Involvement of Mfn2, Bcl2/Bax signaling and mitochondrial viability in the potential protective effect of Royal jelly against mitochondria-mediated ovarian apoptosis by cisplatin in rats

Document Type : Original Article

Authors

1 Department of Biochemistry, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef, Egypt

2 Department of Pharmacology, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt

3 Department of Theriogenology, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef, Egypt

4 Department of Physiology, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef, Egypt

Abstract

Objective(s): The current study aimed to assess cisplatin-mediated ovarian apoptosis in a rat model by Royal jelly (RJ).
Materials and Methods: Thirty female adult albino rats (180-200 g) were divided into three groups (n=10): saline (0.9% NaCl, IP) was given to the control group, the cisplatin group: received (5 mg/kg/once a week IP) for 5 successive weeks, the RJ+Cis. group: received RJ (100 mg/kg/ day PO daily), and Cisplatin (5 mg/kg/once per week IP) for 5 successive weeks. At the end of the experiment, rats were sacrificed and their ovaries were isolated and used for biochemical analysis, molecular investigations and morphometric assessment as well as histological study. Moreover, blood samples were collected for determination of follicle-stimulating hormone (FSH), luteinizing hormone (LH), Estradiol, progesterone and anti-mullerian hormone (AMH).
Results: The current study clarified that RJ given to rats prior to cisplatin significantly increased the ovarian and uterine weights, in addition to follicular count at P˂0.05 compared to rats injected only with cisplatin. Moreover, it restored normal ovarian histological structure with a concurrent reduction in FSH, and LH levels, and increased AMH and ovarian hormone concentrations at P˂0.05 compared to cisplatin group. Also, RJ decreased the ovarian antioxidant/oxidative imbalance harmonized with significant suppression of inducible nitric oxide synthase and increase of quinone oxidoreductase 1 mRNA expression at P˂0.05 compared to cisplatin group.
Conclusion: We concluded that RJ could alleviate mitochondrial-induced ovarian apoptosis caused by cisplatin via increasing anti-apoptotic Bcl2, and diminishing pro-apoptotic Bax with a concomitant increase of Mfn2 mRNA and protein expressions.

Keywords


1. Oktem O, Oktay K. Quantitative assessment of the impact of chemotherapy on ovarian follicle reserve and stromal function. Cancer 2007; 110:2222-2229.
2. Gonfloni S, Di Tella L, Caldarola S, Cannata SM, Klinger FG, Di Bartolomeo C, et al. Inhibition of the c-Abl-TAp63 pathway protects mouse oocytes from chemotherapy-induced death. Nat Med 2009; 15:1179-1185.
3. Dasari S, Tchounwou PB. Cisplatin in cancer therapy: molecular mechanisms of action. Eur J Pharmacol 2014; 740:364-378.
4. Fatima S, Arivarasu NA, Mahmood R. Vitamin C attenuates cisplatin-induced alterations in renal brush border membrane enzymes and phosphate transport. Hum Exp Toxicol 2007; 26:419-426.
5. Ciftci O, Ozdemir I, Vardi N, Gurbuz N. Novel platinum-N-heterocyclic carbene complex is more cardiotoxic than cis-platin in rats. Hum Exp Toxicol 2011; 30:1342-1349.
6. Marhhom E, Cohen I. Fertility preservation options for women with malignancies. Obstet Gynecol Surv 2007; 62:58-72.
7. Meraner V, Gamper EM, Grahmann A, Giesinger JM, Wiesbauer P, Sztankay M, et al. Monitoring physical and psychosocial symptom trajectories in ovarian cancer patients receiving chemotherapy. BMC Cancer 2012; 12:77.
8. Sugiyama S, Hayakawa M, Kato T, Hanaki Y, Shimizu K, Ozawa T. Adverse effects of anti-tumor drug, cisplatin, on rat kidney mitochondria: disturbances in glutathione peroxidase activity. Biochem Biophys Res Commun 1989; 159:1121-1127.
