during cancer development. Nat. Rev. Cancer 2006, 6, 24–37. [Google Scholar] [CrossRef]
  255. Paez, J.; Sellers, W.R. PI3K/PTEN/Akt Pathway. In Signal Transduction in Cancer; Frank, D.A., Ed.; Springer: Boston, MA, USA, 2004; pp. 145–167. [Google Scholar] [CrossRef]
  256. Chang, T.M.; Shan, Y.S.; Chu, P.Y.; Jiang, S.S.; Hung, W.C.; Chen, Y.L.; Tu, H.C.; Lin, H.Y.; Tsai, H.J.; Chen, L.T. The regulatory role of aberrant Phosphatase and Tensin Homologue and Liver Kinase B1 on AKT/mTOR/c-Myc axis in pancreatic neuroendocrine tumors. Oncotarget 2017, 8, 98068–98083. [Google Scholar] [CrossRef]
  257. Farhan, M.; Wang, H.; Gaur, U.; Little, P.J.; Xu, J.; Zheng, W. FOXO signaling pathways as therapeutic targets in cancer. Int. J. Biol. Sci. 2017, 13, 815–827. [Google Scholar] [CrossRef] [PubMed]
  258. Zeng, Y.; Du, W.W.; Wu, Y.; Yang, Z.; Awan, F.M.; Li, X.; Yang, W.; Zhang, C.; Yang, Q.; Yee, A.; et al. A circular RNA binds to and activates AKT phosphorylation and nuclear localization reducing apoptosis and enhancing cardiac repair. Theranostics 2017, 7, 3842–3855. [Google Scholar] [CrossRef]
  259. Tu, F.L.; Guo, X.Q.; Wu, H.X.; He, Z.Y.; Wang, F.; Sun, A.J.; Dai, X.D. Circ-0001313/miRNA-510-5p/AKT2 axis promotes the development and progression of colon cancer. Am. J. Transl. Res. 2020, 12, 281–291. [Google Scholar] [PubMed]
  260. Yue, B.; Wang, J.; Ru, W.; Wu, J.; Cao, X.; Yang, H.; Huang, Y.; Lan, X.; Lei, C.; Huang, B.; et al. The circular RNA circHUWE1 sponges the miR-29b-AKT3 axis to regulate myoblast development. Mol. Ther. Nucleic Acids 2020, 19, 1086–1097. [Google Scholar] [CrossRef]
  261. Peng, Y.K.; Pu, K.; Su, H.X.; Zhang, J.; Zheng, Y.; Ji, R.; Guo, Q.H.; Wang, Y.P.; Guan, Q.L.; Zhou, Y.N. Circular RNA hsa_circ_0010882 promotes the progression of gastric cancer via regulation of the PI3K/Akt/mTOR signaling pathway. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 1142–1151. [Google Scholar] [CrossRef]
  262. Ghafouri-Fard, S.; Khoshbakht, T.; Taheri, M.; Jamali, E. CircITCH: A circular RNA with eminent roles in the carcinogenesis. Front. Oncol. 2021, 11, 774979. [Google Scholar] [CrossRef]
  263. Xue, C.; Li, G.; Lu, J.; Li, L. Crosstalk between circRNAs and the PI3K/AKT signaling pathway in cancer progression. Signal Transduct. Target. Ther. 2021, 6, 400. [Google Scholar] [CrossRef]
  264. Shi, W.; Wang, F. circ_AKT3 knockdown suppresses cisplatin resistance in gastric cancer. Open Med. 2022, 17, 280–291. [Google Scholar] [CrossRef] [PubMed]
  265. Zheng, K.; Xie, H.; Wu, W.; Wen, X.; Zeng, Z.; Shi, Y. CircRNA PIP5K1A promotes the progression of glioma through upregulation of the TCF12/PI3K/AKT pathway by sponging miR-515-5p. Cancer Cell Int. 2021, 21, 27. [Google Scholar] [CrossRef]
  266. Hu, J.; Wang, R.; Liu, Y.; Zhou, J.; Shen, K.; Dai, Y. Baicalein represses cervical cancer cell growth, cell cycle progression and promotes apoptosis via blocking AKT/mTOR pathway by the regulation of circHIAT1/miR-19a-3p axis. OncoTargets Ther. 2021, 14, 905–916. [Google Scholar] [CrossRef] [PubMed]
  267. Cheng, Z.; Liu, G.; Huang, C.; Zhao, X. Upregulation of circRNA_100395 sponges miR-142-3p to inhibit gastric cancer progression by targeting the PI3K/AKT axis. Oncol. Lett. 2021, 21, 419. [Google Scholar] [CrossRef] [PubMed]
  268. Jiang, W.; Zhang, C.; Zhang, X.; Sun, L.; Li, J.; Zuo, J. CircRNA HIPK3 promotes the progression of oral squamous cell carcinoma through upregulation of the NUPR1/PI3K/AKT pathway by sponging miR-637. Ann. Transl. Med. 2021, 9, 860. [Google Scholar] [CrossRef]
  269. Cai, Y.; Li, C.; Peng, F.; Yin, S.; Liang, H.; Su, J.; Li, L.; Yang, A.; Liu, H.; Yang, C.; et al. Downregulation of hsa_circRNA_0001400 helps to promote cell apoptosis through disruption of the circRNA_0001400-miR-326 sponge in cervical cancer cells. Front. Genet. 2021, 12, 779195. [Google Scholar] [CrossRef]
  270. Feng, Y.; Yan, B.; Cheng, H.; Wu, J.; Chen, Q.; Duan, Y.; Zhang, P.; Zheng, D.; Lin, G.; Zhuo, Y. Knockdown circ_0040414 inhibits inflammation, apoptosis and promotes the proliferation of cardiomyocytes via miR-186-5p/PTEN/AKT axis in chronic heart failure. Cell Biol. Int. 2021, 45, 2304–2315. [Google Scholar] [CrossRef]
  271. Ling, Z.; Fang, Z.G.; Wu, J.Y.; Liu, J.J. The depletion of Circ-PRKDC enhances autophagy and apoptosis in T-cell acute lymphoblastic leukemia via microRNA-653-5p/Reelin mediation of the PI3K/AKT/mTOR signaling pathway. Kaohsiung J. Med. Sci. 2021, 37, 392–401. [Google Scholar] [CrossRef]
  272. Gao, L.; Dou, Z.C.; Ren, W.H.; Li, S.M.; Liang, X.; Zhi, K.Q. CircCDR1as upregulates autophagy under hypoxia to promote tumor cell survival via AKT/ERK(1/2)/mTOR signaling pathways in oral squamous cell carcinomas. Cell Death Dis. 2019, 10, 745. [Google Scholar] [CrossRef]
  273. Yang, T.; Shen, P.; Chen, Q.; Wu, P.; Yuan, H.; Ge, W.; Meng, L.; Huang, X.; Fu, Y.; Zhang, Y.; et al. FUS-induced circRHOBTB3 facilitates cell proliferation via miR-600/NACC1 mediated autophagy response in pancreatic ductal adenocarcinoma. J. Exp. Clin. Cancer Res. 2021, 40, 261. [Google Scholar] [CrossRef] [PubMed]
  274. Yin, J.; Tong, F.; Ye, Y.; Hu, T.; Xu, L.; Zhang, L.; Zhu, J.; Pang, Z. Hsa_circRNA_103124 upregulation in Crohn’s disease promotes cell proliferation and inhibits autophagy by regulating the Hsa-miR-650/AKT2 signaling pathway. Front. Genet. 2021, 12, 753161. [Google Scholar] [CrossRef] [PubMed]
  275. Meng, L.; Liu, S.; Ding, P.; Chang, S.; Sang, M. Circular RNA ciRS-7 inhibits autophagy of ESCC cells by functioning as miR-1299 sponge to target EGFR signaling. J. Cell. Biochem. 2020, 121, 1039–1049. [Google Scholar] [CrossRef] [PubMed]
  276. Gao, W.; Guo, H.; Niu, M.; Zheng, X.; Zhang, Y.; Xue, X.; Bo, Y.; Guan, X.; Li, Z.; Guo, Y.; et al. circPARD3 drives malignant progression and chemoresistance of laryngeal squamous cell carcinoma by inhibiting autophagy through the PRKCI-Akt-mTOR pathway. Mol. Cancer 2020, 19, 166. [Google Scholar] [CrossRef] [PubMed]
  277. Du, W.W.; Yang, W.; Chen, Y.; Wu, Z.K.; Foster, F.S.; Yang, Z.; Li, X.; Yang, B.B. Foxo3 circular RNA promotes cardiac senescence by modulating multiple factors associated with stress and senescence responses. Eur. Heart J. 2017, 38, 1402–1412. [Google Scholar] [CrossRef] [Green Version]
  278. Zhang, Y.; Zhao, H.; Zhang, L. Identification of the tumorsuppressive function of circular RNA FOXO3 in nonsmall cell lung cancer through sponging miR155. Mol. Med. Rep. 2018, 17, 7692–7700. [Google Scholar] [CrossRef]
  279. Zhang, K.; Zhang, M.; Zhao, H.; Yan, B.; Zhang, D.; Liang, J. S100A4 regulates motility and invasiveness of human esophageal squamous cell carcinoma through modulating the AKT/Slug signal pathway. Dis. Esophagus 2012, 25, 731–739. [Google Scholar] [CrossRef]
