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 -- 2236 2023-03-22 12:07:29 |
2 format change + 1 word(s) 2237 2023-03-23 02:30:12 | |
3 All is fine. Meta information modification 2237 2023-03-24 01:26:57 |

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.
Wu, J.; Ma, L.; Cao, Y. Alternative Polyadenylation in Plants. Encyclopedia. Available online: https://encyclopedia.pub/entry/42429 (accessed on 16 May 2024).
Wu J, Ma L, Cao Y. Alternative Polyadenylation in Plants. Encyclopedia. Available at: https://encyclopedia.pub/entry/42429. Accessed May 16, 2024.
Wu, Jing, Ligeng Ma, Ying Cao. "Alternative Polyadenylation in Plants" Encyclopedia, https://encyclopedia.pub/entry/42429 (accessed May 16, 2024).
Wu, J., Ma, L., & Cao, Y. (2023, March 22). Alternative Polyadenylation in Plants. In Encyclopedia. https://encyclopedia.pub/entry/42429
Wu, Jing, et al. "Alternative Polyadenylation in Plants." Encyclopedia. Web. 22 March, 2023.
Alternative Polyadenylation in Plants
Edit

The polyadenylation (poly(A)) tail of mRNA is an essential feature that is required to mediate its nuclear export, stability, translation efficiency, and subcellular localization. Most genes have at least two mRNA isoforms via alternative splicing (AS) or alternative polyadenylation (APA), which increases the diversity of transcriptome and proteome. 

gene expression alternative polyadenylation stress response plants

1. Introduction

An essential step in the maturation of mRNA is 3′-end-processing. The 3′-end carries a series of adenine residues called the polyadenylation (poly(A)) tail in almost all eukaryotic mRNAs. The maturation of a RNA from a protein-coding gene requires the addition of the poly(A) tail at its 3′-end, and this process is highly conserved in eukaryotes [1][2]. As a level of post-transcriptional regulation of gene expression, 3′-end-processing affects many aspects of the regulation of gene expression, including nuclear export, stability, translation efficiency, and protein localization of an mRNA [3][4][5][6].
The 3′-end-processing of an mRNA in eukaryotes can be divided into two steps, cleavage and polyadenylation, which are accomplished by a large 3′-end-processing complex that includes four sub-complexes (Figure 1), and is very conserved in yeast, animals, and plants [7][8]. The regulation of polyadenylation involves a complicated interplay between the numerous cis-elements surrounding the poly(A) site and trans-acting factors, of which several are RNA-binding proteins. In vertebrates, the most prominent polyadenylation signal is the A(A/U)UAAA motif, typically located 15–30 nt upstream of the poly(A) site, and upwards of 80% of genes have this consensus sequence feature at the 3′-end of pre-mRNA which can be recognized by the CPSF (cleavage and polyadenylation specificity factor) complex [9]; at the same time, the CstF (cleavage stimulation factor) complex binds to the U-rich and GU-rich motifs downstream of the poly(A) site [10], and the CFI (cleavage factor I) complex binds to the UGUA motif upstream of the poly(A) site [11]. Compared to the three sub-complexes, the ability of the CFII (cleavage factor II) complex to bind RNA has been less studied.
Figure 1. Core factors involved in 3′-end-processing. The core cleavage and polyadenylation factors are similar and can be divided into four sub-complexes in yeast, animals, and plants.
The structure of the cleavage and polyadenylation complexes from yeast and animals shares roughly similar core components, yet with some differences (Figure 1), with yeast lacking the CFI complex and Cst50 that is present in vertebrates, while vertebrates lack the Hrp1 that is in yeast [12][13][14][15][16][17][18][19]. In contrast, the understanding in higher plants relies mainly on the analysis of their homologous proteins in yeast and animals [20][21][22][23][24][25][26][27][28][29][30]. Based on the present understanding, it seems that the overall composition of the 3′-end-processing complex from higher plants is relatively similar to that of vertebrates; it misses the CFIm59 in the CFI complex [20]. Another striking feature is the large number of gene duplications for genes encoding homologous proteins for the 3′-end-processing complex, especially for the homologous proteins of PCF11 and PAP1, with four each (Figure 1), which also implies that the regulatory mechanisms of polyadenylation in plants may be more exquisite and complicated [22][31].
In eukaryotes, most genes have more than one poly(A) site, and the presence of two or more poly(A) sites in a gene leads to the production of different isoforms of transcripts, which is known as alternative polyadenylation (APA) [32]. APA is present in more than 84% of yeast genes and more than 70% of animal genes [33][34][35], while it is present more than 70% of genes in the lower plant Chlamydomonas reinhardtii and the model plant Arabidopsis thaliana, and at least 50% of rice exhibits APA [36][37][38]. This suggests that APA is widespread in eukaryotes and implies that this mode of regulation is important for the regulation of gene expression. When APA occurs in upstream regions of the mRNA, it often leads to the generation of a truncated protein, thereby affecting the function of a full-length protein; however, when it occurs in 3′UTR, it may lead to a change in the stability of mRNA or translation efficiency [39].

2. Molecular Mechanisms for APA-Mediated Responses

The mechanisms for the APA-mediated stress response have rarely been reported in plants; however, there are some reports from animals and yeast for the APA-mediated regulation of gene function.

2.1. Influence Full-Length Transcripts

The poly(A) tail is added to the newly generated mRNA to indicate the termination of transcription and therefore determines the coding region of a protein. There are fewer studies on 5′UTR APA and CDS APA; more extensively studied is intronic APA, where polyadenylation occurring at the intron generally quickly encounters the termination codon to form a truncated protein, such that the truncated protein may be partially active or completely lose function [40]. Intronic APA events are common in human immune cells and severely dysregulated in cancer cells [41][42], suggesting an important role of intronic APA in the precise regulation of gene expression.
Intronic APA events are often associated with competition between splicing and 3′-end-processing machinery on introns, even though eukaryote gene expression is generally considered to be co-transcribed [43]. There are interactions between splicing factor and polyadenylation factor [44][45], which may underlie the competition between them. Intronic APA events often occur in the first two introns, which often produce abnormally transcript-encoding short truncated protein and thus reduce the proportion of functional transcripts [46], or play a dominant negative role as the truncated protein inhibits the function of the full-length protein [47].
A certain percentage of introns are not fully spliced and a cryptic polyadenylation signal is recognized in the retained introns, which produces the truncated proteins under stress. Those retained introns are characterized by a length greater than the average length of introns, which may be close to the 5′UTR, implying that introns with weaker splicing signals may undergo intron retention under stress and be recognized by the 3′-end-formation machinery, which may act as competition for the function of full length and truncated proteins [48][49][50].

2.2. Influence RNA Fate and Translation Efficiency

In contrast to intronic APA, APA occurring on the 3′UTR does not change the protein-coding sequence, but this does not mean that 3′UTR APA produces weaker effects than other forms of APA for the function of the target gene [51]. On the contrary, 3′UTR APA can alter key sequences that regulate RNA export and translation efficiency, and therefore, the regulation of 3′UTR length by APA is an important determinant for RNA fate determination. This mode of regulation is general, as more than 50% of the genes in the human genome show 3′UTR APA events [35].

2.2.1. RNA Stability

Studies in yeast have shown that mRNA stability is negatively correlated to the length of 3′UTR, with transcripts with short half-lives being twice as long as the most stable transcripts [52]. The 3′UTR contains multiple cis-elements and is subject to complex regulation by trans-acting factors, such as microRNA or RNA-binding protein [53].
The more studied factors are the role of microRNA on the stability of mRNA with 3′UTR APA. MicroRNAs are short RNAs of about 20 nucleotides in size that regulate the post-transcriptional silencing of target genes [54]. The 3′UTR APA event often alters the sequence of the UTR in the mRNA to contain or not contain the microRNA-binding site. For example, Hsp70.3 promotes the use of the proximal site in the 3′UTR, which likewise increases the expression of Hsp70.3, thereby improving cell survival under heat shock conditions [55]. The shortening in NLRP3 3′UTR leads to the overactivation of NLRP3 and exacerbates the inflammatory response [56]. The up-regulation of the gene expression level for the genes with 3′UTR shortening is caused by avoiding microRNA-mediated degradation when shortening the 3′UTR.
In addition to the microRNA, RNA-binding protein also acts via the 3′UTR as a post-transcriptional regulator. The binding of RNA-binding protein to mRNA may lead to the degradation of mRNA. The 3′UTR contains multiple cis-elements, ARE (Adenylate/uridylate Reich element) is the most well-known of these, being present in 5–8% of human genes and involved in regulating many important physiological processes [57]. Mouse cells use the proximal poly(A) site under arsenic stress and enhance the degradation of long 3′UTR transcription during recovery. The degradation of long 3′UTR transcription is due to the binding of TIA1 to the U-rich motif of the long 3′UTR, promoting SG recruitment and leading to enhanced mRNA decay [58]. Thus, the use of the proximal poly(A) site under stress facilitates the retention of transcript abundance.

2.2.2. RNA Export

The transport of mRNA from the nucleus to the cytoplasm is a key process in the expression of genes in eukaryotes, and this process is closely controlled by the 3′UTR [59]. Biotic and abiotic stresses trigger the disruption of transcription termination (DoTT), which results in a read-through transcript that extends to the next gene instead of stopping at the 3′-end of the previous gene [60]. The read-through transcripts are strongly enriched in chromatin and soluble nuclear extracts and therefore cannot be efficiently exported to the cytoplasm. Thus, DoTT substantially regulates gene expression by inhibiting RNA export, which is associated with an overall decrease in the level of protein [61]. Under salt stress, 10% of human-encoded proteins produce transcriptional read-through events, and the poly(A) signal intensity of these genes is usually below the average, which may be the underlying cause of the read-through [62]. HSV-1 infection was followed by a significant transcriptional read-through of the interferon regulatory factor gene IRF1, which is important for the immune response against viruses, suggesting that transcriptional read-through may be one of the methods by which the virus escapes from host immunity [61].

2.2.3. Translation Efficiency

Most mRNAs perform their functions at the protein level, and the 3′UTR can also regulate protein expression by affecting translation efficiency. In addition to participating in the regulation of RNA stability, microRNAs, and RNA-binding proteins, long 3′UTR also inhibits protein translation efficiency [63]. In general, the 3′UTR of proliferating cells is shorter than that of differentiating cells in higher animals [64]; correspondingly, mRNA isoforms with shorter 3′UTR exhibit higher translation efficiency [65]. Yeast grown on rich medium tended to express shorter transcripts compared to that on a mini medium, and shorter 3′UTR correlated with up-regulation of genes participating in translation [33]. Under oxidative stress, C/EBPγ, the target of mTOR signaling, appears shortened in 3′UTR; short transcripts have high translational efficiency, and high levels of C/EBPγ expression control redox homeostasis [66]. Therefore, 3′UTR APA can also rapidly regulate the expression of genes by regulating translation efficiency in subtle changes in the cellular and surrounding environment.
In summary, the poly(A) site usage of many genes changes significantly under stress, such as the usage alteration of poly(A) site from the 3′UTR to the 5′UTR, introns and exons, and of course other positions in the 3′UTR. Genes usually escape from microRNA control due to the shortened 3′UTR and thus is up-regulated at the RNA level, which may facilitate the rapid response of resistance to stress, and 3′UTR shortening also improves translation efficiency and thus increases the expression at the protein level due to the presence of the binding motif in the 3′UTR for RNA-binding protein (Figure 2). The intronic APA and transcriptional read-through may down-regulate gene expression by reducing the ratio of functional transcripts or affecting the output of transcripts (Figure 2), and even transcriptional read-through may form long non-coding RNAs to regulate the expression of peripheral genes. At present, there are few studies on 5′UTR APA and exon APA, and they are also a strategy to reduce gene expression.
Figure 2. Molecular mechanisms for APA-mediated responses. Upstream-region APA often produces abnormally transcript-encoding short truncated proteins or is directly degraded by NMD-mediated pathway, thereby reducing the proportion of functional transcript. However, 3′UTR APA generally affects protein levels by influencing nuclear export, RNA stability, and translation efficiency.

3. An Overview of the Role of APA in Response to Biotic and Abiotic Stresses

The regulation of gene expression is one of key steps for the development and response to environmental changes in eukaryotes. Plants require more precise regulation of gene expression than animals and yeast because they have smaller transcription units and intergenic regions [67]. Several recent reviews have summarized the role of APA on the regulation of plant growth and development [67][68][69]. Here, the researchers focus on the role of APA in the plant stress response.
Stresses are commonly found in the environments where the organisms live, grow, and develop. Based on the characteristics of these stresses, they can be classified as biotic and abiotic stresses, which seriously affect the survival of plants and animals [70]. As sessile organisms, plants are more susceptible to stresses than animals. Throughout their life cycle, plants are constantly exposed to a variety of external stimuli. As a result, crop yields are affected to varying degrees by environmental stresses such as drought, salt, heavy metal, and hypoxic stresses. At the same time, plants also actively respond to environmental changes through the regulation of gene expression [71]. To overcome inevitable harsh environmental challenges, plants have evolved multiple gene-regulatory mechanisms to avoid injury.
The studies indicate that a wide range of APA events occur under biotic and abiotic stresses in plants, suggesting that APA may act as a positive post-transcriptional regulation in response to stress in plants (Table 1). However, more detailed molecular mechanisms remain to be explored.
Table 1. Alternative polyadenylation mediates stress responses in plants.

Stress

Species

Target Genes

APA Types

Associated Polyadenylation

Factors

References

Hypoxic

Arabidopsis thaliana

 

5′UTR APA

CDS APA

Intronic APA

3′UTR APA

 

[72]

Drought

Arabidopsis thaliana

 

3′UTR extension

FPA

[73]

Salt

Arabidopsis thaliana

Eutrema salsugineum

Sorghum

AKR2

AT3G47610

CIPK21 MAP3Kδ4

3′UTR APA

FIP1

CPSF30

[74][75][76][77]

N starvation

Arabidopsis thaliana

Chlamydomonas

NRT1.1

CPSF30

3′UTR APA

Intronic APA

FIP1

CPSF30

[28][78][79][80]

Temperature

Arabidopsis thaliana

SVK

3′UTR extension

 

[81]

Pathogens

Rice

Arabidopsis thaliana

Xa1

rTGA2.1

CBP60g

CDS APA

3′UTR APA

FIP1

CPSF30

[82][83][84]

ROS

Arabidopsis thaliana

ERF4

CDS APA

FPA

CPSF30

[85][86][87]

