Abstract
There is accruing data implicating long non-coding RNAs (lncRNAs) in the development and progression of non-communicable diseases such as cancer. These lncRNAs have been implicated in many diverse HIV-host interactions, some of which are beneficial to HIV propagation. The virus-host interactions induce the expression of HIV-regulated long non-coding RNAs, which are implicated in the carcinogenesis process, therefore, it is critical to understand the molecular mechanisms that underpin these HIV-regulated lncRNAs, especially in cancer formation. Herein, we summarize the role of HIV-regulated lncRNAs targeting cancer development-related processes including apoptosis, cell cycle, cell survival signalling, angiogenesis and drug resistance. It is unclear how lncRNAs regulate cancer development, this review also discuss recent discoveries regarding the functions of lncRNAs in cancer biology. Innovative research in this field will be beneficial for the future development of therapeutic strategies targeting long non-coding RNAs that are regulated by HIV, especially in HIV associated cancers.
Keywords: Carcinogenesis, HIV-1 • long non-coding RNA, Oncogenic virus, Cellular process, Drug resistance
1. Introduction
Human immunodeficiency virus-1 (HIV-1) infection is a crucial etiological factor for various non-communicable diseases, which include HIV-associated nephropathies [1,2], cardiomyopathies [3] and cancer [4]. HIV-1 contributes to cancer development directly through oncogenic effects of HIV proteins, such as Trans-activator of transcription (Tat) and indirectly through immunosuppression and promiting coinfections by other oncogenic viruses [[5], [6], [7], [8]]. HIV was discovered in 1983 as the aetiological cause of acquired immunodeficiency syndrome (AIDS), which is a global pandemic that continues to devastate many communities, worldwide [9]. As of 2017, there were 36.9 million people living with HIV, globally [10] with the majority of these infections occurring in the resource-limited countries, especially those in the sub-Saharan Africa [11]. HIV is classified under retroviruses, a subgroup of lentiviruses [12]. This subgroup of viruses targets white blood cells expressing the cluster of differentiation 4 (CD4+) [12]. White blood cells targeted by this virus include T cells, macrophages and dendritic cells [12]. HIV attaches and invades these cells using its glycoproteins, gp120 and gp41, which promote viral entry by fusing with the host co-receptors; namely the C–C chemokine receptor type 5 (CCR5) and C-X-C chemokine receptor type 4 (CXCR4) [13]. Accumulation of this infectious virus decreases CD4+ T cells, resulting in a weakened immune system, which becomes unable to combat the effects of opportunistic pathogens, thus, ultimately leading to the development of full-blown AIDS [12].
The HIV is composed of two forms; type 1 and type 2, with both virus types targeting the same cells, exhibiting similar morphology, tropism, and modes of transmission; however, they differ genetically and antigenically, with HIV type 2 being less pathogenic than HIV type 1 [14]. HIV-1 virus is responsible for the ongoing HIV/AIDS pandemic as it accounts for over 95% of HIV infections, worldwide [15]. HIV-1 virus is categorised into four groups; namely, O (Outlier), M (Major), N (non-M, non-O), and lastly, P group, which was the last group to be discovered [16]. Group M viruses have dominated the global HIV pandemic since its outbreak in 1981 [16]; and the other 3 groups are relatively rare and occur only in certain geographical areas, including Cameroon and Europe [[17], [18], [19]]. HIV-2 virus is reported to be less infectious than HIV-1 and remains generally limited to the Western parts of Africa, but other cases have been reported in Europe [20], the United States of America, as well as in India [21]. HIV type 2 is comprised of nine groups (A - I), HIV-2 subtype data is inadequate and only few recombinants have been studied [22]. Although antiretroviral therapy has prolonged the survival of HIV-affected people, an increasing number of HIV-infected people are at a risk of developing cancer, especially Kaposi's sarcoma, invasive cervical cancer and B-cell lymphomas [23].
Cancer, an umbrella term for malignancies with deregulated cell proliferation, resistance to apoptosis, and enhanced survival signals, has been reported as the second cause of death after cardiovascular diseases [24]. There is a steady increase in cancer-related mortalities and soaring incidence rates despite the current therapeutic advances, and thus, cancer continues to be a global menace [25]. There are over 200 different types of cancers, with unique characteristics and challenges for their treatment [24]. Dalton et al. [26] indicated that 70% of deaths from cancer in 2018 occurred in developing countries, including, but not limited to, South Africa, Zimbabwe and Nigeria. These high incidence rates can be attributed to gaps in current diagnostic and therapeutic strategies, which include chemotherapy, radiation and surgery. The new challenge is the rise of HIV-related cancers, worldwide, especially in Sub-Saharan Africa (SSA). Oncoviral infections are credited as aetiological factors for the development of 12% of human cancers, worldwide, with 80% of cases occurring in developing countries, including SSA [27,28]. Infectious agents such as HIV-1 in certain SSA countries drive the rates of these cancers [29,30]. These oncoviruses can integrate their genomes directly into the host genome or first undergo reverse transcription to DNA (RNA virus) before integration into the host genome [31]. Oncoviruses have developed several strategies to drive virus-induced oncogenesis, and these strategies include inactivation of host tumour suppressors by viral proteins [32], and alteration of host gene expression due to the integration of viral genome into the host genome [33]. Some of these strategies function through a group of RNAs known as Long non-coding RNAs (lncRNAs), which are aberrantly expressed in cancer [34]. These non-coding RNAs (ncRNAs) also promote viral viability by resisting the cellular antiviral response in order to maintain viral latency, promoting viral replication and genome packaging [35]. ncRNAs promote HIV replication because the advanced stage of the HIV infection contributes to immune suppression which is associated with increased cancer risk such as coinfections by other oncogenic viruses which directly cause cancer. Therefore, it is accepted that the progression of HIV to advanced stages is a key factor for HIV-mediated carcinogenesis [5,6]. The effects of some ncRNAs in promoting HIV progression may differ at different stages of the virus lifecycle as summarised in Table 1 [36]. These ncRNAs include, NEAT1, which is identified as a transcriptional regulator that modulates HIV-1 expression by storing excess unspliced instability (INS) transcripts within paraspeckle bodies in the nucleus [46]. Overexpression of NEAT1 leads to increased storage of unspliced HIV-1 RNA transcripts, which serve as a counterbalance for HIV-1 transcription in the cell [46]. Other examples of ncRNAs that showed differential expression at different phases of the virus life cycle include MALAT1, which resulted in an increased HIV-1 transcription by modifying the epigenetic status of HIV-1 genome promoter. In addition to MALAT1, one also finds Epstein-Barr virus-encoded small RNAs (EBERs) [37] and uc002yug.2 long non coding RNA [38] that assist in maintaining viral latency and facilitating HIV-1 replication in host cells. For instance, in surgical lung biopsies of HIV subtype E-infected pediatric patients, Epstein-Barr virus-encoded small RNAs (EBERs) are coexpressed with p24, a HIV core protein that facilitates HIV adsorption, membrane fusion, and entry into the host cells [37]. Additionally, Huan et al. [38] found that uc002yug.2 long non coding RNA has the potential to increase HIV replication, HIV long terminal repeat activity, and the activation of latent HIV in oesophageal squamous cell carcinoma and CD4+ T cells derived from HIV-infected patients. It is also worth noting that HIV alters the transcription of various lncRNAs to regulate the expression of viral genes and inhibits host antiviral defenses [39]. Therefore, the altered transcription of these lncRNAs can result in tumour formation [40].
Table 1.
Long noncoding RNAs deregulated in AIDS-defining cancers.
| Long noncoding RNAs | Classification based on genomic location | Classification based on mode of action | aDifferential expression | Cancer type | Neighboring coding genes | Subcellular localization | Mechanism/s | Reference |
|---|---|---|---|---|---|---|---|---|
| NRON | Antisense lncRNA | Scaffold | Down | T cell leukemia | MVB12B | Cytoplasm | Modulate HIV replication by blocking NFAT transcriptional regulator which regulate HIV gene expression | [45] |
| NEAT1 | LincRNA | Guide | Up | T cell leukemia | FRMD8 and MIR612 | Nucleus | Maintain the integrity of the nuclear paraspeckle to promote HIV replication; enhances cytokine production | [46] |
| ANRIL | Antisense lncRNA | Scaffold | Down | Kaposi sarcoma | CDKN2A and CDKN2B | Nucleus | Epigenetic silencing of INK4B tumor suppresser | [47,48] |
| MEG3 | LincRNA | Signal, Scaffold, Guide and Decoy | Up | Kaposi sarcoma and Liver cancer |
DLK1, RTL1, and DIO3 | Cytoplasm | Increases the transcription of p53 | [[49], [50], [51]] |
| HOTAIR | Antisense lncRNA | Scaffold | Up | Breast cancer, Lung cancer, Prostate cancer and Ovarian cancer |
HOXD | Cytoplasm | Gene silencing by demethylation of H3K4 me3 | [44,51,52] |
| MALAT1 | LincRNA | Signal | Up | Cervical carcinoma | LTBP3 | Nucleus | Increase HIV-1 transcription; promote cell migration and proliferation. | [53,54] |
| H19 | LincRNA | Decoy | Up | Cervical carcinoma | IGF2 | Cytoplasm | Promote cell proliferation | [55,56] |
| GAS5 | LincRNA | Decoy | Down | Breast cancer, Gastric cancer, Lung cancer and Prostate cancer |
E2F4 FGF1 |
Nucleus | Inhibits HIV-1 replication; regulate cell proliferation, apoptosis and invasion |
[57,58] |
| uc002yug.2 | LincRNA | Scaffold and Guide | Up | Oesophageal squamous cell carcinoma | RUNX1 | Nucleus | Support carcinogenesis by promoting alternative splicing of RUNX1; Increase HIV-1 replication |
[59] |
| Fas-AS1 | antisense lncRNA |
Decoy | Up | Acute T-cell leukemia, Uterine carcinoma and Histocytic Lymphoma |
Fas | Nucleus | Inhibits Fas-mediated programmed cell death | [60] |
| NFAT | LincRNA | Signal | Up | B cell lymphoma, Glioblastoma, Pancreatic cancer, Colon cancer and Ovarian cancer |
Interleukin (IL) 2 and Cyclin E |
Cytoplasm | Promote HIV-1 infection | [[61], [62], [63]] |
| LincRNA-p21 | LincRNA | Decoy | Down | Hepatocellular carcinoma, Prostate cancer and Breast cancer | p21 | Cytoplasm | Regulate cell cycle and proliferation | [64,65] |
| lncARSR | LincRNA | Decoy | Up | Renal cancer, Hepatocellular carcinoma and Ovarian cancer | SOX4 | Cytoplasm | Promote cell proliferation, migration and invasion | [66] |
| DLEU1 | Antisense lncRNA | Decoy | UP | Cervical cancer, Colorectal cancer, Gastric cancer and Ovarian cancer |
KPNA3 | Nucleus | Inhibit apoptosis and promote cell proliferation, migration and invasion | [67,68] |
The differential expression are compared to healthy individuals.
In the last few years, researchers have devoted their time and resources in an effort to understand the role played by long non-coding RNAs during cancer development and progression [41]. Therefore, this review examines and analyses HIV-associated lncRNAs that are deregulated in cancer and how they mediate drug resistance, the effect of these lncRNAs in cancer-related processes, which include apoptosis, cell cycle and angiogenesis will also be explored. There have been no reports implicating the role of lncRNAs in carcinogenesis in HIV-2 infected individuals, but there is emerging evidence that implicates lncRNAs in HIV-1-mediated carcinogenesis where they play important roles in regulating gene expression to favour cancer development. Since HIV-1 is more virulent, infects more people than HIV-2 and has direct pro-oncogenic effects, therefore the focus of this review is centered on the impact of lncRNAs on HIV-mediated carcinogenesis, focussing on HIV-1. As shown in Fig. 1, HIV-1 alters the transcription of lncRNAs to promote tumour formation. The figure shows that since lncRNAs control transcriptional processes, they provide an opportunity for HIV to hijack cellular machinery and manipulate gene expression to promote cancer through modulation of lncRNAs.
Fig. 1.
HIV infection deregulate the expression of long noncoding RNAs resulting in the development of cancer. Adapted from Butova et al. [218].
2. Long non coding RNAs
LncRNAs are 200 nucleotides long, non-protein-coding RNA transcripts that may either possess or lack a poly(A) tail, and have no potential of encoding a polypeptide chain. According to the presence or absence of a poly(A) tail at their 3′ ends, they may also possess both exons and introns; however, they lack the potential to code for protein products [41,42]. Based on genomic location, lncRNAs are classified into five types; namely, (a) antisense lncRNAs generated from the antisense strand of protein-coding genes, (b) intergenic lncRNAs transcribed from intergenic regions of both strands, (c) intronic long noncoding RNAs synthesized from introns of protein-coding genes, (d) sense lncRNAs synthesized from the sense strand of protein-coding genes, and (e) lastly, bidirectional lncRNAs synthesized from the opposite strand of the protein-coding genes within 1000 bp of the promoter of the protein coding genes [43,44]. Long noncoding RNAs are also classified based on biogenesis and mode of action as summarised in Table 1. Based on genomic location, nuclear paraspeckle assembly transcript 1 (NEAT1) lncRNA is the most studied, and is classified as Long intergenic non-coding RNA (lincRNA) [46]. HIV promotes NEAT1 expression [46], which functions as an oncogene towards the development and progression of different malignancies, such as colorectal cancer [69], hepatocellular carcinoma [63], lung cancer [70], oesophageal squamous cell carcinoma [71] and glioma [72]. LncRNAs also classified based on their mode of action, thus, there are (i) decoy lncRNAs that titrates away DNA-binding proteins, such as transcription factors, for example, Growth Arrest Specific 5 (GAS5), which is able to bind to the DNA-binding domain of glucocorticoid receptor (GR) and titrates down the amount of GR available to bind to genomic glucocorticoid response elements [73,74] Secondly, one finds (ii) scaffolding lncRNAs that enable two or more proteins to interact, and this is exemplified by non-coding repressor of nuclear factor of activated T cells (NRON), which interacts with human nuclear factor of activated T cells (NFAT) and inhibit its transcriptional activity, NRON also brings the phosphorylated NFAT into a cytoplasmic RNA-protein complex through scaffolding [45]. Moreover, there are (iii) guide lncRNAs, which directs regulatory factors such as chromatin modification enzymes, and this can occur through RNA-DNA interactions or through RNA in synergy with a DNA-binding protein, e.g NEAT1 which recruits epigenetic regulators to their target genes through its role as a guide [45]. Lastly, there are (iv) signalling lncRNAs, which regulate gene expression in response to diverse stimuli, and these are cell type-specific. These are exemplified by Maternally Expressed 3 (MEG3), which binds to cognate genomic regions through RNA-DNA hybrids and modulates the expression of target genes [43,75].
2.1. Cellular roles of lncRNAs
Studies showed that lncRNAs are involved inseveral major biological processes including dosage compensation (e.g Xist), nuclear organization (e.g MALAT1), alternative splicing of pre-mRNA (e.g DGCR5) and regulation of protein expression (e.g GAS5) [74]. A number of proteins are regulated by lncRNAs, including transcription factors which are crucial players in transcriptional regulation. LncRNA GAS5 is known to interact with active glucocorticoid receptors (GRs) through the DNA-binding domain of a transcription factor in order to inhibit their binding to glucocorticoid response element (GRE); consequently, blocking glucocorticoid-GR-GRE interaction induced targeted gene transcription [73,76]. This effect suggests that lncRNAs can act as a binding competitor for DNA Binding Proteins (DBPs) to regulate gene expression [73,76].
Additionally, some lncRNAs are implicated in regulating the expression of many protein-coding genes. For instance, some lncRNAs can directly modulate the expression of genes by binding to chromatin, modifying chromatin complexes or by epigenetically modulating histone proteins to change the expression of homeostasis-related genes, as demonstrated by HOX transcript antisense intergenic RNA (HOTAIR) [77,78]. HOTAIR dysregulation may alter the epigenome, consequently, contributing to the disease initiation and progression through changing cellular homeostasis [77,78]. The expression of HOTAIR in cancer is positively correlated with growth, angiogenesis, progression and drug resistance, largely because it regulates several downstream targets via various signalling pathways, including mTOR signalling pathway [79] and Wnt/β-catenin pathway [80]. HOTAIR mediates these signalling pathways by upregulating the phosphorylation of phosphoinositide 3-kinase (PI3K), serine/threonine kinase (AKT) and mammalian target of rapamycin (mTOR) proteins [79,81].
LncRNAs are also implicated in dosage compensation, X-chromosome inactivation (XCI), which refers to the random selection and transcriptional silence of one of two X-chromosomes in females at the early stages of embryonic development, which is a unique dosage compensation mechanism found in mammals [82]. This process is modulated by X-inactive specific transcript (Xist) lncRNA in association with chromatin modifying complex [83]. Additionally, lncRNAs also play an important role in nuclear organization, and this is exemplified by Wang et al. [84] who showed that Metastasis Associated Lung Adenocarcinoma Transcript 1 (MALAT1) executes its oncogenic activities by shaping the activities of nuclear speckles. Although MALAT1 is non-essential for speckle formation and integrity, the knockdown of this lncRNA has been linked to phenotypic changes including impaired cell cycle progression [85], apoptosis [86] and reduced cell motility [87].
In addition to nuclear organization, lncRNAs are also involved in splicing process, lncRNAs regulate alternative splicing (AS) through different mechanisms, which include, interaction with splicing factors and initiating RNA-RNA duplexes with pre-mRNA molecules to affect splicing events. DiGeorge Syndrome Critical Region Gene 5 (DGCR5) lncRNA is implicated in alternative splicing events and upregulated in oesophageal squamous cell carcinoma (ESCC) [88,89]. Duan et al. [88] showed that DGCR5 interacts with Serine and Arginine Rich Splicing Factor 1 (SRSF1) and regulates the alternative splicing event of Myeloid leukemia 1 (MCL-1), an anti-apoptotic gene of the B-cell lymphoma 2 (Bcl-2) family, which leads to increased ratio of MCL-1L isoform, an oncogene that promotes tumour growth in myeloid cell leukemia. To promote cancer, MCL-1L keeps mitochondrial membranes stable, suppressing the release of cytochrome c,thus, stimulating cell survival and inhibiting apoptosis [90].
There is an increasing evidence that lncRNAs are deregulated by HIV, which is implicated in a number of diseases, including Tuberculosis [91] and HIV-mediated cancers [92]. HIV infection is accompanied by the integration of its genome randomly into the host genome [92]. This action induces structural alterations that promote replication, trigger innate immunity and ultimately weakens the immune system by destroying the white blood cells, thus, allowing the development of AIDS-defining cancers [[92], [93], [94]]. In each of these processes, HIV manipulates host lncRNAs that are involved in innate immunity or cellular response to DNA damage [93]. NRON serves as an example of host lncRNAs that are manipulated by HIV because it negatively regulates HIV replication, knockdown of the NRON by HIV has been shown to increase HIV replication through increasing the activity of nuclear paraspeckle assembly transcript 1 (NFAT1) which is augmented in cervical cancer and Kaposi's sarcoma [95,96].
As the HIV-infected population ages with occurrence among young adults of 15–24 years [97], research on HIV-associated cancers has become an area of focus; however, despite the use of Highly active antiretroviral therapy (HAART), HIV patients are still vulnerable to life threatening diseases such as virus induced cancers [98]. In HIV infected patients, co-infection with oncogenic viruses induces cell transformation, in addition, HIV also encode few proteins with pro-oncogenic effects, thus potentiating tumorigenesis [99]. To potentiate tumorigenesis, Tat protein transactivates the Human papillomavirus (HPV) long control region and enhances the expression of HPV-18 oncoprotein, E7, in cervical cancer cells [7]. Additionally, HIV oncoprotein, Negative factor (Nef), also promote cancer by increasing the expression of cellular myelocytomatosis (c-MYC) expression, which led to genomic instability in Burkitt lymphoma [100].
3. HIV-regulated lncRNAs promote carcinogenesis
It is known that HIV infection is a risk factor for the development of various types of cancers, including non-Hodgkin lymphoma [101], Kaposi's sarcoma [102] and cervical cancer [29,103]. Despite ongoing research, few factors are known to cause cancer among HIV-positive individuals. Immunodeficiency and the prevalence of traditional cancer risk factors (e.g., smoking, infection with oncogenic virus) [104,105] appear to be key risk factors, but evidence is emerging that HIV may also have direct pro-oncogenic effects as well. However, it remains elusive as to whether these factors work independently or in a synergistic manner. It is believed that HIV-associated immunodeficiency exerts its cancer-predisposing effects via two primary mechanisms [1]: reduced body's ability to clear and control oncogenic virus infections and [2] reduced immune surveillance of malignant cells [92,106]. As evidence accumulates, it has been found that HIV itself may exert direct oncogenic effects via its Tat protein [107] involving multifaceted mechanisms including synergism with other oncogenic viruses [107], blockage of tumour suppressor gene function [108] and disruption of cell cycle regulation [109].
Additionally, the development of HIV-associated cancers is partly attributed to deregulation of lncRNAs that modulate viral and host gene expressions. The role of lncRNAs in HIV-mediated carcinogenesis can be virus-dependent, because, although regulatory lncRNAs have been shown to originate from both the virus and the host cells [95], during infection, HIV modulate the expression profiles of both the host and viral lncRNA. The tightly regulated expression of these lncRNA may enable HIV to achieve synchronized gene expression required for replication success [95] thus leading to a weakened immune system that causes persistence of oncoviruses.
Deregulation of oncogenic lncRNAs such as MALAT-1 are linked to most cancers investigated to date and affect major cancer hallmarks [110,111]. To promote cancer, MALAT1 modulates several molecular signalling pathways, including MAPK/ERK [112], PI3K/AKT [113] and Wnt/β-catenin [114], positively influencing tumour cell proliferation, migration and invasion. In addition, HIV-1 also dysregulates lncRNAs to enhance replication and regulate the expression of host genes, bypassing apoptosis and the immune response, resulting in tumour formation [35,39]. LncRNAs deregulated by HIV-1 have crucial roles in several aspects of cancer, and these include regulation of cell survival; for example, NRON has been shown to inhibit cancer proliferation [45,115]. Additionally, lncRNAs deregulated by HIV-1 are also implicated in the regulation of apoptosis e.g p53-dependent lincRNA p-21 which plays a central role in global p53 responses to genomic instability [40].
Several lncRNAs such as GAS5, which is downregulated by HIV-1 viral infection, play a crucial role during carcinogenesis [73,116]. GAS5 expression levels are downregulated in numerous malignant tumours, including breast [117], gastric cancer [118], prostate cancer [119], lung cancer [120], and steosarcoma [121]. The low expression of this transcript increased proliferation and attenuated apoptosis because GAS5 overexpression leads to apoptosis and decrease in S-stage cells in normal cells, while GAS5 silencing has the opposite effect suggesting that GAS5 regulates normal growth stagnation [73,76,122]. Although the precise molecular mechanism of GAS5 in carcinogenesis remains unclear, it is possible that the downregulation of GAS5 in cancer is due to its tumour repressor effects, which include regulating transcription through acting as a decoy by binding to miR-196a-5p, which is associated with increased tumour growth [123,124] and by regulating transcription through histone methylation/demethylation.
NRON, which is involved in the regulation of HIV is also downregulated in triple-negative breast cancer (TNBC) in comparison to healthy tissues, thus indicating the possibility of being a tumour repressor [115]. Niu et al. [115] also showed that NRON overexpression downregulated small NF90-associated RNA (snaR) lncRNA to inhibit cancer cell proliferation. Generally, lncRNA NRON inhibits NFAT, a transcription factor that enhances HIV replication and transcription [45]. NRON also forms a complex with ubiquitin ligase cullin 4B (CUL4B) and Proteasome 26S Subunit, Non-ATPase 11 (PSMD11) to recruit HIV protein Tat which is associated with the occurrence of several tumours, for degradation through ubiquitination, suggesting tumour inhibitory effects of NRON [125].
4. HIV proteins modulate cancer-related lncRNAs
The viral proteins expressed and released by HIV-1-infected cells include Tat, viral envelope glycoprotein (gp120), Nef, reverse transcriptase (RT) and matrix protein p17 [8]. These proteins cause oxidative stress by deregulating oxidative stress pathways, leading to a surge in reactive oxygen species (ROS) production and inducing mitochondrial dysfunction, which gives rise to direct carcinogenic effects [8,[126], [127], [128], [129]]. The HIV proteins are also capable of enhancing tumorigenic potential in already transformed cells, where they can operate alone or together with other oncoproteins, including those from HPV, one of the most common oncoviruses [8]. As a stand alone carcinogenesis inducer, Tat promoted carcinogenesis by reducing the expression of a member of the Rb family, pRb2 and cyclin-dependent kinase inhibitors, p21 and p17, at the mRNA level [130].
Immunodeficiency associated with enhanced susceptibility to viral infections is considered the most important pathway of increased cancer risk in HIV patients [131]. There is substantial evidence which demonstrates that HIV is an oncogenic virus because it causes immunosuppression by permitting the coinfection of other oncogenic viruses [29,132]. To promote cancer, HIV also encode few proteins with pro-oncogenic effects [8,133]. These HIV proteins are implicated in HIV-mediated carcinogenesis and they modulate different cellular players, including lncRNAs (Table 2). As shown in Table 2, some HIV-1 proteins appear to be directly involved in cell transformation and propagation of malignant cells. These HIV-1 oncotranscripts favours neoplastic progression through modulation of different lncRNAs, including LINC00313, uc002yug.2, lincRNA-p21, HEAL and NRON, which are involved in HIV replication, apoptosis, angiogenesis and cell migration. Literature on the interaction of HIV matrix protein p17 with lncRNAs is scarce, but according to Lncrna2target V3.0 prediction tool (http://bio-annotation.cn/lncrna2target/browse.jsp), the first two lncRNAs targeted by p17 are lncRNA152 and NBAT1 which are both involved in carcinogenesis.
Table 2.
HIV proteins that promote carcinogenesis and modulate the expression of lncRNAs.
| HIV protein | Mode of action | LncRNAs | Cancer type | Reference |
|---|---|---|---|---|
| Tat | Induce angiogenesis and tumorigenesis by targeting LINC00313 lncRNAs. Facilitate the uc002yug.2-mediated regulation of HIV-1 reactivation. |
LINC00313 uc002yug.2- |
Kaposi's sarcoma Cervical cancer |
[38,133,134] |
| Gp120 | Invade apoptosis by dysregulating lincRNA-p21. | lincRNA-p21 | Glioblastomas | [135,136] |
| Reverse transcriptase | Enhance lncRNA HEAL through its ability to phosphorylate cyclin-dependent kinase 2 gene (CDK2) required for efficient HIV-1 replication. | HEAL | Murine mammary gland adenocarcinoma | [128,137] |
| Nef | Regulate NRON to enhance HIV replication and promote breast cancer growth and progression. | NRON | Breast cancer | [45,138] |
| p17 | Promote cell migration and invasion. | lncRNA152 NBAT1 | Breast cancer B-cell lymphomas |
[121,139] |
5. HIV-associated cancers and deregulated lncRNAs
HIV attacks the human body in three stages, the first stage is acute infection, which develops few weeks after exposure to the virus through bodily fluids of infected patient. During this stage, the body produces antibodies to fight off the HIV virus that is multiplying at a rapid rate. Once acute infection is over most patients do not experience any symptoms while the virus continues to replicate, this stage is called asymptomatic stage and it lasts for years [140]. Without HAART treatment, asymptomatic stage typically progresses to AIDS in 10 years or longer.
The progression from asymptomatic stage results in immunosuppression due to several factors, which include malfunctioning of innate signalling pathways, an increased viral replication rate and viral load, gradual damage of peripheral CD4+T cells, and reduction of T lymphocytes at mucosal sites, all of which contribute to AIDS development [140]. Based on these hallmarks of HIV infection, it is reasonable to conclude that the progression of HIV to its advanced stages is a key factor for HIV-mediated carcinogenesis because after infection, the host expresses HIV oncoproteins and becomes vulnerable to opportunistic infections resulting with immunodeficiency, which directly and indirectly cause HIV-related cancer such as Kaposi sarcoma, non-Hodgkin lymphoma, and cervical cancer with remarkable level of immunosuppression [[101], [102], [103]].
Early in its pandemic, HIV was linked to the development of cancer, some of the first reported cases included non-Hodgkin lymphoma (NHL) [141] and Kaposi sarcoma (KS) [142], which are now common cancers in AIDS patients [23]. Malignancies that are linked to HIV are classified as AIDS-defining cancers (ADCs) or non–AIDS-defining cancers (NADCs) [143]. ADCs are more common in people living with HIV and they signal progression to the development of AIDS. On the other hand, although NADCs are also associated with HIV, their manifestation does not mean that HIV has progressed to AIDS [143,144]. Kaposi sarcoma, non-Hodgkin lymphoma (NHL) and cervical cancer are regarded as AIDS-defining cancers [23]. Anal cancer, pulmonary cancer, head and neck cancer, liver cancer, Hodgkin lymphoma and lung cancer are regarded as non-AIDS-defining cancers [143,144]. HIV does not contribute directly to the development of NADCs but since HAART does not fully restore health, the suppressed immune system of HIV-infected people with reduced ability to control oncogenic viral processes explains the increased risk of developing NADCs [145].
HIV-infected people have 2800-fold increased risk of developing Kaposi sarcoma, 10-fold elevated risk for NHL and three times elevated risk for cervical cancer in comparison with the overall population [145,146]. Several factors increase the risk of cancer development amongst HIV-infected patients including the expression of HIV oncoproteins, viral coinfection, immunosuppression and high-risk lifestyle choices such as smoking [[145], [146], [147], [148]]. HIV-induced immune suppression increases replication of oncogenic viruses such as hepatitis viruses, HPV and Epstein-Barr virus, which directly cause cancer [[145], [146], [147], [148]]. Many etiological mechanisms contribute to HIV-associated cancers resulting in the imbalance between cellular proliferation and apoptosis. HIV influences cell proliferation by expressing Tat protein, which promotes cell survival by inducing an increase in the levels of proliferation markers such as cyclin A together with a decline in the expression of cell cycle inhibitors such p21, thus dodging apoptosis and favouring cancer development [124,149]. Previously, the carcinogenesis process has been linked to deregulated genes, especially the protein-coding genes, such as Retinoblastoma binding protein 6 (RBBP6) [150,151], Survivin [152] and Death-Associated Protein Kinase 1 (DAPK-1) [153], suggesting that these gene products play pivotal role in cell homeostasis.
Recently, there is an increasing number of lncRNAs that have cellular function and some are linked to tumorigenesis, including HIV-associated cancers [45,94,95,132,135]. HIV has been shown to influence proliferation, inhibiting apoptosis and promoting cell survival by inducing the expression of lncRNAs, which uses their oncogenic nature to promote the expression of anti apoptotic proteins (e.g Bcl-2), maneuvering DNA damage responses and cell cycle check points, thus promoting the development of ADCs [94].
5.1. Deregulated LncRNAs in Kaposi sarcoma
Kaposi sarcoma associated herpes virus (KSHV) infection is aggravated by HIV-1, which is among the most important co-pathogens [30]. KSHV causes Kaposi sarcoma (KS), a cancer of endothelial cells that is a hallmark of the worldwide AIDS crisis [103]. HIV promotes KS pathogenesis by two mechanisms: (i) inducing the production of inflammatory cytokines which causes a profound impairment of the immune system, allowing the development of KS [154], and (ii) the release of HIV proteins, particularly HIV Tat protein which promote human herpesvirus (HHV)-8 replication [155,156]. Furthermore, lncRNAs are also associated with Kaposi sarcoma (Table 3), the dysregulation of these lncRNAs in Kaposi sarcoma is correlated with the degree of cancer prognosis and has been shown to influence cell proliferation, invasiveness, metastasis and regulation of tumor suppressor proteins [155,156]. The association between lncRNAs and HIV infection has been demonstrated to orchestrate and manipulate transcriptional and post-transcriptional effects both in vitro and in vivo, making them a target for clinical use. in vivo studies on molecular mechanisms of lncRNAs in KS are limited, hence, clinical trials to explore and verify the safety and efficacy of lncRNA drugs are far from over. Despite the comparatively small number of in vivo studies currently available, lncRNAs have a pivotal role in cancer, as well as in HIV infection.
Table 3.
LncRNAs associated with the development and progression of ADCs.
| ADCs | Tumour suppressor inhibitor | Cell cycle dysregulation | Apoptosis inhibition |
|---|---|---|---|
| Kaposi sarcoma | PTENP1 [157] | LINC00313 [133] | DLEU2 [157] |
| UCA1 [48] | |||
| Non-Hodgkin lymphoma | TUG1 [158,159] | ZFAS1 [159] | ZFAS1 [159] |
| PANDA [158] | FAS-AS1 [160] | ||
| MALAT1 [161] | MIAT [162] | ||
| Cervical cancer | OIP5-AS1 [163] | DLEU2 [164] | GAS5 [58,165] |
| HOTAIR [166] | |||
| CRNDE [167,168] |
Note: (): Reference.
5.2. Deregulated LncRNAs in non-Hodgkin lymphoma
In HIV-infected individuals, non-hodgkin lymphoma (NHL) accounts for a large proportion of human malignancies. HIV induces immunosuppression and chronic antigenic stimulation, these conditions, coupled with infections such as Epstein-Barr virus which increases the expression of proteins involved in escaping antitumor immunity such as programmed death-ligand 1 (PD-L1), may provide a conducive environment for HIV-induced NHL [6]. Additionally, lncRNAs are implicated but the roles and mechanisms of abnormally expressed lncRNAs in NHL are still unclear. Several lncRNAs whose expression is aberrant in NHL have been examined functionally in an attempt to shed light on their role in various tumor cell biology aspects (Table 3). Huang et al. [169] also added that lncRNAs are also capable of regulating multiple signalling pathways such as Wnt/β-catenin and oncogenic genes through epigenetic mechanisms. Regarding epigenetic mechanisms, it was found that HOTAIR's function is positively correlated with epigenetic regulation through recruitment of Polycomb Repressive Complex 2 (PRC2) proteins (EZH2, SUZ12, and EED), which promote H3K27me3, that is strongly tied to aggressive Diffuse large B-cell lymphoma [169,170]. According to Oh et al. [170], it is possible that lncRNA deregulation can contributes to tumorigenesis by interacting with epigenetic regulators.
5.3. Deregulated LncRNAs in cervical cancer
HIV positive women have a higher prevalence of cervical precancerous lesions. Despite the successful use of HAART in reducing HIV related deaths, HAART prolongs life, increasing the risk of exposure to HPV and allowing for the onset of invasive cervical cancer [171]. Moreover, it has been suggested that lncRNAs play significant roles in cervical cancer progression by sponging miRNAs thus enhancing the oncogenic effect of viral oncoproteins [172]. This effect was shown by Thymopoietin pseudogene 2 (TMPOP2) lncRNA, which upregulated E6/E7 genes by scavenging tumor repressor microRNAs like miR-375 and miR-139 that target HPV E6/E7 mRNA [172]. Other mechanisms deregulated by lncRNAs in cervical cancer are summarised in Table 3. According to Table 3, lncRNAs play a critical role in enhancing the processes involved in viral-mediated cervical cancer.
Additionally, another lncRNA implicated in cervical cancer is HOTAIR, Sharma et al. [173] showed that HPV16 E7 modulate HOTAIR expression and function, this is possible because in order to promote cancer, HOTAIR recruits the chromatin remodelling complex PRC2, creating H3K27me3 marks which induce gene silencing. RNA immunoprecipitation confirmed the interaction between HPV16 E7 and HOTAIR, illustrating one of the mechanisms of HPV16 E7 carcinogenesis [173]. In summary, recent discoveries in this field have revealed the close connection between HIV, HPV and lncRNAs, proving that lncRNAs play a significant role in HPV-induced carcinogenesis.
6. HIV influence the functions of cellular lncRNAs
HIV is implicated in the regulation of decoy lncRNAs, which bind proteins or RNAs, resulting in the negative regulation of protein expression. GAS5, a tumour suppressor decoy lncRNA, is downregulated during HIV-1 infection, GAS5 interacts with microRNA-873 to repress its HIV-1 replication promoting activity [73]. Additionally, HIV has been shown to modulate the expression of lncRNAs involved in direct protein localization; in cancer cells, HIV modulated the expression of NEAT1, which binds to RNA-binding proteins to orchestrate the formation of paraspeckles [46]. The formation of paraspeckles are suggested to be involved in regulating gene expression by nuclear RNA retention [174]. Moreover, HIV can also influence signal lncRNAs, which have binding sites for transcription factors or chromatin-modifying enzymes in order to activate or repress gene expression. Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) lncRNA has been shown to promote HIV through binding to PRC2. Consequently, MALAT1 blocks the binding of Enhancer of zeste homolog 2 (EZH2), a core component of PRC2, to the HIV-1 promoter, making EZH2 unable to mediate the epigenetic silencing of HIV transcription because the progression of HIV is a key factor for HIV-mediated carcinogenesis [54]. Lastly, HIV also influences scaffold lncRNAs that act as organising structure where molecules can bind and interact in order to regulate gene expression. Imam et al. [45] exemplified this effect by showing that NRON, a noncoding repressor of the NFAT, is downregulated due to subsequent HIV-1 infection. NRON has been demonstrated to regulate HIV-1 replication by binding to NFAT as scaffold, thus blocking NFAT to promote HIV-1 infection [45].
7. Cell homeostasis and cell survival processes
In simple terms, cell homeostasis refers to the balance between cellular proliferation and apoptosis. When this balance is disturbed, the system moves from a physiological to a pathological state. Cellular homeostasis ensures organ integrity through the control of cell survival, proliferation, and death, in order to make sure that healthy cells as opposed to cancerous cells, generally stop dividing when they come into contact with neighboring cells [175]. A loss of this conserved genetic program favors neoplastic progression, a key step in malignant transformation. During cellular homeostasis, cell survival is controlled to ensure that cells only grow and divide when required because deregulation of this process is an important hallmark of carcinogenesis. Cell survival is regulated by cell survival pathways, which affect transformation-relevant biological processes, including proliferation, migration and angiogenesis [175].
To understand the role of HIV-regulated lncRNAs in cell homeostasis and cell survival, we have analysed data from various aspects of cancer development pathways/cellular processes including apoptosis, cell cycle, cell survival pathways and angiogenesis. Analysis of data from these processes and cancer drug resistance revealed an overlap between lncRNAs deregulated in HIV-associated cancer and lncRNA impacted by HIV. Examples of these lncRNAs include NEAT1, which regulate proliferation and invasion of cervical cancer cells by targeting AKT/PI3K [176]. Moreover, HIV also affects NEAT1 expression because it plays a role in maintaining the integrity of the nuclear paraspeckle required to promote HIV replication [46]. MALAT1 also serves as an example of those lncRNAs deregulated in HIV associated cancer and impacted by HIV. MALAT1 is an oncogenic lncRNA involved in proliferation, invasion and metastasis in KSHV tumorigenesis [177]. Additionally, highly upregulated MALAT1 promotes HIV transcription in HIV-infected CD4+ T lymphocytes [54].
HIV manipulate host lncRNAS by inducing strong modifications in the cell transcriptome [35]. This strategy enhances HIV's resistance to antiviral immune response, thus promoting the expression of lncRNAs which stimulate a variety of viral-host interactions, which contribute to the growth and spread of the virus [35]. Since the progression of HIV is a key factor for HIV-mediated carcinogenesis, it is possible that during the late stages of HIV infection, HIV alter the expression of tumour suppressor lncRNAs to promote the development and progression of cancer.
7.1. HIV regulated lncRNAs foster apoptosis resistance
Apoptosis is a programmed cell death essential for maintaining cell balance in the human body. One of the hallmarks of cancer cells is their resistance to apoptosis, which plays a key role in the prevention of cancer progression by targeting damaged pro-cancerous cells. Apoptosis initiates a cascade of caspase-related signalling complexes either in response to the intracellular signal or in response to the receptors on the cell surface (extrinsic pathways).
It is now emerging that the overexpression or knockdown of different HIV-regulated lncRNAs in specific types of tumours inhibit or activate the apoptosis process [178]. Fas-antisense 1 (Fas-AS1) is a type of lncRNA that is regulated by HIV, but the molecular mechanism remains to be experimentally validated, to support this notion, Boliar et al. [178] showed that the expression of Fas-AS1 is upregulated in HIV infected cells versus uninfected cells indicating that this variation is HIV infection specific. Fas-AS1 has been shown to inhibit apoptosis in macrophages by targeting effector caspases, interestingly, the down-regulation of Fas-AS1 expression with siRNA treatment activated effector caspase-3/7 only in HIV-infected macrophages without affecting the uninfected cells [178]. Therefore, it is possible that Fas-AS1 can use similar mechanism in cancer cells to either promote or inhibit apoptosis.
Another example is NEAT1 whose expression is altered by HIV-1 infection; this lncRNA is also implicated in imatinib-induced apoptosis inhibition of chronic myeloid leukemia cells [179]. To reverse apoptosis, other HIV-associated lncRNAs induce cell proliferation, which is exemplified by NFAT lncRNA. NFAT promotes cell proliferation through induction of glypican 6 (GPC6), a cell-surface glycoprotein [180]. NFAT directly binds to the GPC6 promoter and stimulates its transcription, to promote cell proliferation, GPC6 triggers cell proliferation by inhibiting canonical β-catenin and increasing non-canonical Wnt5A signalling, which induces p38 mitogen-activated protein kinase (MAPK) and c-Jun N-terminal kinase (JNK) to mediate resistance to apoptosis [180].
7.2. HIV-regulated lncRNAs modulate the cell cycle
The cell cycle in eukaryotes is controlled by regulatory networks, which ensure that specific events occur in an orderly manner through tightly regulated transitions. Contrary to the abundant knowledge of proteins that control the cell cycle [181], very little is known about the role of long non-coding RNAs, especially those that are regulated by HIV in cell-cycle progression. Since a large number of HIV-regulated lncRNAs display deregulated expression in human cancers, it is possible that these lncRNAs regulate the cell cycle. On that note, MALAT1, a lncRNA which promotes HIV transcription [54] has been shown to regulate the expression of cell cycle genes in bone cancer and is required for G1/S and mitotic progression. Additionally, GAS5, a lncRNA that inhibits HIV-1 replication through interaction with miR-873 [73], is also implicated in the cell cycle arrest in normal T-cell lines and human peripheral blood T-cells, GAS5 increased the proportion of cells in G1 while decreasing the proportion of cells in S phase [73].
7.3. HIV regulated lncRNAs are implicated in cell survival pathways
Survival pathways are known to prevent apoptosis, promote cell proliferation and cell survival [182]. HIV-regulated lncRNAs are deregulated in cell survival pathways indicating a need to explore them for biomarker discovery purposes. MALAT1, a lncRNA upregulated during HIV-1 infection, has been shown to promote the transcription of HIV-1 genes, especially those that are involved in cell survival pathways, such as Mitogen-activated protein kinase (MAPK) and phosphatidylinositol 3′-kinase (PI3K)-Akt pathways [183,184]. This survival-promoting effect of MALAT1 has been demonstrated in osteosarcoma [185]; bladder cancer [183] and breast cancer [186]. NEAT1 lncRNA showed a related mechanism in multiple myeloma by promoting cell proliferation through activation of PI3K/AKT pathway [187]. Interestingly, HIV infection alone also promoted cell proliferation, cell survival and angiogenesis by up-regulating ret proto-oncogene (RET) via sponging miR-129–5p, thus activating the PI3K-Akt survival pathway [55,185].
HIV also maneuver host antiviral response by activating survival pathways by negatively regulating LincRNA-p21, which indirectly deactivates cell survival pathways [188]. LincRNA-p21 is induced by p53 and binds to heterogeneous nuclear ribonucleoprotein K (hnRNP K), allowing it to be localized at the promoter of pro-apoptotic genes. As a means of preventing this pro-apoptotic pathway, HIV initiates MAP2K1/ERK2 pathway, which causes hnRNP-K to be sequestered in the cytoplasm. Activated MAP2K1/ERK2 pathway protects HIV-infected cells from p53-induced apoptosis by degrading lincRNA-p21 and sequestering hnRNP-K in the cytoplasm [188]. LincRNA-p21 have been shown to inhibit the growth and progression of non-small-cell lung cancer [189], head and neck squamous cell carcinoma [190], prostate cancer [191], liver cancer [192], gastric cancer [193], oesophageal squamous cell carcinoma [194] and colorectal cancer [195].
Since HIV-regulated lncRNAs are deregulated in cell survival pathways, it can be a promising strategy to target inhibitors of cell survival pathways to alter lncRNAs that are involved in cancer drug resistance. Example of lncRNAs that should be targeted is Activated in Renal cell carcinoma with sunitinib resistance (ARSR). ARSR enhanced drug resistance in numerous tumours such as hepatocellular carcinoma [196,197], osteosarcoma [198] and renal cancer [199] via activation of AKT signalling pathway. Interestingly, the PI3K inhibitor Buparlisib (BKM120) overcame this chemotherapy resistance by inhibiting the AKT, thus resulting in downregulated expression of ARSR long noncoding RNA [198,200]. Although the information on ARSR as a HIV-regulated lncRNA is limited, its downregulation by BKM120 suggests that inhibitors of cell survival pathways can be targeted to alter lncRNAs that are involved in cancer drug resistance.
7.4. HIV-regulated lncRNAs support angiogenesis
The development of new blood vessels (angiogenesis) is required in the pathogenesis of many disorders such as cancer, chronic inflammatory disorders (rheumatoid) and some eye diseases (age-related macular degeneration) [201−202]. Angiogenesis helps tumours to establish a blood supply to satisfy their need for oxygen and nutrients and to accomplish other metabolic functions [201,202]. Angiogenesis takes place during development, regeneration, and wound repair in physiological conditions [203]. The ability of tumours to induce the formation of new blood vessels has been a focus of cancer research for the past years and long noncoding RNAs are grabbing attention as rising stars in this process.
Studies on HIV-1 regulated lncRNAs implicated in promoting angiogenesis are limited but MALAT1, which is involved in HIV-1 viral infection has been shown to promote angiogenesis in breast cancer [204]. Interestingly, knockdown of MALAT1 inhibited breast cancer cell proliferation [204]. Similar effects of MALAT1 in angiogenesis have been demonstrated in hepatocellular carcinoma, gastric cancer and colorectal cancer [[205], [206], [207]].
8. LncRNAs in cancer drug resistance
The problem of drug resistance remains a major challenge as it often leads to therapeutic failure [208]. To date, chemotherapeutic drug resistance mechanisms are still poorly understood. Since lncRNAs regulate gene expression through mechanisms such as chromatin modification, transcriptional, and post-transcriptional regulation [209], which simultaneously occur during the development of drug resistance, therefore, the role of lncRNAs in drug resistance must be critically evaluated, especially in AIDS defining cancers. LncRNAs have been shown to mediate chemoresistance in renal cell carcinoma [210], bladder cancer [211], prostate cancer [212], testicular cancer [210,213] and cervical cancer [214]. LncRNAs contribute to chemoresistance in various ways, including altering drug efflux, preventing DNA damage repair, causing mutations in drug targets and sponging miRNAs [210,215,216].
Targeting other group of non coding RNAs called microRNAs is an interesting mechanism that lncRNAs are using to develop drug resistance across various cancer entities. ARSR lncRNA has been shown to induce drug resistance in Renal cell carcinoma (RCC), as a competing endogenous RNA (ceRNA), ARSR sequestered miR-34 and miR-449 to increase the levels of Anexelekto (AXL) receptor tyrosine kinase and mesenchymal-epithelial transition factor (c-MET), resulting in sunitinib resistance in RCC [199]. Another microRNA related mechanism, which enabled lncRNAs to regulate chemoresistance in cancer cells were described by Li et al. [217], where it was shown that deleted in lymphocytic leukemia 1 lncRNA (DLEU1) lncRNA induced cisplatin resistance in bladder cancer cells by sponging the tumor suppressive miR-99b. Although the information on the implication of HIV associated lncRNAs in drug resistance is limited, according to literature, microRNA could play an important role in regulating lncRNA-mediated drug resistance.
9. Conclusion
Investigation into the effect of HIV on the role and function of lncRNAs is still largely in its infancy. Future studies are required to have a deepened understanding of the role of HIV-regulated lncRNAs in promoting carcinogenesis targeting apoptosis, survival and angiogenesis processes. Since differentially expressed HIV-regulated lncRNAs act as negative or positive regulators in various critical steps of tumorigenesis, understanding of lncRNA-mediated carcinogenesis and drug resistance is of paramount importance. Deciphering the HIV-mediated processes, especially those linked to lncRNA deregulation may assist in the identification of novel diagnostic and prognostic biomarkers, as well as identification of new drug targets for therapeutic purposes.
Author contributions
Conceptualization, Z.M. and S.M., original draft preparation, L.M.; writing—review and editing, L.M and Z.M; revision and proofreading, S.M. All authors have read and agreed to the published version of the manuscript.
Funding
No funding
10. Institutional review board statement
Not applicable.
11. Informed consent statement
Not applicable.
12. Data availability statement
Data sharing is not applicable to this article.
Declaration of competing interest
The authors declare no conflict of interest.
References
- 1.Mbita Z., Hull R., Dlamini Z. Human immunodeficiency virus-1 (HIV-1)-mediated apoptosis: new therapeutic targets. Viruses. 2014 Aug;6(8):3181–3227. doi: 10.3390/v6083181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Reghine É.L., Foresto R.D., Kirsztajn G.M. HIV-related nephropathy: new aspects of an old paradigm. Rev. Assoc. Méd. Bras. 2020 Jan 13;66:s75–81. doi: 10.1590/1806-9282.66.S1.75. [DOI] [PubMed] [Google Scholar]
- 3.Savvoulidis P., Butler J., Kalogeropoulos A. Cardiomyopathy and heart failure in patients with HIV infection. Can. J. Cardiol. 2019 Mar 1;35(3):299–309. doi: 10.1016/j.cjca.2018.10.009. [DOI] [PubMed] [Google Scholar]
- 4.Dhokotera T., Bohlius J., Spoerri A., Egger M., Ncayiyana J., Olago V., Singh E., Sengayi M. The burden of cancers associated with HIV in the South African public health sector, 2004–2014: a record linkage study. Infect. Agents Cancer. 2019 Dec;14(1):1–2. doi: 10.1186/s13027-019-0228-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Brickman C., Palefsky J.M. Cancer in the HIV-infected host: epidemiology and pathogenesis in the antiretroviral era. Curr. HIV AIDS Rep. 2015 Dec;12(4):388–396. doi: 10.1007/s11904-015-0283-7. [DOI] [PubMed] [Google Scholar]
- 6.Puglia M., Stasi C., Da Frè M., Voller F. Prevalence and characteristics of HIV/HBV and HIV/HCV coinfections in Tuscany. Braz. J. Infect. Dis. 2016 Jul;20:330–334. doi: 10.1016/j.bjid.2015.11.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Barillari G., Palladino C., Bacigalupo I., Leone P., Falchi M., Ensoli B. Entrance of the Tat protein of HIV-1 into human uterine cervical carcinoma cells causes upregulation of HPV-E6 expression and a decrease in p53 protein levels. Oncol. Lett. 2016 Oct 1;12(4):2389–2394. doi: 10.3892/ol.2016.4921. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Isaguliants M., Bayurova E., Avdoshina D., Kondrashova A., Chiodi F., Palefsky J.M. Oncogenic effects of HIV-1 proteins, mechanisms behind. Cancers. 2021 Jan;13(2):305. doi: 10.3390/cancers13020305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Chopra N.K., Ni H., Lim V. Past present and future status of HIV-AIDS pandemic problem in world. Microbiol. Infect. Dis. 2019;3(1):1–6. [Google Scholar]
- 10.Zirulnik J.L., Retamar M.S., Perez H.M. HIV-Depression: a shadow over 90-90-90 unaids program. J. Infectiol. 2018 Jul 28;(1):1. [Google Scholar]
- 11.Roth G.A., Abate D., Abate K.H., Abay S.M., Abbafati C., Abbasi N., Abbastabar H., Abd-Allah F., Abdela J., Abdelalim A., Abdollahpour I. Global, regional, and national age-sex-specific mortality for 282 causes of death in 195 countries and territories, 1980–2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet. 2018 Nov 10;392(10159):1736–1788. doi: 10.1016/S0140-6736(18)32203-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Calles N.R., Evans D., Terlonge D. Baylor College of Medicine; Texas, USA: 2010. Pathophysiology of the Human Immunodeficiency Virus. HIV Curriculum for the Health Professional. Baylor Pediatrics International AIDS Iniciative; pp. 7–14. [Google Scholar]
- 13.Barmania F., Potgieter M., Pepper M.S. Mutations in CC chemokine receptor type 5 (CCR5) in South African individuals. Int. J. Infect. Dis. 2013 Dec 1;17(12):e1148–e1153. doi: 10.1016/j.ijid.2013.06.009. [DOI] [PubMed] [Google Scholar]
- 14.Nyamweya S., Hegedus A., Jaye A., Rowland‐Jones S., Flanagan K.L., Macallan D.C. Comparing HIV‐1 and HIV‐2 infection: lessons for viral immunopathogenesis. Rev. Med. Virol. 2013 Jul;23(4):221–240. doi: 10.1002/rmv.1739. [DOI] [PubMed] [Google Scholar]
- 15.NAM HIV-1 subtypes. 2021. https://www.aidsmap.com/about-hiv/hiv-1-subtypes Available at: Accessed.
- 16.Sharp P.M., Hahn B.H. Origins of HIV and the AIDS pandemic. Cold Spring Harbor Perspect. Med. 2011 Sep 1;1(1):a006841. doi: 10.1101/cshperspect.a006841. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Vessière A., Rousset D., Kfutwah A., Leoz M., Depatureaux A., Simon F., Plantier J.C. Diagnosis and monitoring of HIV-1 group O-infected patients in Cameroun. JAIDS J. Acquired Immun. Defic. Syndrom. 2010 Jan 1;53(1):107–110. doi: 10.1097/QAI.0b013e3181b97ec1. [DOI] [PubMed] [Google Scholar]
- 18.Vallari A., Holzmayer V., Harris B., Yamaguchi J., Ngansop C., Makamche F., Mbanya D., Kaptué L., Ndembi N., Gϋrtler L., Devare S. Confirmation of putative HIV-1 group P in Cameroon. J. Virol. 2011 Feb 1;85(3):1403–1407. doi: 10.1128/JVI.02005-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Bbosa N., Kaleebu P., Ssemwanga D. HIV subtype diversity worldwide. Curr. Opin. HIV AIDS. 2019 May 1;14(3):153–160. doi: 10.1097/COH.0000000000000534. [DOI] [PubMed] [Google Scholar]
- 20.Valadas E., Frañ a L., Sousa S., Antunes F. 20 years of HIV-2 infection in Portugal: trends and changes in epidemiology. Clin. Infect. Dis. 2009 Apr 15;48(8):1166–1167. doi: 10.1086/597504. [DOI] [PubMed] [Google Scholar]
- 21.Visseaux B., Damond F., Matheron S., Descamps D., Charpentier C. Hiv-2 molecular epidemiology. Infect. Genet. Evol. 2016 Dec 1;46:233–240. doi: 10.1016/j.meegid.2016.08.010. [DOI] [PubMed] [Google Scholar]
- 22.Ibe S., Yokomaku Y., Shiino T., Tanaka R., Hattori J., Fujisaki S., Iwatani Y., Mamiya N., Utsumi M., Kato S., Hamaguchi M. HIV-2 CRF01_AB: first circulating recombinant form of HIV-2. JAIDS J. Acquired Immun. Defic. Syndrom. 2010 Jul 1;54(3):241–247. doi: 10.1097/QAI.0b013e3181dc98c1. [DOI] [PubMed] [Google Scholar]
- 23.Yarchoan R., Uldrick T.S. HIV-associated cancers and related diseases. N. Engl. J. Med. 2018 Mar 15;378(11):1029–1041. doi: 10.1056/NEJMra1615896. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Bray F., Ferlay J., Soerjomataram I., Siegel R.L., Torre L.A., Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA A Cancer J. Clin. 2018 Nov;68(6):394–424. doi: 10.3322/caac.21492. [DOI] [PubMed] [Google Scholar]
- 25.Cagan R., Meyer P. Rethinking cancer: current challenges and opportunities in cancer research. Dis. Mod. Mech. 2017 Apr 1;10(4):349–352. doi: 10.1242/dmm.030007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Dalton M., Holzman E., Erwin E., Michelen S., Rositch A.F., Kumar S., Vanderpuye V., Yeates K., Liebermann E.J., Ginsburg O. Patient navigation services for cancer care in low-and middle-income countries: a scoping review. PLoS One. 2019 Oct 17;14(10) doi: 10.1371/journal.pone.0223537. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Mesri E.A., Feitelson M.A., Munger K. Human viral oncogenesis: a cancer hallmarks analysis. Cell Host Microbe. 2014 Mar 12;15(3):266–282. doi: 10.1016/j.chom.2014.02.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Mui U.N., Haley C.T., Tyring S.K. Viral oncology: molecular biology and pathogenesis. J. Clin. Med. 2017 Dec;6(12):111. doi: 10.3390/jcm6120111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Parkin D.M., Hämmerl L., Ferlay J., Kantelhardt E.J. Cancer in Africa 2018: the role of infections. Int. J. Cancer. 2020 Apr 15;146(8):2089–2103. doi: 10.1002/ijc.32538. [DOI] [PubMed] [Google Scholar]
- 30.Thakker S., Verma S.C. Co-infections and pathogenesis of KSHV-associated malignancies. Front. Microbiol. 2016 Feb 15;7:151. doi: 10.3389/fmicb.2016.00151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Akram N., Imran M., Noreen M., Ahmed F., Atif M., Fatima Z., Bilal Waqar A. Oncogenic role of tumor viruses in humans. Viral Immunol. 2017 Jan 1;30(1):20–27. doi: 10.1089/vim.2016.0109. [DOI] [PubMed] [Google Scholar]