  280. Yao, J.; Qian, K.; Chen, C.; Liu, X.; Yu, D.; Yan, X.; Liu, T.; Li, S. ZNF139/circZNF139 promotes cell proliferation, migration and invasion via activation of PI3K/AKT pathway in bladder cancer. Aging 2020, 12, 9915–9934. [Google Scholar] [CrossRef]
  281. Wang, X.; Ma, C.; Hou, X.; Zhang, G.; Huang, Y. Circular RNA circ_0002984 promotes cell proliferation and migration by regulating miR-181b-5p/vascular endothelial growth factor axis and PI3K-AKT signaling pathway in oxidized low-density lipoprotein-treated vascular smooth muscle cells. J. Cardiovasc. Pharmacol. 2022, 79, 501–511. [Google Scholar] [CrossRef]
  282. Chen, L.; Zhuo, D.; Yuan, H. Circ_100395 impedes malignancy and glycolysis in papillary thyroid cancer: Involvement of PI3K/AKT/mTOR signaling pathway. Immunol. Lett. 2022, 246, 10–17. [Google Scholar] [CrossRef]
  283. Ou, R.; Mo, L.; Tang, H.; Leng, S.; Zhu, H.; Zhao, L.; Ren, Y.; Xu, Y. circRNA-AKT1 sequesters miR-942-5p to upregulate AKT1 and promote cervical cancer progression. Mol. Ther. Nucleic Acids 2020, 20, 308–322. [Google Scholar] [CrossRef] [PubMed]
  284. Zhang, Z.; Zhao, M.; Wang, G. Hsa_circ_0051079 functions as an oncogene by regulating miR-26a-5p/TGF-β1 in osteosarcoma. Cell Biosci. 2019, 9, 1–11. [Google Scholar] [CrossRef] [PubMed]
  285. Song, Y.Z.; Li, J.F. Circular RNA hsa_circ_0001564 regulates osteosarcoma proliferation and apoptosis by acting miRNA sponge. Biochem. Biophys. Res. Commun. 2018, 495, 2369–2375. [Google Scholar] [CrossRef] [PubMed]
  286. Chen, Z.; Kang, K.; Chen, S.; Wang, S.; Zhang, J.; Zhang, X.; Chen, Z. Circular RNA circ_0017247 promotes melanoma migration and invasion via targeting miR-145. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 1932–1938. [Google Scholar]
  287. Li, C.; Wang, Y.; Chen, K. Circ_0017247 accelerates epithelial mesenchymal transition in non-small cell lung cancer. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 256–263. [Google Scholar]
  288. Luo, Y.; Liu, F.; Guo, J.; Gui, R. Upregulation of circ_0000199 in circulating exosomes is associated with survival outcome in OSCC. Sci. Rep. 2020, 10, 13739. [Google Scholar] [CrossRef] [PubMed]
  289. Hu, J.; Han, Q.; Gu, Y.; Ma, J.; McGrath, M.; Qiao, F.; Chen, B.; Song, C.; Ge, Z. Circular RNA PVT1 expression and its roles in acute lymphoblastic leukemia. Epigenomics 2018, 10, 723–732. [Google Scholar] [CrossRef] [PubMed]
  290. Ren, X.; Yu, J.; Guo, L.; Ma, H. Circular RNA circRHOT1 contributes to pathogenesis of non-small cell lung cancer by epigenetically enhancing C-MYC expression through recruiting KAT5. Aging 2021, 13, 20372–20382. [Google Scholar] [CrossRef]
  291. Yang, X.; Liu, L.; Zou, H.; Zheng, Y.W.; Wang, K.P. circZFR promotes cell proliferation and migration by regulating miR-511/AKT1 axis in hepatocellular carcinoma. Dig. Liver Dis. 2019, 51, 1446–1455. [Google Scholar] [CrossRef]
  292. Zhang, P.; Xue, X.F.; Ling, X.Y.; Yang, Q.; Yu, Y.; Xiao, J.; Wang, Z.N. CircRNA_010763 promotes growth and invasion of lung cancer through serving as a molecular sponge of miR-715 to induce c-Myc expression. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 7310–7319. [Google Scholar] [CrossRef]
  293. Huang, W.; Song, W.; Jiang, Y.; Chen, L.; Lu, H. c-Myc-induced circ-NOTCH1 promotes aggressive phenotypes of nasopharyngeal carcinoma cells by regulating the miR-34c-5p/c-Myc axis. Cell Biol. Int. 2021, 45, 1436–1447. [Google Scholar] [CrossRef] [PubMed]
  294. Zhao, X.; Zhong, Q.; Cheng, X.; Wang, S.; Wu, R.; Leng, X.; Shao, L. miR-449c-5p availability is antagonized by circ-NOTCH1 for MYC-induced NOTCH1 upregulation as well as tumor metastasis and stemness in gastric cancer. J. Cell. Biochem. 2020, 121, 4052–4063. [Google Scholar] [CrossRef] [PubMed]
  295. Xu, G.; Chen, Y.; Fu, M.; Zang, X.; Cang, M.; Niu, Y.; Zhang, W.; Zhang, Y.; Mao, Z.; Shao, M.; et al. Circular RNA CCDC66 promotes gastric cancer progression by regulating c-Myc and TGF-beta signaling pathways. J. Cancer 2020, 11, 2759–2768. [Google Scholar] [CrossRef]
  296. Sun, J.; Zhang, H.; Tao, D.; Xie, F.; Liu, F.; Gu, C.; Wang, M.; Wang, L.; Jiang, G.; Wang, Z.; et al. CircCDYL inhibits the expression of C-MYC to suppress cell growth and migration in bladder cancer. Artif. Cells Nanomed. Biotechnol. 2019, 47, 1349–1356. [Google Scholar] [CrossRef] [PubMed]
  297. Huang, C.; Shu, L.; Zhang, H.; Zhu, X.; Huang, G.; Xu, J. Circ_ZNF512-mediated miR-181d-5p inhibition limits cardiomyocyte autophagy and promotes myocardial ischemia/reperfusion injury through an EGR1/mTORC1/TFEB-based mechanism. J. Med. Chem. 2022, 65, 1808–1821. [Google Scholar] [CrossRef]
  298. Yang, Z.; Huang, C.; Wen, X.; Liu, W.; Huang, X.; Li, Y.; Zang, J.; Weng, Z.; Lu, D.; Tsang, C.K.; et al. Circular RNA circ-FoxO3 attenuates blood-brain barrier damage by inducing autophagy during ischemia/reperfusion. Mol. Ther. 2022, 30, 1275–1287. [Google Scholar] [CrossRef]
  299. Li, Y.; Qin, G.; Du, J.; Yue, P.; Zhang, Y.; Hou, N. circRNA LDLRAD3 enhances the malignant behaviors of NSCLC Cells via the miR-20a-5p-SLC7A5 axis activating the mTORC1 signaling pathway. J. Healthc. Eng. 2022, 2022, 2373580. [Google Scholar] [CrossRef]
  300. Wei, H.; Cao, C.; Wei, X.; Meng, M.; Wu, B.; Meng, L.; Wei, X.; Gu, S.; Li, H. Circular RNA circVEGFC accelerates high glucose-induced vascular endothelial cells apoptosis through miR-338-3p/HIF-1alpha/VEGFA axis. Aging 2020, 12, 14365–14375. [Google Scholar] [CrossRef]
  301. Dang, R.Y.; Liu, F.L.; Li, Y. Circular RNA hsa_circ_0010729 regulates vascular endothelial cell proliferation and apoptosis by targeting the miR-186/HIF-1alpha axis. Biochem. Biophys. Res. Commun. 2017, 490, 104–110. [Google Scholar] [CrossRef]
  302. Zhou, P.; Xie, W.; Huang, H.L.; Huang, R.Q.; Tian, C.; Zhu, H.B.; Dai, Y.H.; Li, Z.Y. circRNA_100859 functions as an oncogene in colon cancer by sponging the miR-217-HIF-1alpha pathway. Aging 2020, 12, 13338–13353. [Google Scholar] [CrossRef]
  303. Ma, X.; Wang, C.; Chen, J.; Wei, D.; Yu, F.; Sun, J. circAGFG1 sponges miR-28-5p to promote non-small-cell lung cancer progression through modulating HIF-1alpha level. Open Med. 2021, 16, 703–717. [Google Scholar] [CrossRef] [PubMed]
  304. Yu, L.; Li, J.; Peng, B.; Cai, P.; Zhao, B.; Chen, Y.; Zhu, H. CircASXL1 knockdown restrains hypoxia-induced DDP resistance and NSCLC progression by sponging miR-206. Cancer Manag. Res. 2021, 13, 5077–5089. [Google Scholar] [CrossRef] [PubMed]
  305. Zhai, Z.; Fu, Q.; Liu, C.; Zhang, X.; Jia, P.; Xia, P.; Liu, P.; Liao, S.; Qin, T.; Zhang, H. Emerging roles of hsa-circ-0046600 targeting the miR-640/HIF-1alpha signalling pathway in the progression of HCC. OncoTargets Ther. 2019, 12, 9291–9302. [Google Scholar] [CrossRef]