9. Brady HR, Kone BC, Stromski ME, Zeidel ML, Giebisch G, Gullans SR. Mitochondrial injury: an early event in cisplatin toxicity to renal proximal tubules. Am J Physiol 1990; 258:F1181-1187.
10. Leibbrandt ME, Wolfgang GH, Metz AL, Ozobia AA, Haskins JR. Critical subcellular targets of cisplatin and related platinum analogs in rat renal proximal tubule cells. Kidney Int 1995; 48:761-770.
11. Deavall DG, Martin EA, Horner JM, Roberts R. Drug-induced oxidative stress and toxicity. J Toxicol 2012; 2012:645460.
12. Sancho-Martinez SM, Prieto-Garcia L, Prieto M, Lopez-Novoa JM, Lopez-Hernandez FJ. Subcellular targets of cisplatin cytotoxicity: an integrated view. Pharmacol Ther 2012; 136:35-55.
13. Marullo R, Werner E, Degtyareva N, Moore B, Altavilla G, Ramalingam SS, et al. Cisplatin induces a mitochondrial-ROS response that contributes to cytotoxicity depending on mitochondrial redox status and bioenergetic functions. PLoS One 2013; 8:e81162.
14. Chen W, Xu X, Wang L, Bai G, Xiang W. Low expression of Mfn2 is associated with mitochondrial damage and apoptosis of ovarian tissues in the premature ovarian failure model. PLoS One 2015; 10:e0136421.
15. Meirow D. Reproduction post-chemotherapy in young cancer patients. Mol Cell Endocrinol 2000; 169:123-131.
16. Tangir J, Zelterman D, Ma W, Schwartz PE. Reproductive function after conservative surgery and chemotherapy for malignant germ cell tumors of the ovary. Obstet Gynecol 2003; 101:251-257.
17. Testing and interpreting measures of ovarian reserve: a committee opinion. Fertil Steril 2015; 103:e9-e17.
18. Van Rooij IA, Broekmans FJ, te Velde ER, Fauser BC, Bancsi LF, de Jong FH, et al. Serum anti-Mullerian hormone levels: a novel measure of ovarian reserve. Hum Reprod 2002; 17:3065-3071.
19. Anderson RA, Cameron DA. Pretreatment serum anti-mullerian hormone predicts long-term ovarian function and bone mass after chemotherapy for early breast cancer. J Clin Endocrinol Metab 2011; 96:1336-1343.
20. Ray WZ, Murphy RK, Santosa K, Johnson PJ, Mackinnon SE. Medial pectoral nerve to axillary nerve neurotization following traumatic brachial plexus injuries: indications and clinical outcomes. Hand 2012; 7:59-65.
21. Bonomi M, Somigliana E, Cacciatore C, Busnelli M, Rossetti R, Bonetti S, et al. Blood cell mitochondrial DNA content and premature ovarian aging. PLoS One 2012; 7:e42423.
22. Paradies G, Paradies V, Ruggiero FM, Petrosillo G. Protective role of melatonin in mitochondrial dysfunction and related disorders. Archives of toxicology 2015; 89:923-939.
23. Hirata H, Lopes GS, Jurkiewicz A, Garcez-do-Carmo L, Smaili SS. Bcl-2 modulates endoplasmic reticulum and mitochondrial calcium stores in PC12 cells. Neurochemical research 2012; 37:238-243.
24. Stary CM, Sun X, Ouyang Y, Li L, Giffard RG. miR-29a differentially regulates cell survival in astrocytes from cornu ammonis 1 and dentate gyrus by targeting VDAC1. Mitochondrion 2016; 30:248-254.
25. Pan Z, Gollahon L. Paclitaxel attenuates Bcl-2 resistance to apoptosis in breast cancer cells through an endoplasmic reticulum-mediated calcium release in a dosage dependent manner. Biochemical and biophysical research communications 2013; 432:431-437.
26. Cui C, Merritt R, Fu L, Pan Z. Targeting calcium signaling in cancer therapy. Acta pharmaceutica sinica B 2017; 7:3-17.
27. Arbel N, Shoshan-Barmatz V. Voltage-dependent anion channel 1-based peptides interact with Bcl-2 to prevent antiapoptotic activity. Journal of Biological Chemistry 2010; 285:6053-6062.
28. Yuan Z, Syed MA, Panchal D, Joo M, Colonna M, Brantly M, et al. Triggering receptor expressed on myeloid cells 1 (TREM-1)-mediated Bcl-2 induction prolongs macrophage survival. Journal of Biological Chemistry 2014; 289:15118-15129.
29. Bach D, Pich S, Soriano FX, Vega N, Baumgartner B, Oriola J, et al. Mitofusin-2 determines mitochondrial network architecture and mitochondrial metabolism. A novel regulatory mechanism altered in obesity. J Biol Chem 2003; 278:17190-17197.
30. Ishihara N, Eura Y, Mihara K. Mitofusin 1 and 2 play distinct roles in mitochondrial fusion reactions via GTPase activity. J Cell Sci 2004; 117:6535-6546.
31.de Brito OM, Scorrano L. Mitofusin 2 tethers endoplasmic reticulum to mitochondria. Nature 2008; 456:605-610.
32. Luo Q, Huo P, Wang L, Wu X. The influencing mechanism of mTOR signal pathway mediated by mitofusin-2 in development of follicle. European review for medical and pharmacological sciences 2018; 22:2212-2217.
33. Zorzano A, Hernandez-Alvarez MI, Sebastian D, Munoz JP. Mitofusin 2 as a driver that controls energy metabolism and insulin signaling. Antioxid Redox Signal 2015; 22:1020-1031.
34. Zhao N, Zhang Y, Liu Q, Xiang W. Mfn2 affects embryo development via mitochondrial dysfunction and apoptosis. PloS one 2015; 10:e0125680.
35. Pasupuleti VR, Sammugam L, Ramesh N, Gan SH. Honey, Propolis, and Royal Jelly: A Comprehensive Review of Their Biological Actions and Health Benefits. Oxid Med Cell Longev 2017; 2017:1259510.
36. Ghanbari E, Khazaei MR, Khazaei M, Nejati V. Royal jelly promotes ovarian follicles growth and increases steroid hormones in immature rats. International journal of fertility & sterility 2018; 11:263.
37. Hashimoto M, Kanda M, Ikeno K, Hayashi Y, Nakamura T, Ogawa Y, et al. Oral administration of royal jelly facilitates mRNA expression of glial cell line-derived neurotrophic factor and neurofilament H in the hippocampus of the adult mouse brain. Biosci Biotechnol Biochem 2005; 69:800-805.
38. Nagai T, Inoue R. Preparation and the functional properties of water extract and alkaline extract of royal jelly. Food Chemistry 2004; 84:181-186.
39. Ramadan MF, Al-Ghamdi A. Bioactive compounds and health-promoting properties of royal jelly: A review. Journal of Functional Foods 2012; 4:39-52.
40. Guo H, Ekusa A, Iwai K, Yonekura M, Takahata Y, Morimatsu F. Royal jelly peptides inhibit lipid peroxidation in vitro and in vivo. Journal of nutritional science and vitaminology 2008; 54:191-195.
41. Viuda‐Martos M, Ruiz‐Navajas Y, Fernández‐López J, Pérez‐Álvarez J. Functional properties of honey, propolis, and royal jelly. Journal of food science 2008; 73:R117-R124.
42. Tamura S, Kono T, Harada C, Yamaguchi K, Moriyama T. Estimation and characterisation of major royal jelly proteins obtained from the honeybee Apis merifera. Food Chemistry 2009; 114:1491-1497.
