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 -- 1921 2022-07-07 22:58:57 |
2 format -1 word(s) 1920 2022-07-08 03:10:21 |

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.
Chihanga, T.;  Vicente-Muñoz, S.;  Ruiz-Torres, S.;  Pal, B.;  Sertorio, M.;  Andreassen, P.R.;  Khoury, R.;  Mehta, P.;  Davies, S.M.;  Lane, A.N.; et al. Dysregulation of Mitochondrial Metabolism in Fanconi Anemia-Deficient Cells. Encyclopedia. Available online: https://encyclopedia.pub/entry/24921 (accessed on 03 May 2024).
Chihanga T,  Vicente-Muñoz S,  Ruiz-Torres S,  Pal B,  Sertorio M,  Andreassen PR, et al. Dysregulation of Mitochondrial Metabolism in Fanconi Anemia-Deficient Cells. Encyclopedia. Available at: https://encyclopedia.pub/entry/24921. Accessed May 03, 2024.
Chihanga, Tafadzwa, Sara Vicente-Muñoz, Sonya Ruiz-Torres, Bidisha Pal, Mathieu Sertorio, Paul R. Andreassen, Ruby Khoury, Parinda Mehta, Stella M. Davies, Andrew N. Lane, et al. "Dysregulation of Mitochondrial Metabolism in Fanconi Anemia-Deficient Cells" Encyclopedia, https://encyclopedia.pub/entry/24921 (accessed May 03, 2024).
Chihanga, T.,  Vicente-Muñoz, S.,  Ruiz-Torres, S.,  Pal, B.,  Sertorio, M.,  Andreassen, P.R.,  Khoury, R.,  Mehta, P.,  Davies, S.M.,  Lane, A.N.,  Romick-Rosendale, L.E., & Wells, S.I. (2022, July 07). Dysregulation of Mitochondrial Metabolism in Fanconi Anemia-Deficient Cells. In Encyclopedia. https://encyclopedia.pub/entry/24921
Chihanga, Tafadzwa, et al. "Dysregulation of Mitochondrial Metabolism in Fanconi Anemia-Deficient Cells." Encyclopedia. Web. 07 July, 2022.
Dysregulation of Mitochondrial Metabolism in Fanconi Anemia-Deficient Cells
Edit

Fanconi anemia (FA) pathway deficiency is associated with dysregulated cellular metabolism in addition to defects in interstrand crosslinks (ICLs) repair, and metabolic abnormalities are an important potential contributor to the observed clinical phenotypes. Metabolic dysregulation is reflected by changes in mitochondrial structure and function with reduced energy production and an increase in oxidative stress and defective mitophagy. Furthermore, mutations in FA proteins cause increased aldehyde load and subsequent aldehyde induced damage, resulting in reduced cellular capacity for aldehyde detoxification, and hyperproduction of, and sensitivity to, inflammatory cytokines. Metabolic reprogramming in individuals with FA may be linked to an array of phenotypes poorly explained by deficient DNA repair, including short stature, insulin resistance, thyroid dysfunction, abnormal body mass index (BMI) and dyslipidemia. Metabolism is a dynamic process which is essential for cell viability, from maintaining membrane potentials, provision of metabolic energy in the form of ATP via oxidation of nutrients (catabolism) for cell maintenance and repair, to cell proliferation that requires ATP to drive the formation of complex macromolecules (anabolism), and tissue specific activities such as contraction of muscle and generation of action potentials in the brain. Nutrient uptake and utilization are commonly altered in cancers and many show a strong dependence on glutamine.

Dysregulation Fanconi anemia Mitochondrial

1. Introduction

Fanconi anemia (FA) is a generally autosomal recessive orphan disease caused by germline loss of function mutations in any one of more than 20 genes associated with the FA DNA repair pathway, although rare cases display X-linked or dominant negative inheritance [1]. FA affects 1 in 160,000 individuals, with a variable life expectancy of 20 years [2][3]. The FA pathway functions classically in the repair of DNA (interstrand crosslinks) ICLs [1]. ICLs are covalent adducts between DNA strands which block transcription and replication and can lead to double strand DNA breaks [4]. Continuous removal of ICLs is required for sustained cell survival. ICLs form as a response to either exogenous or endogenous crosslinkers. Exogenous crosslinkers include chemotherapeutics such as mitomycin C, cisplatin, melphalan, psoralens, tobacco use, broiled meat and alcohol consumption [5][6][7]. Endogenous crosslinkers include natural byproducts of mitochondrial and fat metabolism, metabolism of tobacco smoke or ingested alcohol, all of which can produce reactive oxygen species (ROS) [8] and aldehydes [9][10]. Cells from individuals with FA are highly sensitive to DNA crosslinkers, resulting in increased chromosomal abnormalities and accumulation of cells arrested in the G2/M phase of the cell cycle [5][11]. Individuals with FA are unable to properly repair ICLs, and exhibit congenital defects and progressive bone marrow failure, often early in life. Moreover, patients with FA demonstrate increased susceptibility to human papillomavirus (HPV) and SV40 infection [12][13]. Bone marrow failure in FA can evolve into leukemia [9][14][15][16][17][18] and persons with FA have a markedly elevated frequency of early aggressive keratinocyte-based squamous cell carcinomas (SCCs) of the head and neck, esophagus, anogenital tract, and skin with advancing age compared to the general population [1][19][20][21][22][23][24][25]. Other non-canonical activities reported for the FA pathway include the stabilization of stalled replication forks [26], control of mitosis and cytokinesis [27][28], suppression of non-homologous end-joining (NHEJ) [29], clearance of damaged mitochondria by mitophagy [30], clearance of viruses by autophagy [30], regulation of the HPV life cycle [13][21][24][31][32][33][34][35], maintenance of cellular redox reactions [36] and metabolic regulation [37][38][39].

2. Mitochondrial Activities Play a Role in Oncogenesis

Mitochondria are cellular organelles responsible for oxygen-dependent energy metabolism and are a main source and target of ROS formation. The transport of ADP, phosphate and protons across the inner membrane normally accelerates the rate of electron transport and most of the oxygen consumed by the respiratory electron chain is reduced to water. mtDNA encodes some but not all of the respiratory enzyme subunits that are essential for oxidative phosphorylation and rRNA and tRNA needed for mitochondrial protein synthesis. Nuclear DNA encodes the remaining mitochondrial proteins [40]. Mutations in mtDNA are thought to significantly stimulate oxidative phosphorylation, support neoplastic transformation, and fulfill the sustained bioenergetic demands of cancer cells [41]. Meta-analysis of 20 different cancer types from 859 non-FA patients showed that 66% harbored mutations in mtDNA [42]. Included in this analysis were adult leukemia (9/24) 38% and head and neck cancers (337/467) 72%, which harbored mutations in mtDNA [42]. Although the role of these mutations in cancer and metastasis remains unknown, they may increase energy metabolism and ROS generation and support cell survival. Several studies have uncovered links between FA gene products and mitochondrial dysfunction [38][43][44][45][46][47][48], and oxidative stress-derived mitochondrial dysfunction in combination with decreased scavenging of endogenous aldehydes [10], increased lipid peroxidation [49] and impaired ATP production [38][44][50], have emerged as metabolic phenotypic hallmarks of FA [44]. More detailed FA metabolic studies are needed to determine required compensatory reprogramming such as increased lactic fermentation which provides metabolic energy, intermediates for proliferative anabolism, and more potent tumor-supporting microenvironments by acidification and negative regulation of immune cell function [51].
FA has recently been identified as a mitochondrial disease (MD) given these and other connections between FA proteins and impaired mitochondrial activities [52]. MDs are a group of disorders associated with mutations in nuclear and mitochondrial DNA and consequent impaired oxidative phosphorylation. MDs are characterized by a number of clinical pathologies including short stature, exercise intolerance and hypertrophic or dilated cardiomyopathy [53]. Although FA shares limited clinical similarities with MDs aside from short stature, mitochondrial dysfunction is a hallmark in both instances [52].

3. FA Proteins Localize to Mitochondria

Multiple FA proteins are detectable in mitochondria and regulate mitochondrial metabolic function through physical interactions with other mitochondrial proteins, such as the peroxidase peroxiredoxin-3 (PRDX3) [45], and ATP synthase (ATP5α), a subunit of the mitochondrial ATPase. In FANCA, C and G deficient cells, constitutive oxidative stress suppressed mitochondrial activities by reducing the transmembrane potential, oxygen consumption rate, ATP production, and ROS detoxification [45]. FANCA and FANCC deficient subtypes also suppressed PRDX3, a member of a family of antioxidant enzymes which regulate physiological levels of hydrogen peroxide (H2O2) [45]. FANCG physically interacts with PRDX3, and FANCG mutated cells harbored a distorted mitochondrial structure and reduced thioredoxin-dependent peroxidase activity [45]. Overexpression of PRDX3 restored the resistance of FANCG-deficient cells to oxidative stress, while PRDX3 downregulation increased sensitivity to mitomycin C. FANCA and C deficient subtypes also harbored decreased PRDX3 expression, indicating an as of yet unknown functional interaction between other FA proteins and PRDX3 [45]. Furthermore, a physical interaction between FANCD2 and ATP5α was reported to be essential for optimal ATP synthesis [39]. FANCD2 deficient cells harbored reduced mitochondrial ATP production due to inappropriate ATP5α localization [39]. In addition, FANCD2 localizes to mitochondria in a process mediated by the ATPase Family AAA Domain-Containing Protein 3A (ATAD3) [54], a member of the mitochondrial nucleoid complex that also includes Mitochondrial Transcription Factor A (Tfam) and Mitochondrial Tu Translation Elongation Factor (Tufm). This complex is essential for mtDNA-encoded gene transcription, translation and mitochondrial biosynthesis [54][55][56], and might therefore play a key role in the maintenance of mitochondrial activities. Indeed, genetic deletion of Fancd2 in murine hematopoietic stem and progenitor cells (HSPCs) led to a significant increase in mitochondrial number, mitochondrial protein synthesis, enzyme activity of mitochondrially encoded respiratory complexes and, consequently, OXPHOS and mtROS levels in HSPCs [54]. Increased mitochondrial number and proteins may cause an imbalance in nuclear and mitochondrially encoded factors that is further exacerbated by a significant increase in mitochondrial stress related proteins and a deregulated mitochondrial stress response in FANCD2 -deficient cells [54][57].

4. ROS May Be a Cause or Consequence of Mitochondrial Abnormalities in FA

ROS are produced by several endogenous sources that require oxygen [58]. H2O2 is generated by a wide variety of oxygen-dependent oxidation reactions, as well as by dismutation of superoxide [59]. Oxygen radicals, which include superoxide, oxidize macromolecules such as lipids and proteins, and can generate adducts between DNA strands [60]. Some of these negative effects can be countered by antioxidant defenses such as upregulation of the antioxidant detoxification components NADPH quinone oxidoreductase-1 and Redox factor-1 (Ref-1) [58][61]. Thus, oxidative stress is defined by the imbalance between oxidants and antioxidants [49][62]. The mitochondrial respiratory chain is the major producer of ROS, as a byproduct of ATP production facilitated by NADH oxidases [63]. Other endogenous enzymatic reactions not limited to the mitochondria which produce ROS are prostaglandin synthesis, phagocytosis, and the cytochrome P450 system [64]. In addition, exogenous agents such as metals, therapeutic agents, radiation and environmental toxins such as benzo(a)pyrene also produce ROS [64][65].
It has been suggested that mitochondria in FA deficient cells are shifted to a semi-resting state, where ATP production is defective and the rate of oxygen consumption is decreased due to compromised Complex I activity [44][66]. FANCA-deficient cells displayed defects in mitochondrial respiratory chain Complex I activity, resulting in diminished ATP production [66]. Impaired oxygen consumption and reduced mitochondrial membrane potential as a consequence of low ATP production have been identified as phenotypes of FANCA, FANCC and FANCD2 deficient cells [44]. Overexpression of superoxide dismutase 1 (SOD1) rescued oxygen uptake and respiration capacity in FA cells. Some mitochondrial enzymes responsible for ROS clearance were unable to respond to H2O2 in FA cells, suggesting impairment of the mitochondrial detoxifying machinery [44]. Indeed, in pathological conditions such as FA, buildup and/or failure to detoxify ROS further diminish mitochondrial activities [62][67][68]. Accumulation of ROS dissipates the transmembrane potential, which leads to lower ATP levels in FA deficient cells as compared to their genetically corrected counterparts. The low transmembrane potential and overproduction of ROS could be a result of alterations in mitochondrial morphology, including membrane thinning or rupture, abnormal shapes and mitophagy in FA cells. Treatment of the same cells with H2O2 further increased mitochondrial fragmentation. In turn, addition of the ROS scavenger N-acetyl-cysteine (NAC) lowered the production of ROS in FA-depleted cells [44]. Importantly, the sensitivity of FA deficient cells to crosslinking agents such as mitomycin C was reduced in the presence of ROS scavengers. Pretreatment of FA deficient cells with NAC significantly improved ATP production, restored oxygen consumption and stimulated increased resistance to MMC, albeit in the absence of rescue of abnormal mitochondrial morphologies [44]. These data suggest that some of the mitochondrially induced ROS-related phenotypes of FA deficient cells are reversible [44][66].

5. Increased ROS Exacerbate DNA Damage in FA

ROS at low concentrations can have beneficial roles in signal transduction [69][70] and organismal defense against pathogens [71][72]. However, ROS at high concentrations can induce oxidation and damage of DNA, protein and lipid, with subsequent pro-inflammatory cytokine production, apoptosis, autophagy and/or necrosis. Elevated ROS also induces chronic inflammation which promotes cancer initiation and may further increase cancer risk [73][74]. Chronic inflammation and infection (e.g., persistent HPV infection) can cause the production of chemokines and cytokines, which in turn promote cellular transformation [75], by-pass of tumor suppressor activity [76] and proliferation and angiogenesis [77][78]. ROS stimulates pro-inflammatory chemokine and cytokine production, which initiates a positive feedback loop resulting in further ROS accumulation and an inflammatory oncogenic environment [79]. Interestingly, ROS-dependent lipid peroxidation results in aldehyde production, especially malondialdehyde (MDA) and 4-hydroxynonenal (4HNE), both of which stimulate ICLs and hyper-mutagenicity [70][80]. Heightened endogenous aldehyde production via ROS and lipid peroxidation may therefore co-operate with the classical ICL repair defects to produce structural variants and chromosomal abnormalities that are hallmarks of FA [81][82].
Damaged mitochondria in FA cells are also more likely to rupture, inducing apoptosis at lower levels of stress than in normal cells. The FANCA, FANCC, FANCD2, FANCF, FANCL, BRCA1/FANCS and BRCA2/FANCD1 proteins aid in Parkin-mediated mitophagy, implicating this non-canonical role of FA proteins in disease pathologies [30][47]. Defective mitophagy, in turn, leads to the accumulation of damaged mitochondria and concomitantly increased intracellular oxidative stress [83]. Interestingly, in murine embryonic fibroblasts (MEFs), FANCC is essential for host immunity against herpes simplex virus type 1 mutant strain and Sindbis virus and plays a role in virophagy as well as autophagy [30]. Overall, these data suggest that genetic FA defects lead to the accumulation of ROS, as well as mitochondrial abnormalities with impaired antioxidant defenses [84], thus further damaging mitochondria to diminish cellular respiration and ATP synthesis.

References

  1. Niraj, J.; Färkkilä, A.; D’Andrea, A.D. The Fanconi Anemia Pathway in Cancer. Annu. Rev. Cancer Biol. 2019, 3, 457–478.
  2. Auerbach, A.D. Diagnosis of Fanconi Anemia by Diepoxybutane Analysis. Curr. Protoc. Hum. Genet. 2015, 85, 8.7.1–8.7.17.
  3. Bagby, G.C. Multifunctional Fanconi proteins, inflammation and the Fanconi phenotype. EBioMedicine 2016, 8, 10–11.
  4. Kottemann, M.C.; Smogorzewska, A. Fanconi anaemia and the repair of Watson and Crick DNA crosslinks. Nature 2013, 493, 356–363.
  5. Andreassen, P.R.; Ren, K. Fanconi Anemia Proteins, DNA Interstrand Crosslink Repair Pathways, and Cancer Therapy. Curr. Cancer Drug Targets 2009, 9, 101–117.
  6. Ma, B.; Stepanov, I.; Hecht, S.S. Recent Studies on DNA Adducts Resulting from Human Exposure to Tobacco Smoke. Toxics 2019, 7, 16.
  7. Rageul, J.; Kim, H. Fanconi anemia and the underlying causes of genomic instability. Environ. Mol. Mutagen. 2020, 61, 693–708.
  8. Joenje, H.; Oostra, B. Effect of oxygen tension on chromosomal aberrations in Fanconi anaemia. Qual. Life Res. 1983, 65, 99–101.
  9. Garaycoechea, J.I.; Crossan, G.; Langevin, F.; Daly, M.; Arends, M.J.; Patel, K.J. Genotoxic consequences of endogenous aldehydes on mouse haematopoietic stem cell function. Nature 2012, 489, 571–575.
  10. Langevin, F.; Crossan, G.; Rosado, I.V.; Arends, M.J.; Patel, K.J. Fancd2 counteracts the toxic effects of naturally produced aldehydes in mice. Nature 2011, 475, 53–58.
  11. Grompe, M.; D’Andrea, A. Fanconi anemia and DNA repair. Hum. Mol. Genet. 2001, 10, 2253–2259.
  12. Comar, M.; De Rocco, D.; Cappelli, E.; Zanotta, N.; Bottega, R.; Svahn, J.; Farruggia, P.; Misuraca, A.; Corsolini, F.; Dufour, C.; et al. Fanconi Anemia Patients Are More Susceptible to Infection with Tumor Virus SV40. PLoS ONE 2013, 8, e79683.
  13. Sauter, S.L.; Wells, S.I.; Zhang, X.; Hoskins, E.E.; Davies, S.M.; Myers, K.C.; Mueller, R.; Panicker, G.; Unger, E.; Sivaprasad, U.; et al. Oral Human Papillomavirus Is Common in Individuals with Fanconi Anemia. Cancer Epidemiol. Biomark. Prev. 2015, 24, 864–872.
  14. Butturini AGale, R.P.; Verlander, P.C.; Adler-Brecher, B.; Gillio, A.P.; Auerbach, A.D. Hematologic abnormalities in Fanconi anemia: An International Fanconi Anemia Registry study. Blood 1994, 84, 1650–1655.
  15. Hess, C.J.; Gale, R.P.; Verlander, P.C.; Adler-Brecher, B.; Gillio, A.P.; Auerbach, A.D. Hypermethylation of the FANCC and FANCL promoter regions in sporadic acute leukaemia. Anal. Cell. Pathol. 2008, 30, 299–306.
  16. Li, J.; Sejas, D.P.; Zhang, X.; Qiu, Y.; Nattamai, K.J.; Rani, R.; Rathbun, K.R.; Geiger, H.; Williams, D.A.; Bagby, G.C.; et al. TNF-α induces leukemic clonal evolution ex vivo in Fanconi anemia group C murine stem cells. J. Clin. Investig. 2007, 117, 3283–3295.
  17. Savage, S.A.; Walsh, M.F. Myelodysplastic Syndrome, Acute Myeloid Leukemia, and Cancer Surveillance in Fanconi Anemia. Hematol. Oncol. Clin. N. Am. 2018, 32, 657–668.
  18. Tischkowitz, M.D.; Morgan, N.; Grimwade, D.; Eddy, C.; Ball, S.; Vorechovsky, I.; Langabeer, S.; Stöger, R.; Hodgson, S.V.; Mathew, C. Deletion and reduced expression of the Fanconi anemia FANCA gene in sporadic acute myeloid leukemia. Leukemia 2004, 18, 420–425.
  19. Kutler, D.I.; Auerbach, A.D.; Satagopan, J.; Giampietro, P.F.; Batish, S.D.; Huvos, A.G.; Goberdhan, A.; Shah, J.P.; Singh, B. High Incidence of Head and Neck Squamous Cell Carcinoma in Patients with Fanconi Anemia. Arch. Otolaryngol. Head Neck Surg. 2003, 129, 106–112.
  20. Kutler, D.I.; Patel, K.R.; Auerbach, A.D.; Kennedy, J.; Lach, F.P.; Sanborn, E.; Cohen, M.A.; Kuhel, W.I.; Smogorzewska, A. Natural history and management of Fanconi anemia patients with head and neck cancer: A 10-year follow-up. Laryngoscope 2016, 126, 870–879.
  21. Kutler, D.I.; Singh, B.; Satagopan, J.; Batish, S.D.; Berwick, M.; Giampietro, P.F.; Hanenberg, H.; Auerbach, A. A 20-year perspective on the International Fanconi Anemia Registry (IFAR). Blood 2003, 101, 1249–1256.
  22. Rosenberg, P.S.; Greene, M.H.; Alter, B.P. Cancer incidence in persons with Fanconi anemia. Blood 2003, 101, 822–826.
  23. Scheckenbach, K.; Wagenmann, M.; Freund, M.; Schipper, J.; Hanenberg, H.; Schethenbach, K. Squamous Cell Carcinomas of the Head and Neck in Fanconi Anemia: Risk, Prevention, Therapy, and the Need for Guidelines. Klin. Pädiatrie 2012, 224, 132–138.
  24. Velleuer, E.; Dietrich, R. Fanconi anemia: Young patients at high risk for squamous cell carcinoma. Mol. Cell. Pediatr. 2014, 1, 9.
  25. Alter, B.P.; Giri, N.; Savage, S.; Rosenberg, P. Cancer in the National Cancer Institute inherited bone marrow failure syndrome cohort after fifteen years of follow-up. Haematologica 2017, 103, 30–39.
  26. Schlacher, K.; Wu, H.; Jasin, M. A Distinct Replication Fork Protection Pathway Connects Fanconi Anemia Tumor Suppressors to RAD51-BRCA1/2. Cancer Cell 2012, 22, 106–116.
  27. Naim, V.; Rosselli, F. The FANC pathway and mitosis: A replication legacy. Cell Cycle 2009, 8, 2907–2912.
  28. Vinciguerra, P.; Godinho, S.A.; Parmar, K.; Pellman, D.; D’Andrea, A.D. Cytokinesis failure occurs in Fanconi anemia pathway–deficient murine and human bone marrow hematopoietic cells. J. Clin. Investig. 2010, 120, 3834–3842.
  29. Adamo, A.; Collis, S.J.; Adelman, C.A.; Silva, N.; Horejsi, Z.; Ward, J.D.; Martinez-Perez, E.; Boulton, S.J.; La Volpe, A. Preventing Nonhomologous End Joining Suppresses DNA Repair Defects of Fanconi Anemia. Mol. Cell 2010, 39, 25–35.
  30. Sumpter, R., Jr.; Sirasanagandla, S.; Fernández, Á.F.; Wei, Y.; Dong, X.; Franco, L.H.; Zou, Z.; Marchal, C.; Lee, M.Y.; Clapp, D.W.; et al. Fanconi Anemia Proteins Function in Mitophagy and Immunity. Cell 2016, 165, 867–881.
  31. Myers, K.C.; Sauter, S.; Zhang, X.; Bleesing, J.J.; Davies, S.M.; Wells, S.I.; Mehta, P.A.; Kumar, A.; Marmer, D.; Marsh, R.; et al. Impaired immune function in children and adults with Fanconi anemia. Pediatr. Blood Cancer 2017, 64, e26599.
  32. D’Souza, A.M.; Mark, J.; DeMarcantonio, M.; Leino, D.; Sisson, R.; Geller, J.I. Pediatric laryngeal carcinoma in a heterozygous carrier of Fanconi anemia. Pediatr. Blood Cancer 2017, 64, e26463.
  33. Khanal, S.; Galloway, D.A. High-risk human papillomavirus oncogenes disrupt the Fanconi anemia DNA repair pathway by impairing localization and de-ubiquitination of FancD2. PLoS Pathog. 2019, 15, e1007442.
  34. Spardy, N.; Duensing, A.; Charles, D.; Haines, N.; Nakahara, T.; Lambert, P.F.; Duensing, S. The Human Papillomavirus Type 16 E7 Oncoprotein Activates the Fanconi Anemia (FA) Pathway and Causes Accelerated Chromosomal Instability in FA Cells. J. Virol. 2007, 81, 13265–13270.
  35. Kutler, D.I.; Wreesmann, V.B.; Goberdhan, A.; Ben-Porat, L.; Satagopan, J.; Ngai, I.; Huvos, A.G.; Giampietro, P.; Levran, O.; Pujara, K.; et al. Human Papillomavirus DNA and p53 Polymorphisms in Squamous Cell Carcinomas from Fanconi Anemia Patients. JNCI J. Natl. Cancer Inst. 2003, 95, 1718–1721.
  36. Ruppitsch, W.; Meißlitzer, C.; Weirich-Schwaiger, H.; Klocker, H.; Scheidereit, C.; Schweiger, M.; Hirsch-Kauffmann, M. The role of oxygen metabolism for the pathological phenotype of Fanconi anemia. Qual. Life Res. 1997, 99, 710–719.
  37. Bartlett, A.L.; Romick-Rosendale, L.; Nelson, A.; Abdullah, S.; Luebbering, N.; Bartlett, J.; Brusadelli, M.; Palumbo, J.S.; Lake, K.; Litts, B.; et al. Tryptophan metabolism is dysregulated in individuals with Fanconi anemia. Blood Adv. 2021, 5, 250–261.
  38. Cappelli, E.; Cuccarolo, P.; Stroppiana, G.; Miano, M.; Bottega, R.; Cossu, V.; Degan, P.; Ravera, S. Defects in mitochondrial energetic function compels Fanconi Anaemia cells to glycolytic metabolism. Biochim. Biophys. Acta Mol. Basis Dis. 2017, 1863, 1214–1221.
  39. Jayabal, P.; Manoj, N.; Nepal, M.; Shen, Y.; Che, R.; Turkson, J.; Fei, P. Involvement of FANCD2 in Energy Metabolism via ATP5α. Sci. Rep. 2017, 7, 4921.
  40. Wallace, D.C. Mitochondria and cancer. Nat. Rev. Cancer 2012, 12, 685–698.
  41. Park, J.S.; Sharma, L.K.; Li, H.; Xiang, R.; Holstein, D.; Wu, J.; Lechleiter, J.; Naylor, S.L.; Deng, J.J.; Lu, J.; et al. A heteroplasmic, not homoplasmic, mitochondrial DNA mutation promotes tumorigenesis via alteration in reactive oxygen species generation and apoptosis. Hum. Mol. Genet. 2009, 18, 1578–1589.
  42. Lee, H.-C. Somatic alterations in mitochondrial DNA and mitochondrial dysfunction in gastric cancer progression. World J. Gastroenterol. 2014, 20, 3950–3959.
  43. Cuccarolo, P.; Viaggi, S.; Degan, P. New insights into redox response modulation in Fanconi’s anemia cells by hydrogen peroxide and glutathione depletors. FEBS J. 2012, 279, 2479–2494.
  44. Kumari, U.; Jun, W.Y.; Bay, B.H.; Lyakhovich, A. Evidence of mitochondrial dysfunction and impaired ROS detoxifying machinery in Fanconi Anemia cells. Oncogene 2013, 33, 165–172.
  45. Mukhopadhyay, S.S.; Leung, K.S.; Hicks, M.J.; Hastings, P.J.; Youssoufian, H.; Plon, S.E. Defective mitochondrial peroxiredoxin-3 results in sensitivity to oxidative stress in Fanconi anemia. J. Cell Biol. 2006, 175, 225–235.
  46. Pallardó, F.V.; Lloret, A.; Lebel, M.; D’Ischia, M.; Cogger, V.C.; Le Couteur, D.; Gadaleta, M.N.; Castello, G.; Pagano, G. Mitochondrial dysfunction in some oxidative stress-related genetic diseases: Ataxia-Telangiectasia, Down Syndrome, Fanconi Anaemia and Werner Syndrome. Biogerontology 2010, 11, 401–419.
  47. Solanki, A.; Rajendran, A.; Mohan, S.; Raj, R.; Vundinti, B.R. Mitochondrial DNA variations and mitochondrial dysfunction in Fanconi anemia. PLoS ONE 2020, 15, e0227603.
  48. Milletti, G.; Strocchio, L.; Pagliara, D.; Girardi, K.; Carta, R.; Mastronuzzi, A.; Locatelli, F.; Nazio, F. Canonical and Noncanonical Roles of Fanconi Anemia Proteins: Implications in Cancer Predisposition. Cancers 2020, 12, 2684.
  49. Marnett, L.J. Oxyradicals and DNA damage. Carcinogenesis 2000, 21, 361–370.
  50. Lyakhovich, A. Damaged mitochondria and overproduction of ROS in Fanconi anemia cells. Rare Dis. 2013, 1, e24048.
  51. Tu, V.; Ayari, A.; O’Connor, R. Beyond the Lactate Paradox: How Lactate and Acidity Impact T Cell Therapies against Cancer. Antibodies 2021, 10, 25.
  52. Pagano, G.; Tiano, L.; Pallardó, F.V.; Lyakhovich, A.; Mukhopadhyay, S.S.; Di Bartolomeo, P.; Zatterale, A.; Trifuoggi, M. Re-definition and supporting evidence toward Fanconi Anemia as a mitochondrial disease: Prospects for new design in clinical management. Redox Biol. 2021, 40, 101860.
  53. DiMauro, S.; Hirano, M. Pathogenesis and Treatment of Mitochondrial Disorders. Adv. Exp. Med. Biol. 2009, 652, 139–170.
  54. Chatla, S.; Du, W.; Wilson, A.F.; Meetei, A.R.; Pang, Q. Fancd2-deficient hematopoietic stem and progenitor cells depend on augmented mitochondrial translation for survival and proliferation. Stem Cell Res. 2019, 40, 101550.
  55. Bogenhagen, D.F.; Rousseau, D.; Burke, S. The Layered Structure of Human Mitochondrial DNA Nucleoids. J. Biol. Chem. 2008, 283, 3665–3675.
  56. He, J.; Cooper, H.M.; Reyes, A.; Di Re, M.; Sembongi, H.; Litwin, T.R.; Gao, J.; Neuman, K.C.; Fearnley, I.M.; Spinazzola, A.; et al. Mitochondrial nucleoid interacting proteins support mitochondrial protein synthesis. Nucleic Acids Res. 2012, 40, 6109–6121.
  57. Fernandes, P.; Miotto, B.; Saint-Ruf, C.; Said, M.; Barra, V.; Nähse, V.; Ravera, S.; Cappelli, E.; Naim, V. FANCD2 modulates the mitochondrial stress response to prevent common fragile site instability. Commun. Biol. 2021, 4, 127.
  58. Ray, P.D.; Huang, B.-W.; Tsuji, Y. Reactive oxygen species (ROS) homeostasis and redox regulation in cellular signaling. Cell. Signal. 2012, 24, 981–990.
  59. Wang, Y.; Branicky, R.; Noe, A.; Hekimi, S. Superoxide dismutases: Dual roles in controlling ROS damage and regulating ROS signaling. J. Cell Biol. 2018, 217, 1915–1928.
  60. Wang, Y. Bulky DNA Lesions Induced by Reactive Oxygen Species. Chem. Res. Toxicol. 2008, 21, 276–281.
  61. Zahra, K.F.; Lefter, R.; Ali, A.; Abdellah, E.-C.; Trus, C.; Ciobica, A.; Timofte, D. The Involvement of the Oxidative Stress Status in Cancer Pathology: A Double View on the Role of the Antioxidants. Oxid. Med. Cell. Longev. 2021, 2021, 9965916.
  62. Park, S.-J.; Ciccone, S.L.M.; Beck, B.D.; Hwang, B.; Freie, B.; Clapp, D.W.; Lee, S.-H. Oxidative Stress/Damage Induces Multimerization and Interaction of Fanconi Anemia Proteins. J. Biol. Chem. 2004, 279, 30053–30059.
  63. Beswick, R.A.; Dorrance, A.M.; Leite, R.; Webb, R.C. NADH/NADPH Oxidase and Enhanced Superoxide Production in the Mineralocorticoid Hypertensive Rat. Hypertension 2001, 38, 1107–1111.
  64. Snezhkina, A.V.; Kudryavtseva, A.V.; Kardymon, O.L.; Savvateeva, M.V.; Melnikova, N.V.; Krasnov, G.S.; Dmitriev, A.A. ROS Generation and Antioxidant Defense Systems in Normal and Malignant Cells. Oxid. Med. Cell. Longev. 2019, 2019, 6175804.
  65. Ji, K.; Xing, C.; Jiang, F.; Wang, X.; Guo, H.; Nan, J.; Qian, L.; Yang, P.; Lin, J.; Li, M.; et al. Benzopyrene induces oxidative stress and endothelial progenitor cell dysfunction via the activation of the NF-κB pathway. Int. J. Mol. Med. 2013, 31, 922–930.
  66. Ravera, S.; Vaccaro, D.; Cuccarolo, P.; Columbaro, M.; Capanni, C.; Bartolucci, M.; Panfoli, I.; Morelli, A.; Dufour, C.; Cappelli, E.; et al. Mitochondrial respiratory chain Complex I defects in Fanconi anemia complementation group A. Biochimie 2013, 95, 1828–1837.
  67. Du, W.; Adam, Z.; Rani, R.; Zhang, X.; Pang, Q. Oxidative Stress in Fanconi Anemia Hematopoiesis and Disease Progression. Antioxid. Redox Signal. 2008, 10, 1909–1921.
  68. Degan, P.; Bonassi, S.; De Caterina, M.; Korkina, L.G.; Pinto, L.; Scopacasa, F.; Zatterale, A.; Calzone, R.; Pagano, G. In vivo accumulation of 8-hydroxy-2’-deoxyguanosine in DNA correlates with release of reactive oxygen species in Fanconi’s anaemia families. Carcinogenesis 1995, 16, 735–742.
  69. D’Autréaux, B.; Toledano, M.B. ROS as signalling molecules: Mechanisms that generate specificity in ROS homeostasis. Nat. Rev. Mol. Cell Biol. 2007, 8, 813–824.
  70. Shen, X.; Li, L. Mutagenic repair of DNA interstrand crosslinks. Environ. Mol. Mutagen. 2010, 51, 493–499.
  71. Leto, T.L.; Morand, S.; Hurt, D.; Ueyama, T. Targeting and Regulation of Reactive Oxygen Species Generation by Nox Family NADPH Oxidases. Antioxid. Redox Signal. 2009, 11, 2607–2619.
  72. Yang, H.-C.; Cheng, M.-L.; Ho, H.-Y.; Chiu, D.T.-Y. The microbicidal and cytoregulatory roles of NADPH oxidases. Microbes Infect. 2011, 13, 109–120.
  73. Balkwill, F.; Mantovani, A. Inflammation and cancer: Back to Virchow? Lancet 2001, 357, 539–545.
  74. Korniluk, A.; Koper, O.; Kemona, H.; Dymicka-Piekarska, V. From inflammation to cancer. Ir. J. Med. Sci. 2017, 186, 57–62.
  75. Komori, A.; Yatsunami, J.; Suganuma, M.; Okabe, S.; Abe, S.; Sakai, A.; Sasaki, K.; Fujiki, H. Tumor necrosis factor acts as a tumor promoter in BALB/3T3 cell transformation. Cancer Res. 1993, 53, 1982–1985.
  76. Hudson, J.D.; Shoaibi, M.A.; Maestro, R.; Carnero, A.; Hannon, G.J.; Beach, D.H. A Proinflammatory Cytokine Inhibits P53 Tumor Suppressor Activity. J. Exp. Med. 1999, 190, 1375–1382.
  77. Keane, M.; Strieter, R. The Role of CXC Chemokines in the Regulation of Angiogenesis. Chem. Immunol. Allergy 1999, 72, 86–101.
  78. Raman, D.; Baugher, P.J.; Thu, Y.M.; Richmond, A. Role of chemokines in tumor growth. Cancer Lett. 2007, 256, 137–165.
  79. Jaiswal, M.; LaRusso, N.F.; Burgart, L.J.; Gores, G.J. Inflammatory cytokines induce DNA damage and inhibit DNA repair in cholangiocarcinoma cells by a nitric oxide-dependent mechanism. Cancer Res. 2000, 60, 184.
  80. Yau, T.M. Mutagenicity and cytotoxicity of malonaldehyde in mammalian cells. Mech. Ageing Dev. 1979, 11, 137–144.
  81. Yang, Z.; Wu, X.S.; Wei, Y.; Polyanskaya, S.A.; Iyer, S.V.; Jung, M.; Lach, F.P.; Adelman, E.R.; Klingbeil, O.; Milazzo, J.P.; et al. Transcriptional Silencing of ALDH2 Confers a Dependency on Fanconi Anemia Proteins in Acute Myeloid Leukemia. Cancer Discov. 2021, 11, 2300–2315.
  82. Auerbach, A.D. Fanconi anemia and its diagnosis. Mutat. Res. Fundam. Mol. Mech. Mutagen. 2009, 668, 4–10.
  83. Drake, L.E.; Springer, M.Z.; Poole, L.P.; Kim, C.; Macleod, K.F. Expanding perspectives on the significance of mitophagy in cancer. Semin. Cancer Biol. 2017, 47, 110–124.
  84. Hadjur, S.; Ung, K.; Wadsworth, L.; Dimmick, J.; Rajcan-Separovic, E.; Scott, R.W.; Buchwald, M.; Jirik, F.R. Defective hematopoiesis and hepatic steatosis in mice with combined deficiencies of the genes encoding Fancc and Cu/Zn superoxide dismutase. Blood 2001, 98, 1003–1011.
More
Information
Subjects: Biology
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: 538
Revisions: 2 times (View History)
Update Date: 08 Jul 2022
1000/1000