Skip navigation

DSpace

機構典藏 DSpace 系統致力於保存各式數位資料(如:文字、圖片、PDF)並使其易於取用。

點此認識 DSpace
DSpace logo
English
中文
  • 瀏覽論文
    • 校院系所
    • 出版年
    • 作者
    • 標題
    • 關鍵字
    • 指導教授
  • 搜尋 TDR
  • 授權 Q&A
    • 我的頁面
    • 接受 E-mail 通知
    • 編輯個人資料
  1. NTU Theses and Dissertations Repository
  2. 生命科學院
  3. 基因體與系統生物學學位學程
請用此 Handle URI 來引用此文件: http://tdr.lib.ntu.edu.tw/jspui/handle/123456789/87364
完整後設資料紀錄
DC 欄位值語言
dc.contributor.advisor蘇剛毅zh_TW
dc.contributor.advisorKang-Yi Suen
dc.contributor.author廖耿楙zh_TW
dc.contributor.authorKeng-Mao Liaoen
dc.date.accessioned2023-05-18T17:18:10Z-
dc.date.available2025-07-31-
dc.date.copyright2023-06-14-
dc.date.issued2023-
dc.date.submitted2023-02-14-
dc.identifier.citationPart I:
1. Li, Z. et al. Aging and age-related diseases: from mechanisms to therapeutic strategies. Biogerontology 22, 165-187 (2021).
2. UN. World Population Ageing 2020 Highlights: Living arrangements of older persons. https://www.un.org/development/desa/pd/sites/www.un.org.development.desa.pd/files/files/documents/2020/Sep/un_pop_2020_pf_ageing_10_key_messages.pdf. Published 2020. Accessed.
3. Gasek, N.S., Kuchel, G.A., Kirkland, J.L. & Xu, M. Strategies for Targeting Senescent Cells in Human Disease. Nat Aging 1, 870-879 (2021).
4. Kumari, R. & Jat, P. Mechanisms of Cellular Senescence: Cell Cycle Arrest and Senescence Associated Secretory Phenotype. Front Cell Dev Biol 9, 645593 (2021).
5. Fischer, M. Census and evaluation of p53 target genes. Oncogene 36, 3943-3956 (2017).
6. Herranz, N. & Gil, J. Mechanisms and functions of cellular senescence. J Clin Invest 128, 1238-1246 (2018).
7. Mylonas, A. & O'Loghlen, A. Cellular Senescence and Ageing: Mechanisms and Interventions. Front Aging 3, 866718 (2022).
8. Roger, L., Tomas, F. & Gire, V. Mechanisms and Regulation of Cellular Senescence. Int J Mol Sci 22 (2021).
9. Coppe, J.P., Desprez, P.Y., Krtolica, A. & Campisi, J. The senescence-associated secretory phenotype: the dark side of tumor suppression. Annu Rev Pathol 5, 99-118 (2010).
10. Hudgins, A.D. et al. Age- and Tissue-Specific Expression of Senescence Biomarkers in Mice. Front Genet 9, 59 (2018).
11. Robbins, E., Levine, E.M. & Eagle, H. Morphologic changes accompanying senescence of cultured human diploid cells. J Exp Med 131, 1211-1222 (1970).
12. Kusumoto, D. et al. Anti-senescent drug screening by deep learning-based morphology senescence scoring. Nature Communications 12 (2021).
13. Yang, J. et al. Cell size and growth rate are major determinants of replicative lifespan. Cell Cycle 10, 144-155 (2011).
14. Kirsch, V., Ramge, J.M., Schoppa, A., Ignatius, A. & Riegger, J. In Vitro Characterization of Doxorubicin-Mediated Stress-Induced Premature Senescence in Human Chondrocytes. Cells 11 (2022).
15. Sapega, O. et al. Distinct phenotypes and 'bystander' effects of senescent tumour cells induced by docetaxel or immunomodulatory cytokines. Int J Oncol 53, 1997-2009 (2018).
16. Son, H.N., Chi, H.N.Q., Chung, D.C. & Long, L.T. Morphological changes during replicative senescence in bovine ovarian granulosa cells. Cell Cycle 18, 1490-1497 (2019).
17. Schmid, N. et al. Insights into replicative senescence of human testicular peritubular cells. Sci Rep 9, 15052 (2019).
