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 -- 3195 2022-06-08 23:10:58 |
2 format correct -32 word(s) 3163 2022-06-09 06:01:56 |

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.
Nualart, F.; , .; Ramírez, E. Two Faces of Vitamin C: AA vs. DHA. Encyclopedia. Available online: https://encyclopedia.pub/entry/23855 (accessed on 30 June 2024).
Nualart F,  , Ramírez E. Two Faces of Vitamin C: AA vs. DHA. Encyclopedia. Available at: https://encyclopedia.pub/entry/23855. Accessed June 30, 2024.
Nualart, Francisco, , Eder Ramírez. "Two Faces of Vitamin C: AA vs. DHA" Encyclopedia, https://encyclopedia.pub/entry/23855 (accessed June 30, 2024).
Nualart, F., , ., & Ramírez, E. (2022, June 08). Two Faces of Vitamin C: AA vs. DHA. In Encyclopedia. https://encyclopedia.pub/entry/23855
Nualart, Francisco, et al. "Two Faces of Vitamin C: AA vs. DHA." Encyclopedia. Web. 08 June, 2022.
Two Faces of Vitamin C: AA vs. DHA
Edit

Historically, vitamin C has been associated with many regulatory processes that involve specific signaling pathways. Among the most studied signaling pathways are those involved in the regulation of aging, differentiation, neurotransmission, proliferation, and cell death processes in cancer. This wide variety of regulatory effects is due to the fact that vitamin C has a dual mechanism of action. The reduced form of vitamin C (ascorbic acid, AA) is an essential micronutrient of small size; it is soluble in water and has two dissociable protons with pKa values of 4.2 and 11.8. At physiological pH, its reduced form predominates as the monovalent ascorbate anion (AA); when it loses the second proton, it is oxidized to dehydroascorbic acid (DHA).

vitamin C signal transduction cancer cell death necroptosis

1. Introduction

The reduced form of vitamin C (ascorbic acid, AA) is an essential micronutrient of small size; it is soluble in water and has two dissociable protons with pKa values of 4.2 and 11.8. At physiological pH, its reduced form predominates as the monovalent ascorbate anion (AA); when it loses the second proton, it is oxidized to dehydroascorbic acid (DHA) [1][2][3]. Most mammals can synthesize vitamin C from D-glucose in the liver, except guinea pigs, bats, and higher primates, including humans, due to the absence of the enzyme L-gulonolactone oxidase, which catalyzes the last step of the biosynthesis of vitamin C [4]. Therefore, to meet the body’s requirements, vitamin C must be incorporated into the diet [1]. The best-known function of vitamin C is as an antioxidant agent that can act as a cofactor of enzymatic reactions involved in the synthesis of catecholamines, carnitine, cholesterol, amino acids, and some hormonal peptides, as well as in the maintenance of brain function and the protection of central nervous system (CNS) structures [1][3][5][6][7].
AA uptake in different cells is performed by the sodium-ascorbate cotransporters SVCT1 and SVCT2, which stereospecifically transport the reduced form of vitamin C, L-ascorbate [8][9][10][11]. Vitamin C can also be transported in its oxidized form, DHA, through the facilitative glucose transporters GLUT1, GLUT2, GLUT3, GLUT4, and GLUT8 [12][13][14][15][16]. However, for a long time, it has been postulated that the contribution of DHA to the accumulation of vitamin C in tissues is relatively low [3][17][18][19].

2. Molecular Pathways Regulated by Vitamin C

One of the first targets for vitamin C was discovered via its relationship to the NF‒κB pathway. In this pathway, vitamin C has an inhibitory function; in studies carried out in endothelial cells, millimolar doses of AA inhibited NF-κB and IL-8 activation in response to tumor necrosis factor (TNF) [20]. In this entry, the authors also evaluated the toxicity generated by high doses of vitamin C supplementation and did not detect cell damage or lipid peroxidation [20][21]. Furthermore, they were able to determine that the inhibition of the NF-κB pathway was not due to the antioxidant activity of vitamin C, but rather to the direct inhibition of IκB kinase α/β (IKKα/β) [20][21][22][23]. In line with this notion, IKKα/β is a kinase responsible for the phosphorylation of IκBα protein that maintains NF-κB-p65 in the cytoplasm [24]. IκBα phosphorylation is a signal for proteasomal degradation of this protein, allowing NF-κB-p65 nuclear translocation (Figure 1A), triggering the activation of specific genes [24][25][26]. In line with these findings, it was postulated that AA is a regulator of IKKα/β activity; however, subsequent studies determined that AA has no action on IKKα/β [22][27]. Interestingly, it was shown that DHA was a regulator of IKKα/β mediated by directly binding to this kinase, inhibiting it, and finally controlling the activity of NF-κB [27]. This function of DHA was determined through immunoprecipitation experiments using p-IκBα-GST where derivatives of vitamin C, AA, DHA, oxalic acid, and threonic acid were used. Only treatment with DHA inhibited IκBα-phosphorylation, and this inhibition was mediated by DHA directly blocking the activity of IKKα/β and p38, likely competing for the binding of ATP to the active site of IKKβ [22][27]. Given this evidence, it was concluded that vitamin C has a dual action against reactive oxygen species (ROS). Intracellularly, AA would fulfill its antioxidant function by neutralizing ROS, generating DHA. Thus, the intracellular accumulation of DHA would block the activation of NF-κB, involving vitamin C in signaling processes that control inflammatory responses and cell death among others (Figure 1A).
Figure 1. Integrative vision of the principal molecular pathways regulated by vitamin C. (A) Scheme of vitamin C recycling in normal cells or cells with oxidative stress. Under normal conditions, AA concentrations remain homeostatically stable due to the efficient recycling of DHA by specialized cells. However, under conditions of oxidative stress or inefficient recycling, an accumulation of intracellular DHA can occur. DHA would target the inhibition of IKK α/β, metabolic enzymes such as GAPDH, as well as the production of genotoxic stress, resulting in the induction of cell death. (B) Intracellular effects of vitamin C on signaling pathways associated with cell death. The physiological levels of AA would have a protective function intracellularly, favoring the inhibition of apoptosis by inducing overexpression of antiapoptotic genes, as well as caspases. At the same time, AA could maintain RIPK1 in its inhibited state, which favors cell survival. (C) Under pathophysiological or acute oxidative stress conditions, ROS overload induces a massive oxidation of AA to DHA, intracellularly. The accumulation of DHA results in the inhibition of IKK α/β, and p38, which can trigger the activation of RIPK1 and cell death due to necroptosis, in cells that accumulate high concentrations of vitamin C, such as neurons. AA: ascorbic acid; DHA: dehydroascorbic acid; SVCT2s: sodium-dependent vitamin C transporter; GLUTs: glucose transporters; RIPK1: receptor-interacting serine/threonine-protein kinase 1; MK2: p38MAPK-activated protein kinase 2; TRADD: TNFR1-associated death domain protein; TRAF2: TNF receptor associated factor 2.
Another kinase-dependent pathway that is regulated by vitamin C is that of the mitogen-activated protein kinases (MAPK), which involves three other MAPK-dependent pathways, extracellular signal-regulated kinases (ERK), c-Jun N-terminal kinases (JNK), and p38 kinase, which are involved in proliferation, differentiation, and apoptosis, respectively [10][28][29]. The first studies examining the relationship between vitamin C and MAPK concluded that vitamin C was involved in in vitro cell death processes, but the mechanism of action was unknown. Thus, the possible regulation of MAPK-ERK mediated by vitamin C was analyzed. For this, leukemia cell lines were treated with AA (0-500 µM) for 1 to 3 h in order to analyze ERK activation by in vitro phosphorylation assays. In cells treated with concentrations as low as 100 μM AA, phosphorylation of ERK and therefore activation was induced [10][30]. Thus, it was proposed that the regulation of ERK mediated by vitamin C would be associated with eventual apoptotic processes that are observed in certain tumor lines when treated with vitamin C because ERK activation is associated with proliferative processes and cell death. However, to date, it has been shown that the use of pharmacological doses of AA induces tumor death from conventional necrosis due to the extracellular generation of H2O2 [31][32], as discussed in detail later. At the same time, it has also been shown that AA can antagonize apoptosis in cancer cells induced by classical mechanisms, such as treatment with doxorubidicin, TRAIL, or FAS [33][34][35]. In line with this notion, treatment with physiological doses of AA in neuronal cultures induces overexpression of antiapoptotic genes, such as Bcl-2, and decreases the expression of proapoptotic genes, such as Bax and caspase 8 [19]. Thus, the current evidence suggests that physiological doses of AA could inhibit apoptosis rather than activate this death pathway. Furthermore, AA-mediated ERK activation could be associated with neuronal arborization mechanisms, which would be an indicator of neuronal “good health” [10].

References

  1. Padayatty, S.J.; Levine, M. Vitamin C: The known and the unknown and Goldilocks. Oral Dis. 2016, 22, 463–493.
  2. May, J.M. Vitamin C transport and its role in the central nervous system. Subcell. Biochem. 2012, 56, 85–103.
  3. Nualart, F.; Mack, L.; Garcia, A.; Cisternas, P.; Bongarzone, E.R.; Heitzer, M.; Jara, N.; Martinez, F.; Ferrada, L.; Espinoza, F.; et al. Vitamin C Transporters, Recycling and the Bystander Effect in the Nervous System: SVCT2 versus Gluts. J. Stem Cell Res. Ther. 2014, 4, 209.
  4. Nishikimi, M.; Fukuyama, R.; Minoshima, S.; Shimizu, N.; Yagi, K. Cloning and chromosomal mapping of the human nonfunctional gene for L-gulono-gamma-lactone oxidase, the enzyme for L-ascorbic acid biosynthesis missing in man. J. Biol. Chem. 1994, 269, 13685–13688.
  5. Harrison, F.E.; May, J.M. Vitamin C function in the brain: Vital role of the ascorbate transporter SVCT2. Free Radic. Biol. Med. 2009, 46, 719–730.
  6. Rice, M.E. Ascorbate regulation and its neuroprotective role in the brain. Trends Neurosci. 2000, 23, 209–216.
  7. Ngo, B.; Van Riper, J.M.; Cantley, L.C.; Yun, J. Targeting cancer vulnerabilities with high-dose vitamin C. Nat. Rev. Cancer 2019, 19, 271–282.
  8. Tsukaguchi, H.; Tokui, T.; Mackenzie, B.; Berger, U.V.; Chen, X.Z.; Wang, Y.; Brubaker, R.F.; Hediger, M.A. A family of mammalian Na+-dependent L-ascorbic acid transporters. Nature 1999, 399, 70–75.
  9. Silva-Alvarez, C.; Salazar, K.; Cisternas, P.; Martinez, F.; Liour, S.; Jara, N.; Bertinat, R.; Nualart, F. Apical Polarization of SVCT2 in Apical Radial Glial Cells and Progenitors During Brain Development. Mol. Neurobiol. 2017, 54, 5449–5467.
  10. Salazar, K.; Martinez, M.; Ulloa, V.; Bertinat, R.; Martinez, F.; Jara, N.; Espinoza, F.; Bongarzone, E.R.; Nualart, F. SVCT2 Overexpression in Neuroblastoma Cells Induces Cellular Branching that is Associated with ERK Signaling. Mol. Neurobiol. 2016, 53, 6668–6679.
  11. Wang, Y.; Mackenzie, B.; Tsukaguchi, H.; Weremowicz, S.; Morton, C.C.; Hediger, M.A. Human vitamin C (L-ascorbic acid) transporter SVCT1. Biochem. Biophys. Res. Commun. 2000, 267, 488–494.
  12. Ferrada, L.; Salazar, K.; Nualart, F. Metabolic control by dehydroascorbic acid: Questions and controversies in cancer cells. J. Cell Physiol. 2019, 234, 19331–19338.
  13. Nualart, F.J.; Rivas, C.I.; Montecinos, V.P.; Godoy, A.S.; Guaiquil, V.H.; Golde, D.W.; Vera, J.C. Recycling of vitamin C by a bystander effect. J. Biol. Chem. 2003, 278, 10128–10133.
  14. Rumsey, S.C.; Daruwala, R.; Al-Hasani, H.; Zarnowski, M.J.; Simpson, I.A.; Levine, M. Dehydroascorbic acid transport by GLUT4 in Xenopus oocytes and isolated rat adipocytes. J. Biol. Chem. 2000, 275, 28246–28253.
  15. Rumsey, S.C.; Kwon, O.; Xu, G.W.; Burant, C.F.; Simpson, I.; Levine, M. Glucose transporter isoforms GLUT1 and GLUT3 transport dehydroascorbic acid. J. Biol. Chem. 1997, 272, 18982–18989.
  16. Corpe, C.P.; Eck, P.; Wang, J.; Al-Hasani, H.; Levine, M. Intestinal dehydroascorbic acid (DHA) transport mediated by the facilitative sugar transporters, GLUT2 and GLUT8. J. Biol. Chem. 2013, 288, 9092–9101.
  17. Cisternas, P.; Silva-Alvarez, C.; Martinez, F.; Fernandez, E.; Ferrada, L.; Oyarce, K.; Salazar, K.; Bolanos, J.P.; Nualart, F. The oxidized form of vitamin C, dehydroascorbic acid, regulates neuronal energy metabolism. J. Neurochem. 2014, 129, 663–671.
  18. Garcia-Krauss, A.; Ferrada, L.; Astuya, A.; Salazar, K.; Cisternas, P.; Martinez, F.; Ramirez, E.; Nualart, F. Dehydroascorbic Acid Promotes Cell Death in Neurons Under Oxidative Stress: A Protective Role for Astrocytes. Mol. Neurobiol. 2016, 53, 5847–5863.
  19. Ferrada, L.; Barahona, M.J.; Salazar, K.; Vandenabeele, P.; Nualart, F. Vitamin C controls neuronal necroptosis under oxidative stress. Redox Biol. 2020, 29, 101408.
  20. Bowie, A.G.; O’Neill, L.A. Vitamin C inhibits NF-kappa B activation by TNF via the activation of p38 mitogen-activated protein kinase. J. Immunol. 2000, 165, 7180–7188.
  21. Bowie, A.; O’Neill, L.A. Vitamin C inhibits NF kappa B activation in endothelial cells. Biochem. Soc. Trans. 1997, 25, 131S.
  22. Carcamo, J.M.; Pedraza, A.; Borquez-Ojeda, O.; Golde, D.W. Vitamin C suppresses TNF alpha-induced NF kappa B activation by inhibiting I kappa B alpha phosphorylation. Biochemistry 2002, 41, 12995–13002.
  23. Abhilash, P.A.; Harikrishnan, R.; Indira, M. Ascorbic acid suppresses endotoxemia and NF-kappaB signaling cascade in alcoholic liver fibrosis in guinea pigs: A mechanistic approach. Toxicol. Appl. Pharmacol. 2014, 274, 215–224.
  24. Yi, F.; Frazzette, N.; Cruz, A.C.; Klebanoff, C.A.; Siegel, R.M. Beyond Cell Death: New Functions for TNF Family Cytokines in Autoimmunity and Tumor Immunotherapy. Trends Mol. Med. 2018, 24, 642–653.
  25. Malek, R.; Borowicz, K.K.; Jargiello, M.; Czuczwar, S.J. Role of nuclear factor kappaB in the central nervous system. Pharmacol. Rep. 2007, 59, 25–33.
  26. Engelmann, C.; Weih, F.; Haenold, R. Role of nuclear factor kappa B in central nervous system regeneration. Neural Regen. Res. 2014, 9, 707–711.
  27. Carcamo, J.M.; Pedraza, A.; Borquez-Ojeda, O.; Zhang, B.; Sanchez, R.; Golde, D.W. Vitamin C is a kinase inhibitor: Dehydroascorbic acid inhibits IkappaBalpha kinase beta. Mol. Cell. Biol. 2004, 24, 6645–6652.
  28. Lee, S.A.; Son, Y.O.; Kook, S.H.; Choi, K.C.; Lee, J.C. Ascorbic acid increases the activity and synthesis of tyrosinase in B16F10 cells through activation of p38 mitogen-activated protein kinase. Arch. Dermatol. Res. 2011, 303, 669–678.
  29. Lei, F.X.; Jin, L.; Liu, X.Y.; Lai, F.; Yan, X.G.; Farrelly, M.; Guo, S.T.; Zhao, X.H.; Zhang, X.D. RIP1 protects melanoma cells from apoptosis induced by BRAF/MEK inhibitors. Cell Death Dis. 2018, 9, 679.
  30. Park, S.; Park, C.H.; Hahm, E.R.; Kim, K.; Kimler, B.F.; Lee, S.J.; Park, H.K.; Lee, S.H.; Kim, W.S.; Jung, C.W.; et al. Activation of Raf1 and the ERK pathway in response to l-ascorbic acid in acute myeloid leukemia cells. Cell Signal. 2005, 17, 111–119.
  31. Chen, Q.; Espey, M.G.; Sun, A.Y.; Pooput, C.; Kirk, K.L.; Krishna, M.C.; Khosh, D.B.; Drisko, J.; Levine, M. Pharmacologic doses of ascorbate act as a prooxidant and decrease growth of aggressive tumor xenografts in mice. Proc. Natl. Acad. Sci. USA 2008, 105, 11105–11109.
  32. Chen, Q.; Espey, M.G.; Krishna, M.C.; Mitchell, J.B.; Corpe, C.P.; Buettner, G.R.; Shacter, E.; Levine, M. Pharmacologic ascorbic acid concentrations selectively kill cancer cells: Action as a pro-drug to deliver hydrogen peroxide to tissues. Proc. Natl. Acad. Sci. USA 2005, 102, 13604–13609.
  33. Heaney, M.L.; Gardner, J.R.; Karasavvas, N.; Golde, D.W.; Scheinberg, D.A.; Smith, E.A.; O’Connor, O.A. Vitamin C antagonizes the cytotoxic effects of antineoplastic drugs. Cancer Res. 2008, 68, 8031–8038.
  34. Perez-Cruz, I.; Carcamo, J.M.; Golde, D.W. Vitamin C inhibits FAS-induced apoptosis in monocytes and U937 cells. Blood 2003, 102, 336–343.
  35. Perez-Cruz, I.; Carcamo, J.M.; Golde, D.W. Caspase-8 dependent TRAIL-induced apoptosis in cancer cell lines is inhibited by vitamin C and catalase. Apoptosis 2007, 12, 225–234.
  36. Zinkel, S.; Gross, A.; Yang, E. BCL2 family in DNA damage and cell cycle control. Cell Death Differ. 2006, 13, 1351–1359.
  37. Nguyen, M.D.; Mushynski, W.E.; Julien, J.P. Cycling at the interface between neurodevelopment and neurodegeneration. Cell Death Differ. 2002, 9, 1294–1306.
  38. El Banna, N.; Hatem, E.; Heneman-Masurel, A.; Leger, T.; Baille, D.; Vernis, L.; Garcia, C.; Martineau, S.; Dupuy, C.; Vagner, S.; et al. Redox modifications of cysteine-containing proteins, cell cycle arrest and translation inhibition: Involvement in vitamin C-induced breast cancer cell death. Redox. Biol. 2019, 26, 101290.
  39. Belin, S.; Kaya, F.; Duisit, G.; Giacometti, S.; Ciccolini, J.; Fontes, M. Antiproliferative effect of ascorbic acid is associated with the inhibition of genes necessary to cell cycle progression. PLoS ONE 2009, 4, e4409.
  40. Martinotti, S.; Ranzato, E.; Parodi, M.; Vitale, M.; Burlando, B. Combination of ascorbate/epigallocatechin-3-gallate/gemcitabine synergistically induces cell cycle deregulation and apoptosis in mesothelioma cells. Toxicol. Appl. Pharmacol. 2014, 274, 35–41.
  41. Felipe, K.B.; Benites, J.; Glorieux, C.; Sid, B.; Valenzuela, M.; Kviecinski, M.R.; Pedrosa, R.C.; Valderrama, J.A.; Leveque, P.; Gallez, B.; et al. Antiproliferative effects of phenylaminonaphthoquinones are increased by ascorbate and associated with the appearance of a senescent phenotype in human bladder cancer cells. Biochem. Biophys. Res. Commun. 2013, 433, 573–578.
  42. McConnell, M.J.; Herst, P.M. Ascorbate combination therapy: New tool in the anticancer toolbox? Sci. Transl. Med. 2014, 6, 222fs6.
  43. Alexander, B.; Fishman, A.I.; Eshghi, M.; Choudhury, M.; Konno, S. Induction of cell death in renal cell carcinoma with combination of D-fraction and vitamin C. Integr. Cancer Ther. 2013, 12, 442–448.
  44. Herst, P.M.; Broadley, K.W.; Harper, J.L.; McConnell, M.J. Pharmacological concentrations of ascorbate radiosensitize glioblastoma multiforme primary cells by increasing oxidative DNA damage and inhibiting G2/M arrest. Free. Radic. Biol. Med. 2012, 52, 1486–1493.
  45. Moteki, H.; Shimamura, Y.; Kimura, M.; Ogihara, M. Signal transduction pathway for L-ascorbic acid- and L-ascorbic acid 2-glucoside-induced DNA synthesis and cell proliferation in primary cultures of adult rat hepatocytes. Eur. J. Pharmacol. 2012, 683, 276–284.
  46. Handy, D.E.; Castro, R.; Loscalzo, J. Epigenetic modifications: Basic mechanisms and role in cardiovascular disease. Circulation 2011, 123, 2145–2156.
  47. Lu, C.; Thompson, C.B. Metabolic regulation of epigenetics. Cell Metab. 2012, 16, 9–17.
  48. Huang, S.; Xu, Y.M.; Lau, A.T.Y. Epigenetic effects of the 13 vitamins. Curr. Pharmacol. Rep. 2018, 4, 453–467.
  49. Kuiper, C.; Vissers, M.C. Ascorbate as a co-factor for fe- and 2-oxoglutarate dependent dioxygenases: Physiological activity in tumor growth and progression. Front. Oncol. 2014, 4, 359.
  50. Melamed, P.; Yosefzon, Y.; David, C.; Tsukerman, A.; Pnueli, L. Tet Enzymes, Variants, and Differential Effects on Function. Front. Cell Dev. Biol. 2018, 6, 22.
  51. Carr, A.C.; Frei, B. Toward a new recommended dietary allowance for vitamin C based on antioxidant and health effects in humans. Am. J. Clin. Nutr. 1999, 69, 1086–1107.
  52. Farrow, S.C.; Facchini, P.J. Functional diversity of 2-oxoglutarate/Fe(II)-dependent dioxygenases in plant metabolism. Front. Plant. Sci. 2014, 5, 524.
  53. Myllyla, R.; Majamaa, K.; Gunzler, V.; Hanauske-Abel, H.M.; Kivirikko, K.I. Ascorbate is consumed stoichiometrically in the uncoupled reactions catalyzed by prolyl 4-hydroxylase and lysyl hydroxylase. J. Biol. Chem. 1984, 259, 5403–5405.
  54. de Jong, L.; Albracht, S.P.; Kemp, A. Prolyl 4-hydroxylase activity in relation to the oxidation state of enzyme-bound iron. The role of ascorbate in peptidyl proline hydroxylation. Biochim. Biophys. Acta 1982, 704, 326–332.
  55. Cimmino, L.; Neel, B.G.; Aifantis, I. Vitamin C in Stem Cell Reprogramming and Cancer. Trends Cell Biol. 2018, 28, 698–708.
  56. Lane, D.J.; Chikhani, S.; Richardson, V.; Richardson, D.R. Transferrin iron uptake is stimulated by ascorbate via an intracellular reductive mechanism. Biochim. Biophys. Acta 2013, 1833, 1527–1541.
  57. Tsukada, Y.; Fang, J.; Erdjument-Bromage, H.; Warren, M.E.; Borchers, C.H.; Tempst, P.; Zhang, Y. Histone demethylation by a family of JmjC domain-containing proteins. Nature 2006, 439, 811–816.
  58. Yin, R.; Mao, S.Q.; Zhao, B.; Chong, Z.; Yang, Y.; Zhao, C.; Zhang, D.; Huang, H.; Gao, J.; Li, Z.; et al. Ascorbic acid enhances Tet-mediated 5-methylcytosine oxidation and promotes DNA demethylation in mammals. J. Am. Chem. Soc. 2013, 135, 10396–10403.
  59. Cho, Y.S.; Challa, S.; Moquin, D.; Genga, R.; Ray, T.D.; Guildford, M.; Chan, F.K. Phosphorylation-driven assembly of the RIP1-RIP3 complex regulates programmed necrosis and virus-induced inflammation. Cell 2009, 137, 1112–1123.
  60. Zhang, D.W.; Shao, J.; Lin, J.; Zhang, N.; Lu, B.J.; Lin, S.C.; Dong, M.Q.; Han, J. RIP3, an energy metabolism regulator that switches TNF-induced cell death from apoptosis to necrosis. Science 2009, 325, 332–336.
  61. He, S.; Wang, L.; Miao, L.; Wang, T.; Du, F.; Zhao, L.; Wang, X. Receptor interacting protein kinase-3 determines cellular necrotic response to TNF-alpha. Cell 2009, 137, 1100–1111.
  62. Wang, L.; Du, F.; Wang, X. TNF-alpha induces two distinct caspase-8 activation pathways. Cell 2008, 133, 693–703.
  63. Pasparakis, M.; Vandenabeele, P. Necroptosis and its role in inflammation. Nature 2015, 517, 311–320.
  64. Vanden Berghe, T.; Kaiser, W.J.; Bertrand, M.J.; Vandenabeele, P. Molecular crosstalk between apoptosis, necroptosis, and survival signaling. Mol. Cell. Oncol. 2015, 2, e975093.
  65. Murphy, J.M.; Czabotar, P.E.; Hildebrand, J.M.; Lucet, I.S.; Zhang, J.G.; Alvarez-Diaz, S.; Lewis, R.; Lalaoui, N.; Metcalf, D.; Webb, A.I.; et al. The pseudokinase MLKL mediates necroptosis via a molecular switch mechanism. Immunity 2013, 39, 443–453.
  66. Sun, L.; Wang, H.; Wang, Z.; He, S.; Chen, S.; Liao, D.; Wang, L.; Yan, J.; Liu, W.; Lei, X.; et al. Mixed lineage kinase domain-like protein mediates necrosis signaling downstream of RIP3 kinase. Cell 2012, 148, 213–227.
  67. Hildebrand, J.M.; Tanzer, M.C.; Lucet, I.S.; Young, S.N.; Spall, S.K.; Sharma, P.; Pierotti, C.; Garnier, J.M.; Dobson, R.C.; Webb, A.I.; et al. Activation of the pseudokinase MLKL unleashes the four-helix bundle domain to induce membrane localization and necroptotic cell death. Proc. Natl. Acad. Sci. USA 2014, 111, 15072–15077.
  68. Zhang, Y.; Chen, X.; Gueydan, C.; Han, J. Plasma membrane changes during programmed cell deaths. Cell Res. 2018, 28, 9–21.
  69. Meng, H.; Liu, Z.; Li, X.; Wang, H.; Jin, T.; Wu, G.; Shan, B.; Christofferson, D.E.; Qi, C.; Yu, Q.; et al. Death-domain dimerization-mediated activation of RIPK1 controls necroptosis and RIPK1-dependent apoptosis. Proc. Natl. Acad. Sci. USA 2018, 115, E2001–E2009.
  70. Amin, P.; Florez, M.; Najafov, A.; Pan, H.; Geng, J.; Ofengeim, D.; Dziedzic, S.A.; Wang, H.; Barrett, V.J.; Ito, Y.; et al. Regulation of a distinct activated RIPK1 intermediate bridging complex I and complex II in TNFalpha-mediated apoptosis. Proc. Natl. Acad. Sci. USA 2018.
  71. Remijsen, Q.; Goossens, V.; Grootjans, S.; Van den Haute, C.; Vanlangenakker, N.; Dondelinger, Y.; Roelandt, R.; Bruggeman, I.; Goncalves, A.; Bertrand, M.J.; et al. Depletion of RIPK3 or MLKL blocks TNF-driven necroptosis and switches towards a delayed RIPK1 kinase-dependent apoptosis. Cell Death Dis. 2014, 5, e1004.
  72. Degterev, A.; Yuan, J. Expansion and evolution of cell death programmes. Nat. Rev. Mol. Cell Biol. 2008, 9, 378–390.
  73. Degterev, A.; Hitomi, J.; Germscheid, M.; Ch’en, I.L.; Korkina, O.; Teng, X.; Abbott, D.; Cuny, G.D.; Yuan, C.; Wagner, G.; et al. Identification of RIP1 kinase as a specific cellular target of necrostatins. Nat. Chem. Biol. 2008, 4, 313–321.
  74. de Almagro, M.C.; Goncharov, T.; Izrael-Tomasevic, A.; Duttler, S.; Kist, M.; Varfolomeev, E.; Wu, X.; Lee, W.P.; Murray, J.; Webster, J.D.; et al. Coordinated ubiquitination and phosphorylation of RIP1 regulates necroptotic cell death. Cell Death Differ. 2017, 24, 26–37.
  75. Bertrand, M.J.; Milutinovic, S.; Dickson, K.M.; Ho, W.C.; Boudreault, A.; Durkin, J.; Gillard, J.W.; Jaquith, J.B.; Morris, S.J.; Barker, P.A. cIAP1 and cIAP2 facilitate cancer cell survival by functioning as E3 ligases that promote RIP1 ubiquitination. Mol. Cell 2008, 30, 689–700.
  76. Dondelinger, Y.; Jouan-Lanhouet, S.; Divert, T.; Theatre, E.; Bertin, J.; Gough, P.J.; Giansanti, P.; Heck, A.J.; Dejardin, E.; Vandenabeele, P.; et al. NF-kappaB-Independent Role of IKKalpha/IKKbeta in Preventing RIPK1 Kinase-Dependent Apoptotic and Necroptotic Cell Death during TNF Signaling. Mol. Cell 2015, 60, 63–76.
  77. Dondelinger, Y.; Delanghe, T.; Rojas-Rivera, D.; Priem, D.; Delvaeye, T.; Bruggeman, I.; Van Herreweghe, F.; Vandenabeele, P.; Bertrand, M.J.M. MK2 phosphorylation of RIPK1 regulates TNF-mediated cell death. Nat. Cell Biol. 2017, 19, 1237–1247.
  78. Jaco, I.; Annibaldi, A.; Lalaoui, N.; Wilson, R.; Tenev, T.; Laurien, L.; Kim, C.; Jamal, K.; Wicky John, S.; Liccardi, G.; et al. MK2 Phosphorylates RIPK1 to Prevent TNF-Induced Cell Death. Mol. Cell 2017, 66, 698–710.e695.
  79. Shan, B.; Pan, H.; Najafov, A.; Yuan, J. Necroptosis in development and diseases. Genes Dev. 2018, 32, 327–340.
  80. Christofferson, D.E.; Li, Y.; Yuan, J. Control of life-or-death decisions by RIP1 kinase. Annu. Rev. Physiol. 2014, 76, 129–150.
  81. Dhar-Mascareno, M.; Carcamo, J.M.; Golde, D.W. Hypoxia-reoxygenation-induced mitochondrial damage and apoptosis in human endothelial cells are inhibited by vitamin C. Free. Radic. Biol. Med. 2005, 38, 1311–1322.
  82. Guaiquil, V.H.; Golde, D.W.; Beckles, D.L.; Mascareno, E.J.; Siddiqui, M.A. Vitamin C inhibits hypoxia-induced damage and apoptotic signaling pathways in cardiomyocytes and ischemic hearts. Free. Radic. Biol. Med. 2004, 37, 1419–1429.
  83. Han, J.M.; Chang, B.J.; Li, T.Z.; Choe, N.H.; Quan, F.S.; Jang, B.J.; Cho, I.H.; Hong, H.N.; Lee, J.H. Protective effects of ascorbic acid against lead-induced apoptotic neurodegeneration in the developing rat hippocampus in vivo. Brain Res. 2007, 1185, 68–74.
  84. Tian, W.; Wang, Y.; Xu, Y.; Guo, X.; Wang, B.; Sun, L.; Liu, L.; Cui, F.; Zhuang, Q.; Bao, X.; et al. The hypoxia-inducible factor renders cancer cells more sensitive to vitamin C-induced toxicity. J. Biol. Chem. 2014, 289, 3339–3351.
  85. Evans, M.K.; Tovmasyan, A.; Batinic-Haberle, I.; Devi, G.R. Mn porphyrin in combination with ascorbate acts as a pro-oxidant and mediates caspase-independent cancer cell death. Free Radic. Biol. Med. 2014, 68, 302–314.
  86. Yun, J.; Mullarky, E.; Lu, C.; Bosch, K.N.; Kavalier, A.; Rivera, K.; Roper, J.; Chio, I.I.C.; Giannopoulou, E.G.; Rago, C.; et al. Vitamin C selectively kills KRAS and BRAF mutant colorectal cancer cells by targeting GAPDH. Science 2015, 350, 1391–1396.
  87. Sarosiek, K.A.; Fraser, C.; Muthalagu, N.; Bhola, P.D.; Chang, W.; McBrayer, S.K.; Cantlon, A.; Fisch, S.; Golomb-Mello, G.; Ryan, J.A.; et al. Developmental Regulation of Mitochondrial Apoptosis by c-Myc Governs Age- and Tissue-Specific Sensitivity to Cancer Therapeutics. Cancer Cell 2017, 31, 142–156.
  88. Kole, A.J.; Annis, R.P.; Deshmukh, M. Mature neurons: Equipped for survival. Cell Death Dis. 2013, 4, e689.
More
Information
Subjects: Cell 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: 566
Revisions: 2 times (View History)
Update Date: 14 Jun 2022
1000/1000
Video Production Service