Minireview Exploring the Biological Cycle of Vitamin B3 and Its Influence on Oxidative Stress: Further Molecular and Clinical Aspects
Abstract
:1. The Biological Cycle of Niacin
1.1. Poly (ADP-Ribose) Polymerases (PARPs)
1.2. Sirtuins
1.3. Cluster of Differentiation: 38 and 157
2. Involvement of NAD and NAD(P) in Apoptosis and Gene Expression
3. Mechanism of NAD in the DNA Damage Response
4. Does Oxidative Stress Influence Fertilization Rate?
5. Is There a Relationship between Oxidative Stress and Gut Dysbacteriosis?
6. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Aguilera-Méndez, A.; Fernández-Lainez, C.; Ibarra-González, I.; Fernandez-Mejia, C. The chemistry and biochemistry of niacin (B3). In B Vitamins and Folate: Chemistry, Analysis, Function and Effects; RSC Publishing: Cambridge, UK, 2012; pp. 108–126. [Google Scholar]
- Ying, W. NAD+/NADH and NADP+/NADPH in cellular functions and cell death: Regulation and biological consequences. Antioxid. Redox Signal. 2007, 10, 179–206. [Google Scholar] [CrossRef] [Green Version]
- Karthikeyan, K.; Thappa, D.M. Pellagra and skin. Int. J. Derm. 2002, 41, 476–481. [Google Scholar] [CrossRef]
- Hegyi, J.; Schwartz, R.A.; Hegyi, V. Pellagra: Dermatitis, dementia, and diarrhea. Int. J. Derm. 2004, 43, 1–5. [Google Scholar] [CrossRef] [PubMed]
- Altschul, R.; Hoffer, A.; Stephen, J.D. Influence of nicotinic acid on serum cholesterol in man. Arch. Biochem. Biophys. 1955, 54, 558–559. [Google Scholar] [CrossRef]
- Carlson, L.A. Studies on the effect of nicotinic acid on catecholamine stimulated lipolysis in adipose tissue in vitro. Acta Med. Scand. 1963, 173, 719–722. [Google Scholar] [CrossRef]
- Carlson, L.A.; Orö, L. The effect of nicotinic acid on the plasma free fatty acids demonstration of a metabolic type of sympathicolysis. Acta Med. Scand. 1962, 172, 641–645. [Google Scholar] [CrossRef]
- Carlson, L.A.; Hanngren, Å. Initial distribution in mice of 3H-labeled nicotinic acid studied with autoradiography. Life Sci. 1964, 3, 867–871. [Google Scholar] [CrossRef]
- Butcher, R.W.; Baird, C.E.; Sutherland, E.W. Effects of lipolytic and antilipolytic substances on adenosine 3′,5′-monophosphate levels in isolated fat cells. J. Biol. Chem. 1968, 243, 1705–1712. [Google Scholar]
- Aktories, K.; Schultz, G.; Jakobs, K.H. Regulation of adenylate cyclase activity in hamster adipocytes. Naunyn. Schmiedebergs. Arch. Pharm. 1980, 312, 167–173. [Google Scholar] [CrossRef]
- Aktories, K.; Schultz, G.; Jakobs, K.H. Inactivation of the guanine nucleotide regulatory site mediating inhibition of the adenylate cyclase in hamster adipocytes. Naunyn. Schmiedebergs. Arch. Pharm. 1982, 321, 247–252. [Google Scholar] [CrossRef]
- Lorenzen, A.; Stannek, C.; Lang, H.; Andrianov, V.; Kalvins, I.; Schwabe, U. Characterization of a G protein-coupled receptor for nicotinic acid. Mol. Pharm. 2001, 59, 349–357. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kamanna, V.S.; Kashyap, M.L. Nicotinic acid (niacin) receptor agonists: Will they be useful therapeutic agents? Am. J. Cardiol. 2007, 100, S53–S61. [Google Scholar] [CrossRef] [PubMed]
- Offermanns, S. The nicotinic acid receptor GPR109A (HM74A or PUMA-G) as a new therapeutic target. Trends Pharm. Sci. 2006, 27, 384–390. [Google Scholar] [CrossRef] [PubMed]
- Blad, C.C.; Ahmed, K.; IJzerman, A.P.; Offermanns, S. 7 - biological and pharmacological roles of HCA receptors. In Pharmacology of G Protein Coupled Receptors; Richard, R.N., Ed.; Academic Press: Cambridge, MA, USA, 2011; pp. 219–250. [Google Scholar]
- Ahmed, K. Biological roles and therapeutic potential of hydroxy-carboxylic Acid receptors. Front. Endocrinol. 2011, 2, 51. [Google Scholar] [CrossRef] [Green Version]
- Suzuki, K.; Kaneko-Kawano, T. Biological roles and therapeutic potential of G protein-coupled receptors for free fatty acids and metabolic intermediates. J. Phys. Fit. Sports Med. 2016, 5, 213–227. [Google Scholar] [CrossRef]
- Selkrig, J.; Wong, P.; Zhang, X.; Pettersson, S. Metabolic tinkering by the gut microbiome: Implications for brain development and function. Gut Microbes 2014, 5, 369–380. [Google Scholar] [CrossRef]
- Graff, E.C.; Fang, H.; Wanders, D.; Judd, R.L. Anti-inflammatory effects of the hydroxycarboxylic acid receptor 2. Metabolism 2016, 65, 102–113. [Google Scholar] [CrossRef]
- Tolhurst, G.; Heffron, H.; Lam, Y.S.; Parker, H.E.; Habib, A.M.; Diakogiannaki, E.; Cameron, J.; Grosse, J.; Reimann, F.; Gribble, F.M. Short-chain fatty acids stimulate glucagon-like peptide-1 secretion via the G-protein-coupled receptor FFAR2. Diabetes 2012, 61, 364–371. [Google Scholar] [CrossRef] [Green Version]
- Samuel, B.S.; Shaito, A.; Motoike, T.; Rey, F.E.; Backhed, F.; Manchester, J.K.; Hammer, R.E.; Williams, S.C.; Crowley, J.; Yanagisawa, M.; et al. Effects of the gut microbiota on host adiposity are modulated by the short-chain fatty-acid binding G protein-coupled receptor, Gpr41. Proc. Natl. Acad. Sci. USA 2008, 105, 16767–16772. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kimura, I.; Ozawa, K.; Inoue, D.; Imamura, T.; Kimura, K.; Maeda, T.; Terasawa, K.; Kashihara, D.; Hirano, K.; Tani, T.; et al. The gut microbiota suppresses insulin-mediated fat accumulation via the short-chain fatty acid receptor GPR43. Nat. Commun. 2013, 4, 1829. [Google Scholar] [CrossRef] [Green Version]
- Bellahcene, M.; O’Dowd, J.F.; Wargent, E.T.; Zaibi, M.S.; Hislop, D.C.; Ngala, R.A.; Smith, D.M.; Cawthorne, M.A.; Stocker, C.J.; Arch, J.R.S. Male mice that lack the G-protein-coupled receptor GPR41 have low energy expenditure and increased body fat content. Br. J. Nutr. 2013, 109, 1755–1764. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Giri, B.; Belanger, K.; Seamon, M.; Bradley, E.; Purohit, S.; Chong, R.; Morgan, J.C.; Baban, B.; Wakade, C. Niacin ameliorates neuro-inflammation in Parkinson’s disease via GPR109A. Int. J. Mol. Sci. 2019, 20, 4559. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ilie, O.-D.; Ciobica, A.; McKenna, J.; Doroftei, B.; Mavroudis, I. Minireview on the relations between gut microflora and Parkinson’s disease: Further biochemical (oxidative stress), inflammatory, and neurological particularities. Oxid. Med. Cell. Longev. 2020, 2020, 4518023. [Google Scholar] [CrossRef] [PubMed]
- Gasperi, V.; Sibilano, M.; Savini, I.; Catani, M.V. Niacin in the central nervous system: An update of biological aspects and clinical applications. Int. J. Mol. Sci. 2019, 20, 974. [Google Scholar] [CrossRef] [Green Version]
- Bieganowski, P.; Brenner, C. Discoveries of nicotinamide riboside as a nutrient and conserved nrk genes establish a preiss-handler independent route to NAD+ in fungi and humans. Cell 2004, 117, 495–502. [Google Scholar] [CrossRef] [Green Version]
- Houtkooper, R.H.; Cantó, C.; Wanders, R.J.; Auwerx, J. The secret life of NAD+: An old metabolite controlling new metabolic signaling pathways. Endocr. Rev. 2010, 31, 194–223. [Google Scholar] [CrossRef] [Green Version]
- Brazill, J.M.; Li, C.; Zhu, Y.; Zhai, R.G. NMNAT: It’s an NAD(+) synthase… It’s a chaperone… It’s a neuroprotector. Curr. Opin. Genet. Dev. 2017, 44, 156–162. [Google Scholar] [CrossRef]
- Cantó, C.; Menzies, K.J.; Auwerx, J. NAD(+) metabolism and the control of energy homeostasis: A balancing act between mitochondria and the nucleus. Cell Metab. 2015, 22, 31–53. [Google Scholar] [CrossRef] [Green Version]
- Botting, N.P. Chemistry and neurochemistry of the kynurenine pathway of tryptophan metabolism. Chem. Soc. Rev. 1995, 24, 401–412. [Google Scholar] [CrossRef]
- Yang, Y.; Sauve, A.A. NAD(+) metabolism: Bioenergetics, signaling and manipulation for therapy. Biochim. Biophys. Acta 2016, 1864, 1787–1800. [Google Scholar] [CrossRef] [Green Version]
- Balmus, I.-M.; Ilie-Dumitru, O.; Ciobica, A.; Cojocariu, R.-O.; Stanciu, C.; Trifan, A.; Cimpeanu, M.; Cimpeanu, C.; Gorgan, L. Irritable bowel syndrome between molecular approach and clinical expertise—Searching for gap fillers in the oxidative stress way of thinking. Medicina 2020, 56, 38. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Davila, A.; Liu, L.; Chellappa, K.; Redpath, P.; Nakamaru-Ogiso, E.; Paolella, L.M.; Zhang, Z.; Migaud, M.E.; Rabinowitz, J.D.; Baur, J.A. Nicotinamide adenine dinucleotide is transported into mammalian mitochondria. Elife 2018, 7, e33246. [Google Scholar] [CrossRef] [PubMed]
- Dölle, C.; Niere, M.; Lohndal, E.; Ziegler, M. Visualization of subcellular NAD pools and intra-organellar protein localization by poly-ADP-ribose formation. Cell. Mol. Life Sci. 2010, 67, 433–443. [Google Scholar] [CrossRef] [PubMed]
- Agrimi, G.; Russo, A.; Scarcia, P.; Palmieri, F. The human gene SLC25A17 encodes a peroxisomal transporter of coenzyme A, FAD and NAD+. Biochem. J. 2012, 443, 241–247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Senesi, S.; Csala, M.; Marcolongo, P.; Fulceri, R.; Mandl, J.; Bánhegyi, G.; Benedetti, A. Hexose-6-phosphate dehydrogenase in the endoplasmic reticulum. Biol. Chem. 2009, 391, 1–8. [Google Scholar] [CrossRef] [PubMed]
- Fabrizio, G.; Di Paola, S.; Stilla, A.; Giannotta, M.; Ruggiero, C.; Menzel, S.; Koch-Nolte, F.; Sallese, M.; Di Girolamo, M. ARTC1-mediated ADP-ribosylation of GRP78/BiP: A new player in endoplasmic-reticulum stress responses. Cell. Mol. Life Sci. 2015, 72, 1209–1225. [Google Scholar] [CrossRef]
- Nikiforov, A.; Kulikova, V.; Ziegler, M. The human NAD metabolome: Functions, metabolism and compartmentalization. Crit. Rev. Biochem. Mol. Biol. 2015, 50, 284–297. [Google Scholar] [CrossRef] [Green Version]
- Kirkland, J.B.; Meyer-Ficca, M.L. Chapter three - Niacin. In New Research and Developments of Water-Soluble Vitamins; Eskin, M.N.A., Ed.; Academic Press: Cambridge, MA, USA, 2018; pp. 83–149. [Google Scholar]
- Morales, J.; Li, L.; Fattah, F.J.; Dong, Y.; Bey, E.A.; Patel, M.; Gao, J.; Boothman, D.A. Review of poly (ADP-ribose) polymerase (PARP) mechanisms of action and rationale for targeting in cancer and other diseases. Crit. Rev. Eukaryot. Gene Expr. 2014, 24, 15–28. [Google Scholar] [CrossRef] [Green Version]
- Cohen, M.S.; Chang, P. Insights into the biogenesis, function, and regulation of ADP-ribosylation. Nat. Chem. Biol. 2018, 14, 236–243. [Google Scholar] [CrossRef] [Green Version]
- Palazzo, L.; Mikoč, A.; Ahel, I. ADP-ribosylation: New facets of an ancient modification. Febs J. 2017, 284, 2932–2946. [Google Scholar] [CrossRef]
- Perina, D.; Mikoč, A.; Ahel, J.; Ćetković, H.; Žaja, R.; Ahel, I. Distribution of protein poly(ADP-ribosyl)ation systems across all domains of life. DNA Repair 2014, 23, 4–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vivelo, C.A.; Leung, A.K.L. Proteomics approaches to identify mono-(ADP-ribosyl)ated and poly(ADP-ribosyl)ated proteins. Proteomics 2015, 15, 203–217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Brown, J.S.; O’Carrigan, B.; Jackson, S.P.; Yap, T.A. Targeting DNA repair in cancer: Beyond PARP inhibitors. Cancer Discov. 2017, 7, 20–37. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jubin, T.; Kadam, A.; Jariwala, M.; Bhatt, S.; Sutariya, S.; Gani, A.R.; Gautam, S.; Begum, R. The PARP family: Insights into functional aspects of poly (ADP-ribose) polymerase-1 in cell growth and survival. Cell Prolif. 2016, 49, 421–437. [Google Scholar] [CrossRef]
- Jacobson, M.; Jacobson, E. Therapeutic targeting of the PARPs: Future challenges, future opportunities. In PARP as a Therapeutic Target; CRC Press: Bota Roton, FL, USA, 2002; pp. 321–328. [Google Scholar]
- Kouzarides, T. Acetylation: A regulatory modification to rival phosphorylation? Embo J. 2000, 19, 1176–1179. [Google Scholar] [CrossRef]
- Yang, X.-J.; Seto, E. HATs and HDACs: From structure, function and regulation to novel strategies for therapy and prevention. Oncogene 2007, 26, 5310–5318. [Google Scholar] [CrossRef]
- Poulose, N.; Raju, R. Sirtuin regulation in aging and injury. Biochim. Biophys. Acta 2015, 1852, 2442–2455. [Google Scholar] [CrossRef] [Green Version]
- Michishita, E.; Park, J.Y.; Burneskis, J.M.; Barrett, J.C.; Horikawa, I. Evolutionarily conserved and nonconserved cellular localizations and functions of human SIRT proteins. Mol. Biol. Cell 2005, 16, 4623–4635. [Google Scholar] [CrossRef] [Green Version]
- Michan, S.; Sinclair, D. Sirtuins in mammals: Insights into their biological function. Biochem. J. 2007, 404, 1–13. [Google Scholar] [CrossRef] [Green Version]
- Haigis, M.C.; Sinclair, D.A. Mammalian sirtuins: Biological insights and disease relevance. Annu. Rev. Pathol. 2010, 5, 253–295. [Google Scholar] [CrossRef] [Green Version]
- Houtkooper, R.H. NAD+ as a pharmacological tool to boost sirtuin activity bt - sirtuins. In Sirtuins; Houtkooper, R.H., Ed.; Springer: Dordrecht, The Netherlands, 2016; pp. 29–40. [Google Scholar]
- Mesquita, I.; Vergnes, B.; Silvestre, R. Alterations on cellular redox states upon infection and implications for host cell homeostasis BT - Metabolic interaction in infection. In Metabolic Interaction in Infection; Silvestre, R., Torrado, E., Eds.; Springer International Publishing: Cham, Switzerland, 2018; pp. 197–220. [Google Scholar] [CrossRef]
- Quarona, V.; Zaccarello, G.; Chillemi, A.; Brunetti, E.; Singh, V.K.; Ferrero, E.; Funaro, A.; Horenstein, A.L.; Malavasi, F. CD38 and CD157: A long journey from activation markers to multifunctional molecules. Cytom. Part B Clin. Cytom. 2013, 84B, 207–217. [Google Scholar] [CrossRef] [PubMed]
- Clapham, D.E. Calcium Signaling. Cell 2007, 131, 1047–1058. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, H.C. Physiological functions of cyclic ADP-ribose and NAADP as calcium messengers. Annu. Rev. Pharm. Toxicol. 2001, 41, 317–345. [Google Scholar] [CrossRef] [PubMed]
- Malavasi, F.; Deaglio, S.; Funaro, A.; Ferrero, E.; Horenstein, A.L.; Ortolan, E.; Vaisitti, T.; Aydin, S. Evolution and function of the ADP ribosyl cyclase/CD38 gene family in physiology and pathology. Physiol. Rev. 2008, 88, 841–886. [Google Scholar] [CrossRef] [Green Version]
- Li, P.-L.; Tang, W.-X.; Valdivia, H.H.; Zou, A.-P.; Campbell, W.B. cADP-ribose activates reconstituted ryanodine receptors from coronary arterial smooth muscle. Am. J. Physiol. Circ. Physiol. 2001, 280, H208–H215. [Google Scholar] [CrossRef]
- Patel, S.; Marchant, J.S.; Brailoiu, E. Two-pore channels: Regulation by NAADP and customized roles in triggering calcium signals. Cell Calcium 2010, 47, 480–490. [Google Scholar] [CrossRef] [Green Version]
- Perraud, A.-L.; Fleig, A.; Dunn, C.A.; Bagley, L.A.; Launay, P.; Schmitz, C.; Stokes, A.J.; Zhu, Q.; Bessman, M.J.; Penner, R.; et al. ADP-ribose gating of the calcium-permeable LTRPC2 channel revealed by Nudix motif homology. Nature 2001, 411, 595–599. [Google Scholar] [CrossRef]
- Cui, C.; Merritt, R.; Fu, L.; Pan, Z. Targeting calcium signaling in cancer therapy. Acta Pharm. Sin. B 2017, 7, 3–17. [Google Scholar] [CrossRef]
- Zong, W.-X.; Ditsworth, D.; Bauer, D.E.; Wang, Z.-Q.; Thompson, C.B. Alkylating DNA damage stimulates a regulated form of necrotic cell death. Genes Dev. 2004, 18, 1272–1282. [Google Scholar] [CrossRef] [Green Version]
- Zong, W.-X.; Thompson, C.B. Necrotic death as a cell fate. Genes Dev. 2006, 20, 1–15. [Google Scholar] [CrossRef] [Green Version]
- Bellac, C.L.; Coimbra, R.S.; Christen, S.; Leib, S.L. Inhibition of the kynurenine-NAD+ pathway leads to energy failure and exacerbates apoptosis in pneumococcal meningitis. J. Neuropathol. Exp. Neurol. 2010, 69, 1096–1104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Takeuchi, M.; Yamamoto, T. Apoptosis induced by NAD depletion is inhibited by KN-93 in a CaMKII-independent manner. Exp. Cell Res. 2015, 335, 62–67. [Google Scholar] [CrossRef] [PubMed]
- Wright, S.C.; Wei, Q.S.; Kinder, D.H.; Larrick, J.W. Biochemical pathways of apoptosis: Nicotinamide adenine dinucleotide-deficient cells are resistant to tumor necrosis factor or ultraviolet light activation of the 24-kD apoptotic protease and DNA fragmentation. J. Exp. Med. 1996, 183, 463–471. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Asher, G.; Lotem, J.; Cohen, B.; Sachs, L.; Shaul, Y. Regulation of p53 stability and p53-dependent apoptosis by NADH quinone oxidoreductase 1. Proc. Natl. Acad. Sci. USA 2001, 98, 1188–1193. [Google Scholar] [CrossRef] [PubMed]
- Xie, L.; Yu, S.; Wang, Z.; Yang, K.; Liu, Z.; Li, C.; Liang, Y. nicotinamide adenine dinucleotide protects against spinal cord ischemia reperfusion injury-induced apoptosis by blocking autophagy. Oxid. Med. Cell. Longev. 2017, 2017, 7063874. [Google Scholar] [CrossRef]
- Vaquero, E.C.; Edderkaoui, M.; Pandol, S.J.; Gukovsky, I.; Gukovskaya, A.S. Reactive oxygen species produced by Nad(P)H oxidase inhibit apoptosis in pancreatic cancer cells. J. Biol. Chem. 2004, 279, 34643–34654. [Google Scholar] [CrossRef] [Green Version]
- Chong, Z.Z.; Lin, S.-H.; Maiese, K. The NAD+ precursor nicotinamide governs neuronal survival during oxidative stress through protein kinase B coupled to FOXO3a and mitochondrial membrane potential. J. Cereb. Blood Flow Metab. 2004, 24, 728–743. [Google Scholar] [CrossRef] [Green Version]
- Lu, L.; Tang, L.E.; Wei, W.; Hong, Y.; Chen, H.; Ying, W.; Chen, S. Nicotinamide mononucleotide improves energy activity and survival rate in an in vitro model of Parkinson’s disease. Exp. Med. 2014, 8, 943–950. [Google Scholar] [CrossRef] [Green Version]
- Gendron, M.C.; Schrantz, N.; Métivier, D.; Kroemer, G.; Maciorowska, Z.; Sureau, F.; Koester, S.; Petit, P.X. Oxidation of pyridine nucleotides during Fas- and ceramide-induced apoptosis in Jurkat cells: Correlation with changes in mitochondria, glutathione depletion, intracellular acidification and caspase 3 activation. Biochem. J. 2001, 353, 357–367. [Google Scholar]
- Zoratti, M.; Szabò, I. The mitochondrial permeability transition. Biochim. Biophys. Acta - Rev. Biomembr. 1995, 1241, 139–176. [Google Scholar] [CrossRef]
- Jaeschke, H.; Kleinwaechter, C.; Wendel, A. NADH-Dependent reductive stress and ferritin-bound iron in allyl alcohol-induced lipid peroxidation in vivo: The protective effect of vitamin E. Chem. Biol. Interact. 1992, 81, 57–68. [Google Scholar] [CrossRef]
- Yeung, F.; Hoberg, J.E.; Ramsey, C.S.; Keller, M.D.; Jones, D.R.; Frye, R.A.; Mayo, M.W. Modulation of NF-kappaB-dependent transcription and cell survival by the SIRT1 deacetylase. Embo J. 2004, 23, 2369–2380. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kil, I.S.; Shin, S.W.; Yeo, H.S.; Lee, Y.S.; Park, J.-W. Mitochondrial NADP+ - dependent isocitrate dehydrogenase protects cadmium-induced apoptosis. Mol. Pharm. 2006, 70, 1053–1061. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, J.H.; Park, J.-W. Oxalomalate regulates ionizing radiation-induced apoptosis in mice. Free Radic. Biol. Med. 2007, 42, 44–51. [Google Scholar] [CrossRef] [PubMed]
- Ramos, K.L.; Colquhoun, A. Protective role of glucose-6-phosphate dehydrogenase activity in the metabolic response of C6 rat glioma cells to polyunsaturated fatty acid exposure. Glia 2003, 43, 149–166. [Google Scholar] [CrossRef] [PubMed]
- Abramov, A.Y.; Duchen, M.R. The role of an astrocytic NADPH oxidase in the neurotoxicity of amyloid beta peptides. Philos. Trans. R. Soc. Lond. B. Biol. Sci. 2005, 360, 2309–2314. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Abramov, A.Y.; Canevari, L.; Duchen, M.R. Beta-amyloid peptides induce mitochondrial dysfunction and oxidative stress in astrocytes and death of neurons through activation of NADPH oxidase. J. Neurosci. 2004, 24, 565–575. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Abramov, A.Y.; Scorziello, A.; Duchen, M.R. Three distinct mechanisms generate oxygen free radicals in neurons and contribute to cell death during anoxia and reoxygenation. J. Neurosci. 2007, 27, 1129–1138. [Google Scholar] [CrossRef]
- Ryter, S.W.; Kim, H.P.; Hoetzel, A.; Park, J.W.; Nakahira, K.; Wang, X.; Choi, A.M.K. mechanisms of cell death in oxidative stress. Antioxid. Redox Signal. 2006, 9, 49–89. [Google Scholar] [CrossRef]
- Andreone, T.L.; O’Connor, M.; Denenberg, A.; Hake, P.W.; Zingarelli, B. Poly(ADP-Ribose) polymerase-1 regulates activation of activator protein-1 in murine fibroblasts. J. Immunol. 2003, 170, 2113–2120. [Google Scholar] [CrossRef] [Green Version]
- Ha, H.C.; Hester, L.D.; Snyder, S.H. Poly(ADP-ribose) polymerase-1 dependence of stress-induced transcription factors and associated gene expression in glia. Proc. Natl. Acad. Sci. USA 2002, 99, 3270–3275. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, M.Y.; Zhang, T.; Kraus, W.L. Poly(ADP-ribosyl)ation by PARP-1: ‘PAR-laying’ NAD+ into a nuclear signal. Genes Dev. 2005, 19, 1951–1967. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, M.; Naidu, P.; Yu, Y.; Berger, N.A.; Kannan, P. Dual regulation of AP-2alpha transcriptional activation by poly(ADP-ribose) polymerase-1. Biochem. J. 2004, 382, 323–329. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.; Oh, H.J.; Lau, Y.-F.C. The poly(ADP-ribose) polymerase 1 interacts with Sry and modulates its biological functions. Mol. Cell. Endocrinol. 2006, 257–258, 35–46. [Google Scholar] [CrossRef]
- Martín-Oliva, D.; O’Valle, F.; Muñoz-Gámez, J.A.; Valenzuela, M.T.; Nuñez, M.I.; Aguilar, M.; de Almodóvar, J.M.R.; del Moral, R.G.; Oliver, F.J. Crosstalk between PARP-1 and NF-κB modulates the promotion of skin neoplasia. Oncogene 2004, 23, 5275–5283. [Google Scholar] [CrossRef] [Green Version]
- Kim, M.Y.; Mauro, S.; Gévry, N.; Lis, J.T.; Kraus, W.L. NAD+-dependent modulation of chromatin structure and transcription by nucleosome binding properties of PARP-1. Cell 2004, 119, 803–814. [Google Scholar] [CrossRef] [Green Version]
- D’Amours, D.; Desnoyers, S.; D’Silva, I.; Poirier, G.G. Poly(ADP-ribosyl)ation reactions in the regulation of nuclear functions. Biochem. J. 1999, 342, 249–268. [Google Scholar] [CrossRef] [Green Version]
- Poirier, G.G.; de Murcia, G.; Jongstra-Bilen, J.; Niedergang, C.; Mandel, P. Poly(ADP-ribosyl)ation of polynucleosomes causes relaxation of chromatin structure. Proc. Natl. Acad. Sci. USA 1982, 79, 3423–3427. [Google Scholar] [CrossRef] [Green Version]
- Meisterernst, M.; Stelzer, G.; Roeder, R.G. Poly(ADP-ribose) polymerase enhances activator-dependent transcription. Proc. Natl. Acad. Sci. USA 1997, 94, 2261–2265. [Google Scholar] [CrossRef] [Green Version]
- Oei, S.L.; Griesenbeck, J.; Schweiger, M.; Ziegler, M. Regulation of RNA polymerase II-dependent Transcription by Poly(ADP-ribosyl)ation of transcription factors. J. Biol. Chem. 1998, 273, 31644–31647. [Google Scholar] [CrossRef] [Green Version]
- Oei, S.L.; Griesenbeck, J.; Ziegler, M.; Schweiger, M. A novel function of poly(ADP-ribosyl)ation: Silencing of RNA polymerase II-dependent transcription. Biochemistry 1998, 37, 1465–1469. [Google Scholar] [CrossRef] [PubMed]
- Amiri, K.I.; Ha, H.C.; Smulson, M.E.; Richmond, A. Differential regulation of CXC ligand 1 transcription in melanoma cell lines by poly(ADP-ribose) polymerase-1. Oncogene 2006, 25, 7714–7722. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yu, Z.; Kuncewicz, T.; Dubinsky, W.P.; Kone, B.C. Nitric oxide-dependent negative feedback of PARP-1 trans-activation of the inducible nitric-oxide synthase gene. J. Biol. Chem. 2006, 281, 9101–9109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Reale, A.; De Matteis, G.; Galleazzi, G.; Zampieri, M.; Caiafa, P. Modulation of DNMT1 activity by ADP-ribose polymers. Oncogene 2005, 24, 13–19. [Google Scholar] [CrossRef] [Green Version]
- Zardo, G.; D’Erme, M.; Reale, A.; Strom, R.; Perilli, M.; Caiafa, P. Does poly(ADP-ribosyl)ation regulate the DNA methylation pattern? Biochemistry 1997, 36, 7937–7943. [Google Scholar] [CrossRef]
- Zardo, G.; Marenzi, S.; Perilli, M.; Caiafa, P. Inhibition of poly(ADP-ribosyl)ation introduces an anomalous methylation pattern in transfected foreign DNA. Faseb J. 1999, 13, 1518–1522. [Google Scholar] [CrossRef]
- Zardo, G.; Reale, A.; Passananti, C.; Pradhan, S.; Buontempo, S.; de Matteis, G.; Adams, R.L.P.; Caiafa, P. Inhibition of poly(ADP-ribosyl)ation induces DNA hypermethylation: A possible molecular mechanism. Faseb J. 2002, 16, 1319–1321. [Google Scholar] [CrossRef] [Green Version]
- Luo, J.; Nikolaev, A.Y.; Imai, S.; Chen, D.; Su, F.; Shiloh, A.; Guarente, L.; Gu, W. Negative control of p53 by Sir2α promotes cell survival under stress. Cell 2001, 107, 137–148. [Google Scholar] [CrossRef] [Green Version]
- Motta, M.C.; Divecha, N.; Lemieux, M.; Kamel, C.; Chen, D.; Gu, W.; Bultsma, Y.; McBurney, M.; Guarente, L. Mammalian SIRT1 Represses forkhead transcription factors. Cell 2004, 116, 551–563. [Google Scholar] [CrossRef] [Green Version]
- Nakae, J.; Cao, Y.; Daitoku, H.; Fukamizu, A.; Ogawa, W.; Yano, Y.; Hayashi, Y. The LXXLL motif of murine forkhead transcription factor FoxO1 mediates Sirt1-dependent transcriptional activity. J. Clin. Investig. 2006, 116, 2473–2483. [Google Scholar] [CrossRef] [Green Version]
- Dai, J.M.; Wang, Z.Y.; Sun, D.C.; Lin, R.X.; Wang, S.Q. SIRT1 interacts with p73 and suppresses p73-dependent transcriptional activity. J. Cell. Physiol. 2007, 210, 161–166. [Google Scholar] [CrossRef] [PubMed]
- Pagans, S.; Pedal, A.; North, B.J.; Kaehlcke, K.; Marshall, B.L.; Dorr, A.; Hetzer-Egger, C.; Henklein, P.; Frye, R.; McBurney, M.W.; et al. SIRT1 regulates HIV transcription via tat deacetylation. PLoS Biol. 2005, 3, e41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ford, E.; Voit, R.; Liszt, G.; Magin, C.; Grummt, I.; Guarente, L. Mammalian Sir2 homolog SIRT7 is an activator of RNA polymerase I transcription. Genes Dev. 2006, 20, 1075–1080. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Muth, V.; Nadaud, S.; Grummt, I.; Voit, R. Acetylation of TAF(I)68, a subunit of TIF-IB/SL1, activates RNA polymerase I transcription. Embo J. 2001, 20, 1353–1362. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Poljsak, B.; Milisav, I. NAD+ as the link between oxidative stress, inflammation, caloric restriction, exercise, DNA repair, longevity, and health span. Rejuvenation Res. 2016, 19, 406–413. [Google Scholar] [CrossRef] [Green Version]
- Donmez, G.; Guarente, L. Aging and disease: Connections to sirtuins. Aging Cell 2010, 9, 285–290. [Google Scholar] [CrossRef]
- Johnson, S.; Imai, S.-I. NAD (+) biosynthesis, aging, and disease. F1000Research 2018, 7, 132. [Google Scholar] [CrossRef] [Green Version]
- Zhu, X.-H.; Lu, M.; Lee, B.-Y.; Ugurbil, K.; Chen, W. In vivo NAD assay reveals the intracellular NAD contents and redox state in healthy human brain and their age dependences. Proc. Natl. Acad. Sci. USA 2015, 112, 2876–2881. [Google Scholar] [CrossRef] [Green Version]
- Frederick, D.W.; Loro, E.; Liu, L.; Davila Jr, A.; Chellappa, K.; Silverman, I.M.; Quinn 3rd, W.J.; Gosai, S.J.; Tichy, E.D.; Davis, J.G.; et al. Loss of NAD Homeostasis Leads to Progressive and Reversible Degeneration of Skeletal Muscle. Cell Metab. 2016, 24, 269–282. [Google Scholar] [CrossRef] [Green Version]
- Stein, L.R.; Imai, S. Specific ablation of Nampt in adult neural stem cells recapitulates their functional defects during aging. Embo J. 2014, 33, 1321–1340. [Google Scholar] [CrossRef] [Green Version]
- van der Veer, E.; Ho, C.; O’Neil, C.; Barbosa, N.; Scott, R.; Cregan, S.P.; Pickering, J.G. Extension of human cell lifespan by nicotinamide phosphoribosyltransferase. J. Biol. Chem. 2007, 282, 10841–10845. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chang, H.-C.; Guarente, L. SIRT1 mediates central circadian control in the SCN by a mechanism that decays with aging. Cell 2013, 153, 1448–1460. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bai, P. Biology of poly(ADP-Ribose) polymerases: The factotums of cell maintenance. Mol. Cell 2015, 58, 947–958. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Camacho-Pereira, J.; Tarragó, M.G.; Chini, C.C.S.; Nin, V.; Escande, C.; Warner, G.M.; Puranik, A.S.; Schoon, R.A.; Reid, J.M.; Galina, A.; et al. CD38 dictates age-related NAD decline and mitochondrial dysfunction through an SIRT3-dependent mechanism. Cell Metab. 2016, 23, 1127–1139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Imai, S.; Guarente, L. NAD+ and sirtuins in aging and disease. Trends Cell Biol. 2014, 24, 464–471. [Google Scholar] [CrossRef]
- Abbotts, R.; Wilson, D.M., 3rd. Coordination of DNA single strand break repair. Free Radic. Biol. Med. 2017, 107, 228–244. [Google Scholar] [CrossRef]
- Tulić, L.; Vidaković, S.; Tulić, I.; Ćurčić, M.; Stojnić, J.; Jeremić, K. Oxidative stress markers in GnRH agonist and antagonist protocols in IVF. J. Med. Biochem. 2017, 36, 163–170. [Google Scholar] [CrossRef] [Green Version]
- Siristatidis, C.; Vogiatzi, P.; Varounis, C.; Askoxylaki, M.; Chrelias, C.; Papantoniou, N. The effect of reactive oxygen species on embryo quality in IVF. In Vivo 2016, 30, 149–153. [Google Scholar]
- Nishihara, T.; Matsumoto, K.; Hosoi, Y.; Morimoto, Y. Evaluation of antioxidant status and oxidative stress markers in follicular fluid for human in vitro fertilization outcome. Reprod. Med. Biol. 2018, 17, 481–486. [Google Scholar] [CrossRef] [Green Version]
- Borowiecka, M.; Wojsiat, J.; Polac, I.; Radwan, M.; Radwan, P.; Zbikowska, H.M. Oxidative stress markers in follicular fluid of women undergoing in vitro fertilization and embryo transfer. Syst. Biol. Reprod. Med. 2012, 58, 301–305. [Google Scholar] [CrossRef] [Green Version]
- Kumar, S.; Mishra, V.; Thaker, R.; Gor, M.; Perumal, S.; Joshi, P.; Sheth, H.; Shaikh, I.; Gautam, A.K.; Verma, Y. Role of environmental factors & oxidative stress with respect to in vitro fertilization outcome. Indian J. Med. Res. 2018, 148, S125–S133. [Google Scholar] [PubMed]
- Singh, A.K.; Chattopadhyay, R.; Chakravarty, B.; Chaudhury, K. Markers of oxidative stress in follicular fluid of women with endometriosis and tubal infertility undergoing IVF. Reprod. Toxicol. 2013, 42, 116–124. [Google Scholar] [CrossRef] [PubMed]
- Luddi, A.; Capaldo, A.; Focarelli, R.; Gori, M.; Morgante, G.; Piomboni, P.; De Leo, V. Antioxidants reduce oxidative stress in follicular fluid of aged women undergoing IVF. Reprod. Biol. Endocrinol. 2016, 14, 57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, S.; Sun, M.; Yu, L.; Wang, Y.; Yao, Y.; Wang, D. Niacin inhibits apoptosis and rescues premature ovarian failure. Cell. Physiol. Biochem. 2018, 50, 2060–2070. [Google Scholar] [CrossRef]
- Jandhyala, S.M.; Talukdar, R.; Subramanyam, C.; Vuyyuru, H.; Sasikala, M.; Nageshwar Reddy, D. Role of the normal gut microbiota. World J. Gastroenterol. 2015, 21, 8787–8803. [Google Scholar] [CrossRef]
- Lefter, R.; Ciobica, A.; Timofte, D.; Stanciu, C.; Trifan, A. A descriptive review on the prevalence of gastrointestinal disturbances and their multiple associations in autism spectrum disorder. Medicina 2019, 56, 11. [Google Scholar] [CrossRef] [Green Version]
- Hritcu, L.; Dumitru, I.; Padurariu, M.; Ciobica, A.; Spataru, C.; Spataru, C.; Stefanescu, G.; Stefanescu, C.; Grecu-Gabos, C. The modulation of oxytocin and cortisol levels in major depression disorder and irritable bowel syndrome. Rev. Chim. 2020, 71, 150–154. [Google Scholar] [CrossRef]
- Sfarti, C.; Ciobica, A.; Balmus, I.-M.; Ilie, O.-D.; Trifan, A.; Petrea, O.; Cojocariu, C.; Gîrleanu, I.; Sîngeap, A.M.; Stanciu, C. Systemic oxidative stress markers in cirrhotic patients with hepatic encephalopathy: Possible connections with systemic ammoniemia. Medicina 2020, 56, 196. [Google Scholar] [CrossRef] [Green Version]
- Zhang, J.; Zhao, X.; Jiang, Y.; Zhao, W.; Guo, T.; Cao, Y.; Teng, J.; Hao, X.; Zhao, J.; Yang, Z. Antioxidant status and gut microbiota change in an aging mouse model as influenced by exopolysaccharide produced by Lactobacillus plantarum YW11 isolated from Tibetan kefir. J. Dairy Sci. 2017, 100, 6025–6041. [Google Scholar] [CrossRef]
- Zhao, J.; Tian, F.; Yan, S.; Zhai, Q.; Zhang, H.; Chen, W. Lactobacillus plantarum CCFM10 alleviating oxidative stress and restoring the gut microbiota in d-galactose-induced aging mice. Food Funct. 2018, 9, 917–924. [Google Scholar] [CrossRef]
- Jose, S.; Bhalla, P.; Suraishkumar, G.K. Oxidative stress decreases the redox ratio and folate content in the gut microbe, enterococcus durans (MTCC 3031). Sci. Rep. 2018, 8, 12138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Reese, A.T.; Cho, E.H.; Klitzman, B.; Nichols, S.P.; Wisniewski, N.A.; Villa, M.M.; Durand, H.K.; Jiang, S.; Midani, F.S.; Nimmagadda, S.N.; et al. Antibiotic-induced changes in the microbiota disrupt redox dynamics in the gut. Elife 2018, 7, e35987. [Google Scholar] [CrossRef] [PubMed]
- Ge, Y.; Lin, S.; Li, B.; Yang, Y.; Tang, X.; Shi, Y.; Sun, J.; Le, G. oxidized pork induces oxidative stress and inflammation by altering gut microbiota in mice. Mol. Nutr. Food Res. 2020, 64, 1901012. [Google Scholar] [CrossRef] [PubMed]
- de Sousa, A.R.; de Castro Moreira, M.E.; Grancieri, M.; Toledo, R.C.L.; de Oliveira Araújo, F.; Mantovani, H.C.; Queiroz, V.A.V.; Martino, H.S.D. Extruded sorghum (Sorghum bicolor L.) improves gut microbiota, reduces inflammation, and oxidative stress in obese rats fed a high-fat diet. J. Funct. Foods 2019, 58, 282–291. [Google Scholar] [CrossRef]
- Yuzefpolskaya, M.; Bohn, B.; Nasiri, M.; Zuver, A.M.; Onat, D.D.; Royzman, E.A.; Nwokocha, J.; Mabasa, M.; Pinsino, A.; Brunjes, D.; et al. Gut microbiota, endotoxemia, inflammation, and oxidative stress in patients with heart failure, left ventricular assist device, and transplant. J. Heart Lung Transplant. 2020. [Google Scholar] [CrossRef]
- Kadariswantiningsih, I.; Thaha, M.; Wibisono, C.; Empitu, M. Thiol-producing microbiota of the intestine modulate oxidative stress and inflammation in Chronic Kidney Disease. Bali Med. J. 2019, 8, 307. [Google Scholar] [CrossRef]
- Bhalla, P.; Rengaswamy, R.; Karunagaran, D.; Suraishkumar, G.K.; Sahoo, S. Silver nanoparticle induced oxidative stress augments anticancer gut bacterial metabolites production. bioRxiv 2019, 658609. [Google Scholar] [CrossRef]
- Chi, L.; Bian, X.; Gao, B.; Tu, P.; Ru, H.; Lu, K. The effects of an environmentally relevant level of arsenic on the gut microbiome and its functional metagenome. Toxicol. Sci. 2017, 160, 193–204. [Google Scholar] [CrossRef]
- Stefanescu, C.; Ciobica, A. The relevance of oxidative stress status in first episode and recurrent depression. J. Affect. Disord. 2012, 143, 34–38. [Google Scholar] [CrossRef]
- Padurariu, M.; Ciobica, A.; Hritcu, L.; Stoica, B.; Bild, W.; Stefanescu, C. Changes of some oxidative stress markers in the serum of patients with mild cognitive impairment and Alzheimer’s disease. Neurosci. Lett. 2010, 469, 6–10. [Google Scholar] [CrossRef]
- Balmuș, I.-M.; Strungaru, S.-A.; Ciobica, A.; Nicoara, M.-N.; Dobrin, R.; Plavan, G.; Ștefănescu, C. Preliminary data on the interaction between some biometals and oxidative stress status in mild cognitive impairment and Alzheimer’s disease patients. Oxid. Med. Cell. Longev. 2017, 2017, 7156928. [Google Scholar] [CrossRef] [PubMed]
- Balmus, I.M.; Ciobica, A.; Antioch, I.; Dobrin, R.; Timofte, D. Oxidative stress implications in the affective disorders: Main biomarkers, animal models relevance, genetic perspectives, and antioxidant approaches. Oxid. Med. Cell. Longev. 2016, 2016, 3975101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Balmus, I.M.; Ciobica, A.; Trifan, A.; Stanciu, C. The implications of oxidative stress and antioxidant therapies in inflammatory bowel disease: Clinical aspects and animal models. Saudi J. Gastroenterol. 2016, 22, 3–17. [Google Scholar] [CrossRef] [PubMed]
- Ciobica, A.; Padurariu, M.; Dobrin, I.; Stefanescu, C.; Dobrin, R. Oxidative stress in schizophrenia - Focusing on the main markers. Psychiatr. Danub. 2011, 23, 237–245. [Google Scholar]
- Padurariu, M.; Ciobica, A.; Lefter, R.; Serban, I.; Stefanescu, C.; Chirita, R. The oxidative stress hypothesis in Alzheimer’s disease. Psychiatr. Danub. 2013, 25, 401–409. [Google Scholar]
- Aktas, B.; Aslim, B. Gut-lung axis and dysbiosis in COVID-19. Turk. J. Biol. Turk Biyol. Derg. 2020, 44, 265–272. [Google Scholar]
- Guan, W.; Ni, Z.; Hu, Y.; Liang, W.; Ou, C.; He, J.; Liu, L.; Shan, H.; Lei, C.; Hui, D.S.C.; et al. Clinical characteristics of coronavirus disease 2019 in China. N. Engl. J. Med. 2020, 382, 1708–1720. [Google Scholar] [CrossRef]
- Huang, C.; Wang, Y.; Li, X.; Ren, L.; Zhao, J.; Hu, Y.; Zhang, L.; Fan, G.; Xu, J.; Gu, X.; et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 2020, 395, 497–506. [Google Scholar] [CrossRef] [Green Version]
- Chen, N.; Zhou, M.; Dong, X.; Qu, J.; Gong, F.; Han, Y.; Qiu, Y.; Wang, J.; Liu, Y.; Wei, Y.; et al. Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: A descriptive study. Lancet 2020, 395, 507–513. [Google Scholar] [CrossRef] [Green Version]
- Xu, X.-W.; Wu, X.-X.; Jiang, X.-G.; Xu, K.-J.; Ying, L.-J.; Ma, C.-L.; Li, S.-B.; Wang, H.-Y.; Zhang, S.; Gao, H.-N.; et al. Clinical findings in a group of patients infected with the 2019 novel coronavirus (SARS-Cov-2) outside of Wuhan, China: Retrospective case series. BMJ 2020, 368, m606. [Google Scholar] [CrossRef] [Green Version]
- Jin, X.; Lian, J.-S.; Hu, J.-H.; Gao, J.; Zheng, L.; Zhang, Y.-M.; Hao, S.-R.; Jia, H.-Y.; Cai, H.; Zhang, X.-L.; et al. Epidemiological, clinical and virological characteristics of 74 cases of coronavirus-infected disease 2019 (COVID-19) with gastrointestinal symptoms. Gut 2020, 69, 1002–1009. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhou, F.; Yu, T.; Du, R.; Fan, G.; Liu, Y.; Liu, Z.; Xiang, J.; Wang, Y.; Song, B.; Gu, X.; et al. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: A retrospective cohort study. Lancet 2020, 395, 1054–1062. [Google Scholar] [CrossRef]
- Wang, D.; Hu, B.; Hu, C.; Zhu, F.; Liu, X.; Zhang, J.; Wang, B.; Xiang, H.; Cheng, Z.; Xiong, Y.; et al. Clinical characteristics of 138 hospitalized patients with 2019 novel coronavirus–Infected pneumonia in Wuhan, China. JAMA 2020, 323, 1061–1069. [Google Scholar] [CrossRef] [PubMed]
- Lin, L.; Jiang, X.; Zhang, Z.; Huang, S.; Zhang, Z.; Fang, Z.; Gu, Z.; Gao, L.; Shi, H.; Mai, L.; et al. Gastrointestinal symptoms of 95 cases with SARS-CoV-2 infection. Gut 2020, 69, 997–1001. [Google Scholar] [CrossRef]
- Zhang, H.; Kang, Z.; Gong, H.; Xu, D.; Wang, J.; Li, Z.; Li, Z.; Cui, X.; Xiao, J.; Zhan, J.; et al. Digestive system is a potential route of COVID-19: An analysis of single-cell coexpression pattern of key proteins in viral entry process. Gut 2020, 69, 1010–1018. [Google Scholar] [CrossRef]
- Ong, J.; Young, B.E.; Ong, S. COVID-19 in gastroenterology: A clinical perspective. Gut 2020, 69, 1144–1145. [Google Scholar] [CrossRef]
- Song, Y.; Liu, P.; Shi, X.L.; Chu, Y.L.; Zhang, J.; Xia, J.; Gao, X.Z.; Qu, T.; Wang, M.Y. SARS-CoV-2 induced diarrhoea as onset symptom in patient with COVID-19. Gut 2020, 69, 1143–1144. [Google Scholar] [CrossRef] [Green Version]
- Liang, W.; Feng, Z.; Rao, S.; Xiao, C.; Xue, X.; Lin, Z.; Zhang, Q.; Qi, W. Diarrhoea may be underestimated: A missing link in 2019 novel coronavirus. Gut 2020, 69, 1141–1143. [Google Scholar] [CrossRef] [Green Version]
- Kibune-Nagasako, C.; García-Montes, C.; Silva-Lorena, S.L.; Aparecida-Mesquita, M. Irritable bowel syndrome subtypes: Clinical and psychological features, body mass index and comorbidities. Rev. Española Enferm. Dig. 2016, 108, 59–64. [Google Scholar] [CrossRef] [Green Version]
- Martin, C.R.; Osadchiy, V.; Kalani, A.; Mayer, E.A. The brain-gut-microbiome axis. Cell. Mol. Gastroenterol. Hepatol. 2018, 6, 133–148. [Google Scholar] [CrossRef] [Green Version]
- Chen, T.; Wu, D.; Chen, H.; Yan, W.; Yang, D.; Chen, G.; Ma, K.; Xu, D.; Yu, H.; Wang, H.; et al. Clinical characteristics of 113 deceased patients with coronavirus disease 2019: Retrospective study. BMJ 2020, 368, m1091. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Magro, F.; Lopes, J.; Borralho, P.; Lopes, S.; Coelho, R.; Cotter, J.; de Castro, F.D.; de Sousa, H.T.; Salgado, M.; Andrade, P.; et al. Comparison of different histological indexes in the assessment of UC activity and their accuracy regarding endoscopic outcomes and faecal calprotectin levels. Gut 2019, 68, 594–603. [Google Scholar] [CrossRef] [PubMed]
- Effenberger, M.; Grabherr, F.; Mayr, L.; Schwaerzler, J.; Nairz, M.; Seifert, M.; Hilbe, R.; Seiwald, S.; Scholl-Buergi, S.; Fritsche, G.; et al. Faecal calprotectin indicates intestinal inflammation in COVID-19. Gut 2020, 69, 1543–1544. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Taxonera, C.; Sagastagoitia, I.; Alba, C.; Mañas, N.; Olivares, D.; Rey, E. 2019 novel coronavirus disease (COVID-19) in patients with inflammatory bowel diseases. Aliment. Pharm. 2020, 52, 276–283. [Google Scholar] [CrossRef] [PubMed]
- Mazza, S.; Sorce, A.; Peyvandi, F.; Vecchi, M.; Caprioli, F. A fatal case of COVID-19 pneumonia occurring in a patient with severe acute ulcerative colitis. Gut 2020, 69, 1148–1149. [Google Scholar] [CrossRef] [PubMed]
- Wu, Y.; Guo, C.; Tang, L.; Hong, Z.; Zhou, J.; Dong, X.; Yin, H.; Xiao, Q.; Tang, Y.; Qu, X.; et al. Prolonged presence of SARS-CoV-2 viral RNA in faecal samples. Lancet Gastroenterol. Hepatol. 2020, 5, 434–435. [Google Scholar] [CrossRef]
- Holshue, M.L.; DeBolt, C.; Lindquist, S.; Lofy, K.H.; Wiesman, J.; Bruce, H.; Spitters, C.; Ericson, K.; Wilkerson, S.; Tural, A.; et al. First Case of 2019 Novel Coronavirus in the United States. N. Engl. J. Med. 2020, 382, 929–936. [Google Scholar] [CrossRef] [PubMed]
- Zheng, S.; Fan, J.; Yu, F.; Feng, B.; Lou, B.; Zou, Q.; Xie, G.; Lin, S.; Wang, R.; Yang, X.; et al. Viral load dynamics and disease severity in patients infected with SARS-CoV-2 in Zhejiang province, China, January-March 2020: Retrospective cohort study. BMJ 2020, 369, m1443. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sriwijitalai, W.; Wiwanitkit, V. COVID-19 in forensic medicine unit personnel: Observation from Thailand. J. Forensic Leg. Med. 2020, 72, 101964. [Google Scholar] [CrossRef] [PubMed]
- Xiao, F.; Tang, M.; Zheng, X.; Liu, Y.; Li, X.; Shan, H. Evidence for gastrointestinal infection of SARS-CoV-2. Gastroenterology 2020, 158, 1831–1833. [Google Scholar] [CrossRef]
- Gao, Q.Y.; Chen, Y.X.; Fang, J.Y. 2019 novel coronavirus infection and gastrointestinal tract. J. Dig. Dis. 2020, 21, 125–126. [Google Scholar] [CrossRef] [Green Version]
- Yang, Y.; Shen, C.; Li, J.; Yuan, J.; Yang, M.; Wang, F.; Li, G.; Li, Y.; Xing, L.; Peng, L.; et al. Exuberant elevation of IP-10, MCP-3 and IL-1ra during SARS-CoV-2 infection is associated with disease severity and fatal outcome. medRxiv 2020. [Google Scholar] [CrossRef] [Green Version]
- Gou, W.; Fu, Y.; Yue, L.; Chen, G.; Cai, X.; Shuai, M.; Xu, F.; Yi, X.; Chen, H.; Zhu, Y.J.; et al. Gut microbiota may underlie the predisposition of healthy individuals to COVID-19. medRxiv 2020. [Google Scholar] [CrossRef] [Green Version]
- Devaux, C.A.; Rolain, J.-M.; Raoult, D. ACE2 receptor polymorphism: Susceptibility to SARS-CoV-2, hypertension, multi-organ failure, and COVID-19 disease outcome. J. Microbiol. Immunol. Infect. 2020, 53, 425–435. [Google Scholar] [CrossRef] [PubMed]
- Hashimoto, T.; Perlot, T.; Rehman, A.; Trichereau, J.; Ishiguro, H.; Paolino, M.; Sigl, V.; Hanada, T.; Hanada, R.; Lipinski, S.; et al. ACE2 links amino acid malnutrition to microbial ecology and intestinal inflammation. Nature 2012, 487, 477–481. [Google Scholar] [CrossRef]
- Xu, H.; Zhong, L.; Deng, J.; Peng, J.; Dan, H.; Zeng, X.; Li, T.; Chen, Q. High expression of ACE2 receptor of 2019-nCoV on the epithelial cells of oral mucosa. Int. J. Oral Sci. 2020, 12, 8. [Google Scholar] [CrossRef]
- Zheng, M.; Gao, Y.; Wang, G.; Song, G.; Liu, S.; Sun, D.; Xu, Y.; Tian, Z. Functional exhaustion of antiviral lymphocytes in COVID-19 patients. Cell. Mol. Immunol. 2020, 17, 533–535. [Google Scholar] [CrossRef] [Green Version]
- Wang, F.; Nie, J.; Wang, H.; Zhao, Q.; Xiong, Y.; Deng, L.; Song, S.; Ma, Z.; Mo, P.; Zhang, Y. Characteristics of peripheral lymphocyte subset alteration in COVID-19 pneumonia. J. Infect. Dis. 2020, 221, 1762–1769. [Google Scholar] [CrossRef] [Green Version]
- Bellono, N.W.; Bayrer, J.R.; Leitch, D.B.; Castro, J.; Zhang, C.; O’Donnell, T.A.; Brierley, S.M.; Ingraham, H.A.; Julius, D. Enterochromaffin cells are gut chemosensors that couple to sensory neural pathways. Cell 2017, 170, 185–198. [Google Scholar] [CrossRef] [Green Version]
- Rios, D.; Wood, M.B.; Li, J.; Chassaing, B.; Gewirtz, A.T.; Williams, I.R. Antigen sampling by intestinal M cells is the principal pathway initiating mucosal IgA production to commensal enteric bacteria. Mucosal Immunol. 2016, 9, 907–916. [Google Scholar] [CrossRef] [Green Version]
- Yano, J.M.; Yu, K.; Donaldson, G.P.; Shastri, G.G.; Ann, P.; Ma, L.; Nagler, C.R.; Ismagilov, R.F.; Mazmanian, S.K.; Hsiao, E.Y. Indigenous bacteria from the gut microbiota regulate host serotonin biosynthesis. Cell 2015, 161, 264–276. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zang, R.; Castro, M.F.G.; McCune, B.T.; Zeng, Q.; Rothlauf, P.W.; Sonnek, N.M.; Liu, Z.; Brulois, K.F.; Wang, X.; Greenberg, H.B.; et al. TMPRSS2 and TMPRSS4 promote SARS-CoV-2 infection of human small intestinal enterocytes. Sci. Immunol. 2020, 5, eabc3582. [Google Scholar] [CrossRef] [PubMed]
- Lamers, M.M.; Beumer, J.; van der Vaart, J.; Knoops, K.; Puschhof, J.; Breugem, T.I.; Ravelli, R.B.G.; Paul van Schayck, J.; Mykytyn, A.Z.; Duimel, H.Q.; et al. SARS-CoV-2 productively infects human gut enterocytes. Science 2020, 369, 50–54. [Google Scholar] [CrossRef] [PubMed]
- Xu, Y.; Li, X.; Zhu, B.; Liang, H.; Fang, C.; Gong, Y.; Guo, Q.; Sun, X.; Zhao, D.; Shen, J.; et al. Characteristics of pediatric SARS-CoV-2 infection and potential evidence for persistent fecal viral shedding. Nat. Med. 2020, 26, 502–505. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, W.; Xu, Y.; Gao, R.; Lu, R.; Han, K.; Wu, G.; Tan, W. Detection of SARS-CoV-2 in different types of clinical specimens. JAMA 2020, 323, 1843–1844. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Long, Q.-X.; Liu, B.-Z.; Deng, H.-J.; Wu, G.-C.; Deng, K.; Chen, Y.-K.; Liao, P.; Qiu, J.-F.; Lin, Y.; Cai, X.-F.; et al. Antibody responses to SARS-CoV-2 in patients with COVID-19. Nat. Med. 2020, 26, 845–848. [Google Scholar] [CrossRef]
- Carrouel, F.; Conte, M.P.; Fisher, J.; Gonçalves, L.S.; Dussart, C.; Llodra, J.C.; Bourgeois, D. COVID-19: A recommendation to examine the effect of mouthrinses with β-cyclodextrin combined with citrox in preventing infection and progression. J. Clin. Med. 2020, 9, 1126. [Google Scholar] [CrossRef] [Green Version]
- Zhou, D.; Dai, S.-M.; Tong, Q. COVID-19: A recommendation to examine the effect of hydroxychloroquine in preventing infection and progression. J. Antimicrob. Chemother. 2020, 75, 1667–1670. [Google Scholar] [CrossRef]
- Bescos, R.; Ashworth, A.; Cutler, C.; Brookes, Z.L.; Belfield, L.; Rodiles, A.; Casas-Agustench, P.; Farnham, G.; Liddle, L.; Burleigh, M.; et al. Effects of Chlorhexidine mouthwash on the oral microbiome. Sci. Rep. 2020, 10, 5254. [Google Scholar] [CrossRef] [Green Version]
- Dhar, D.; Mohanty, A. Gut microbiota and Covid-19- possible link and implications. Virus Res. 2020, 285, 198018. [Google Scholar] [CrossRef]
PARP | Subcellular Localization | Biological Role(s) [48] |
---|---|---|
PARP-1 | Nucleus | Gene regulation DNA damage response |
PARP-2 | Nucleus | DNA repair Base-excision repair |
PARP-3 | Nucleus | DNA repair |
PARP-4 | Cytosol | DNA repair Cell death Inflammation |
PARP-5a | Nucleus, cytosol | Wnt signaling pathways Cell division mRNA and protein transport Telomerase regulation Protein ubiquitination |
PARP-5b | Nucleus, cytosol | Wnt signaling pathways Telomerase regulation Protein ubiquitination |
PARP-6 | unknown | ADP-ribosyltransferase activity |
PARP-7 | unknown | Hormonal processes Embryonic development and morphogenesis |
PARP-8 | unknown | ADP-ribosyltransferase activity |
PARP-9 | Nucleus, cytosol | DNA repair Cell migration Response to interferon γ |
PARP-10 | Nucleus; cytosol | Cell proliferation Chromatin assembly regulation |
PARP-11 | unknown | ADP-ribosyltransferase activity |
PARP-12 | Nucleus | ADP-ribosyltransferase activity Nucleic acid binding Zinc ion binding |
PARP-13 | Cytosol, nucleus, plasma membrane, Golgi apparatus | Innate immune response |
PARP-14 | Cytosol, nucleus, plasma membrane | Transcriptional regulation |
PARP-15 | Nucleus | Transcriptional regulation |
PARP-16 | Endoplasmic reticulum Nuclear membrane | Response to unfolded proteins |
Sirtuin | Biological Role | Reference |
---|---|---|
SIRT1 | Metabolism Stress | [53,54] |
SIRT2 | Cell cycle | |
SIRT3 | Thermogenesis ATP production | |
SIRT4 | Insulin secretion | |
SIRT5 | Urea cycle | |
SIRT6 | Base-excision repair and metabolism | |
SIRT7 | rDNA |
DNA Repair Mechanism | Involvement | Reference |
---|---|---|
Base-excision repair | DNA polymerase β interaction | [122] |
X-ray repair cross-complementing protein 1 recruitment | ||
Activator protein 1 | ||
PARP-1 binding (PARP-2) | ||
X-repair cros-complementing protein 1 interaction (PARG) | ||
DNA ligase III binding (PARP-1) | ||
Nucleotide-excision repair | DNA repair protein comlementing XP-A cells association (PARP-1) | |
DNA damage-binding protein 2 (PARP-1) | ||
Xeroderma pigmentosum, complementation group C (PARP-1) | ||
Mismatch repair | DNA mismatch repair protein MutS Homolog 3 interaction (PARP-1) | |
Homologous recombination repair | Breast cancer type 1 susceptibility protein recruitment (PARP-1) | [33] |
Non-homologous end-joining repair | DNA-dependent protein kinase, catalytic subunit, Ku80/Ku70 heterodimer interaction (PARP-1) |
© 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Doroftei, B.; Ilie, O.-D.; Cojocariu, R.-O.; Ciobica, A.; Maftei, R.; Grab, D.; Anton, E.; McKenna, J.; Dhunna, N.; Simionescu, G. Minireview Exploring the Biological Cycle of Vitamin B3 and Its Influence on Oxidative Stress: Further Molecular and Clinical Aspects. Molecules 2020, 25, 3323. https://doi.org/10.3390/molecules25153323
Doroftei B, Ilie O-D, Cojocariu R-O, Ciobica A, Maftei R, Grab D, Anton E, McKenna J, Dhunna N, Simionescu G. Minireview Exploring the Biological Cycle of Vitamin B3 and Its Influence on Oxidative Stress: Further Molecular and Clinical Aspects. Molecules. 2020; 25(15):3323. https://doi.org/10.3390/molecules25153323
Chicago/Turabian StyleDoroftei, Bogdan, Ovidiu-Dumitru Ilie, Roxana-Oana Cojocariu, Alin Ciobica, Radu Maftei, Delia Grab, Emil Anton, Jack McKenna, Nitasha Dhunna, and Gabriela Simionescu. 2020. "Minireview Exploring the Biological Cycle of Vitamin B3 and Its Influence on Oxidative Stress: Further Molecular and Clinical Aspects" Molecules 25, no. 15: 3323. https://doi.org/10.3390/molecules25153323