Editorial Open Access
Copyright ©The Author(s) 2024. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Meta-Anal. Sep 18, 2024; 12(3): 97757
Published online Sep 18, 2024. doi: 10.13105/wjma.v12.i3.97757
Role of long non-coding RNAs in non-alcoholic fatty liver disease
Anju Mullath, Department of Gastroenterology, Aster Royal Hospital, Muscat 133, Muscat, Oman
Murali Krishna, Department of Urology, Army Hospital, R & R, Delhi Cantonment 110010, Delhi, India
ORCID number: Anju Mullath (0000-0003-0813-2203); Murali Krishna (0000-0002-9590-5798).
Author contributions: Mullath A conceptualized and researched the study; Krishna M contributed to the research and wrote the manuscript.
Conflict-of-interest statement: All the authors declare that they have no conflict of interest.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Murali Krishna, DNB, MBBS, MD, Assistant Professor, Surgeon, Department of Urology, Army Hospital, R & R, Dhaula Kuan, New Delhi, Delhi Cantonment 110010, Delhi, India. murali276@yahoo.com
Received: June 7, 2024
Revised: August 6, 2024
Accepted: August 15, 2024
Published online: September 18, 2024
Processing time: 98 Days and 3 Hours

Abstract

Non-alcoholic fatty liver disease (NAFLD) is emerging as a common cause of chronic liver disease in children and adults. NAFLD can progress to steatohepatitis and potentially even hepatocellular carcinoma. Early identification of patients at risk for progressive disease is crucial for managing NAFLD. Recent studies have identified long noncoding RNAs (lncRNAs), circular RNAs, and microRNAs as playing important roles in the pathogenesis of NAFLD. These noncoding RNAs are involved in modulating several metabolic pathways such as hepatic glucose and lipid metabolism, oxidative stress, and even carcinogenesis. Elevated levels of lncARSR and lncRNA nuclear-enriched abundant transcript 1 have been found in patients with NAFLD. In addition, lncRNAs such as PRYP4-3 and RP11-128N14.5 can distinguish patients with NAFLD from healthy individuals. Increased MEG3 expression has been observed in both NAFLD and non-alcoholic steatohepatitis, suggesting that it may help predict patients at risk for disease progression. With advances in transcriptomics, we may discover additional targets to help in the identification and prognostication of NAFLD.

Key Words: Long noncoding RNA; Non-alcoholic fatty liver disease; Plasmacytoma variant translocation 1; Nuclear-enriched abundant transcript 1; Muscle- and adipose-associated long intergenic non-coding RNA; H19

Core Tip: Non-alcoholic fatty liver disease (NAFLD) is one of the most common liver ailments. Early diagnosis and treatment can help mitigate its impact on the liver. The role of long non-coding RNAs (lncRNAs) in the pathogenesis of NAFLD has been the subject of research for some time. LncRNAs such as plasmacytoma variant translocation 1, nuclear-enriched abundant transcript 1, muscle- and adipose-associated long intergenic non-coding RNA, and H19 have been shown to play important roles in the disease process of NAFLD.



INTRODUCTION

Non-alcoholic fatty liver disease (NAFLD) has emerged as one of the most prevalent causes of chronic liver disease globally. However, a lack of awareness surrounding this seemingly benign condition exacerbates the issue. NAFLD frequently manifests as part of metabolic syndrome, which includes obesity and type 2 diabetes[1]. The spectrum of NAFLD encompasses non-alcoholic steatohepatitis, cirrhosis, and hepatocellular carcinoma.

Long non-coding RNAs (lncRNAs) are transcripts exceeding 200 nucleotides and are intricately involved in various functions such as post-transcriptional regulation and epigenetic modifications. Recent research has uncovered the role lncRNAs play in several disease processes. These molecules hold promise as agents for diagnosis and prognostication in the near future.

ROLE OF LNCRNA IN NAFLD

Among non-coding RNAs, lncRNAs are believed to play a crucial role in regulating hepatic gluconeogenesis, inflammation, hepatic regeneration, and fibrosis. Several candidates have been identified thus far, including plasmacytoma variant translocation 1 (PVT1), nuclear-enriched abundant transcript 1 (NEAT1), PRYP4.3, RP 11-128N14.5, HCG18, and MEG3[2]. In this editorial, we will delve into four promising markers that may potentially revolutionize the diagnosis and management of NAFLD in the future.

PVT1

LncRNA PVT1 has garnered significant attention in the context of NAFLD owing to its multifaceted roles in hepatic pathophysiology. Studies have elucidated its involvement in diverse biological processes, including hepatic lipid metabolism, inflammation, and fibrosis, all of which are central to the progression of NAFLD. The dysregulation of PVT1 has been implicated in exacerbating liver injury and fibrosis severity in patients with NAFLD[3]. Moreover, elevated PVT1 expression levels have been correlated with the presence of steatosis and the degree of liver inflammation in NAFLD cohorts[4]. PVT1 affects liver fat metabolism by reducing the levels of microRNA (miR)-20a-5p. Mir-20a-5p acts by suppressing the expression of CD36, which, in turn, reduces lipid accumulation by binding to the 3′-untranslated region[5]. CD36 plays a crucial role in regulating inflammation, fatty acid oxidation, and intracellular fatty acid homeostasis.

Emerging evidence suggests that PVT1 holds promise as a diagnostic and prognostic biomarker for NAFLD. Its overexpression in hepatic tissues and circulation has been proposed as a potential indicator of the onset and progression of NAFLD[6]. Additionally, aberrant expression of PVT1 has been associated with adverse clinical outcomes, including liver cirrhosis and hepatocellular carcinoma, further underscoring its significance in the pathogenesis of NAFLD[7].

Targeting PVT1 presents a novel therapeutic avenue for managing NAFLD. Inhibition of PVT1 expression has shown promise in ameliorating hepatic steatosis, inflammation, and fibrosis in preclinical models, suggesting its potential as a therapeutic target for NAFLD intervention[8].

NEAT1

LncRNA NEAT1 has emerged as a pivotal player in the pathogenesis of NAFLD. Dysregulation of NEAT1 has been implicated in various aspects of the progression of NAFLD, including hepatic lipid metabolism, inflammation, and fibrosis[9]. Studies have revealed elevated NEAT1 expression levels in the liver tissues and circulation of patients with NAFLD, suggesting its potential as a diagnostic biomarker for the disease[10].

NEAT1 exerts its effects through diverse mechanisms, including the regulation of gene expression, chromatin organization, and nuclear structure maintenance[11]. In NAFLD, NEAT1 has been shown to modulate key signaling pathways involved in lipid accumulation, hepatic steatosis, and inflammation, thereby contributing to the pathogenesis of the disease[12]. NEAT1 Levels were found to be increased in patients with NAFLD, and higher NEAT1 Levels correlated with reduced levels of miR-212-5p. This miRNA is known to inhibit the activity of fatty acid synthase, and its inhibition has been shown to result in triglyceride accumulation in mouse primary hepatocytes[13].

Furthermore, NEAT1 has been implicated in promoting hepatic fibrosis through its interaction with various proteins and miRNAs, highlighting its multifaceted role in the progression of NAFLD[14]. Given its intricate involvement in the pathophysiology of NAFLD, NEAT1 represents a promising therapeutic target for developing novel interventions aimed at mitigating disease progression and improving clinical outcomes.

MUSCLE- AND ADIPOSE-ASSOCIATED LONG INTERGENIC NON-CODING RNA (MAYA)

LncRNA MAYA has emerged as a novel regulator of metabolic disorders, including NAFLD. The expression of MAYA is dysregulated in patients with NAFLD, suggesting its potential role in disease pathogenesis[15]. Recent studies have implicated MAYA in modulating lipid metabolism, insulin sensitivity, and inflammation in hepatic tissues, thereby contributing to the development and progression and NAFLD[15]. MAYA regulates the Hippo signaling pathway, which, when activated, leads to the repression of downstream transcriptional co-activators YAP and TAZ. The Hippo signaling pathway also plays a role in liver fibrosis.

Additionally, MAYA has been proposed as a diagnostic biomarker for NAFLD owing to its differential expression patterns in liver tissues and circulation of patients with NAFLD compared to healthy controls[16]. Further elucidation of the molecular mechanisms and functional significance of MAYA may unveil novel therapeutic targets for NAFLD intervention.

H19

LncRNA H19 has garnered considerable attention in the realm of NAFLD owing to its regulatory roles in hepatic lipid metabolism and inflammation. H19 expression levels are dysregulated in patients with NAFLD, suggesting its involvement in disease pathogenesis[17]. Mechanistically, H19 has been implicated in modulating key signaling pathways related to lipid accumulation, hepatic steatosis, and inflammation in the liver[18]. H19 downregulates the expression of miR-130a, which further inhibits the expression of PPARγ, and promotes steatosis by upregulating the transcription factor MLXIPL (also known as ChREBP)[17]. Furthermore, aberrant H19 expression has been correlated with the severity of liver injury and fibrosis in NAFLD cohorts[19]. Given its potential as a diagnostic and prognostic biomarker, further investigation into the functional significance of H19 in the pathophysiology of NAFLD may unveil novel therapeutic targets for disease intervention.

Table 1 provides a brief overview of other lncRNAs that are upregulated or downregulated in NAFLD[2].

Table 1 Various long non-coding RNAs implicated in non-alcoholic fatty liver disease.
Gene
Principal functions
Upregulated/downregulated
Gm9795Endoplasmic reticulum stress
Promoting inflammatory response
Upregulated
Platr4Promoting inflammatory responseUpregulated
HOTAIRPromoting lipid accumulationUpregulated
LncTNFPromoting inflammationUpregulated
Gm15622Promoting lipid accumulationUpregulated
LncHR1Preventing the accumulation of fatty acids and triglycerideDownregulated
SRD5A3-AS1Promoting cell proliferation, steatosis, inflammation and fibrosisDownregulated
Gm16551Promoting de novo lipogenesisDownregulated
AC012668Promoting lipid accumulationDownregulated
Mirt2Promoting hepatic steatosisDownregulated
CONCLUSION

NAFLD is emerging as the leading liver ailment with far-reaching effects on patient morbidity and mortality. Early markers for diagnosis and prognostication will help in design and implementation of appropriate steps to ameliorate the condition. LncRNAs can play an important role in both these aspects, and further research is required to improve our understanding of their functions and potential applications.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country of origin: India

Peer-review report’s classification

Scientific Quality: Grade C, Grade D

Novelty: Grade B, Grade C

Creativity or Innovation: Grade B, Grade C

Scientific Significance: Grade B, Grade C

P-Reviewer: Pan Y S-Editor: Liu JH L-Editor: A P-Editor: Wang WB

References
1.  Vilar-Gomez E. NAFLD and liver-related events: does type 2 diabetes have a key role? Lancet Gastroenterol Hepatol. 2023;8:777-778.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
2.  Shi N, Sun K, Tang H, Mao J. The impact and role of identified long noncoding RNAs in nonalcoholic fatty liver disease: A narrative review. J Clin Lab Anal. 2023;37:e24943.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
3.  Yu F, Dong B, Dong P, He Y, Zheng J, Xu P. Hypoxia induces the activation of hepatic stellate cells through the PVT1-miR-152-ATG14 signaling pathway. Mol Cell Biochem. 2020;465:115-123.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 12]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
4.  Yi K, Hou M, Yuan J, Yang L, Zeng X, Xi M, Chen J. LncRNA PVT1 epigenetically stabilizes and post-transcriptionally regulates FOXM1 by acting as a microRNA sponge and thus promotes malignant behaviors of ovarian cancer cells. Am J Transl Res. 2020;12:2860-2874.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Wang X, Ma Y, Yang LY, Zhao D. MicroRNA-20a-5p Ameliorates Non-alcoholic Fatty Liver Disease via Inhibiting the Expression of CD36. Front Cell Dev Biol. 2020;8:596329.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 24]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
6.  Li S, Zhao X, Cheng S, Li J, Bai X, Meng X. Downregulating long non-coding RNA PVT1 expression inhibited the viability, migration and phenotypic switch of PDGF-BB-treated human aortic smooth muscle cells via targeting miR-27b-3p. Hum Cell. 2021;34:335-348.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 17]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
7.  Yan M, Pan XF, Liu Y, Zhao S, Gong WQ, Liu W. Long noncoding RNA PVT1 promotes metastasis via miR-484 sponging in osteosarcoma cells. Eur Rev Med Pharmacol Sci. 2020;24:2229-2238.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 7]  [Reference Citation Analysis (0)]
8.  Zhu Y, Liu Y, Xiao B, Cai H, Liu M, Ma L, Yin H, Wang F. The circular RNA PVT1/miR-203/HOXD3 pathway promotes the progression of human hepatocellular carcinoma. Biol Open. 2019;8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 29]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
9.  Bu FT, Wang A, Zhu Y, You HM, Zhang YF, Meng XM, Huang C, Li J. LncRNA NEAT1: Shedding light on mechanisms and opportunities in liver diseases. Liver Int. 2020;40:2612-2626.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 42]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
10.  Ye J, Lin Y, Yu Y, Sun D. LncRNA NEAT1/microRNA-129-5p/SOCS2 axis regulates liver fibrosis in alcoholic steatohepatitis. J Transl Med. 2020;18:445.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 45]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
11.  Dong Y, Hu M, Tan K, Dai R. ZNF143 inhibits hepatocyte mitophagy and promotes non-alcoholic fatty liver disease by targeting increased lncRNA NEAT1 expression to activate ROCK2 pathway. Epigenetics. 2023;18:2239592.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
12.  Nguyen TM, Kabotyanski EB, Reineke LC, Shao J, Xiong F, Lee JH, Dubrulle J, Johnson H, Stossi F, Tsoi PS, Choi KJ, Ellis AG, Zhao N, Cao J, Adewunmi O, Ferreon JC, Ferreon ACM, Neilson JR, Mancini MA, Chen X, Kim J, Ma L, Li W, Rosen JM. The SINEB1 element in the long non-coding RNA Malat1 is necessary for TDP-43 proteostasis. Nucleic Acids Res. 2020;48:2621-2642.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 39]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
13.  Zhu Y, Tan JK, Wong SK, Goon JA. Therapeutic Effects of microRNAs on Nonalcoholic Fatty Liver Disease (NAFLD) and Nonalcoholic Steatohepatitis (NASH): A Systematic Review and Meta-Analysis. Int J Mol Sci. 2023;24.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
14.  Huang S, Xu Y, Ge X, Xu B, Peng W, Jiang X, Shen L, Xia L. Long noncoding RNA NEAT1 accelerates the proliferation and fibrosis in diabetic nephropathy through activating Akt/mTOR signaling pathway. J Cell Physiol. 2019;234:11200-11207.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 82]  [Article Influence: 13.7]  [Reference Citation Analysis (0)]
15.  Yuan P, Qi X, Song A, Ma M, Zhang X, Lu C, Bian M, Lian N, He J, Zheng S, Jin H. LncRNA MAYA promotes iron overload and hepatocyte senescence through inhibition of YAP in non-alcoholic fatty liver disease. J Cell Mol Med. 2021;25:7354-7366.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 19]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
16.  Liu L, An X, Li Z, Song Y, Li L, Zuo S, Liu N, Yang G, Wang H, Cheng X, Zhang Y, Yang X, Wang J. The H19 long noncoding RNA is a novel negative regulator of cardiomyocyte hypertrophy. Cardiovasc Res. 2016;111:56-65.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 164]  [Cited by in F6Publishing: 174]  [Article Influence: 21.8]  [Reference Citation Analysis (0)]
17.  Yang Z, Zhang T, Han S, Kusumanchi P, Huda N, Jiang Y, Liangpunsakul S. Long noncoding RNA H19 - a new player in the pathogenesis of liver diseases. Transl Res. 2021;230:139-150.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 19]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
18.  Wang Y, Tai YL, Way G, Zeng J, Zhao D, Su L, Jiang X, Jackson KG, Wang X, Gurley EC, Liu J, Liu J, Chen W, Wang XY, Sanyal AJ, Hylemon PB, Zhou H. RNA binding protein HuR protects against NAFLD by suppressing long noncoding RNA H19 expression. Cell Biosci. 2022;12:172.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Reference Citation Analysis (0)]
19.  Nascimbeni F, Dalla Salda A, Carubbi F. Energy balance, glucose and lipid metabolism, cardiovascular risk and liver disease burden in adult patients with type 1 Gaucher disease. Blood Cells Mol Dis. 2018;68:74-80.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 21]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]