Next Article in Journal
Identification of Bottle Gourd (Lagenaria siceraria) OVATE Family Genes and Functional Characterization of LsOVATE1
Next Article in Special Issue
Tyrosine Kinase Inhibitors Target B Lymphocytes
Previous Article in Journal
Expression of a Salt-Tolerant Pseudolysin in Yeast for Efficient Protein Hydrolysis under High-Salt Conditions
Previous Article in Special Issue
Weil’s Disease—Immunopathogenesis, Multiple Organ Failure, and Potential Role of Gut Microbiota
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

DNA Repair and Immune Response: Editorial

by
Valentyn Oksenych
Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway
Biomolecules 2023, 13(1), 84; https://doi.org/10.3390/biom13010084
Submission received: 27 December 2022 / Accepted: 29 December 2022 / Published: 30 December 2022
(This article belongs to the Collection DNA Repair and Immune Response)
Developing B and T lymphocytes requires programmed DNA double-strand breaks followed by the activation of the DNA damage response (DDR) pathway and DNA repair. It is necessary for the DNA recombination process during the V(D)J recombination in maturating B and T lymphocytes to ensure the final assembly of the B cell receptor and T cell receptor genes [1,2,3,4,5,6,7]. The primary DNA repair process controlling V(D)J recombination is non-homologous DNA end-joining (NHEJ). Two additional processes specific to B cell maturation include DNA lesions and DNA repair. One is class-switch recombination (CSR), which is required to change the immunoglobulin class that also involves NHEJ and base excision repair (BER). The somatic hypermutation (SHM) process helps to increase the specificity of immunoglobulins (antibodies) and depends on different DNA repair pathways (BER and mismatch repair, MMR) [1,2].
There are several known NHEJ factors, but the extensive genetic interaction between the NHEJ-NHEJ, NHEJ-DDR, and DDR-DDR genes suggests that additional NHEJ and DDR proteins will be discovered, and currently, their functions are not known due to the complex compensatory mechanisms of the other factors. One central NHEJ gene with multiple known genetic interactions encodes an X-ray repair cross-complementing 4-like factor (XLF). It interacts genetically with several NHEJ factors, i.e., DNA-dependent protein kinase, catalytic subunit (DNA-PKcs) [8,9,10], a paralogue of XRCC4 and XLF (PAXX) [2,10,11,12,13,14,15,16,17,18,19,20], and a Modulator of retrovirus infection (MRI) [12,21,22]. Moreover, XLF has overlapping or complementary functions (functional redundancy or interacts genetically) with several DDR factors, i.e., Ataxia telangiectasia mutated (ATM) [23], histone H2AX [23], a mediator of DNA damage checkpoint 1 (MDC1) [24], p53-binding protein 1 (53BP1) [25,26], Really interesting new gene (RING) finger protein 8 (RNF8) and RNF168 [27], and Shieldin [28].
In the current Topical Collection, Raquel Gago-Fuentes et al. published a research article focusing on the genetic interaction between the NHEJ factors XLF, PAXX, and DNA-PKcs during the development of the central nervous system in mice. In particular, neural stem and progenitor cell populations were isolated from mice lacking one or two NHEJ factors. Using self-renewal capacity and differentiation and proliferation assays, it was concluded that XLF, PAXX, and DNA-PKcs are required for the early stages of neurodevelopment in mammals [29].
Richard Frock et al. published a Review article on the DNA repair process [30]. In addition to classical NHEJ, this review also focuses on alternative end-joining (A-EJ, see also [31]), and the DNA repair pathway choice in the G0/G1 phases of the cell cycle, especially in developing a B lymphocyte research model in the context of the recombination-activating gene (RAG)-mediated DNA breaks [30].
Valentyn Oksenych et al. published a research article on the roles of acetyltransferases General control nondepressible 5 (GCN5, or lysine acetyltransferase 2A, KAT2A) and p300/CBP-associated factor (PCAF, or KAT2B) in B cell development in mice [32]. Using a genetically modified mouse model combining germline knockout, conditional knockout, and knock-in mutations, as well as an overall loss-of-function strategy, the authors found that both GCN5 and PCAF are necessary for the B lymphocyte maturation in vivo. The count of mature B cells in mice lacking GCN5 was significantly reduced compared to the wild-type (WT) controls. The combined inactivation of GCN5 and PCAF resulted in even more dramatic effects observed in the spleen, blood, and bone marrow with even lower numbers of B cells when compared to single knockouts. Moreover, GCN5 is required for the immunoglobulin (Ig) class switch recombination process ex vivo, and the switching from IgM to IgG3 was significantly reduced in B cells lacking GCN5 protein [32]. Further studies are required to elucidate the mechanisms underlying the functions of GCN5 and PCAF in B cell development.
Sucheng Zhu et al. published a research article on the development of new methods to detect DNA modification [33]. The article focuses on three methods based on iodine-induced specific DNA cleavage. The new methods are dedicated to saving time and labor for the detection of DNA phosphorothioate modifications [33].
Finally, Pavlo Petakh et al. published a review article on Weil’s disease [34]. Leptospirosis is a zoonotic disease causing 60,000 deaths annually. The review describes immunopathogenesis, the influence of cytokines and genetic susceptibility on the course of the disease, and the role of gut microbiota in the clinical course. Additionally, it suggests that modulating the gut microbiota by probiotics or fecal microbiota transplantation is a possible area of further scientific research [34].
Thus, the Topical Collection highlighted the recent discoveries in DNA repair, lymphocyte maturation, development of novel methods, and immune response during disease. Additional DNA repair factors with roles in the development of the immune and nervous systems, as well as protection from genomic instability and cancer, will be discovered in the future, for example, by taking into consideration a complex genetic interaction between the DNA repair genes and the pathways in mammalian cells.

Conflicts of Interest

The author declares no conflict of interest.

References

  1. Alt, F.W.; Zhang, Y.; Meng, F.L.; Guo, C.; Schwer, B. Mechanisms of programmed DNA lesions and genomic instability in the immune system. Cell 2013, 152, 417–429. [Google Scholar] [CrossRef] [Green Version]
  2. Castaneda-Zegarra, S.; Fernandez-Berrocal, M.; Tkachov, M.; Yao, R.; Upfold, N.L.E.; Oksenych, V. Genetic interaction between the non-homologous end-joining factors during B and T lymphocyte development: In vivo mouse models. Scand. J. Immunol. 2020, 92, e12936. [Google Scholar] [CrossRef]
  3. Kumar, V.; Alt, F.W.; Oksenych, V. Functional overlaps between XLF and the ATM-dependent DNA double strand break response. DNA Repair 2014, 16, 11–22. [Google Scholar] [CrossRef] [Green Version]
  4. Wang, X.S.; Lee, B.J.; Zha, S. The recent advances in non-homologous end-joining through the lens of lymphocyte development. DNA Repair 2020, 94, 102874. [Google Scholar] [CrossRef]
  5. Libri, A.; Marton, T.; Deriano, L. The (Lack of) DNA Double-Strand Break Repair Pathway Choice During V(D)J Recombination. Front. Genet. 2021, 12, 823943. [Google Scholar] [CrossRef]
  6. Oksenych, V.; Kainov, D.E. DNA Damage Response. Biomolecules 2021, 11, 123. [Google Scholar] [CrossRef]
  7. Ragunathan, K.; Upfold, N.L.E.; Oksenych, V. Interaction between Fibroblasts and Immune Cells Following DNA Damage Induced by Ionizing Radiation. Int. J. Mol. Sci. 2020, 21, 8635. [Google Scholar] [CrossRef]
  8. Oksenych, V.; Kumar, V.; Liu, X.; Guo, C.; Schwer, B.; Zha, S.; Alt, F.W. Functional redundancy between the XLF and DNA-PKcs DNA repair factors in V(D)J recombination and nonhomologous DNA end joining. Proc. Natl. Acad. Sci. USA 2013, 110, 2234–2239. [Google Scholar] [CrossRef] [Green Version]
  9. Xing, M.; Bjoras, M.; Daniel, J.A.; Alt, F.W.; Oksenych, V. Synthetic lethality between murine DNA repair factors XLF and DNA-PKcs is rescued by inactivation of Ku70. DNA Repair 2017, 57, 133–138. [Google Scholar] [CrossRef]
  10. Xing, M.; Oksenych, V. Genetic interaction between DNA repair factors PAXX, XLF, XRCC4 and DNA-PKcs in human cells. FEBS Open Bio 2019, 9, 1315–1326. [Google Scholar] [CrossRef]
  11. Castaneda-Zegarra, S.; Xing, M.; Gago-Fuentes, R.; Saeterstad, S.; Oksenych, V. Synthetic lethality between DNA repair factors Xlf and Paxx is rescued by inactivation of Trp53. DNA Repair 2019, 73, 164–169. [Google Scholar] [CrossRef]
  12. Castaneda-Zegarra, S.; Zhang, Q.; Alirezaylavasani, A.; Fernandez-Berrocal, M.; Yao, R.; Oksenych, V. Leaky severe combined immunodeficiency in mice lacking non-homologous end joining factors XLF and MRI. Aging 2020, 12, 23578–23597. [Google Scholar] [CrossRef]
  13. Dewan, A.; Xing, M.; Lundbæk, M.B.; Gago-Fuentes, R.; Beck, C.; Aas, P.A.; Liabakk, N.B.; Sæterstad, S.; Chau, K.T.; Kavli, B.M.; et al. Robust DNA repair in PAXX-deficient mammalian cells. FEBS Open Bio 2018, 8, 442–448. [Google Scholar] [CrossRef]
  14. Gago-Fuentes, R.; Xing, M.; Sæterstad, S.; Sarno, A.; Dewan, A.; Beck, C.; Bradamante, S.; Bjørås, M.; Oksenych, V. Normal development of mice lacking PAXX, the paralogue of XRCC4 and XLF. FEBS Open Bio 2018, 8, 426–434. [Google Scholar] [CrossRef]
  15. Balmus, G.; Barros, A.C.; Wijnhoven, P.W.; Lescale, C.; Hasse, H.L.; Boroviak, K.; Le Sage, C.; Doe, B.; Speak, A.O.; Galli, A.; et al. Synthetic lethality between PAXX and XLF in mammalian development. Genes Dev. 2016, 30, 2152–2157. [Google Scholar] [CrossRef] [Green Version]
  16. Lescale, C.; Hasse, H.L.; Blackford, A.N.; Balmus, G.; Bianchi, J.J.; Yu, W.; Bacoccina, L.; Jarade, A.; Clouin, C.; Sivapalan, R.; et al. Specific Roles of XRCC4 Paralogs PAXX and XLF during V(D)J Recombination. Cell Rep. 2016, 16, 2967–2979. [Google Scholar] [CrossRef] [Green Version]
  17. Liu, X.; Shao, Z.; Jiang, W.; Lee, B.J.; Zha, S. PAXX promotes KU accumulation at DNA breaks and is essential for end-joining in XLF-deficient mice. Nat. Commun. 2017, 8, 13816. [Google Scholar] [CrossRef] [Green Version]
  18. Abramowski, V.; Etienne, O.; Elsaid, R.; Yang, J.; Berland, A.; Kermasson, L.; Roch, B.; Musilli, S.; Moussu, J.P.; Lipson-Ruffert, K.; et al. PAXX and Xlf interplay revealed by impaired CNS development and immunodeficiency of double KO mice. Cell Death Differ. 2018, 25, 444–452. [Google Scholar] [CrossRef] [Green Version]
  19. Hung, P.J.; Chen, B.R.; George, R.; Liberman, C.; Morales, A.J.; Colon-Ortiz, P.; Tyler, J.K.; Sleckman, B.P.; Bredemeyer, A.L. Deficiency of XLF and PAXX prevents DNA double-strand break repair by non-homologous end joining in lymphocytes. Cell Cycle 2017, 16, 286–295. [Google Scholar] [CrossRef] [Green Version]
  20. Kumar, V.; Alt, F.W.; Frock, R.L. PAXX and XLF DNA repair factors are functionally redundant in joining DNA breaks in a G1-arrested progenitor B-cell line. Proc. Natl. Acad. Sci. USA 2016, 113, 10619–10624. [Google Scholar] [CrossRef]
  21. Hung, P.J.; Johnson, B.; Chen, B.R.; Byrum, A.K.; Bredemeyer, A.L.; Yewdell, W.T.; Johnson, T.E.; Lee, B.J.; Deivasigamani, S.; Hindi, I.; et al. MRI Is a DNA Damage Response Adaptor during Classical Non-homologous End Joining. Mol. Cell 2018, 71, 332–342.e8. [Google Scholar] [CrossRef]
  22. Castañeda-Zegarra, S.; Huse, C.; Røsand, Ø.; Sarno, A.; Xing, M.; Gago-Fuentes, R.; Zhang, Q.; Alirezaylavasani, A.; Werner, J.; Ji, P.; et al. Generation of a Mouse Model Lacking the Non-Homologous End-Joining Factor Mri/Cyren. Biomolecules 2019, 9, 798. [Google Scholar] [CrossRef] [Green Version]
  23. Zha, S.; Guo, C.; Boboila, C.; Oksenych, V.; Cheng, H.L.; Zhang, Y.; Wesemann, D.R.; Yuen, G.; Patel, H.; Goff, P.H.; et al. ATM damage response and XLF repair factor are functionally redundant in joining DNA breaks. Nature 2011, 469, 250–254. [Google Scholar] [CrossRef] [Green Version]
  24. Beck, C.; Castaneda-Zegarra, S.; Huse, C.; Xing, M.; Oksenych, V. Mediator of DNA Damage Checkpoint Protein 1 Facilitates V(D)J Recombination in Cells Lacking DNA Repair Factor XLF. Biomolecules 2019, 10, 60. [Google Scholar] [CrossRef] [Green Version]
  25. Oksenych, V.; Alt, F.W.; Kumar, V.; Schwer, B.; Wesemann, D.R.; Hansen, E.; Patel, H.; Su, A.; Guo, C. Functional redundancy between repair factor XLF and damage response mediator 53BP1 in V(D)J recombination and DNA repair. Proc. Natl. Acad. Sci. USA 2012, 109, 2455–2460. [Google Scholar] [CrossRef] [Green Version]
  26. Liu, X.; Jiang, W.; Dubois, R.L.; Yamamoto, K.; Wolner, Z.; Zha, S. Overlapping functions between XLF repair protein and 53BP1 DNA damage response factor in end joining and lymphocyte development. Proc. Natl. Acad. Sci. USA 2012, 109, 3903–3908. [Google Scholar] [CrossRef] [Green Version]
  27. Chen, B.R.; Wang, Y.; Shen, Z.J.; Bennett, A.; Hindi, I.; Tyler, J.K.; Sleckman, B.P. The RNF8 and RNF168 Ubiquitin Ligases Regulate Pro- and Anti-Resection Activities at Broken DNA Ends During Non-Homologous End Joining. DNA Repair 2021, 108, 103217. [Google Scholar] [CrossRef]
  28. Vincendeau, E.; Wei, W.; Zhang, X.; Planchais, C.; Yu, W.; Lenden-Hasse, H.; Cokelaer, T.; Pipoli da Fonseca, J.; Mouquet, H.; Adams, D.J.; et al. SHLD1 is dispensable for 53BP1-dependent V(D)J recombination but critical for productive class switch recombination. Nat. Commun. 2022, 13, 3707. [Google Scholar] [CrossRef]
  29. Gago-Fuentes, R.; Oksenych, V. Non-Homologous End Joining Factors XLF, PAXX and DNA-PKcs Maintain the Neural Stem and Progenitor Cell Population. Biomolecules 2020, 11, 20. [Google Scholar] [CrossRef]
  30. Frock, R.L.; Sadeghi, C.; Meng, J.; Wang, J.L. DNA End Joining: G0-ing to the Core. Biomolecules 2021, 11, 1487. [Google Scholar] [CrossRef]
  31. Boboila, C.; Oksenych, V.; Gostissa, M.; Wang, J.H.; Zha, S.; Zhang, Y.; Chai, H.; Lee, C.S.; Jankovic, M.; Saez, L.M.; et al. Robust chromosomal DNA repair via alternative end-joining in the absence of X-ray repair cross-complementing protein 1 (XRCC1). Proc. Natl. Acad. Sci. USA 2012, 109, 2473–2478. [Google Scholar] [CrossRef] [Green Version]
  32. Oksenych, V.; Su, D.; Daniel, J.A. Acetyltransferases GCN5 and PCAF Are Required for B Lymphocyte Maturation in Mice. Biomolecules 2021, 12, 61. [Google Scholar] [CrossRef]
  33. Zhu, S.; Zheng, T.; Kong, L.; Li, J.; Cao, B.; DeMott, M.S.; Sun, Y.; Chen, Y.; Deng, Z.; Dedon, P.C.; et al. Development of Methods Derived from Iodine-Induced Specific Cleavage for Identification and Quantitation of DNA Phosphorothioate Modifications. Biomolecules 2020, 10, 1491. [Google Scholar] [CrossRef]
  34. Petakh, P.; Isevych, V.; Kamyshnyi, A.; Oksenych, V. Weil’s Disease-Immunopathogenesis, Multiple Organ Failure, and Potential Role of Gut Microbiota. Biomolecules 2022, 12, 1831. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Oksenych, V. DNA Repair and Immune Response: Editorial. Biomolecules 2023, 13, 84. https://doi.org/10.3390/biom13010084

AMA Style

Oksenych V. DNA Repair and Immune Response: Editorial. Biomolecules. 2023; 13(1):84. https://doi.org/10.3390/biom13010084

Chicago/Turabian Style

Oksenych, Valentyn. 2023. "DNA Repair and Immune Response: Editorial" Biomolecules 13, no. 1: 84. https://doi.org/10.3390/biom13010084

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop