REFERENCIAS BIBLIOGRÁFICAS

  • Dimauro S, Davidzon G. Mitochondrial DNA and disease. Ann Med. 2005;37(3):222-32.
  • Reddy PH. Mitochondrial medicine for aging and neurodegenerative diseases. Neuromolecular Med. 2008;10(4):291–315.
  • Karbowski M, Neutzner A. Neurodegeneration as a consequence of failed mitochondrial maintenance. Acta Neuropathol. 2012;123(2):157–171.
  • Limongelli G, Masarone D, D’Alessandro R, Elliott PM. Mitochondrial diseases and the heart: an overview of molecular basis, diagnosis, treatment and clinical course. Future Cardiol. 2012;8(1):71–88.
  • Bhatti JS, Bhatti GK, Reddy PH. Mitochondrial dysfunction and oxidative stress in metabolic disorders – A step towards mitochondria based therapeutic strategies. Biochim Biophys Acta Mol
  • Basis Dis. 2017 May;1863(5):1066-1077.
  • Li, X., Zhang, W., Cao, Q. et al. Mitochondrial dysfunction in fibrotic diseases. Cell Death Discov. 6, 80 (2020)
  • C Almeida, L Gaspar, B Santos, A Barros-Viegas, S Carmo Silva, C Travassos, F Teixeira, J Moita, C Cavadas, A R Álvaro ERJ Open Research 2021 7: 50;
  • Mao P, Reddy PH. Is multiple sclerosis a mitochondrial disease? Biochim Biophys Acta. 2010;1802(1):66–79.
  • Maiese K, Morhan SD, Chong ZZ. Oxidative stress biology and cell injury during type 1 and type 2 diabetes mellitus. Curr Neurovasc Res. 2007;4(1):63–71.
  • Palmieri L, Persico AM. Mitochondrial dysfunction in autism spectrum disorders: cause or effect? Biochim Biophys Acta. 2010;1797(6–7):1130–1137.
  • Chitkara DK, Nurko S, Shoffner JM, Buie T, Flores A. Abnormalities in gastrointestinal motility are associated with diseases of oxidative phosphorylation in children. Am J Gastroenterol. 2003;98(4):871–877.
  • Cordero MD, de Miguel M, Carmona-Lopez I, Bonal P, Campa F, Moreno-Fernandez AM. Oxidative stress and mitochondrial dysfunction in fibromyalgia. Neuro Endocrinol Lett. 2010;31(2):169–173.
  • Ashida H, Mimuro H, Ogawa M, et al. Cell death and infection: a double-edged sword for host and pathogen survival. J Cell Biol. 2011;195(6):931–942.
  • Kroenke K, Wood DR, Mangelsdorff AD, Meier NJ, Powell JB. Chronic fatigue in primary care: prevalence, patient characteristics, and outcome. JAMA. 1988;260(7):929–934.
  • Morrison JD. Fatigue as a presenting complaint in family practice. J Family Pract. 1980;10(5):795–801.
  • Wang Y, Yang Q, Wang H, Zhu J, Cong L, Li H, Sun Y. NAD+ deficiency and mitochondrial dysfunction in granulosa cells of women with polycystic ovary syndrome‡. Biol Reprod. 2021 Aug 3;105(2):371-380.
  • Shannon Chiang, Nady Braidy, Sanaz Maleki, Sean Lal, Des R. Richardson, Michael L.-H. Huang,
  • Mechanisms of impaired mitochondrial homeostasis and NAD+ metabolism in a model of mitochondrial heart disease exhibiting redox active iron accumulation, Redox Biology, Volume 46, 2021, 102038, ISSN 2213-2317.
  • Villavicencio-Queijeiro Alexa. La mitocondria como fábrica de cofactores: biosíntesis de grupo hemo, centros Fe-S y nucleótidos de flavina (FMN/FAD).
    Chambon, P., Weill, J. D. & Mandel, P. Nicotinamide mononucleotide activation of new DNA-dependent polyadenylic acid synthesizing nuclear enzyme. Biochem. Biophys. Res. Commun. 11, 39–43 (1963).
  • Imai, S., Armstrong, C. M., Kaeberlein, M. & Guarente, L. Transcriptional silencing and longevity protein Sir2 is an NAD-dependent histone deacetylase. Nature 403, 795–800 (2000).
  • Spiteller G. Is lipid peroxidation of polyunsaturated acids the only source of free radicals that induce aging and age-related diseases? Rejuvenation Res. 2010;13(1):91–103.
  • Stadtman E. Introduction to serial reviews on oxidatively modified proteins in aging and disease. Free Radic Biol Med. 2002;32(9):789.
  • Divakaruni AS, Brand MD. The regulation and physiology of mitochondrial proton leak. Physiology (Bethesda) 2011;26(3):192–205.
  • Duchen MR, Szabadkai G. Roles of mitochondria in human disease. Essays Biochem. 2010;47:115–137
  • Popov LD. Biogénesis mitocondrial: una actualización. J Célula Mol Med. 2020 mayo;24(9):4892-4899.
  • Twig G, Shirihai OS. The interplay between mitochondrial dynamics and mitophagy. Antioxid Redox Signal. 2011;14(10):1939–1951.
  • Popov LD. Mitochondrial biogenesis: An update. J Cell Mol Med. 2020 May;24(9):4892-4899..
  • Oka SI, Sabry AD, Cawley KM, Warren JS. Multiple Levels of PGC-1α Dysregulation in Heart Failure. Front Cardiovasc Med. 2020 Jan 30;7:2.
  • Gao J, Liu P, Shen Z, Xu K, Wu C, Tian F, Chen M, Wang L, Li P. Morroniside Promotes PGC-1α-Mediated Cholesterol Efflux in Sodium Palmitate or High Glucose-Induced Mouse Renal Tubular
  • Epithelial Cells. Biomed Res Int. 2021 Aug 24;2021:9942152.
  • Rius-Pérez S, Torres-Cuevas I, Millán I, Ortega ÁL, Pérez S. PGC-1α, Inflammation, and Oxidative Stress: An Integrative View in Metabolism. Oxid Med Cell Longev. 2020 Mar 9;2020:1452696.
  • Lauschke VM, Ingelman-Sundberg M. The Importance of Patient-Specific Factors for Hepatic Drug Response and Toxicity. Int J Mol Sci. 2016 Oct 12;17(10):1714.
  • Stoker ML, Newport E, Hulit JC, West AP, Morten KJ. Impact of pharmacological agents on mitochondrial function: a growing opportunity? Biochem Soc Trans. 2019 Dec 20;47(6):1757-1772.
  • Varga ZV, Ferdinandy P, Liaudet L, Pacher P. Drug-induced mitochondrial dysfunction and cardiotoxicity. Am J Physiol Heart Circ Physiol. 2015 Nov;309(9):H1453-67.
  • Morén C, Juárez-Flores DL, Cardellach F, Garrabou G. The Role of Therapeutic Drugs on Acquired Mitochondrial Toxicity. Curr Drug Metab. 2016;17(7):648-62.
  • Parikh S, Saneto R, Falk MJ, Anselm I, Cohen BH, Haas R, Medicine Society TM. A modern approach to the treatment of mitochondrial disease. Curr Treat Options Neurol. 2009 Nov;11(6):414-30.
  • Kerr DS. Treatment of mitochondrial electron transport chain disorders: a review of clinical trials over the past decade. Mol Genet Metab. 2010;99(3):246–255.
  • Nicolson GL. Mitochondrial Dysfunction and Chronic Disease: Treatment With Natural Supplements. Integr Med (Encinitas). 2014 Aug;13(4):35-43.
  • E. Wesselink, W.A.C. Koekkoek, S. Grefte, R.F. Witkamp, A.R.H. van Zanten, Feeding mitochondria: Potential role of nutritional components to improve critical illness convalescence,u Clinical
  • Nutrition, Volume 38, Issue 3, 2019, Pages 982-995.
  • Villa-Bellosta R. Dietary magnesium supplementation improves lifespan in a mouse model of progeria. EMBO Mol Med. 2020 Oct 7;12(10):e12423.
  • Chicco AJ, Sparagna GC. Role of cardiolipin alterations in mitochondrial dysfunction and disease. Am J Physiol Cell Physiol. 2007;292(1):C33–C44.
  • Afshordel S, Hagl S, Werner D, Röhner N, Kögel D, Bazan NG, Eckert GP. Omega-3 polyunsaturated fatty acids improve mitochondrial dysfunction in brain aging–impact of Bcl-2 and NPD-1 like metabolites. Prostaglandins Leukot Essent Fatty Acids. 2015 Jan;92:23-31.
  • Tarnopolsky MA, Roy BD, MacDonald JR. A randomized, controlled trial of creatine monohydrate in patients with mitochondrial cytopathies. Muscle Nerve. 1997;20:1502–1509.
  • Quinzii CM, Lopez LC, Naini A, DiMauro S, Hirano M. Human CoQ10 deficiencies. Biofactors. 2008;32:113–118. 3
  • Glover EI, Martin J, Maher A, Thornhill RE, Moran GR, Tarnopolsky MA. A randomized trial of coenzyme Q10 in mitochondrial disorders. Muscle Nerve. 2010;42:739–748.
  • Geier DA, Geier MR. L-carnitine exposure and mitochondrial function in human neuronal cells. Neurochem Res. 2013 Nov;38(11):2336-41.
  • Lettieri-Barbato D, Cannata SM, Casagrande V, Ciriolo MR, Aquilano K. Time-controlled fasting prevents aging-like mitochondrial changes induced by persistent dietary fat overload in skeletal muscle. PLoS One. 2018 May 9;13(5):e0195912.
  • Haupt S, Eckstein ML, Wolf A, Zimmer RT, Wachsmuth NB, Moser O. Eat, Train, Sleep-Retreat? Hormonal Interactions of Intermittent Fasting, Exercise and Circadian Rhythm. Biomolecules. 2021 Mar 30;11(4):516.