  306. Wang, J.; Huang, K.; Shi, L.; Zhang, Q.; Zhang, S. CircPVT1 promoted the progression of breast cancer by regulating MiR-29a-3p-Mediated AGR2-HIF-1alpha pathway. Cancer Manag. Res. 2020, 12, 11477–11490. [Google Scholar] [CrossRef] [PubMed]
  307. Tan, Y.; Du, B.; Zhan, Y.; Wang, K.; Wang, X.; Chen, B.; Wei, X.; Xiao, J. Antitumor effects of circ-EPHB4 in hepatocellular carcinoma via inhibition of HIF-1alpha. Mol. Carcinog. 2019, 58, 875–886. [Google Scholar] [CrossRef]
  308. Lin, X.; Sun, R.; Zhao, X.; Zhu, D.; Zhao, X.; Gu, Q.; Dong, X.; Zhang, D.; Zhang, Y.; Li, Y.; et al. C-myc overexpression drives melanoma metastasis by promoting vasculogenic mimicry via c-myc/snail/Bax signaling. J. Mol. Med. 2017, 95, 53–67. [Google Scholar] [CrossRef]
  309. Bai, Y.; Rao, H.; Chen, W.; Luo, X.; Tong, C.; Qi, H. Profiles of circular RNAs in human placenta and their potential roles related to preeclampsia. Biol. Reprod. 2018, 98, 705–712. [Google Scholar] [CrossRef] [Green Version]
  310. Pu, Z.; Lu, J.; Yang, X. Emerging roles of circular RNAs in vascular smooth muscle cell dysfunction. Front. Genet. 2021, 12, 749296. [Google Scholar] [CrossRef]
  311. Yang, H.; Wang, H.; Shang, H.; Chen, X.; Yang, S.; Qu, Y.; Ding, J.; Li, X. Circular RNA circ_0000950 promotes neuron apoptosis, suppresses neurite outgrowth and elevates inflammatory cytokines levels via directly sponging miR-103 in Alzheimer’s disease. Cell Cycle 2019, 18, 2197–2214. [Google Scholar] [CrossRef]
  312. Zhang, M.; Bian, Z. The emerging role of circular RNAs in Alzheimer’s disease and Parkinson’s disease. Front. Aging Neurosci. 2021, 13, 691512. [Google Scholar] [CrossRef]
  313. Chen, T.; Wang, X.; Li, C.; Zhang, H.; Liu, Y.; Han, D.; Li, Y.; Li, Z.; Luo, D.; Zhang, N.; et al. CircHIF1A regulated by FUS accelerates triple-negative breast cancer progression by modulating NFIB expression and translocation. Oncogene 2021, 40, 2756–2771. [Google Scholar] [CrossRef] [PubMed]
  314. Wang, X.B.; Li, P.B.; Guo, S.F.; Yang, Q.S.; Chen, Z.X.; Wang, D.; Shi, S.B. circRNA_0006393 promotes osteogenesis in glucocorticoidinduced osteoporosis by sponging miR1455p and upregulating FOXO1. Mol. Med. Rep. 2019, 20, 2851–2858. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Overview of AKT, AKT effectors, lncRNAs, and circRNAs regulating diverse cancer cell functions.
Figure 1. Overview of AKT, AKT effectors, lncRNAs, and circRNAs regulating diverse cancer cell functions.
Cells 11 02940 g001
Figure 2. Strategy for connecting database-predicted AKT-, AKT effector-targeting lncRNAs and circRNAs to their regulating cell functions. By searching LncTarD [38] and circBase [61], these AKT- and AKT effector-targeting lncRNA and circRNA candidates were retrieved by individual input of gene names for AKT1, AKT2, and AKT3, as well as AKT effectors. Subsequently, they were applied to a literature survey by Google Scholar and PubMed to check their potential cell functions.
Figure 2. Strategy for connecting database-predicted AKT-, AKT effector-targeting lncRNAs and circRNAs to their regulating cell functions. By searching LncTarD [38] and circBase [61], these AKT- and AKT effector-targeting lncRNA and circRNA candidates were retrieved by individual input of gene names for AKT1, AKT2, and AKT3, as well as AKT effectors. Subsequently, they were applied to a literature survey by Google Scholar and PubMed to check their potential cell functions.
