Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Jan 1.
Published in final edited form as: Biochim Biophys Acta. 2015 Jul 2;1859(1):209–221. doi: 10.1016/j.bbagrm.2015.06.013

Long noncoding RNAs in Diseases of Aging

Jiyoung Kim 1, Kyoung Mi Kim 1, Ji Heon Noh 1, Je-Hyun Yoon 1, Kotb Abdelmohsen 1,*, Myriam Gorospe 1,*
PMCID: PMC4698248  NIHMSID: NIHMS709002  PMID: 26141605

Abstract

Aging is a process during which progressive deteriorating of cells, tissues, and organs over time lead to loss of function, disease, and death. Towards the goal of extending human health span, there is escalating interest in understanding the mechanisms that govern aging-associated pathologies. Adequate regulation of expression of coding and noncoding genes is critical for maintaining organism homeostasis and preventing disease processes. Long noncoding RNAs (lncRNAs) are increasingly recognized as key regulators of gene expression at all levels – transcriptional, post-transcriptional and post-translational. In this review, we discuss our emerging understanding of lncRNAs implicated in aging illnesses. We focus on diseases arising from age-driven impairment in energy metabolism (obesity, diabetes), the declining capacity to respond homeostatically to proliferative and damaging stimuli (cancer, immune dysfunction), and neurodegeneration. We identify the lncRNAs involved in these ailments and discuss the rising interest in lncRNAs as diagnostic and therapeutic targets to ameliorate age-associated pathologies and prolong health.

Keywords: senescence, age-associated disease, ribonucleoprotein complexes, transcriptional control, post-transcriptional gene regulation, differentiation, myocytes, mRNA translation, mRNA stability, transcriptome

1. INTRODUCTION

Aging is a complex biological process during which the function of cells, tissues, and organs declines progressively, leading to a reduction in normal activity and an increase in age-related diseases [1, 2]. These declines can be categorized in a variety of ways, but they are generally believed to arise from changes in tissue composition leading to imbalanced energy metabolism, homeostatic dysregulation, and neurodegeneration [3]. Aging phenotypes are driven by changes in gene expression programs. Although such programs are robustly regulated by classic regulators of gene expression, such as DNA- and RNA-binding proteins, there is rising recognition that a new class of potent gene regulatory factors, noncoding RNAs (lncRNAs), also directly and profoundly affects gene expression programs with aging.

1.1. LncRNAs

NcRNAs of all sizes have been known for several decades [4-8]. However, the advent of high-throughput methodologies has provided a comprehensive view of the vast collection of ncRNAs which are pervasively transcribed across more than 75% of genome [9, 10]. This large and heterogeneous class of transcripts contains many small RNAs, including small nucleolar RNAs (snoRNAs), microRNAs (miRNAs), small interfering RNAs (siRNAs), small nuclear RNAs (snRNAs), and Piwi-interacting RNA (piRNA), but the vast majority of ncRNAs are longer than 200 nt and are thus designated as long ncRNAs (lncRNAs) [11-13]. LncRNAs have emerged as potent regulators of gene expression at different levels, including chromatin remodeling, transcriptional control, mRNA stability, mRNA translation, microRNA function, and protein metabolism [12, 14-16]. Through their influence on gene expression, lncRNAs are increasingly recognized as players with essential roles in molecular and cellular processes such as proliferation, apoptosis, differentiation, and senescence; in this regard, the impact of lncRNAs on the cellular and molecular basis of aging was recently reviewed [17]. LncRNAs also underlie important pathologic processes that increase with aging, including metabolic imbalances, neurodegeneration, cardiovascular disease, and cancer [18-23].

1.2. LncRNA classification and function

Unlike protein-coding RNAs and small regulatory RNAs, lncRNAs show little sequence or motif conservation among species [12, 24, 25]. Instead, the genetic position relative to protein-coding sequences and their functional conservation are considered to be important features to understand lncRNA biology and function [11, 12, 26, 27]. There is no universally adopted method to classify lncRNAs, but based on their transcriptional position and direction, lncRNAs can be categorized as antisense, intronic, intergenic, and divergent lncRNAs [28]. Antisense lncRNAs are synthesized in reverse direction relative to the protein-coding mRNA, resulting in overlaps of varying lengths between the two transcripts, and intronic lncRNA transcripts are transcribed from introns of a parental gene [28, 29]. Long intergenic lncRNAs (lincRNAs) are those with annotated positions localized between two protein-coding genes and are at least 5 kb away from protein-coding genes [11, 12, 28, 30, 31]. Finally, divergent lncRNAs are short transcripts usually synthesized from the promoter region of a protein-coding mRNA, but in the opposite direction [32].

Despite the fast progress in lncRNA transcript identification and annotation [33, 34], elucidating their molecular and biological roles has been challenging and, thus far, they have proven to vary widely [35]. Nonetheless, some functions are beginning to emerge. Nuclear lncRNAs have been extensively implicated in chromatin remodeling, histone modification, X-chromosome inactivation, transcription, splicing, imprinting, and nuclear organization [36-39]. These nuclear functions are achieved by the lncRNA-mediated recruitment of chromatin modifiers to specific chromatin regions, sometimes guided by the lncRNA to a specific DNA site, with the lncRNA serving as a scaffold for the assembly of multiple chromatin modifiers [36-39]. LncRNAs can also be directly implicated in the process of pre-mRNA transcription, as shown for lncRNAs transcribed from enhancer regions (enhancer lncRNA [eRNA]), promoter regions (promoter-associated lncRNA [paRNA]), and transcriptional start site regions (transcription start site-associated lnRNA [TSSaRNA]) [40]. LncRNAs can also bind to nascent pre-mRNAs and influence their splicing and processing [41]. Cytoplasmic functions of lncRNAs include many diverse post-transcriptional steps [15]. LncRNAs can interact with RNA-binding proteins (RBPs), microRNAs and mRNAs to function as decoys or sponges, influence mRNA turnover rate, subcytoplasmic mRNA localization, and translation of target mRNAs [15, 42, 43], affect the stability of certain proteins by assembling components of ubiquitin-mediated proteolysis [16, 44, 45], and affect protein function by serving as scaffolds to assemble multiprotein complexes [41]. These examples illustrate the rich and versatile mechanisms whereby lncRNAs affect gene expression virtually at all levels –from early changes in chromatin architecture to the proteolysis of encoded proteins. In this review we focus on lncRNAs implicated in age-related diseases, their mechanism of actions and possible future interventions.

2. DISEASES OF AGING ASSOCIATED WITH DECLINING ENERGY METABOLISM

With advancing age, there is an overall decline in the body’s ability to store energy, mobilize energy stores, sense changes in energy availability, and utilize energy. The age-associated increases in adiposity, reduced pancreatic function, and loss of muscle mass lead to major pathologic conditions seen in the elderly. In this section, we discuss lncRNAs implicated in the pathogenesis of diabetes and obesity [46, 47] as well as conditions arising from mitochondria dysfunction and muscle loss (Table 1).

Table 1.

LncRNAs in aging-associated diabetes, obesity, mitochondrial disease, and muscle dysfunction

LncRNA Processes Targets Relevance to Aging, Other Implications Refs.

Obesity

lnc-RAPs Adipogenesis (various) Many lnc-RAPs induced by PPARγ (a TF
reduced with age) and C/EBPα
[55, 56]

lnc-RAP-1,
Firre
Adipogenesis genes Slc25a12, Eef1a1,
Atf4, Ppp1r10 (on
chromosomes 2, 9, 15,
17), and hnRNP U
Helps organize nuclear architecture across
chromosomes
[57]

naPINK1 Obesity, diabetes,
neurodegeneration,
muscle activity
PINK1 mRNA, encoding
a mitochondrial kinase
In inactive muscle, low PINK1 mRNA and
high naPINK1; after endurance training,
high PINK1 mRNA and low naPINK1
[58-60]

H19 Fat deposition and
fat metabolism
Imprinting of gene
cluster that includes
IGF2
High IGF2 and H19 due to loss of
imprinting during aging linked to fat
deposition and cancer
[61, 62]

linc-DMRT2 Adipogenesis Undetermined Low in fat tissue of obese humans [63]

linc-TP53I13 Adipogenesis Undetermined Low in fat tissue of obese humans [63]


Diabetes

KCNQ1OT1 Diabetes Undetermined High in pancreatic islets of diabetic
individuals
[74]

HI-LNC45 Diabetes Undetermined Low in pancreatic islets of diabetic
individuals
[74]

HI-LNC78,
HI-LNC80
Sensing glucose
levels
Undetermined Elevated by high glucose [74]

HI-LNC25 β cell function GLIS3 mRNA β cell function in adults [74, 75]

ANRIL Proliferation
capacity of β
cells(?)
PRC1, PRC2, p16INK4A
and p15INK4B
ANRIL identified as hotspot of T2D
mutations, suppresses (via PRC1,2)
transcription of tumor suppressors and
senescence inducers p16INK4A and p15INK4B
[76-78]

Mitochondria

LNCND5,
LNCND6,
LNCCYTB
Mitochondrial gene
regulation
Undetermined Encoded by mitochondrial genome [83-85]

RMRP Mitochondria
functionl
Undetermined Encoded by nuclear genome [86]

LIPCAR Myocardial
infarction
Undetermined Encoded by mitochondrial genome, levels
in blood have prognostic value
[88-91]

Muscle physiology

H19 Skeletal muscle
regeneration(?)
Suppresses BMP
pathway, TFs SMAD1,5
replication factor CDC6
H19 levels selectively upregulated in old
muscle, hosts miR-675, which elicits some
of H19 actions
[92]

linc-MD1 Muscle regeneration
and sarcopenia
Decoys miR-133 and
miR-135
Decoys miR-133, alleviating repression of
HuR/ELAVL1
[96-99]

SIRT1 AS Myogenesis Myoblast Sirt1 mRNA SIRT1 helps maintain a robust myogenic
program, SIRT1 AS prevents this
[100-102]

MALAT1 Myoblast
proliferation
SRSF1, SRSF2,
hnRNP C, TP53
MALAT1 levels reduced by Myostatin,
showing deficiency with age
[103-105]

Yam-1 Repression of
myogenesis
Wnt7b
Yam-1 harbors miR-715
TF YY1 (implicated in muscle wasting,
cancer, diabetes, chronic heart failure)
promotes Yam-1 expression
[107-108]

LncRNAs involved in aging-related decline in energy metabolism (column 1), the ageassociated diseases and disease processes in which they are implicated (column 2), the direct molecular targets of the lncRNAs (column 3), and the relevance of the lncRNAs to energy metabolism and aging processes (column 4) are listed.

2.1. LncRNAs affecting fat metabolism

The distribution and function of the adipose tissue change dramatically throughout life. Besides its immune and endocrine actions and its roles in thermoregulation, mechanical protection, and tissue regeneration, the adipose tissue is a major source of energy. The size of the adipose tissue increases during the adult years and has mainly a subcutaneous distribution. It declines in old age and progressively adopts a visceral distribution with accumulation in the liver, muscle, and bone marrow [48-51]. This extensive remodeling is accomplished by developmental processes governed by transcriptional regulatory factors whose expression and function are impaired with aging: the CCAAT/enhancer binding protein (C/EBP)β, peroxisome proliferator-activated receptor (PPAR)γ, and C/EBPα [52]. Posttranscriptionally, adipogenesis is also regulated by RBPs such as Sam68 [53] and by numerous miRNAs, reviewed in [54]. In this section we discuss lncRNAs involved in controlling fat tissue metabolism and adipogenesis.

Sun and colleagues [55] first identified lnc-RAPs as lncRNAs that showed regulated expression during adipogenesis. Among ~175 lnc-RAPs upregulated during differentiation, the pro-adipogenic transcription factors PPARγ and CEBPα were found to bind to the promoter regions of 23 and 34 of them, respectively. Silencing lnc-RAPs 1 through 9 impaired adipogenesis, as determined by a lower accumulation of lipid and reduced expression of adipogenic markers including PPARγ [55]. The direct impact of individual lnc-RAPs on aging of adipose tissue warrant direct study, particularly as PPARγ levels decline in advancing age [56].

LncRNA Firre (functional intergenic repeating RNA element), also known as lnc-RAP-1 was shown to interact in trans with chromosomes 2, 9, 15, and 17 in regions overlapping with genes Slc25a12, Eef1a1, Atf4, and Ppp1r10, all encoding adipogenic proteins. Firre also interacts with hnRNP (heterogeneous nuclear ribonucleoprotein) U in mouse adipose tissue and in adipocyte lysates. Downregulation of hnRNP U disrupted Firre localization on the inter-chromosomal loci, suggesting that Firre helps organize the nuclear architecture across chromosomes [57]. The potential impact of Firre on age-related adipose tissue function remains to be investigated.

naPINK1 is a natural antisense lncRNA to PINK1, a gene that encodes a PTEN (phosphatase and tensin homologue deleted on chromosome 10)-induced putative kinase 1. A mitochondrial kinase, PINK1 protects cells against oxidative stress-induced apoptosis and displayed aberrant function in age-related diseases like diabetes and neurodegeneration [58, 59]. Gene expression analysis of skeletal muscles indicated that PINK1 mRNA was expressed with a pattern opposite to that of naPINK1. In healthy volunteers, inactive muscle expressed low PINK1 mRNA and high naPINK1, while endurance training elevated PINK1 mRNA and lowered naPINK1 expression levels [60]. Since the PINK1 locus, linked to neurodegenerative disease, is dysregulated during inactivity, obesity, and type 2 diabetes [60], disruption of naPINK1 expression may impact upon these common age-related disease processes.

The lncRNA H19 controls imprinting of a conserved cluster of genes that includes IGF2 (insulin-like growth factor 2), involved in fat metabolism and fat deposition. In adult mice, low Igf2 abundance was associated with increased fat deposition and occasional obesity, while loss of imprinting of the Igf2-H19 locus during aging was found to enhance the expression of IGF2 and H19 [61]. This regulatory paradigm in old prostate tissue was linked to cancer [62], but the age-related alteration may be associated broadly with fat deposition and metabolism.

Other lncRNAs related to adipogenesis have also been identified. For instance, linc-DMRT2 and linc-TP53I13 are regulated by lipopolysaccharide and downregulated in adipose tissue of obese humans [63]. Future studies will establish the function of lncRNAs in age-related diseases derived from loss of homeostasis of the adipose tissue.

2.2. LncRNAs affecting diabetes

Type 2 diabetes (T2D) increases in the elderly, accelerating the body’s impairment in sensing energy levels, storing energy, and mobilizing energy out of storage [64-66]. A correlation between T2D and aging of pancreatic islet cells has been proposed to account at least in part for the rise in T2D in the elderly [67-69]. T2D is characterized by insulin resistance and low levels of circulating insulin, which reduces the availability of energy from glucose in peripheral tissues and elevates circulating glucose levels, causing other important problems like stroke, atherosclerosis, and heart attack [70]. Numerous lncRNAs were identified in human β cells, and many of them have been dynamically integrated into diabetes-regulatory programs [71]. Transcriptome-wide sequencing of pancreatic β-cells and islets of Langerhans identified several lncRNAs that might be involved in the development or progression of age-related diabetes [72, 73]. However, their exact function in diabetes remains to be investigated. In this section, we explore lncRNAs that may have an impact on diabetes.

Compared with the levels in control individuals, diabetic individuals displayed higher levels of lncRNA KCNQ1OT1 and lower levels of lncRNA HI-LNC45 in pancreatic islets. In addition, the levels of lncRNAs HI-LNC78 and HI-LNC80 in islet cells increased in the presence of high glucose, indicating that these lncRNAs may be involved in sensing blood glucose levels [74]. The lncRNA HI-LNC25, also selectively expressed in islets and β cells, enhanced the levels of GLIS3 mRNA, which encodes the GLIS3 transcription factor [74]. GLIS3 is required for normal β-cell function in adults [75]. Future studies are needed to fully understand the roles of these lncRNAs in diabetes.

Also known as CDKN2B-AS1, the lncRNA ANRIL interacts with PRC (polycomb repressive complex)1 and PRC2 to suppress transcription of the tumor suppressors and senescence inducers p16INK4A and p15INK4B [76-79]. Accordingly, silencing ANRIL in fibroblasts inhibited proliferation and triggered senescence [78]. Genome-wide association studies (GWAS) identified ANRIL as a hotspot of mutations in T2D [76, 80], suggesting that ANRIL may also contribute to regulating β cell proliferation capacity with age.

2.3. LncRNAs affecting diseases of mitochondrial dysfunction

The gradual age-driven decline in mitochondrial function causes tissue damage, reduces lifespan, and triggers a number of diseases such as myopathies, neuropathies, and diabetes [81, 82]. Several mitochondrial lncRNAs have been identified, such as LNCND5, LNCND6, and LNCCYTB, and they appear to be involved in mitochondrial gene regulation [83-85]. Additionally, some nuclear-encoded lncRNAs, such as RMRP, can be transported from the nucleus to the mitochondria and regulate mitochondrial gene expression and other mitochondrial functions [86]. Thus, mitochondrial lncRNAs involved in maintaining healthy and functional mitochondria may provide important information for aging-associated declines and pathologies.

Myocardial infarction and congestive heart failure are far more common in the elderly [87]. In a recent study, global transcriptome analysis of plasma RNA indicated that the mitochondrial lncRNA LIPCAR is downregulated early after myocardial infarction (MI), is upregulated during later stages following MI, and its levels in circulation may predict survival in patients with heart failure [88, 89]. Future analysis will ascertain the relevance and function of LIPCAR in age-related cardiovascular pathologies, as well as the decline in mitochondrial function with age [90] in this system.

2.4. LncRNAs affecting age-associated muscle pathology

Muscle dysfunction is commonly observed in the elderly, leading to the aging-associated frailty and sarcopenia [89]. Numerous lncRNAs associated with age-related muscle disorders are emerging.

The aforementioned imprinted lncRNA H19 is implicated in cellular differentiation [92]. H19 is expressed from the maternal allele and is highly abundant in embryonic tissues. Unlike most tissues, which experience strong repression of H19 levels after birth and throughout old age, skeletal muscle maintains high H19 levels, suggesting an important role of H19 in skeletal muscle function [92]. H19 and its encoded miRNAs, miR-675-3p and miR-675-5p, are upregulated during myoblast differentiation in vitro and muscle regeneration in vivo. H19 promotes muscle differentiation in mouse and human primary muscle cells and in cultured C2C12 mouse myoblasts. In addition, miR-675-3p and miR-675-5p mediate H19 function in muscle differentiation and regeneration by negatively regulating the bone morphogenetic protein (BMP) pathway, the transcription factors SMAD1 and SMAD5, and the DNA replication initiation factor CDC6 [92]. Similarly, H19 can bind let-7 and limit its availability, acting as a molecular sponge; the decreased levels of H19 levels in muscle from T2D individuals and in response to acute hyperinsulinemia increases let-7 availability in muscle cells [93, 94]. Thus, H19 may have an essential role in skeletal muscle regeneration, a process that is impaired in the elderly.

The muscle-specific linc-MD1 displays a decoy activity for miR-133, limiting its repressive impact on the Elavl1 mRNA (encoding the RBP HuR) in mouse myoblasts [95]. A feed-forward regulatory mechanism was suggested, whereby HuR enhanced linc-MD1 sponge activity by facilitating its recruitment of miR-133 and miR-135 and thereby modulated differentiation [96]. HuR is expressed at low levels in differentiated muscle and plays a direct role in muscle wasting and sarcopenia [97-99]. Thus, linc-MD1 functions during the decline in muscle regeneration via HuR.

SIRT1 AS is a natural antisense lncRNA that was recently found to play a role in myogenesis. The levels of Sirt1 mRNA and Sirt1 AS lncRNA decrease gradually during C2C12 myogenic differentiation. Overexpression of Sirt1 AS enhanced the levels of the NAD-dependent deacetylase SIRT1 (sirtuin-1). Interestingly, downregulation of SIRT1 by miR-34a was opposed by Sirtl AS in C2C12 cells [100]. The fact that SIRT1 helps prevent senescence and aging [101], specifically maintaining a robust myogenic program [102], connects Sirt1 AS with muscle aging.

The levels of MALAT1 (metastasis-associated lung adenocarcinoma transcript 1) rise during differentiation of mouse and human muscle cells, and MALAT1 silencing suppressed the proliferation of myoblasts and endothelial cells [103, 104]. Malat1 levels declined significantly following treatment of mouse gastrocnemius muscle with recombinant myostatin [103]. Given the impact of myostatin deficiency on muscle performance with age [105, 106], these findings suggest that MALAT1 may be a downstream target of myostatin and a regulator of myogenesis influencing muscle aging.

The transcription factor YY1 (YIN YANG 1) was implicated in diseases that involve muscle weakness and wasting, including cancer, diabetes and chronic heart failure [107]. Genome-wide mapping of the binding sites of transcription factor YY1 in C2C12 myoblasts revealed a large YY1-regulated lincRNA locus that included the muscle-associated lincRNA Yam-1. YY1 repressed myogenesis at least in part by promoting the expression of Yam-1, which harbored the microRNA miR-715. Increasing Yam-1 elevated miR-715 abundance in myoblasts, which in turn repressed production of a key target, the myogenic factor WNT7b [108]. By contrast, the lncRNA Dum [developmental pluripotency-associated 2 (DPPA2), upstream binding muscle lncRNA] activates myogenesis in C2C12 myoblasts [109]. Following transcriptional activation by the myogenic transcription factor MYOD, Dum interacts with DNA methyltransferases leading to the silencing of the neighboring gene DPPA2, which encodes a repressor of myogenesis [109].

3. DISEASES OF AGING ARISING FROM LOSS OF HOMEOSTASIS

The ability of the body to restore functions and internal homeostasis declines with age, leading to the development of cancer and other diseases associated with impaired ability to respond to cell and tissue damage [110-112]. In this section, we review examples of lncRNAs associated with cancer and the stress and immune responses (Table 2).

Table 2.

LncRNAs in aging-associated cancer, stress susceptibility, and immune dysfunction

LncRNA Processes Targets Relevance to Aging, Other Implications Refs.

Cancer

H19 Cancer, apoptosis,
proliferation, aging
IGF2, many proliferation
genes
Igf2/H19 imprinting lost with aging, H19
and its host, miR-675, elevated with aging
and cancer; upregulated by MYC
[120-126]

MEG3 Growth inhibition MDM2 MEG3 overexpression lowers cancer cell
growth, MEG3 silencing promotes it
[127-132]

MALAT1 Cancer, senescence TP53, SRSF1, SRSF2,
hnRNP C
SP factors relevant to aging, senescence;
High MALAT1 in lung and liver cancers
[133-138]

ANRIL Cancer, senescence p16INK4A and p15INK4B,
PRC1, PRC2
Represses p16INK4A and p15INK4B , p21,
KLF2; pro-tumorigenic in prostate and
liver by silencing miR-449a and miR-99a
[140-144]

HOTAIR Cancer, senescence ubiquitin/proteolysis
factors, PRC2
Prognostic in colorectal, cervical and
endometrial carcinomas
[16,146-
149]

XIST Cancer, senescence X chromosome Prognostic in ovarian cancer chemotherapy [151-153]

BCYRN1 Cancer,
neurodegeneration
Undetermined Decreased in brain of old individuals,
highly expressed in numerous cancers
[154-156]

GAS5 Growth inhibition Glucocorticoid Receptor Decreased in breast and liver cancers [157, 158]

MIR31HG Senescence miR-31 targets Harbors miR-31 (down in ovarian cancer
and leukemia)
[159-164]

7SL Prevents autophagy
and senescence
Binds to TP53 mRNA in
competition with HuR
Protumorigenic by lowering p53 [17, 43,
155]

HULC Tumorigenesis Liver and colon cancer [166, 167]

lncRNA-LET Tumor suppression Binds, destabilizes NF90 Downregulated by hypoxia and HIF-1α;
modulates NF-90-governed SASP
[44, 168]

PCGEM1 Anti-apoptosis Undetermined (indirectly
TP53, p21, others)
Promotes prostate cancer cell survival after
chemotherapy
[169, 170]

PVT1 Anti-apoptosis,
growth inhibition
Undetermined Upregulated by MYC; prognostic of
response to chemotherapy
[171, 172]


Stress Response

lincRNA-p21 Response to DNA
damage, hypoxia
CTNNB mRNA, JUNB
mRNA
Induced by TP53 and hypoxia, implicated
in senescence and growth arrest
[176, 179]

PINT DNA damage,
tumor suppression
SUZ12, epigenetic
silencing by PRC2
Induced by TP53, reduces proliferation,
low in colon cancer
[180]

PANDA Cell proliferation,
growth inhibition
PRC1, PRC2 Interacts with PRC1,2 in dividing cells,
represses p21 and growth inhibitory genes;
released from PRC1,2 in senescent cells
[181]

lncRNA-JADE DNA damage BRCA1 Upregulated via ATM, activates JADE1
transcriptionally
[183, 184]

lncRNA-LET Hypoxic stress Binds, destabilizes NF90 Downregulated by hypoxia and HIF-1α;
modulates NF-90-governed SASP
[44]

lncRNA-ROR DNA damage hnRNP I Represses TP53 translation [185]

LSINCTs DNA damage Undetermined Highly expressed in lung and breast cancer [186]

Immune Response

lincRNA-Cox2 Immune response hnRNP A/B, hnRNP
A2/B1
Upregulated by LPS and Pam3CSK4 in
macrophages, linked to NF-κB function
[190, 191]

Lethe Induced by TNFα Undetermined Pseudogene-encoded lncRNA transcribed
by NFκB, represses NFκB target genes
[193]

THRIL Repressed by TNFα hnRNP L Induced TNFα transcriptionally [194]

NEAT1 Viral infection SFPQ Induces IL8, linked to cognitive loss [195-197]

lnc-DC Immune response STAT3 Implicated in tumorigenesis, aging [198-200]

NeST Viral infection Undetermined Inflammation, neurological disease [201-204]

LncRNAs involved in aging-related decline in body homeostasis (column 1), the age-associated disease processes in which they are implicated (column 2), the direct molecular targets of the lncRNAs (column 3), and the relevance of the lncRNAs to homeostatic processes (column 4) are listed.

3.1. LncRNAs affecting cancer

LncRNAs have been implicated in carcinogenesis, which rises with advancing age. More than 70% of cancer cases are expected to be diagnosed in people aged over 65 years old by 2030 [113]. Here, we review the involvement of lncRNAs in cancer.

As mentioned earlier, H19 plays roles in proliferation, cancer cell growth, apoptosis, and aging, and is regulated via imprinting of the Igf2/H19 locus [114-118]. Loss of imprinting during aging of prostate tissues enhanced the expression of H19 and IGF2, a factor linked to aging and cancer [62, 119]. Expression of H19 is elevated in several human malignancies, including bladder, breast and colon cancers. H19 is highly expressed in low-grade bladder carcinoma patients, suggesting that H19 can be used as a marker for early recurrence in bladder carcinoma and metastasis [120, 121]. Expression of H19 and the inhibitor of DNA binding/differentiation 2 (ID2) correlated positively in bladder cancer, and H19 overexpression increased bladder cancer growth at least in part by elevating ID2 expression levels [122]. In breast cancer, the oncoprotein MYC, which is involved in cancer cell senescence, binds to and facilitates histone acetylation and transcriptional initiation of the H19 promoter [123, 124]. Overexpression of H19 enhanced breast cancer cell tumorigenesis and subcutaneous injection of H19-overexpressing breast cancer cells into immunodeficient (scid) mice showed that H19 overexpression promotes tumor formation and progression [125]. Both H19 and the microRNA it harbors, miR-675, are upregulated in human colon cancer cell lines and primary human colorectal cancer. miR-675 lowers expression of the tumor suppressor pRB (retinoblastoma protein) and inhibiting miR-675 reduced colon cancer cell growth and colony formation [126]. These findings suggest that H19 enhances tumorigenesis at least in part via miR-675.

The lncRNA MEG3 lowers cell proliferation by reducing the abundance of MDM2, an inhibitor of the tumor suppressor TP53 [127]. MEG3 is highly expressed in normal liver, but is weakly expressed in liver cancer and pituitary cancer [128-130]. Ectopic expression of MEG3 suppressed the growth of cancer cell lines HeLa, MCF-7, and H4, and hepatocellular carcinoma (HCC) [131, 132] and downregulation of MEG3 in bladder cancer increased cell proliferation and inhibited apoptosis [130], supporting a tumor suppressor function for MEG3.

Human MALAT1 is downregulated in senescent cells, and its silencing in young cells triggered cell senescence by enhancing TP53 expression [133, 134]. The oncogenic influence of MALAT1 was associated with its ability to modulate serine/arginine (SR) splicing factors, which are closely associated with senescence and aging [135, 136]. The notion that altered MALAT1 levels may impact upon tumorigenesis is supported by its high expression levels in several cancers, including lung and liver carcinoma [137, 138].

ANRIL promotes cell cycle arrest and inhibits senescence by repressing transcription of the tumor suppressor gene p15 (INK4B) [77, 78, 139]. GWAS analysis identified ANRIL as a risk locus for several cancers including breast carcinoma [76, 80]. Indeed, ANRIL was upregulated in prostate cancer cells, promoted non-small cell lung cancer cell proliferation by lowering expression of KLF2 and p21, and was proposed to be a marker of poor prognosis in gastric cancer [140-142]. Interestingly, ANRIL promoted tumor growth by preventing the action of miR-99a, a microRNA that inhibits HCC cell proliferation, and miR-449a, which induces prostate cancer cell cycle arrest and senescence [143, 144].

The Hox antisense intergenic RNA (HOTAIR) [143] regulates gene expression via polycomb-dependent chromatin modification and ubiquitin-mediated proteolysis [16, 146], and has been implicated in cell proliferation, senescence, and cancer [147]. Its oncogenic function is largely mediated by the recruitment of chromatin modifiers such as PRC2 to alter histone H3 trimethylation at lysine 27 (H3K27me3) and modulate gene transcription. HOTAIR levels are associated with poor prognosis in colorectal, cervical and endometrial carcinomas [146, 148, 149]. It is highly expressed in primary breast tumors and metastases and appears to serve as a predictor of metastasis and death [150].

The lncRNA XIST (X inactive-specific transcript) regulates dosage compensation that balances X-linked gene expression between males and females [151]. XIST is downregulated in senescent cells and upregulated in breast cancer, where the numbers of XIST-covered domains in chromosomes correlates with genetic abnormalities [152]. XIST levels correlate with Taxol sensitivity, thus possibly serving as a marker for chemotherapeutic responses in ovarian cancer [153].

BCYRN1 (brain cytoplasmic RNA 1, BC200) is a 200-nt-long ncRNA originally identified as a neuronal RNA. BCYRN1 levels decrease by >60% in cortical areas between the ages of 49 and 86, but is upregulated in AD brains compared to age-matched normal brains, as mentioned below (section 4) [154]. BCYRN1 is highly expressed in cancers of the breast, cervix, esophagus, lung, ovary, parotid, and tongue, but not in the corresponding normal tissues [155, 156]. These data suggest that altered expression of BCYRN1 is associated with both age-associated cancer and neurodegeneration.

Another lncRNA associated with both cancer and neurodegeneration, the growth arrest-specific 5 (GAS5), suppressed cell growth by acting as decoy for the transcription factor glucocorticoid receptor (GR), and thereby inhibited gene expression and induced apoptosis [157]. GAS5 was found to be downregulated in human HCC and breast cancer [158]. These data suggest that low GAS5 expression levels may promote tumorigenesis.

MIR31HG (LOC554202) harbors miR-31, a microRNA upregulated in senescent HUVECs (human umbilical vein endothelial cells) [159, 160]. miR-31 is downregulated in various cancers, including ovarian carcinoma and chronic myeloid leukemia cells, wherein miR-31 modulated cell proliferation and survival [161-164]. These findings suggest that MIR31HG and miR-31 are expressed with similar patterns [159], and its upregulation in senescent cells might be required to suppress tumorigenesis. It is not clear if MIR31HG functions in cancer independently of miR-31.

The lncRNA 7SL is highly expressed in several cancers [155]. Although its impact on aging has not been studied directly, 7SL silencing promoted autophagy and senescence, both hallmarks of aging and growth suppression [17]. 7SL competes with HuR for binding to TP53 mRNA and thus suppresses TP53 translation [43], supporting the notion that 7SL can promote tumorigenesis by reducing the levels of a major tumor suppressor protein.

Numerous additional lncRNAs associated with tumorigenesis have been reported (reviewed in [165]). For instance, HULC is highly expressed in hepatocytes, HCC, and colorectal carcinomas that metastasize in the liver and may function in liver cancer by sponging miR-372 [166, 167]. Hypoxia-induced histone deacetylation lowered the production of lncRNA-LET (‘low expression in tumor’) in several cancers including carcinomas of the liver, lung, and colon. Ectopic expression of lncRNA-LET inhibited metastases in a mouse xenograft model, while its depletion increased invasion [44]. Lowering lncRNA-LET was also associated with the stabilization of the nuclear factor 90 protein (NF90), which is required for hypoxia-induced cancer cell invasion and for the senescence-associated secretory phenotype (SASP) [168]. The lncRNA PCGEM1 (prostate cancer gene expression marker 1) was upregulated in prostate cancer and inhibited doxorubicin-induced apoptosis in LNCaP cells by reducing the levels of TP53, p21, cleaved caspase 7, and cleaved PARP [169, 170], suggesting that PCGEM1 may regulate cellular senescence. Like H19, the oncogenic lncRNA PVT1 is upregulated by MYC and is highly expressed in transformed cells [171]. Inhibition of PVT1 or MYC expression inhibited proliferation and induced apoptosis of breast and ovarian cancer cell lines. In addition, the enhancement in PVT1 levels was associated with reduced survival in patients treated with apoptotic agents [172]. The direct involvement of these lncRNAs in age-associated processes is unknown.

3.2. lncRNAs and the declining stress response

Environmental stress can cause an accumulation of cellular damage and disruption of cell functions leading to an acceleration of organismal aging. Thus, adequate responses to stresses such as DNA damage and unfavorable oxygen concentration are crucial for enhancing cell survival, preventing age-related disorders, and increasing lifespan [173, 174]. The levels of many lncRNAs change following exposure to stress agents, in turn affecting various cellular responses. In this section, we will review the regulatory function of lncRNAs in stress responses relevant to the aging process.

A number of TP53-inducible lncRNAs were characterized by the Rinn laboratory [175]. Among them, lincRNA-p21 mediated TP53-dependent transcriptional repression, as depletion of lincRNA-p21 inhibited apoptosis and cell cycle arrest after doxorubicin treatment, while lincRNA-p21 overexpression promoted spontaneous apoptosis in the absence of stress. LincRNA-p21 elicited these effects by interacting with the RBP hnRNP K to repress TP53-target gene transcription [176]. Depletion of lincRNA-p21 inhibited the production of lactate and glucose by hypoxia and reduced the levels of mRNAs encoding LDHA (lactate dehydrogenase A) and GLUT1 (glucose transporter 1). LincRNA-p21 expression levels were induced by hypoxia and HIF-1α (hypoxia-inducible factor 1α) in many human cancer cell lines; reciprocally, silencing lincRNA-p21 destabilized HIF-1α protein levels by increasing its ubiquitination and degradation [45]. In addition, lncRNA-p21 interacts with and suppresses the translation of CTNNB and JUNB mRNAs [177] (encoding the proliferative, senescence-inhibitory factor β-catenin [178] and the broad modulator proliferation JUNB [179], respectively) by recruiting the translation repressors RCK and FMRP [177]. Further roles of stress-inducible lncRNA-p21 in aging pathologies remain to be investigated.

Increased expression of lncRNA PINT, another TP53-inducible lncRNA also named lincRNA-Mkln1, lowers caspase-3 and caspase-7 levels, while silencing PINT induces their abundance. PINT interacts with SUZ12, a component of the PRC2 complex involved in histone modification and transcriptional repression, thereby contributing to the repression of certain genes. PINT is upregulated in human colorectal cancer and is required for proliferation of the human cancer cell line HCT116, linking this stress-responsive lncRNA to tumor-associated gene regulation [180].

DNA damage also induces PANDA (p21-associated ncRNA DNA damage-activated) in a TP53-dependent manner [181]. In proliferating cells, PANDA recruits PRC1 and PRC2 complexes to repress transcription of pro-senescence PRC target genes, such as those that encode CDKN1A (p21) and PANDA itself. In senescent cells, PRC1 and PRC2 complexes dissociate and the levels of PANDA and CDKN1A increase; under these conditions, PANDA blocks proliferation by associating with NF-YA, while CDKN1A solidifies growth arrest by activating pRB. Accordingly, depletion of the lncRNA PANDA leads to exit from senescence [182].

Treatment with the radiomimetic drug and inducer of DNA damage neocarzinostatin (NCS) upregulated many lncRNAs, including lncRNA-JADE (located next to JADE1), in an ATM-dependent manner. LncRNA-JADE depletion inhibits JADE1 induction by NCS and sensitizes cells from NCS-induced apoptosis, while lncRNA-JADE overexpression promotes JADE1 expression and inhibits apoptosis. In addition, LncRNA-JADE interacts with the transcription co-activator BRCA1, a major player in cancer, senescence and aging [183, 184], supporting the notion that cancer cells may achieve resistance to apoptosis by modulating stress and ATM-dependent lncRNA expression.

LncRNA-LET is downregulated in cancers, as mentioned above, via transcriptional repression during hypoxia. Interaction of lncRNA-LET with the RBP NF90 promoted NF90 ubiquitination and degradation [44] and thus the hypoxia-lowered lncRNA-LET caused NF90 upregulation. Interestingly, overexpression of lncRNA-LET or depletion of NF90 suppressed HIF-1α mRNA induction following hypoxia [44].

Other lncRNAs may also modulate TP53 expression under stress conditions. For instance, depletion of lncRNA-ROR enhances TP53 translation in the absence of stress, while its upregulation with stress or its ectopic expression suppresses TP53 translation through its direct interaction with the RBP hnRNP I. Interestingly, the promoter region of lncRNA-ROR contains TP53-binding sites and TP53 overexpression promotes lncRNA-ROR transcription, suggesting the existence of negative feedback regulation [185]. Finally, the expression levels of long stress-induced non-coding transcripts (LSINCTs) increase under stress conditions such as DNA damage. These lncRNAs are highly expressed in lung and breast cancer cell lines [186] and may rise in aging tissues, given that DNA damage accumulates with age.

3.3. lncRNAs affecting age-associated immune decline

Aberrant immune responses are a hallmark of aging and age-associated diseases, in part associated with the continuously secretion of proinflammatory cytokines by senescent cells [187, 188]. Recently, lncRNAs were reported to influence cytokine production and to modulate the subcellular localization of transcription factors involved in the cellular defense mechanisms against pathogenic and viral infections. The impact of lncRNAs on the immune response appears to be relatively well conserved across species [189]. Recent findings of lncRNAs linked to impaired immune responses in aging pathologies are detailed below.

Among the lncRNAs involved in the immune response, lincRNA-Cox2 was found upregulated after treatment with LPS (lipopolysaccharide) or the Toll-like receptor (TLR2/TLR1) agonist Pam3CSK4 in bone marrow-derived macrophages (BMDM), and required the transcription factor NFκB and the TLR adaptor Myd88. lincRNA-Cox2 selectively repressed the transcription of immune genes via its interaction with the RBPs hnRNP A/B and hnRNP A2/B1, while it enhanced IL6 expression via TLRs [190]. These results demonstrate that lincRNA-COX2 can activate and repress immune response genes in macrophages, and mediates the actions of NFκB, a major player in aging and age-related diseases [191]. Expression of COX2 was elevated by the lncRNA PACER (p50-associated COX-2 extragenic RNA), which binds to p53, a repressive subunit of NF-κB and blocks its association with the COX2 gene promoter, facilitating the transcriptional upregulation of COX2 expression by NF-κB [192].

The proinflammatory cytokine TNFα, linked to inflammation, aging, age-related diseases, and cellular senescence [193], induces the expression of several lncRNAs, including Lethe, a pseudogene-encoded lncRNA transcribed by NFκB. Depletion of Lethe led to an upregulation of NFKBIA and NFKB2, suggesting that Lethe may act as a repressor of NFκB activity. Accordingly, ectopic expression of Lethe repressed the induction of NFκB targets like Il6, Sod2, Il8, and Nfkbia [194], further supporting the view that Lethe participates in negative feedback modulation of NFκB target genes regulated by TNFα to prevent a sustained immune response. By contrast, lncRNA THRIL (TNFα- and hnRNP L-related immunoregulatory lincRNA) declined after TNFα treatment. After complexing with its binding partner hnRNP L, THRIL induced transcription of TNFα via its promoter [195]. These findings underscore the involvement of lncRNAs in networks regulated by NFκB and TNFα, two major regulators of aging, age-associated disease, and immune responses.

Expression of NEAT1 is induced by polyI:C. Depletion of NEAT1 inhibits the rise in IL8 and CCL5 mRNAs upon polyI:C treatment, while ectopic expression of NEAT1 is sufficient to induce IL8 and CCL5 expression without further stimuli. In addition, NEAT1 binds SFPQ, a transcriptional repressor of the IL8 gene. These data suggest that NEAT1 induced by polyI:C or viral infection might sequester SFPQ from the IL8 promoter region, activating IL8 transcription in response to an immune stimulus [196]. A similar mechanism may contribute to upregulating IL8 in the elderly, a process associated with cognitive dysfunction [197, 198].

Other lncRNAs tied less directly with aging include lnc-DC, which interacts with STAT3, a transcription factor closely implicated in the immune response, tumorigenesis and aging [199-201], and NeST, which controls Theiler’s virus infection and resistance to Salmonella infection in mice by elevating transcription of the cytokine IFNγ [202]. NeST may participate in aging-associated processes through its impact on the expression of IFNγ, a cytokine closely linked to age-related diseases, and through Theiler’s virus links to inflammation and neurological diseases [203-205].

4. AGING-ASSOCIATED NEURODEGENERATION

Neurodegenerative disorders in the elderly include Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease (HD), and Amyotrophic lateral sclerosis (ALS). Recently, several articles reported the role of lncRNAs in this group of diseases, as well as in schizophrenia, spinocerebellar ataxia, sciatic nerve injury, and neurogenesis [206]. In this section we will discuss lncRNAs affecting age-related neurological diseases (Table 3).

Table 3.

LncRNAs in aging-associated neurodegeneration.

LncRNA Processes Targets Relevance to Aging, Other Implications Refs.

BACE1AS AD BACE1 mRNA, HuD Competes with miR-485-5p for binding to,
and repressing BACE1 mRNA
[207-210]

lncRNA-17A AD Modulates alternative
splicing of GABABR2
pre-mRNA
Repressed GABABR2 variant A, promoted
variant B, and enhanced accumulation of
peptides Aβ42 and Aβ40
[211]

BCYRN1 AD Undetermined Upregulated in AD brain cortex [154]

GOMAFU
(MIAT)
QKI and SRSF1 Modulates alternative splicing of DISC1
(linked to age-related neurological
disorder), and ERBB4 pre-mRNAs
[212-214]

BC089918 Sciatic nerve injury Undetermined Downregulated in elderly, proposed as
cause of impaired nerve injury, recovery
[215]

RMST Neurogenesis,
neurodegeneration
hnRNP A2/B1, SOX2 Transcriptionally repressed by REST,
RMST is essential for neurogenesis
[216, 217]

GAS5 Neurodegeneration,
cancer
GR decoy Induced by psychogenic stress in
hippocampus of aged mouse, linked to
impaired learning and exploration
[30, 218,
219]

LncRNAs involved in neurodegeneration (column 1), the specific disease processes in which they are implicated (column 2), the direct molecular targets (column 3), and the relevance of the lncRNAs to neurodegeneration and aging-relevant events (column 4) are listed.

BACE1-antisense transcript (BACE1-AS) has extensive complementarity with BACE1 mRNA and enhances BACE1 mRNA stability by protecting it from degradation by factors including miR-485-5p [207, 208]. BACE1 protein levels and activity increase with brain aging and AD; in AD, high BACE1 levels correlate with elevated BACE1-AS abundance, suggesting that BACE1-AS could play a role in AD development or progression [207, 209, 210].

Also highly abundant in individuals with AD, lncRNA-17A is associated with a defect in the alternative splicing of GABABR2 (γ-aminobutyric acid B receptor 2). Overexpression of lncRNA-17A repressed GABABR2 variant A, promoted expression of variant B, and favored the accumulation of peptides Aβ42 and Aβ40, derived from cleavage of the amyloid precursor protein (APP) and closely implicated in AD pathogenesis. These findings indicate that lncRNA-17A may play a role in GABA signaling and Aβ production [211]. Also in AD, BCYRN1 was found to decrease by >60% in cortical areas between the ages of 49 and 86, but was upregulated in AD brains compared to age-matched normal brains [154].

The lncRNA GOMAFU (also known as myocardial infarction-associated transcript, MIAT) was downregulated in mouse cortical neurons and neurons derived from human induced pluripotent stem cell (iPSC) upon KCl-induced depolarization. GOMAFU interacts with QKI and SRSF1 to modulate the alternative splicing of DISC1 and ERBB4 pre-mRNAs, suggesting that by downregulating GOMAFU, potassium-activated ion channels make splicing factors available to carry out alternative splicing [212]. These could be linked to changes in alternative splicing of genes associated with age-related neurological disorders such as DISC1, for which some splice variants were associated with age-related recurrent major depression [213, 214].

Sciatic nerve injury led to differential expression of many lncRNAs, including BC089918, which was downregulated upon injury and its depletion promoted the outgrowth of dorsal root ganglion neurons. This function of BC089918 in the recovery of neurons from sciatic nerve injury has potential relevance to nerve injury and recovery in the elderly [215].

Expression of the brain-specific lncRNA RMST (rhabdomyosarcoma 2-associated transcript), which is transcriptionally repressed by the neuronal transcription factor REST, is essential for neurogenesis. In neural cells, RMST associated with the transcription factor SOX2 and the complex was necessary for the recruitment of SOX2 to appropriate DNA regions to implement a neurogenic program. RMST also interacts with the hnRNP A2/B1, whose aberrant function is associated with neurodegeneration [216, 217]

As mentioned above, the lncRNA GAS5 was associated both with cancer and with neurodegeneration, at least in part by acting as decoy for the transcription factor GR and thus inhibiting GR-driven gene expression programs and inducing apoptosis [30, 218, 219]. Gas5 was recently found to be highly expressed in the hippocampus of aged mice and mice subject to psychogenic stress (stress triggered by behavioral or emotional factors). Higher levels of Gas5 were associated with spatial-learning impairments and reduced novelty-induced exploration. These findings suggest a possible impact of lncRNA GAS5 on age-related behavior impairment and stress-induced changes in hippocampal function [218].

5. CONCLUDING REMARKS AND PERSPECTIVES

Numerous diseases that arise with advancing age, including obesity, diabetes, frailty, cancer, and neurodegeneration, are characterized by altered patterns of expressed transcripts, both coding and noncoding. As discussed in this review, lncRNAs affecting aging pathologies can modulate protein profiles via transcriptional and posttranscriptional regulatory processes (Figure 1). Through this broad range of influences, lncRNAs can regulate age-associated disease processes at the cell, tissue, organ, and system levels. Indeed, a few animal models are beginning to emerge supporting roles for lncRNAs in aging physiology and pathology at the organism level. For example, mouse lncRNAs from the myosin heavy chain 7 (Myh7) loci were found to be cardioprotective [220], deletion of Xist in mouse hematopoietic cells caused blood cancers, highlighting a tumor suppressive function for Xist [221], and knockdown of the neuronal lncRNA tuna in zebrafish recaputulated the impaired locomotor function seen in the brains of Huntington’s disease patients [221].

Figure 1. Molecular mechanisms of action of lncRNAs implicated in age-associated diseases.

Figure 1

In the nucleus, many lncRNAs involved in age-associated diseases elicit changes in chromatin or modulate the organization of chromosomes, control (enhance or repress) transcription by acting on transcription factors, or can modulate splicing by acting upon splicing regulatory factors. In the cytoplasm, lncRNAs implicated in age-associated diseases can modulate positively or negatively the stability or translation of subsets of mRNAs, alter the turnover of proteins, function as sponges/decoys for microRNAs or proteins, or influence mitochondrial metabolism.

It is therefore essential to understand comprehensively the spectrum of lncRNAs implicated in age-associated diseases, as well as the mechanisms that control lncRNA expression and localization. Given that their function is often linked to their interaction with other molecules, it is also critical to elucidate systematically the factors with which these lncRNAs interact: the proteins, DNA elements, mRNAs, microRNAs, and other noncoding RNAs that form complexes with lncRNAs. Not all lncRNAs are well conserved phylogenetically, but their structure and function does appear to be maintained with evolution [11, 223]. Therefore, assessment of these complexes in the context of the cell and the organism will provide valuable insight towards understanding the mechanisms of action of lncRNAs in aging pathologies.

LncRNAs are rapidly becoming molecules of exceptional interest in disease diagnosis and prognosis. The ease of detection of many lncRNAs in various bodily fluids, particularly blood, make this class of molecules attractive targets for different clinical applications; for example, lncRNA-ROR was recently found to be mobilized into extracellular vesicles, an event that was discovered to protect hepatocellular cancer against therapeutic interventions [224]. In addition, while lncRNAs are assumed not to produce large proteins, many lncRNAs are predicted to have short open reading frames that may give rise to small peptides that have thus been difficult to study. However, the development of better technical and computational methods [225] is facilitating the identification of micropeptides expressed from lncRNAs. One recent example is the skeletal muscle-specific micropeptide myoregulin (MLN), expressed from a 138-nucleotide open reading frame in an intron of a mouse lncRNA [226]. MLN was found to be a strong modulator of the muscle calcium system and thus modulated muscle contraction [226]; given its high conservation between mouse and human, MLN may be relevant to the decline in muscle function seen with human aging. Other lncRNA-encoded micropeptides of potential clinical interest are likely to emerge, as their analysis expands and deepens [227, 228].

With advances in our understanding of lncRNA expression and function, we can expect to identify classes of target lncRNAs of therapeutic interest that can be increased or suppressed for clinical benefit. At the same time, we are witnessing fast progress in technologies to detect aberrant lncRNAs and deliver lncRNA-relevant therapy with increasing precision and efficacy. In this regard, revolutionary advances in the design of adenoviral and AAV (adenoviral-associated) vectors for RNA delivery, the development of cell-specific RNA aptamer drugs, nanotechnology, and the widespread adoption of CRISPR (clustered regularly interspaced palindromic repeats)-Cas9-mediated interventions are enhancing our ability to repress and enhance gene expression with increasing accuracy. Thus, interventions to reduce the levels of harmful lncRNAs or elevate beneficial lncRNAs in selective tissues at specific times may soon become a reality.

Supplementary Material

NIHMS709002-supplement.docx (130.9KB, docx)

HIGHLIGHTS.

  • Long noncoding (lnc)RNAs regulate gene expression programs in aging pathologies

  • LncRNAs govern aging-impaired energy metabolism (obesity, diabetes)

  • LncRNAs affect aging-impaired proliferative and immune responses (cancer, immunity)

  • LncRNAs modulate age-related neurodegeneration

  • LncRNAs involved in diseases of aging represent promising therapeutic targets

ACKNOWLEDGEMENTS

This work was supported in its entirety by the National Institute on Aging-Intramural Research Program of the National Institutes of Health.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

REFERENCES

  • [1].Vural DC, Morrison G, Mahadevan L. Aging in complex interdependency networks. Phys. Rev. E Stat. Nonlin. Soft Matter Phys. 2014;89:022811. doi: 10.1103/PhysRevE.89.022811. [DOI] [PubMed] [Google Scholar]
  • [2].Lopez-Otin C, Blasco MA, Partridge L, Serrano M, Kroemer G. The hallmarks of aging. Cell. 2013;153:1194–1217. doi: 10.1016/j.cell.2013.05.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [3].Ferrucci L, Hesdorffer C, Bandinelli S, Simonsick EM. Frailty as a nexus between the biology of aging, environmental conditions and clinical geriatrics. Public Health Rev. 2010;32:475–488. [Google Scholar]
  • [4].Weinberg RA, Penman S. Small molecular weight monodisperse nuclear RNA. J. Mol. Biol. 1968;38:289–304. doi: 10.1016/0022-2836(68)90387-2. [DOI] [PubMed] [Google Scholar]
  • [5].Paul J, Duerksen JD. Chromatin-associated RNA content of heterochromatin and euchromatin. Mol. Cell. Biochem. 1975;9:9–16. doi: 10.1007/BF01731728. [DOI] [PubMed] [Google Scholar]
  • [6].Salditt-Georgieff M, Harpold MM, Wilson MC, Darnell JE., Jr Large heterogeneous nuclear ribonucleic acid has three times as many 5' caps as polyadenylic acid segments, and most caps do not enter polyribosomes. Mol. Cell. Biol. 1981;1:179–187. doi: 10.1128/mcb.1.2.179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [7].Slditt-Georgieff M, Darnell JE., Jr. Further evidence that the majority of primary nuclear RNA transcripts in mammalian cells do not contribute to mRNA. Mol. Cell. Biol. 1982;2:701–707. doi: 10.1128/mcb.2.6.701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [8].Lai F, Orom UA, Cesaroni M, Beringer M, Taatjes DJ, Blobel GA, Shiekhattar R. Activating RNAs associate with Mediator to enhance chromatin architecture and transcription. Nature. 2013;494:497–501. doi: 10.1038/nature11884. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [9].Eddy SR. Non-coding RNA genes and the modern RNA world. Nat. Rev. Genet. 2001;2:919–929. doi: 10.1038/35103511. [DOI] [PubMed] [Google Scholar]
  • [10].Djebali S, Davis CA, Merkel A, Dobin A, Lassmann T, Mortazavi A, Tanzer A, Lagarde J, Lin W, Schlesinger F, Xue C, Marinov GK, Khatun J, Williams BA, Zaleski C, Rozowsky J, Röder M, Kokocinski F, Abdelhamid RF, Alioto T, Antoshechkin I, Baer MT, Bar NS, Batut P, Bell K, Bell I, Chakrabortty S, Chen X, Curado J, Chrast, J., Derrien T, Drenkow J, Dumais E, Dumais J, Duttagupta R, Falconnet E, Fastuca M, Fejes-Toth K, Ferreira P, Foissac S, Fullwood MJ, Gao H, Gonzalez D, Gordon A, Gunawardena H, Howald C, Jha S, Johnson R, Kapranov P, King B, Kingswood C, Luo OJ, Park E, Persaud K, Preall JB, Ribeca P, Risk B, Robyr D, Sammeth M, Schaffer L, See LH, Shahab A, Skancke J, Suzuki AM, Takahashi H, Tilgner H, Trout D, Walters N, Wang H, Wrobel J, Yu Y, Ruan X, Hayashizaki Y, Harrow J, Gerstein M, Hubbard T, Reymond A, Antonarakis SE, Hannon G, Giddings MC, Ruan Y, Wold B, Carninci P, Guigó R, Gingeras TR. Landscape of transcription in human cells. Nature. 2012;489:101–108. doi: 10.1038/nature11233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [11].Guttman M, Amit I, Garber M, French C, Lin MF, Feldser D, Huarte M, Zuk O, Carey BW, Cassady JP, Cabili MN, Jaenisch R, Mikkelsen TS, Jacks T, Hacohen N, Bernstein BE, Kellis M, Regev A, Rinn JL, Lander ES. Chromatin signature reveals over a thousand highly conserved large non-coding RNAs in mammals. Nature. 2009;458:223–227. doi: 10.1038/nature07672. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Ulitsky I, Bartel DP. lincRNAs: Genomics, Evolution, and Mechanisms. Cell. 2013;154:26–46. doi: 10.1016/j.cell.2013.06.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].The FANTOM Consortium*. Carninci† P, Kasukawa T, Katayama S, Gough† J, C. Frith† M, Maeda N, Oyama R, Ravasi† T, Lenhard† B, Wells† C, Kodzius R, Shimokawa K, Bajic† VB, Brenner SE, Batalov S, Forrest ARR, Zavolan M, Davis MJ, Wilming LG, Aidinis V, Allen JE, Ambesi-Impiombato A, Apweiler R, Aturaliya RN, Bailey TL, Bansal M, Baxter L, Beisel KW, Bersano T, Bono H, Chalk AM, Chiu KP, Choudhary V, Christoffels A, Clutterbuck DR, Crowe ML, Dalla E, Dalrymple BP, de Bono B, Della Gatta G, di Bernardo D, Down T, Engstrom P, Fagiolini M, Faulkner G, Fletcher CF, Fukushima T, Furuno M, Futaki S, Gariboldi M, Georgii-Hemming P, Gingeras TR, Gojobori T, Green RE, Gustincich S, Harbers M, Hayashi Y, Hensch TK, Hirokawa N, Hill D, Huminiecki L, Iacono M, Ikeo K, Iwama A, Ishikawa T, Jakt M, Kanapin A, Katoh M, Kawasawa Y, Kelso J, Kitamura H, Kitano H, Kollias G, Krishnan SPT, Kruger A, Kummerfeld SK, Kurochkin IV, Lareau LF, Lazarevic D, Lipovich L, Liu J, Liuni S, McWilliam S, Madan Babu M, Madera M, Marchionni L, Matsuda H, Matsuzawa S, Miki H, Mignone F, Miyake S, Morris K, Mottagui-Tabar S, Mulder N, Nakano N, Nakauchi H, Ng P, Nilsson R, Nishiguchi S, Nishikawa S, Nori F, Ohara O, Okazaki Y, Orlando V, Pang KC, Pavan WJ, Pavesi G, Pesole G, Petrovsky N, Piazza S, Reed J, Reid JF, Ring BZ, Ringwald M, Rost B, Ruan Y, Salzberg SL, Sandelin A, Schneider C, Schönbach C, Sekiguchi K, Semple CAM, Seno S, Sessa L, Sheng Y, Shibata Y, Shimada H, Shimada K, Silva D, Sinclair B, Sperling S, Stupka E, Sugiura K, Sultana R, Takenaka Y, Taki K, Tammoja K, Tan SL, Tang S, Taylor MS, Tegner J, Teichmann SA, Ueda HR, van Nimwegen E, Verardo R, Wei CL, Yagi K, Yamanishi H, Zabarovsky E, Zhu S, Zimmer A, Hide W, Bult† C, Grimmond SM, Teasdale RD, Liu† ET, Brusic V, Quackenbush† J, Wahlestedt† C, Mattick† JS, Hume† DA, RIKEN Genome Exploration Research Group and Genome Science Group (Genome Network Project Core Group)*. Kai C, Sasaki D, Tomaru Y, Fukuda S, Kanamori-Katayama M, Suzuki† M, Aoki J, Arakawa T, Iida J, Imamura K, Itoh M, Kato T, Kawaji H, Kawagashira N, Kawashima T, Kojima M, Kondo S, Konno H, Nakano K, Ninomiya N, Nishio T, Okada M, Plessy C, Shibata K, Shiraki T, Suzuki S, Tagami M, Waki K, Watahiki A, Okamura-Oho Y, Suzuki H, Kawai J, Hayashizaki Y. The Transcriptional Landscape of the Mammalian Genome. Science. 2005;309:1559–1563. doi: 10.1126/science.1112014. [DOI] [PubMed] [Google Scholar]
  • [14].Yang L, Lin C, Jin C, Yang JC, Tanasa B, Li W, Merkurjev D, Ohgi KA, Meng D, Zhang J, Evans CP, Rosenfeld MG. lncRNA-dependent mechanisms of androgen-receptor-regulated gene activation programs. Nature. 2013;500:598–602. doi: 10.1038/nature12451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [15].Yoon JH, Abdelmohsen K, Gorospe M. Posttranscriptional gene regulation by long noncoding RNA. J. Mol. Biol. 2013;425:3723–3730. doi: 10.1016/j.jmb.2012.11.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [16].Yoon JH, Abdelmohsen K, Kim J, Yang X, Martindale JL, Tominaga-Yamanaka K, White EJ, Orjalo AV, Rinn JL, Kreft SG, Wilson GM, Gorospe M. Scaffold function of long non-coding RNA HOTAIR in protein ubiquitination. Nat. Commun. 2013;4:2939. doi: 10.1038/ncomms3939. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [17].Grammatikakis I, Panda AC, Abdelmohsen K, Gorospe M. Long noncoding RNAs (lncRNAs) and the molecular hallmarks of aging. Aging. 2014;6:992–1009. doi: 10.18632/aging.100710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [18].Qureshi IA, Mehler MF. Non-coding RNA networks underlying cognitive disorders across the lifespan. Trends Mol. Med. 2011;17:337–346. doi: 10.1016/j.molmed.2011.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [19].Qureshi IA, Mattick JS, Mehler MF. Long non-coding RNAs in nervous system function and disease. Brain Res. 2010;1338:20–35. doi: 10.1016/j.brainres.2010.03.110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [20].Gutschner T, Diederichs S. The hallmarks of cancer: A long non-coding RNA point of view. RNA Biol. 2012;9:703–719. doi: 10.4161/rna.20481. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [21].Esteller M. Non-coding RNAs in human disease. Nat. Rev. Genet. 2011;12:861–874. doi: 10.1038/nrg3074. [DOI] [PubMed] [Google Scholar]
  • [22].Huarte M, Rinn JL. Large non-coding RNAs: missing links in cancer? Hum. Mol. Genet. 2010;19:152–161. doi: 10.1093/hmg/ddq353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [23].Greco S, Gorospe M, Martelli F. Noncoding RNA in age-related cardiovascular diseases. J. Mol. Cell. Cardiol. 2015 doi: 10.1016/j.yjmcc.2015.01.011. [epub ahead of print] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [24].Ulitsky I, Shkumatava A, Jan CH, Sive H, Bartel DP. Conserved function of lincRNAs in vertebrate embryonic development despite rapid sequence evolution. Cell. 2011;147:1537–1550. doi: 10.1016/j.cell.2011.11.055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [25].Cabili MN, Trapnell C, Goff L, Koziol M, Tazon-Vega B, Regev A, Rinn JL. Integrative annotation of human large intergenic noncoding RNAs reveals global properties and specific subclasses. Genes Dev. 2011;25:1915–1927. doi: 10.1101/gad.17446611. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].Novikova IV, Hennelly SP, Sanbonmatsu KY. Sizing up long non-coding RNAs do lncRNAs have secondary and tertiary structure? Bioarchitecture. 2012;2:189–199. doi: 10.4161/bioa.22592. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Wilusz JE, JnBaptiste CK, Lu LY, Kuhn CD, Joshua-Tor L, Sharp PA. A triple helix stabilizes the 3’ ends of long noncoding RNAs that lack poly(A) tails. Genes Dev. 2012;26:2392–2407. doi: 10.1101/gad.204438.112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [28].Rinn JL, Chang HY. Genome regulation by long noncoding RNAs. Ann. Rev. Biochem. 2012;81:145–166. doi: 10.1146/annurev-biochem-051410-092902. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [29].Faghihi MA, Wahlestedt C. Regulatory roles of natural antisense transcripts. Nat Rev Mol Cell Biol. 2009;10:637–643. doi: 10.1038/nrm2738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [30].Wapinski O, Chang HY. Long noncoding RNAs and human disease. Trends Cell Biol. 2011;21:354–361. doi: 10.1016/j.tcb.2011.04.001. [DOI] [PubMed] [Google Scholar]
  • [31].Marques AC, Hughes J, Graham B, Kowalczyk MS, Higgs DR, Ponting CP. Chromatin signatures at transcriptional start sites separate two equally populated yet distinct classes of intergenic long noncoding RNAs. Genome Biol. 2013;14:131–144. doi: 10.1186/gb-2013-14-11-r131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [32].Alla AS, M. Alan C, Benoit M, Sumeeta G, David AO, Matthew GG, Albert EA, Charles L, Phillip AS, Cosmas CG, Richard AY. Divergent transcription of long noncoding RNA/mRNA gene pairs in embryonic stem cells. Proc. Natl. Acad. Sci. USA. 2013;110:2876–2881. doi: 10.1073/pnas.1221904110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [33].Dinger ME, Pang KC, Mercer TR, Crowe ML, Grimmond SM, Mattick JS. NRED: a database of long noncoding RNA expression. Nucleic acids Res. 2009;37:D122–126. doi: 10.1093/nar/gkn617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [34].Derrien T, Johnson R, Bussotti G, Tanzer A, Djebali S, Tilgner H, Guernec G, Martin D, Merkel A, Knowles DG, Lagarde J, Veeravalli L, Ruan X, Ruan Y, Lassmann T, Carninci P, Brown JB, Lipovich L, Gonzalez JM, Thomas M, Davis CA, Shiekhattar R, Gingeras TR, Hubbard TJ, Notredame C, Harrow J, Guigo R. The GENCODE v7 catalog of human long noncoding RNAs: analysis of their gene structure, evolution, and expression. Genome Res. 2012;22:1775–1789. doi: 10.1101/gr.132159.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [35].Wang KC, Chang HY. Molecular Mechanisms of Long Noncoding RNAs. Mol. Cell. 2011;43:904–914. doi: 10.1016/j.molcel.2011.08.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [36].Quinodoz S, Guttman M. Long noncoding RNAs: an emerging link between gene regulation and nuclear organization. Trends Cell Biol. 2014;24:651–663. doi: 10.1016/j.tcb.2014.08.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [37].Popadin K, Gutierrez-Arcelus M, Dermitzakis ET, Antonarakis SE. Genetic and epigenetic regulation of human lincRNA gene expression. Am. J. Hum. Genet. 2013;93:1015–1026. doi: 10.1016/j.ajhg.2013.10.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [38].Lee JT. Epigenetic regulation by long noncoding RNAs. Science. 2012;338:1435–1439. doi: 10.1126/science.1231776. [DOI] [PubMed] [Google Scholar]
  • [39].Marchese FP, Huarte M. Long non-coding RNAs and chromatin modifiers: Their place in the epigenetic code. Epigenetics. 2014;9:21–26. doi: 10.4161/epi.27472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [40].Guil S, Esteller M. Cis-acting noncoding RNAs: friends and foes. Nat. Struct. Mol. Biol. 2012;19:1068–1075. doi: 10.1038/nsmb.2428. [DOI] [PubMed] [Google Scholar]
  • [41].Geisler S, Coller J. RNA in unexpected places: long non-coding RNA functions in diverse cellular contexts. Nat. Rev. Mol. Cell. Biol. 2013;14:699–712. doi: 10.1038/nrm3679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Ørom UA, Shiekhattar R. Noncoding RNAs and enhancers: complications of a long-distance relationship. Trends Genet. 2011;10:433–439. doi: 10.1016/j.tig.2011.06.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [43].Abdelmohsen K, Panda AC, Kang MJ, Guo R, Kim J, Grammatikakis I, Yoon JH, Dudekula DB, Noh JH, Yang X, Martindale JL, Gorospe M. 7SL RNA represses p53 translation by competing with HuR. Nucleic Acids Res. 2014;42:10099–10111. doi: 10.1093/nar/gku686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [44].Yang F, Huo XS, Yuan SX, Zhang L, Zhou WP, Wang F, Sun SH. Repression of the long noncoding RNA-LET by histone deacetylase 3 contributes to hypoxia-mediated metastasis. Mol. Cell. 2013;49:1083–1096. doi: 10.1016/j.molcel.2013.01.010. [DOI] [PubMed] [Google Scholar]
  • [45].Yang F, Zhang H, Mei Y, Wu M. Reciprocal regulation of HIF-1alpha and lincRNA-p21 modulates the Warburg effect. Mol. Cell. 2014;53:88–100. doi: 10.1016/j.molcel.2013.11.004. [DOI] [PubMed] [Google Scholar]
  • [46].Kawada T, Otsuka T, Endo T, Kon Y. Prevalence of the metabolic syndrome and its relationship with diabetes mellitus by aging. The aging male. 2011;14:203–206. doi: 10.3109/13685538.2011.565092. [DOI] [PubMed] [Google Scholar]
  • [47].Kornfeld JW, Brüning JC. Regulation of metabolism by long, non-coding RNAs. Front Genet. 2014;5:57. doi: 10.3389/fgene.2014.00057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [48].Slawik M, Vidal-Puig AJ. Lipotoxicity, overnutrition and energy metabolism in aging. Ageing Res. Rev. 2006;5:144–164. doi: 10.1016/j.arr.2006.03.004. [DOI] [PubMed] [Google Scholar]
  • [49].Cartwright MJ, Tchkonia T, Kirkland JL. Aging in adipocytes: Potential impact of inherent, depot-specific mechanisms. Exp. Gerontol. 2007;42:463–471. doi: 10.1016/j.exger.2007.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [50].Kuk JL, Saunders TJ, Davidson LE, Ross R. Age-related changes in total and regional fat distribution. Ageing Res. Rev. 2009;8:339–348. doi: 10.1016/j.arr.2009.06.001. [DOI] [PubMed] [Google Scholar]
  • [51].Raguso CA, Kyle U, Kossovsky MP, Roynette C, Paoloni-Giacobino A, Hans D, Genton L, Pichard C. A 3-year longitudinal study on body composition changes in the elderly: Role of physical exercise. Clin. Nutr. 2006;25:573–580. doi: 10.1016/j.clnu.2005.10.013. [DOI] [PubMed] [Google Scholar]
  • [52].Karagiannides I, Tchkonia T, Dobson DE, Steppan CM, Cummins P, Chan G, Salvatori K, Hadzopoulou-Cladaras M, Kirkland JL. Altered expression of C/EBP family members results in decreased adipogenesis with aging. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2001;280:R1772–1780. doi: 10.1152/ajpregu.2001.280.6.R1772. [DOI] [PubMed] [Google Scholar]
  • [53].Huot ME, Vogel G, Zabarauskas A, Ngo CT, Coulombe-Huntington J, Majewski J, Richard S. The Sam68 STAR RNA-binding protein regulates mTOR alternative splicing during adipogenesis. Mol. Cell. 2012;46:187–199. doi: 10.1016/j.molcel.2012.02.007. [DOI] [PubMed] [Google Scholar]
  • [54].Ling H, Li X, Yao CH, Hu B, Liao D, Feng S, Wen G, Zhang L. The physiological and pathophysiological roles of adipocyte miRNAs. Biochem. Cell Biol. 2013;91:195–202. doi: 10.1139/bcb-2012-0053. [DOI] [PubMed] [Google Scholar]
  • [55].Sun L, Goff LA, Trapnell C, Alexander R, Lo KA, Hacisuleyman E, Sauvageau M, Tazon-Vega B, Kelley DR, Hendrickson DG, Yuan B, Kellis M, Lodish HF, Rinn JL. Long noncoding RNAs regulate adipogenesis. Proc. Natl. Acad. Sci. USA. 2013;110:3387–3392. doi: 10.1073/pnas.1222643110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [56].Ye P, Zhang XJ, Wang ZJ, Zhang C. Effect of aging on the expression of peroxisome proliferator-activated receptor gamma and the possible relation to insulin resistance. Gerontology. 2006;52:69–75. doi: 10.1159/000090951. [DOI] [PubMed] [Google Scholar]
  • [57].Hacisuleyman E, Goff LA, Trapnell C, Williams A, Henao-Mejia J, Sun L, McClanahan P, Hendrickson DG, Sauvageau M, Kelley DR, Morse M, Engreitz J, Lander ES, Guttman M, Lodish HF, Flavell R, Raj A, Rinn JL. Topological organization of multichromosomal regions by the long intergenic noncoding RNA Firre. Nat. Struct. Mol. Biol. 2014;21:198–206. doi: 10.1038/nsmb.2764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [58].Deng H, Jankovic J, Guo Y, Xie W, Le W. Small interfering RNA targeting the PINK1 induces apoptosis in dopaminergic cells SH-SY5Y. Biochem. Biophys. Res. Comm. 2005;337:1133–1138. doi: 10.1016/j.bbrc.2005.09.178. [DOI] [PubMed] [Google Scholar]
  • [59].Valente EM, Abou-Sleiman PM, Caputo V, Muqit MM, Harvey K, Gispert S, Ali Z, Del Turco D, Bentivoglio AR, Healy DG, Albanese A, Nussbaum R, Gonzalez-Maldonado R, Deller T, Salvi S, Cortelli P, Gilks WP, Latchman DS, Harvey RJ, Dallapiccola B, Auburger G, Wood NW. Hereditary early-onset Parkinson's disease caused by mutations in PINK1. Science. 2004;304:1158–1160. doi: 10.1126/science.1096284. [DOI] [PubMed] [Google Scholar]
  • [60].Scheele C, Nielsen AR, Walden TB, Sewell DA, Fischer CP, Brogan RJ, Petrovic N, Larsson O, Tesch PA, Wennmalm K, Hutchinson DS, Cannon B, Wahlestedt C, Pedersen BK, Timmons JA. Altered regulation of the PINK1 locus: a link between type 2 diabetes and neurodegeneration? FASEB J. 2007;21:3653–3665. doi: 10.1096/fj.07-8520com. [DOI] [PubMed] [Google Scholar]
  • [61].Jones BK, Levorse J, Tilghman SM. Deletion of a nuclease-sensitive region between the Igf2 and H19 genes leads to Igf2 misregulation and increased adiposity. Hum. Mol. Genet. 2001;10:807–814. doi: 10.1093/hmg/10.8.807. [DOI] [PubMed] [Google Scholar]
  • [62].Fu VX, Dobosy JR, Desotelle JA, Almassi N, Ewald JA, Srinivasan R, Berres M, Svaren J, Weindruch R, Jarrard DF. Aging and cancer-related loss of insulin-like growth factor 2 imprinting in the mouse and human prostate. Cancer Res. 2008;68:6797–6802. doi: 10.1158/0008-5472.CAN-08-1714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [63].Liu Y, Ferguson JF, Xue C, Ballantyne RL, Silverman IM, Gosai SJ, Serfecz J, Morley MP, Gregory BD, Li M, Reilly MP. Tissue-specific RNA-Seq in human evoked inflammation identifies blood and adipose LincRNA signatures of cardiometabolic diseases. Arterioscler. Thromb. Vasc. Biol. 2014;34:902–912. doi: 10.1161/ATVBAHA.113.303123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [64].Morley JE, Mooradian AD, Rosenthal MJ, Kaiser FE. Diabetes mellitus in elderly patients. Is it different? Am. J. Med. 1987;83:533–544. doi: 10.1016/0002-9343(87)90767-4. [DOI] [PubMed] [Google Scholar]
  • [65].Hartnell JM, Morley JE, Mooradian AD. Reduction of alkali-induced white blood cell DNA unwinding rate: a potential biomarker of aging. J. Gerontol. 1989;44:B125–130. doi: 10.1093/geronj/44.5.b125. [DOI] [PubMed] [Google Scholar]
  • [66].Gunasekaran U, Gannon M. Type 2 diabetes and the aging pancreatic beta cell. Aging. 2011;3:565–575. doi: 10.18632/aging.100350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [67].Coon PJ, Rogus EM, Drinkwater D, Muller DC, Goldberg AP. Role of body fat distribution in the decline in insulin sensitivity and glucose tolerance with age. J. Clin. Endocrinol. Metab. 1992;75:1125–1132. doi: 10.1210/jcem.75.4.1400882. [DOI] [PubMed] [Google Scholar]
  • [68].Bourey RE, Kohrt WM, Kirwan JP, Staten MA, King DS, Holloszy JO. Relationship between glucose tolerance and glucose-stimulated insulin response in 65-year-olds. J. Gerontol. 1993;48:M122–127. doi: 10.1093/geronj/48.4.m122. [DOI] [PubMed] [Google Scholar]
  • [69].Ammon HP, Fahmy A, Mark M, Wahl MA, Youssif N. The effect of glucose on insulin release and ion movements in isolated pancreatic islets of rats in old age. J. Physiol. 1987;384:347–354. doi: 10.1113/jphysiol.1987.sp016458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [70].Prentki M, Nolan CJ. Islet beta cell failure in type 2 diabetes. J. Clin. Invest. 2006;116:1802–1812. doi: 10.1172/JCI29103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [71].Moran I, Akerman I, van de Bunt M, Xie R, Benazra M, Nammo T, Arnes L, Nakic N, Garcia-Hurtado J, Rodriguez-Segui S, Pasquali L, Sauty-Colace C, Beucher A, Scharfmann R, van Arensbergen J, Johnson PR, Berry A, Lee C, Harkins T, Gmyr V, Pattou F, Kerr-Conte J, Piemonti L, Berney T, Hanley N, Gloyn AL, Sussel L, Langman L, Brayman KL, Sander M, McCarthy MI, Ravassard P, Ferrer J. Human beta cell transcriptome analysis uncovers lncRNAs that are tissue-specific, dynamically regulated, and abnormally expressed in type 2 diabetes. Cell Metab. 2012;16:435–448. doi: 10.1016/j.cmet.2012.08.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [72].Ku GM, Kim H, Vaughn IW, Hangauer MJ, Myung Oh C, German MS, McManus MT. Research resource: RNA-Seq reveals unique features of the pancreatic beta-cell transcriptome. Mol. Endocrinol. 2012;26:1783–1792. doi: 10.1210/me.2012-1176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [73].Voight BF, Scott LJ, Steinthorsdottir V, Morris AP, Dina C, Welch RP, Zeggini E, Huth C, Aulchenko YS, Thorleifsson G, McCulloch LJ, Ferreira T, Grallert H, Amin N, Wu GM, Willer CJ, Raychaudhuri S, McCarroll SA, Langenberg C, Hofmann OM, Dupuis J, Qi L, Segre AV, van Hoek M, Navarro P, Ardlie K, Balkau B, Benediktsson R, Bennett AJ, Blagieva R, Boerwinkle E, Bonnycastle LL, Bostrom KB, Bravenboer B, Bumpstead S, Burtt NP, Charpentier G, Chines PS, Cornelis M, Couper DJ, Crawford G, Doney ASF, Elliott KS, Elliott AL, Erdos MR, Fox CS, Franklin CS, Ganser M, Gieger C, Grarup N, Green T, Griffin S, Groves CJ, Guiducci C, Hadjadj S, Hassanali N, Herder C, Isomaa B, Jackson AU, Johnson PRV, Jorgensen T, Kao WHL, Klopp N, Kong A, Kraft P, Kuusisto J, Lauritzen T, Li M, Lieverse A, Lindgren CM, Lyssenko V, Marre M, Meitinger T, Midthjell K, Morken MA, Narisu N, Nilsson P, Owen KR, Payne F, Perry JRB, Petersen AK, Platou C, Proenca C, Prokopenko I, Rathmann W, Rayner NW, Robertson NR, Rocheleau G, Roden M, Sampson MJ, Saxena R, Shields BM, Shrader P, Sigurdsson G, Sparso T, Strassburger K, Stringham HM, Sun Q, Swift AJ, Thorand B, Tichet J, Tuomi T, van Dam RM, van Haeften TW, van Herpt T, van Vliet-Ostaptchouk JV, Walters GB, Weedon MN, Wijmenga C, Witteman J, Bergman RN, Cauchi S, Collins FS, Gloyn AL, Gyllensten U, Hansen T, Hide WA, Hitman GA, Hofman A, Hunter DJ, Hveem K, Laakso M, Mohlke KL, Morris AD, Palmer CNA, Pramstaller PP, Rudan I, Sijbrands E, Stein LD, Jaakko T, Uitterlinden A, Walker M, Wareham NJ, Watanabe RM, Abecasis GR, Boehm BO, Campbell H, Daly MJ, Hattersley AT, Hu FB, Meigs JB, Pankow JS, Pedersen O, Wichmann HE, Barroso I, Florez JC, Frayling TM, Groop L, Sladek R, Thorsteinsdottir U, Wilson JF, Illig T, Froguel P, van Duijn CM, Stefansson K, Altshuler D, Boehnke M, McCarthy MI, Investigators M, Consortium G. Twelve type 2 diabetes susceptibility loci identified through large-scale association analysis. Nat. Genet. 2010;42:579–589. doi: 10.1038/ng.609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [74].Moran I, Akerman I, van de Bunt M, Xie R, Benazra M, Nammo T, Arnes L, Nakic N, Garcia-Hurtado J, Rodriguez-Segui S, Pasquali L, Sauty-Colace C, Beucher A, Scharfmann R, van Arensbergen J, Johnson PR, Berry A, Lee C, Harkins T, Gmyr V, Pattou F, Kerr-Conte J, Piemonti L, Berney T, Hanley N, Gloyn AL, Sussel L, Langman L, Brayman KL, Sander M, McCarthy MI, Ravassard P, Ferrer J. Human beta cell transcriptome analysis uncovers lncRNAs that are tissue-specific, dynamically regulated, and abnormally expressed in type 2 diabetes. Cell Metab. 2012;16:435–448. doi: 10.1016/j.cmet.2012.08.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [75].Yang Y, Chang BH, Chan L. Sustained expression of the transcription factor GLIS3 is required for normal beta cell function in adults. EMBO Mol. Med. 2013;5:92–104. doi: 10.1002/emmm.201201398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [76].Hannou SA, Wouters K, Paumelle1 R, Staels B. Functional genomics of the CDKN2A/B locus in cardiovascular and metabolic disease: what have we learned from GWASs? Trends Endocrinol. Metab. 2015;26:176–184. doi: 10.1016/j.tem.2015.01.008. [DOI] [PubMed] [Google Scholar]
  • [77].Latres E, Malumbres M, Sotillo R, Martin J, Ortega S, Martin-Caballero J, Flores JM, Cordon-Cardo C, Barbacid M. Limited overlapping roles of P15 (INK4b) and P18 (INK4c) cell cycle inhibitors in proliferation and tumorigenesis. EMBO J. 2000;19:3496–3506. doi: 10.1093/emboj/19.13.3496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [78].Kotake Y, Nakagawa T, Kitagawa K, Suzuki S, Liu N, Kitagawa M, Xiong Y. Long non-coding RNA ANRIL is required for the PRC2 recruitment to and silencing of p15 (INK4B) tumor suppressor gene. Oncogene. 2011;30:1956–1962. doi: 10.1038/onc.2010.568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [79].Pasmant E, Laurendeau I, Heron D, Vidaud M, Vidaud D, Biéche I. Characterization of a germ-line deletion, including the entire INK4/ARF locus, in a melanoma-neural system tumor family: Identification of ANRIL, an antisense noncoding RNA whose expression coclusters with ARF. Cancer Res. 2007;67:3963–3969. doi: 10.1158/0008-5472.CAN-06-2004. [DOI] [PubMed] [Google Scholar]
  • [80].Pasmant E, Sabbagh A, Vidaud M, Biéche I. ANRIL, a long noncoding RNA, is an unexpected major hotspot in GWAS. FASEB J. 2011;2:444–448. doi: 10.1096/fj.10-172452. [DOI] [PubMed] [Google Scholar]
  • [81].Larsson NG. Somatic mitochondrial DNA mutations in mammalian aging. Ann. Rev. Biochem. 2010;79:683–706. doi: 10.1146/annurev-biochem-060408-093701. [DOI] [PubMed] [Google Scholar]
  • [82].Lee HC, Wei YH. Mitochondria and aging. Adv. Exp. Med. Biol. 2012;942:311–327. doi: 10.1007/978-94-007-2869-1_14. [DOI] [PubMed] [Google Scholar]
  • [83].Rackham O, Shearwood AM, Mercer TR, Davies SM, Mattick JS, Filipovska A. Long noncoding RNAs are generated from the mitochondrial genome and regulated by nuclear-encoded proteins. RNA. 2011;17:2085–2093. doi: 10.1261/rna.029405.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [84].Mercer TR, Neph S, Dinger ME, Crawford J, Smith MA, Shearwood AM, Haugen E, Bracken CP, Rackham O, Stamatoyannopoulos JA, Filipovska A, Mattick JS. The human mitochondrial transcriptome. Cell. 2011;146:645–658. doi: 10.1016/j.cell.2011.06.051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [85].Rackham O, Filipovska A. Analysis of the human mitochondrial transcriptome using directional deep sequencing and parallel analysis of RNA ends. Methods Mol. Biol. 2014;1125:263–275. doi: 10.1007/978-1-62703-971-0_21. [DOI] [PubMed] [Google Scholar]
  • [86].Rackham O, Shearwood AM, Mercer TR, Davies SM, Mattick JS, Filipovska A. Long noncoding RNAs are generated from the mitochondrial genome and regulated by nuclear-encoded proteins. RNA. 2011;17:2085–2093. doi: 10.1261/rna.029405.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [87].Martin AN, Li YD. RNase MRP RNA and human genetic diseases. Cell Res. 2007;17:219–226. doi: 10.1038/sj.cr.7310120. [DOI] [PubMed] [Google Scholar]
  • [88].Dorn GW., 2nd LIPCAR: a mitochondrial lnc in the noncoding RNA chain? Circ. Res. 2014;114:1548–1550. doi: 10.1161/CIRCRESAHA.114.304028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [89].Kumarswamy R, Bauters C, Volkmann I, Maury F, Fetisch J, Holzmann A, Lemesle G, de Groote P, Pinet F, Thum T. Circulating long noncoding RNA, LIPCAR, predicts survival in patients with heart failure. Circ. Res. 2014;114:1569–1575. doi: 10.1161/CIRCRESAHA.114.303915. [DOI] [PubMed] [Google Scholar]
  • [90].Chistiakov DA, Sobenin IA, Revin VV, Orekhov AN, Bobryshev YV. Mitochondrial aging and age-related dysfunction of mitochondria. BioMed Res. Int. 2014;2014:238463. doi: 10.1155/2014/238463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [91].Lim S, Kim JH, Yoon JW, Kang SM, Choi SH, Park YJ, Kim KW, Lim JY, Park KS, Jang HC. Sarcopenic obesity: prevalence and association with metabolic syndrome in the Korean Longitudinal Study on Health and Aging (KLoSHA) Diabetes Care. 2010;33:1652–1654. doi: 10.2337/dc10-0107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [92].Dey BK, Pfeifer K, Dutta A. The H19 long noncoding RNA gives rise to microRNAs miR-675-3p and miR-675-5p to promote skeletal muscle differentiation and regeneration. Genes Dev. 2014;28:491–501. doi: 10.1101/gad.234419.113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [93].Kallen AN, Zhou XB, Xu J, Qiao C, Ma J, Yan L, Lu L, Liu C, Yi JS, Zhang H, Min W, Bennett AM, Gregory RI, Ding Y, Huang Y. The imprinted H19 lncRNA antagonizes let-7 microRNAs. Mol. Cell. 2013;52:101–112. doi: 10.1016/j.molcel.2013.08.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [94].Gao Y, Wu F, Zhou J, Yan L, Jurczak MJ, Lee HY, Yang L, Mueller M, Zhou XB, Dandolo L, Szendroedi J, Roden M, Flannery C, Taylor H, Carmichael GG, Shulman G, Huang Y. The H19/let-7 double-negative feedback loop contributes to glucose metabolism in muscle cells. Nucleic Acids Res. 2014;42:13799–137811. doi: 10.1093/nar/gku1160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [95].Cesana M, Cacchiarelli D, Legnini I, Santini T, Sthandier O, Chinappi M, Tramontano A, Bozzoni I. A long noncoding RNA controls muscle differentiation by functioning as a competing endogenous RNA. Cell. 2011;147:358–369. doi: 10.1016/j.cell.2011.09.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [96].Legnini I, Morlando M, Mangiavacchi A, Fatica A, Bozzoni I. A feedforward regulatory loop between HuR and the long noncoding RNA linc-MD1 controls early phases of myogenesis. Mol. Cell. 2014;53:506–514. doi: 10.1016/j.molcel.2013.12.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [97].Masuda K, Marasa B, Martindale JL, Halushka MK, Gorospe M. Tissue- and age-dependent expression of RNA-binding proteins that influence mRNA turnover and translation. Aging. 2009;26:681–698. doi: 10.18632/aging.100073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [98].Marco SD, Mazroui R, Dallaire P, Chittur S, Tenenbaum SA, Radzioch D, Marette A, Gallouzi IE. NF-kappa B-mediated MyoD decay during muscle wasting requires nitric oxide synthase mRNA stabilization, HuR protein, and nitric oxide release. Mol. Cell. Biol. 2005;25:6533–6545. doi: 10.1128/MCB.25.15.6533-6545.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [99].Hall DT, Ma JF, Marco SD, Gallouzi IE. Inducible nitric oxide synthase (iNOS) in muscle wasting syndrome, sarcopenia, and cachexia. Aging. 2011;3:702–715. doi: 10.18632/aging.100358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [100].Wang Y, Pang WJ, Wei N, Xiong Y, Wu WJ, Zhao CZ, Shen QW, Yang GS. Identification, stability and expression of Sirt1 antisense long non-coding RNA. Gene. 2014;539:117–124. doi: 10.1016/j.gene.2014.01.037. [DOI] [PubMed] [Google Scholar]
  • [101].Gong H, Pang J, Han YW, Dai Y, Dai DP, Cai JP, Zhang TM. Age-dependent tissue expression patterns of Sirt1 in senescence-accelerated mice. Mol. Med. Rep. 2014;10:3296–3302. doi: 10.3892/mmr.2014.2648. [DOI] [PubMed] [Google Scholar]
  • [102].Pardo PS, Boriek AM. The physiological roles of Sirt1 in skeletal muscle. Aging. 2011;3:430–437. doi: 10.18632/aging.100312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [103].Watts R, Johnsen VL, Shearer J, Hittel DS. Myostatin-induced inhibition of the long noncoding RNA Malat1 is associated with decreased myogenesis. Am. J. Physiol. Cell. Physiol. 2013;304:C995–1001. doi: 10.1152/ajpcell.00392.2012. [DOI] [PubMed] [Google Scholar]
  • [104].Michalik KM, You X, Manavski Y, Doddaballapur A, Zörnig M, Braun T, John D, Ponomareva Y, Chen W, Uchida S, Boon RA, Dimmeler S. Long noncoding RNA MALAT1 regulates endothelial cell function and vessel growth. Circ Res. 2014;114:1389–1397. doi: 10.1161/CIRCRESAHA.114.303265. [DOI] [PubMed] [Google Scholar]
  • [105].Schirwis E, Agbulut O, Vadrot N, Mouisel E, Hourde C, Bonnieu A, Butler-Browne G, Amthor H, Ferry A. The beneficial effect of myostatin deficiency on maximal muscle force and power is attenuated with age. Exp. Gerontol. 2013;48:183–190. doi: 10.1016/j.exger.2012.11.008. [DOI] [PubMed] [Google Scholar]
  • [106].McKay BR, Ogborn DI, Bellamy LM, Tarnopolsky MA, Parise G. Myostatin is associated with age-related human muscle stem cell dysfunction. FASEB J. 2012;26:2509–2521. doi: 10.1096/fj.11-198663. [DOI] [PubMed] [Google Scholar]
  • [107].Soares RJ, Cagnin S, Chemello F, Silvestrin M, Musaro A, de Pitta D, Lanfranchi G, Sandri M. Involvement of microRNAs in the regulation of muscle wasting during catabolic conditions. J. Biol. Chem. 2014;289:21909–21925. doi: 10.1074/jbc.M114.561845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [108].Lu L, Sun K, Chen X, Zhao Y, Wang L, Zhou L, Sun H, Wang H. Genome-wide survey by ChIP-seq reveals YY1 regulation of lincRNAs in skeletal myogenesis. EMBO J. 2013;32:2575–2588. doi: 10.1038/emboj.2013.182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [109].Wang L, Zhao Y, Bao X, Zhu X, Kwok YK, Sun K, Chen X, Huang Y, Jauch R, Esteban MA, Sun H, Wang H. LncRNA Dum interacts with Dnmts to regulate Dppa2 expression during myogenic differentiation and muscle regeneration. Cell Res. 2015;25:335–350. doi: 10.1038/cr.2015.21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [110].Xu W, Li Y, Liu C, Zhao S. Protein lysine acetylation guards metabolic homeostasis to fight against cancer. Oncogene. 2014;33:2279–2285. doi: 10.1038/onc.2013.163. [DOI] [PubMed] [Google Scholar]
  • [111].Singh SR. Stem cell niche in tissue homeostasis, aging and cancer. Curr. Med. Chem. 2012;19:5965–5974. doi: 10.2174/092986712804485917. [DOI] [PubMed] [Google Scholar]
  • [112].Marques O, da Silva BM, Porto G, Lopes C. Iron homeostasis in breast cancer. Cancer Lett. 2014;347:1–14. doi: 10.1016/j.canlet.2014.01.029. [DOI] [PubMed] [Google Scholar]
  • [113].Edwards BK, Howe HL, Ries LA, Thun MJ, Rosenberg HM, Yancik R, Wingo PA, Jemal A, Feigal EG. Annual report to the nation on the status of cancer, 1973-1999, featuring implications of age and aging on U.S. cancer burden. Cancer. 2002;94:2766–2792. doi: 10.1002/cncr.10593. [DOI] [PubMed] [Google Scholar]
  • [114].Thorvaldsen JL, Duran KL, Bartolomei MS. Deletion of the H19 differentially methylated domain results in loss of imprinted expression of H19 and Igf2. Genes Dev. 1998;12:3693–3702. doi: 10.1101/gad.12.23.3693. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [115].Gabory A, Jammes H, Dandolo L. The H19 locus: Role of an imprinted non-coding RNA in growth and development. Bioessays. 2010;32:473–480. doi: 10.1002/bies.200900170. [DOI] [PubMed] [Google Scholar]
  • [116].Matouk IJ, DeGroot N, Mezan S, Ayesh S, Abu-Iail R, Hochberg A, Galun E. The H19 Non-Coding RNA Is Essential for Human Tumor Growth. PloS One. 2007;2:e845. doi: 10.1371/journal.pone.0000845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [117].Issa JPJ, Vertino PM, Boehm CD, Newsham IF, Baylin SB. Switch from monoallelic to biallelic human IGF2 promoter methylation during aging and carcinogenesis. Proc. Natl. Acad. Sci. USA. 1996;93:11757–11762. doi: 10.1073/pnas.93.21.11757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [118].Tsang WP, Ng EK, Ng SS, Jin H, Yu J, Sung JJ, Kwok TT. Oncofetal H19-derived miR-675 regulates tumor suppressor RB in human colorectal cancer. Carcinogenesis. 2010;31:350–358. doi: 10.1093/carcin/bgp181. [DOI] [PubMed] [Google Scholar]
  • [119].Christofori G, Naik P, Hanahan D. A second signal supplied by insulin-like growth factor II in oncogene-induced tumorigenesis. Nature. 1994;369:414–418. doi: 10.1038/369414a0. [DOI] [PubMed] [Google Scholar]
  • [120].Ariel I, Sughayer M, Fellig Y, Pizov G, Ayesh S, Podeh D, Libdeh BA, Levy C, Birman T, Tykocinski ML, de Groot N, Hochberg A. The imprinted H19 gene is a marker of early recurrence in human bladder carcinoma. Mol. Pathol. 2000;53:320–323. doi: 10.1136/mp.53.6.320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [121].Luo M, Li Z, Wang W, Zeng Y, Liu Z, Qiu J. Long non-coding RNA H19 increases bladder cancer metastasis by associating with EZH2 and inhibiting E-cadherin expression. Cancer Lett. 2013;333:213–221. doi: 10.1016/j.canlet.2013.01.033. [DOI] [PubMed] [Google Scholar]
  • [122].Luo M, Li Z, Wang W, Zeng Y, Liu Z, Qiu J. Upregulated H19 contributes to bladder cancer cell proliferation by regulating ID2 expression. FEBS J. 2013;280:1709–1716. doi: 10.1111/febs.12185. [DOI] [PubMed] [Google Scholar]
  • [123].Guney I, Sedivy JM. Cellular senescence, epigenetic switches and c-Myc. Cell Cycle. 2006;5:2319–2323. doi: 10.4161/cc.5.20.3348. [DOI] [PubMed] [Google Scholar]
  • [124].Barsyte-Lovejoy D, Lau SK, Boutros PC, Khosravi F, Jurisica I, Andrulis IL, Tsao MS, Penn LZ. The c-Myc oncogene directly induces the H19 noncoding RNA by allele-specific binding to potentiate tumorigenesis. Cancer Res. 2006;66:5330–5337. doi: 10.1158/0008-5472.CAN-06-0037. [DOI] [PubMed] [Google Scholar]
  • [125].Lottin S, Adriaenssens E, Dupressoir T, Berteaux N, Montpellier C, Coll J, Dugimont T, Curgy JJ. Overexpression of an ectopic H19 gene enhances the tumorigenic properties of breast cancer cells. Carcinogenesis. 2002;23:1885–1895. doi: 10.1093/carcin/23.11.1885. [DOI] [PubMed] [Google Scholar]
  • [126].Tsang WP, Ng EK, Ng SS, Jin H, Yu J, Sung JJ, Kwok TT. Oncofetal H19-derived miR-675 regulates tumor suppressor RB in human colorectal cancer. Carcinogenesis. 2010;31:350–358. doi: 10.1093/carcin/bgp181. [DOI] [PubMed] [Google Scholar]
  • [127].Zhou Y, Zhong Y, Wang Y, Zhang X, Batista DL, Gejman R, Ansell PJ, Zhao J, Weng C, Klibanski A. Activation of p53 by MEG3 non-coding RNA. J. Biol. Chem. 2007;282:24731–24742. doi: 10.1074/jbc.M702029200. [DOI] [PubMed] [Google Scholar]
  • [128].Braconi C, Kogure T, Valeri N, Huang N, Nuovo G, Costinean S, Negrini M, Miotto E, Croce CM, Patel T. microRNA-29 can regulate expression of the long non-coding RNA gene MEG3 in hepatocellular cancer. Oncogene. 2011;30:4750–4756. doi: 10.1038/onc.2011.193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [129].Zhang X, Zhou YL, Mehta KR, Danila DC, Scolavino S, Johnson SR, Klibanski A. A pituitary-derived MEG3 isoform functions as a growth suppressor in tumor cells. J. Clin. Endocronol. Metab. 2003;88:5119–5126. doi: 10.1210/jc.2003-030222. [DOI] [PubMed] [Google Scholar]
  • [130].Ying L, Huang YR, Chen HG, Wang YW, Xia L, Chen YH, Liu YD, Qiu F. Downregulated MEG3 activates autophagy and increases cell proliferation in bladder cancer. Mol. Biosyst. 2013;9:407–411. doi: 10.1039/c2mb25386k. [DOI] [PubMed] [Google Scholar]
  • [131].Braconi C, Kogure T, Valeri N, Huang N, Nuovo G, Costinean S, Negrini M, Miotto E, Croce CM, Patel T. microRNA-29 can regulate expression of the long non-coding RNA gene MEG3 in hepatocellular cancer. Oncogene. 2011;30:4750–4756. doi: 10.1038/onc.2011.193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [132].Zhang X, Zhou YL, Mehta KR, Danila DC, Scolavino S, Johnson SR, Klibanski A. A pituitary-derived MEG3 isoform functions as a growth suppressor in tumor cells. J. Clin. Endocronol. Metab. 2003;88:5119–5126. doi: 10.1210/jc.2003-030222. [DOI] [PubMed] [Google Scholar]
  • [133].Abdelmohsen K, Panda A, Kang MJ, Xu J, Selimyan R, Yoon JH, Martindale JL, De S, Wood WH, 3rd, Becker KG, Gorospe M. Senescence-associated lncRNAs: senescence-associated long noncoding RNAs. Aging Cell. 2013;12:890–900. doi: 10.1111/acel.12115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [134].Tripathi V, Shen Z, Chakraborty A, Giri S, Freier SM, Wu XL, Zhang YQ, Gorospe M, Prasanth SG, Lal A, Prasanth KV. Long Noncoding RNA MALAT1 Controls Cell Cycle Progression by Regulating the Expression of Oncogenic Transcription Factor B-MYB. PloS Genet. 2013;9:e1003368. doi: 10.1371/journal.pgen.1003368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [135].Tripathi V, Ellis JD, Shen Z, Song DY, Pan Q, Watt AT, Freier SM, Bennett CF, Sharma A, Bubulya PA, Blencowe BJ, Prasanth SG, Prasanth KV. The nuclear-retained noncoding RNA MALAT1 regulates alternative splicing by modulating SR splicing factor phosphorylation. Mol. Cell. 2010;39:925–938. doi: 10.1016/j.molcel.2010.08.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [136].Holly AC, Melzer D, Pilling LC, Fellows AC, Tanaka T, Ferrucci L, Harries LW. Changes in splicing factor expression are associated with advancing age in man. Mech. Ageing Dev. 2013;134:356–366. doi: 10.1016/j.mad.2013.05.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [137].Qi P, Du X. The long non-coding RNAs, a new cancer diagnostic and therapeutic gold mine. Mod. Pathol. 2013;26:155–165. doi: 10.1038/modpathol.2012.160. [DOI] [PubMed] [Google Scholar]
  • [138].Luo JH, Ren B, Keryanov S, Tseng GC, Rao UN, Monga SP, Strom S, Demetris AJ, Nalesnik M, Yu YP, Ranganathan S, Michalopoulos GK. Transcriptomic and genomic analysis of human hepatocellular carcinomas and hepatoblastomas. Hepatology. 2006;44:1012–1024. doi: 10.1002/hep.21328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [139].Peters G. An INKlination for epigenetic control of senescence. Nat. Struct. Mol. Biol. 2008;15:1133–1134. doi: 10.1038/nsmb1108-1133. [DOI] [PubMed] [Google Scholar]
  • [140].Yap KL, Li S, Munoz-Cabello AM, Raguz S, Zeng L, Mujtaba S, Gil J, Walsh MJ, Zhou MM. Molecular interplay of the noncoding RNA ANRIL and methylated histone H3 lysine 27 by polycomb CBX7 in transcriptional silencing of INK4a. Mol. Cell. 2010;38:662–674. doi: 10.1016/j.molcel.2010.03.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [141].Nie F, Sun M, Yang J, Xie M, Xu T, Xia R, Liu Y, Liu X, Zhang E, Lu K, Shu Y. Long Noncoding RNA ANRIL Promotes Non-Small Cell Lung Cancer Cell Proliferation and Inhibits Apoptosis by Silencing KLF2 and P21 Expression. Mol. Cancer Ther. 2014;1:268–277. doi: 10.1158/1535-7163.MCT-14-0492. [DOI] [PubMed] [Google Scholar]
  • [142].Zhang EB, Kong R, Yin DD, You LH, Sun M, Han L, Xu TP, Xia R, Yang JS, De W, Chen J. Long noncoding RNA ANRIL indicates a poor prognosis of gastric cancer and promotes tumor growth by epigenetically silencing of miR-99a/miR-449a. Oncotarget. 2014;5:2276–2292. doi: 10.18632/oncotarget.1902. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [143].Li D, Liu X, Lin L, Hou J, Li N, Wang C, Wang P, Zhang Q, Zhang P, Zhou W, Wang Z, Ding G, Zhuang SM, Zheng L, Tao W, Cao X. MicroRNA-99a inhibits hepatocellular carcinoma growth and correlates with prognosis of patients with hepatocellular carcinoma. J. Biol. Chem. 2011;286:36677–36685. doi: 10.1074/jbc.M111.270561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [144].Noonan EJ, Place RF, Basak S, Pookot D, Li LC. miR-449a causes Rb-dependent cell cycle arrest and senescence in prostate cancer cells. Oncotarget. 2010;1:349–358. doi: 10.18632/oncotarget.167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [145].Rinn RL, Kertesz M, Wang JK, Squazzo SL, Xu X, Brugmann SA, Goodnough LH, Helms JA, Farnham PJ, Segal E, Chang HY. Functional demarcation of active and silent chromatin domains in human HOX loci by noncoding RNAs. Cell. 2007;129:1311–1323. doi: 10.1016/j.cell.2007.05.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [146].Kogo R, Shimamura T, Mimori K, Kawahara K, Imoto S, Sudo T, Tanaka F, Shibata K, Suzuki A, Komune S, Miyano S, Mori M. Long noncoding RNA HOTAIR regulates polycomb-dependent chromatin modification and is associated with poor prognosis in colorectal cancers. Cancer Res. 2011;71:6320–6326. doi: 10.1158/0008-5472.CAN-11-1021. [DOI] [PubMed] [Google Scholar]
  • [147].Zhang JX, Han L, Bao ZS, Wang YY, Chen LY, Yan W, Yu SZ, Pu PY, Liu N, You YP, Jiang T, Kang CS, Chinese Glioma Cooperative G. HOTAIR, a cell cycle-associated long noncoding RNA and a strong predictor of survival, is preferentially expressed in classical and mesenchymal glioma. Neuro-oncology. 2013;15:1595–1603. doi: 10.1093/neuonc/not131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [148].Huang L, Liao LM, Liu AW, Wu JB, Cheng XL, Lin JX, Zheng M. Overexpression of long noncoding RNA HOTAIR predicts a poor prognosis in patients with cervical cancer. Arch. Gynecol. Obstet. 2014;290:717–723. doi: 10.1007/s00404-014-3236-2. [DOI] [PubMed] [Google Scholar]
  • [149].He X, Bao W, Li X, Chen Z, Che Q, Wang H, Wan XP. The long non-coding RNA HOTAIR is upregulated in endometrial carcinoma and correlates with poor prognosis. Int. J. Mol. Med. 2014;33:325–332. doi: 10.3892/ijmm.2013.1570. [DOI] [PubMed] [Google Scholar]
  • [150].Gupta RA, Shah N, Wang KC, Kim J, Horlings HM, Wong DJ, Tsai MC, Hung T, Argani P, Rinn JL, Wang Y, Brzoska P, Kong B, Li R, West RB, van de Vijver MJ, Sukumar S, Chang HY. Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature. 2010;464:1071–1076. doi: 10.1038/nature08975. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [151].Heard E, Disteche CM. Dosage compensation in mammals: fine-tuning the expression of the X chromosome. Genes Dev. 2006;20:1848–1867. doi: 10.1101/gad.1422906. [DOI] [PubMed] [Google Scholar]
  • [152].Vincent-Salomon A, Ganem-Elbaz C, Manie E, Raynal V, Sastre-Garau X, Stoppa-Lyonnet D, Stern MH, Heard E. X inactive-specific transcript RNA coating and genetic instability of the X chromosome in BRCA1 breast tumors. Cancer Res. 2007;67:5134–5140. doi: 10.1158/0008-5472.CAN-07-0465. [DOI] [PubMed] [Google Scholar]
  • [153].Huang KC, Rao PH, Lau CC, Heard E, Ng SK, Brown C, Mok SC, Berkowitz RS, Ng SW. Relationship of XIST expression and responses of ovarian cancer to chemotherapy. Mol. Cancer Ther. 2002;1:769–776. [PubMed] [Google Scholar]
  • [154].Mus E, Hof PR, Tiedge H. Dendritic BC200 RNA in aging and in Alzheimer's disease. Proc. Natl. Acad. Sci. USA. 2007;104:10679–10684. doi: 10.1073/pnas.0701532104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [155].Chen W, Bocker W, Brosius J, Tiedge H. Expression of neural BC200 RNA in human tumours. J. Pathol. 1997;183:345–351. doi: 10.1002/(SICI)1096-9896(199711)183:3<345::AID-PATH930>3.0.CO;2-8. [DOI] [PubMed] [Google Scholar]
  • [156].Iacoangeli A, Lin Y, Morley EJ, Muslimov IA, Bianchi R, Reilly J, Weedon J, Diallo R, Bocker W, Tiedge H. BC200 RNA in invasive and preinvasive breast cancer. Carcinogenesis. 2004;25:2125–2133. doi: 10.1093/carcin/bgh228. [DOI] [PubMed] [Google Scholar]
  • [157].Mourtada-Maarabouni M, Pickard MR, Hedge VL, Farzaneh F, Williams GT. GAS5, a non-protein-coding RNA, controls apoptosis and is downregulated in breast cancer. Oncogene. 2009;28:195–208. doi: 10.1038/onc.2008.373. [DOI] [PubMed] [Google Scholar]
  • [158].Tu ZQ, Li RJ, Mei JZ, Li XH. Down-regulation of long non-coding RNA GAS5 is associated with the prognosis of hepatocellular carcinoma. Int. J. Clin. Exp. Pathol. 2014;7:4303–4309. [PMC free article] [PubMed] [Google Scholar]
  • [159].Augoff K, McCue B, Plow EF, Sossey-Alaoui K. miR-31 and its host gene lncRNA LOC554202 are regulated by promoter hypermethylation in triple-negative breast cancer. Mol. Cancer. 2012;11:5. doi: 10.1186/1476-4598-11-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [160].Dellago H, Preschitz-Kammerhofer B, Terlecki-Zaniewicz L, Schreiner C, Fortschegger K, Chang MW, Hackl M, Monteforte R, Kuhnel H, Schosserer M, Gruber F, Tschachler E, Scheideler M, Grillari-Voglauer R, Grillari J, Wieser M. High levels of oncomiR-21 contribute to the senescence-induced growth arrest in normal human cells and its knock-down increases the replicative lifespan. Aging Cell. 2013;12:446–458. doi: 10.1111/acel.12069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [161].Mitamura T, Watari H, Wang L, Kanno H, Hassan MK, Miyazaki M, Katoh Y, Kimura T, Tanino M, Nishihara H, Tanaka S, Sakuragi N. Downregulation of miRNA-31 induces taxane resistance in ovarian cancer cells through increase of receptor tyrosine kinase MET. Oncogenesis. 2013;2:e40. doi: 10.1038/oncsis.2013.3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [162].Rokah OH, Granot G, Ovcharenko A, Modai S, Pasmanik-Chor M, Toren A, Shomron N, Shpilberg O. Downregulation of miR-31, miR-155, and miR-564 in chronic myeloid leukemia cells. PloS one. 2012;7:e35501. doi: 10.1371/journal.pone.0035501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [163].Korner C, Keklikoglou I, Bender C, Worner A, Munstermann E, Wiemann S. MicroRNA-31 sensitizes human breast cells to apoptosis by direct targeting of protein kinase C epsilon (PKCepsilon) J. Biol. Chem. 2013;288:8750–8761. doi: 10.1074/jbc.M112.414128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [164].Zhong Z, Dong Z, Yang L, Chen X, Gong Z. MicroRNA-31-5p modulates cell cycle by targeting human mutL homolog 1 in human cancer cells. Tumour Biol. 2013;34:1959–1965. doi: 10.1007/s13277-013-0741-z. [DOI] [PubMed] [Google Scholar]
  • [165].Yang G, Lu X, Yuan L. LncRNA: a link between RNA and cancer. Biochim. Biophys. Acta. 2014;1839:1097–1109. doi: 10.1016/j.bbagrm.2014.08.012. [DOI] [PubMed] [Google Scholar]
  • [166].Panzitt K, Tschernatsch MMO, Guelly C, Moustafa T, Stradner M, Strohmaier HM, Buck CR, Denk H, Schroeder R, Trauner M, Zatloukal K. Characterization of HULC, a novel gene with striking up-regulation in hepatocellular carcinoma, as noncoding RNA. Gastroenterology. 2007;132:330–342. doi: 10.1053/j.gastro.2006.08.026. [DOI] [PubMed] [Google Scholar]
  • [167].Wang J, Liu X, Wu H, Ni P, Gu Z, Qiao Y, Chen N, Sun F, Fan Q. CREB up-regulates long non-coding RNA, HULC expression through interaction with microRNA-372 in liver cancer. Nucleic Acids Res. 2010;38:5366–5383. doi: 10.1093/nar/gkq285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [168].Tominaga-Yamanaka K, Abdelmohsen K, Martindale JL, Yang X, Taub DD, Gorospe M. NF90 coordinately represses the senescence-associated secretory phenotype. Aging. 2012;4:695–708. doi: 10.18632/aging.100497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [169].Petrovics G, Zhang W, Makarem M, Street JP, Connelly R, Sun L, Sesterhenn IA, Srikantan V, Moul JW, Srivastava S. Elevated expression of PCGEM1, a prostate-specific gene with cell growth-promoting function, is associated with high-risk prostate cancer patients. Oncogene. 2004;23:605–611. doi: 10.1038/sj.onc.1207069. [DOI] [PubMed] [Google Scholar]
  • [170].Fu X, Ravindranath L, Tran N, Petrovics G, Srivastava S. Regulation of apoptosis by a prostate-specific and prostate cancer-associated noncoding gene. PCGEM1, DNA Cell Biol. 2006;25:135–141. doi: 10.1089/dna.2006.25.135. [DOI] [PubMed] [Google Scholar]
  • [171].Carramusa L, Contino F, Ferro A, Minafra L, Perconti G, Giallongo A, Feo S. The PVT-1 oncogene is a Myc protein target that is overexpressed in transformed cells. J. Cell. Physiol. 2007;213:511–518. doi: 10.1002/jcp.21133. [DOI] [PubMed] [Google Scholar]
  • [172].Guan Y, Kuo WL, Stilwell JL, Takano H, Lapuk AV, Fridlyand J, Mao JH, Yu M, Miller MA, Santos JL, Kalloger SE, Carlson JW, Ginzinger DG, Celniker SE, Mills GB, Huntsman DG, Gray JW. Amplification of PVT1 contributes to the pathophysiology of ovarian and breast cancer. Clin. Cancer Res. 2007;13:5745–5755. doi: 10.1158/1078-0432.CCR-06-2882. [DOI] [PubMed] [Google Scholar]
  • [173].Merksamer PI, Liu YF, He WJ, Hirschey MD, Chen D, Verdin E. The sirtuins, oxidative stress and aging: an emerging link. Aging-US. 2013;5:144–150. doi: 10.18632/aging.100544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [174].Sohal RS, Weindruch R. Oxidative stress, caloric restriction, and aging. Science. 1996;273:59–63. doi: 10.1126/science.273.5271.59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [175].Huarte M, Guttman M, Feldser D, Garber M, Koziol MJ, Kenzelmann-Broz D, Khalil AM, Zuk O, Amit I, Rabani M, Attardi LD, Regev A, Lander ES, Jacks T, Rinn JL. A large intergenic noncoding RNA induced by p53 mediates global gene repression in the p53 response. Cell. 2010;142:409–419. doi: 10.1016/j.cell.2010.06.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [176].Huarte M, Rinn JL. Large non-coding RNAs: missing links in cancer? Hum. Mol. Genet. 2010;19:R152–161. doi: 10.1093/hmg/ddq353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [177].Yoon JH, Abdelmohsen K, Srikantan S, Yang X, Martindale JL, De S, Huarte M, Zhan M, Becker KG, Gorospe M. LincRNA-p21 suppresses target mRNA translation. Mol. Cell. 2012;47:648–655. doi: 10.1016/j.molcel.2012.06.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [178].Zhang DY, Wang HJ, Tan YZ. Wnt/beta-catenin signaling induces the aging of mesenchymal stem cells through the DNA damage response and the p53/p21 pathway. PloS One. 2011;6:e21397. doi: 10.1371/journal.pone.0021397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [179].Piechaczyk M, Farrás R. Regulation and function of JunB in cell proliferation. Biochem Soc Trans. 2008;36:864–867. doi: 10.1042/BST0360864. [DOI] [PubMed] [Google Scholar]
  • [180].Marin-Bejar O, Marchese FP, Athie A, Sanchez Y, Gonzalez J, Segura V, Huang L, Moreno I, Navarro A, Monzo M, Garcia-Foncillas J, Rinn JL, Guo S, Huarte M. Pint lincRNA connects the p53 pathway with epigenetic silencing by the Polycomb repressive complex 2. Genome Biol. 2013;14:R104. doi: 10.1186/gb-2013-14-9-r104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [181].Hung T, Wang Y, Lin MF, Koegel AK, Kotake Y, Grant GD, Horlings HM, Shah N, Umbricht C, Wang P, Wang Y, Kong B, Langerød A, Børresen-Dale AL, Kim SK, van de Vijver M, Sukumar S, Whitfield ML, Kellis M, Xiong Y, Wong DJ, Chang HY. Extensive and coordinated transcription of noncoding RNAs within cell-cycle promoters. Nat. Genet. 2011;43:621–629. doi: 10.1038/ng.848. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [182].Puvvula PK, Desetty RD, Pineau P, Marchio A, Moon A, Dejean A, Bischof O. Long noncoding RNA PANDA and scaffold-attachment-factor SAFA control senescence entry and exit. Nat. Commun. 2014;5:5323. doi: 10.1038/ncomms6323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [183].Wan G, Hu X, Liu Y, Han C, Sood AK, Calin GA, Zhang X, Lu X. A novel non-coding RNA lncRNA-JADE connects DNA damage signalling to histone H4 acetylation. EMBO J. 2013;32:2833–2847. doi: 10.1038/emboj.2013.221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [184].Lowndes NF. The interplay between BRCA1 and 53BP1 influences death, aging, senescence and cancer. DNA Repair. 2010;9:1112–1116. doi: 10.1016/j.dnarep.2010.07.012. [DOI] [PubMed] [Google Scholar]
  • [185].Zhang A, Zhou N, Huang J, Liu Q, Fukuda K, Ma D, Lu Z, Bai C, Watabe K, Mo YY. The human long non-coding RNA-RoR is a p53 repressor in response to DNA damage. Cell Res. 2013;23:340–350. doi: 10.1038/cr.2012.164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [186].Silva JM, Perez DS, Pritchett JR, Hailing ML, Tang H, Smith DI. Identification of Long stress-induced non-coding transcripts that have altered expression in cancer. Genomics. 2010;95:355–362. doi: 10.1016/j.ygeno.2010.02.009. [DOI] [PubMed] [Google Scholar]
  • [187].Coppe JP, Desprez PY, Krtolica A, Campisi J. The senescence-associated secretory phenotype: the dark side of tumor suppression. Annu. Rev. Pathol. 2010;5:99–118. doi: 10.1146/annurev-pathol-121808-102144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [188].Boraschi D, Aguado MT, Dutel C, Goronzy J, Louis J, Grubeck-Loebenstein B, Rappuoli R, Del Giudice G. The Gracefully Aging Immune System. Sci. Transl. Med. 2013;5 doi: 10.1126/scitranslmed.3005624. [DOI] [PubMed] [Google Scholar]
  • [189].Wei N, Pang WJ, Wang Y, Xiong Y, Xu RX, Wu WJ, Zhao CZ, Yang GS. Knockdown of PU.1 mRNA and AS lncRNA regulates expression of immune-related genes in zebrafish Danio rerio. Dev. Comp. Immunol. 2014;44:315–319. doi: 10.1016/j.dci.2014.01.015. [DOI] [PubMed] [Google Scholar]
  • [190].Carpenter S, Aiello D, Atianand MK, Ricci EP, Gandhi P, Hall LL, Byron M, Monks B, Henry-Bezy M, Lawrence JB, O'Neill LA, Moore MJ, Caffrey DR, Fitzgerald KA. A long noncoding RNA mediates both activation and repression of immune response genes. Science. 2013;341:789–792. doi: 10.1126/science.1240925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [191].Tilstra JS, Clauson CL, Niedernhofer LJ, Robbins PD. NF-kappaB in Aging and Disease. Aging Dis. 2011;2:449–465. [PMC free article] [PubMed] [Google Scholar]
  • [192].Elife . p50-associated COX-2 extragenic RNA (PACER) activates COX-2 gene expression by occluding repressive NF-κB complexes. Vol. 3. Emerson BM2; Krawczyk M1: Apr 29, 2014. p. e01776. doi: 10.7554/eLife.01776. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [193].Ratliff M, Alter S, Frasca D, Blomberg BB, Riley RL. In senescence, age-associated B cells secrete TNFalpha and inhibit survival of B-cell precursors. Aging Cell. 2013;12:303–311. doi: 10.1111/acel.12055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [194].Rapicavoli NA, Qu K, Zhang J, Mikhail M, Laberge RM, Chang HY. A mammalian pseudogene lncRNA at the interface of inflammation and anti-inflammatory therapeutics. eLife. 2013;2:e00762. doi: 10.7554/eLife.00762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [195].Li ZH, Chao TC, Chang KY, Lin NW, Patil VS, Shimizu C, Head SR, Burns JC, Rana TM. The long noncoding RNA THRIL regulates TNF alpha expression through its interaction with hnRNPL, Proc. Natl. Acad. Sci. USA. 2014;111:1002–1007. doi: 10.1073/pnas.1313768111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [196].Imamura K, Imamachi N, Akizuki G, Kumakura M, Kawaguchi A, Nagata K, Kato A, Kawaguchi Y, Sato H, Yoneda M, Kai C, Yada T, Suzuki Y, Yamada T, Ozawa T, Kaneki K, Inoue T, Kobayashi M, Kodama T, Wada Y, Sekimizu K, Akimitsu N. Long noncoding RNA NEAT1-dependent SFPQ relocation from promoter region to paraspeckle mediates IL8 expression upon immune stimuli. Mol. Cell. 2014;53:393–406. doi: 10.1016/j.molcel.2014.01.009. [DOI] [PubMed] [Google Scholar]
  • [197].Clark JA, Peterson TC. Cytokine production and aging: overproduction of IL-8 in elderly males in response to lipopolysaccharide. Mech. Ageing Dev. 1994;77:127–139. doi: 10.1016/0047-6374(94)90020-5. [DOI] [PubMed] [Google Scholar]
  • [198].Baune BT, Ponath G, Golledge J, Varga G, Arolt V, Rothermundt M, Berger K. Association between IL-8 cytokine and cognitive performance in an elderly general population the MEMO-Study. Neurobiol. Aging. 2008;29:937–944. doi: 10.1016/j.neurobiolaging.2006.12.003. [DOI] [PubMed] [Google Scholar]
  • [199].Wang P, Xue Y, Han Y, Lin L, Wu C, Xu S, Jiang Z, Xu J, Liu Q, Cao X. The STAT3-binding long noncoding RNA lnc-DC controls human dendritic cell differentiation. Science. 2014;344:310–313. doi: 10.1126/science.1251456. [DOI] [PubMed] [Google Scholar]
  • [200].Kida H, Ihara S, Kumanogoh A. Involvement of STAT3 in immune evasion during lung tumorigenesis. Oncoimmunology. 2013;2:e22653. doi: 10.4161/onci.22653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [201].Gray GK, McFarland BC, Nozell SE, Benveniste EN. NF-kappaB and STAT3 in glioblastoma: therapeutic targets coming of age. Expert Rev. Neurother. 2014;14:1293–1306. doi: 10.1586/14737175.2014.964211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [202].Gomez JA, Wapinski OL, Yang YW, Bureau JF, Gopinath S, Monack DM, Chang HY, Brahic M, Kirkegaard K. The NeST Long ncRNA Controls Microbial Susceptibility and Epigenetic Activation of the Interferon-gamma Locus. Cell. 2013;152:743–754. doi: 10.1016/j.cell.2013.01.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [203].Bureau JF, Montagutelli X, Bihl F, Lefebvre S, Guenet JL, Brahic M. Mapping loci influencing the persistence of Theiler's virus in the murine central nervous system. Nat Genet. 1993;5:87–91. doi: 10.1038/ng0993-87. [DOI] [PubMed] [Google Scholar]
  • [204].Bihl F, Brahic M, Bureau JF. Two loci, Tmevp2 and Tmevp3, located on the telomeric region of chromosome 10, control the persistence of Theiler's virus in the central nervous system of mice. Genetics. 1999;152:385–392. doi: 10.1093/genetics/152.1.385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [205].Oxenkrug G. Interferon-gamma - Inducible Inflammation: Contribution to Aging and Aging-Associated Psychiatric Disorders. Aging Dis. 2011;2:474–486. [PMC free article] [PubMed] [Google Scholar]
  • [206].Ng SY, Lin L, Soh BS, Stanton LW. Long noncoding RNAs in development and disease of the central nervous system. Trends Genet. 2013;29:461–468. doi: 10.1016/j.tig.2013.03.002. [DOI] [PubMed] [Google Scholar]
  • [207].Faghihi MA, Modarresi F, Khalil AM, Wood DE, Sahagan BG, Morgan TE, Finch CE, Laurent GS, Kenny PJ, Wahlestedt C. Expression of a noncoding RNA is elevated in Alzheimer's disease and drives rapid feed-forward regulation of beta-secretase. Nat. Med. 2008;14:723–730. doi: 10.1038/nm1784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [208].Faghihi MA, Zhang M, Huang J, Modarresi F, Van der Brug MP, Nalls MA, Cookson MR, St-Laurent G, Wahlestedt C. Evidence for natural antisense transcript-mediated inhibition of microRNA function. Genome Biol. 2010;11 doi: 10.1186/gb-2010-11-5-r56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [209].Modarresi F, Faghihi MA, Patel NS, Sahagan BG, Wahlestedt C, Lopez-Toledano MA. Knockdown of BACE1-AS Nonprotein-Coding Transcript Modulates Beta-Amyloid-Related Hippocampal Neurogenesis. Int. J. Alzheimers Dis. 2011;2011:929042. doi: 10.4061/2011/929042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [210].Fukumoto H, Cheung BS, Hyman BT, Irizarry MC. beta-secretase protein and activity are increased in the neocortex in Alzheimer disease. Arch. Neurol. 2002;59:1381–1389. doi: 10.1001/archneur.59.9.1381. [DOI] [PubMed] [Google Scholar]
  • [211].Massone S, Vassallo I, Fiorino G, Castelnuovo M, Barbieri F, Borghi R, Tabaton M, Robello M, Gatta E, Russo C, Florio T, Dieci G, Cancedda R, Pagano A. 17A, a novel non-coding RNA, regulates GABA B alternative splicing and signaling in response to inflammatory stimuli and in Alzheimer disease. Neurobiol. Dis. 2011;41:308–317. doi: 10.1016/j.nbd.2010.09.019. [DOI] [PubMed] [Google Scholar]
  • [212].Barry G, Briggs JA, Vanichkina DP, Poth EM, Beveridge NJ, Ratnu VS, Nayler SP, Nones K, Hu J, Bredy TW, Nakagawa S, Rigo F, Taft RJ, Cairns MJ, Blackshaw S, Wolvetang EJ, Mattick JS. The long non-coding RNA Gomafu is acutely regulated in response to neuronal activation and involved in schizophrenia-associated alternative splicing. Mol. Psychiatry. 2014;19:486–494. doi: 10.1038/mp.2013.45. [DOI] [PubMed] [Google Scholar]
  • [213].Kimbro LB, Mangione CM, Steers WN, Duru OK, McEwen L, Karter A, Ettner SL. Depression and All-Cause Mortality in Persons with Diabetes Mellitus: Are Older Adults at Higher Risk? Results from the Translating Research Into Action for Diabetes Study. J. Am. Geriatr. Soc. 2014;62:1017–1022. doi: 10.1111/jgs.12833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [214].Thomson PA, MacIntyre DJ, Hamilton G, Dominiczak A, Smith BH, Morris A, Evans KL, Porteous DJ. Association of DISC1 variants with age of onset in a population-based sample of recurrent major depression. Mol. Psychiatry. 2013;18:745–747. doi: 10.1038/mp.2012.117. [DOI] [PubMed] [Google Scholar]
  • [215].Yu B, Zhou S, Hu W, Qian T, Gao R, Ding G, Ding F, Gu X. Altered long noncoding RNA expressions in dorsal root ganglion after rat sciatic nerve injury. Neurosci. Lett. 2013;534:117–122. doi: 10.1016/j.neulet.2012.12.014. [DOI] [PubMed] [Google Scholar]
  • [216].Ng SY, Bogu GK, Soh BS, Stanton LW. The long noncoding RNA RMST interacts with SOX2 to regulate neurogenesis. Mol. Cell. 2013;51:349–359. doi: 10.1016/j.molcel.2013.07.017. [DOI] [PubMed] [Google Scholar]
  • [217].Kim HJ, Kim NC, Wang YD, Scarborough EA, Moore J, Diaz Z, MacLea KS, Freibaum B, Li S, Molliex A, Kanagaraj AP, Carter R, Boylan KB, Wojtas AM, Rademakers R, Pinkus JL, Greenberg SA, Trojanowski JQ, Traynor BJ, Smith BN, Topp S, Gkazi AS, Miller J, Shaw CE, Kottlors M, Kirschner J, Pestronk A, Li YR, Ford AF, Gitler AD, Benatar M, King OD, Kimonis VE, Ross ED, Shorter C.C Weihl, J., Taylor JP. Mutations in prion-like domains in hnRNPA2B1 and hnRNPA1 cause multisystem proteinopathy and ALS. Nature. 2013;495:467–473. doi: 10.1038/nature11922. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [218].Meier I, Fellini L, Jakovcevski M, Schachner M, Morellini F. Expression of the snoRNA Host Gene gas5 in the Hippocampus Is Upregulated by Age and Psychogenic Stress and Correlates with Reduced Novelty-Induced Behavior in C57BL/6 Mice. Hippocampus. 2010;20:1027–1036. doi: 10.1002/hipo.20701. [DOI] [PubMed] [Google Scholar]
  • [219].Mourtada-Maarabouni M, Pickard MR, Hedge VL, Farzaneh F, Williams GT. GAS5, a non-protein-coding RNA, controls apoptosis and is downregulated in breast cancer. Oncogene. 2009;28:195–208. doi: 10.1038/onc.2008.373. [DOI] [PubMed] [Google Scholar]
  • [220].Han P, Li W, Lin CH, Yang J, Shang C, Nurnberg ST, Jin KK, Xu W, Lin CY, Lin CJ, Xiong Y, Chien HC, Zhou B, Ashley E, Bernstein D, Chen PS, Chen HS, Quertermous T, Chang CP. A long noncoding RNA protects the heart from pathological hypertrophy. Nature. 2014;514:102–106. doi: 10.1038/nature13596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [221].Yildirim E, Kirby JE, Brown DE, Mercier FE, Sadreyev RI, Scadden DT, Lee JT. Xist RNA is a potent suppressor of hematologic cancer in mice. Cell. 2013;152:727–742. doi: 10.1016/j.cell.2013.01.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [222].Lin N, Chang KY, Li Z, Gates K, Rana ZA, Dang J, Zhang D, Han T, Yang CS, Cunningham TJ, Head SR, Duester G, Dong PD, Rana TM. An evolutionarily conserved long noncoding RNA TUNA controls pluripotency and neural lineage commitment. Mol Cell. 2014;53:1005–1019. doi: 10.1016/j.molcel.2014.01.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [223].Ulitsky I, Shkumatava A, Jan CH, Sive H, Bartel DP. Conserved function of lincRNAs in vertebrate embryonic development despite rapid sequence evolution. Cell. 2011;147:1537–1550. doi: 10.1016/j.cell.2011.11.055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [224].Takahashi K, Yan IK, Kogure T, Haga H, Patel T. Extracellular vesicle-mediated transfer of long non-coding RNA ROR modulates chemosensitivity in human hepatocellular cancer. FEBS Open Bio. 2014;4:458–467. doi: 10.1016/j.fob.2014.04.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [225].Bazzini AA, Johnstone TG, Christiano R, Mackowiak SD, Obermayer B, Fleming ES, Vejnar CE, Lee MT, Rajewsky N, Walther TC, Giraldez AJ. Identification of small ORFs in vertebrates using ribosome footprinting and evolutionary conservation. EMBO J. 2014;33:981–993. doi: 10.1002/embj.201488411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [226].Anderson DM, Anderson KM, Chang CL, Makarewich CA, Nelson BR, McAnally JR, Kasaragod P, Shelton JM, Liou J, Bassel-Duby R, Olson EN. A micropeptide encoded by a putative long noncoding RNA regulates muscle performance. Cell. 2015;160:595–606. doi: 10.1016/j.cell.2015.01.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [227].Slavoff SA, Mitchell AJ, Schwaid AG, Cabili MN, Ma J, Levin JZ, Karger AD, Budnik BA, Rinn JL, Saghatelian A. Peptidomic discovery of short open reading frame-encoded peptides in human cells. Nat. Chem. Biol. 2013;9:59–64. doi: 10.1038/nchembio.1120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [228].Andrews SJ, Rothnagel JA. Emerging evidence for functional peptides encoded by short open reading frames. Nat. Rev. Genet. 2014;15:193–204. doi: 10.1038/nrg3520. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

NIHMS709002-supplement.docx (130.9KB, docx)

RESOURCES