References

  1. Wang, R.; Zheng, D.; Yehia, G.; Tian, B. A compendium of conserved cleavage and polyadenylation events in mammalian genes. Genome Res. 2018, 28, 1427–1441.
  2. Passmore, L.A.; Coller, J. Roles of mRNA poly(A) tails in regulation of eukaryotic gene expression. Nat. Rev. Mol. Cell Biol. 2022, 23, 93–106.
  3. Neve, J.; Burger, K.; Li, W.; Hoque, M.; Patel, R.; Tian, B.; Gullerova, M.; Furger, A. Subcellular RNA profiling links splicing and nuclear DICER1 to alternative cleavage and polyadenylation. Genome Res. 2016, 26, 24–35.
  4. Graham, R.R.; Kyogoku, C.; Sigurdsson, S.; Vlasova, I.A.; Davies, L.R.L.; Baechler, E.C.; Plenge, R.M.; Koeuth, T.; Ortmann, W.A.; Hom, G.; et al. Three functional variants of IFN regulatory factor 5 (IRF5) define risk and protective haplotypes for human lupus. Proc. Natl. Acad. Sci. USA 2007, 104, 6758–6763.
  5. Hartwick, E.W.; Costantino, D.A.; MacFadden, A.; Nix, J.C.; Tian, S.; Das, R.; Kieft, J.S. Ribosome-induced RNA conformational changes in a viral 3′-UTR sense and regulate translation levels. Nat. Commun. 2018, 9, 5074.
  6. Berkovits, B.D.; Mayr, C. Alternative 3′ UTRs act as scaffolds to regulate membrane protein localization. Nature 2015, 522, 363–367.
  7. Kumar, A.; Clerici, M.; Muckenfuss, L.M.; Passmore, L.A.; Jinek, M. Mechanistic insights into mRNA 3′-end processing. Curr. Opin. Struct. Biol. 2019, 59, 143–150.
  8. Hunt, A.G. mRNA 3′ end formation in plants: Novel connections to growth, development and environmental responses. Wiley Interdiscip. Rev. RNA 2020, 11, e1575.
  9. Clerici, M.; Faini, M.; Muckenfuss, L.M.; Aebersold, R.; Jinek, M. Structural basis of AAUAAA polyadenylation signal recognition by the human CPSF complex. Nat. Struct. Mol. Biol. 2018, 25, 135–138.
  10. Yang, W.; Hsu, P.L.; Yang, F.; Song, J.-E.; Varani, G. Reconstitution of the CstF complex unveils a regulatory role for CstF-50 in recognition of 3′-end processing signals. Nucleic Acids Res. 2018, 46, 493–503.
  11. Yang, Q.; Gilmartin, G.M.; Doublie, S. Structural basis of UGUA recognition by the Nudix protein CFI(m)25 and implications for a regulatory role in mRNA 3′ processing. Proc. Natl. Acad. Sci. USA 2010, 107, 10062–10067.
  12. Gross, S.; Moore, C. Five subunits are required for reconstitution of the cleavage and polyadenylation activities of Saccharomyces cerevisiae cleavage factor I. Proc. Natl. Acad. Sci. USA 2001, 98, 6080–6085.
  13. Zhao, J.; Kessler, M.M.; Moore, C.L. Cleavage factor II of Saccharomyces cerevisiae contains homologues to subunits of the mammalian Cleavage/polyadenylation specificity factor and exhibits sequence-specific, ATP-dependent interaction with precursor RNA. J. Biol. Chem. 1997, 272, 10831–10838.
  14. Zhao, J.; Kessler, M.; Helmling, S.; O’Connor, J.P.; Moore, C. Pta1, a component of yeast CF II, is required for both cleavage and poly(A) addition of mRNA precursor. Mol. Cell. Biol. 1999, 19, 7733–7740.
  15. Preker, P.J.; Ohnacker, M.; Minvielle-Sebastia, L.; Keller, W. A multisubunit 3′ end processing factor from yeast containing poly(A) polymerase and homologues of the subunits of mammalian cleavage and polyadenylation specificity factor. EMBO J. 1997, 16, 4727–4737.
  16. Ruegsegger, U.; Blank, D.; Keller, W. Human pre-mRNA cleavage factor Im is related to spliceosomal SR proteins and can be reconstituted in vitro from recombinant subunits. Mol. Cell 1998, 1, 243–253.
  17. de Vries, H.; Rüegsegger, U.; Hübner, W.; Friedlein, A.; Langen, H.; Keller, W. Human pre-mRNA cleavage factor IIm contains homologs of yeast proteins and bridges two other cleavage factors. EMBO J. 2000, 19, 5895–5904.
  18. Takagaki, Y.; Manley, J.L. Complex protein interactions within the human polyadenylation machinery identify a novel component. Mol. Cell. Biol. 2000, 20, 1515–1525.
  19. Clerici, M.; Faini, M.; Aebersold, R.; Jinek, M. Structural insights into the assembly and polyA signal recognition mechanism of the human CPSF complex. eLife 2017, 6, e33111.
  20. Yu, Z.; Hong, L.; Li, Q.Q. Signatures of mRNA alternative polyadenylation in Arabidopsis leaf development. Front. Genet. 2022, 13, 863253.
  21. Xing, D.; Zhao, H.; Li, Q.Q. Arabidopsis CLP1-SIMILAR PROTEIN3, an ortholog of human polyadenylation factor CLP1, functions in gametophyte, embryo, and postembryonic development. Plant Physiol. 2008, 148, 2059–2069.
  22. Xing, D.; Zhao, H.; Xu, R.; Li, Q.Q. Arabidopsis PCFS4, a homologue of yeast polyadenylation factor Pcf11p, regulates FCA alternative processing and promotes flowering time. Plant J. 2008, 54, 899–910.
  23. Yao, Y.L.; Song, L.H.; Katz, Y.; Galili, G. Cloning and characterization of Arabidopsis homologues of the animal CstF complex that regulates 3′ mRNA cleavage and polyadenylation. J. Exp. Bot. 2002, 53, 2277–2278.
  24. Herr, A.J.; Molnar, A.; Jones, A.; Baulcombe, D.C. Defective RNA processing enhances RNA silencing and influences flowering of Arabidopsis. Proc. Natl. Acad. Sci. USA 2006, 103, 14994–15001.
  25. Chakrabarti, M.; Hunt, A.G. CPSF30 at the interface of alternative polyadenylation and cellular signaling in plants. Biomolecules 2015, 5, 1151–1168.
  26. Elliott, B.J.; Dattaroy, T.; Meeks-Midkiff, L.R.; Forbes, K.P.; Hunt, A.G. An interaction between an Arabidopsis poly(A) polymerase and a homologue of the 100 kDa subunit of CPSF. Plant Mol. Biol. 2003, 51, 373–384.
  27. Xu, R.Q.; Ye, X.F.; Li, Q.S.Q. AtCPSF73-II gene encoding an Arabidopsis homolog of CPSF 73 kDa subunit is critical for early embryo development. Gene 2004, 324, 35–45.
  28. Li, Z.; Wang, R.; Gao, Y.; Wang, C.; Zhao, L.; Xu, N.; Chen, K.-E.; Qi, S.; Zhang, M.; Tsay, Y.-F.; et al. The Arabidopsis CPSF30-L gene plays an essential role in nitrate signaling and regulates the nitrate transceptor gene NRT1.1. New Phytol. 2017, 216, 1205–1222.
  29. Forbes, K.P.; Addepalli, B.; Hunt, A.G. An Arabidopsis Fip1 homolog interacts with RNA and provides conceptual links with a number of other polyadenylation factor subunits. J. Biol. Chem. 2006, 281, 176–186.
  30. Simpson, G.G.; Dijkwel, P.P.; Quesada, V.; Henderson, I.; Dean, C. FY is an RNA 3′ end-processing factor that interacts with FCA to control the Arabidopsis floral transition. Cell 2003, 113, 777–787.
  31. Vi, S.L.; Trost, G.; Lange, P.; Czesnick, H.; Rao, N.; Lieber, D.; Laux, T.; Gray, W.M.; Manley, J.L.; Groth, D.; et al. Target specificity among canonical nuclear poly(A) polymerases in plants modulates organ growth and pathogen response. Proc. Natl. Acad. Sci. USA 2013, 110, 13994–13999.
  32. Ozsolak, F.; Kapranov, P.; Foissac, S.; Kim, S.W.; Fishilevich, E.; Monaghan, A.P.; John, B.; Milos, P.M. Comprehensive polyadenylation site maps in yeast and human reveal pervasive alternative polyadenylation. Cell 2010, 143, 1018–1029.
  33. Liu, X.; Hoque, M.; Larochelle, M.; Lemay, J.-F.; Yurko, N.; Manley, J.L.; Bachand, F.; Tian, B. Comparative analysis of alternative polyadenylation in S. cerevisiae and S. pombe. Genome Res. 2017, 27, 1685–1695.
  34. Derti, A.; Garrett-Engele, P.; MacIsaac, K.D.; Stevens, R.C.; Sriram, S.; Chen, R.; Rohl, C.A.; Johnson, J.M.; Babak, T. A quantitative atlas of polyadenylation in five mammals. Genome Res. 2012, 22, 1173–1183.
  35. Hoque, M.; Ji, Z.; Zheng, D.; Luo, W.; Li, W.; You, B.; Park, J.Y.; Yehia, G.; Tian, B. Analysis of alternative cleavage and polyadenylation by 3′ region extraction and deep sequencing. Nat. Methods 2013, 10, 133–139.
  36. Bell, S.A.; Shen, C.; Brown, A.; Hunt, A.G. Experimental Genome-Wide Determination of RNA Polyadenylation in Chlamydomonas reinhardtii. PLoS ONE 2016, 11, e0146107.
  37. Wu, X.; Liu, M.; Downie, B.; Liang, C.; Ji, G.; Li, Q.Q.; Hunt, A.G. Genome-wide landscape of polyadenylation in Arabidopsis provides evidence for extensive alternative polyadenylation. Proc. Natl. Acad. Sci. USA 2011, 108, 12533–12538.
  38. Shen, Y.; Ji, G.; Haas, B.J.; Wu, X.; Zheng, J.; Reese, G.J.; Li, Q.Q. Genome level analysis of rice mRNA 3′-end processing signals and alternative polyadenylation. Nucleic Acids Res. 2008, 36, 3150–3161.
  39. Elkon, R.; Ugalde, A.P.; Agami, R. Alternative cleavage and polyadenylation: Extent, regulation and function. Nat. Rev. Genet. 2013, 14, 496–506.
  40. Tsuchiya, T.; Eulgem, T. An alternative polyadenylation mechanism coopted to the Arabidopsis RPP7 gene through intronic retrotransposon domestication. Proc. Natl. Acad. Sci. USA 2013, 110, E3535–E3543.
  41. Ma, X.; Cheng, S.; Ding, R.; Zhao, Z.; Zou, X.; Guang, S.; Wang, Q.; Jing, H.; Yu, C.; Ni, T.; et al. ipaQTL-atlas: An atlas of intronic polyadenylation quantitative trait loci across human tissues. Nucleic Acids Res. 2022, 51, 1046–1052.
  42. Zhao, Z.; Xu, Q.; Wei, R.; Wang, W.; Ding, D.; Yang, Y.; Yao, J.; Zhang, L.; Hu, Y.-Q.; Wei, G.; et al. Cancer-associated dynamics and potential regulators of intronic polyadenylation revealed by IPAFinder using standard RNA-seq data. Genome Res. 2021, 31, 2095–2106.
  43. Yang, J.; Cao, Y.; Ma, L. Co-Transcriptional RNA Processing in Plants: Exploring from the Perspective of Polyadenylation. Int. J. Mol. Sci. 2021, 22, 3300.
  44. Kyburz, A.; Friedlein, A.; Langen, H.; Keller, W. Direct interactions between subunits of CPSF and the U2 snRNP contribute to the coupling of pre-mRNA 3′ end processing and splicing. Mol. Cell 2006, 23, 195–205.
  45. Müller-McNicoll, M.; Botti, V.; de Jesus Domingues, A.M.; Brandl, H.; Schwich, O.D.; Steiner, M.C.; Curk, T.; Poser, I.; Zarnack, K.; Neugebauer, K.M. SR proteins are NXF1 adaptors that link alternative RNA processing to mRNA export. Genes Dev. 2016, 30, 553–566.
  46. Dubbury, S.J.; Boutz, P.L.; Sharp, P.A. CDK12 regulates DNA repair genes by suppressing intronic polyadenylation. Nature 2018, 564, 141–145.
  47. Vorlová, S.; Rocco, G.; LeFave, C.V.; Jodelka, F.M.; Hess, K.; Hastings, M.L.; Henke, E.; Cartegni, L. Induction of antagonistic soluble decoy receptor tyrosine kinases by intronic polyA activation. Mol. Cell 2011, 43, 927–939.
  48. Adam, S.; Polo, S.E.; Almouzni, G. Transcription recovery after DNA damage requires chromatin priming by the H3.3 histone chaperone HIRA. Cell 2013, 155, 963.
  49. Devany, E.; Park, J.Y.; Murphy, M.R.; Zakusilo, G.; Baquero, J.; Zhang, X.; Hoque, M.; Tian, B.; Kleiman, F.E. Intronic cleavage and polyadenylation regulates gene expression during DNA damage response through U1 snRNA. Cell Discov. 2016, 2, 16013.
  50. Krajewska, M.; Dries, R.; Grassetti, A.V.; Dust, S.; Gao, Y.; Huang, H.; Sharma, B.; Day, D.S.; Kwiatkowski, N.; Pomaville, M.; et al. CDK12 loss in cancer cells affects DNA damage response genes through premature cleavage and polyadenylation. Nat. Commun. 2019, 10, 1757.
  51. Mayr, C. What are 3′ UTRs doing? Cold Spring Harb. Perspect. Biol. 2019, 11, a034728.
  52. Geisberg, J.V.; Moqtaderi, Z.; Fan, X.; Ozsolak, F.; Struhl, K. Global analysis of mRNA isoform half-lives reveals stabilizing and destabilizing elements in yeast. Cell 2014, 156, 812–824.
  53. Srivastava, A.K.; Lu, Y.; Zinta, G.; Lang, Z.; Zhu, J.-K. UTR-dependent control of gene expression in plants. Trends Plant Sci. 2018, 23, 248–259.
  54. Axtell, M.J.; Meyers, B.C. Revisiting criteria for plant microRNA annotation in the era of big data. Plant Cell 2018, 30, 272–284.
  55. Deka, K.; Saha, S. Heat stress induced arginylation of HuR promotes alternative polyadenylation of Hsp70.3 by regulating HuR stability and RNA binding. Cell Death Differ. 2021, 28, 730–747.
  56. Zheng, T.; Tan, Y.; Qiu, J.; Xie, Z.; Hu, X.; Zhang, J.; Na, N. Alternative polyadenylation trans-factor FIP1 exacerbates UUO/IRI-induced kidney injury and contributes to AKI-CKD transition via ROS-NLRP3 axis. Cell Death Dis. 2021, 12, 512.
  57. Ripin, N.; Boudet, J.; Duszczyk, M.M.; Hinniger, A.; Faller, M.; Krepl, M.; Gadi, A.; Schneider, R.J.; Šponer, J.; Meisner-Kober, N.C.; et al. Molecular basis for AU-rich element recognition and dimerization by the HuR C-terminal RRM. Proc. Natl. Acad. Sci. USA 2019, 116, 2935–2944.
  58. Zheng, D.; Wang, R.; Ding, Q.; Wang, T.; Xie, B.; Wei, L.; Zhong, Z.; Tian, B. Cellular stress alters 3′ UTR landscape through alternative polyadenylation and isoform-specific degradation. Nature Commun. 2018, 9, 2268.
  59. Wolf, E.J.; Miles, A.; Lee, E.S.; Nabeel-Shah, S.; Greenblatt, J.F.; Palazzo, A.F.; Tropepe, V.; Emili, A. MKRN2 Physically Interacts with GLE1 to Regulate mRNA Export and Zebrafish Retinal Development. Cell Rep. 2020, 31, 107693.
  60. Rosa-Mercado, N.A.; Steitz, J.A. Who let the DoGs out?—Biogenesis of stress-induced readthrough transcripts. Trends Biochem. Sci. 2022, 47, 206–217.
  61. Hennig, T.; Michalski, M.; Rutkowski, A.J.; Djakovic, L.; Whisnant, A.W.; Friedl, M.-S.; Jha, B.A.; Baptista, M.A.P.; L’Hernault, A.; Erhard, F.; et al. HSV-1-induced disruption of transcription termination resembles a cellular stress response but selectively increases chromatin accessibility downstream of genes. PLoS Pathog. 2018, 14, e1006954.
  62. Vilborg, A.; Passarelli, M.; Yario, T.A.; Tycowski, K.T.; Steitz, J.A. Widespread inducible transcription downstream of human genes. Mol. Cell 2015, 59, 449–461.
  63. Mayr, C. Regulation by 3′-Untranslated regions. Annu. Rev. Genet. 2017, 51, 171–194.
  64. Sandberg, R.; Neilson, J.R.; Sarma, A.; Sharp, P.A.; Burge, C.B. Proliferating cells express mRNAs with shortened 3′ untranslated regions and fewer microRNA target sites. Science 2008, 320, 1643–1647.
  65. Fu, Y.; Chen, L.; Chen, C.; Ge, Y.; Kang, M.; Song, Z.; Li, J.; Feng, Y.; Huo, Z.; He, G.; et al. Crosstalk between alternative polyadenylation and miRNAs in the regulation of protein translational efficiency. Genome Res. 2018, 28, 1656–1663.
  66. Huggins, C.J.; Mayekar, M.K.; Martin, N.; Saylor, K.L.; Gonit, M.; Jailwala, P.; Kasoji, M.; Haines, D.C.; Quiñones, O.A.; Johnson, P.F. C/EBPγ is a critical regulator of cellular stress response networks through heterodimerization with ATF4. Mol. Cell. Biol. 2015, 36, 693–713.
  67. Hunt, A.G. Review: Mechanisms underlying alternative polyadenylation in plants—Looking in the right places. Plant Sci. 2022, 324, 111430.
  68. Deng, X.; Cao, X. Roles of pre-mRNA splicing and polyadenylation in plant development. Curr. Opin. Plant Biol. 2017, 35, 45–53.
  69. Hornyik, C.; Duc, C.; Rataj, K.; Terzi, L.C.; Simpson, G.G. Alternative polyadenylation of antisense RNAs and flowering time control. Biochem. Soc. Trans. 2010, 38, 1077–1081.
  70. Saijo, Y.; Loo, E.P.I. Plant immunity in signal integration between biotic and abiotic stress responses. New Phytol. 2020, 225, 87–104.
  71. VanWallendael, A.; Soltani, A.; Emery, N.C.; Peixoto, M.M.; Olsen, J.; Lowry, D.B. A molecular view of plant local adaptation: Incorporating stress-response networks. Annu. Rev. Plant Biol. 2019, 70, 559–583.
  72. de Lorenzo, L.; Sorenson, R.; Bailey-Serres, J.; Hunt, A.G. Noncanonical alternative polyadenylation contributes to gene regulation in response to hypoxia. Plant Cell 2017, 29, 1262–1277.
  73. Sun, H.-X.; Li, Y.; Niu, Q.-W.; Chua, N.-H. Dehydration stress extends mRNA 3′ untranslated regions with noncoding RNA functions in Arabidopsis. Genome Res. 2017, 27, 1427–1436.
  74. Téllez-Robledo, B.; Manzano, C.; Saez, A.; Navarro-Neila, S.; Silva-Navas, J.; de Lorenzo, L.; González-García, M.; Toribio, R.; Hunt, A.G.; Baigorri, R.; et al. The polyadenylation factor FIP1 is important for plant development and root responses to abiotic stresses. Plant J. 2019, 99, 1203–1219.
  75. Yu, Z.; Lin, J.; Li, Q.Q. Transcriptome analyses of FY mutants reveal its role in mRNA alternative polyadenylation. Plant Cell 2019, 31, 2332–2352.
  76. Chakrabarti, M.; Lorenzo, L.; Abdel-Ghany, S.E.; Reddy, A.S.N.; Hunt, A.G. Wide-ranging transcriptome remodelling mediated by alternative polyadenylation in response to abiotic stresses in Sorghum. Plant J. 2020, 102, 916–930.
  77. Ma, H.; Cai, L.; Lin, J.; Zhou, K.; Li, Q.Q. Divergence in the regulation of the salt tolerant response between Arabidopsis thaliana and its halophytic relative Eutrema Salsugineum by mRNA alternative polyadenylation. Front. Plant Sci. 2022, 13, 866054.
  78. Conesa, C.M.; Saez, A.; Navarro-Neila, S.; de Lorenzo, L.; Hunt, A.G.; Sepúlveda, E.B.; Baigorri, R.; Garcia-Mina, J.M.; Zamarreño, A.M.; Sacristán, S.; et al. Alternative Polyadenylation and Salicylic Acid Modulate Root Responses to Low Nitrogen Availability. Plants 2020, 9, 251.
  79. Higuera, J.J.; Fernandez, E.; Galvan, A. Chlamydomonas NZF1, a tandem-repeated zinc finger factor involved in nitrate signalling by controlling the regulatory gene NIT2. Plant Cell Environ. 2014, 37, 2139–2150.
  80. Wang, C.; Zhang, W.; Li, Z.; Li, Z.; Bi, Y.; Crawford, N.M.; Wang, Y. FIP1 Plays an Important Role in Nitrate Signaling and Regulates CIPK8 and CIPK23 Expression in Arabidopsis. Front. Plant Sci. 2018, 9, 593.
  81. Kindgren, P.; Ard, R.; Ivanov, M.; Marquardt, S. Transcriptional read-through of the long non-coding RNA SVALKA governs plant cold acclimation. Nat. Commun. 2018, 9, 4561.
  82. Zhou, Q.; Fu, H.; Yang, D.; Ye, C.; Zhu, S.; Lin, J.; Ye, W.; Ji, G.; Ye, X.; Wu, X.; et al. Differential alternative polyadenylation contributes to the developmental divergence between two rice subspecies, japonica and indica. Plant J. 2019, 98, 260–276.
  83. Peng, S.; Guo, D.; Guo, Y.; Zhao, H.; Mei, J.; Han, Y.; Guan, R.; Wang, T.; Song, T.; Sun, K.; et al. Constitutive expresser of pathogenesis-related genes 5 is an RNA-binding protein controlling plant immunity via an RNA processing complex. Plant Cell 2022, 34, 1724–1744.
  84. Bruggeman, Q.; Garmier, M.; de Bont, L.; Soubigou-Taconnat, L.; Mazubert, C.; Benhamed, M.; Raynaud, C.; Bergounioux, C.; Delarue, M.; Wang, G.; et al. The polyadenylation factor subunit CLEAVAGE AND POLYADENYLATION SPECIFICITY FACTOR30: A key factor of programmed cell death and a regulator of immunity in Arabidopsis. Plant Physiol. 2014, 165, 732–746.
  85. Cao, J.; Ye, C.; Hao, G.; Dabney-Smith, C.; Hunt, A.G.; Li, Q.Q. Root Hair Single Cell Type Specific Profiles of Gene Expression and Alternative Polyadenylation Under Cadmium Stress. Front. Plant Sci. 2019, 10, 589.
  86. Lyons, R.; Iwase, A.; Gänsewig, T.; Sherstnev, A.; Duc, C.; Barton, G.J.; Hanada, K.; Higuchi-Takeuchi, M.; Matsui, M.; Sugimoto, K.; et al. The RNA-binding protein FPA regulates flg22-triggered defense responses and transcription factor activity by alternative polyadenylation. Sci. Rep. 2013, 3, 2866.
  87. Zhang, J.; Addepalli, B.; Yun, K.-Y.; Hunt, A.G.; Xu, R.; Rao, S.; Li, Q.Q.; Falcone, D.L. A polyadenylation factor subunit implicated in regulating oxidative signaling in Arabidopsis thaliana. PLoS ONE 2008, 3, e2410.
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: 277
Revisions: 3 times (View History)
Update Date: 24 Mar 2023
1000/1000