Cells 11 02940 g002
Figure 3. Schematic overview. AKT, its effectors, and all database lncRNAs mentioned were shown at the points of the AKT pathway that they were involved in, as shown in Table 1 and Table 4. No FOXO-targeting lncRNA was available by LncTarD searching (13 June 2022).
Figure 3. Schematic overview. AKT, its effectors, and all database lncRNAs mentioned were shown at the points of the AKT pathway that they were involved in, as shown in Table 1 and Table 4. No FOXO-targeting lncRNA was available by LncTarD searching (13 June 2022).
Cells 11 02940 g003
Table 1. AKT1-, AKT2-, AKT3-targeting database lncRNAs.
Table 1. AKT1-, AKT2-, AKT3-targeting database lncRNAs.
AKT1AKT2AKT3
UpregulateENST00113
MALAT1
CDKN2B-AS1
HULC
LUCAT1
AFAP1-AS1
LINC00462
LOXL1-AS1
AB073614
H19
SPRY4-IT1
lncRNA-p3134FEZF1-AS1
DownregulateGAS5
RP11-708H21.4
FOXD2-AS1
LINC00312
--
lncRNAs targeting several AKT effectors were retrieved from the LncTarD database [38] (accessed on 13 June 2022).
Table 2. Connecting AKT1-, AKT2-, and AKT3-targeting database lncRNAs to cell functions.
Table 2. Connecting AKT1-, AKT2-, and AKT3-targeting database lncRNAs to cell functions.
Cell Functions
AKT1/2/3-Targeting lncRNAsApoptosisAutophagyER StressMitochondrial
Morphogenesis
FerroptosisNecroptosisDNA
Damage
Response
SenescenceMigration
AKT1ENST00113[62][63][62]
MALAT1[64][65][66][64][67][68]
GAS5[69,70][71,72][73][74][75][76][77][78]
CDKN2B-AS1[79][80][81][82][79,82][83]
HULC[84,85][86][87][88][89,90]
LUCAT1[91][91][92][93][94][91]
RP11-708H21.4[95][95]
AFAP1-AS1[96][96]
LINC00462[97][98]
LOXL1-AS1[99][99]
FOXD2-AS1[100][100]
AB073614[101][102]
H19[103][104][105][106][107][105][108][109][104]
SPRY4-IT1[110][111][112]
LINC00312[113][53][114]
AKT2lncRNA-p3134[115]
AKT3FEZF1-AS1[116][117]
Literature could not be found on Google Scholar or PubMed (12 June 2022). The lncRNAs targeting several AKT effectors were retrieved from the LncTarD database [38] (accessed on 13 June 2022).
Table 3. Connecting AKT effectors to lncRNA-regulated cell functions.
Table 3. Connecting AKT effectors to lncRNA-regulated cell functions.
Cell Functions
ApoptosisAutophagyER StressMitochondrial
Morphogenesis
FerroptosisNecroptosisDNA
Damage
Response
SenescenceMigration
FOXO
c-Myc
mTORC1
mTORC2
SREBP1
HIF
c-Myc
mTORC1
SREBP1
HIF
c-MycFOXO
c-Myc
HIF
c-Mycc-Myc
HIF
c-My
cmTORC1
FOXO
c-Myc
mTORC1
Different AKT effectors may regulate various cell functions. Literature could not be found by searching Google Scholar and PubMed (12 June 2022).
Table 4. AKT effector-targeting database lncRNAs.
Table 4. AKT effector-targeting database lncRNAs.
AKT Effectors
LncRNAsc-MycmTORS6K1SREBP1HIF
UpregulationPVT1
HOTTIP
CRNDE
CCAT2
HNF1A-AS1 SNHG1
NEAT1H19
CERNA2
TUG1
PCAT1 LINC-ROR
FILNC1
THORLNC
MALAT1
ENST00113
HOTAIR
PVT1
H19
CRNDE
HULC
lncRNA-p3134
UCA1
HOTAIR
PCGEM1
LNCARSRHOTAIR
RAB4B-EGLN2
MEG3
RPL13AP23
DownregulationHULC
PCAT1
lncRNA-BCAT1 PCAT6
UCA1
RP11-708H21.4
GAS5
HOTAIR
RP11-708H21.4 CPS1-IT1
MIR31HG
MALAT1
FOXO, mTORC1 (RPTOR, MLST8, AKT1S1, and DEPTOR), S6K2, and 4EBP1 targeted by lncRNAs were omitted because they were not available after the retrieval of the LncTarD database (http://bio-bigdata.hrbmu.edu.cn/LncTarD/ or https://lnctard.bio-database.com/) [38] (accessed on 13 June 2022).
