2020
DOI: 10.1101/2020.06.12.148890
|View full text |Cite
Preprint
|
Sign up to set email alerts
|

Nuclear deformation causes DNA damage by increasing replication stress

Abstract: Cancer metastasis, i.e., the spreading of tumor cells from the primary tumor to distant organs, is responsible for the vast majority of cancer deaths. In the process, cancer cells migrate through narrow interstitial spaces substantially smaller in cross-section than the cell. During such confined migration, cancer cells experience extensive nuclear deformation, nuclear envelope rupture, and DNA damage. The molecular mechanisms responsible for the confined migrationinduced DNA damage remain incompletely underst… Show more

Help me understand this report
View published versions

Search citation statements

Order By: Relevance

Paper Sections

Select...
2
2
1

Citation Types

2
29
0

Year Published

2020
2020
2022
2022

Publication Types

Select...
7

Relationship

1
6

Authors

Journals

citations
Cited by 19 publications
(31 citation statements)
references
References 74 publications
2
29
0
Order By: Relevance
“…Close associations between NE rupture and the loss of lamin A/C and lamin B1 as well as the mutation of nuclear lamins exist in HGPS and cancer (Earle et al, 2020;Penfield et al, 2018). Cancer cells suffer more NE rupture, DNA damage and nuclear deformation through confining space (Denais et al, 2016;Kidiyoor et al, 2020;Shah et al, 2021). The confined migration and compression force increased replication stress and promoted replication fork stalling (Shah et al, 2021).…”
Section: Ne Stressmentioning
confidence: 99%
See 1 more Smart Citation
“…Close associations between NE rupture and the loss of lamin A/C and lamin B1 as well as the mutation of nuclear lamins exist in HGPS and cancer (Earle et al, 2020;Penfield et al, 2018). Cancer cells suffer more NE rupture, DNA damage and nuclear deformation through confining space (Denais et al, 2016;Kidiyoor et al, 2020;Shah et al, 2021). The confined migration and compression force increased replication stress and promoted replication fork stalling (Shah et al, 2021).…”
Section: Ne Stressmentioning
confidence: 99%
“…Cancer cells suffer more NE rupture, DNA damage and nuclear deformation through confining space (Denais et al, 2016;Kidiyoor et al, 2020;Shah et al, 2021). The confined migration and compression force increased replication stress and promoted replication fork stalling (Shah et al, 2021). In addition, the ATR defective cells generated NE rupture, perinuclear cGAS (cyclic GMP-AMP synthase) accumulation and aberrant perinuclear chromatin status during interstitial migration or mechanical force loading (Kidiyoor et al, 2020).…”
Section: Ne Stressmentioning
confidence: 99%
“…Through treating the nucleus as an elastic-fluid system wherein the fluid surrounding the chromatin can be squeezed out of the nucleus, their model showed that outflow of mobile repair proteins due to the constricted migration was sufficient to explain the experimental data on increased damage sites [47]. Recent experimental work has shown similarly that nuclear deformation alone can cause increased DNA damage [110]. In their model, nuclear rupture was assumed to merely delay the ability of the repair factors to return to their original locations.…”
Section: Nuclear Blebbing and Rupturementioning
confidence: 99%
“…We know that efficient DNA repair mechanisms to counteract the effects of replicative stress and other sources of damage are central to avoiding both premature ageing and cancer ( Rossi et al, 2008 ; Jeggo et al, 2016 ; Sperka et al, 2012 ; Adams et al, 2015 ). Recent in vitro studies using cancer cell lines, dendritic cells, as well as primary stem cells have shown that migration through micro capillaries or extreme constrictions imparts mechanical stress on nuclei, and this can be a source of DNA damage and genome instability ( Denais et al, 2016 ; Raab et al, 2016 ; Irianto et al, 2017a ; Smith et al, 2019 ; Nader, 2020 ; Shah et al, 2017 ; Shah, 2020 ; Kirby and Lammerding, 2018 ). These studies reveal an assortment of mechanisms by which mechanical stress results in DNA damage.…”
Section: Introductionmentioning
confidence: 99%