Review Article Volume 20 Issue 2 - 2025

Role of Food-Its Influence on Modulation of Therapies Including Chemotherapy’s Outcomes

Faiza Abdur Rab*

Assistant Professor, Department of Food Science and Technology, University of Karachi, Karachi, Pakistan

*Corresponding Author: Faiza Abdur Rab, Assistant Professor, Department of Food Science and Technology, University of Karachi, Karachi, Pakistan.
Received: September 04, 2025; Published: September 29, 2025



Food provides basic ingredients that a given living body can or cannot synthesis to enable it to perform its normal functions, in addition to providing energy. Another under-acknowledged role of food is to protect the given body from illness and to cure current ailment on regular use. Many non-contagious diseases share the same interconnected genomic or epigenetically driven molecular pathways leading to cause illnesses as well as to decide the fate of illnesses and associated complications whereas food plays a critical role in deciding the extent of severity and the fate of illnesses and its associated complications mainly by effecting oxidative stress shifts in the body modulating genes networks operations’ regulation involving genomic and/or involving epigenetic driven pathways by effecting homeostasis. The effect of the therapies can be modulated by type of processed or un-processed food intake. This piece of work reveals the missing understanding in the treatment strategies that are being used to treat most of the non-contagious diseases, including cancer. For this reason, patients are being dying not of diseases but due to inappropriate approach of therapies and food recommendations and complications associated with the therapies and their management. This paper also provides some understanding on current cancer therapy strategies responsible for intensifying the cancer illness and its associated consequences.

This paper is based on current understanding of knowledge, revealing the underlying molecular mechanisms responsible for causing illnesses as well as for deciding fate of illnesses and associated complications sharing the same interconnected genomics and/or involving epigenetic driven molecular pathways in addition to discussing some appropriate universal food combination models’ features constituting the food therapy that can be helpful to modulate the outcome of other therapies particularly cancer therapies leading to complete cure.

 Keywords: Food; Modulation of Therapies; Chemotherapy; Genes Networks Operations’ Regulation; Non-Contagious Diseases; Food Therapy; Desi Food; Epigenetics; Universal Evolutionary Capacitor; Consumers’ Safety; Natural Health; Stress Chemistry; Knowledge Gap

  1. Sensoy I. “A review on the food digestion in the digestive tract and the used in vitro models”. Current Research in Food Science 4 (2021): 308-319.
  2. MacDonald L., et al. “Food and therapeutic product interactions - a therapeutic perspective”. Journal of Pharmacy and Pharmaceutical Sciences3 (2009): 367-377.
  3. Abdur Rab F and Hassan A. “Tourism, health promoting food domain and technology applications: individual’s genes reservoir, environmental change and food in natural health context”. In: Hassan, A. (eds) Handbook of Technology Application in Tourism in Asia. Springer, Singapore (2022): 1159-1200.
  4. Rab FA. “Drug-disease relationship and role of the food in healthy living”. EC Nutrition8 (2018): 543-548.
  5. Rab FA. “Genome-nutrifortified diets-their disease protection and remedy potential”. Journal of Probiotics and Health 2 (2018): 204.
  6. Rab FA. “Food items biologically tailored to meet nutritional deficiency challenge during Covid 19 Pandemic”. Journal of Probiotics and Health 9 (2021): 233.
  7. Bryant DM and Mostov KE. “From cells to organs: building polarized tissue”. Nature Reviews. Molecular Cell Biology11 (2008): 887-901.
  8. Alberts B., et al. “Molecular biology of the cell”. 4th New York: Garland Science. From DNA to RNA (2002).
  9. Bishop AL., et al. “Phenotypic heterogeneity can enhance rare-cell survival in 'stress-sensitive' yeast populations”. Molecular Microbiology2 (2007): 507-520.
  10. Faiza Abdur Rab. Research thesis titled Phenotypic variation in stress resistance between individual cells in isogenic populations of Saccharomyces cerevisiae was submitted at University of Nottingham United Kingdom which was funded by National Institute of Health (NIH) US Department of Health and Human Services whereas Full PhD Tuition Fees was supported by Developing Solution PhD Tuition Fees Scholarship 2003 offered by University of Nottingham, United Kingdom and boarding and lodging was supported for two years only by University of Karachi Pakistan s Overseas PhD Scholarship Scheme 2003 (2007).
  11. Schlessinger A., et al. “Protein disorder--a breakthrough invention of evolution?”. Current Opinion in Structural Biology3 (2011): 412-418.
  12. Jeansonne NE. “Yeast as a model system for mammalian seven-transmembrane segment receptors”. Proceedings of the Society for Experimental Biology and Medicine. Society for Experimental Biology and Medicine (New York) N.Y.1 (1994): 35-44.
  13. Petranovic D., et al. “Prospects of yeast systems biology for human health: integrating lipid, protein and energy metabolism”. FEMS Yeast Research8 (2010): 1046-1059.
  14. Vanderwaeren L., et al. “Saccharomyces cerevisiae as a model system for eukaryotic cell biology, from cell cycle control to DNA damage response”. International Journal of Molecular Sciences 19 (2022): 11665.
  15. Rab FA. “Environmentally modulated evolution through genetic regulation”. Information systems for biotechnology ISB report Virginia Tech (2014).
  16. Sumner ER., et al. “Cell cycle- and age-dependent activation of Sod1p drives the formation of stress resistant cell subpopulations within clonal yeast cultures”. Molecular Microbiology3 (2003): 857-870.
  17. Lim L and Song J. “A novel SOD1-dependent mechanism for the iron-induced production of toxic SOD1 and oxidative stress that initiates ALS”. bioRxiv (2015).
  18. De Freitas JM., et al. “Yeast lacking Cu-Zn superoxide dismutase show altered iron homeostasis. Role of oxidative stress in iron metabolism”. The Journal of Biological Chemistry16 (2000): 11645-11649.
  19. King JL and Jukes TH. “Non-Darwinian evolution”. Science (New York N.Y.)3881 (1969): 788-798.
  20. Palazzo AF and Kejiou NS. “Non-Darwinian molecular biology”. Frontiers in Genetics 13 (2022): 831068.
  21. Ubeda F and Wilkins JF. “Imprinted genes and human disease: an evolutionary perspective”. Advances in Experimental Medicine and Biology 626 (2008): 101-115.
  22. Bergman A and Siegal ML. “Evolutionary capacitance as a general feature of complex gene networks”. Nature 6948 (2003): 549-552.
  23. Levy SF and Siegal ML. “Network hubs buffer environmental variation in Saccharomyces cerevisiae”. PLoS Biology11 (2008): e264.
  24. Masel J and Siegal ML. “Robustness: mechanisms and consequences”. Trends in Genetics: TIG9 (2009): 395-403.
  25. Che M., et al. “Expanding roles of superoxide dismutases in cell regulation and cancer”. Drug Discovery Today1 (2016): 143-149.
  26. Reddi AR and Culotta VC. “SOD1 integrates signals from oxygen and glucose to repress respiration”. Cell 1-2 (2013): 224-235.
  27. Eleutherio ECA., et al. “SOD1, more than just an antioxidant”. Archives of Biochemistry and Biophysics 697 (2021): 108701.
  28. Picazo C., et al. “Regulation of metabolism, stress response, and sod1 activity by cytosolic thioredoxins in yeast depends on growth phase”. Advances in Redox Research 9 (2023): 100081.
  29. Xu J., et al. “Nuclear SOD1 in growth control, oxidative stress response, amyotrophic lateral sclerosis, and cancer”. Antioxidants (Basel, Switzerland)2 (2022): 427.
  30. Park JH., et al. “SOD1 deficiency: a novel syndrome distinct from amyotrophic lateral sclerosis”. Brain: A Journal of Neurology8 (2019): 2230-2237.
  31. Liochev SI and Fridovich I. “CO2 enhanced peroxidase activity of SOD1: the effects of pH”. Free Radical Biology and Medicine11 (2004): 1444-1447.
  32. Ranguelova K., et al. “Kinetics of the oxidation of reduced Cu,Zn-superoxide dismutase by peroxymonocarbonate”. Free Radical Biology and Medicine3 (2012): 589-594.
  33. Zhao H., et al. “Dynamic imaging of cellular pH and redox homeostasis with a genetically encoded dual-functional biosensor, pHaROS, in yeast”. The Journal of Biological Chemistry43 (2019): 15768-15780.
  34. Sumner ER and Avery SV. “Phenotypic heterogeneity: differential stress resistance among individual cells of yeast Saccharomyces cerevisiae”. Microbiology 2 (2002): 345-351.
  35. Smith MC., et al. “Glutathione and Gts1p drive beneficial variability in the cadmium resistances of individual yeast cells”. Molecular Microbiology 3 (2007): 699-712.
  36. Sipos H., et al. “Impaired regulation of pH homeostasis by oxidative stress in rat brain capillary endothelial cells”. Cellular and Molecular Neurobiology 1 (2005): 141-151.
  37. Rab FA. “Is sugar a necessary or an accessory”. EC Nutrition4 (2018): 236-237.
  38. Rab FA. “Hurdles in progression of knowledge and its global impact”. Global Journal of Research and Review 1 (2020): 47.
  39. Rab FA. “Halal or Haram-new challenges for religious scholars Muslim world and food supply chain stake holders”. International Journal of Nutritional Science and Food Technology 6 (2020): 3.
  40. Vazquez-Jienez A., et al. “Characterization of intrinsic and extrinsic noise effects in positively regulated genes”. Journal of Biological Systems3 (2019): 1-16.
  41. Lindemann K. “Most of the human genome isn’t being actively studied” (2018).
  42. Rabilloud T., et al. “Oxidative stress response: a proteomic view”. Expert Review of Proteomics 6 (2005): 949-956.
  43. Ryter SW., et al. “Mechanisms of cell death in oxidative stress”. Antioxidants and Redox Signaling 1 (2007): 49-89.
  44. Santos LC., et al. “Mitochondrial origins of fractional control in regulated cell death”. Nature Communications 1 (2019): 1313.
  45. Joseph PV., et al. “Emerging role of nutri-epigenetics in inflammation and cancer”. Oncology Nursing Forum6 (2016): 784-788.
  46. Orij R., et al. “Genome-wide analysis of intracellular pH reveals quantitative control of cell division rate by pH(c) in Saccharomyces cerevisiae”. Genome Biology9 (2012): R80.
  47. Parsons LB. “Multiclonal tumor origin: Evidence and implications”. Mutation Research/Reviews in Mutation Research 777 (2018): 1-18.
  48. Wang X., et al. “Sodium oligomannate therapeutically remodels gut microbiota and suppresses gut bacterial amino acids-shaped neuroinflammation to inhibit Alzheimer’s disease progression”. Cell Research 10 (2019): 787-803.
  49. Coskun P., et al. “Metabolic and growth rate alterations in lymphoblastic cell lines discriminate between down syndrome and Alzheimer’s disease”. Journal of Alzheimer's Disease 2 (2017): 737-748.
  50. Chiorcea-Paquim A., et al. “Electrochemistry of Alzheimer disease amyloid beta peptides”. Current Medicinal Chemistry33 (2018): 4066-4083.
  51. Mittal M., et al. “Reactive oxygen species in inflammation and tissue injury”. Antioxidants and Redox Signaling 7 (2014): 1126-1167.
  52. Lei Y., et al. “Redox regulation of inflammation: old elements, a new story”. Medicinal Research Reviews 2 (2015): 306-340.
  53. Zohoori FV. “Chapter 1: Nutrition and diet”. Monographs in Oral Science 28 (2020): 1-13.
  54. Larosa V and Remacle C. “Insights into the respiratory chain and oxidative stress”. Bioscience Reports5 (2018): BSR20171492.
  55. Furukawa Y and O'Halloran TV. “Posttranslational modifications in Cu,Zn-superoxide dismutase and mutations associated with amyotrophic lateral sclerosis”. Antioxidants and Redox Signaling5-6 (2006): 847-867.
  56. Brown NM., et al. “Oxygen and the copper chaperone CCS regulate posttranslational activation of Cu, Zn superoxide dismutase”. Proceedings of the National Academy of Sciences of the United States of America15 (2004): 5518-5523.
  57. Niesor EJ., et al. “Red blood cell membrane cholesterol may be a key regulator of sickle cell disease microvascular complications”. Membranes11 (2022): 1134.
  58. Buchwald H., et al. “Plasma cholesterol: an influencing factor in red blood cell oxygen release and cellular oxygen availability”. Journal of the American College of Surgeons 5 (2000): 490-497.
  59. Nitin S. “HbA1c and factors other than diabetes mellitus affecting it”. Singapore Medical Journal8 (2010): 616-622.
  60. Anastasiadi AT., et al. “Molecular modifications to mitigate oxidative stress and improve red blood cell storability”. Frontiers in Physiology 15 (2024): 1499308.
  61. Rifkind JM., et al. “The pathophysiology of extracellular hemoglobin associated with enhanced oxidative reactions”. Frontiers in Physiology 5 (2015): 500.
  62. Obeagu EI., et al. “Oxidative stress's impact on red blood cells: Unveiling implications for health and disease”. Medicine9 (2024): e37360.
  63. Merino B., et al. “Intestinal fructose and glucose metabolism in health and disease”. Nutrients1 (2019): 94.
  64. Huttunen JK. “Fructose in medicine. A review with particular reference to diabetes mellitus”. Postgraduate Medical Journal552 (1971): 654-659.
  65. Chen X., et al. “Fructose metabolism in cancer: Molecular mechanisms and therapeutic implications”. International Journal of Medical Sciences11 (2025): 2852-2876.
  66. Bollig-Fischer A., et al. “Oncogene activation induces metabolic transformation resulting in insulin-independence in human breast cancer cells”. PloS one3 (2011): e17959.
  67. Petersen MC and Shulman GI. “Mechanisms of insulin action and insulin resistance”. Physiological Reviews4 (2018): 2133-2223.
  68. Saltiel AR. “Insulin signaling in health and disease”. The Journal of Clinical Investigation1 (2021): e142241.
  69. Douard V and Ferraris RP. “Regulation of the fructose transporter GLUT5 in health and disease”. American Journal of Physiology. Endocrinology and Metabolism2 (2008): E227-E237.
  70. Geidl-Flueck B and Gerber PA. “Insights into the hexose liver metabolism-glucose versus fructose”. Nutrients 9 (2017): 1026.
  71. Sato T., et al. “Acute fructose intake suppresses fasting-induced hepatic gluconeogenesis through the AKT-FoxO1 pathway”. Biochemistry and Biophysics Reports 18 (2019): 100638.
  72. Liu R., et al. “Oxidative stress in cancer immunotherapy: molecular mechanisms and potential applications”. Antioxidants (Basel) Switzerland5 (2022): 853.
  73. Ojha A., et al. “Comparative study of oxidative stress in cancer patients occupationally exposed to the mixture of pesticides”. Discover Oncology1 (2024): 526.
  74. Conklin KA. “Chemotherapy-associated oxidative stress: impact on chemotherapeutic effectiveness”. Integrative Cancer Therapies4 (2004): 294-300.
  75. Zhang J., et al. “Oxidative stress response induced by chemotherapy in leukemia treatment”. Molecular and Clinical Oncology3 (2018): 391-399.
  76. Druzhkova I., et al. “Tracing of intracellular pH in cancer cells in response to Taxol treatment”. Cell Cycle 16 (2021): 1540-1551.
  77. Shirmanova MV., et al. “Chemotherapy with cisplatin: insights into intracellular pH and metabolic landscape of cancer cells in vitro and in vivo”. Scientific Reports1 (2017): 8911.
  78. Künili İE., et al. “Bioactive compounds in fermented foods: a systematic narrative review”. Frontiers in Nutrition 12 (2025): 1625816.
  79. Leeuwendaal NK., et al. “Fermented foods, health and the gut microbiome”. Nutrients 7 (2022): 1527.
  80. Zhang P. “Influence of foods and nutrition on the gut microbiome and implications for intestinal health”. International Journal of Molecular Sciences17 (2022): 9588.
  81. Gamrath L., et al. “Role of the microbiome and diet for response to cancer checkpoint immunotherapy: a narrative review of clinical trials”. Current Oncology Reports1 (2025): 45-58.
  82. Gulliver EL., et al. “Review article: the future of microbiome-based therapeutics”. Alimentary Pharmacology and Therapeutics 2 (2022): 192-208.
  83. Kunst C., et al. “The influence of gut microbiota on oxidative stress and the immune system”. Biomedicines5 (2023): 1388.
  84. Lee JY., et al. “The microbiome and gut homeostasis”. Science (New York, N.Y.)6601 (2022): eabp9960.
  85. Lee JY., et al. “The human gut microbiome in health and disease: time for a new chapter?”. Infection and Immunity11 (2024): e0030224.
  86. Gupte A and Mumper RJ. “Elevated copper and oxidative stress in cancer cells as a target for cancer treatment”. Cancer Treatment Reviews1 (2009): 32-46.
  87. Arfin S., et al. “Oxidative stress in cancer cell metabolism”. Antioxidants (Basel, Switzerland)5 (2021): 642.
  88. Brown JS., et al. “Updating the definition of cancer”. Molecular Cancer Research: MCR11 (2023): 1142-1147.
  89. Cooper GM. “The cell: A molecular approach”. 2nd Sunderland (MA): Sinauer Associates; The Development and Causes of Cancer (2000).
  90. Shipitsin M and Polyak K. “The cancer stem cell hypothesis: in search of definitions, markers, and relevance”. Laboratory Investigation; A Journal of Technical Methods and Pathology5 (2008): 459-463.
  91. Iqbal MJ., et al. “Interplay of oxidative stress, cellular communication and signaling pathways in cancer”. Cell Communication and Signaling: CCS1 (2024): 7.
  92. Aboelella NS., et al. “Oxidative stress in the tumor microenvironment and its relevance to cancer immunotherapy”. Cancers 5 (2021): 986.
  93. Salnikow K. “Role of iron in cancer”. Seminars in Cancer Biology 76 (2021): 189-194.
  94. Xi Y., et al. “Mechanisms of induction of tumors by cholesterol and potential therapeutic prospects”. Biomedicine and Pharmacotherapy = Biomedecine and Pharmacotherapie 144 (2021): 112277.
  95. Saleh EAM., et al. “Oxidative stress affects the beginning of the growth of cancer cells through a variety of routes”. Pathology, Research and Practice 249 (2023): 154664.
  96. Noh J., et al. “Amplification of oxidative stress by a dual stimuli-responsive hybrid drug enhances cancer cell death”. Nature Communications 6 (2015): 6907.
  97. Hayes JD., et al. “Oxidative stress in cancer”. Cancer Cell2 (2020): 167-197.
  98. Zhao Q., et al. “AKR1B1-dependent fructose metabolism enhances malignancy of cancer cells”. Cell Death and Differentiation12 (2024): 1611-1624.
  99. Ting KKY. “Fructose-induced metabolic reprogramming of cancer cells”. Frontiers in Immunology 15 (2024): 1375461.
  100. Nakagawa T., et al. “Fructose contributes to the Warburg effect for cancer growth”. Cancer and Metabolism 8 (2020): 16.
  101. Mao Y., et al. “Metabolic reprogramming, sensing, and cancer therapy”. Cell Reports12 (2024): 115064.
  102. Koltai T and Fliegel L. “Fructose, another sweet for cancer: a context acting nutrient hypothesis”. Gene Expression 2 (2023): 141-155.
  103. Chen HW., et al. “Inhibition of cell growth by oxygenated derivatives of cholesterol”. Nature5474 (1974): 419-421.
  104. Stanhope KL and Havel PJ. “Fructose consumption: potential mechanisms for its effects to increase visceral adiposity and induce dyslipidemia and insulin resistance”. Current Opinion in Lipidology1 (2008): 16-24.
  105. Menezes-Santos M., et al. “Copper deficiency associated with glycemic control in individuals with type 2 diabetes mellitus”. Biological Trace Element Research1 (2025): 119-126.
  106. Odom H. “Copper deficiency reduces insulin receptor and AKT activation in house hepatocytes”. Undergraduate Honors Thesis. University of Nebraska - Lincoln (2024).
  107. Tan PY and Soma Roy, M. “Dietary copper and selenium are associated with insulin resistance in overweight and obese Malaysian adults”. Nutrition Research (New York, N.Y.) 93 (2021): 38-47.
  108. Kuang H., et al. “The impact of egg nutrient composition and its consumption on cholesterol homeostasis”. Cholesterol (2018): 6303810.
  109. Skrivan M., et al. “Effect of various copper supplements to feed of laying hens on cu content in eggs, liver, excreta, soil, and herbage”. Archives of Environmental Contamination and Toxicology2 (2006): 280-283.
  110. Ward C., et al. “The impact of tumour pH on cancer progression: strategies for clinical intervention”. Exploration of Targeted Anti-Tumor Therapy2 (2020): 71-100.
  111. Hao G., et al. “Manipulating extracellular tumour pH: an effective target for cancer therapy”. RSC Advances39 (2018): 22182-22192.
  112. Koltai T. “Cancer: fundamentals behind pH targeting and the double-edged approach”. OncoTargets and Therapy 9 (2016): 6343-6360.
  113. Aredia F and Scovassi AI. “Manipulation of intracellular pH in cancer cells by NHE1 inhibitors”. Protein and Peptide Letters 12 (2016): 1123-1129.
  114. Lee S and Shanti A. “Effect of exogenous pH on cell growth of breast cancer cells”. International Journal of Molecular Sciences18 (2021): 9910.
  115. White KA., et al. “Cancer cell behaviors mediated by dysregulated pH dynamics at a glance”. Journal of Cell Science4 (2017): 663-669.
  116. Tafech A and Stéphanou A. “On the importance of acidity in cancer cells and therapy”. Biology4 (2024): 225.
  117. Damaghi M., et al. “pH sensing and regulation in cancer”. Frontiers in Physiology 4 (2013): 370.
  118. Olżyńska A., et al. “Tail-oxidized cholesterol enhances membrane permeability for small solutes”. Langmuir: The ACS Journal of Surfaces and Colloids 35 (2020): 10438-10447.
  119. Yang Y-T., et al. “Characterization of cholesterol-depleted or -restored cell membranes by depth-sensing nano-indentation”. Soft Matter 8 (2011): 682-687.
  120. Giraldo-Lorza JM., et al. “The influence of cholesterol on membrane targeted bioactive peptides: modulating peptide activity through changes in bilayer biophysical properties”. Membranes10 (2024): 220.
  121. Kulig W., et al. “Oxidation of cholesterol changes the permeability of lipid membranes”. Biophysical Journal3 (2017): 377a.
  122. Zipser B., et al. “Cholesterol and its derivatives, are the principal steroids isolated from the leech species Hirudo medicinalis”. Comparative Biochemistry and Physiology. Part C, Pharmacology, Toxicology and Endocrinology2 (1998): 269-282.
  123. Liu S., et al. “SOD1 promotes cell proliferation and metastasis in non-small cell lung cancer via an miR-409-3p/SOD1/SETDB1 epigenetic regulatory feedforward loop”. Frontiers in Cell and Developmental Biology 8 (2020): 213.
  124. König S., et al. “Superoxide dismutase 1 mediates adaptation to the tumor microenvironment of glioma cells via mammalian target of rapamycin complex”. Cell Death Discovery1 (2024): 379.
  125. Harris N., et al. “Overexpressed Sod1p acts either to reduce or to increase the lifespans and stress resistance of yeast, depending on whether it is Cu(2+)-deficient or an active Cu, Zn-superoxide dismutase”. Aging Cell1 (2005): 41-52.
  126. Magrì A., et al. “Overexpression of human SOD1 in VDAC1-less yeast restores mitochondrial functionality modulating beta-barrel outer membrane protein genes”. Biochimica et Biophysica Acta 6 (2016): 789-798.
  127. Wawryn J., et al. “Deficiency in superoxide dismutases shortens life span of yeast cells”. Acta Biochimica Polonica2 (1999): 249-253.
  128. Tasić D., et al. “Effects of fructose and stress on rat renal copper metabolism and antioxidant enzymes function”. International Journal of Molecular Sciences16 (2022): 9023.
  129. Klevay LM. “Iron overload can induce mild copper deficiency”. Journal of Trace Elements in Medicine and Biology: Organ of the Society for Minerals and Trace Elements (GMS)4 (2001): 237-240.
  130. Fisher AL., et al. “Iron loading induces cholesterol synthesis and sensitizes endothelial cells to TNFα-mediated apoptosis”. The Journal of Biological Chemistry4 (2021): 101156.
  131. Lee J., et al. “High iron consumption modifies the hepatic transcriptome related to cholesterol metabolism”. Journal of Medicinal Food9 (2024): 895-900.
  132. Fouani L., et al. “Metals and metastasis: Exploiting the role of metals in cancer metastasis to develop novel anti-metastatic agents”. Pharmacological Research 115 (2017): 275-287.
  133. Shan D., et al. “Copper in cancer: friend or foe? Metabolism, dysregulation, and therapeutic opportunities”. Cancer Communications (London, England) 5 (2025): 577-607.
  134. Kamiya T. “Copper in the tumor microenvironment and tumor metastasis”. Journal of Clinical Biochemistry and Nutrition 1 (2022): 22-28.
  135. Xiao M., et al. “Functional significance of cholesterol metabolism in cancer: from threat to treatment”. Experimental and Molecular Medicine9 (2023): 1982-1995.
  136. Ding X., et al. “The role of cholesterol metabolism in cancer”. American Journal of Cancer Research2 (2019): 219-227.
  137. Hordyjewska A., et al. “The many "faces" of copper in medicine and treatment”. Biometals: An International Journal on the Role of Metal Ions in Biology, Biochemistry, and Medicine4 (2014): 611-621.
  138. Ikonen E and Zhou X. “Cholesterol transport between cellular membranes: A balancing act between interconnected lipid fluxes”. Developmental Cell10 (2021): 1430-1436.
  139. Chen Y., et al. “Regulation of intracellular cholesterol distribution by Na/K-ATPase”. The Journal of Biological Chemistry22 (2009): 14881-14890.
  140. Le Grimellec C and Leblanc G. “Effect of membrane cholesterol on potassium transport in Mycoplasma mycoides var. Capri (PG3)”. Biochimica et Biophysica Acta1 (1978): 152-163.
  141. Franco R., et al. “Glutathione depletion is necessary for apoptosis in lymphoid cells independent of reactive oxygen species formation”. The Journal of Biological Chemistry42 (2007): 30452-30465.
  142. Hatori Y., et al. “Functional partnership of the copper export machinery and glutathione balance in human cells”. The Journal of Biological Chemistry32 (2012): 26678-26687.
  143. Ghibelli L., et al. “Non-oxidative loss of glutathione in apoptosis via GSH extrusion”. Biochemical and Biophysical Research Communications 1 (1995): 313-320.
  144. Boggs SE., et al. “Glutathione levels determine apoptosis in macrophages”. Biochemical and Biophysical Research Communications 2 (1998): 229-233.
  145. Circu ML and Aw TY. “Glutathione and apoptosis”. Free Radical Research8 (2008): 689-706.
  146. Maryon EB., et al. “Cellular glutathione plays a key role in copper uptake mediated by human copper transporter 1”. American Journal of Physiology. Cell Physiology8 (2013): C768-C779.
  147. Gustafsson J., et al. “Cholesterol synthesis in patients with glutathione deficiency”. European Journal of Clinical Investigation4 (1990): 470-474.
  148. Le Jeune N., et al. “Influence of glutathione depletion on plasma membrane cholesterol esterification and on Tc-99m-sestamibi and Tc-99m-tetrofosmin uptakes: a comparative study in sensitive U-87-MG and multidrug-resistant MRP1 human glioma cells”. Cancer Biotherapy and Radiopharmaceuticals4 (2004): 411-421.
  149. McCay PB., et al. “Glutathione-dependent inhibition of lipid peroxidation by a soluble, heat-labile factor not glutathione peroxidase”. Federation Proceedings2 (1981): 199-205.
  150. Lee JY., et al. “Lipid metabolism and ferroptosis”. Biology3 (2021): 184.
  151. Ursini F and Maiorino M. “Lipid peroxidation and ferroptosis: The role of GSH and GPx4”. Free Radical Biology and Medicine 152 (2020): 175-185.
  152. Ashkaran F., et al. “Mutation/metal deficiency in the "electrostatic loop" enhanced aggregation process in apo/holo SOD1 variants: implications for ALS diseases”. BMC Chemistry1 (2024): 177.
  153. Homma K., et al. “SOD1 as a molecular switch for initiating the homeostatic ER stress response under zinc deficiency”. Molecular Cell1 (2013): 75-86.
  154. Damiano S., et al. “Metabolism regulation and redox state: insight into the role of superoxide dismutase 1”. International Journal of Molecular Sciences18 (2020): 6606.
  155. Raha S and Robinson BH. “Mitochondria, oxygen free radicals, and apoptosis”. American Journal of Medical Genetics 1 (2001): 62-70.
  156. Azad N and Iyer AKV. “Reactive oxygen species and apoptosis”. In: Laher, I. (eds) Systems Biology of Free Radicals and Antioxidants. Springer, Berlin, Heidelberg (2014).
  157. Chen Q., et al. “The late increase in intracellular free radical oxygen species during apoptosis is associated with cytochrome c release, caspase activation, and mitochondrial dysfunction”. Cell Death and Differentiation3 (2003): 323-334.
  158. Li Y., et al. “The ferroptosis inhibitor liproxstatin-1 ameliorates LPS-induced cognitive impairment in mice”. Nutrients21 (2022): 4599.
  159. Cronin SJF., et al. “The role of iron regulation in immunometabolism and immune-related disease”. Frontiers in Molecular Biosciences 6 (2019): 116.
  160. Porto G and De Sousa M. “Iron overload and immunity”. World Journal of Gastroenterology35 (2007): 4707-4715.
  161. Obeagu EI. “Iron homeostasis and health: understanding its role beyond blood health - a narrative review”. Annals of Medicine and Surgery (2012)6 (2025): 3362-3371.
  162. Keiko M., et al. “Iron overload effects on immune system through the cytokine secretion by macrophage”. Blood 21 (2013): 1047.
  163. Walker EM Jr and Walker SM. “Effects of iron overload on the immune system”. Annals of Clinical and Laboratory Science4 (2000): 354-365.
  164. Acharya GK., et al. “Autoimmune hepatitis: Diagnostic dilemma when it is disguised as iron overload syndrome”. Journal of Clinical and Experimental Hepatology 3 (2017): 269-273.
  165. Falahatian M., et al. “Hereditary hemochromatosis associated with autoimmune hemolytic anemia; A case report”. Journal of Preventive Epidemiology 1 (2019): e02.
  166. Yazdali Koylu N., et al. “In the presence of autoantibodies and iron overload, do not judge a book by its cover: A case report”. Hepatology Forum2 (2021): 76-79.
  167. Zandman-Goddard G and Shoenfeld Y. “Ferritin in autoimmune diseases”. Autoimmunity Reviews7 (2007): 457-463.
  168. Fibach E and Rachmilewitz EA. “Iron overload in hematological disorders”. Presse Medicale (Paris, France: 1983) 12.2 (2017): e296-e305.
  169. Hsu CC., et al. “Iron overload disorders”. Hepatology Communications8 (2022): 1842-1854.
  170. Pang N., et al. “Iron overload causes macrophages to produce a pro-inflammatory phenotype in the synovium of hemophiliac arthritis via the acetyl-p53 pathway”. Haemophilia: The Official Journal of the World Federation of Hemophilia1 (2024): 195-203.
  171. Ghezzi P. “Role of glutathione in immunity and inflammation in the lung”. International Journal of General Medicine 4 (2011): 105-113.
  172. Tan M., et al. “Glutathione system enhancement for cardiac protection: pharmacological options against oxidative stress and ferroptosis”. Cell Death and Disease2 (2023): 131.
  173. Rajasekaran NS., et al. “Chronic depletion of glutathione (GSH) and minimal modification of LDL in vivo: its prevention by glutathione mono ester (GME) therapy”. Biochimica et Biophysica Acta1-2 (2005): 103-112.
  174. Yang X., et al. “Inhibition of glutathione production induces macrophage CD36 expression and enhances cellular-oxidized low density lipoprotein (oxLDL) uptake”. The Journal of Biological Chemistry36 (2015): 21788-21799.
  175. Saito Y., et al. “Cholesterol is more readily oxidized than phospholipid linoleates in cell membranes to produce cholesterol hydroperoxides”. Free Radical Biology and Medicine 211 (2024): 89-95.
  176. Lushchak VI. “Glutathione homeostasis and functions: potential targets for medical interventions”. Journal of Amino Acids (2012): 736837.
  177. Catalgol B and Kartal Ozer N. “Lipid rafts and redox regulation of cellular signaling in cholesterol induced atherosclerosis”. Current Cardiology Reviews4 (2010): 309-324.
  178. Matuz-Mares D., et al. “Glutathione participation in the prevention of cardiovascular diseases”. Antioxidants (Basel, Switzerland) 8 (2021): 1220.
  179. Shan XQ., et al. “Glutathione-dependent protection against oxidative injury”. Pharmacology and Therapeutics1 (1990): 61-71.
  180. Averill-Bates DA. “The antioxidant glutathione”. Vitamins and Hormones 121 (2023): 109-141.
  181. Lee LR., et al. “Glutathione accelerates the cell cycle and cellular reprogramming in plant regeneration”. bioRxiv (2024).
  182. Diaz Vivancos P., et al. “A nuclear glutathione cycle within the cell cycle”. The Biochemical Journal2 (2010): 169-178.
  183. Pallardó FV., et al. “Role of nuclear glutathione as a key regulator of cell proliferation”. Molecular Aspects of Medicine 1-2 (2009): 77-85.
  184. Ayala A., et al. “Lipid peroxidation: production, metabolism, and signaling mechanisms of malondialdehyde and 4-hydroxy-2-nonenal”. Oxidative Medicine and Cellular Longevity (2014): 360438.
  185. Benedusi V., et al. “The peroxisome proliferator-activated receptor γ (PPARγ) controls natural protective mechanisms against lipid peroxidation in amyotrophic lateral sclerosis”. The Journal of Biological Chemistry43 (2012): 35899-35911.
  186. Nam TG. “Lipid peroxidation and its toxicological implications”. Toxicological Research1 (2011): 1-6.
  187. Li Pomi F., et al. “Oxidative stress and skin diseases: The role of lipid peroxidation”. Antioxidants5 (2025): 555.
  188. Lee SH., et al. “Lipid peroxidation-derived modification and its effect on the activity of glutathione peroxidase 1”. Free Radical Biology and Medicine 208 (2023): 252-259.
  189. Nardella MI., et al. “Oxidation of human copper chaperone atox1 and disulfide bond cleavage by cisplatin and glutathione”. International Journal of Molecular Sciences18 (2019): 4390.
  190. Guan D., et al. “Copper in cancer: From pathogenesis to therapy”. Biomedicine and Pharmacotherapy = Biomedecine and Pharmacotherapie 163 (2023): 114791.
  191. Juarez JC., et al. “Copper binding by tetrathiomolybdate attenuates angiogenesis and tumor cell proliferation through the inhibition of superoxide dismutase 1”. Clinical Cancer Research: An Official Journal of the American Association for Cancer Research16 (2006): 4974-4982.
  192. Tang X., et al. “Copper in cancer: from limiting nutrient to therapeutic target”. Frontiers in Oncology 13 (2023): 1209156.
  193. Badran O., et al. “The impact of iron on cancer-related immune functions in oncology: molecular mechanisms and clinical evidence”. Cancers 24 (2024): 4156.
  194. Shi Y., et al. “Effect of metal loading and subcellular pH on net charge of superoxide dismutase-1”. Journal of Molecular Biology22 (2013): 4388-4404.
  195. Byström R., et al. “SOD1 mutations targeting surface hydrogen bonds promote amyotrophic lateral sclerosis without reducing apo-state stability”. The Journal of Biological Chemistry25 (2010): 19544-19552.
  196. Perry JJ., et al. “The structural biochemistry of the superoxide dismutases”. Biochimica et Biophysica Acta2 (2010): 245-262.
  197. Sehati S., et al. “Metabolic alterations in yeast lacking copper-zinc superoxide dismutase”. Free Radical Biology and Medicine11 (2011): 1591-1598.
  198. Ansenberger-Fricano K., et al. “The peroxidase activity of mitochondrial superoxide dismutase”. Free Radical Biology and Medicine 54 (2013): 116-124.
  199. Arnesano F., et al. “The unusually stable quaternary structure of human Cu,Zn-superoxide dismutase 1 is controlled by both metal occupancy and disulfide status”. The Journal of Biological Chemistry 46 (2004): 47998-48003.
  200. Ramirez DC., et al. “Cu,Zn-superoxide dismutase-driven free radical modifications: copper- and carbonate radical anion-initiated protein radical chemistry”. The Biochemical Journal1 (2009): 341-353.
  201. Zhang H., et al. “Bicarbonate enhances peroxidase activity of Cu,Zn-superoxide dismutase. Role of carbonate anion radical and scavenging of carbonate anion radical by metalloporphyrin antioxidant enzyme mimetics”. The Journal of Biological Chemistry2 (2002): 1013-1020.
  202. Hink HU., et al. “Peroxidase properties of extracellular superoxide dismutase: role of uric acid in modulating in vivo activity”. Arteriosclerosis, Thrombosis, and Vascular Biology9 (2002): 1402-1408.
  203. Sankarapandi S and Zweier JL. “Bicarbonate is required for the peroxidase function of Cu, Zn-superoxide dismutase at physiological pH”. The Journal of Biological Chemistry3 (1999): 1226-1232.
  204. Li Y., et al. “Iron and copper: critical executioners of ferroptosis, cuproptosis and other forms of cell death”. Cell Communication and Signaling: CCS1 (2023): 327.
  205. Troost FJ., et al. “Iron supplements inhibit zinc but not copper absorption in vivo in ileostomy subjects”. The American Journal of Clinical Nutrition 5 (2003): 1018-1023.
  206. Aracena P., et al. “Iron and glutathione at the crossroad of redox metabolism in neurons”. Biological Research1 (2006): 157-165.
  207. Núñez MT., et al. “Progressive iron accumulation induces a biphasic change in the glutathione content of neuroblastoma cells”. Free Radical Biology and Medicine7 (2004): 953-960.
  208. Han P., et al. “Activation of chicken liver fructose-1,6-bisphosphatase by oxidized glutathione”. FEBS Letters2 (1986): 347-351.
  209. Halliwell B and Foyer CH. “Properties and physiological function of a glutathione reductase purified from spinach leaves by affinity chromatography”. Planta1 (1978): 9-17.
  210. Song M., et al. “High fructose feeding induces copper deficiency in Sprague-Dawley rats: a novel mechanism for obesity related fatty liver”. Journal of Hepatology2 (2012): 433-440.
  211. Moreno JA and Hong E. “A single oral dose of fructose induces some features of metabolic syndrome in rats: role of oxidative stress”. Nutrition, Metabolism, and Cardiovascular Diseases: NMCD6 (2013): 536-542.
  212. Silva JM., et al. “Prevention of nitrofurantoin-induced cytotoxicity in isolated hepatocytes by fructose”. Archives of Biochemistry and Biophysics 2 (1991): 313-318.
  213. Kulig W., et al. “Cholesterol oxidation products and their biological importance”. Chemistry and Physics of Lipids 199 (2016): 144-160.
  214. Kim JW., et al. “An integrated view of lipid metabolism in ferroptosis revisited via lipidomic analysis”. Experimental and Molecular Medicine8 (2023): 1620-1631.
  215. Chen X., et al. “Ferroptosis by Lipid Peroxidation: The Tip of the Iceberg?”. Frontiers in Cell and Developmental Biology 9 (2021): 646890.
  216. Li X., et al. “Iron accumulation and lipid peroxidation: implication of ferroptosis in hepatocellular carcinoma”. Frontiers in Endocrinology 14 (2024): 1319969.
  217. Yang WS and Stockwell BR. “Ferroptosis: Death by lipid peroxidation”. Trends in Cell Biology3 (2016): 165-176.
  218. Endale HT., et al. “ROS induced lipid peroxidation and their role in ferroptosis”. Frontiers in Cell and Developmental Biology 11 (2023): 1226044.
  219. Yang X., et al. “Ferroptosis as a new tool for tumor suppression through lipid peroxidation”. Communications Biology1 (2024): 1475.
  220. Conrad M., et al. “Regulation of lipid peroxidation and ferroptosis in diverse species”. Genes and Development 9-10 (2018): 602-619.
  221. Coradduzza D., et al. “Ferroptosis and senescence: A systematic review”. International Journal of Molecular Sciences4 (2023): 3658.
  222. Cheng Z., et al. “Ferroptosis in non-alcoholic liver disease: Molecular mechanisms and therapeutic implications”. Frontiers in Nutrition 10 (2023): 1090338.
  223. Montllor-Albalate C., et al. “Sod1 integrates oxygen availability to redox regulate NADPH production and the thiol redoxome”. Proceedings of the National Academy of Sciences of the United States of America1 (2022): e2023328119.
  224. Patra KC and Hay N. “The pentose phosphate pathway and cancer”. Trends in Biochemical Sciences8 (2014): 347-354.
  225. Krause N and Wegner A. “Fructose metabolism in cancer”. Cells12 (2020): 2635.
  226. Chen H., et al. “Mechanisms and active substances of targeting lipid peroxidation in ferroptosis regulation”. Food Science and Human Wellness5 (2024): 2502-2518.
  227. Gao L., et al. “Bioinformatics analysis reveals SOD1 is a prognostic factor in lung adenocarcinoma”. Translational Cancer Research10 (2024): 5522-5534.
  228. Sangani RG and Ghio AJ. “Iron, human growth, and the global epidemic of obesity”. Nutrients10 (2013): 4231-4249.
  229. Manz DH., et al. “Iron and cancer: recent insights”. Annals of the New York Academy of Sciences1 (2016): 149-161.
  230. Islam S., et al. “Iron overload and breast cancer: iron chelation as a potential therapeutic approach”. Life (Basel, Switzerland)7 (2022): 963.
  231. Basak T and Kanwar RK. “Iron imbalance in cancer: Intersection of deficiency and overload”. Cancer Medicine20 (2022): 3837-3853.
  232. Ngamchuea K., et al. “The Copper(II)-Catalyzed Oxidation of Glutathione”. Chemistry (Weinheim an der Bergstrasse, Germany) 44 (2016): 15937-15944.
  233. Friesen C., et al. “A critical role of glutathione in determining apoptosis sensitivity and resistance in leukemia cells”. Cell Death and Differentiation 1 (2004): S73-S85.
  234. Cazanave S., et al. “High hepatic glutathione stores alleviate Fas-induced apoptosis in mice”. Journal of Hepatology5 (2007): 858-868.
  235. Armstrong JS., et al. “Role of glutathione depletion and reactive oxygen species generation in apoptotic signaling in a human B lymphoma cell line”. Cell Death and Differentiation3 (2002): 252-263.
  236. Sensoy I. “A review on the food digestion in the digestive tract and the used in vitro models”. Current Research in Food Science 4 (2021): 308-319.
  237. MacDonald L., et al. “Food and therapeutic product interactions - a therapeutic perspective”. Journal of Pharmacy and Pharmaceutical Sciences3 (2009): 367-377.
  238. Abdur Rab F and Hassan A. “Tourism, health promoting food domain and technology applications: individual’s genes reservoir, environmental change and food in natural health context”. In: Hassan, A. (eds) Handbook of Technology Application in Tourism in Asia. Springer, Singapore (2022): 1159-1200.
  239. Rab FA. “Drug-disease relationship and role of the food in healthy living”. EC Nutrition8 (2018): 543-548.
  240. Rab FA. “Genome-nutrifortified diets-their disease protection and remedy potential”. Journal of Probiotics and Health 2 (2018): 204.
  241. Rab FA. “Food items biologically tailored to meet nutritional deficiency challenge during Covid 19 Pandemic”. Journal of Probiotics and Health 9 (2021): 233.
  242. Bryant DM and Mostov KE. “From cells to organs: building polarized tissue”. Nature Reviews. Molecular Cell Biology11 (2008): 887-901.
  243. Alberts B., et al. “Molecular biology of the cell”. 4th New York: Garland Science. From DNA to RNA (2002).
  244. Bishop AL., et al. “Phenotypic heterogeneity can enhance rare-cell survival in 'stress-sensitive' yeast populations”. Molecular Microbiology2 (2007): 507-520.
  245. Faiza Abdur Rab. Research thesis titled Phenotypic variation in stress resistance between individual cells in isogenic populations of Saccharomyces cerevisiae was submitted at University of Nottingham United Kingdom which was funded by National Institute of Health (NIH) US Department of Health and Human Services whereas Full PhD Tuition Fees was supported by Developing Solution PhD Tuition Fees Scholarship 2003 offered by University of Nottingham, United Kingdom and boarding and lodging was supported for two years only by University of Karachi Pakistan s Overseas PhD Scholarship Scheme 2003 (2007).
  246. Schlessinger A., et al. “Protein disorder--a breakthrough invention of evolution?”. Current Opinion in Structural Biology3 (2011): 412-418.
  247. Jeansonne NE. “Yeast as a model system for mammalian seven-transmembrane segment receptors”. Proceedings of the Society for Experimental Biology and Medicine. Society for Experimental Biology and Medicine (New York) N.Y.1 (1994): 35-44.
  248. Petranovic D., et al. “Prospects of yeast systems biology for human health: integrating lipid, protein and energy metabolism”. FEMS Yeast Research8 (2010): 1046-1059.
  249. Vanderwaeren L., et al. “Saccharomyces cerevisiae as a model system for eukaryotic cell biology, from cell cycle control to DNA damage response”. International Journal of Molecular Sciences 19 (2022): 11665.
  250. Rab FA. “Environmentally modulated evolution through genetic regulation”. Information systems for biotechnology ISB report Virginia Tech (2014).
  251. Sumner ER., et al. “Cell cycle- and age-dependent activation of Sod1p drives the formation of stress resistant cell subpopulations within clonal yeast cultures”. Molecular Microbiology3 (2003): 857-870.
  252. Lim L and Song J. “A novel SOD1-dependent mechanism for the iron-induced production of toxic SOD1 and oxidative stress that initiates ALS”. bioRxiv (2015).
  253. De Freitas JM., et al. “Yeast lacking Cu-Zn superoxide dismutase show altered iron homeostasis. Role of oxidative stress in iron metabolism”. The Journal of Biological Chemistry16 (2000): 11645-11649.
  254. King JL and Jukes TH. “Non-Darwinian evolution”. Science (New York N.Y.)3881 (1969): 788-798.
  255. Palazzo AF and Kejiou NS. “Non-Darwinian molecular biology”. Frontiers in Genetics 13 (2022): 831068.
  256. Ubeda F and Wilkins JF. “Imprinted genes and human disease: an evolutionary perspective”. Advances in Experimental Medicine and Biology 626 (2008): 101-115.
  257. Bergman A and Siegal ML. “Evolutionary capacitance as a general feature of complex gene networks”. Nature 6948 (2003): 549-552.
  258. Levy SF and Siegal ML. “Network hubs buffer environmental variation in Saccharomyces cerevisiae”. PLoS Biology11 (2008): e264.
  259. Masel J and Siegal ML. “Robustness: mechanisms and consequences”. Trends in Genetics: TIG9 (2009): 395-403.
  260. Che M., et al. “Expanding roles of superoxide dismutases in cell regulation and cancer”. Drug Discovery Today1 (2016): 143-149.
  261. Reddi AR and Culotta VC. “SOD1 integrates signals from oxygen and glucose to repress respiration”. Cell 1-2 (2013): 224-235.
  262. Eleutherio ECA., et al. “SOD1, more than just an antioxidant”. Archives of Biochemistry and Biophysics 697 (2021): 108701.
  263. Picazo C., et al. “Regulation of metabolism, stress response, and sod1 activity by cytosolic thioredoxins in yeast depends on growth phase”. Advances in Redox Research 9 (2023): 100081.
  264. Xu J., et al. “Nuclear SOD1 in growth control, oxidative stress response, amyotrophic lateral sclerosis, and cancer”. Antioxidants (Basel, Switzerland)2 (2022): 427.
  265. Park JH., et al. “SOD1 deficiency: a novel syndrome distinct from amyotrophic lateral sclerosis”. Brain: A Journal of Neurology8 (2019): 2230-2237.
  266. Liochev SI and Fridovich I. “CO2 enhanced peroxidase activity of SOD1: the effects of pH”. Free Radical Biology and Medicine11 (2004): 1444-1447.
  267. Ranguelova K., et al. “Kinetics of the oxidation of reduced Cu,Zn-superoxide dismutase by peroxymonocarbonate”. Free Radical Biology and Medicine3 (2012): 589-594.
  268. Zhao H., et al. “Dynamic imaging of cellular pH and redox homeostasis with a genetically encoded dual-functional biosensor, pHaROS, in yeast”. The Journal of Biological Chemistry43 (2019): 15768-15780.
  269. Sumner ER and Avery SV. “Phenotypic heterogeneity: differential stress resistance among individual cells of yeast Saccharomyces cerevisiae”. Microbiology 2 (2002): 345-351.
  270. Smith MC., et al. “Glutathione and Gts1p drive beneficial variability in the cadmium resistances of individual yeast cells”. Molecular Microbiology 3 (2007): 699-712.
  271. Sipos H., et al. “Impaired regulation of pH homeostasis by oxidative stress in rat brain capillary endothelial cells”. Cellular and Molecular Neurobiology 1 (2005): 141-151.
  272. Rab FA. “Is sugar a necessary or an accessory”. EC Nutrition4 (2018): 236-237.
  273. Rab FA. “Hurdles in progression of knowledge and its global impact”. Global Journal of Research and Review 1 (2020): 47.
  274. Rab FA. “Halal or Haram-new challenges for religious scholars Muslim world and food supply chain stake holders”. International Journal of Nutritional Science and Food Technology 6 (2020): 3.
  275. Vazquez-Jienez A., et al. “Characterization of intrinsic and extrinsic noise effects in positively regulated genes”. Journal of Biological Systems3 (2019): 1-16.
  276. Lindemann K. “Most of the human genome isn’t being actively studied” (2018).
  277. Rabilloud T., et al. “Oxidative stress response: a proteomic view”. Expert Review of Proteomics 6 (2005): 949-956.
  278. Ryter SW., et al. “Mechanisms of cell death in oxidative stress”. Antioxidants and Redox Signaling 1 (2007): 49-89.
  279. Santos LC., et al. “Mitochondrial origins of fractional control in regulated cell death”. Nature Communications 1 (2019): 1313.
  280. Joseph PV., et al. “Emerging role of nutri-epigenetics in inflammation and cancer”. Oncology Nursing Forum6 (2016): 784-788.
  281. Orij R., et al. “Genome-wide analysis of intracellular pH reveals quantitative control of cell division rate by pH(c) in Saccharomyces cerevisiae”. Genome Biology9 (2012): R80.
  282. Parsons LB. “Multiclonal tumor origin: Evidence and implications”. Mutation Research/Reviews in Mutation Research 777 (2018): 1-18.
  283. Wang X., et al. “Sodium oligomannate therapeutically remodels gut microbiota and suppresses gut bacterial amino acids-shaped neuroinflammation to inhibit Alzheimer’s disease progression”. Cell Research 10 (2019): 787-803.
  284. Coskun P., et al. “Metabolic and growth rate alterations in lymphoblastic cell lines discriminate between down syndrome and Alzheimer’s disease”. Journal of Alzheimer's Disease 2 (2017): 737-748.
  285. Chiorcea-Paquim A., et al. “Electrochemistry of Alzheimer disease amyloid beta peptides”. Current Medicinal Chemistry33 (2018): 4066-4083.
  286. Mittal M., et al. “Reactive oxygen species in inflammation and tissue injury”. Antioxidants and Redox Signaling 7 (2014): 1126-1167.
  287. Lei Y., et al. “Redox regulation of inflammation: old elements, a new story”. Medicinal Research Reviews 2 (2015): 306-340.
  288. Zohoori FV. “Chapter 1: Nutrition and diet”. Monographs in Oral Science 28 (2020): 1-13.
  289. Larosa V and Remacle C. “Insights into the respiratory chain and oxidative stress”. Bioscience Reports5 (2018): BSR20171492.
  290. Furukawa Y and O'Halloran TV. “Posttranslational modifications in Cu,Zn-superoxide dismutase and mutations associated with amyotrophic lateral sclerosis”. Antioxidants and Redox Signaling5-6 (2006): 847-867.
  291. Brown NM., et al. “Oxygen and the copper chaperone CCS regulate posttranslational activation of Cu, Zn superoxide dismutase”. Proceedings of the National Academy of Sciences of the United States of America15 (2004): 5518-5523.
  292. Niesor EJ., et al. “Red blood cell membrane cholesterol may be a key regulator of sickle cell disease microvascular complications”. Membranes11 (2022): 1134.
  293. Buchwald H., et al. “Plasma cholesterol: an influencing factor in red blood cell oxygen release and cellular oxygen availability”. Journal of the American College of Surgeons 5 (2000): 490-497.
  294. Nitin S. “HbA1c and factors other than diabetes mellitus affecting it”. Singapore Medical Journal8 (2010): 616-622.
  295. Anastasiadi AT., et al. “Molecular modifications to mitigate oxidative stress and improve red blood cell storability”. Frontiers in Physiology 15 (2024): 1499308.
  296. Rifkind JM., et al. “The pathophysiology of extracellular hemoglobin associated with enhanced oxidative reactions”. Frontiers in Physiology 5 (2015): 500.
  297. Obeagu EI., et al. “Oxidative stress's impact on red blood cells: Unveiling implications for health and disease”. Medicine9 (2024): e37360.
  298. Merino B., et al. “Intestinal fructose and glucose metabolism in health and disease”. Nutrients1 (2019): 94.
  299. Huttunen JK. “Fructose in medicine. A review with particular reference to diabetes mellitus”. Postgraduate Medical Journal552 (1971): 654-659.
  300. Chen X., et al. “Fructose metabolism in cancer: Molecular mechanisms and therapeutic implications”. International Journal of Medical Sciences11 (2025): 2852-2876.
  301. Bollig-Fischer A., et al. “Oncogene activation induces metabolic transformation resulting in insulin-independence in human breast cancer cells”. PloS one3 (2011): e17959.
  302. Petersen MC and Shulman GI. “Mechanisms of insulin action and insulin resistance”. Physiological Reviews4 (2018): 2133-2223.
  303. Saltiel AR. “Insulin signaling in health and disease”. The Journal of Clinical Investigation1 (2021): e142241.
  304. Douard V and Ferraris RP. “Regulation of the fructose transporter GLUT5 in health and disease”. American Journal of Physiology. Endocrinology and Metabolism2 (2008): E227-E237.
  305. Geidl-Flueck B and Gerber PA. “Insights into the hexose liver metabolism-glucose versus fructose”. Nutrients 9 (2017): 1026.
  306. Sato T., et al. “Acute fructose intake suppresses fasting-induced hepatic gluconeogenesis through the AKT-FoxO1 pathway”. Biochemistry and Biophysics Reports 18 (2019): 100638.
  307. Liu R., et al. “Oxidative stress in cancer immunotherapy: molecular mechanisms and potential applications”. Antioxidants (Basel) Switzerland5 (2022): 853.
  308. Ojha A., et al. “Comparative study of oxidative stress in cancer patients occupationally exposed to the mixture of pesticides”. Discover Oncology1 (2024): 526.
  309. Conklin KA. “Chemotherapy-associated oxidative stress: impact on chemotherapeutic effectiveness”. Integrative Cancer Therapies4 (2004): 294-300.
  310. Zhang J., et al. “Oxidative stress response induced by chemotherapy in leukemia treatment”. Molecular and Clinical Oncology3 (2018): 391-399.
  311. Druzhkova I., et al. “Tracing of intracellular pH in cancer cells in response to Taxol treatment”. Cell Cycle 16 (2021): 1540-1551.
  312. Shirmanova MV., et al. “Chemotherapy with cisplatin: insights into intracellular pH and metabolic landscape of cancer cells in vitro and in vivo”. Scientific Reports1 (2017): 8911.
  313. Künili İE., et al. “Bioactive compounds in fermented foods: a systematic narrative review”. Frontiers in Nutrition 12 (2025): 1625816.
  314. Leeuwendaal NK., et al. “Fermented foods, health and the gut microbiome”. Nutrients 7 (2022): 1527.
  315. Zhang P. “Influence of foods and nutrition on the gut microbiome and implications for intestinal health”. International Journal of Molecular Sciences17 (2022): 9588.
  316. Gamrath L., et al. “Role of the microbiome and diet for response to cancer checkpoint immunotherapy: a narrative review of clinical trials”. Current Oncology Reports1 (2025): 45-58.
  317. Gulliver EL., et al. “Review article: the future of microbiome-based therapeutics”. Alimentary Pharmacology and Therapeutics 2 (2022): 192-208.
  318. Kunst C., et al. “The influence of gut microbiota on oxidative stress and the immune system”. Biomedicines5 (2023): 1388.
  319. Lee JY., et al. “The microbiome and gut homeostasis”. Science (New York, N.Y.)6601 (2022): eabp9960.
  320. Lee JY., et al. “The human gut microbiome in health and disease: time for a new chapter?”. Infection and Immunity11 (2024): e0030224.
  321. Gupte A and Mumper RJ. “Elevated copper and oxidative stress in cancer cells as a target for cancer treatment”. Cancer Treatment Reviews1 (2009): 32-46.
  322. Arfin S., et al. “Oxidative stress in cancer cell metabolism”. Antioxidants (Basel, Switzerland)5 (2021): 642.
  323. Brown JS., et al. “Updating the definition of cancer”. Molecular Cancer Research: MCR11 (2023): 1142-1147.
  324. Cooper GM. “The cell: A molecular approach”. 2nd Sunderland (MA): Sinauer Associates; The Development and Causes of Cancer (2000).
  325. Shipitsin M and Polyak K. “The cancer stem cell hypothesis: in search of definitions, markers, and relevance”. Laboratory Investigation; A Journal of Technical Methods and Pathology5 (2008): 459-463.
  326. Iqbal MJ., et al. “Interplay of oxidative stress, cellular communication and signaling pathways in cancer”. Cell Communication and Signaling: CCS1 (2024): 7.
  327. Aboelella NS., et al. “Oxidative stress in the tumor microenvironment and its relevance to cancer immunotherapy”. Cancers 5 (2021): 986.
  328. Salnikow K. “Role of iron in cancer”. Seminars in Cancer Biology 76 (2021): 189-194.
  329. Xi Y., et al. “Mechanisms of induction of tumors by cholesterol and potential therapeutic prospects”. Biomedicine and Pharmacotherapy = Biomedecine and Pharmacotherapie 144 (2021): 112277.
  330. Saleh EAM., et al. “Oxidative stress affects the beginning of the growth of cancer cells through a variety of routes”. Pathology, Research and Practice 249 (2023): 154664.
  331. Noh J., et al. “Amplification of oxidative stress by a dual stimuli-responsive hybrid drug enhances cancer cell death”. Nature Communications 6 (2015): 6907.
  332. Hayes JD., et al. “Oxidative stress in cancer”. Cancer Cell2 (2020): 167-197.
  333. Zhao Q., et al. “AKR1B1-dependent fructose metabolism enhances malignancy of cancer cells”. Cell Death and Differentiation12 (2024): 1611-1624.
  334. Ting KKY. “Fructose-induced metabolic reprogramming of cancer cells”. Frontiers in Immunology 15 (2024): 1375461.
  335. Nakagawa T., et al. “Fructose contributes to the Warburg effect for cancer growth”. Cancer and Metabolism 8 (2020): 16.
  336. Mao Y., et al. “Metabolic reprogramming, sensing, and cancer therapy”. Cell Reports12 (2024): 115064.
  337. Koltai T and Fliegel L. “Fructose, another sweet for cancer: a context acting nutrient hypothesis”. Gene Expression 2 (2023): 141-155.
  338. Chen HW., et al. “Inhibition of cell growth by oxygenated derivatives of cholesterol”. Nature5474 (1974): 419-421.
  339. Stanhope KL and Havel PJ. “Fructose consumption: potential mechanisms for its effects to increase visceral adiposity and induce dyslipidemia and insulin resistance”. Current Opinion in Lipidology1 (2008): 16-24.
  340. Menezes-Santos M., et al. “Copper deficiency associated with glycemic control in individuals with type 2 diabetes mellitus”. Biological Trace Element Research1 (2025): 119-126.
  341. Odom H. “Copper deficiency reduces insulin receptor and AKT activation in house hepatocytes”. Undergraduate Honors Thesis. University of Nebraska - Lincoln (2024).
  342. Tan PY and Soma Roy, M. “Dietary copper and selenium are associated with insulin resistance in overweight and obese Malaysian adults”. Nutrition Research (New York, N.Y.) 93 (2021): 38-47.
  343. Kuang H., et al. “The impact of egg nutrient composition and its consumption on cholesterol homeostasis”. Cholesterol (2018): 6303810.
  344. Skrivan M., et al. “Effect of various copper supplements to feed of laying hens on cu content in eggs, liver, excreta, soil, and herbage”. Archives of Environmental Contamination and Toxicology2 (2006): 280-283.
  345. Ward C., et al. “The impact of tumour pH on cancer progression: strategies for clinical intervention”. Exploration of Targeted Anti-Tumor Therapy2 (2020): 71-100.
  346. Hao G., et al. “Manipulating extracellular tumour pH: an effective target for cancer therapy”. RSC Advances39 (2018): 22182-22192.
  347. Koltai T. “Cancer: fundamentals behind pH targeting and the double-edged approach”. OncoTargets and Therapy 9 (2016): 6343-6360.
  348. Olżyńska A., et al. “Tail-oxidized cholesterol enhances membrane permeability for small solutes”. Langmuir: The ACS Journal of Surfaces and Colloids 35 (2020): 10438-10447.
  349. Yang Y-T., et al. “Characterization of cholesterol-depleted or -restored cell membranes by depth-sensing nano-indentation”. Soft Matter 8 (2011): 682-687.
  350. Giraldo-Lorza JM., et al. “The influence of cholesterol on membrane targeted bioactive peptides: modulating peptide activity through changes in bilayer biophysical properties”. Membranes10 (2024): 220.
  351. Aredia F and Scovassi AI. “Manipulation of intracellular pH in cancer cells by NHE1 inhibitors”. Protein and Peptide Letters 12 (2016): 1123-1129.
  352. Lee S and Shanti A. “Effect of exogenous pH on cell growth of breast cancer cells”. International Journal of Molecular Sciences18 (2021): 9910.
  353. White KA., et al. “Cancer cell behaviors mediated by dysregulated pH dynamics at a glance”. Journal of Cell Science4 (2017): 663-669.
  354. Tafech A and Stéphanou A. “On the importance of acidity in cancer cells and therapy”. Biology4 (2024): 225.
  355. Damaghi M., et al. “pH sensing and regulation in cancer”. Frontiers in Physiology 4 (2013): 370.
  356. Kulig W., et al. “Oxidation of cholesterol changes the permeability of lipid membranes”. Biophysical Journal3 (2017): 377a.
  357. Zipser B., et al. “Cholesterol and its derivatives, are the principal steroids isolated from the leech species Hirudo medicinalis”. Comparative Biochemistry and Physiology. Part C, Pharmacology, Toxicology and Endocrinology2 (1998): 269-282.
  358. Liu S., et al. “SOD1 promotes cell proliferation and metastasis in non-small cell lung cancer via an miR-409-3p/SOD1/SETDB1 epigenetic regulatory feedforward loop”. Frontiers in Cell and Developmental Biology 8 (2020): 213.
  359. König S., et al. “Superoxide dismutase 1 mediates adaptation to the tumor microenvironment of glioma cells via mammalian target of rapamycin complex”. Cell Death Discovery1 (2024): 379.
  360. Harris N., et al. “Overexpressed Sod1p acts either to reduce or to increase the lifespans and stress resistance of yeast, depending on whether it is Cu(2+)-deficient or an active Cu, Zn-superoxide dismutase”. Aging Cell1 (2005): 41-52.
  361. Magrì A., et al. “Overexpression of human SOD1 in VDAC1-less yeast restores mitochondrial functionality modulating beta-barrel outer membrane protein genes”. Biochimica et Biophysica Acta 6 (2016): 789-798.
  362. Wawryn J., et al. “Deficiency in superoxide dismutases shortens life span of yeast cells”. Acta Biochimica Polonica2 (1999): 249-253.
  363. Tasić D., et al. “Effects of fructose and stress on rat renal copper metabolism and antioxidant enzymes function”. International Journal of Molecular Sciences16 (2022): 9023.
  364. Klevay LM. “Iron overload can induce mild copper deficiency”. Journal of Trace Elements in Medicine and Biology: Organ of the Society for Minerals and Trace Elements (GMS)4 (2001): 237-240.
  365. Fisher AL., et al. “Iron loading induces cholesterol synthesis and sensitizes endothelial cells to TNFα-mediated apoptosis”. The Journal of Biological Chemistry4 (2021): 101156.
  366. Lee J., et al. “High iron consumption modifies the hepatic transcriptome related to cholesterol metabolism”. Journal of Medicinal Food9 (2024): 895-900.
  367. Fouani L., et al. “Metals and metastasis: Exploiting the role of metals in cancer metastasis to develop novel anti-metastatic agents”. Pharmacological Research 115 (2017): 275-287.
  368. Shan D., et al. “Copper in cancer: friend or foe? Metabolism, dysregulation, and therapeutic opportunities”. Cancer Communications (London, England) 5 (2025): 577-607.
  369. Kamiya T. “Copper in the tumor microenvironment and tumor metastasis”. Journal of Clinical Biochemistry and Nutrition 1 (2022): 22-28.
  370. Xiao M., et al. “Functional significance of cholesterol metabolism in cancer: from threat to treatment”. Experimental and Molecular Medicine9 (2023): 1982-1995.
  371. Ding X., et al. “The role of cholesterol metabolism in cancer”. American Journal of Cancer Research2 (2019): 219-227.
  372. Hordyjewska A., et al. “The many "faces" of copper in medicine and treatment”. Biometals: An International Journal on the Role of Metal Ions in Biology, Biochemistry, and Medicine4 (2014): 611-621.
  373. Ikonen E and Zhou X. “Cholesterol transport between cellular membranes: A balancing act between interconnected lipid fluxes”. Developmental Cell10 (2021): 1430-1436.
  374. Chen Y., et al. “Regulation of intracellular cholesterol distribution by Na/K-ATPase”. The Journal of Biological Chemistry22 (2009): 14881-14890.
  375. Le Grimellec C and Leblanc G. “Effect of membrane cholesterol on potassium transport in Mycoplasma mycoides var. Capri (PG3)”. Biochimica et Biophysica Acta1 (1978): 152-163.
  376. Franco R., et al. “Glutathione depletion is necessary for apoptosis in lymphoid cells independent of reactive oxygen species formation”. The Journal of Biological Chemistry42 (2007): 30452-30465.
  377. Hatori Y., et al. “Functional partnership of the copper export machinery and glutathione balance in human cells”. The Journal of Biological Chemistry32 (2012): 26678-26687.
  378. Ghibelli L., et al. “Non-oxidative loss of glutathione in apoptosis via GSH extrusion”. Biochemical and Biophysical Research Communications 1 (1995): 313-320.
  379. Boggs SE., et al. “Glutathione levels determine apoptosis in macrophages”. Biochemical and Biophysical Research Communications 2 (1998): 229-233.
  380. Circu ML and Aw TY. “Glutathione and apoptosis”. Free Radical Research8 (2008): 689-706.
  381. Maryon EB., et al. “Cellular glutathione plays a key role in copper uptake mediated by human copper transporter 1”. American Journal of Physiology. Cell Physiology8 (2013): C768-C779.
  382. Gustafsson J., et al. “Cholesterol synthesis in patients with glutathione deficiency”. European Journal of Clinical Investigation4 (1990): 470-474.
  383. Le Jeune N., et al. “Influence of glutathione depletion on plasma membrane cholesterol esterification and on Tc-99m-sestamibi and Tc-99m-tetrofosmin uptakes: a comparative study in sensitive U-87-MG and multidrug-resistant MRP1 human glioma cells”. Cancer Biotherapy and Radiopharmaceuticals4 (2004): 411-421.
  384. McCay PB., et al. “Glutathione-dependent inhibition of lipid peroxidation by a soluble, heat-labile factor not glutathione peroxidase”. Federation Proceedings2 (1981): 199-205.
  385. Lee JY., et al. “Lipid metabolism and ferroptosis”. Biology3 (2021): 184.
  386. Ursini F and Maiorino M. “Lipid peroxidation and ferroptosis: The role of GSH and GPx4”. Free Radical Biology and Medicine 152 (2020): 175-185.
  387. Ashkaran F., et al. “Mutation/metal deficiency in the "electrostatic loop" enhanced aggregation process in apo/holo SOD1 variants: implications for ALS diseases”. BMC Chemistry1 (2024): 177.
  388. Homma K., et al. “SOD1 as a molecular switch for initiating the homeostatic ER stress response under zinc deficiency”. Molecular Cell1 (2013): 75-86.
  389. Damiano S., et al. “Metabolism regulation and redox state: insight into the role of superoxide dismutase 1”. International Journal of Molecular Sciences18 (2020): 6606.
  390. Raha S and Robinson BH. “Mitochondria, oxygen free radicals, and apoptosis”. American Journal of Medical Genetics 1 (2001): 62-70.
  391. Azad N and Iyer AKV. “Reactive oxygen species and apoptosis”. In: Laher, I. (eds) Systems Biology of Free Radicals and Antioxidants. Springer, Berlin, Heidelberg (2014).
  392. Chen Q., et al. “The late increase in intracellular free radical oxygen species during apoptosis is associated with cytochrome c release, caspase activation, and mitochondrial dysfunction”. Cell Death and Differentiation3 (2003): 323-334.
  393. Li Y., et al. “The ferroptosis inhibitor liproxstatin-1 ameliorates LPS-induced cognitive impairment in mice”. Nutrients21 (2022): 4599.
  394. Cronin SJF., et al. “The role of iron regulation in immunometabolism and immune-related disease”. Frontiers in Molecular Biosciences 6 (2019): 116.
  395. Porto G and De Sousa M. “Iron overload and immunity”. World Journal of Gastroenterology35 (2007): 4707-4715.
  396. Obeagu EI. “Iron homeostasis and health: understanding its role beyond blood health - a narrative review”. Annals of Medicine and Surgery (2012)6 (2025): 3362-3371.
  397. Keiko M., et al. “Iron overload effects on immune system through the cytokine secretion by macrophage”. Blood 21 (2013): 1047.
  398. Walker EM Jr and Walker SM. “Effects of iron overload on the immune system”. Annals of Clinical and Laboratory Science4 (2000): 354-365.
  399. Acharya GK., et al. “Autoimmune hepatitis: Diagnostic dilemma when it is disguised as iron overload syndrome”. Journal of Clinical and Experimental Hepatology 3 (2017): 269-273.
  400. Falahatian M., et al. “Hereditary hemochromatosis associated with autoimmune hemolytic anemia; A case report”. Journal of Preventive Epidemiology 1 (2019): e02.
  401. Yazdali Koylu N., et al. “In the presence of autoantibodies and iron overload, do not judge a book by its cover: A case report”. Hepatology Forum2 (2021): 76-79.
  402. Zandman-Goddard G and Shoenfeld Y. “Ferritin in autoimmune diseases”. Autoimmunity Reviews7 (2007): 457-463.
  403. Fibach E and Rachmilewitz EA. “Iron overload in hematological disorders”. Presse Medicale (Paris, France: 1983) 12.2 (2017): e296-e305.
  404. Hsu CC., et al. “Iron overload disorders”. Hepatology Communications8 (2022): 1842-1854.
  405. Pang N., et al. “Iron overload causes macrophages to produce a pro-inflammatory phenotype in the synovium of hemophiliac arthritis via the acetyl-p53 pathway”. Haemophilia: The Official Journal of the World Federation of Hemophilia1 (2024): 195-203.
  406. Ghezzi P. “Role of glutathione in immunity and inflammation in the lung”. International Journal of General Medicine 4 (2011): 105-113.
  407. Tan M., et al. “Glutathione system enhancement for cardiac protection: pharmacological options against oxidative stress and ferroptosis”. Cell Death and Disease2 (2023): 131.
  408. Rajasekaran NS., et al. “Chronic depletion of glutathione (GSH) and minimal modification of LDL in vivo: its prevention by glutathione mono ester (GME) therapy”. Biochimica et Biophysica Acta1-2 (2005): 103-112.
  409. Yang X., et al. “Inhibition of glutathione production induces macrophage CD36 expression and enhances cellular-oxidized low density lipoprotein (oxLDL) uptake”. The Journal of Biological Chemistry36 (2015): 21788-21799.
  410. Saito Y., et al. “Cholesterol is more readily oxidized than phospholipid linoleates in cell membranes to produce cholesterol hydroperoxides”. Free Radical Biology and Medicine 211 (2024): 89-95.
  411. Lushchak VI. “Glutathione homeostasis and functions: potential targets for medical interventions”. Journal of Amino Acids (2012): 736837.
  412. Catalgol B and Kartal Ozer N. “Lipid rafts and redox regulation of cellular signaling in cholesterol induced atherosclerosis”. Current Cardiology Reviews4 (2010): 309-324.
  413. Matuz-Mares D., et al. “Glutathione participation in the prevention of cardiovascular diseases”. Antioxidants (Basel, Switzerland) 8 (2021): 1220.
  414. Shan XQ., et al. “Glutathione-dependent protection against oxidative injury”. Pharmacology and Therapeutics1 (1990): 61-71.
  415. Averill-Bates DA. “The antioxidant glutathione”. Vitamins and Hormones 121 (2023): 109-141.
  416. Lee LR., et al. “Glutathione accelerates the cell cycle and cellular reprogramming in plant regeneration”. bioRxiv (2024).
  417. Diaz Vivancos P., et al. “A nuclear glutathione cycle within the cell cycle”. The Biochemical Journal2 (2010): 169-178.
  418. Pallardó FV., et al. “Role of nuclear glutathione as a key regulator of cell proliferation”. Molecular Aspects of Medicine 1-2 (2009): 77-85.
  419. Ayala A., et al. “Lipid peroxidation: production, metabolism, and signaling mechanisms of malondialdehyde and 4-hydroxy-2-nonenal”. Oxidative Medicine and Cellular Longevity (2014): 360438.
  420. Benedusi V., et al. “The peroxisome proliferator-activated receptor γ (PPARγ) controls natural protective mechanisms against lipid peroxidation in amyotrophic lateral sclerosis”. The Journal of Biological Chemistry43 (2012): 35899-35911.
  421. Nam TG. “Lipid peroxidation and its toxicological implications”. Toxicological Research1 (2011): 1-6.
  422. Li Pomi F., et al. “Oxidative stress and skin diseases: The role of lipid peroxidation”. Antioxidants5 (2025): 555.
  423. Lee SH., et al. “Lipid peroxidation-derived modification and its effect on the activity of glutathione peroxidase 1”. Free Radical Biology and Medicine 208 (2023): 252-259.
  424. Nardella MI., et al. “Oxidation of human copper chaperone atox1 and disulfide bond cleavage by cisplatin and glutathione”. International Journal of Molecular Sciences18 (2019): 4390.
  425. Guan D., et al. “Copper in cancer: From pathogenesis to therapy”. Biomedicine and Pharmacotherapy = Biomedecine and Pharmacotherapie 163 (2023): 114791.
  426. Juarez JC., et al. “Copper binding by tetrathiomolybdate attenuates angiogenesis and tumor cell proliferation through the inhibition of superoxide dismutase 1”. Clinical Cancer Research: An Official Journal of the American Association for Cancer Research16 (2006): 4974-4982.
  427. Tang X., et al. “Copper in cancer: from limiting nutrient to therapeutic target”. Frontiers in Oncology 13 (2023): 1209156.
  428. Badran O., et al. “The impact of iron on cancer-related immune functions in oncology: molecular mechanisms and clinical evidence”. Cancers 24 (2024): 4156.
  429. Shi Y., et al. “Effect of metal loading and subcellular pH on net charge of superoxide dismutase-1”. Journal of Molecular Biology22 (2013): 4388-4404.
  430. Byström R., et al. “SOD1 mutations targeting surface hydrogen bonds promote amyotrophic lateral sclerosis without reducing apo-state stability”. The Journal of Biological Chemistry25 (2010): 19544-19552.
  431. Perry JJ., et al. “The structural biochemistry of the superoxide dismutases”. Biochimica et Biophysica Acta2 (2010): 245-262.
  432. Sehati S., et al. “Metabolic alterations in yeast lacking copper-zinc superoxide dismutase”. Free Radical Biology and Medicine11 (2011): 1591-1598.
  433. Ansenberger-Fricano K., et al. “The peroxidase activity of mitochondrial superoxide dismutase”. Free Radical Biology and Medicine 54 (2013): 116-124.
  434. Arnesano F., et al. “The unusually stable quaternary structure of human Cu,Zn-superoxide dismutase 1 is controlled by both metal occupancy and disulfide status”. The Journal of Biological Chemistry 46 (2004): 47998-48003.
  435. Ramirez DC., et al. “Cu,Zn-superoxide dismutase-driven free radical modifications: copper- and carbonate radical anion-initiated protein radical chemistry”. The Biochemical Journal1 (2009): 341-353.
  436. Zhang H., et al. “Bicarbonate enhances peroxidase activity of Cu,Zn-superoxide dismutase. Role of carbonate anion radical and scavenging of carbonate anion radical by metalloporphyrin antioxidant enzyme mimetics”. The Journal of Biological Chemistry2 (2002): 1013-1020.
  437. Hink HU., et al. “Peroxidase properties of extracellular superoxide dismutase: role of uric acid in modulating in vivo activity”. Arteriosclerosis, Thrombosis, and Vascular Biology9 (2002): 1402-1408.
  438. Sankarapandi S and Zweier JL. “Bicarbonate is required for the peroxidase function of Cu, Zn-superoxide dismutase at physiological pH”. The Journal of Biological Chemistry3 (1999): 1226-1232.
  439. Li Y., et al. “Iron and copper: critical executioners of ferroptosis, cuproptosis and other forms of cell death”. Cell Communication and Signaling: CCS1 (2023): 327.
  440. Troost FJ., et al. “Iron supplements inhibit zinc but not copper absorption in vivo in ileostomy subjects”. The American Journal of Clinical Nutrition 5 (2003): 1018-1023.
  441. Aracena P., et al. “Iron and glutathione at the crossroad of redox metabolism in neurons”. Biological Research1 (2006): 157-165.
  442. Núñez MT., et al. “Progressive iron accumulation induces a biphasic change in the glutathione content of neuroblastoma cells”. Free Radical Biology and Medicine7 (2004): 953-960.
  443. Han P., et al. “Activation of chicken liver fructose-1,6-bisphosphatase by oxidized glutathione”. FEBS Letters2 (1986): 347-351.
  444. Halliwell B and Foyer CH. “Properties and physiological function of a glutathione reductase purified from spinach leaves by affinity chromatography”. Planta1 (1978): 9-17.
  445. Song M., et al. “High fructose feeding induces copper deficiency in Sprague-Dawley rats: a novel mechanism for obesity related fatty liver”. Journal of Hepatology2 (2012): 433-440.
  446. Moreno JA and Hong E. “A single oral dose of fructose induces some features of metabolic syndrome in rats: role of oxidative stress”. Nutrition, Metabolism, and Cardiovascular Diseases: NMCD6 (2013): 536-542.
  447. Silva JM., et al. “Prevention of nitrofurantoin-induced cytotoxicity in isolated hepatocytes by fructose”. Archives of Biochemistry and Biophysics 2 (1991): 313-318.
  448. Kulig W., et al. “Cholesterol oxidation products and their biological importance”. Chemistry and Physics of Lipids 199 (2016): 144-160.
  449. Kim JW., et al. “An integrated view of lipid metabolism in ferroptosis revisited via lipidomic analysis”. Experimental and Molecular Medicine8 (2023): 1620-1631.
  450. Chen X., et al. “Ferroptosis by Lipid Peroxidation: The Tip of the Iceberg?”. Frontiers in Cell and Developmental Biology 9 (2021): 646890.
  451. Li X., et al. “Iron accumulation and lipid peroxidation: implication of ferroptosis in hepatocellular carcinoma”. Frontiers in Endocrinology 14 (2024): 1319969.
  452. Yang WS and Stockwell BR. “Ferroptosis: Death by lipid peroxidation”. Trends in Cell Biology3 (2016): 165-176.
  453. Endale HT., et al. “ROS induced lipid peroxidation and their role in ferroptosis”. Frontiers in Cell and Developmental Biology 11 (2023): 1226044.
  454. Yang X., et al. “Ferroptosis as a new tool for tumor suppression through lipid peroxidation”. Communications Biology1 (2024): 1475.
  455. Conrad M., et al. “Regulation of lipid peroxidation and ferroptosis in diverse species”. Genes and Development 9-10 (2018): 602-619.
  456. Coradduzza D., et al. “Ferroptosis and senescence: A systematic review”. International Journal of Molecular Sciences4 (2023): 3658.
  457. Cheng Z., et al. “Ferroptosis in non-alcoholic liver disease: Molecular mechanisms and therapeutic implications”. Frontiers in Nutrition 10 (2023): 1090338.
  458. Montllor-Albalate C., et al. “Sod1 integrates oxygen availability to redox regulate NADPH production and the thiol redoxome”. Proceedings of the National Academy of Sciences of the United States of America1 (2022): e2023328119.
  459. Patra KC and Hay N. “The pentose phosphate pathway and cancer”. Trends in Biochemical Sciences8 (2014): 347-354.
  460. Krause N and Wegner A. “Fructose metabolism in cancer”. Cells12 (2020): 2635.
  461. Chen H., et al. “Mechanisms and active substances of targeting lipid peroxidation in ferroptosis regulation”. Food Science and Human Wellness5 (2024): 2502-2518.
  462. Gao L., et al. “Bioinformatics analysis reveals SOD1 is a prognostic factor in lung adenocarcinoma”. Translational Cancer Research10 (2024): 5522-5534.
  463. Sangani RG and Ghio AJ. “Iron, human growth, and the global epidemic of obesity”. Nutrients10 (2013): 4231-4249.
  464. Manz DH., et al. “Iron and cancer: recent insights”. Annals of the New York Academy of Sciences1 (2016): 149-161.
  465. Islam S., et al. “Iron overload and breast cancer: iron chelation as a potential therapeutic approach”. Life (Basel, Switzerland)7 (2022): 963.
  466. Basak T and Kanwar RK. “Iron imbalance in cancer: Intersection of deficiency and overload”. Cancer Medicine20 (2022): 3837-3853.
  467. Ngamchuea K., et al. “The Copper(II)-Catalyzed Oxidation of Glutathione”. Chemistry (Weinheim an der Bergstrasse, Germany) 44 (2016): 15937-15944.
  468. Friesen C., et al. “A critical role of glutathione in determining apoptosis sensitivity and resistance in leukemia cells”. Cell Death and Differentiation 1 (2004): S73-S85.
  469. Cazanave S., et al. “High hepatic glutathione stores alleviate Fas-induced apoptosis in mice”. Journal of Hepatology5 (2007): 858-868.
  470. Armstrong JS., et al. “Role of glutathione depletion and reactive oxygen species generation in apoptotic signaling in a human B lymphoma cell line”. Cell Death and Differentiation3 (2002): 252-263.
  471. Franco R and Cidlowski JA. “Apoptosis and glutathione: beyond an antioxidant”. Cell Death and Differentiation10 (2009): 1303-1314.
  472. Liu XQ., et al. “Hypoxia and ferroptosis”. Cellular Signalling 122 (2024): 111328.
  473. Feng S., et al. “The mechanism of ferroptosis and its related diseases”. Molecular Biomedicine1 (2023): 33.
  474. Chen X. “Organelle-specific Mechanisms of Ferroptosis”. In: Tang, D. (eds) Ferroptosis in Health and Disease. Springer, Cham (2023).
  475. Zhang LL., et al. “The underlying pathological mechanism of ferroptosis in the development of cardiovascular disease”. Frontiers in Cardiovascular Medicine 9 (2022): 964034.
  476. Brasil AA., et al. “The involvement of GSH in the activation of human Sod1 linked to FALS in chronologically aged yeast cells”. FEMS Yeast Research5 (2013): 433-440.
  477. Boyd SD., et al. “Quantifying the interaction between copper-zinc superoxide dismutase (Sod1) and its copper chaperone (Ccs1)”. Journal of Proteomics and Bioinformatics4 (2018): 473.
  478. Furukawa Y. “A pathological link between dysregulated copper binding in Cu/Zn-superoxide dismutase and amyotrophic lateral sclerosis”. Journal of Clinical Biochemistry and Nutrition2 (2022): 73-77.
  479. Kamiya T. “Role of copper and SOD3-mediated extracellular redox regulation in tumor progression”. Journal of Clinical Biochemistry and Nutrition1 (2024): 1-6.
  480. Trumbull KA and Beckman JS. “A role for copper in the toxicity of zinc-deficient superoxide dismutase to motor neurons in amyotrophic lateral sclerosis”. Antioxidants and Redox Signaling7 (2009): 1627-1639.
  481. González M., et al. “Expression of copper-related genes in response to copper load”. The American Journal of Clinical Nutrition3 (2008): 830S-834S.
  482. Roy N and Paira P. “Glutathione depletion and stalwart anticancer activity of metallotherapeutics inducing programmed cell death: opening a new window for cancer therapy”. ACS Omega19 (2024): 20670-20701.
  483. Kennedy L., et al. “Role of glutathione in cancer: from mechanisms to therapies”. Biomolecules 10 (2020): 1429.
  484. Grek CL., et al. “Causes and consequences of cysteine S-glutathionylation”. The Journal of Biological Chemistry 37 (2013): 26497-26504.
  485. Xiong Y., et al. “S-glutathionylation: from molecular mechanisms to health outcomes”. Antioxidants and Redox Signaling1 (2011): 233-270.
  486. Pal D., et al. “Role of protein S-Glutathionylation in cancer progression and development of resistance to anti-cancer drugs”. Archives of Biochemistry and Biophysics 704 (2021): 108890.
  487. Kalinina E. “Glutathione-dependent pathways in cancer cells”. International Journal of Molecular Sciences15 (2024): 8423.
  488. Lee JH and Paik HD. “Anticancer and immunomodulatory activity of egg proteins and peptides: a review”. Poultry Science12 (2019): 6505-6516.
  489. Franco R and Cidlowski JA. “Apoptosis and glutathione: beyond an antioxidant”. Cell Death and Differentiation10 (2009): 1303-1314.
  490. Liu XQ., et al. “Hypoxia and ferroptosis”. Cellular Signalling 122 (2024): 111328.
  491. Feng S., et al. “The mechanism of ferroptosis and its related diseases”. Molecular Biomedicine1 (2023): 33.
  492. Chen X. “Organelle-specific Mechanisms of Ferroptosis”. In: Tang, D. (eds) Ferroptosis in Health and Disease. Springer, Cham (2023).
  493. Zhang LL., et al. “The underlying pathological mechanism of ferroptosis in the development of cardiovascular disease”. Frontiers in Cardiovascular Medicine 9 (2022): 964034.
  494. Brasil AA., et al. “The involvement of GSH in the activation of human Sod1 linked to FALS in chronologically aged yeast cells”. FEMS Yeast Research5 (2013): 433-440.
  495. Boyd SD., et al. “Quantifying the interaction between copper-zinc superoxide dismutase (Sod1) and its copper chaperone (Ccs1)”. Journal of Proteomics and Bioinformatics4 (2018): 473.
  496. Furukawa Y. “A pathological link between dysregulated copper binding in Cu/Zn-superoxide dismutase and amyotrophic lateral sclerosis”. Journal of Clinical Biochemistry and Nutrition2 (2022): 73-77.
  497. Kamiya T. “Role of copper and SOD3-mediated extracellular redox regulation in tumor progression”. Journal of Clinical Biochemistry and Nutrition1 (2024): 1-6.
  498. Trumbull KA and Beckman JS. “A role for copper in the toxicity of zinc-deficient superoxide dismutase to motor neurons in amyotrophic lateral sclerosis”. Antioxidants and Redox Signaling7 (2009): 1627-1639.
  499. González M., et al. “Expression of copper-related genes in response to copper load”. The American Journal of Clinical Nutrition3 (2008): 830S-834S.
  500. Roy N and Paira P. “Glutathione depletion and stalwart anticancer activity of metallotherapeutics inducing programmed cell death: opening a new window for cancer therapy”. ACS Omega19 (2024): 20670-20701.
  501. Kennedy L., et al. “Role of glutathione in cancer: from mechanisms to therapies”. Biomolecules 10 (2020): 1429.
  502. Grek CL., et al. “Causes and consequences of cysteine S-glutathionylation”. The Journal of Biological Chemistry 37 (2013): 26497-26504.
  503. Xiong Y., et al. “S-glutathionylation: from molecular mechanisms to health outcomes”. Antioxidants and Redox Signaling1 (2011): 233-270.
  504. Pal D., et al. “Role of protein S-Glutathionylation in cancer progression and development of resistance to anti-cancer drugs”. Archives of Biochemistry and Biophysics 704 (2021): 108890.
  505. Kalinina E. “Glutathione-dependent pathways in cancer cells”. International Journal of Molecular Sciences15 (2024): 8423.
  506. Lee JH and Paik HD. “Anticancer and immunomodulatory activity of egg proteins and peptides: a review”. Poultry Science12 (2019): 6505-6516.

Faiza Abdur Rab. “Role of Food-Its Influence on Modulation of Therapies Including Chemotherapy’s Outcomes”. EC Nutrition  20.2 (2025): 01-31.