axon regeneration
Recently Published Documents


TOTAL DOCUMENTS

1025
(FIVE YEARS 273)

H-INDEX

86
(FIVE YEARS 11)

BMC Genomics ◽  
2022 ◽  
Vol 23 (1) ◽  
Author(s):  
Sergei Reverdatto ◽  
Aparna Prasad ◽  
Jamie L. Belrose ◽  
Xiang Zhang ◽  
Morgan A. Sammons ◽  
...  

Abstract Background Because some of its CNS neurons (e.g., retinal ganglion cells after optic nerve crush (ONC)) regenerate axons throughout life, whereas others (e.g., hindbrain neurons after spinal cord injury (SCI)) lose this capacity as tadpoles metamorphose into frogs, the South African claw-toed frog, Xenopus laevis, offers unique opportunities for exploring differences between regenerative and non-regenerative responses to CNS injury within the same organism. An earlier, three-way RNA-seq study (frog ONC eye, tadpole SCI hindbrain, frog SCI hindbrain) identified genes that regulate chromatin accessibility among those that were differentially expressed in regenerative vs non-regenerative CNS [11]. The current study used whole genome bisulfite sequencing (WGBS) of DNA collected from these same animals at the peak period of axon regeneration to study the extent to which DNA methylation could potentially underlie differences in chromatin accessibility between regenerative and non-regenerative CNS. Results Consistent with the hypothesis that DNA of regenerative CNS is more accessible than that of non-regenerative CNS, DNA from both the regenerative tadpole hindbrain and frog eye was less methylated than that of the non-regenerative frog hindbrain. Also, consistent with observations of CNS injury in mammals, DNA methylation in non-regenerative frog hindbrain decreased after SCI. However, contrary to expectations that the level of DNA methylation would decrease even further with axotomy in regenerative CNS, DNA methylation in these regions instead increased with injury. Injury-induced differences in CpG methylation in regenerative CNS became especially enriched in gene promoter regions, whereas non-CpG methylation differences were more evenly distributed across promoter regions, intergenic, and intragenic regions. In non-regenerative CNS, tissue-related (i.e., regenerative vs. non-regenerative CNS) and injury-induced decreases in promoter region CpG methylation were significantly correlated with increased RNA expression, but the injury-induced, increased CpG methylation seen in regenerative CNS across promoter regions was not, suggesting it was associated with increased rather than decreased chromatin accessibility. This hypothesis received support from observations that in regenerative CNS, many genes exhibiting increased, injury-induced, promoter-associated CpG-methylation also exhibited increased RNA expression and association with histone markers for active promoters and enhancers. DNA immunoprecipitation for 5hmC in optic nerve regeneration found that the promoter-associated increases seen in CpG methylation were distinct from those exhibiting changes in 5hmC. Conclusions Although seemingly paradoxical, the increased injury-associated DNA methylation seen in regenerative CNS has many parallels in stem cells and cancer. Thus, these axotomy-induced changes in DNA methylation in regenerative CNS provide evidence for a novel epigenetic state favoring successful over unsuccessful CNS axon regeneration. The datasets described in this study should help lay the foundations for future studies of the molecular and cellular mechanisms involved. The insights gained should, in turn, help point the way to novel therapeutic approaches for treating CNS injury in mammals.


2022 ◽  
Vol 17 (6) ◽  
pp. 1318
Author(s):  
Jin-Zhu Bai ◽  
Yi-Xin Wang ◽  
Zhen Lyu ◽  
Guang-Hao Zhang ◽  
Xiao-Lin Huo

2022 ◽  
Vol 17 (5) ◽  
pp. 989
Author(s):  
SanjoyK Bhattacharya ◽  
Sean Meehan
Keyword(s):  

2022 ◽  
Vol 17 (7) ◽  
pp. 1518
Author(s):  
Xiao-Song Gu ◽  
Song-Lin Zhou ◽  
Ting-Ting Guo ◽  
Ying Zhao ◽  
Wei-Xiao Huang ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document