Acessibilidade / Reportar erro

Interchromosomal translocation in neural progenitor cells exposed to L1 retrotransposition

Abstract

LINE-1 (L1) elements are a class of transposons, comprising approximately 19% and 21% of the mouse and human genomes, respectively. L1 retrotransposons can reverse transcribe their own RNA sequence into a de novo DNA copy integrated into a new genomic location. This activity, known as retrotransposition, may induce genomic alterations, such as insertions and deletions. Interestingly, L1s can retrotranspose and generate more de novo L1 copies in brains than in other somatic tissues. Here, we describe for the first time interchromosomal translocation triggered by ectopic L1 retrotransposition in neural progenitor cells. Such an observation adds to the studies in neurological and psychiatric diseases that exhibited variation in L1 activity between diseased brains compared with controls, suggesting that L1 activity could be detrimental when de-regulated.

Keywords:
L1 retrotransposition; neural progenitor cells; translocations; brain mosaicism

About 20% of the human, rat and mouse genomes are composed of LINE-1 (L1) retrotransposons (Lander et al., 2001Lander ES, Linton LM, Birren B, Nusbaum C, Zody MC, Baldwin J, Devon K, Dewar K, Doyle M, FitzHugh W et al. (2001) Initial sequencing and analysis of the human genome. Nature 409:860-921.; Waterston et al., 2002Waterston RH, Lindblad-Toh K, Birney E, Rogers J, Abril JF, Agarwal P, Agarwala R, Ainscough R, Alexandersson M, An P et al. (2002) Initial sequencing and comparative analysis of the mouse genome. Nature 420:520-562.; Gibbs et al., 2004Gibbs RA, Weinstock GM, Metzker ML, Muzny DM, Sodergren EJ, Scherer S, Scott G, Steffen D, Worley KC, Burch PE et al. (2004) Genome sequence of the brown norway rat yields insights into mammalian evolution. Nature 428:493-521.). Although most L1 elements are retrotransposition defective (Grimaldi et al., 1984Grimaldi G, Skowronski J and Singer MF (1984) Defining the beginning and end of KpnI family segments. Embo J 3:1753-1759.; Moran and Gilbert, 2002Moran JV and Gilbert N (2002) Mammalian LINE-1 retrotransposons and related elements. In: Craig NL, Craigie R, Gellert M and Lambowitz AM (eds). Mobile DNA. Wiley Online, Hoboken, NJ, chapter 35. DOI: 10.1128/9781555817954.ch35.
https://doi.org/10.1128/9781555817954.ch...
), the average human genome is estimated to harbor around 100 active L1 elements, whereas the rat contains roughly 400 and the mouse genome around 3,000 active L1 elements (DeBerardinis et al., 1998DeBerardinis RJ, Goodier JL, Ostertag EM and Kazazian HH Jr. (1998) Rapid amplification of a retrotransposon subfamily is evolving the mouse genome. Nat Genet 20:288-290.; Goodier et al., 2001Goodier JL, Ostertag EM, Du K and Kazazian HHJr. (2001) A novel active L1 retrotransposon subfamily in the mouse. Genome Res 11:1677-1685.; Brouha et al., 2003Brouha B, Schustak J, Badge RM, Lutz-Prigge S, Farley AH, Moran JV and Kazazian HH Jr. (2003) Hot L1s account for the bulk of retrotransposition in the human population. Proc Natl Acad Sci U S A 100:5280-5285.; Penzkofer et al., 2005Penzkofer T, Dandekar T and Zemojtel T (2005) L1Base: From functional annotation to prediction of active LINE-1 elements. Nucleic Acids Res 33:D498-500.). Active L1 retrotransposons can affect the genome in various ways, creating insertions, deletions, processed pseudogenes, new splice sites, or fine-tuning gene expression, revealing incredible flexibility for genetic manipulation upon integration (Kazazian, 1998Kazazian HH Jr. (1998) Mobile elements and disease. Curr Opin Genet Dev 8:343-350.; Esnault et al., 2000Esnault C, Maestre J and Heidmann T (2000) Human LINE retrotransposons generate processed pseudogenes. Nat Genet 24:363-367.; Wei et al., 2001Wei W, Gilbert N, Ooi SL, Lawler JF, Ostertag EM, Kazazian HH, Boeke JD and Moran JV (2001) Human L1 retrotransposition: Cis preference versus trans complementation. Mol Cell Biol 21:1429-1439.; Dewannieux et al., 2003Dewannieux M, Esnault C and Heidmann T (2003) LINE-mediated retrotransposition of marked Alu sequences. Nat Genet 35:41-48.; Perepelitsa-Belancio and Deininger, 2003Perepelitsa-Belancio V and Deininger P (2003) RNA truncation by premature polyadenylation attenuates human mobile element activity. Nat Genet 35:363-366.; Han et al., 2004Han JS, Szak ST and Boeke JD (2004) Transcriptional disruption by the L1 retrotransposon and implications for mammalian transcriptomes. Nature 429:268-274.). Human L1s structurally resemble those present in rodent genomes, and the advent of a cultured cell retrotransposition assay has revealed that human L1s can retrotranspose in a variety of mammalian cell lines (Moran et al., 1996Moran JV, Holmes SE, Naas TP, DeBerardinis RJ, Boeke JD and Kazazian HH Jr. (1996) High frequency retrotransposition in cultured mammalian cells. Cell 87:917-927.; Wei et al., 2000Wei W, Morrish TA, Alisch RS and Moran JV (2000) A transient assay reveals that cultured human cells can accommodate multiple LINE-1 retrotransposition events. Anal Biochem 284:435-438.; Morrish et al., 2002Morrish TA, Gilbert N, Myers JS, Vincent BJ, Stamato TD, Taccioli GE, Batzer MA and Moran JV (2002) DNA repair mediated by endonuclease-independent LINE-1 retrotransposition. Nat Genet 31:159-165.; Han and Boeke, 2004Han JS and Boeke JD (2004) A highly active synthetic mammalian retrotransposon. Nature 429:314-318.; Muotri, 2016Muotri AR (2016) L1 Retrotransposition in neural progenitor cells. Methods Mol Biol 1400:157-163. ; Macia et al., 2017Macia A, Widmann TJ, Heras SR, Ayllon V, Sanchez L, Benkaddour-Boumzaouad M, Munoz-Lopez M, Rubio A, Amador-Cubero S, Blanco-Jimenez E et al. (2017) Engineered LINE-1 retrotransposition in nondividing human neurons. Genome Res 27:335-348. ).