43. Sugiyama T, Takahashi K, Mori H. Royal jelly acid, 10-hydroxy-trans-2-decenoic acid, as a modulator of the innate immune responses. Endocr Metab Immune Disord Drug Targets 2012; 12:368-376.
44. Ibrahim A, Eldaim MAA, Abdel-Daim MM. Nephroprotective effect of bee honey and royal jelly against subchronic cisplatin toxicity in rats. Cytotechnology 2016; 68:1039-1048.
45. Matsunaga Y, Sakata Y, Yago T, Nakamura H, Shimizu T, Takeda Y. Effects of Glucose with Casein Peptide Supplementation on Post-Exercise Muscle Glycogen Resynthesis in C57BL/6J Mice. Nutrients 2018; 10:753.
46. Liang C, Curry BJ, Brown PL, Zemel MB. Leucine modulates mitochondrial biogenesis and SIRT1-AMPK signaling in C2C12 myotubes. Journal of nutrition and metabolism 2014; 2014.
47. Yoshida M, Hayashi K, Watadani R, Okano Y, Tanimura K, Kotoh J, et al. Royal jelly improves hyperglycemia in obese/diabetic KK-Ay mice. Journal of Veterinary Medical Science 2016:16-0458.
48. Takahashi Y, Hijikata K, Seike K, Nakano S, Banjo M, Sato Y, et al. Effects of Royal Jelly Administration on Endurance Training-Induced Mitochondrial Adaptations in Skeletal Muscle. Nutrients 2018; 10:1735.
49. Chtourou Y, Aouey B, Kebieche M, Fetoui H. Protective role of naringin against cisplatin induced oxidative stress, inflammatory response and apoptosis in rat striatum via suppressing ROS-mediated NF-kappaB and P53 signaling pathways. Chem Biol Interact 2015; 239:76-86.
50. Ahmed MM, El-Shazly SA, Alkafafy ME, Mohamed AA, Mousa AA. Protective potential of royal jelly against cadmium-induced infertility in male rats. Andrologia 2018; 50:e12996.
51. Warren B. Theory and Practice of Histological Techniques. Pathology 1996; 28:381.
52. Patil SR, Ravindra S, Patil R, Londonkar R, Patil SB. Nicotine induced ovarian and uterine changes in albino mice. Indian journal of physiology and pharmacology 1998; 42:503-508.
53. Li X, Kang X, Deng Q, Cai J, Wang Z. Combination of a GnRH agonist with an antagonist prevents flare-up effects and protects primordial ovarian follicles in the rat ovary from cisplatin-induced toxicity: a controlled experimental animal study. Reproductive Biology and Endocrinology 2013; 11:16.
54. Beutler E, Duron O, Kelly BM. Improved method for the determination of blood glutathione. J Lab Clin Med 1963; 61:882-888.
55. Aebi H. Catalase in vitro. Methods Enzymol 1984; 105:121-126.
56. Nishikimi M, Appaji N, Yagi K. The occurrence of superoxide anion in the reaction of reduced phenazine methosulfate and molecular oxygen. Biochem Biophys Res Commun 1972; 46:849-854.
57. Koracevic D, Koracevic G, Djordjevic V, Andrejevic S, Cosic V. Method for the measurement of antioxidant activity in human fluids. J Clin Pathol 2001; 54:356-361.
58. Erel O. A new automated colorimetric method for measuring total oxidant status. Clin Biochem 2005; 38:1103-1111.
59. Meng B, Li J, Cao H. Antioxidant and antiinflammatory activities of curcumin on diabetes mellitus and its complications. Curr Pharm Des 2013; 19:2101-2113.
60. Franco JL, Braga HC, Stringari J, Missau FC, Posser T, Mendes BG, et al. Mercurial-induced hydrogen peroxide generation in mouse brain mitochondria: protective effects of quercetin. Chemical research in toxicology 2007; 20:1919-1926.
61. Burton K. A study of the conditions and mechanism of the diphenylamine reaction for the colorimetric estimation of deoxyribonucleic acid. Biochem J 1956; 62:315-323.
62. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 2001; 25:402-408.
63. Gasic S, Vucevic D, Vasilijic S, Antunovic M, Chinou I, Colic M. Evaluation of the immunomodulatory activities of royal jelly components in vitro. Immunopharmacology and Immunotoxicology 2007; 29:521-536.
64. Cayir K, Karadeniz A, Yildirim A, Kalkan Y, Karakoc A, Keles M, et al. Protective effect of L-carnitine against cisplatin-induced liver and kidney oxidant injury in rats. Open Medicine 2009; 4:184-191.
65. Smith J. Erlotinib: Small-molecule targeted therapy in the treatmentof non-small-cell lung cancer. Clinical therapeutics 2005; 27:1513-1534.
66. Crona DJ, Faso A, Nishijima TF, McGraw KA, Galsky MD, Milowsky MI. A systematic review of strategies to prevent cisplatin‐induced nephrotoxicity. The oncologist 2017; 22:609-619.
67. Binnetoglu D, Hacimuftuoglu A, Aricioglu F. Neuroprotective effects of agmatine in antineoplastic drugs induced neurotoxicity: In vitro study. Life sciences 2019; 221:311-318.
68. Adefisayo MA, Adeyemi WJ, Alabi QK. Combined but not single administration of vitamin C and l-carnitine ameliorates cisplatin-induced gastric mucosa damage in male rats. Canadian journal of physiology and pharmacology 2018; 96:830-838.
69. Han SE, Park MJ, Kim HJ, Kim HG, Kim CW, Joo BS, et al. Establishment of Effective Mouse Model of Premature Ovarian Failure Considering Treatment Duration of Anticancer Drugs and Natural Recovery Time. Journal of menopausal medicine 2018; 24:196-203.
70. Ozdamar S, Taskin MI, Onder GO, Kaymak E, Baran M, Yay A. Progesterone decreases the extent of ovarian damage caused by cisplatin in an experimental rat model. Advances in clinical and experimental medicine: official organ Wroclaw Medical University 2019; 28:25-33.
71. Said RS, Mantawy EM, El-Demerdash E. Mechanistic perspective of protective effects of resveratrol against cisplatin-induced ovarian injury in rats: emphasis on anti-inflammatory and anti-apoptotic effects. Naunyn-Schmiedeberg’s archives of pharmacology 2019:1-14.
72. Altuner D, Gulaboglu M, Yapca OE, Cetin N. The effect of mirtazapine on cisplatin-induced oxidative damage and infertility in rat ovaries. The Scientific World Journal 2013; 2013.
73. Hoyer PB. Damage to ovarian development and function. Cell and tissue research 2005; 322:99-106.
74. Mishra B, Ortiz L, Luderer U. Charged iron particles, components of space radiation, destroy ovarian follicles. Human Reproduction 2016; 31:1816-1826.
75. Jayaprakasan K, Campbell B, Hopkisson J, Johnson I, Raine-Fenning N. A prospective, comparative analysis of anti-Müllerian hormone, inhibin-B, and three-dimensional ultrasound determinants of ovarian reserve in the prediction of poor response to controlled ovarian stimulation. Fertility and sterility 2010; 93:855-864.
76. Yeh J, Kim B, Liang YJ, Peresie J. Müllerian inhibiting substance as a novel biomarker of cisplatin-induced ovarian damage. Biochemical and biophysical research communications 2006; 348:337-344.
77. Li X, Yang S, Lv X, Sun H, Weng J, Liang Y, et al. The mechanism of mesna in protection from cisplatin-induced ovarian damage in female rats. Journal of gynecologic oncology 2013; 24:177-185.
78. ANTUNES LMG, DARIN JDAC, BIANCHI MDLP. Protective effects of vitamin C against cisplatin-induced nephrotoxicity and lipid peroxidation in adult rats: a dose-dependent study. Pharmacological Research 2000; 41:405-411.
79. Wozniak K, Czechowska A, Blasiak J. Cisplatin-evoked DNA fragmentation in normal and cancer cells and its modulation by free radical scavengers and the tyrosine kinase inhibitor STI571. Chemico-biological interactions 2004; 147:309-318.
80. Deavall DG, Martin EA, Horner JM, Roberts R. Drug-induced oxidative stress and toxicity. Journal of toxicology 2012; 2012.
81. Rashed LA, Hashem RM, Soliman HM. Oxytocin inhibits NADPH oxidase and P38 MAPK in cisplatin-induced nephrotoxicity. Biomedicine & Pharmacotherapy 2011; 65:474-480.
82. Jacob KD, Hooten NN, Trzeciak AR, Evans MK. Markers of oxidant stress that are clinically relevant in aging and age-related disease. Mechanisms of ageing and development 2013; 134:139-157.
83. Omar HA, Mohamed WR, Arafa E-SA, Shehata BA, El Sherbiny GA, Arab HH, et al. Hesperidin alleviates cisplatin-induced hepatotoxicity in rats without inhibiting its antitumor activity. Pharmacological Reports 2016; 68:349-356.
84. Chtourou Y, Aouey B, Kebieche M, Fetoui H. Protective role of naringin against cisplatin induced oxidative stress, inflammatory response and apoptosis in rat striatum via suppressing ROS-mediated NF-κB and P53 signaling pathways. Chemico-biological interactions 2015; 239:76-86.
85. Krishna MB, Joseph A, Thomas PL, Dsilva B, Pillai SM, Laloraya M. Impaired arginine metabolism coupled to a defective redox conduit contributes to low plasma nitric oxide in polycystic ovary syndrome. Cellular Physiology and Biochemistry 2017; 43:1880-1892.
86. Caglayan C, Kandemir FM, Yıldırım S, Kucukler S, Kılınc MA, Saglam YS. Zingerone ameliorates cisplatin‐induced ovarian and uterine toxicity via suppression of sex hormone imbalances, oxidative stress, inflammation and apoptosis in female wistar rats. Biomedicine & Pharmacotherapy 2018; 102:517-530.
87. Cortés-Rojo C, R Rodriguez-Orozco A. Importance of oxidative damage on the electron transport chain for the rational use of mitochondria-targeted antioxidants. Mini reviews in medicinal chemistry 2011; 11:625-632.
88. Tanabe M, Tamura H, Taketani T, Okada M, Lee L, Tamura I, et al. Melatonin protects the integrity of granulosa cells by reducing oxidative stress in nuclei, mitochondria, and plasma membranes in mice. Journal of Reproduction and Development 2014.
89. Park C, Choi EO, Kim GY, Hwang HJ, Kim BW, Yoo YH, et al. Protective Effect of Baicalein on Oxidative Stress-induced DNA Damage and Apoptosis in RT4-D6P2T Schwann Cells. Int J Med Sci 2019; 16:8-16.
90. Peitsch M, Polzar B, Stephan H, Crompton T, MacDonald HR, Mannherz H, et al. Characterization of the endogenous deoxyribonuclease involved in nuclear DNA degradation during apoptosis (programmed cell death). The EMBO journal 1993; 12:371-377.
91. Xiong Y, Zhao Q, Gu L, Liu C, Wang C. Shenqi Fuzheng Injection Reverses Cisplatin Resistance through Mitofusin-2-Mediated Cell Cycle Arrest and Apoptosis in A549/DDP Cells. Evidence-Based Complementary and Alternative Medicine 2018; 2018.
92. Desagher S, Martinou J-C. Mitochondria as the central control point of apoptosis. Trends in cell biology 2000; 10:369-377.
93. Jiang G-J, Pan L, Huang X-Y, Han M, Wen J-K, Sun F-Z. Expression of HSG is essential for mouse blastocyst formation. Biochemical and biophysical research communications 2005; 335:351-355.
94. Ngoh GA, Papanicolaou KN, Walsh K. Loss of mitofusin 2 promotes endoplasmic reticulum stress. Journal of Biological Chemistry 2012; 287:20321-20332.
95. Duarte A, Poderoso C, Cooke M, Soria G, Maciel FC, Gottifredi V, et al. Mitochondrial fusion is essential for steroid biosynthesis. PLoS One 2012; 7:e45829.
96. Karadeniz A, Simsek N, Karakus E, Yildirim S, Kara A, Can I, et al. Royal jelly modulates oxidative stress and apoptosis in liver and kidneys of rats treated with cisplatin. Oxidative medicine and cellular longevity 2011; 2011.
97. Pavel CI, Mărghitaş LA, Bobiş O, Dezmirean DS, Şapcaliu A, Radoi I, et al. Biological activities of royal jelly-review. Scientific Papers Animal Science and Biotechnologies 2011; 44:108-118.
98. Imai M, Qin J, Yamakawa N, Miyado K, Umezawa A, Takahashi Y. Molecular Alterations during Female Reproductive Aging: Can Aged Oocytes Remind Youth?  Embryology-Updates and Highlights on Classic Topics: IntechOpen; 2012.
99. Kamakura M. Royalactin induces queen differentiation in honeybees. Nature 2011; 473:478.
100. Husein M, Kridli R. Reproductive responses following royal jelly treatment administered orally or intramuscularly into progesterone-treated Awassi ewes. Animal reproduction science 2002; 74:45-53.
101. El-Nekeety AA, El-Kholy W, Abbas NF, Ebaid A, Amra HA, Abdel-Wahhab MA. Efficacy of royal jelly against the oxidative stress of fumonisin in rats. Toxicon 2007; 50:256-269.
102. You M-M, Chen Y-F, Pan Y-M, Liu Y-C, Tu J, Wang K, et al. Royal jelly attenuates LPS-induced inflammation in BV-2 microglial cells through modulating NF-κB and p38/JNK signaling pathways. Mediators of inflammation 2018; 2018.
103. Dinkova-Kostova AT, Talalay P. Persuasive evidence that quinone reductase type 1 (DT diaphorase) protects cells against the toxicity of electrophiles and reactive forms of oxygen. Free Radical Biology and Medicine 2000; 29:231-240.
104. Landi L, Fiorentini D, Galli MC, Segura-Aguilar J, Beyer RE. DT-Diaphorase maintains the reduced state of ubiquinones in lipid vesicles thereby promoting their antioxidant function. Free Radical Biology and Medicine 1997; 22:329-335.
105. Beyer RE, Segura-Aguilar J, Di Bernardo S, Cavazzoni M, Fato R, Fiorentini D, et al. The role of DT-diaphorase in the maintenance of the reduced antioxidant form of coenzyme Q in membrane systems. Proceedings of the National Academy of Sciences 1996; 93:2528-2532.
106. Vidoni S, Zanna C, Rugolo M, Sarzi E, Lenaers G. Why mitochondria must fuse to maintain their genome integrity. Antioxidants & redox signaling 2013; 19:379-388.
107. Wakai T, Harada Y, Miyado K, Kono T. Mitochondrial dynamics controlled by mitofusins define organelle positioning and movement during mouse oocyte maturation. Molecular human reproduction 2014; 20:1090-1100.
108. Zhou H, Hu S, Jin Q, Shi C, Zhang Y, Zhu P, et al. Mff‐Dependent Mitochondrial Fission Contributes to the Pathogenesis of Cardiac Microvasculature Ischemia/Reperfusion Injury via Induction of mROS‐Mediated Cardiolipin Oxidation and HK 2/VDAC 1 Disassociation‐Involved mPTP Opening. Journal of the American Heart Association 2017; 6:e005328.