- 32.Tornesello M.L., Annunziata C., Tornesello A.L., Buonaguro L., Buonaguro F.M. Human oncoviruses and p53 tumor suppressor pathway deregulation at the origin of human cancers. Cancers. 2018 Jul;10(7):213. doi: 10.3390/cancers10070213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Guven-Maiorov E., Tsai C.J., Nussinov R. Oncoviruses can drive cancer by rewiring signaling pathways through interface mimicry. Front. Oncol. 2019 Nov 15;9:1236. doi: 10.3389/fonc.2019.01236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Fang Y., Fullwood M.J. Roles, functions, and mechanisms of long non-coding RNAs in cancer. Dev. Reprod. Biol. 2016 Feb 1;14(1):42–54. doi: 10.1016/j.gpb.2015.09.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Fortes P., Morris K.V. Long noncoding RNAs in viral infections. Virus Res. 2016 Jan 2;212 doi: 10.1016/j.virusres.2015.10.002. 1-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Ray R.M., Morris K.V. Long non-coding RNAs mechanisms of action in HIV-1 modulation and the identification of novel therapeutic targets. Non-coding RNA. 2020 Mar;6(1):12. doi: 10.3390/ncrna6010012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Bhoopat L., Rangkakulnuwat S., Ya-In C, Bhoopat T. Relationship of cell bearing EBER and p24 antigens in biopsy-proven lymphocytic interstitial pneumonia in HIV-1 subtype E infected children. Appl. Immunohistochem. Mol. Morphol. 2011 Dec 1;19(6):547–551. doi: 10.1097/PAI.0b013e31821bfc34. [DOI] [PubMed] [Google Scholar]
- 38.Huan C., Li Z., Ning S., Wang H., Yu X.F., Zhang W. Long non-coding RNA uc002yug. 2 activates HIV-1 latency through regulation of mRNA levels of various RUNX1 isoforms and increased Tat expression. J. Virol. 2018 Apr 13;92(9) doi: 10.1128/JVI.01844-17. e01844-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Ouyang J., Hu J., Chen J.L. lncRNAs regulate the innate immune response to viral infection. Wiley Interdisciplinary Reviews: RNA. 2016 Jan;7(1):129–143. doi: 10.1002/wrna.1321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Sanchez Calle A., Kawamura Y., Yamamoto Y., Takeshita F., Ochiya T. Emerging roles of long non‐coding RNA in cancer. Cancer Sci. 2018 Jul;109(7):2093–2100. doi: 10.1111/cas.13642. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Wang W.T., Han C., Sun Y.M., Chen T.Q., Chen Y.Q. Noncoding RNAs in cancer therapy resistance and targeted drug development. J. Hematol. Oncol. 2019 Dec;12(1):1–5. doi: 10.1186/s13045-019-0748-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Yang L., Duff M.O., Graveley B.R., Carmichael G.G., Chen L.L. Genomewide characterization of non-polyadenylated RNAs. Genome Biol. 2011 Feb;12(2):1–4. doi: 10.1186/gb-2011-12-2-r16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Ma L., Bajic V.B., Zhang Z. On the classification of long non-coding RNAs. RNA Biol. 2013 Jun 1;10(6):924–933. doi: 10.4161/rna.24604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Fatica A., Bozzoni I. Long non-coding RNAs: new players in cell differentiation and development. Nat. Rev. Genet. 2014 Jan;15(1):7–21. doi: 10.1038/nrg3606. [DOI] [PubMed] [Google Scholar]
- 45.Imam H., Shahr Bano A., Patel P., Holla P., Jameel S. The lncRNA NRON modulates HIV-1 replication in a NFAT-dependent manner and is differentially regulated by early and late viral proteins. Sci. Rep. 2015 Mar 2;5(1) doi: 10.1038/srep08639. 1-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Zhang Q., Chen C.Y., Yedavalli V.S., Jeang K.T. NEAT1 long non-coding RNA and paraspeckle bodies modulate HIV-1 posttranscriptional expression. mBio. 2013 Jan 29;4(1) doi: 10.1128/mBio.00596-12. e00596-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Congrains A., Kamide K., Ohishi M., Rakugi H. ANRIL: molecular mechanisms and implications in human health. Int. J. Mol. Sci. 2013 Jan;14(1):1278–1292. doi: 10.3390/ijms14011278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Sethuraman S., Gay L.A., Jain V., Haecker I., Renne R. microRNA dependent and independent deregulation of long non-coding RNAs by an oncogenic herpesvirus. PLoS Pathog. 2017 Jul 17;13(7) doi: 10.1371/journal.ppat.1006508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Zhou Y., Zhang X., Klibanski A. MEG3 noncoding RNA: a tumor suppressor. J. Mol. Endocrinol. 2012 Jun 1;48(3):R45–R53. doi: 10.1530/JME-12-0008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Hsieh P.F., Yu C.C., Chu P.M., Hsieh P.L. Long non-coding RNA MEG3 in cellular stemness. Int. J. Mol. Sci. 2021 Jan;22(10):5348. doi: 10.3390/ijms22105348. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Tornesello M.L., Faraonio R., Buonaguro L., Annunziata C., Starita N., Cerasuolo A., Pezzuto F., Tornesello A.L., Buonaguro F.M. The role of microRNAs, long non-coding RNAs, and circular RNAs in cervical cancer. Front. Oncol. 2020:150. doi: 10.3389/fonc.2020.00150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Cantile M., Di Bonito M., Tracey De Bellis M., Botti G. Functional interaction among lncRNA HOTAIR and microRNAs in cancer and other human diseases. Cancers. 2021 Jan;13(3):570. doi: 10.3390/cancers13030570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Jiang Y., Li Y., Fang S., Jiang B., Qin C., Xie P., Zhou G., Li G. The role of MALAT1 correlates with HPV in cervical cancer. Oncol. Lett. 2014 Jun 1;7(6):2135–2141. doi: 10.3892/ol.2014.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Qu D., Sun W.W., Li L., Ma L., Sun L., Jin X., Li T., Hou W., Wang J.H. Long noncoding RNA MALAT1 releases epigenetic silencing of HIV-1 replication by displacing the polycomb repressive complex 2 from binding to the LTR promoter. Nucleic Acids Res. 2019 Apr 8;47(6):3013–3027. doi: 10.1093/nar/gkz117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Liang W.C., Fu W.M., Wong C.W., Wang Y., Wang W.M., Hu G.X., Zhang L., Xiao L.J., Wan D.C., Zhang J.F., Waye M.M. The lncRNA H19 promotes epithelial to mesenchymal transition by functioning as miRNA sponges in colorectal cancer. Oncotarget. 2015 Sep 8;6(26) doi: 10.18632/oncotarget.4154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Xin H., Li M., Cheng X., Wang T., Liu X., Zhang Y. Suppression of long non-coding RNA H19 inhibits proliferation, cell migration and invasion in human cervical cancer cells. Trop. J. Pharmaceut. Res. 2018;17(7):1249–1253. [Google Scholar]
- 57.Zhou Y., Chen B. GAS5-mediated regulation of cell signaling. Mol. Med. Rep. 2020 Oct 1;22(4):3049–3056. doi: 10.3892/mmr.2020.11435. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Ji J., Dai X., Yeung S.C., He X. The role of long non-coding RNA GAS5 in cancers. Cancer Manag. Res. 2019;11:2729. doi: 10.2147/CMAR.S189052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Wu H., Zheng J., Deng J., Zhang L., Li N., Li W., Li F., Lu J., Zhou Y. LincRNA-uc002yug. 2 involves in alternative splicing of RUNX1 and serves as a predictor for esophageal cancer and prognosis. Oncogene. 2015 Sep;34(36):4723–4734. doi: 10.1038/onc.2014.400. [DOI] [PubMed] [Google Scholar]
- 60.Yan M.D., Hong C.C., Lai G.M., Cheng A.L., Lin Y.W., Chuang S.E. Identification and characterization of a novel gene Saf transcribed from the opposite strand of Fas. Hum. Mol. Genet. 2005 Jun 1;14(11):1465–1474. doi: 10.1093/hmg/ddi156. [DOI] [PubMed] [Google Scholar]
- 61.Teixeira L.K., Carrossini N., Sécca C., Kroll J.E., DaCunha D.C., Faget D.V., Carvalho L.D., de Souza S.J., Viola J.P. NFAT1 transcription factor regulates cell cycle progression and cyclin E expression in B lymphocytes. Cell Cycle. 2016 Sep 1;15(17):2346–2359. doi: 10.1080/15384101.2016.1203485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Xin B., Ji K.Q., Liu Y.S., Zhao X.D. NFAT overexpression correlates with CA72-4 and poor prognosis of ovarian clear-cell carcinoma subtype. Reprod. Sci. 2021 Mar;28(3):745–756. doi: 10.1007/s43032-020-00368-3. [DOI] [PubMed] [Google Scholar]
- 63.Guo S., Chen W., Luo Y., Ren F., Zhong T., Rong M., Dang Y., Feng Z., Chen G. Clinical implication of long non-coding RNA NEAT1 expression in hepatocellular carcinoma patients. Int. J. Clin. Exp. Pathol. 2015;8(5):5395. [PMC free article] [PubMed] [Google Scholar]
- 64.Dimitrova N., Zamudio J.R., Jong R.M., Soukup D., Resnick R., Sarma K., Ward A.J., Raj A., Lee J.T., Sharp P.A., Jacks T. LincRNA-p21 activates p21 in cis to promote Polycomb target gene expression and to enforce the G1/S checkpoint. Mol. Cell. 2014 Jun 5;54(5):777–790. doi: 10.1016/j.molcel.2014.04.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Chen S., Liang H., Yang H., Zhou K., Xu L., Liu J., Lai B., Song L., Luo H., Peng J., Liu Z. LincRNa-p21: function and mechanism in cancer. Med. Oncol. 2017 May;34(5):1–8. doi: 10.1007/s12032-017-0959-5. [DOI] [PubMed] [Google Scholar]
- 66.Liao C., Long Z., Zhang X., Cheng J., Qi F., Wu S., Huang T. LncARSR sponges miR-129-5p to promote proliferation and metastasis of bladder cancer cells through increasing SOX4 expression. Int. J. Biol. Sci. 2020;16(1):1. doi: 10.7150/ijbs.39461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Liu C., Tian X., Zhang J., Jiang L. Long non-coding RNA DLEU1 promotes proliferation and invasion by interacting with miR-381 and enhancing HOXA13 expression in cervical cancer. Front. Genet. 2018:629. doi: 10.3389/fgene.2018.00629. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Liu T., Han Z., Li H., Zhu Y., Sun Z., Zhu A. LncRNA DLEU1 contributes to colorectal cancer progression via activation of KPNA3. Mol. Cancer. 2018 Dec;17(1):1–3. doi: 10.1186/s12943-018-0873-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Li Y., Li Y., Chen W., He F., Tan Z., Zheng J., Wang W., Zhao Q., Li J. NEAT expression is associated with tumor recurrence and unfavorable prognosis in colorectal cancer. Oncotarget. 2015 Sep 29;6(29) doi: 10.18632/oncotarget.4737. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Hu X., Bao J., Wang Z., Zhang Z., Gu P., Tao F., Cui D., Jiang W. The plasma lncRNA acting as fingerprint in non-small-cell lung cancer. Tumor Biol. 2016 Mar;37(3):3497–3504. doi: 10.1007/s13277-015-4023-9. [DOI] [PubMed] [Google Scholar]
- 71.Yuequan J., Shifeng C., Bing Z. Prognostic factors and family history for survival of esophageal squamous cell carcinoma patients after surgery. Ann. Thorac. Surg. 2010 Sep 1;90(3):908–913. doi: 10.1016/j.athoracsur.2010.05.060. [DOI] [PubMed] [Google Scholar]
- 72.Zhang J., Li Y., Liu Y., Xu G., Hei Y., Lu X., Liu W. Long non coding RNA NEAT1 regulates glioma cell proliferation and apoptosis by competitively binding to microRNA 324 5p and upregulating KCTD20 expression. Oncol. Rep. 2021 Jul 1;46(1):1–7. doi: 10.3892/or.2021.8076. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Kino T., Hurt D.E., Ichijo T., Nader N., Chrousos G.P. Noncoding RNA gas5 is a growth arrest–and starvation-associated repressor of the glucocorticoid receptor. Sci. Signal. 2010 Feb 2;3(107):ra8. doi: 10.1126/scisignal.2000568. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Choudhari R., Sedano M.J., Harrison A.L., Subramani R., Lin K.Y., Ramos E.I., Lakshmanaswamy R., Gadad S.S. Long noncoding RNAs in cancer: from discovery to therapeutic targets. Adv. Clin. Chem. 2020 Jan 1;95:105–147. doi: 10.1016/bs.acc.2019.08.003. [DOI] [PubMed] [Google Scholar]
- 75.Quinodoz S., Guttman M. Long noncoding RNAs: an emerging link between gene regulation and nuclear organization. Trends Cell Biol. 2014 Nov 1;24(11):651–663. doi: 10.1016/j.tcb.2014.08.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Ma C., Shi X., Zhu Q., Li Q., Liu Y., Yao Y., Song Y. The growth arrest-specific transcript 5 (GAS5): a pivotal tumor suppressor long noncoding RNA in human cancers. Tumor Biol. 2016 Feb;37(2):1437–1444. doi: 10.1007/s13277-015-4521-9. [DOI] [PubMed] [Google Scholar]
- 77.Gupta R.A., Shah N., Wang K.C., Kim J., Horlings H.M., Wong D.J., Tsai M.C., Hung T., Argani P., Rinn J.L., Wang Y. Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature. 2010 Apr;464(7291):1071–1076. doi: 10.1038/nature08975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Bhan A., Mandal S.S. LncRNA HOTAIR: a master regulator of chromatin dynamics and cancer. Biochim. Biophys. Acta, Rev. Cancer. 2015 Aug 1;1856(1):151–164. doi: 10.1016/j.bbcan.2015.07.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.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 Dec 25;468(4):561–567. doi: 10.1016/j.bbrc.2015.10.129. [DOI] [PubMed] [Google Scholar]
- 80.Li J., Yang S., Su N., Wang Y., Yu J., Qiu H., He X. Overexpression of long non-coding RNA HOTAIR leads to chemoresistance by activating the Wnt/β-catenin pathway in human ovarian cancer. Tumor Biol. 2016 Feb;37(2):2057–2065. doi: 10.1007/s13277-015-3998-6. [DOI] [PubMed] [Google Scholar]
- 81.Li Z., Qian J., Li J., Zhu C. Knockdown of lncRNA-HOTAIR downregulates the drug-resistance of breast cancer cells to doxorubicin via the PI3K/AKT/mTOR signaling pathway. Exp. Ther. Med. 2019 Jul 1;18(1):435–442. doi: 10.3892/etm.2019.7629. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Yu B., van Tol H.T., Stout T.A., Roelen B.A. Initiation of X chromosome inactivation during bovine embryo development. Cells. 2020 Apr 19;9(4):1016. doi: 10.3390/cells9041016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Wang W., Min L., Qiu X., Wu X., Liu C., Ma J., Zhang D., Zhu L. Biological function of long non-coding RNA (LncRNA) Xist. Front. Cell Dev. Biol. 2021 Jun 10;9 doi: 10.3389/fcell.2021.645647. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Wang X., Liu C., Zhang S., Yan H., Zhang L., Jiang A., Liu Y., Feng Y., Li D., Guo Y., Hu X. N6-methyladenosine modification of MALAT1 promotes metastasis via reshaping nuclear speckles. Dev. Cell. 2021 Mar 8;56(5):702–715. doi: 10.1016/j.devcel.2021.01.015. [DOI] [PubMed] [Google Scholar]
- 85.Tripathi V., Shen Z., Chakraborty A., Giri S., Freier S.M., Wu X., Zhang Y., Gorospe M., Prasanth S.G., Lal A., Prasanth K.V. Long noncoding RNA MALAT1 controls cell cycle progression by regulating the expression of oncogenic transcription factor B-MYB. PLoS Genet. 2013 Mar 21;9(3) doi: 10.1371/journal.pgen.1003368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Wang Y., Zhang Q. vol. 31. Cancer Biotherapy & Radiopharmaceuticals; 2020 Dec. (Long Noncoding RNA MALAT1 Knockdown Inhibits Proliferation, Migration, and Invasion and Promotes Apoptosis in Non-small-cell Lung Cancer Cells through Regulating miR-515-3p/TRIM65 axis). [DOI] [PubMed] [Google Scholar]
- 87.Gutschner T., Hämmerle M., Eißmann M., Hsu J., Kim Y., Hung G., Revenko A., Arun G., Stentrup M., Groß M., Zörnig M. The noncoding RNA MALAT1 is a critical regulator of the metastasis phenotype of lung cancer cells. Cancer Res. 2013 Feb 1;73(3):1180–1189. doi: 10.1158/0008-5472.CAN-12-2850. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Duan Y., Jia Y., Wang J., Liu T., Cheng Z., Sang M., Qin J., Liu L. Long noncoding RNA DGCR5 involves in tumorigenesis of esophageal squamous cell carcinoma via SRSF1-mediated alternative splicing of Mcl-1. Cell Death Dis. 2021 Jun 7;12(6):1–4. doi: 10.1038/s41419-021-03858-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Ouyang J., Zhong Y., Zhang Y., Yang L., Wu P., Hou X., Xiong F., Li X., Zhang S., Gong Z., He Y. Long non-coding RNAs are involved in alternative splicing and promote cancer progression. Br. J. Cancer. 2021 Nov;8:1–2. doi: 10.1038/s41416-021-01600-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Shimazu T., Degenhardt K., Nur-E-Kamal A., Zhang J., Yoshida T., Zhang Y., Mathew R., White E., Inouye M. NBK/BIK antagonizes MCL-1 and BCL-XL and activates BAK-mediated apoptosis in response to protein synthesis inhibition. Genes Dev. 2007 Apr 15;21(8):929–941. doi: 10.1101/gad.1522007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Bell L.C., Pollara G., Pascoe M., Tomlinson G.S., Lehloenya R.J., Roe J., Meldau R., Miller R.F., Ramsay A., Chain B.M., Dheda K. In vivo molecular dissection of the effects of HIV-1 in active tuberculosis. PLoS Pathog. 2016 Mar 17;12(3) doi: 10.1371/journal.ppat.1005469. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Dubrow R., Silverberg M.J., Park L.S., Crothers K., Justice A.C. HIV infection, aging, and immune function: implications for cancer risk and prevention. Curr. Opin. Oncol. 2012 Sep;24(5):506. doi: 10.1097/CCO.0b013e328355e131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Iijima K., Kobayashi J., Ishizaka Y. Structural alteration of DNA induced by viral protein R of HIV-1 triggers the DNA damage response. Retrovirology. 2018 Dec;15(1):1–25. doi: 10.1186/s12977-018-0391-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Shen L., Wu C., Zhang J., Xu H., Liu X., Wu X., Wang T., Mao L. Roles and potential applications of lncRNAs in HIV infection. Int. J. Infect. Dis. 2020;92:97–104. doi: 10.1016/j.ijid.2020.01.006. Mar 1. [DOI] [PubMed] [Google Scholar]
- 95.Lazar D.C., Morris K.V., Saayman S.M. The emerging role of long non-coding RNAs in HIV infection. Virus Res. 2016 Jan 2;212:114–126. doi: 10.1016/j.virusres.2015.07.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Ram B.M., Dolpady J., Kulkarni R., Usha R., Bhoria U., Poli U.R., Islam M., Trehanpati N., Ramakrishna G. Human papillomavirus (HPV) oncoprotein E6 facilitates Calcineurin-Nuclear factor for activated T cells 2 (NFAT2) signaling to promote cellular proliferation in cervical cell carcinoma. Exp. Cell Res. 2018 Jan 1;362(1):132–141. doi: 10.1016/j.yexcr.2017.11.010. [DOI] [PubMed] [Google Scholar]
- 97.Gaumer G., Jordan M., Sherafat-Kazemzadeh R., Hariharan D., Bosman V., Nandakumar A.K. The role of economic factors and risky behavior for youth and young adults in the HIV epidemic in 29 low-and middle-income countries. J. Global Health Rep. 2021 Apr 19;5 [Google Scholar]
- 98.Shmakova A., Germini D., Vassetzky Y. HIV‐1, HAART and cancer: a complex relationship. Int. J. Cancer. 2020 May 15;146(10):2666–2679. doi: 10.1002/ijc.32730. [DOI] [PubMed] [Google Scholar]
- 99.Loarca L., Fraietta J.A., Pirrone V., Szep Z., Wigdahl B. Human immunodeficiency and virus (HIV) infection and cancer. HIV/AIDS contemp. Chall. 2017 Feb;22:1. [Google Scholar]
- 100.Mdletshe N., Nel A., Shires K., Mowla S. HIV Nef enhances the expression of oncogenic c-MYC and activation-induced cytidine deaminase in Burkitt lymphoma cells, promoting genomic instability. Infect. Agents Cancer. 2020 Dec;15(1) 1-0. [Google Scholar]
- 101.Gibson T.M., Morton L.M., Shiels M.S., Clarke C.A., Engels E.A. Risk of non-Hodgkin lymphoma subtypes in HIV-infected people during the HAART era: a population-based study. AIDS (Lond.) 2014 Sep 24;28(15):2313. doi: 10.1097/QAD.0000000000000428. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Gonçalves P.H., Uldrick T.S., Yarchoan R. HIV-associated Kaposi sarcoma and related diseases. AIDS (Lond.) 2017 Sep 10;31(14):1903. doi: 10.1097/QAD.0000000000001567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Vangipuram R., Tyring S.K. HIV/AIDS-Associated Viral Oncogenesis; 2019. AIDS-Associated Malignancies; pp. 1–21. [DOI] [PubMed] [Google Scholar]
- 104.Grulich A.E., Jin F., Poynten I.M., Vajdic C.M. HIV, cancer, and aging. Sex. Health. 2011 Jul 29;8(4):521–525. doi: 10.1071/SH11048. [DOI] [PubMed] [Google Scholar]
- 105.Lifson A.R., Lando H.A. Smoking and HIV: prevalence, health risks, and cessation strategies. Curr. HIV AIDS Rep. 2012 Sep;9(3):223–230. doi: 10.1007/s11904-012-0121-0. [DOI] [PubMed] [Google Scholar]
- 106.Borges Á.H., Dubrow R., Silverberg M.J. Factors contributing to risk for cancer among HIV-infected individuals, and evidence that earlier cART will alter this risk. Curr. Opin. HIV AIDS. 2014 Jan;9(1):34. doi: 10.1097/COH.0000000000000025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Zhou F., Xue M., Qin D., Zhu X., Wang C., Zhu J., Hao T., Cheng L., Chen X., Bai Z., Feng N. HIV-1 Tat promotes Kaposi's sarcoma-associated herpesvirus (KSHV) vIL-6-induced angiogenesis and tumorigenesis by regulating PI3K/PTEN/AKT/GSK-3β signaling pathway. PLoS One. 2013 Jan 2;8(1) doi: 10.1371/journal.pone.0053145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Harrod R., Nacsa J., Van Lint C., Hansen J., Karpova T., McNally J., Franchini G. Human immunodeficiency virus type-1 Tat/co-activator acetyltransferase interactions inhibit p53Lys-320acetylation and p53-responsive transcription. J. Biol. Chem. 2003 Apr 4;278(14):12310–12318. doi: 10.1074/jbc.M211167200. [DOI] [PubMed] [Google Scholar]
- 109.Amini S., Khalili K., Sawaya B.E. Effect of HIV-1 Vpr on cell cycle regulators. DNA Cell Biol. 2004 Apr 1;23(4):249–260. doi: 10.1089/104454904773819833. [DOI] [PubMed] [Google Scholar]
- 110.Schmitt A.M., Chang H.Y. Long noncoding RNAs in cancer pathways. Cancer Cell. 2016 Apr 11;29(4):452–463. doi: 10.1016/j.ccell.2016.03.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Li Z.X., Zhu Q.N., Zhang H.B., Hu Y., Wang G., Zhu Y.S. MALAT1: a potential biomarker in cancer. Cancer Manag. Res. 2018;10:6757. doi: 10.2147/CMAR.S169406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Liu S., Yan G., Zhang J., Yu L. Knockdown of long noncoding RNA (lncRNA) metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) inhibits proliferation, migration, and invasion and promotes apoptosis by targeting miR-124 in retinoblastoma. Oncol. Res. 2018;26(4):581. doi: 10.3727/096504017X14953948675403. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 113.Dong Y., Liang G., Yuan B.O., Yang C., Gao R., Zhou X. MALAT1 promotes the proliferation and metastasis of osteosarcoma cells by activating the PI3K/Akt pathway. Tumor Biol. 2015 Mar;36(3):1477–1486. doi: 10.1007/s13277-014-2631-4. [DOI] [PubMed] [Google Scholar]
- 114.Liang J., Liang L., Ouyang K., Li Z., Yi X. MALAT 1 induces tongue cancer cells' EMT and inhibits apoptosis through Wnt/β‐catenin signaling pathway. J. Oral Pathol. Med. 2017 Feb;46(2):98–105. doi: 10.1111/jop.12466. [DOI] [PubMed] [Google Scholar]
- 115.Niu L., Fan Q., Yan M., Wang L. LncRNA NRON down-regulates lncRNA snaR and inhibits cancer cell proliferation in TNBC. Biosci. Rep. 2019 May 1;39(5) doi: 10.1042/BSR20190468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Chen L., Chen L., Zuo L., Gao Z., Shi Y., Yuan P., Han S., Yin J., Peng B., He X., Liu W. Long noncoding RNA GAS5 inhibits HIV-1 replication through interaction with miR-873. AIDS Res. Hum. Retrovir. 2018 Jun 1;34(6):544–549. doi: 10.1089/AID.2017.0177. [DOI] [PubMed] [Google Scholar]
- 117.Mourtada-Maarabouni M., Pickard M.R., Hedge V.L., Farzaneh F., Williams G.T. GAS5, a non-protein-coding RNA, controls apoptosis and is downregulated in breast cancer. Oncogene. 2009 Jan;28(2):195–208. doi: 10.1038/onc.2008.373. [DOI] [PubMed] [Google Scholar]
- 118.Sun M., Jin F.Y., Xia R., Kong R., Li J.H., Xu T.P., Liu Y.W., Zhang E.B., Liu X.H., De W. Decreased expression of long noncoding RNA GAS5 indicates a poor prognosis and promotes cell proliferation in gastric cancer. BMC Cancer. 2014 Dec;14(1):1–2. doi: 10.1186/1471-2407-14-319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Pickard M.R., Mourtada-Maarabouni M., Williams G.T. Long non-coding RNA GAS5 regulates apoptosis in prostate cancer cell lines. Biochim. Biophys. Acta, Mol. Basis Dis. 2013 Oct 1;1832(10):1613–1623. doi: 10.1016/j.bbadis.2013.05.005. [DOI] [PubMed] [Google Scholar]
- 120.Tan Q., Zuo J., Qiu S., Yu Y., Zhou H., Li N., Wang H., Liang C., Yu M., Tu J. Identification of circulating long non-coding RNA GAS5 as a potential biomarker for non-small cell lung cancer diagnosisnon-small cell lung cancer, long non-coding RNA, plasma, GAS5, biomarker. Int. J. Oncol. 2017 May 1;50(5):1729–1738. doi: 10.3892/ijo.2017.3925. [DOI] [PubMed] [Google Scholar]
- 121.Ye K., Wang S., Zhang H., Han H., Ma B., Nan W. Long noncoding RNA GAS5 suppresses cell growth and epithelial–mesenchymal transition in osteosarcoma by regulating the miR‐221/ARHI pathway. J. Cell. Biochem. 2017 Dec;118(12):4772–4781. doi: 10.1002/jcb.26145. [DOI] [PubMed] [Google Scholar]
- 122.Mourtada-Maarabouni M., Hedge V.L., Kirkham L., Farzaneh F., Williams G.T. Growth arrest in human T-cells is controlled by the non-coding RNA growth-arrest-specific transcript 5 (GAS5) J. Cell Sci. 2008 Apr 1;121(7):939–946. doi: 10.1242/jcs.024646. [DOI] [PubMed] [Google Scholar]
- 123.Zhao X., Wang P., Liu J., Zheng J., Liu Y., Chen J., Xue Y. Gas5 exerts tumor-suppressive functions in human glioma cells by targeting miR-222. Mol. Ther. 2015 Dec 1;23(12):1899–1911. doi: 10.1038/mt.2015.170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Zhao H., Yu H., Zheng J., Ning N., Tang F., Yang Y., Wang Y. Lowly-expressed lncRNA GAS5 facilitates progression of ovarian cancer through targeting miR-196-5p and thereby regulating HOXA5. Gynecol. Oncol. 2018 Nov 1;151(2):345–355. doi: 10.1016/j.ygyno.2018.08.032. [DOI] [PubMed] [Google Scholar]
- 125.Li J., Chen C., Ma X., Geng G., Liu B., Zhang Y., Zhang S., Zhong F., Liu C., Yin Y., Cai W. Long noncoding RNA NRON contributes to HIV-1 latency by specifically inducing tat protein degradation. Nat. Commun. 2016 Jun 13;7(1) doi: 10.1038/ncomms11730. 1-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Ivanov A.V., Valuev-Elliston V.T., Ivanova O.N., Kochetkov S.N., Starodubova E.S., Bartosch B., Isaguliants M.G. vol. 2016. 2016. Oxidative Stress during HIV Infection: Mechanisms and Consequences. (Oxidative Medicine and Cellular Longevity). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Caccuri F., Giordano F., Barone I., Mazzuca P., Giagulli C., Andò S., Caruso A., Marsico S. HIV-1 matrix protein p17 and its variants promote human triple negative breast cancer cell aggressiveness. Infect. Agents Cancer. 2017 Dec;12(1):1–9. doi: 10.1186/s13027-017-0160-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Bayurova E., Jansons J., Skrastina D., Smirnova O., Mezale D., Kostyusheva A., Kostyushev D., Petkov S., Podschwadt P., Valuev-Elliston V., Sasinovich S. HIV-1 reverse transcriptase promotes tumor growth and metastasis formation via ROS-dependent upregulation of twist. Oxid. Med. Cell. Longev. 2019 Dec;2:2019. doi: 10.1155/2019/6016278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Santerre M., Chatila W., Wang Y., Mukerjee R., Sawaya B.E. HIV-1 Nef promotes cell proliferation and microRNA dysregulation in lung cells. Cell Cycle. 2019 Jan 17;18(2):130–142. doi: 10.1080/15384101.2018.1557487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Nyagol J., Leucci E., Omnis A., De Falco G., Tigli C., Sanseverino F., Torriccelli M., Palummo N., Pacenti L., Santopietro R., Spina D. The effects of HIV-1 Tat protein on cell cycle during cervical carcinogenesis. Cancer Biol. Ther. 2006 Jun 1;5(6):684–690. doi: 10.4161/cbt.5.6.2907. [DOI] [PubMed] [Google Scholar]
- 131.Bruyand M., Thiébaut R., Lawson-Ayayi S., Joly P., Sasco A.J., Mercié P., Pellegrin J.L., Neau D., Dabis F., Morlat P., Chêne G. Role of uncontrolled HIV RNA level and immunodeficiency in the occurrence of malignancy in HIV-infected patients during the combination antiretroviral therapy era: agence Nationale de Recherche sur le Sida (ANRS) CO3 Aquitaine Cohort. Clin. Infect. Dis. 2009 Oct 1;49(7):1109–1116. doi: 10.1086/605594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Anampa J., Barta S.K., Haigentz M., Sparano J.A. vol. 1. Elsevier; 2020 Jan. Human immunodeficiency virus (HIV) infection and cancer; pp. 894–903. (InAbeloff's Clinical Oncology). [Google Scholar]
- 133.Yang W.S., Lin T.Y., Chang L., Yeh W.W., Huang S.C., Chen T.Y., Hsieh Y.T., Chen S.T., Li W.C., Pan C.C., Campbell M. HIV-1 Tat interacts with a Kaposi's sarcoma-associated herpesvirus reactivation-upregulated antiangiogenic long noncoding RNA, LINC00313, and antagonizes its function. J. Virol. 2020 Jan 17;94(3) doi: 10.1128/JVI.01280-19. e01280-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Vendeville A., Rayne F., Bonhoure A., Bettache N., Montcourrier P., Beaumelle B. HIV-1 Tat enters T cells using coated pits before translocating from acidified endosomes and eliciting biological responses. Mol. Biol. Cell. 2004 May;15(5):2347–2360. doi: 10.1091/mbc.E03-12-0921. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Barichievy S., Naidoo J., Boullé M., Scholefield J., Parihar S.P., Coussens A.K., Brombacher F., Sigal A., Mhlanga M.M. Viral apoptosis evasion via the MAPK pathway by use of a host long noncoding RNA. Front. Cell. Infect. Microbiol. 2018 Aug 3;8:263. doi: 10.3389/fcimb.2018.00263. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Valentín-Guillama G., López S., Kucheryavykh Y.V., Chorna N.E., Pérez J., Ortiz-Rivera J., Inyushin M., Makarov V., Valentín-Acevedo A., Quinones-Hinojosa A., Boukli N. HIV-1 envelope protein gp120 promotes proliferation and the activation of glycolysis in glioma cell. Cancers. 2018 Sep;10(9):301. doi: 10.3390/cancers10090301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Leng J., Ho H.P., Buzon M.J., Pereyra F., Walker B.D., Xu G.Y., Chang E.J., Lichterfeld M. A cell-intrinsic inhibitor of HIV-1 reverse transcription in CD4+ T cells from elite controllers. Cell Host Microbe. 2014 Jun 11;15(6):717–728. doi: 10.1016/j.chom.2014.05.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Manninen A., Renkema G.H., Saksela K. Synergistic activation of NFAT by HIV-1 nef and the Ras/MAPK pathway. J. Biol. Chem. 2000 Jun 2;275(22):16513–16517. doi: 10.1074/jbc.M910032199. [DOI] [PubMed] [Google Scholar]
- 139.Dolcetti R., Giagulli C., He W., Selleri M., Caccuri F., Eyzaguirre L.M., Mazzuca P., Corbellini S., Campilongo F., Marsico S., Giombini E. Role of HIV-1 matrix protein p17 variants in lymphoma pathogenesis. Proc. Natl. Acad. Sci. USA. 2015 Nov 17;112(46):14331–14336. doi: 10.1073/pnas.1514748112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Elfaki M.G. Immunosuppression induced by HIV infection. Biol. Med. 2014 Jul 1;6(3):1. [Google Scholar]
- 141.Somay K., Çöpür S., Osmanbaşoğlu E., Masyan H., Arslan H., Akay O.M., Tekin S., Ferhanoğlu B. HIV-associated non Hodgkin lymphoma: a case series study from Turkey. African J. Infect. Dis. 2020;14(2):42–47. doi: 10.21010/ajid.v14i2.7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Colafigli M., Bonadies A., Ferraresi V., Tonachella R., Cristaudo A., Latini A. Kaposi Sarcoma in HIV-infected patients: an infectious-dermatological outpatient service experience. Infect. Dis. Trop. Med. 2017;3:e410. [Google Scholar]
- 143.Silverberg M.J., Abrams D.I. AIDS-defining and non-AIDS-defining malignancies: cancer occurrence in the antiretroviral therapy era. Curr. Opin. Oncol. 2007 Sep 1;19(5):446–451. doi: 10.1097/CCO.0b013e3282c8c90d. [DOI] [PubMed] [Google Scholar]
- 144.Mitsuyasu R.T. Non–AIDS-defining cancers. Topics Antiviral Med. 2014 Jun;22(3):660. [PMC free article] [PubMed] [Google Scholar]
- 145.Wang C.C., Silverberg M.J., Abrams D.I. Non-AIDS-defining malignancies in the HIV-infected population. Curr. Infect. Dis. Rep. 2014 Jun;16(6):1–7. doi: 10.1007/s11908-014-0406-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Engels E.A., Biggar R.J., Hall H.I., Cross H., Crutchfield A., Finch J.L., Grigg R., Hylton T., Pawlish K.S., McNeel T.S., Goedert J.J. Cancer risk in people infected with human immunodeficiency virus in the United States. Int. J. Cancer. 2008 Jul 1;123(1):187–194. doi: 10.1002/ijc.23487. [DOI] [PubMed] [Google Scholar]
- 147.Javadi S., Menias C.O., Karbasian N., Shaaban A., Shah K., Osman A., Jensen C.T., Lubner M.G., Gaballah A.H., Elsayes K.M. HIV-related malignancies and mimics: imaging findings and management. Radiographics. 2018 Nov;38(7):2051–2068. doi: 10.1148/rg.2018180149. [DOI] [PubMed] [Google Scholar]
- 148.Salters K.A., Cescon A., Zhang W., Ogilvie G., Murray M.C., Coldman A., Hamm J., Chiu C.G., Montaner J.S., Wiseman S.M., Money D. Cancer incidence among HIV‐positive women in British Columbia, Canada: heightened risk of virus‐related malignancies. HIV Med. 2016 Mar;17(3):188–195. doi: 10.1111/hiv.12290. [DOI] [PubMed] [Google Scholar]
- 149.Huynh D., Vincan E., Mantamadiotis T., Purcell D., Chan C.K., Ramsay R. Oncogenic properties of HIV-Tat in colorectal cancer cells. Curr. HIV Res. 2007 Jul 1;5(4):403–409. doi: 10.2174/157016207781023974. [DOI] [PubMed] [Google Scholar]
- 150.Mbita Z., Naicker S., Goetsch S., Dlamini Z. The association of RBBP6 variant 3 expressions with apoptosis in human immunodeficiency virus-associated nephropathy (HIVAN) Exp. Mol. Pathol. 2015 Aug 1;99(1):74–80. doi: 10.1016/j.yexmp.2015.04.005. [DOI] [PubMed] [Google Scholar]
- 151.Makgoo L., Laka K., Mbita Z. Downregulation of RBBP6 variant 1 during arsenic trioxide-mediated cell cycle arrest and curcumin-induced apoptosis in MCF-7 breast cancer cells. Future Science OA. 2019 Jul 30;5(8):FSO409. doi: 10.2144/fsoa-2019-0047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Laka K., Makgoo L., Mbita Z. Survivin splice variants in arsenic trioxide (As2O3)-Induced deactivation of PI3K and MAPK cell signalling pathways in MCF-7 cells. Genes. 2019 Jan;10(1):41. doi: 10.3390/genes10010041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Huang Y., Chen L., Guo L., Hupp T.R., Lin Y. Evaluating DAPK as a therapeutic target. Apoptosis. 2014 Feb;19(2):371–386. doi: 10.1007/s10495-013-0919-2. [DOI] [PubMed] [Google Scholar]
- 154.Rezaei N., Hedayat M., Aghamohammadi A., Nichols K.E. Primary immunodeficiency diseases associated with increased susceptibility to viral infections and malignancies. J. Allergy Clin. Immunol. 2011 Jun 1;127(6):1329–1341. doi: 10.1016/j.jaci.2011.02.047. [DOI] [PubMed] [Google Scholar]
- 155.Ensoli B., Gendelman R., Markham P., Fiorelli V., Colombini S., Raffeld M., Cafaro A., Chang H.K., Brady J.N., Gallo R.C. Synergy between basic fibroblast growth factor and HIV-1 Tat protein in induction of Kaposi's sarcoma. Nature. 1994 Oct;371(6499):674–680. doi: 10.1038/371674a0. [DOI] [PubMed] [Google Scholar]
- 156.Foreman K.E. Kaposi's sarcoma: the role of HHV-8 and HIV-1 in pathogenesis. Expet Rev. Mol. Med. 2001 Mar;3(9):1–7. doi: 10.1017/S1462399401002733. [DOI] [PubMed] [Google Scholar]
- 157.Sethuraman S., Thomas M., Gay L.A., Renne R. Computational analysis of ribonomics datasets identifies long non-coding RNA targets of γ-herpesviral miRNAs. Nucleic Acids Res. 2018 Sep 19;46(16):8574–8589. doi: 10.1093/nar/gky459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Wang Y., Zhang M., Xu H., Wang Y., Li Z., Chang Y., Wang X., Fu X., Zhou Z., Yang S., Wang B. Discovery and validation of the tumor-suppressive function of long noncoding RNA PANDA in human diffuse large B-cell lymphoma through the inactivation of MAPK/ERK signaling pathway. Oncotarget. 2017 Sep 22;8(42) doi: 10.18632/oncotarget.20053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Baytak E., Gong Q., Akman B., Yuan H., Chan W.C., Küçük C. Whole transcriptome analysis reveals dysregulated oncogenic lncRNAs in natural killer/T-cell lymphoma and establishes MIR155HG as a target of PRDM1. Tumor Biol. 2017 Apr;39(5) doi: 10.1177/1010428317701648. [DOI] [PubMed] [Google Scholar]
- 160.Sehgal L., Mathur R., Braun F.K., Wise J.F., Berkova Z., Neelapu S., Kwak L.W., Samaniego F. FAS-antisense 1 lncRNA and production of soluble versus membrane Fas in B-cell lymphoma. Leukemia. 2014 Dec;28(12):2376–2387. doi: 10.1038/leu.2014.126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Wang X., Sehgal L., Jain N., Khashab T., Mathur R., Samaniego F. LncRNA MALAT1 promotes development of mantle cell lymphoma by associating with EZH2. J. Transl. Med. 2016 Dec;14(1):1–4. doi: 10.1186/s12967-016-1100-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Sattari A., Siddiqui H., Moshiri F., Ngankeu A., Nakamura T., Kipps T.J., Croce C.M. Upregulation of long noncoding RNA MIAT in aggressive form of chronic lymphocytic leukemias. Oncotarget. 2016 Aug 23;7(34) doi: 10.18632/oncotarget.11099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Kim J., Abdelmohsen K., Yang X., De S., Grammatikakis I., Noh J.H., Gorospe M. LncRNA OIP5-AS1/cyrano sponges RNA-binding protein HuR. Nucleic Acids Res. 2016 Mar 18;44(5):2378–2392. doi: 10.1093/nar/gkw017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.He M., Wang Y., Cai J., Xie Y., Tao C., Jiang Y., Li H., Song F. LncRNA DLEU2 promotes cervical cancer cell proliferation by regulating cell cycle and NOTCH pathway. Exp. Cell Res. 2021 May 1;402(1) doi: 10.1016/j.yexcr.2021.112551. [DOI] [PubMed] [Google Scholar]
- 165.Yang W., Hong L., Xu X., Wang Q., Huang J., Jiang L. LncRNA GAS5 suppresses the tumorigenesis of cervical cancer by downregulating miR-196a and miR-205. Tumor Biol. 2017 Jun;39(7) doi: 10.1177/1010428317711315. [DOI] [PubMed] [Google Scholar]
- 166.Zhou Y., Wang Y., Lin M., Wu D., Zhao M. LncRNA HOTAIR promotes proliferation and inhibits apoptosis by sponging miR-214-3p in HPV16 positive cervical cancer cells. Cancer Cell Int. 2021 Dec;21(1):1–5. doi: 10.1186/s12935-021-02103-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Cáceres-Durán M.Á., Ribeiro-dos-Santos Â, Vidal A.F. Roles and mechanisms of the long noncoding RNAs in cervical cancer. Int. J. Mol. Sci. 2020 Jan;21(24):9742. doi: 10.3390/ijms21249742. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Yang H.Y., Huang C.P., Cao M.M., Wang Y.F., Liu Y. Long non-coding RNA CRNDE may be associated with poor prognosis by promoting proliferation and inhibiting apoptosis of cervical cancer cells through targeting PI3K/AKT. Neoplasma. 2018 Sep 4;65(6):872–880. doi: 10.4149/neo_2018_171225N841. [DOI] [PubMed] [Google Scholar]
- 169.Huang X., Qian W., Ye X. Long noncoding RNAs in diffuse large B-cell lymphoma: current advances and perspectives. OncoTargets Ther. 2020;13:4295. doi: 10.2147/OTT.S253330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Oh E.J., Kim S.H., Yang W.I., Ko Y.H., Yoon S.O. Long non-coding RNA HOTAIR expression in diffuse large B-cell lymphoma: in relation to polycomb repressive complex pathway proteins and H3K27 trimethylation. J. Pathol. Translat. Med. 2016 Sep;50(5):369. doi: 10.4132/jptm.2016.06.06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Adler D.H. The impact of HAART on HPV-related cervical disease. Curr. HIV Res. 2010;8:493–497. doi: 10.2174/157016210793499240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.He H., Liu X., Liu Y., Zhang M., Lai Y., Hao Y., Wang Q., Shi D., Wang N., Luo X.G., Ma W. Human papillomavirus E6/E7 and long noncoding RNA TMPOP2 mutually upregulated gene expression in cervical cancer cells. J. Virol. 2019 Apr 3;93(8):e01808–e01818. doi: 10.1128/JVI.01808-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Sharma S., Mandal P., Sadhukhan T., Roy Chowdhury R., Ranjan Mondal N., Chakravarty B., Chatterjee T., Roy S., Sengupta S. Bridging links between long noncoding RNA HOTAIR and HPV oncoprotein E7 in cervical cancer pathogenesis. Sci. Rep. 2015 Jul 8;5(1):1–5. doi: 10.1038/srep11724. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Clemson C.M., Hutchinson J.N., Sara S.A., Ensminger A.W., Fox A.H., Chess A., Lawrence J.B. An architectural role for a nuclear noncoding RNA: NEAT1 RNA is essential for the structure of paraspeckles. Mol. Cell. 2009 Mar 27;33(6):717–726. doi: 10.1016/j.molcel.2009.01.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Mehta S., Campbell H., Drummond C.J., Li K., Murray K., Slatter T., Bourdon J.C., Braithwaite A.W. Adaptive homeostasis and the p53 isoform network. EMBO Rep. 2021 Dec 6;22(12) doi: 10.15252/embr.202153085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Guo H.M., Yang S.H., Zhao S.Z., Li L., Yan M.T., Fan M.C. LncRNA NEAT1 regulates cervical carcinoma proliferation and invasion by targeting AKT/PI3K. Eur. Rev. Med. Pharmacol. Sci. 2018 Jul 1;22(13):4090–4097. doi: 10.26355/eurrev_201807_15400. [DOI] [PubMed] [Google Scholar]
- 177.Naipauer J., García Solá M.E., Salyakina D., Rosario S., Williams S., Coso O., Abba M.C., Mesri E.A., Lacunza E. A non-coding RNA network involved in KSHV tumorigenesis. Front. Oncol. 2021 Jun 16;11:2217. doi: 10.3389/fonc.2021.687629. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Boliar S., Gludish D.W., Jambo K.C., Kamng’ona R., Mvaya L., Mwandumba H.C., Russell D.G. Inhibition of the lncRNA SAF drives activation of apoptotic effector caspases in HIV-1–infected human macrophages. Proc. Natl. Acad. Sci. USA. 2019 Apr 9;116(15):7431–7438. doi: 10.1073/pnas.1818662116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Zeng C., Liu S., Lu S., Yu X., Lai J., Wu Y., Chen S., Wang L., Yu Z., Luo G., Li Y. The c-Myc-regulated lncRNA NEAT1 and paraspeckles modulate imatinib-induced apoptosis in CML cells. Mol. Cancer. 2018 Dec;17(1):1–6. doi: 10.1186/s12943-018-0884-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Yiu G.K., Kaunisto A., Chin Y.R., Toker A. NFAT promotes carcinoma invasive migration through glypican-6. Biochem. J. 2011 Nov 15;440(1):157–166. doi: 10.1042/BJ20110530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Barnum KJ, O'Connell MJ. Cell cycle regulation by checkpoints. InCell Cycle Control 2014 (pp. 29-40). Humana Press, New York, NY. [DOI] [PMC free article] [PubMed]
- 182.Janku F., Yap T.A., Meric-Bernstam F. Targeting the PI3K pathway in cancer: are we making headway? Nat. Rev. Clin. Oncol. 2018 May;15(5):273–291. doi: 10.1038/nrclinonc.2018.28. [DOI] [PubMed] [Google Scholar]
- 183.Wu X.S., Wang X.A., Wu W.G., Hu Y.P., Li M.L., Ding Q., Weng H., Shu Y.J., Liu T.Y., Jiang L., Cao Y. MALAT1 promotes the proliferation and metastasis of gallbladder cancer cells by activating the ERK/MAPK pathway. Cancer Biol. Ther. 2014 Jun 1;15(6):806–814. doi: 10.4161/cbt.28584. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Wang C., Mao Z.P., Wang L., Wu G.H., Zhang F.H., Wang D.Y., Shi J.L. Long non-coding RNA MALAT1 promotes cholangiocarcinoma cell proliferation and invasion by activating PI3K/Akt pathway. Neoplasma. 2017 Jan 1;64(5):725–731. doi: 10.4149/neo_2017_510. [DOI] [PubMed] [Google Scholar]
- 185.Chen Y., Huang W., Sun W., Zheng B., Wang C., Luo Z., Wang J., Yan W. LncRNA MALAT1 promotes cancer metastasis in osteosarcoma via activation of the PI3K-Akt signaling pathway. Cell. Physiol. Biochem. 2018;51(3):1313–1326. doi: 10.1159/000495550. [DOI] [PubMed] [Google Scholar]
- 186.Xu S., Sui S., Zhang J., Bai N., Shi Q., Zhang G., Gao S., You Z., Zhan C., Liu F., Pang D. Downregulation of long noncoding RNA MALAT1 induces epithelial-to-mesenchymal transition via the PI3K-AKT pathway in breast cancer. Int. J. Clin. Exp. Pathol. 2015;8(5):4881. [PMC free article] [PubMed] [Google Scholar]
- 187.Xu H., Li J., Zhou Z.G. NEAT1 promotes cell proliferation in multiple myeloma by activating PI3K/AKT pathway. Eur. Rev. Med. Pharmacol. Sci. 2018 Oct 1;22(19):6403–6411. doi: 10.26355/eurrev_201810_16053. [DOI] [PubMed] [Google Scholar]
- 188.Liu X., Xu M., Li P., Zhang W., Zeng L.H., Yang Y., Yang G. Roles of lncRNAs in the transcription regulation of HIV-1. Biomed. J. 2022 Mar;29 doi: 10.1016/j.bj.2022.03.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Ao X., Jiang M., Zhou J., Liang H., Xia H., Chen G. lincRNA p21 inhibits the progression of non small cell lung cancer via targeting miR 17 5p. Oncol. Rep. 2019 Feb 1;41(2):789–800. doi: 10.3892/or.2018.6900. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Jin S., Yang X., Li J., Yang W., Ma H., Zhang Z. p53-targeted lincRNA-p21 acts as a tumor suppressor by inhibiting JAK2/STAT3 signaling pathways in head and neck squamous cell carcinoma. Mol. Cancer. 2019 Dec;18(1):1–8. doi: 10.1186/s12943-019-0993-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Wang X., Xu Y., Wang X., Jiang C., Han S., Dong K., Shen M., Xu D. Linc RNA‐p21 suppresses development of human prostate cancer through inhibition of PKM 2. Cell Prolif. 2017 Dec;50(6) doi: 10.1111/cpr.12395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Wang T., Liu J., Li S., Yuan Z., Huang X. Effects of knockout of lincRNA-p21 on the proliferation, migration and invasion ability of HepG2 liver cancer cells. Oncol. Lett. 2019 Jun 1;17(6):5103–5107. doi: 10.3892/ol.2019.10201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Chen Y., Wei G., Xia H., Yu H., Tang Q., Bi F. Down regulation of lincRNA-p21 contributes to gastric cancer development through Hippo-independent activation of YAP. Oncotarget. 2017 Sep 8;8(38) doi: 10.18632/oncotarget.19130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Zhang Y., Miao Y., Shang M., Liu M., Liu R., Pan E., Pu Y., Yin L. LincRNA-p21 leads to G1 arrest by p53 pathway in esophageal squamous cell carcinoma. Cancer Manag. Res. 2019;11:6201. doi: 10.2147/CMAR.S197557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Liu T., Zhang J., Chai Z., Wang G., Cui N., Zhou B. Ginkgo biloba extract EGb 761–induced upregulation of LincRNA-p21 inhibits colorectal cancer metastasis by associating with EZH2. Oncotarget. 2017 Oct 31;8(53) doi: 10.18632/oncotarget.21345. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 196.Li Y., Ye Y., Feng B., Qi Y. Long noncoding RNA lncARSR promotes doxorubicin resistance in hepatocellular carcinoma via modulating PTEN‐PI3K/Akt pathway. J. Cell. Biochem. 2017 Dec;118(12):4498–4507. doi: 10.1002/jcb.26107. [DOI] [PubMed] [Google Scholar]
- 197.Chi Y., Gong Z., Xin H., Wang Z., Liu Z. Long noncoding RNA lncARSR promotes nonalcoholic fatty liver disease and hepatocellular carcinoma by promoting YAP1 and activating the IRS2/AKT pathway. J. Transl. Med. 2020 Dec;18(1) doi: 10.1186/s12967-020-02225-y. 1-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Shen P., Cheng Y. Long noncoding RNA lncARSR confers resistance to Adriamycin and promotes osteosarcoma progression. Cell Death Dis. 2020 May 13;11(5):1–2. doi: 10.1038/s41419-020-2573-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Qu L.E., Ding J., Chen C., Wu Z.J., Liu B., Gao Y., Chen W., Liu F., Sun W., Li X.F., Wang X. Exosome-transmitted lncARSR promotes sunitinib resistance in renal cancer by acting as a competing endogenous RNA. Cancer Cell. 2016 May 9;29(5):653–668. doi: 10.1016/j.ccell.2016.03.004. [DOI] [PubMed] [Google Scholar]
- 200.Hu Y., Guo R., Wei J., Zhou Y., Ji W., Liu J., Zhi X., Zhang J. Effects of PI3K inhibitor NVP-BKM120 on overcoming drug resistance and eliminating cancer stem cells in human breast cancer cells. Cell Death Dis. 2015 Dec;6(12) doi: 10.1038/cddis.2015.363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Radomska-Leśniewska D.M., Bałan B.J., Skopiński P. Angiogenesis modulation by exogenous antioxidants. Central-European J. Immunol. 2017;42(4):370. doi: 10.5114/ceji.2017.72804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Potente M., Gerhardt H., Carmeliet P. Basic and therapeutic aspects of angiogenesis. Cell. 2011 Sep 16;146(6):873–887. doi: 10.1016/j.cell.2011.08.039. [DOI] [PubMed] [Google Scholar]
- 203.Ma Q., Reiter R.J., Chen Y. Role of melatonin in controlling angiogenesis under physiological and pathological conditions. Angiogenesis. 2020 May;23(2):91–104. doi: 10.1007/s10456-019-09689-7. [DOI] [PubMed] [Google Scholar]
- 204.Huang X.J., Xia Y., He G.F., Zheng L.L., Cai Y.P., Yin Y., Wu Q. MALAT1 promotes angiogenesis of breast cancer. Oncol. Rep. 2018 Nov 1;40(5):2683–2689. doi: 10.3892/or.2018.6705. [DOI] [PubMed] [Google Scholar]
- 205.Li Y., Wu Z., Yuan J., Sun L., Lin L., Huang N., Bin J., Liao Y., Liao W. Long non-coding RNA MALAT1 promotes gastric cancer tumorigenicity and metastasis by regulating vasculogenic mimicry and angiogenesis. Cancer Lett. 2017 Jun 1;395:31–44. doi: 10.1016/j.canlet.2017.02.035. [DOI] [PubMed] [Google Scholar]
- 206.Sun Z., Ou C., Liu J., Chen C., Zhou Q., Yang S., Li G., Wang G., Song J., Li Z., Zhang Z. YAP1-induced MALAT1 promotes epithelial–mesenchymal transition and angiogenesis by sponging miR-126-5p in colorectal cancer. Oncogene. 2019 Apr;38(14):2627–2644. doi: 10.1038/s41388-018-0628-y. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 207.Malakar P., Shilo A., Mogilevsky A., Stein I., Pikarsky E., Nevo Y., Benyamini H., Elgavish S., Zong X., Prasanth K.V., Karni R. Long noncoding RNA MALAT1 promotes hepatocellular carcinoma development by SRSF1 upregulation and mTOR ActivationMALAT1 is a proto-oncogene in HCC development. Cancer Res. 2017 Mar 1;77(5):1155–1167. doi: 10.1158/0008-5472.CAN-16-1508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Alfarouk K.O., Stock C.M., Taylor S., Walsh M., Muddathir A.K., Verduzco D., Bashir A.H., Mohammed O.Y., Elhassan G.O., Harguindey S., Reshkin S.J. Resistance to cancer chemotherapy: failure in drug response from ADME to P-gp. Cancer Cell Int. 2015 Dec;15(1):1–3. doi: 10.1186/s12935-015-0221-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.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 Feb;22(2):96–118. doi: 10.1038/s41580-020-00315-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Liu K., Gao L., Ma X., Huang J.J., Chen J., Zeng L., Ashby C.R., Zou C., Chen Z.S. Long non-coding RNAs regulate drug resistance in cancer. Mol. Cancer. 2020 Dec;19(1):1–3. doi: 10.1186/s12943-020-01162-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Yu G., Zhou H., Yao W., Meng L., Lang B. lncRNA TUG1 promotes cisplatin resistance by regulating CCND2 via epigenetically silencing miR-194-5p in bladder cancer. Mol. Ther. Nucleic Acids. 2019 Jun 7;16:257–271. doi: 10.1016/j.omtn.2019.02.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Wang X., Yang B., Ma B. The UCA1/miR-204/Sirt1 axis modulates docetaxel sensitivity of prostate cancer cells. Cancer Chemother. Pharmacol. 2016 Nov;78(5):1025–1031. doi: 10.1007/s00280-016-3158-8. [DOI] [PubMed] [Google Scholar]
- 213.Wei J., Gan Y., Peng D., Jiang X., Kitazawa R., Xiang Y., Dai Y., Tang Y., Yang J. Long non‐coding RNA H19 promotes TDRG1 expression and cisplatin resistance by sequestering miRNA‐106b‐5p in seminoma. Cancer Med. 2018 Dec;7(12):6247–6257. doi: 10.1002/cam4.1871. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Liu Y., Guo R., Qiao Y., Han L., Liu M. LncRNA NNT-AS1 contributes to the cisplatin resistance of cervical cancer through NNT-AS1/miR-186/HMGB1 axis. Cancer Cell Int. 2020 Dec;20(1):1–2. doi: 10.1186/s12935-020-01278-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Chen Q.N., Wei C.C., Wang Z.X., Sun M. Long non-coding RNAs in anti-cancer drug resistance. Oncotarget. 2017 Jan 3;8(1):1925. doi: 10.18632/oncotarget.12461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Bukowski K., Kciuk M., Kontek R. Mechanisms of multidrug resistance in cancer chemotherapy. Int. J. Mol. Sci. 2020 Jan;21(9):3233. doi: 10.3390/ijms21093233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Li Y., Shi B., Dong F., Zhu X., Liu B., Liu Y. Long non-coding RNA DLEU1 promotes cell proliferation, invasion, and confers cisplatin resistance in bladder cancer by regulating the miR-99b/HS3ST3B1 axis. Front. Genet. 2019 Mar 29;10:280. doi: 10.3389/fgene.2019.00280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218.Butova R., Vychytilova-Faltejskova P., Souckova A., Sevcikova S., Hajek R. Long non-coding RNAs in multiple myeloma. Non-coding RNA. 2019 Jan 24;5(1):13. doi: 10.3390/ncrna5010013. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Data sharing is not applicable to this article.