Previous studies have indicated that L1 retrotransposition can occur in germ cells or in early embryogenesis before the germ line becomes a distinct lineage (Ostertag et al., 2002Ostertag EM, DeBerardinis RJ, Goodier JL, Zhang Y, Yang N, Gerton GL and Kazazian HH Jr. (2002) A mouse model of human L1 retrotransposition. Nat Genet 32:655-660.; Prak et al., 2003Prak ET, Dodson AW, Farkash EA and Kazazian HH Jr. (2003) Tracking an embryonic L1 retrotransposition event. Proc Natl Acad Sci U S A 1001:832-1837.; Garcia-Perez et al., 2007Garcia-Perez JL, Marchetto MC, Muotri AR, Coufal NG, Gage FH, O’Shea KS and Moran JV (2007) LINE-1 retrotransposition in human embryonic stem cells. Hum Mol Genet 16:1569-1577. ), and cultured cell retrotransposition assay has revealed that human and mouse L1 elements can retrotranspose in a variety of transformed or immortalized cultured cell lines (Moran et al., 1996Moran JV, Holmes SE, Naas TP, DeBerardinis RJ, Boeke JD and Kazazian HH Jr. (1996) High frequency retrotransposition in cultured mammalian cells. Cell 87:917-927.; Morrish et al., 2002Morrish TA, Gilbert N, Myers JS, Vincent BJ, Stamato TD, Taccioli GE, Batzer MA and Moran JV (2002) DNA repair mediated by endonuclease-independent LINE-1 retrotransposition. Nat Genet 31:159-165.; Han et al., 2004Han JS, Szak ST and Boeke JD (2004) Transcriptional disruption by the L1 retrotransposon and implications for mammalian transcriptomes. Nature 429:268-274.). Previous data showed that neuronal progenitor cells (NPCs) support L1 retrotransposition during neuronal differentiation in vitro and in vivo, resulting in a L1 genetic mosaic nervous system (Muotri et al., 2005Muotri AR, Chu VT, Marchetto MC, Deng W, Moran JV and Gage FH (2005) Somatic mosaicism in neuronal precursor cells mediated by L1 retrotransposition. Nature 435:903-910.; Muotri, 2016Muotri AR (2016) L1 Retrotransposition in neural progenitor cells. Methods Mol Biol 1400:157-163. ; Macia et al., 2017Macia A, Widmann TJ, Heras SR, Ayllon V, Sanchez L, Benkaddour-Boumzaouad M, Munoz-Lopez M, Rubio A, Amador-Cubero S, Blanco-Jimenez E et al. (2017) Engineered LINE-1 retrotransposition in nondividing human neurons. Genome Res 27:335-348. ). In NPCs in culture, L1 retrotransposons can insert near to or within neuron-associated genes, such as olfactory receptors, ion channel-associated genes, and cadherin receptors. An L1 insertion in the promoter region of the Psd-93 gene, encoding a post-synaptic density protein involved in different aspects of synapse formation (Conroy et al., 2003Conroy WG, Liu Z, Nai Q, Coggan JS and Berg DK (2003) PDZ-containing proteins provide a functional postsynaptic scaffold for nicotinic receptors in neurons. Neuron 38:759-771.; Tao et al., 2003Tao YX, Rumbaugh G, Wang GD, Petralia RS, Zhao C, Kauer FW, Tao F, Zhuo M, Wenthold RJ, Raja SN et al. (2003) Impaired NMDA receptor-mediated postsynaptic function and blunted NMDA receptor-dependent persistent pain in mice lacking postsynaptic density-93 protein. J Neurosci 23:6703-6712.; Parker et al., 2004Parker MJ, Zhao S, Bredt DS, Sanes JR and Feng G (2004) PSD93 regulates synaptic stability at neuronal cholinergic synapses. J Neurosci 24:378-388.), significantly increased gene expression level and, consequently, accelerated neuronal maturation in culture (Muotri et al., 2005Muotri AR (2016) L1 Retrotransposition in neural progenitor cells. Methods Mol Biol 1400:157-163. ). An analysis of the sequence data from several L1 insertions in NPCs indicated that the integration process might be regulated and probably targeted open chromatin regions, but size of the target genes might also be a factor (Thomas et al., 2012Thomas CA, Paquola AC and Muotri AR (2012) LINE-1 retrotransposition in the nervous system. Annu Rev Cell Dev Biol 28:555-573. ). It is challenging to determine or precisely predict the consequences of L1 endogenous retrotransposition in neurons. Virtually any RNA molecule can be subject to in trans retrotransposition if hijacked by L1 machinery (Esnault et al., 2000Esnault C, Maestre J and Heidmann T (2000) Human LINE retrotransposons generate processed pseudogenes. Nat Genet 24:363-367.; Wei et al., 2001Wei W, Gilbert N, Ooi SL, Lawler JF, Ostertag EM, Kazazian HH, Boeke JD and Moran JV (2001) Human L1 retrotransposition: Cis preference versus trans complementation. Mol Cell Biol 21:1429-1439.; Kajikawa and Okada, 2002Kajikawa M and Okada N (2002) LINEs mobilize SINEs in the eel through a shared 3’ sequence. Cell 111:433-444.; Dewannieux et al., 2003Dewannieux M, Esnault C and Heidmann T (2003) LINE-mediated retrotransposition of marked Alu sequences. Nat Genet 35:41-48.). Thus, each developing neuron can potentially carry several L1-mediated events, and if some of the resulting insertions occur in genes expressed during neuronal development, it is possible that neuronal networks during brain development could be significantly impacted by de novo L1 insertions (Muotri and Gage, 2006Muotri AR and Gage FH (2006) Generation of neuronal variability and complexity. Nature 441:1087-1093.; Cao et al., 2006Cao X, Yeo G, Muotri AR, Kuwabara T and Gage FH (2006) Noncoding RNAs in the mammalian central nervous system. Annu Rev Neurosci 29:77-103.; Baillie et al., 2011Baillie JK, Barnett MW, Upton KR, Gerhardt DJ, Richmond TA, De Sapio F, Brennan PM, Rizzu P, Smith S, Fell M et al. (2011) Somatic retrotransposition alters the genetic landscape of the human brain. Nature 479:534-537.; Evrony et al., 2012Evrony GD, Cai X, Lee E, Hills LB, Elhosary PC, Lehmann HS, Parker JJ, Atabay KD, Gilmore EC, Poduri A et al. (2012) Single-neuron sequencing analysis of L1 retrotransposition and somatic mutation in the human brain. Cell 151:483-496. ; Jacob-Hirsch et al., 2018Jacob-Hirsch J, Eyal E, Knisbacher BA, Roth J, Cesarkas K, Dor C, Farage-Barhom S, Kunik V, Simon AJ, Gal M et al. (2018) Whole-genome sequencing reveals principles of brain retrotransposition in neurodevelopmental disorders. Cell Res 28:187-203. ; Shpyleva et al.,2018Shpyleva S, Melnyk S, Pavliv O, Pogribny I and Jill James S (2018) Overexpression of LINE-1 retrotransposons in autism brain. Mol Neurobiol 55:1740-1749. ; Moszczynska, 2020Moszczynska A (2020) Differential Responses of LINE-1 in the dentate gyrus, striatum and prefrontal cortex to chronic neurotoxic Methamphetamine: A study in rat brain. Genes (Basel) 11:364. ; Bundo and Iwamoto, 2023Bundo M and Iwamoto K (2023) A method for detection of somatic LINE-1 insertions at the single-cell level from postmortem human brain. Methods Mol Biol 2577:147-159.). The characterization of L1 insertions in NPCs reveals if the neural intracellular milieu can interfere with L1 insertion, helping to pave the way for a better understanding of the role of L1 retrotransposition in the nervous system (Thomas et al.,2012Thomas CA, Paquola AC and Muotri AR (2012) LINE-1 retrotransposition in the nervous system. Annu Rev Cell Dev Biol 28:555-573. ; Suarez et al., 2018Suarez NA, Macia A and Muotri AR (2018) LINE-1 retrotransposons in healthy and diseased human brain. Dev Neurobiol 78:434-455. ).

Here, we describe the analysis of an interchromosomal translocation potentially triggered by ectopic L1 retrotransposition in an NPC clone derived from stable rat neural stem cells in culture (HCN). HCN cells represent a heterogeneous population composed of neural stem cells and more differentiated NPCs. An episomal retrotransposition-competent human active L1 element (LRE3) controlled by its endogenous promoter and carried an EGFP reporter construct was used to obtain the Clone C6+ (Muotri et al., 2005Muotri AR, Chu VT, Marchetto MC, Deng W, Moran JV and Gage FH (2005) Somatic mosaicism in neuronal precursor cells mediated by L1 retrotransposition. Nature 435:903-910.). Briefly, the EGFP gene is interrupted by the g-globin IVS2 intron in the same transcriptional orientation as the L1 transcript. This arrangement ensures that EGFP-positive cells will arise only when a transcript initiated from the promoter driving L1 expression is spliced, reverse transcribed, and integrated into chromosomal DNA, thereby allowing expression of the EGFP gene from the pCMV promoter (Moran et al., 1996Moran JV, Holmes SE, Naas TP, DeBerardinis RJ, Boeke JD and Kazazian HH Jr. (1996) High frequency retrotransposition in cultured mammalian cells. Cell 87:917-927.; Ostertag et al., 2000Ostertag EM, Prak ET, DeBerardinis RJ, Moran JV and Kazazian HH Jr. (2000) Determination of L1 retrotransposition kinetics in cultured cells. Nucleic Acids Res 28:1418-1423.) (Figure 1a). Cells harboring the L1 expression constructs were selected by the addition of puromycin to the culture medium, and puromycin-resistant cells were screened for EGFP expression by flow cytometry, as previously described (Muotri et al., 2005Muotri AR, Chu VT, Marchetto MC, Deng W, Moran JV and Gage FH (2005) Somatic mosaicism in neuronal precursor cells mediated by L1 retrotransposition. Nature 435:903-910.). The clone C6+ comes from a single EGFP-positive puromycin-resistant cell that was collected by fluorescence-activated cell sorting (FACS) 7 days post-electroporation (Figure 1b). As previously reported, the EGFP expression was reduced over time due to epigenetic modifications at the L1-EGFP insertion site (Muotri et al., 2005Muotri AR, Chu VT, Marchetto MC, Deng W, Moran JV and Gage FH (2005) Somatic mosaicism in neuronal precursor cells mediated by L1 retrotransposition. Nature 435:903-910.). Sequencing of the PCR product from the clone C6+ confirmed that the intron in the EGFP gene was spliced precisely (Figure 1c).

Figure 1 -
Isolation of clone C6+ by L1-EGFP retrotransposition. a, The retrotransposition-competent human L1 (L1RP) contains a 5’ untranslated region (UTR) that harbors an internal promoter, two open reading frames (ORF1 and ORF2; not drawn to scale), and a 3’ UTR that ends in a poly (A) tail. ORF2 contains an endonuclease (EN) and reverse transcriptase (RT) domains, as well as a cystiyne-rich 3’ end (C). The EGFP retrotransposition indicator cassette consists of a backward copy of the EGFP gene whose expression is controlled by the human cytomegalovirus major immediate early promoter (pCMV) and the herpes simplex virus thymidine kinase polyadenylation sequence (pA). This arrangement ensures that EGFP expression will only become activated upon L1 retrotransposition. The black arrows indicate PCR primers flanking the intron present in the EGFP gene. b, Bright field and UV images are shown of clone C6+ cells containing an L1 retrotransposition event derived from a single cell. Bar = 20 mm. c, PCR analysis of genomic DNA isolated from the clone C6+. PCR was conducted using the primers shown in panel a. The 1243-bp PCR product corresponds to the original L1 vector harboring the intron-containing EGFP indicator cassette. The 343-bp PCR product, diagnostic for the loss of the intron, is indicative of a retrotransposition event. Sequencing of the 343-bp PCR product confirmed the precise splicing of the intron (data not shown).

Previously characterized L1 clones in NPCs showed a neuronal differentiation tendency using a mixed differentiation protocol (Muotri et al., 2005Muotri AR, Chu VT, Marchetto MC, Deng W, Moran JV and Gage FH (2005) Somatic mosaicism in neuronal precursor cells mediated by L1 retrotransposition. Nature 435:903-910.). Then, we next examined whether the clone C6+ also has a similar neuronal predisposition. Interestingly, the ability of this clone to differentiate in neurons is 1,6 times higher compared to the original bulk HCN cells. Moreover, such neuronal tendency is gained mainly at the expense of glial cells since the number of astrocytes in the clone C6+ is reduced by 50% after differentiation (Figure 2).

Figure 2 -
Neural differentiation for the clone C6+. After a mixed differentiation protocol using retinoic acid and serum (see Supplementary Material file Methods Methods. ), cells were stained and quantified for neuronal (Map2ab), astrocytes (GFAP), or oligodendrocytes (RIP) markers. These experiments were performed in triplicates (n=3) and the bars represent standard deviation.

Inverse PCR was used to characterize the post-integration site from clone C6+ (Morrish et al., 2002Morrish TA, Gilbert N, Myers JS, Vincent BJ, Stamato TD, Taccioli GE, Batzer MA and Moran JV (2002) DNA repair mediated by endonuclease-independent LINE-1 retrotransposition. Nat Genet 31:159-165.). The insertion occurred at a preferred L1 EN consensus cleavage site (5’ - TTT/A - 3’) located on chromosome Xq22. This region is flanked by a small region on chromosome 10, suggesting a putative translocation between chromosome X and 10 (t(x;10)) upon L1 retrotransposition. The insertion site has a small TSD and a long polyA. The L1-EGFP element was ORF2 truncated at 6,176 bp (Figure 3). PCR analysis at this site to detect if the inversion was previously present in the parental HCN cells had proven difficult because of the presence of repetitive sequences nearby. Alternatively, we used rat probes for chromosome X in both HCN and clone C6+ mitotic chromosome spreads. In HCN cells, the FISH (fluorescence in situ hybridization) reaction revealed the presence of two signals in two independent chromosomes in HCN cells, suggesting the presence of two X chromosomes, as expected for a female-derived cell line. However, clone C6+ cells displayed an extra signal in a third chromosome (Figure 4). Finally, we use SKY (Spectral Karyotype) to confirm chromosome 10 as the other chromosomal arm in the putative translocation (Figure 4). Interestingly, the SKY also revealed that some cells from the C6+ clone also carry additional non-clonal chromosome aberrations, such as t(12;7) and t(15;6), which displays the potential for this cell line to change after long periods in culture. Such instability was not observed in the parental HCN cells, suggesting that chromosomal instability appears only in NPCs but not in neural stem cells.

Figure 3 -
Schematic representation of the Clone 6+ L1 insertion. Inverse PCR indicated that the L1 was inserted in the antisense orientation of the dystrophin major muscle isoform (chromosome Xq).

Figure 4 -
Inverse PCR putative interchromosomal translocation confirmation in Clone C6+ cells. Isolated cellular chromosomal spread in metaphase hybridized with X paint probe (arrowheads) and a BAC probe control for chromosome 3 (arrows) illustrates the extra fluorescence signal observed in the Clone C6+ but absence in the HCN parental cell. Bottom: spectral analysis of chromosomal spreads in the Clone C6+ revealed a translocation between chromosome X (brown) and chromosome 10 (blue).

Chromosomal translocations caused by L1 retrotransposons were proposed as a mechanism to explain the evolution of recent rodent L1 subfamilies and the occurrence of promoters swapping for non-LTR retroelements in certain avian species (Hayward et al., 1997Hayward BE, Zavanelli M and Furano AV (1997) Recombination creates novel L1 (LINE-1) elements in Rattus norvegicus. Genetics 146:641-654.; Saxton and Martin, 1998Saxton JA and Martin SL (1998) Recombination between subtypes creates a mosaic lineage of LINE-1 that is expressed and actively retrotransposing in the mouse genome. J Mol Biol 280:611-622.; Haas et al., 2001Haas NB, Grabowski JM, North J, Moran JV, Kazazian HH and Burch JB (2001) Subfamilies of CR1 non-LTR retrotransposons have different 5’UTR sequences but are otherwise conserved. Gene 265:175-183.). A de novo L1 insertion using a similar system described here was previously described to map a putative interchromosomal translocation in HeLa cells (Gilbert et al., 2005Gilbert N, Lutz S, Morrish TA and Moran JV (2005) Multiple fates of L1 retrotransposition intermediates in cultured human cells. Mol Cell Biol 25:7780-7795.). The fact that we observed a similar L1 insertion in HeLa and NPCs suggests that the intracellular environment requirements for interchromosomal translocation are present in both cell types. Together with this previous observation, we believe interchromosomal translocation can be regarded as a new genetic strategy by which L1 retrotransposons can shuffle DNA to new genomic locations.

The L1 target in the clone C6+ was inside the Loc317538 in chromosome Xq22, which is a region like the dystrophin major muscle isoform. Alterations in gene expression in this region do not seem to cause the strong neurogenic effect observed in this clone. However, the SKY experiment also revealed other, less frequent chromosomal translocations that may contribute to the final phenotype. While our data was observed using an ectopic L1 reporter construct, it is currently unknown if these other interchromosomal translocations were caused by endogenous L1 retrotransposition.

A chromosomal inversion involving parental L1 retrotransposon polymorphism has been correlated to autism in siblings (Vincent et al., 2006Vincent JB, Horike SI, Choufani S, Paterson AD, Roberts W, Szatmari P, Weksberg R, Fernandez B and Scherer SW (2006) An inversion inv(4)(p12-p15.3) in autistic siblings implicates the 4p GABA receptor gene cluster. J Med Genet 43:429-434.). At a somatic level, early reports applying FISH using individual chromosomal probes in postmitotic neurons revealed an extra signal, additional from two fluorescence spots expected for diploid cells, both in normal and Alzheimer’s brain tissues (Yang et al., 2001Yang Y, Geldmacher DS and Herrup K (2001) DNA replication precedes neuronal cell death in Alzheimer’s disease. J Neurosci 21:2661-2668.). Two interpretations for this phenomenon were proposed: (i) the extra signal could be related to a possible cell-cycle re-entering process associated with neuronal death, which would be more frequent in the disease brain (Herrup and Yang, 2007Herrup K and Yang Y (2007) Cell cycle regulation in the postmitotic neuron: Oxymoron or new biology? Nat Rev Neurosci 8:368-378.; Yang and Herrup, 2007Yang Y and Herrup K (2007) Cell division in the CNS: Protective response or lethal event in post-mitotic neurons? Biochim Biophys Acta 1772:457-466.; Pandey and Vinod, 2022Pandey N and Vinod PK (2022) Model scenarios for cell cycle re-entry in Alzheimer’s disease. iScience 25:104543.; Yuen et al., 2022Yuen SC, Lee SM and Leung SW (2022) Putative factors interfering cell cycle re-entry in Alzheimer’s disease: An omics study with differential expression meta-analytics and co-expression profiling. J Alzheimers Dis 85:1373-1398. ; Wong and Chow, 2022Wong GC-N and Chow KH-M (2022) DNA damage response-associated cell cycle re-rntry and neuronal senescence in brain aging and Alzheimer’s Disease. J Alzheimers Dis. DOI: 10.3233/JAD-220203.
https://doi.org/10.3233/JAD-220203...
), or (ii) the extra signal may represent a gain of a chromosome in a constitutional and physiological aneuploidy process generated during brain development (Rehen et al., 2001Rehen SK, McConnell MJ, Kaushal D, Kingsbury MA, Yang AH and Chun J (2001) Chromosomal variation in neurons of the developing and adult mammalian nervous system. Proc Natl Acad Sci U S A 98:13361-13366., 2005Rehen SK, Yung YC, McCreight MP, Kaushal D, Yang AH, Almeida BS, Kingsbury MA, Cabral KM, McConnell MJ, Anliker B et al. (2005) Constitutional aneuploidy in the normal human brain. J Neurosci 25:2176-2180.; Arendt, 2012Arendt T (2012) Cell cycle activation and aneuploid neurons in Alzheimer’s disease. Mol Neurobiol 46:125-135.; Bajic et al., 2015Bajic V, Spremo-Potparevic B, Zivkovic L, Isenovic ER and Arendt T (2015) Cohesion and the aneuploid phenotype in Alzheimer’s disease: A tale of genome instability. Neurosci Biobehav Rev 55:365-374.). Based on our experimental data, we would like to propose a third, non-exclusive interpretation: the extra signal revealed by FISH in postmitotic neurons may be a consequence of a genetic rearrangement, such as interchromosomal translocation or duplication, caused by L1 retrotransposition in NPCs enrolled in neuronal differentiation. Our hypothesis agrees with the view that terminally differentiated cells, such as neurons, accommodate situations of genetic instability to dispense with the energy cost of maintaining global DNA repair systems (Nouspikel and Hanawalt, 2000Nouspikel T and Hanawalt PC (2000) Terminally differentiated human neurons repair transcribed genes but display attenuated global DNA repair and modulation of repair gene expression. Mol Cell Biol 20:1562-1570., 2002Nouspikel T and Hanawalt PC (2002) DNA repair in terminally differentiated cells. DNA Repair (Amst) 1:59-75., 2003Nouspikel T and Hanawalt PC (2003) When parsimony backfires: Neglecting DNA repair may doom neurons in Alzheimer’s disease. Bioessays 25:168-173.).

Acknowledgments

This work was supported by discretionary funds from the Muotri lab.

References

  • Arendt T (2012) Cell cycle activation and aneuploid neurons in Alzheimer’s disease. Mol Neurobiol 46:125-135.
  • Baillie JK, Barnett MW, Upton KR, Gerhardt DJ, Richmond TA, De Sapio F, Brennan PM, Rizzu P, Smith S, Fell M et al (2011) Somatic retrotransposition alters the genetic landscape of the human brain. Nature 479:534-537.
  • Bajic V, Spremo-Potparevic B, Zivkovic L, Isenovic ER and Arendt T (2015) Cohesion and the aneuploid phenotype in Alzheimer’s disease: A tale of genome instability. Neurosci Biobehav Rev 55:365-374.
  • Brouha B, Schustak J, Badge RM, Lutz-Prigge S, Farley AH, Moran JV and Kazazian HH Jr. (2003) Hot L1s account for the bulk of retrotransposition in the human population. Proc Natl Acad Sci U S A 100:5280-5285.
  • Bundo M and Iwamoto K (2023) A method for detection of somatic LINE-1 insertions at the single-cell level from postmortem human brain. Methods Mol Biol 2577:147-159.
  • Cao X, Yeo G, Muotri AR, Kuwabara T and Gage FH (2006) Noncoding RNAs in the mammalian central nervous system. Annu Rev Neurosci 29:77-103.
  • Conroy WG, Liu Z, Nai Q, Coggan JS and Berg DK (2003) PDZ-containing proteins provide a functional postsynaptic scaffold for nicotinic receptors in neurons. Neuron 38:759-771.
  • DeBerardinis RJ, Goodier JL, Ostertag EM and Kazazian HH Jr. (1998) Rapid amplification of a retrotransposon subfamily is evolving the mouse genome. Nat Genet 20:288-290.
  • Dewannieux M, Esnault C and Heidmann T (2003) LINE-mediated retrotransposition of marked Alu sequences. Nat Genet 35:41-48.
  • Esnault C, Maestre J and Heidmann T (2000) Human LINE retrotransposons generate processed pseudogenes. Nat Genet 24:363-367.
  • Evrony GD, Cai X, Lee E, Hills LB, Elhosary PC, Lehmann HS, Parker JJ, Atabay KD, Gilmore EC, Poduri A et al (2012) Single-neuron sequencing analysis of L1 retrotransposition and somatic mutation in the human brain. Cell 151:483-496.
  • Garcia-Perez JL, Marchetto MC, Muotri AR, Coufal NG, Gage FH, O’Shea KS and Moran JV (2007) LINE-1 retrotransposition in human embryonic stem cells. Hum Mol Genet 16:1569-1577.
  • Gibbs RA, Weinstock GM, Metzker ML, Muzny DM, Sodergren EJ, Scherer S, Scott G, Steffen D, Worley KC, Burch PE et al (2004) Genome sequence of the brown norway rat yields insights into mammalian evolution. Nature 428:493-521.
  • Gilbert N, Lutz S, Morrish TA and Moran JV (2005) Multiple fates of L1 retrotransposition intermediates in cultured human cells. Mol Cell Biol 25:7780-7795.
  • Goodier JL, Ostertag EM, Du K and Kazazian HHJr. (2001) A novel active L1 retrotransposon subfamily in the mouse. Genome Res 11:1677-1685.
  • Grimaldi G, Skowronski J and Singer MF (1984) Defining the beginning and end of KpnI family segments. Embo J 3:1753-1759.
  • Haas NB, Grabowski JM, North J, Moran JV, Kazazian HH and Burch JB (2001) Subfamilies of CR1 non-LTR retrotransposons have different 5’UTR sequences but are otherwise conserved. Gene 265:175-183.
  • Han JS and Boeke JD (2004) A highly active synthetic mammalian retrotransposon. Nature 429:314-318.
  • Han JS, Szak ST and Boeke JD (2004) Transcriptional disruption by the L1 retrotransposon and implications for mammalian transcriptomes. Nature 429:268-274.
  • Hayward BE, Zavanelli M and Furano AV (1997) Recombination creates novel L1 (LINE-1) elements in Rattus norvegicus. Genetics 146:641-654.
  • Herrup K and Yang Y (2007) Cell cycle regulation in the postmitotic neuron: Oxymoron or new biology? Nat Rev Neurosci 8:368-378.
  • Jacob-Hirsch J, Eyal E, Knisbacher BA, Roth J, Cesarkas K, Dor C, Farage-Barhom S, Kunik V, Simon AJ, Gal M et al (2018) Whole-genome sequencing reveals principles of brain retrotransposition in neurodevelopmental disorders. Cell Res 28:187-203.
  • Kajikawa M and Okada N (2002) LINEs mobilize SINEs in the eel through a shared 3’ sequence. Cell 111:433-444.
  • Kazazian HH Jr. (1998) Mobile elements and disease. Curr Opin Genet Dev 8:343-350.
  • Lander ES, Linton LM, Birren B, Nusbaum C, Zody MC, Baldwin J, Devon K, Dewar K, Doyle M, FitzHugh W et al (2001) Initial sequencing and analysis of the human genome. Nature 409:860-921.
  • Macia A, Widmann TJ, Heras SR, Ayllon V, Sanchez L, Benkaddour-Boumzaouad M, Munoz-Lopez M, Rubio A, Amador-Cubero S, Blanco-Jimenez E et al (2017) Engineered LINE-1 retrotransposition in nondividing human neurons. Genome Res 27:335-348.
  • Moran JV and Gilbert N (2002) Mammalian LINE-1 retrotransposons and related elements. In: Craig NL, Craigie R, Gellert M and Lambowitz AM (eds). Mobile DNA. Wiley Online, Hoboken, NJ, chapter 35. DOI: 10.1128/9781555817954.ch35.
    » https://doi.org/10.1128/9781555817954.ch35
  • Moran JV, Holmes SE, Naas TP, DeBerardinis RJ, Boeke JD and Kazazian HH Jr. (1996) High frequency retrotransposition in cultured mammalian cells. Cell 87:917-927.
  • Morrish TA, Gilbert N, Myers JS, Vincent BJ, Stamato TD, Taccioli GE, Batzer MA and Moran JV (2002) DNA repair mediated by endonuclease-independent LINE-1 retrotransposition. Nat Genet 31:159-165.
  • Moszczynska A (2020) Differential Responses of LINE-1 in the dentate gyrus, striatum and prefrontal cortex to chronic neurotoxic Methamphetamine: A study in rat brain. Genes (Basel) 11:364.
  • Muotri AR (2016) L1 Retrotransposition in neural progenitor cells. Methods Mol Biol 1400:157-163.
  • Muotri AR, Chu VT, Marchetto MC, Deng W, Moran JV and Gage FH (2005) Somatic mosaicism in neuronal precursor cells mediated by L1 retrotransposition. Nature 435:903-910.
  • Muotri AR and Gage FH (2006) Generation of neuronal variability and complexity. Nature 441:1087-1093.
  • Nouspikel T and Hanawalt PC (2000) Terminally differentiated human neurons repair transcribed genes but display attenuated global DNA repair and modulation of repair gene expression. Mol Cell Biol 20:1562-1570.
  • Nouspikel T and Hanawalt PC (2002) DNA repair in terminally differentiated cells. DNA Repair (Amst) 1:59-75.
  • Nouspikel T and Hanawalt PC (2003) When parsimony backfires: Neglecting DNA repair may doom neurons in Alzheimer’s disease. Bioessays 25:168-173.
  • Ostertag EM, DeBerardinis RJ, Goodier JL, Zhang Y, Yang N, Gerton GL and Kazazian HH Jr. (2002) A mouse model of human L1 retrotransposition. Nat Genet 32:655-660.
  • Ostertag EM, Prak ET, DeBerardinis RJ, Moran JV and Kazazian HH Jr. (2000) Determination of L1 retrotransposition kinetics in cultured cells. Nucleic Acids Res 28:1418-1423.
  • Pandey N and Vinod PK (2022) Model scenarios for cell cycle re-entry in Alzheimer’s disease. iScience 25:104543.
  • Parker MJ, Zhao S, Bredt DS, Sanes JR and Feng G (2004) PSD93 regulates synaptic stability at neuronal cholinergic synapses. J Neurosci 24:378-388.
  • Penzkofer T, Dandekar T and Zemojtel T (2005) L1Base: From functional annotation to prediction of active LINE-1 elements. Nucleic Acids Res 33:D498-500.
  • Perepelitsa-Belancio V and Deininger P (2003) RNA truncation by premature polyadenylation attenuates human mobile element activity. Nat Genet 35:363-366.
  • Prak ET, Dodson AW, Farkash EA and Kazazian HH Jr. (2003) Tracking an embryonic L1 retrotransposition event. Proc Natl Acad Sci U S A 1001:832-1837.
  • Rehen SK, McConnell MJ, Kaushal D, Kingsbury MA, Yang AH and Chun J (2001) Chromosomal variation in neurons of the developing and adult mammalian nervous system. Proc Natl Acad Sci U S A 98:13361-13366.
  • Rehen SK, Yung YC, McCreight MP, Kaushal D, Yang AH, Almeida BS, Kingsbury MA, Cabral KM, McConnell MJ, Anliker B et al (2005) Constitutional aneuploidy in the normal human brain. J Neurosci 25:2176-2180.
  • Saxton JA and Martin SL (1998) Recombination between subtypes creates a mosaic lineage of LINE-1 that is expressed and actively retrotransposing in the mouse genome. J Mol Biol 280:611-622.
  • Shpyleva S, Melnyk S, Pavliv O, Pogribny I and Jill James S (2018) Overexpression of LINE-1 retrotransposons in autism brain. Mol Neurobiol 55:1740-1749.
  • Suarez NA, Macia A and Muotri AR (2018) LINE-1 retrotransposons in healthy and diseased human brain. Dev Neurobiol 78:434-455.
  • Tao YX, Rumbaugh G, Wang GD, Petralia RS, Zhao C, Kauer FW, Tao F, Zhuo M, Wenthold RJ, Raja SN et al (2003) Impaired NMDA receptor-mediated postsynaptic function and blunted NMDA receptor-dependent persistent pain in mice lacking postsynaptic density-93 protein. J Neurosci 23:6703-6712.
  • Thomas CA, Paquola AC and Muotri AR (2012) LINE-1 retrotransposition in the nervous system. Annu Rev Cell Dev Biol 28:555-573.
  • Vincent JB, Horike SI, Choufani S, Paterson AD, Roberts W, Szatmari P, Weksberg R, Fernandez B and Scherer SW (2006) An inversion inv(4)(p12-p15.3) in autistic siblings implicates the 4p GABA receptor gene cluster. J Med Genet 43:429-434.
  • Waterston RH, Lindblad-Toh K, Birney E, Rogers J, Abril JF, Agarwal P, Agarwala R, Ainscough R, Alexandersson M, An P et al (2002) Initial sequencing and comparative analysis of the mouse genome. Nature 420:520-562.
  • Wei W, Gilbert N, Ooi SL, Lawler JF, Ostertag EM, Kazazian HH, Boeke JD and Moran JV (2001) Human L1 retrotransposition: Cis preference versus trans complementation. Mol Cell Biol 21:1429-1439.
  • Wei W, Morrish TA, Alisch RS and Moran JV (2000) A transient assay reveals that cultured human cells can accommodate multiple LINE-1 retrotransposition events. Anal Biochem 284:435-438.
  • Wong GC-N and Chow KH-M (2022) DNA damage response-associated cell cycle re-rntry and neuronal senescence in brain aging and Alzheimer’s Disease. J Alzheimers Dis. DOI: 10.3233/JAD-220203.
    » https://doi.org/10.3233/JAD-220203
  • Yang Y, Geldmacher DS and Herrup K (2001) DNA replication precedes neuronal cell death in Alzheimer’s disease. J Neurosci 21:2661-2668.
  • Yang Y and Herrup K (2007) Cell division in the CNS: Protective response or lethal event in post-mitotic neurons? Biochim Biophys Acta 1772:457-466.
  • Yuen SC, Lee SM and Leung SW (2022) Putative factors interfering cell cycle re-entry in Alzheimer’s disease: An omics study with differential expression meta-analytics and co-expression profiling. J Alzheimers Dis 85:1373-1398.

Supplementary material

The following online material is available for this article:

Methods.
Associate Editor: Carlos F. M. Menck

Publication Dates

  • Publication in this collection
    30 Jan 2023
  • Date of issue
    2023

History

  • Received
    15 Sept 2022
  • Accepted
    20 Dec 2022
Sociedade Brasileira de Genética Rua Cap. Adelmio Norberto da Silva, 736, 14025-670 Ribeirão Preto SP Brazil, Tel.: (55 16) 3911-4130 / Fax.: (55 16) 3621-3552 - Ribeirão Preto - SP - Brazil
E-mail: editor@gmb.org.br