Table 5. Connecting AKT effectors and database lncRNAs to cell functions.
Table 5. Connecting AKT effectors and database lncRNAs to cell functions.
Cell Functions
AKT EffectorslncRNAsApoptosisAutophagyER StressMitochondrial
Morphogenesis
FerroptosisNecroptosisDNA
Damage
Response
SenescenceMigration
c-MycPVT1[171,172][173][174][175][133][176][177]
HOTTIP[178][179][180][181][179,182]
CRNDE[183][184][185][186][185]
HULC[84,85][86][87][88][89,90]
CCAT2[187][188][188]
HNF1A-AS1[189][190][191]
PCAT1[192][131][193][194]
SNHG1[195][196][197][195]
lncRNA-BCAT1[198]
NEAT1[199,200][199][200][201][202][203][199,200]
H19[103][104][105][106][107][105][108][109][104]
CERNA2[204][205]
PCAT6[206][207][208].[209][209][210]
TUG1[211][212][213][214][215][216][217]
LINC-ROR[218][218][219][220]
FILNC1[118]
THORLNC
mTORC1/2MALAT1[64] [65][66][64][67][68]
(mTOR)ENST00113[62] [63][62]
HOTAIR[221][222,223][224][225][225][223]
PVT1[171,172][173][174][175][133][176][177]
UCA1[226][227][228][229][230][231][232]
RP11-708H21.4[95] [95]
GAS5[69,70][71,72][73][74][75][76][77][78]
H19[103][104][105][106][107][105][108][109][104]
lncRNA-p3134[115]
S6K1/2HOTAIR[221][222,223][224][225][225][223]
RP11-708H21.4[95][95]
PCGEM1[233][234][235]
SREBP1LNCARSR[236][237]
HIFHOTAIR[221][222,223][224][225][225][223]
CPS1-IT1[238][239][240]
MIR31HG[241][242][243]
MEG3[244][245][244][246][247][248][249][250][251]
RPL13AP23
FOXO, mTORC1 complex (including RPTOR, MLST8, AKT1S1, and DEPTOR), S6K2, and 4EBP1 targeted by lncRNAs were omitted because they were not available after the retrieval of the LncTarD database [38] (13 June 2022). mTOR is one of the components of mTORC1. ○ Literature could not be found by searching Google Scholar and PubMed (12 June 2022).
Table 6. AKT1/AKT2/AKT3-targeting database circRNAs.
Table 6. AKT1/AKT2/AKT3-targeting database circRNAs.
AKT1-, AKT2-, AKT3-Targeting CircRNAs
AKT1circ_0101403circ_0101404circ_0033555circ_0033560circ_0033559circ_0033546
circ_0033552circ_0033558circ_0033557circ_0033553circ_0033547circ_0033551
circ_0033548circ_0033556circ_0033549circ_0033554circ_0033550
AKT2circ_0051068circ_0051082circ_0051074circ_0051077circ_0051071circ_0051081
circ_0051073circ_0051080circ_0051070circ_0051075circ_0051069circ_0051072
circ_0051078circ_0051079circ_0051076circ_0008719circ_0005812
AKT3circ_0017242circ_0112774circ_0017251circ_0112785circ_0017249circ_0112773
circ_0006696circ_0017252circ_0017243circ_0112797circ_0112778circ_0112800
circ_0004649circ_0112777circ_0017254circ_0017246circ_0112788circ_0112798
circ_0017250circ_0000199circ_0112770circ_0112782circ_0112787circ_0017247
circ_0017244circ_0112767circ_0112775circ_0017253circ_0112799circ_0112776
circ_0002240circ_0112801circ_0112802circ_0112772circ_0112780circ_0112771
circ_0112791circ_0112766circ_0112783circ_0017245circ_0112790circ_0112786
circ_0112792circ_0112768circ_0112784circ_0017248circ_0112779circ_0112796
circ_0112769circ_0112789circ_0112781circ_0112794circ_0112793circ_0112795
Data were available in the circBase database (http://www.circbase.org/) (accessed on 13 June 2022). CircRNAs in bold font were reported in the literature, as described below.
Table 7. AKT effector-targeting database circRNAs.
Table 7. AKT effector-targeting database circRNAs.
AKT EffectorsAKT Effector-Targeting CircRNAs
FOXO
c-Myccirc_0085535circ_0085533circ_0085534
mTORcirc_0110437circ_0009803circ_0009829circ_0110441circ_0009793circ_0009810circ_0110442circ_0009787
circ_0009779circ_0009834circ_0009801circ_0009795circ_0009813circ_0009809circ_0110447circ_0110418
circ_0009837circ_0009820circ_0009822circ_0009831circ_0009844circ_0009825circ_0009776circ_0009835
circ_0009845circ_0009847circ_0009782circ_0009842circ_0009788circ_0009819circ_0110416circ_0110435
circ_0009823circ_0009832circ_0009839circ_0009785circ_0009830circ_0009840circ_0009808circ_0110438
circ_0009815circ_0009804circ_0009799circ_0009800circ_0009811circ_0009784circ_0009778circ_0009821
circ_0009786circ_0110414circ_0110424circ_0009789circ_0009828circ_0009838circ_0009833circ_0110440
circ_0009780circ_0009777circ_0009802circ_0009806circ_0009796circ_0009798circ_0009794circ_0009791
circ_0110417circ_0006576circ_0009826circ_0110420circ_0110439circ_0009797circ_0009805circ_0009790
circ_0009846circ_0009807circ_0009792circ_0009814circ_0009824circ_0009843circ_0009818circ_0009841
circ_0110415circ_0009817circ_0009783circ_0110443circ_0009812circ_0009781circ_0110419circ_0009827
circ_0009816circ_0009836
RPTOR
MLST8circ_0105204circ_0037498
AKT1S1circ_0051983circ_0000950circ_0051984
DEPTORcirc_0135616circ_0085412circ_0135615circ_0135617
S6K1circ_0008625circ_0044904circ_0044907circ_0044900circ_0044903circ_0107292circ_0107290circ_0044902
circ_0044905circ_0044899circ_0044906circ_0107291circ_0044901
S6K2circ_0023096circ_0023090circ_0023094circ_0023095circ_0023091circ_0023092circ_0023093circ_0023089
SREBP1
HIFcirc_0102309circ_0102321circ_0102322circ_0102317circ_0004817circ_0102315circ_0004623circ_0102323
circ_0102313circ_0006326circ_0005205circ_0032132circ_0032139circ_0102310circ_0102327circ_0032136
circ_0102326circ_0102311circ_0102314circ_0032135circ_0102318circ_0032137circ_0032140circ_0032138
circ_0007976circ_0032133circ_0102325circ_0032134circ_0102312circ_0102320circ_0102316circ_0006393
circ_0102319circ_0102324
mTORC1 consists of the mTOR, RPTOR, MLST8, AKT1S1, and DEPTOR. Bold circRNAs were reported in the literature, as described later. indicates not available in the circBase database (accessed on 13 June 2022). CircRNAs in bold were reported in the literature, as described later.
Table 8. Connecting AKT effectors and database circRNAs to cell functions.
Table 8. Connecting AKT effectors and database circRNAs to cell functions.
Cell Functions
AKT EffectorscircRNAsApoptosisMigration
c-Myccirc_0085533 [308]downregulate
mTORC1/2 (mTOR)circ_0009805 [309], circ_0009792 [310]
mTORC1/2 (AKT1S1)circ_0000950 [311,312]upregulate
HIFcirc_0032138 [313], circ_0006393 [314]upregulate
mTOR and AKT1S1 are two of the components for mTORC1. Only circRNAs for c-Myc, mTOR, AKT1S1, and HIF were available in the circBase database (accessed on 13 June 2022). indicates no available after a Google Scholar and PubMed search (accessed on 13 June 2022).
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Tang, J.-Y.; Chuang, Y.-T.; Shiau, J.-P.; Yang, K.-H.; Chang, F.-R.; Hou, M.-F.; Farooqi, A.A.; Chang, H.-W. Long Noncoding RNAs and Circular RNAs Regulate AKT and Its Effectors to Control Cell Functions of Cancer Cells. Cells 2022, 11, 2940. https://doi.org/10.3390/cells11192940

AMA Style

Tang J-Y, Chuang Y-T, Shiau J-P, Yang K-H, Chang F-R, Hou M-F, Farooqi AA, Chang H-W. Long Noncoding RNAs and Circular RNAs Regulate AKT and Its Effectors to Control Cell Functions of Cancer Cells. Cells. 2022; 11(19):2940. https://doi.org/10.3390/cells11192940

Chicago/Turabian Style

Tang, Jen-Yang, Ya-Ting Chuang, Jun-Ping Shiau, Kun-Han Yang, Fang-Rong Chang, Ming-Feng Hou, Ammad Ahmad Farooqi, and Hsueh-Wei Chang. 2022. "Long Noncoding RNAs and Circular RNAs Regulate AKT and Its Effectors to Control Cell Functions of Cancer Cells" Cells 11, no. 19: 2940. https://doi.org/10.3390/cells11192940

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop