Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 1815 2022-11-02 09:54:59 |
2 format correct -19 word(s) 1796 2022-11-03 02:36:08 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Durge, A.;  Sharma, I.;  Tupe, R.S. Glycation-Associated Diabetic Nephropathy and Long Noncoding RNAs. Encyclopedia. Available online: https://encyclopedia.pub/entry/32491 (accessed on 03 July 2024).
Durge A,  Sharma I,  Tupe RS. Glycation-Associated Diabetic Nephropathy and Long Noncoding RNAs. Encyclopedia. Available at: https://encyclopedia.pub/entry/32491. Accessed July 03, 2024.
Durge, Ankita, Isha Sharma, Rashmi Santosh Tupe. "Glycation-Associated Diabetic Nephropathy and Long Noncoding RNAs" Encyclopedia, https://encyclopedia.pub/entry/32491 (accessed July 03, 2024).
Durge, A.,  Sharma, I., & Tupe, R.S. (2022, November 02). Glycation-Associated Diabetic Nephropathy and Long Noncoding RNAs. In Encyclopedia. https://encyclopedia.pub/entry/32491
Durge, Ankita, et al. "Glycation-Associated Diabetic Nephropathy and Long Noncoding RNAs." Encyclopedia. Web. 02 November, 2022.
Glycation-Associated Diabetic Nephropathy and Long Noncoding RNAs
Edit

The glycation of various biomolecules is the root cause of many pathological conditions associated with diabetic nephropathy and end-stage kidney disease. Glycation imbalances metabolism and increases renal cell injury. Numerous therapeutic measures have narrowed down the adverse effects of endogenous glycation, but efficient and potent measures are miles away. Recent advances in the identification and characterization of noncoding RNAs, especially the long noncoding RNAs (lncRNAs), have opened a mammon of new biology to explore the mitigations for glycation-associated diabetic nephropathy. Furthermore, tissue-specific distribution and condition-specific expression make lncRNA a promising key for second-generation therapeutic interventions.

diabetes diabetic nephropathy glycation long noncoding RNA biomarkers therapeutics

1. Introduction

With the advancement in high-throughput sequencing technologies, including RNA sequencing and next-generation sequencing, researchers’ understanding of genome and transcriptome has improved remarkably. The advanced genome study started by several consortia such as the Human Genome Project, Encyclopedia of DNA element (ENCODE), and Functional Annotation of the mouse/mammalian genome (FANTOM) has eventually publicized that only a small portion (3%) of the complete genome relates to known protein-coding genes. The remaining amount (97%) is the so-called ‘unchartered territory of noncoding RNA’ with small fractions decoded [1]. The established understanding of gene regulation in nature revolves around the central dogma of life (DNA → mRNA → protein); however, the rediscovery of noncoding RNA (ncRNA) genes and their role in the regulation of protein-coding genes has stirred the world [2]. The advanced genomic studies pointed out that most RNA transcripts are noncoding and identified about 35,000 ncRNA transcripts exhibiting signatures such as mRNA, including polyadenylation, capping, and splicing [3].
Moreover, due to their mRNA-like features, these newly identified ncRNA transcripts are difficult to distinguish from protein-coding transcripts. Despite these cumbersome challenges, various phases of these consortia identified almost around 27,919 long noncoding RNAs (lncRNAs) [4]. These findings suggested that human genome transcription is ubiquitous and produces thousands of ncRNA transcripts [5]. The ncRNAs can be further grouped into two classes based on their nucleotide number. One group consists of RNA genes with fewer than 200 nucleotides (microRNAs, small interfering RNAs, Piwi-interacting RNAs, and small nucleolar RNAs) [6], while the other group comprises RNA genes with more than 200 nucleotides, called lncRNAs [7]. The lncRNAs are present abundantly in both animals and plants, and their presence in the plant kingdom can be seen as an example of combinatorial transcriptional regulation [8]. The expression patterns of lncRNAs depend on many biological factors, including high glucose (HG), free fatty acids, growth factors, and inflammatory cytokines; hence, they are considered biomarkers in many pathological conditions (Table 1) [9][10][11][12][13][14][15][16][17][18][19][20][21][22][23][24].
Table 1. LncRNAs involved in various diabetes-associated complications.
Diabetic Complications lncRNA Involved Mode of Action References
Diabetic neuropathy lncRNA NEAT1 Regulate disease progression by targeting two miRNAs, miR-183-5p and miR-433-3p. [9]
lncRNA TP73-AS1 Sponges decreases miR-142 and upregulates HMGB1 expression as well as promotes cell proliferation. Its silencing decreases neuropathic pain. [10]
Diabetic retinopathy lncRNA HOTTIP Induces p38/MAPK signaling and promotes retinal cell inflammatory response and diabetic retinopathy progression. [11]
lncRNA BANCR Regulates cell apoptosis [12]
lncRNA MEG3 Regulates VEGF and TGF-β1 expressions [13]
Inflammatory diabetes complications lncRNA DRAIR Regulates macrophages/monocyte pro/anti-inflammatory phenotypes in T2DM. Down-regulated by diabetogenic factors [14]
Diabetic wound healing lncRNA URIDS Impairs collagen production and crosslinking by interacting with Plod1 and delays wound healing [15]
lncRNA MALAT1 Increases wound healing and upregulate fibroblast activation in diabetic mice by activating the HIF-1α signaling pathway. [16]
Diabetic cardiomyopathy lncRNA HOTAIR Downregulates DCM effects by activating SIRT1 expressions and sponging miR-34a. [17]
lncRNA Kcnq1ot1 Promotes pyroptosis by regulating expressions of miR-214-3p and caspase-1 [18]
lncRNA H19 Regulates high glucose-induced apoptosis by targeting VDAC1. It also improves left ventricular function when overexpressed. [19]
lncRNA Crende Negatively regulates cardiac fibroblast differentiation. Also, its expression is induced by Smad3 in cardiac fibroblasts. It inhibits myofibroblastic gene transcription. [20]
lncRNA TUG1 Its knockdown lessened DCM-induced hypertrophy and diastolic function. Also, its silencing upregulates the expression of some miRNAs [21]
Diabetic nephropathy lncRNA Blnc1 Attenuates renal fibrosis and inflammation and affects oxidative stress by NF-κB and NRF2/HO-1 pathways [22]
lncRNA TCF7 It acts as a sponge against miR-200c and triggers endoplasmic reticulum stress in patients with DN. [23]
lncRNA Gas5 Alleviates cell proliferation and fibrosis sponging miR-221 and upregulates SIRT1 [24]
Abbreviations: lncRNA MEG3, maternally expressed gene 3;, lncRNA Neat1, nuclear paraspeckle assembly target 1; HMGB1, High mobility group box 1; lncRNA URIDS, lncRNA Upregulated in Diabetic Skin; MAPK, Mitogen-activated protein kinase; lncRNA MALAT1, Metastasis-Associated Lung Adenocarcinoma Transcript 1; HIF-1α, Hypoxia-inducing factor - 1α; lncRNA Gas5, Growth Arrest-specific 5; lncRNA DRAIR, diabetes regulated anti-inflammatory RNA; lncRNA Crende, colorectal neoplasia differentially expressed; Smad3, SMAD family member 3lncRNA TUG1, taurine up-regulated gene 1lncRNA H19, Imprinted maternally expressed transcript; TCF7, Transcription factor 7; lncRNA BANCR, B-Raf proto-oncogene serine/threonine kinase-activated non-protein coding RNA; VEGF, vascular endothelial growth factor; SIRT1, silent mating type information regulator 2 homolog 1; DCM, diabetic cardiomyopathy; T2DM, type 2 diabetes mellitus.

1.1. Structural Properties of LncRNAs

Many lncRNAs transcribe and process like mRNAs (Figure 1). The generative pathways (histone-modification patterns, splicing, spliceosome signals, and exons/introns segments) for lncRNAs are similar to those of protein-coding genes [25]. Nevertheless, unlike mRNAs, a significant portion of lncRNAs are predominantly located in the nucleus, then in the cytoplasm [26]. Additionally, lncRNAs are broadly divided into categories based on genomic locations, such as 1. Intergenic lncRNAs (located between two separate genes), 2. Intronic lncRNAs (located between two exons), 3. Sense, or 4. Antisense lncRNAs (located on the same or opposite strands of protein-coding transcripts) and 5. Bidirectional lncRNAs [27][28]. This location-based classification is helpful for structural recognitions and annotations; however, lncRNAs’ biological functions are equally essential for more accurate classification. The majority of the lncRNAs have a half-life of more than 16 h. The intergenic and cis-antisense lncRNAs are more stable than intronic lncRNAs.
Figure 1. Archetypes and characteristics of lncRNAs along with their nuclear or cytoplasmic localizations. The various archetypes explain their mechanism of action in a non-mutually exclusive fashion. LncRNAs exhibit post-transcriptional signatures such as mRNA. Depending upon the splicing, lncRNA can either retain in the nucleus or localize in the cytosol. Abbreviations: LncRNAs, long noncoding RNAs; RNP, ribonucleoprotein; UTR, untranslated region; RMRP, RNA component of mitochondrial RNA-processing endoribonuclease; RBP, RNA-binding protein.

1.2. Functional Properties of LncRNAs

1.2.1. Role in Transcription Regulation

Gene regulation by lncRNAs can be facilitated through one of the two non-mutually exclusive ways: 1. By regulating adjacent gene expression and 2. By regulating chromatin state [29], thereby influencing neighboring gene expression. In both cases, lncRNA can either silences or enhances targeted gene expression [30]. The lncRNA Xist provides an ideal example of transcriptional silencing by spreading on a large portion of genes of one of the two X chromosomes [31]. LncRNAs Gas6 and SWINGN are well-studied examples of enhancer and enhancer-associated lncRNAs, respectively, to promote the transcriptional activation of targeted proteins [32].

1.2.2. Role in Post-Transcriptional Regulation

Regulation of post-transcriptional events comprises targeted protein modifications and their subcellular localization. The process includes mRNA splicing, processing, and nuclear export, with the help of RNA binding proteins (RBP). LncRNAs regulate gene expression at post-transcriptional levels by binding to RBP via RNA sequence motifs. LncRNA-mediated post-transcriptional regulation increases or decreases targeted mRNA and/or protein expression and their nuclear trafficking [33]. Additionally, lncRNAs can alter mRNA stability and splicing in a context-dependent manner [34]. The mRNA splicing regulation by lncRNAs involves either forming lncRNA-RNA hybrids with pre-mRNA or modifying the availability of splicing factors.

1.2.3. Role in Epigenetic Regulation

Many lncRNAs have been seen to regulate the epigenetic architecture of various proteins. These lncRNAs can either guide the epigenetic regulators to specific protein-coding loci or trigger the enhancer-promoter crosstalk. Their role as re-writers of chromatin folding (through chromatin modifying proteins such as methyltransferases and deacetylases) is documented in various studies [35]. Recent advances in detecting lncRNA-chromatin association have lightened their role at the epigenetic level [36]. Owing to their negatively charged surfaces, lncRNAs can neutralize positively charged histone proteins and de-compact the chromatin [37].

2. Understanding the Role of LncRNAs in AGEs-Related DN

Studies have shown that lncRNAs play a significant role in AGEs-mediated metabolic malfunctions [38]. So far, the involvement of lncRNAs-mediated AGEs-RAGE signaling in cancer, the immune system, and neurodegenerative diseases has been studied in detail [39]. However, few studies have reported the role of lncRNAs in glycation-associated diabetic complications [15][40][41][42][43] (Table 2).
Table 2. LncRNAs involved in glycation/AGE-associated diabetic complications.
LncRNA Involved Diabetic Complication Mode of Action References
lncRNA
Arid2-IR
Diabetic retinopathy Regulates oxidative stress, inflammatory responses, and endothelial cell dysfunction via interacting with Smad3 [40]
lncRNA MIAT Diabetic retinopathy AGE-induced HRPCs MIAT and CASP1 expressions increase, followed by the release of IL-1β, IL-18, and suppression of cell viability. [42]
lncRNA MEG3 Diabetic vascular diseases Upregulates in AGE-induced cells and suppresses cell viability and proliferation by modulating the MEG3/miR-193/p21 pathway [41]
lncRNA URIDS Diabetic Wound Healing Upregulates upon AGE induction. Regulates collagen production and deposition by targeting Plod1. It delays the wound healing process. [15]
lncRNA E330013P06 Inflammatory diabetes complications Increases inflammatory response upon AGE induction; by triggering pro-inflammatory gene. It also enhances foam cell formation. [43]
Abbreviations: lncRNA MEG3, maternally expressed gene 3; lncRNA URIDS, lncRNA Upregulated in Diabetic Skin; lncRNA MIAT, myocardial infarction-associated transcript; HRPCs, Human retinal pericytes.

2.1. LncRNA-Mediated RAGE Gene Expression and Signaling

The lncRNAs such as H9, MIAT, and MEG3 significantly regulate the pathogenesis of various diabetic complications. Recently, the role of lncRNA Arid2-IR in AGEs-induced retinal endothelial cells was also observed. By binding to Smad3, LncRNA Arid2-IR regulated levels of oxidative stress, inflammation, and apoptosis [40]. Similarly, lncRNA HOTAIR regulated RAGE expression and inflammation in acute myocardium infarction-induced rat models [44]. Likewise, the delayed process of diabetic wound healing in AGEs-induced fibroblast was associated with lncRNA URIDS [15]. Overexpression and knockdown studies also confirmed the role of another lncRNA MVIH in the AGEs-RAGE signaling pathway responsible for tumor induction and progression in cancer [45]. In AGEs-induced endothelial cells, lncRNA MEG3 regulated cell viability, proliferation, and apoptosis through the lncRNA MEG3/MiR-93/p21 mediated pathway during the onset of diabetic vascular diseases [41]. In a cell-based study on hypoxia/reoxygenation-injured cardiomyocytes, lncRNA SNHG12 down-regulated RAGE, NF-κB expression, and pro-inflammatory responses [46]. In a different context, the lncRNA TP73-AS1-mediated RAGE-HMGB1 signaling pathway upregulated NF-κB expression and pro-inflammatory cytokine levels [12][47]. The above-discussed lncRNAs can be a promising biomarker to detect complications in AGEs-RAGE signaling-mediated metabolism.

2.2. LncRNAs That Regulate AGER Gene Expression and Signaling

Notably, the expression pattern of lncRNA AGER-1 correlates with AGER expression levels (r = 0.360, p = 2.15 × 10−18) possibly by binding and sponging miRNA-185. Animal-based studies confirmed these results with suppressed cell proliferation rate, migration, and colony-forming efficiency in nude mice [48]. In a different context, lncRNA AGER-1 induced cell cycle arrest and promoted apoptosis, thereby inhibiting cell proliferation and migration efficiency of tumor cells in several types of cancer [49]. Yet another study indicated the sponging effect of lncRNA AGER-1 for miR-182, an inhibitor of AGER1 [50]. Recently, a study showed that both AGER and its positive regulator lncRNA AGER-1 have the significant diagnostic potential for lung adenocarcinoma. Both AGER and lncRNA AGER-1 regulate apoptosis, cell migration, and antitumor responses [51]. In summary, lncRNA AGER-1 can be a promising agent against AGEs signaling in metabolic disorders.

References

  1. Palazzo, A.F.; Lee, E.S. Non-Coding RNA: What Is Functional and What Is Junk? Front. Genet. 2015, 5, 1–11.
  2. Dunham, I.; Kundaje, A.; Aldred, S.F.; Collins, P.J.; Davis, C.A.; Doyle, F.; Epstein, C.B.; Frietze, S.; Harrow, J.; Kaul, R.; et al. An Integrated Encyclopedia of DNA Elements in the Human Genome. Nature 2012, 489, 57–74.
  3. Carninci, P.; Kasukawa, T.; Katayama, S.; Gough, J.; Frith, M.C.; Maeda, N.; Oyama, R.; Ravasi, T.; Lenhard, B.; Wells, C.; et al. Molecular Biology: The Transcriptional Landscape of the Mammalian Genome. Science 2005, 309, 1559–1563.
  4. Hon, C.C.; Ramilowski, J.A.; Harshbarger, J.; Bertin, N.; Rackham, O.J.L.; Gough, J.; Denisenko, E.; Schmeier, S.; Poulsen, T.M.; Severin, J.; et al. An Atlas of Human Long Non-Coding RNAs with Accurate 5′ Ends. Nature 2017, 543, 199–204.
  5. Hangauer, M.J.; Vaughn, I.W.; McManus, M.T. Pervasive Transcription of the Human Genome Produces Thousands of Previously Unidentified Long Intergenic Noncoding RNAs. PLoS Genet. 2013, 9, e1003569.
  6. Bushati, N.; Cohen, S.M. MicroRNA Functions. Annu. Rev. Cell Dev. Biol. 2007, 23, 175–205.
  7. Esteller, M. Non-Coding RNAs in Human Disease. Nat. Rev. Genet. 2011, 12, 861–874.
  8. Chen, M. PlncRNADB: A Repository of Plant LncRNAs and LncRNA-RBP Protein Interactions. Curr. Bioinform. 2019, 14, 621–627.
  9. Asadi, G.; Rezaei Varmaziar, F.; Karimi, M.; Rajabinejad, M.; Ranjbar, S.; Gorgin Karaji, A.; Salari, F.; Afshar Hezarkhani, L.; Rezaiemanesh, A. Determination of the Transcriptional Level of Long Non-Coding RNA NEAT-1, Downstream Target MicroRNAs, and Genes Targeted by MicroRNAs in Diabetic Neuropathy Patients. Immunol. Lett. 2021, 232, 20–26.
  10. Zhang, R.; Jin, H.; Lou, F. The Long Non-Coding RNA TP73-AS1 Interacted with MiR-142 to Modulate Brain Glioma Growth through HMGB1/RAGE Pathway. J. Cell. Biochem. 2018, 119, 3007–3016.
  11. Sun, Y.; Liu, Y. LncRNA HOTTIP Improves Diabetic Retinopathy by Regulating the P38-MAPK Pathway. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 2941–2948.
  12. Zhang, X.; Zou, X.; Li, Y.; Wang, Y. Downregulation of lncRNA BANCR participates in the development of retinopathy among diabetic patients. Exp. Ther. Med. 2019, 17, 4132–4138.
  13. Zhang, D.; Qin, H.; Leng, Y.; Li, X.; Zhang, L.; Bai, D.; Meng, Y.; Wang, J. LncRNA MEG3 Overexpression Inhibits the Development of Diabetic Retinopathy by Regulating TGF-β1 and VEGF. Exp. Ther. Med. 2018, 16, 2337–2342.
  14. Reddy, M.A.; Amaram, V.; Das, S.; Tanwar, V.S.; Ganguly, R.; Wang, M.; Lanting, L.; Zhang, L.; Abdollahi, M.; Chen, Z.; et al. LncRNA DRAIR Is Downregulated in Diabetic Monocytes and Modulates the Inflammatory Phenotype via Epigenetic Mechanisms. JCI Insight 2021, 6, 1–22.
  15. Hu, M.; Wu, Y.; Yang, C.; Wang, X.; Wang, W.; Zhou, L.; Zeng, T.; Zhou, J.; Wang, C.; Lao, G.; et al. Novel Long Noncoding RNA Lnc-URIDS Delays Diabetic Wound Healing by Targeting Plod1. Diabetes 2020, 69, 2144–2156.
  16. Liu, X.; Duan, L.; Chen, Y.; Jin, X.; Zhu, N.; Zhou, X.; Wei, H. LncRNA MALAT1 Accelerates Wound Healing of Diabetic Mice Transfused with Modified Autologous Blood via the HIF-1 a Signaling Pathway. Mol. Ther. Nucleic Acid 2019, 17, 504–515.
  17. Gao, L.; Wang, X.; Guo, S.; Xiao, L.; Liang, C.; Wang, Z.; Li, Y.; Liu, Y.; Yao, R.; Liu, Y.; et al. LncRNA HOTAIR Functions as a Competing Endogenous RNA to Upregulate SIRT1 by Sponging MiR-34a in Diabetic Cardiomyopathy. J. Cell. Physiol. 2019, 234, 4944–4958.
  18. Yang, F.; Qin, Y.; Wang, Y.; Li, A.; Lv, J.; Sun, X.; Che, H.; Han, T.; Meng, S.; Bai, Y.; et al. LncRNA KCNQ1OT1 mediates pyroptosis in diabetic cardiomyopathy. Cell. Physiol. Biochem. 2018, 5, 1230–1244.
  19. Li, X.; Wang, H.; Yao, B.; Xu, W.; Chen, J.; Zhou, X. LncRNA H19/MiR-675 Axis Regulates Cardiomyocyte Apoptosis by Targeting VDAC1 in Diabetic Cardiomyopathy. Sci. Rep. 2016, 6, 1–9.
  20. Zheng, D.; Zhang, Y.; Hu, Y.; Guan, J.; Xu, L.; Xiao, W.; Zhong, Q.; Ren, C.; Lu, J.; Liang, J.; et al. Long Noncoding RNA Crnde Attenuates Cardiac Fibrosis via Smad3-Crnde Negative Feedback in Diabetic Cardiomyopathy. FEBS J. 2019, 286, 1645–1655.
  21. Zhao, L.; Li, W.; Zhao, H. Inhibition of Long Non-Coding RNA TUG1 Protects against Diabetic Cardiomyopathy Induced Diastolic Dysfunction by Regulating MiR-499-5p. Am. J. Transl. Res. 2020, 12, 718–730.
  22. Feng, X.; Zhao, J.; Ding, J.; Shen, X.; Zhou, J.; Xu, Z. LncRNA Blnc1 Expression and Its Effect on Renal Fibrosis in Diabetic Nephropathy. Am. J. Transl. Res. 2019, 11, 5664–5672.
  23. Liu, H.; Sun, H. LncRNA TCF7 Triggered Endoplasmic Reticulum Stress through a Sponge Action with MiR-200c in Patients with Diabetic Nephropathy. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 5912–5922.
  24. Ge, X.; Xu, B.; Xu, W.; Xia, L.; Xu, Z.; Shen, L.; Peng, W.; Huang, S. Long Noncoding RNA GAS5 Inhibits Cell Proliferation and Fibrosis in Diabetic Nephropathy by Sponging MiR-221 and Modulating SIRT1 Expression. Aging 2019, 11, 8745–8759.
  25. Derrien, T.; Johnson, R.; Bussotti, G.; Tanzer, A.; Djebali, S.; Tilgner, H.; Guernec, G.; Martin, D.; Merkel, A.; Knowles, D.G.; et al. The GENCODE v7 Catalog of Human Long Noncoding RNAs: Analysis of Their Gene Structure, Evolution, and Expression. Genome Res. 2012, 22, 1775–1789.
  26. Bridges, M.C.; Daulagala, A.C.; Kourtidis, A. LNCcation: LncRNA Localization and Function. J. Cell Biol. 2021, 220, 1–17.
  27. Klapproth, C.; Sen, R.; Stadler, P.F.; Findeiß, S.; Fallmann, J. Common Features in LncRNA Annotation and Classification: A Survey. Non-Coding RNA 2021, 7, 77.
  28. Parasramka, M.A.; Maji, S.; Matsuda, A.; Yan, I.K.; Patel, T. Pharmacology & Therapeutics Long Non-Coding RNAs as Novel Targets for Therapy in Hepatocellular Carcinoma. Pharmacol. Ther. 2016, 161, 67–78.
  29. Su, P.P.; Liu, D.W.; Zhou, S.J.; Chen, H.; Wu, X.M.; Liu, Z.S. Down—Regulation of Risa Improves Podocyte Injury by Enhancing Autophagy in Diabetic Nephropathy. Mil. Med. Res. 2022, 9, 1–13.
  30. Gil, N.; Ulitsky, I. Regulation of Gene Expression by Cis-acting Long non-coding RNAs. Nat. Rev. Genet. 2020, 21, 102–117.
  31. Wutz, A. Gene Silencing in X-Chromosome Inactivation: Advances in Understanding Facultative Heterochromatin Formation. Nat. Rev. Genet. 2011, 12, 542–553.
  32. Grossi, E.; Raimondi, I.; Goñi, E.; González, J.; Marchese, F.P.; Chapaprieta, V.; Martín-Subero, J.I.; Guo, S.; Huarte, M.A. LncRNA-SWI/SNF Complex Crosstalk Controls Transcriptional Activation at Specific Promoter Regions. Nat. Commun. 2020, 11, 1–16.
  33. He, R.; Luo, D.; Mo, Y. Emerging Roles of LncRNAs in the Post-Transcriptional Regulation in Cancer. Genes Dis. 2019, 6, 6–15.
  34. Lee, S.; Kopp, F.; Chang, T.; Sataluri, A.; Chen, B.; Sivakumar, S.; Yu, H.; Xie, Y.; Mendell, J.T.; Comprehensive, C.S. Noncoding RNA NORAD Regulates Genomic Stability by Sequestering PUMILIO proteins. Cell 2017, 164, 69–80.
  35. Morlando, M.; Fatica, A. Alteration of Epigenetic Regulation by Ong Noncoding RNAs in Cancer. Int. J. Mol. Sci. 2018, 19, 570.
  36. Isoda, T.; Moore, A.J.; He, Z.; Chandra, V.; Aida, M.; Denholtz, M.; Piet van Hamburg, J.; Fisch, K.M.; Chang, A.N.; Fahl, S.P.; et al. Non-Coding Transcription Instructs Chromatin Folding and Compartmentalization to Dictate Enhancer-Promoter Communication and T cell fate. Cell 2017, 171, 103–119.
  37. Dueva, R.; Akopyan, K.; Pederiva, C.; Trevisan, D.; Dhanjal, S.; Lindqvist, A.; Farnebo, M.; Dueva, R.; Akopyan, K.; Pederiva, C.; et al. Neutralization of the Positive Charges on Histone tails by RNA Promotes an Open Chromatin Structure. Cell Chem. Biol. 2019, 26, 1436–1449.
  38. Sanchez, C.A.; Kawamura, Y.; Yamamoto, Y.; Takeshita, F.; Ochiya, T. Emerging Roles of Long Non-Coding RNA in Cancer. Cancer Sci. 2018, 109, 2093–2100.
  39. Chrysanthou, M.; Estruch, I.M.; Rietjens, I.M.C.M.; Wichers, H.J.; Hoppenbrouwers, T. In Vitro Methodologies to Study the Role of Advanced Glycation End Products (AGEs) in Neurodegeneration. Nutrients 2022, 14, 363.
  40. Xiao, H.; Yang, H.; Zeng, Y. Long Non-Coding RNA Arid2-IR Affects Advanced Glycation End Products-Induced Human Retinal Endothelial Cell Injury by Binding to Smad3. Int. Ophthalmol. 2020, 40, 1123–1133.
  41. Ju, C.; Sheng, Z.; Wang, Q.; Li, Y.; Wang, X.; Li, S.; Qi, Q.; Yuan, Z. Advanced Glycation End Products of Bovine Serum Albumin Affect the Cell Growth of Human Umbilical Vein Endothelial Cells via Modulation of MEG3/MiR-93/P21 Pathway. Acta Biochim. Biophys. Sin. 2018, 51, 41–50.
  42. Yu, X.; Ma, X.; Lin, W.; Xu, Q.; Zhou, H.; Kuang, H.Y. Long Noncoding RNA MIAT Regulates Primary Human Retinal Pericyte Pyroptosis by Modulating MiR-342–3p Targeting of CASP1 in Diabetic Retinopathy. Exp. Eye Res. 2021, 202, 108300.
  43. Reddy, M.A.; Chen, Z.; Park, J.T.; Wang, M.; Lanting, L.; Zhang, Q.; Bhatt, K.; Leung, A.; Wu, X.; Putta, S.; et al. Regulation of Inflammatory Phenotype in Macrophages by a Diabetes-Induced Long Noncoding RNA. Diabetes 2014, 63, 4249–4261.
  44. Lu, W.; Zhu, L.; Ruan, Z.B.; Wang, M.X.; Ren, Y.; Li, W. HOTAIR Promotes Inflammatory Response after Acute Myocardium Infarction by Upregulating RAGE. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 7423–7430.
  45. Hu, S.; Zheng, Q.; Xiong, J.; Wu, H.; Wang, W.; Zhou, W. Long Non-Coding RNA MVIH Promotes Cell Proliferation, Migration, Invasion through Regulating Multiple Cancer-Related Pathways, and Correlates with Worse Prognosis in Pancreatic Ductal Adenocarcinomas. Am. J. Transl. Res. 2020, 12, 2118–2135.
  46. Lu, P.; Xiao, S.; Chen, S.; Fu, Y.; Zhang, P.; Yao, Y.; Chen, F. LncRNA SNHG12 Downregulates RAGE to Attenuate Hypoxia-Reoxygenation-Induced Apoptosis in H9c2 Cells. Biosci. Biotechnol. Biochem. 2021, 85, 866–873.
  47. Li, S.; Huang, Y.; Huang, Y.; Fu, Y.; Tang, D.; Kang, R.; Zhou, R.; Fan, X.G. The Long Non-Coding RNA TP73-AS1 Modulates HCC Cell Proliferation through MiR-200a-Dependent HMGB1/RAGE Regulation. J. Exp. Clin. Cancer Res. 2017, 36, 1–12.
  48. Pan, Z.; Liu, L.; Nie, W.; Miggin, S.; Qiu, F.; Cao, Y.; Chen, J.; Yang, B.; Zhou, Y.; Lu, J.; et al. Long Non-Coding RNA AGER-1 Functionally Upregulates the Innate Immunity Gene AGER and Approximates Its Anti-Tumor Effect in Lung Cancer. Mol. Carcinog. 2018, 57, 305–318.
  49. Lin, M.; Li, Y.; Xian, J.; Chen, J.; Feng, Y.; Mao, C.; Pan, Y.; Li, Z.; Zeng, Y.; Yang, L.; et al. Long Non-Coding RNA AGER-1 Inhibits Colorectal Cancer Progression through Sponging MiR-182. Int. J. Biol. Markers 2020, 35, 10–18.
  50. Pidíková, P.; Herichová, I. MiRNA Clusters with Up-Regulated Expression in Colorectal Cancer. Cancers 2021, 13, 2979.
  51. Abdelwahab, O.; Awad, N.; Elserafy, M.; Badr, E. A Feature Selection-Based Framework to Identify Biomarkers for Cancer Diagnosis: A Focus on Lung Adenocarcinoma. PLoS ONE 2022, 17, e0269126.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , ,
View Times: 471
Entry Collection: Biopharmaceuticals Technology
Revisions: 2 times (View History)
Update Date: 03 Nov 2022
1000/1000
Video Production Service