18. Sherwood, S.W., Rush, D., Ellsworth, J.L. & Schimke, R.T. Defining cellular senescence in IMR-90 cells: a flow cytometric analysis. Proc Natl Acad Sci U S A 85, 9086-9090 (1988).
19. Neurohr, G.E. et al. Excessive Cell Growth Causes Cytoplasm Dilution And Contributes to Senescence. Cell 176, 1083-1097 e1018 (2019).
20. Lanz, M.C. et al. Increasing cell size remodels the proteome and promotes senescence. Mol Cell 82, 3255-3269 e3258 (2022).
21. Haynes, M.K. et al. Detection of intracellular granularity induction in prostate cancer cell lines by small molecules using the HyperCyt high-throughput flow cytometry system. J Biomol Screen 14, 596-609 (2009).
22. Bielak-Zmijewska, A. et al. A comparison of replicative senescence and doxorubicin-induced premature senescence of vascular smooth muscle cells isolated from human aorta. Biogerontology 15, 47-64 (2014).
23. Psaroudis, R.T. et al. CD26 is a senescence marker associated with reduced immunopotency of human adipose tissue-derived multipotent mesenchymal stromal cells. Stem Cell Res Ther 13, 358 (2022).
24. Kurz, D.J., Decary, S., Hong, Y. & Erusalimsky, J.D. Senescence-associated (beta)-galactosidase reflects an increase in lysosomal mass during replicative ageing of human endothelial cells. J Cell Sci 113 ( Pt 20), 3613-3622 (2000).
25. Gonzalez-Gualda, E., Baker, A.G., Fruk, L. & Munoz-Espin, D. A guide to assessing cellular senescence in vitro and in vivo. FEBS J 288, 56-80 (2021).
26. Bojko, A., Czarnecka-Herok, J., Charzynska, A., Dabrowski, M. & Sikora, E. Diversity of the Senescence Phenotype of Cancer Cells Treated with Chemotherapeutic Agents. Cells 8 (2019).
27. Dimri, G.P. et al. A biomarker that identifies senescent human cells in culture and in aging skin in vivo. Proc Natl Acad Sci U S A 92, 9363-9367 (1995).
28. Lee, B.Y. et al. Senescence-associated beta-galactosidase is lysosomal beta-galactosidase. Aging Cell 5, 187-195 (2006).
29. Chakravarti, D., LaBella, K.A. & DePinho, R.A. Telomeres: history, health, and hallmarks of aging. Cell 184, 306-322 (2021).
30. Hayflick, L. & Moorhead, P.S. The serial cultivation of human diploid cell strains. Exp Cell Res 25, 585-621 (1961).
31. Harley, C.B., Futcher, A.B. & Greider, C.W. Telomeres shorten during ageing of human fibroblasts. Nature 345, 458-460 (1990).
32. Herbig, U., Jobling, W.A., Chen, B.P., Chen, D.J. & Sedivy, J.M. Telomere shortening triggers senescence of human cells through a pathway involving ATM, p53, and p21(CIP1), but not p16(INK4a). Mol Cell 14, 501-513 (2004).
33. Serrano, M., Lin, A.W., McCurrach, M.E., Beach, D. & Lowe, S.W. Oncogenic ras provokes premature cell senescence associated with accumulation of p53 and p16INK4a. Cell 88, 593-602 (1997).
34. Baek, K.H. & Ryeom, S. Detection of Oncogene-Induced Senescence In Vivo. Methods Mol Biol 1534, 185-198 (2017).
35. Harman, D. Aging: a theory based on free radical and radiation chemistry. J Gerontol 11, 298-300 (1956).
36. Kodama, R. et al. ROS-generating oxidases Nox1 and Nox4 contribute to oncogenic Ras-induced premature senescence. Genes Cells 18, 32-41 (2013).
37. Chang, T.C., Hsu, M.F. & Wu, K.K. High glucose induces bone marrow-derived mesenchymal stem cell senescence by upregulating autophagy. PLoS One 10, e0126537 (2015).
38. Zhang, Y. et al. Mice deficient in both Mn superoxide dismutase and glutathione peroxidase-1 have increased oxidative damage and a greater incidence of pathology but no reduction in longevity. J Gerontol A Biol Sci Med Sci 64, 1212-1220 (2009).
39. Kwon, M.J., Lee, K.Y., Lee, H.W., Kim, J.H. & Kim, T.Y. SOD3 Variant, R213G, Altered SOD3 Function, Leading to ROS-Mediated Inflammation and Damage in Multiple Organs of Premature Aging Mice. Antioxid Redox Signal 23, 985-999 (2015).
40. Kawaguchi, K., Hashimoto, M. & Sugimoto, M. An antioxidant suppressed lung cellular senescence and enhanced pulmonary function in aged mice. Biochem Biophys Res Commun 541, 43-49 (2021).
41. Davalli, P., Mitic, T., Caporali, A., Lauriola, A. & D'Arca, D. ROS, Cell Senescence, and Novel Molecular Mechanisms in Aging and Age-Related Diseases. Oxid Med Cell Longev 2016, 3565127 (2016).
42. Osellame, L.D., Blacker, T.S. & Duchen, M.R. Cellular and molecular mechanisms of mitochondrial function. Best Pract Res Clin Endocrinol Metab 26, 711-723 (2012).
43. Correia-Melo, C. et al. Mitochondria are required for pro-ageing features of the senescent phenotype. EMBO J 35, 724-742 (2016).
44. Rambold, A.S., Kostelecky, B., Elia, N. & Lippincott-Schwartz, J. Tubular network formation protects mitochondria from autophagosomal degradation during nutrient starvation. Proc Natl Acad Sci U S A 108, 10190-10195 (2011).
45. Gomes, L.C. & Scorrano, L. Mitochondrial elongation during autophagy: a stereotypical response to survive in difficult times. Autophagy 7, 1251-1253 (2011).
46. Yao, C.H. et al. Mitochondrial fusion supports increased oxidative phosphorylation during cell proliferation. Elife 8 (2019).
47. Yoo, S.M. et al. FKBP8 LIRL-dependent mitochondrial fragmentation facilitates mitophagy under stress conditions. FASEB J 34, 2944-2957 (2020).
48. Taguchi, N., Ishihara, N., Jofuku, A., Oka, T. & Mihara, K. Mitotic phosphorylation of dynamin-related GTPase Drp1 participates in mitochondrial fission. J Biol Chem 282, 11521-11529 (2007).
49. Pisani, D.F. et al. Mitochondrial fission is associated with UCP1 activity in human brite/beige adipocytes. Mol Metab 7, 35-44 (2018).
50. Chen, H. et al. Mitofusins Mfn1 and Mfn2 coordinately regulate mitochondrial fusion and are essential for embryonic development. J Cell Biol 160, 189-200 (2003).
51. Kushnareva, Y.E. et al. Loss of OPA1 disturbs cellular calcium homeostasis and sensitizes for excitotoxicity. Cell Death Differ 20, 353-365 (2013).
52. Wu, W. et al. OPA1 overexpression ameliorates mitochondrial cristae remodeling, mitochondrial dysfunction, and neuronal apoptosis in prion diseases. Cell Death Dis 10, 710 (2019).
53. Davies, V.J. et al. Opa1 deficiency in a mouse model of autosomal dominant optic atrophy impairs mitochondrial morphology, optic nerve structure and visual function. Hum Mol Genet 16, 1307-1318 (2007).
54. Song, Z., Chen, H., Fiket, M., Alexander, C. & Chan, D.C. OPA1 processing controls mitochondrial fusion and is regulated by mRNA splicing, membrane potential, and Yme1L. J Cell Biol 178, 749-755 (2007).
55. Wang, R. et al. Identification of new OPA1 cleavage site reveals that short isoforms regulate mitochondrial fusion. Mol Biol Cell 32, 157-168 (2021).
56. Hu, C. et al. OPA1 and MICOS Regulate mitochondrial crista dynamics and formation. Cell Death Dis 11, 940 (2020).
57. Patten, D.A. et al. OPA1-dependent cristae modulation is essential for cellular adaptation to metabolic demand. EMBO J 33, 2676-2691 (2014).
58. Quintana-Cabrera, R. et al. The cristae modulator Optic atrophy 1 requires mitochondrial ATP synthase oligomers to safeguard mitochondrial function. Nat Commun 9, 3399 (2018).
59. Loson, O.C., Song, Z., Chen, H. & Chan, D.C. Fis1, Mff, MiD49, and MiD51 mediate Drp1 recruitment in mitochondrial fission. Mol Biol Cell 24, 659-667 (2013).
60. Hu, J. et al. ROCK1 activation-mediated mitochondrial translocation of Drp1 and cofilin are required for arnidiol-induced mitochondrial fission and apoptosis. J Exp Clin Cancer Res 39, 37 (2020).
61. Chang, C.R. & Blackstone, C. Dynamic regulation of mitochondrial fission through modification of the dynamin-related protein Drp1. Ann N Y Acad Sci 1201, 34-39 (2010).
62. Jin, J.Y., Wei, X.X., Zhi, X.L., Wang, X.H. & Meng, D. Drp1-dependent mitochondrial fission in cardiovascular disease. Acta Pharmacol Sin 42, 655-664 (2021).
63. Yu, T., Jhun, B.S. & Yoon, Y. High-glucose stimulation increases reactive oxygen species production through the calcium and mitogen-activated protein kinase-mediated activation of mitochondrial fission. Antioxid Redox Signal 14, 425-437 (2011).
64. Qi, X., Disatnik, M.H., Shen, N., Sobel, R.A. & Mochly-Rosen, D. Aberrant mitochondrial fission in neurons induced by protein kinase Cdelta under oxidative stress conditions in vivo. Mol Biol Cell 22, 256-265 (2011).
65. Bo, T. et al. Calmodulin-dependent protein kinase II (CaMKII) mediates radiation-induced mitochondrial fission by regulating the phosphorylation of dynamin-related protein 1 (Drp1) at serine 616. Biochem Biophys Res Commun 495, 1601-1607 (2018).
66. Brand, C.S., Tan, V.P., Brown, J.H. & Miyamoto, S. RhoA regulates Drp1 mediated mitochondrial fission through ROCK to protect cardiomyocytes. Cell Signal 50, 48-57 (2018).
67. Jahani-Asl, A. & Slack, R.S. The phosphorylation state of Drp1 determines cell fate. EMBO Rep 8, 912-913 (2007).
68. Cereghetti, G.M. et al. Dephosphorylation by calcineurin regulates translocation of Drp1 to mitochondria. Proc Natl Acad Sci U S A 105, 15803-15808 (2008).
69. Jhun, B.S. et al. Protein kinase D activation induces mitochondrial fragmentation and dysfunction in cardiomyocytes. J Physiol 596, 827-855 (2018).
70. Xu, S. et al. CaMKII induces permeability transition through Drp1 phosphorylation during chronic beta-AR stimulation. Nat Commun 7, 13189 (2016).
71. Wasiak, S., Zunino, R. & McBride, H.M. Bax/Bak promote sumoylation of DRP1 and its stable association with mitochondria during apoptotic cell death. J Cell Biol 177, 439-450 (2007).
72. Prudent, J. et al. MAPL SUMOylation of Drp1 Stabilizes an ER/Mitochondrial Platform Required for Cell Death. Mol Cell 59, 941-955 (2015).
73. Guo, C., Wilkinson, K.A., Evans, A.J., Rubin, P.P. & Henley, J.M. SENP3-mediated deSUMOylation of Drp1 facilitates interaction with Mff to promote cell death. Sci Rep 7, 43811 (2017).
74. Guo, C. et al. SENP3-mediated deSUMOylation of dynamin-related protein 1 promotes cell death following ischaemia. EMBO J 32, 1514-1528 (2013).
75. Rape, M. Ubiquitylation at the crossroads of development and disease. Nat Rev Mol Cell Biol 19, 59-70 (2018).
76. Lavie, J. et al. Ubiquitin-Dependent Degradation of Mitochondrial Proteins Regulates Energy Metabolism. Cell Rep 23, 2852-2863 (2018).
77. Yonashiro, R. et al. A novel mitochondrial ubiquitin ligase plays a critical role in mitochondrial dynamics. EMBO J 25, 3618-3626 (2006).
78. Karbowski, M., Neutzner, A. & Youle, R.J. The mitochondrial E3 ubiquitin ligase MARCH5 is required for Drp1 dependent mitochondrial division. J Cell Biol 178, 71-84 (2007).
79. Das, R., Kamal, I.M., Das, S., Chakrabarti, S. & Chakrabarti, O. MITOL-mediated DRP1 ubiquitylation and degradation promotes mitochondrial hyperfusion in a CMT2A-linked MFN2 mutant. J Cell Sci 135 (2022).
80. Wang, H. et al. Parkin ubiquitinates Drp1 for proteasome-dependent degradation: implication of dysregulated mitochondrial dynamics in Parkinson disease. J Biol Chem 286, 11649-11658 (2011).
81. Zhang, Y., Qin, Z., Sun, W., Chu, F. & Zhou, F. Function of Protein S-Palmitoylation in Immunity and Immune-Related Diseases. Front Immunol 12, 661202 (2021).
82. Napoli, E. et al. Zdhhc13-dependent Drp1 S-palmitoylation impacts brain bioenergetics, anxiety, coordination and motor skills. Sci Rep 7, 12796 (2017).
83. Cho, D.H. et al. S-nitrosylation of Drp1 mediates beta-amyloid-related mitochondrial fission and neuronal injury. Science 324, 102-105 (2009).
84. Lee, D.S. & Kim, J.E. PDI-mediated S-nitrosylation of DRP1 facilitates DRP1-S616 phosphorylation and mitochondrial fission in CA1 neurons. Cell Death Dis 9, 869 (2018).
85. Zhao, L., Feng, Z., Yang, X. & Liu, J. The regulatory roles of O-GlcNAcylation in mitochondrial homeostasis and metabolic syndrome. Free Radic Res 50, 1080-1088 (2016).
86. Gawlowski, T. et al. Modulation of dynamin-related protein 1 (DRP1) function by increased O-linked-beta-N-acetylglucosamine modification (O-GlcNAc) in cardiac myocytes. J Biol Chem 287, 30024-30034 (2012).
87. Park, S.J. et al. Increased O-GlcNAcylation of Drp1 by amyloid-beta promotes mitochondrial fission and dysfunction in neuronal cells. Mol Brain 14, 6 (2021).
88. Toyama, E.Q. et al. Metabolism. AMP-activated protein kinase mediates mitochondrial fission in response to energy stress. Science 351, 275-281 (2016).
89. Yu, Y. et al. Fis1 phosphorylation by Met promotes mitochondrial fission and hepatocellular carcinoma metastasis. Signal Transduct Target Ther 6, 401 (2021).
90. Denoth Lippuner, A., Julou, T. & Barral, Y. Budding yeast as a model organism to study the effects of age. FEMS Microbiol Rev 38, 300-325 (2014).
91. Kitagaki, H., Araki, Y., Funato, K. & Shimoi, H. Ethanol-induced death in yeast exhibits features of apoptosis mediated by mitochondrial fission pathway. FEBS Lett 581, 2935-2942 (2007).
92. Piper, P.W. et al. The shortened replicative life span of prohibitin mutants of yeast appears to be due to defective mitochondrial segregation in old mother cells. Aging Cell 1, 149-157 (2002).
93. Scheckhuber, C.Q. et al. Reducing mitochondrial fission results in increased life span and fitness of two fungal ageing models. Nat Cell Biol 9, 99-105 (2007).
94. Palermo, V., Falcone, C. & Mazzoni, C. Apoptosis and aging in mitochondrial morphology mutants of S. cerevisiae. Folia Microbiol (Praha) 52, 479-483 (2007).
95. Cheng, W.C. et al. Fis1 deficiency selects for compensatory mutations responsible for cell death and growth control defects. Cell Death Differ 15, 1838-1846 (2008).
96. Palermo, V., Falcone, C., Calvani, M. & Mazzoni, C. Acetyl-L-carnitine protects yeast cells from apoptosis and aging and inhibits mitochondrial fission. Aging Cell 9, 570-579 (2010).
97. Regmi, S.G., Rolland, S.G. & Conradt, B. Age-dependent changes in mitochondrial morphology and volume are not predictors of lifespan. Aging (Albany NY) 6, 118-130 (2014).
98. Byrne, J.J. et al. Disruption of mitochondrial dynamics affects behaviour and lifespan in Caenorhabditis elegans. Cell Mol Life Sci 76, 1967-1985 (2019).
99. Chaudhari, S.N. & Kipreos, E.T. Increased mitochondrial fusion allows the survival of older animals in diverse C. elegans longevity pathways. Nat Commun 8, 182 (2017).
100. Princz, A., Pelisch, F. & Tavernarakis, N. SUMO promotes longevity and maintains mitochondrial homeostasis during ageing in Caenorhabditis elegans. Sci Rep 10, 15513 (2020).
101. Ryu, D. et al. Urolithin A induces mitophagy and prolongs lifespan in C. elegans and increases muscle function in rodents. Nat Med 22, 879-888 (2016).
102. Rana, A., Rera, M. & Walker, D.W. Parkin overexpression during aging reduces proteotoxicity, alters mitochondrial dynamics, and extends lifespan. Proc Natl Acad Sci U S A 110, 8638-8643 (2013).
103. Lee, T.T. et al. Loss of Fis1 impairs proteostasis during skeletal muscle aging in Drosophila. Aging Cell 20, e13379 (2021).
104. Rana, A. et al. Promoting Drp1-mediated mitochondrial fission in midlife prolongs healthy lifespan of Drosophila melanogaster. Nat Commun 8, 448 (2017).
105. Mai, S., Klinkenberg, M., Auburger, G., Bereiter-Hahn, J. & Jendrach, M. Decreased expression of Drp1 and Fis1 mediates mitochondrial elongation in senescent cells and enhances resistance to oxidative stress through PINK1. J Cell Sci 123, 917-926 (2010).
106. Park, Y.Y. et al. Loss of MARCH5 mitochondrial E3 ubiquitin ligase induces cellular senescence through dynamin-related protein 1 and mitofusin 1. J Cell Sci 123, 619-626 (2010).
107. Kim, Y.Y. et al. p53 regulates mitochondrial dynamics by inhibiting Drp1 translocation into mitochondria during cellular senescence. FASEB J 34, 2451-2464 (2020).
108. Hubackova, S. et al. Selective elimination of senescent cells by mitochondrial targeting is regulated by ANT2. Cell Death Differ 26, 276-290 (2019).
109. Yoon, Y.S. et al. Formation of elongated giant mitochondria in DFO-induced cellular senescence: involvement of enhanced fusion process through modulation of Fis1. J Cell Physiol 209, 468-480 (2006).
110. Son, J.M. et al. Mitofusin 1 and optic atrophy 1 shift metabolism to mitochondrial respiration during aging. Aging Cell 16, 1136-1145 (2017).
111. Wang, D. et al. Apoptotic transition of senescent cells accompanied with mitochondrial hyper-function. Oncotarget 7, 28286-28300 (2016).
112. Fujita, Y., Iketani, M., Ito, M. & Ohsawa, I. Temporal changes in mitochondrial function and reactive oxygen species generation during the development of replicative senescence in human fibroblasts. Exp Gerontol 165, 111866 (2022).
113. Yu, B. et al. Mitochondrial phosphatase PGAM5 modulates cellular senescence by regulating mitochondrial dynamics. Nat Commun 11, 2549 (2020).
114. Baker, D.J. et al. Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders. Nature 479, 232-236 (2011).
115. Lafontaine, J. et al. Senolytic Targeting of Bcl-2 Anti-Apoptotic Family Increases Cell Death in Irradiated Sarcoma Cells. Cancers (Basel) 13 (2021).
116. Fuhrmann-Stroissnigg, H. et al. Identification of HSP90 inhibitors as a novel class of senolytics. Nat Commun 8, 422 (2017).
117. Kirkland, J.L. & Tchkonia, T. Senolytic drugs: from discovery to translation. J Intern Med 288, 518-536 (2020).
118. Lamming, D.W., Ye, L., Sabatini, D.M. & Baur, J.A. Rapalogs and mTOR inhibitors as anti-aging therapeutics. J Clin Invest 123, 980-989 (2013).
119. Tilstra, J.S. et al. NF-kappaB inhibition delays DNA damage-induced senescence and aging in mice. J Clin Invest 122, 2601-2612 (2012).
120. Wang, Y.W. et al. Metformin: a review of its potential indications. Drug Des Devel Ther 11, 2421-2429 (2017).
121. Martin-Montalvo, A. et al. Metformin improves healthspan and lifespan in mice. Nat Commun 4, 2192 (2013).
122. Amor, C. et al. Senolytic CAR T cells reverse senescence-associated pathologies. Nature 583, 127-132 (2020).
123. Guderyon, M.J. et al. Mobilization-based transplantation of young-donor hematopoietic stem cells extends lifespan in mice. Aging Cell 19, e13110 (2020).
124. Shyu, Y.C. et al. Genetic Disruption of KLF1 K74 SUMOylation in Hematopoietic System Promotes Healthy Longevity in Mice. Adv Sci (Weinh) 9, e2201409 (2022).
125. Lee, Y.H. et al. Targeting Mitochondrial Metabolism as a Strategy to Treat Senescence. Cells 10 (2021).
126. Ren, X. et al. Resveratrol Ameliorates Mitochondrial Elongation via Drp1/Parkin/PINK1 Signaling in Senescent-Like Cardiomyocytes. Oxid Med Cell Longev 2017, 4175353 (2017).
127. Kuang, Y. et al. Metformin prevents against oxidative stress-induced senescence in human periodontal ligament cells. Biogerontology 21, 13-27 (2020).
128. Wang, Y. et al. Metformin Improves Mitochondrial Respiratory Activity through Activation of AMPK. Cell Rep 29, 1511-1523 e1515 (2019).
129. Li, M., You, L., Xue, J. & Lu, Y. Ionizing Radiation-Induced Cellular Senescence in Normal, Non-transformed Cells and the Involved DNA Damage Response: A Mini Review. Front Pharmacol 9, 522 (2018).
130. Feng, M. et al. Aspirin ameliorates the long-term adverse effects of doxorubicin through suppression of cellular senescence. FASEB Bioadv 1, 579-590 (2019).
131. Masterson, J.C. & O'Dea, S. 5-Bromo-2-deoxyuridine activates DNA damage signalling responses and induces a senescence-like phenotype in p16-null lung cancer cells. Anticancer Drugs 18, 1053-1068 (2007).
132. Marazita, M.C., Dugour, A., Marquioni-Ramella, M.D., Figueroa, J.M. & Suburo, A.M. Oxidative stress-induced premature senescence dysregulates VEGF and CFH expression in retinal pigment epithelial cells: Implications for Age-related Macular Degeneration. Redox Biol 7, 78-87 (2016).
133. Liu, X.L., Ding, J. & Meng, L.H. Oncogene-induced senescence: a double edged sword in cancer. Acta Pharmacol Sin 39, 1553-1558 (2018).
134. Harrison, K.F.J.M.C.D.E. The Mouse in Biomedical Research: Chapter 20-Mouse Model in Aging Research. 639-668.
135. Xu, Q. et al. The flavonoid procyanidin C1 has senotherapeutic activity and increases lifespan in mice. Nat Metab 3, 1706-1726 (2021).
136. Hashimoto, M. et al. Elimination of p19(ARF)-expressing cells enhances pulmonary function in mice. JCI Insight 1, e87732 (2016).
137. Schafer, M.J. et al. Cellular senescence mediates fibrotic pulmonary disease. Nat Commun 8, 14532 (2017).
138. Cai, Y. et al. Elimination of senescent cells by beta-galactosidase-targeted prodrug attenuates inflammation and restores physical function in aged mice. Cell Res 30, 574-589 (2020).
139. Zhang, L. et al. Fisetin Alleviated Bleomycin-Induced Pulmonary Fibrosis Partly by Rescuing Alveolar Epithelial Cells From Senescence. Front Pharmacol 11, 553690 (2020).
140. Zhang, X. et al. Roxithromycin attenuates bleomycin-induced pulmonary fibrosis by targeting senescent cells. Acta Pharmacol Sin 42, 2058-2068 (2021).
141. Xu, M. et al. Transplanted Senescent Cells Induce an Osteoarthritis-Like Condition in Mice. J Gerontol A Biol Sci Med Sci 72, 780-785 (2017).
142. Kim, S.R. et al. Transplanted senescent renal scattered tubular-like cells induce injury in the mouse kidney. Am J Physiol Renal Physiol 318, F1167-F1176 (2020).
143. Xu, M. et al. Senolytics improve physical function and increase lifespan in old age. Nat Med 24, 1246-1256 (2018).
144. Zavadskis, S. et al. Effect of Diphenyleneiodonium Chloride on Intracellular Reactive Oxygen Species Metabolism with Emphasis on NADPH Oxidase and Mitochondria in Two Therapeutically Relevant Human Cell Types. Pharmaceutics 13 (2020).
145. Stuehr, D.J. et al. Inhibition of macrophage and endothelial cell nitric oxide synthase by diphenyleneiodonium and its analogs. FASEB J 5, 98-103 (1991).
146. Suzuki, H. et al. The NADPH oxidase inhibitor diphenyleneiodonium activates the human TRPA1 nociceptor. Am J Physiol Cell Physiol 307, C384-394 (2014).
147. Ye, C. et al. Identification of a novel small-molecule agonist for human G protein-coupled receptor 3. J Pharmacol Exp Ther 349, 437-443 (2014).
148. Li, Y. & Trush, M.A. Diphenyleneiodonium, an NAD(P)H oxidase inhibitor, also potently inhibits mitochondrial reactive oxygen species production. Biochem Biophys Res Commun 253, 295-299 (1998).
149. Guo, B. et al. Expression, regulation and function of Egr1 during implantation and decidualization in mice. Cell Cycle 13, 2626-2640 (2014).
150. Ju, W.K. et al. Intraocular pressure elevation induces mitochondrial fission and triggers OPA1 release in glaucomatous optic nerve. Invest Ophthalmol Vis Sci 49, 4903-4911 (2008).
151. Kitaoka, Y. et al. Effects of Nrf2 deficiency on mitochondrial oxidative stress in aged skeletal muscle. Physiol Rep 7, e13998 (2019).
152. Massey, N. et al. Organic dust-induced mitochondrial dysfunction could be targeted via cGAS-STING or cytoplasmic NOX-2 inhibition using microglial cells and brain slice culture models. Cell Tissue Res 384, 465-486 (2021).
153. Vercellino, I. & Sazanov, L.A. The assembly, regulation and function of the mitochondrial respiratory chain. Nat Rev Mol Cell Biol 23, 141-161 (2022).
154. Ross, H.H. et al. Bromodeoxyuridine induces senescence in neural stem and progenitor cells. Stem Cells 26, 3218-3227 (2008).
155. Biran, A. et al. Quantitative identification of senescent cells in aging and disease. Aging Cell 16, 661-671 (2017).
156. Yang, H., Wang, H., Ren, J., Chen, Q. & Chen, Z.J. cGAS is essential for cellular senescence. Proc Natl Acad Sci U S A 114, E4612-E4620 (2017).
157. Xu, S. et al. MicroRNA-33 promotes the replicative senescence of mouse embryonic fibroblasts by suppressing CDK6. Biochem Biophys Res Commun 473, 1064-1070 (2016).
158. Sheedfar, F., Di Biase, S., Koonen, D. & Vinciguerra, M. Liver diseases and aging: friends or foes? Aging Cell 12, 950-954 (2013).
159. Naik, P.N. et al. Pulmonary fibrosis induced by gamma-herpesvirus in aged mice is associated with increased fibroblast responsiveness to transforming growth factor-beta. J Gerontol A Biol Sci Med Sci 67, 714-725 (2012).
160. WHO. Ageing and health (2021). https://www.who.int/News-Room/Fact-Sheets/Detail/Ageing-and-Health
161. Kimberg, D.V., Loud, A.V. & Wiener, J. Cortisone-induced alterations in mitochondrial function and structure. J Cell Biol 37, 63-79 (1968).
162. Zeng, K.W. et al. Small molecule induces mitochondrial fusion for neuroprotection via targeting CK2 without affecting its conventional kinase activity. Signal Transduct Target Ther 6, 71 (2021).
163. Chen, M. et al. Mitophagy receptor FUNDC1 regulates mitochondrial dynamics and mitophagy. Autophagy 12, 689-702 (2016).
164. Citrin, D.E. et al. Role of type II pneumocyte senescence in radiation-induced lung fibrosis. J Natl Cancer Inst 105, 1474-1484 (2013).
165. Lyublinskaya, O. et al. Induction of Premature Cell Senescence Stimulated by High Doses of Antioxidants Is Mediated by Endoplasmic Reticulum Stress. Int J Mol Sci 22 (2021).
166. Wang, H. et al. Lung specific homing of diphenyleneiodonium chloride improves pulmonary fibrosis by inhibiting macrophage M2 metabolic program. J Adv Res 44, 213-225 (2023).
167. Kim, S.J. et al. Mitochondrial peptides modulate mitochondrial function during cellular senescence. Aging (Albany NY) 10, 1239-1256 (2018).
168. Wang, R. et al. Rapamycin inhibits the secretory phenotype of senescent cells by a Nrf2-independent mechanism. Aging Cell 16, 564-574 (2017).
169. Mishra, P. & Chan, D.C. Mitochondrial dynamics and inheritance during cell division, development and disease. Nat Rev Mol Cell Biol 15, 634-646 (2014).
170. Duvezin-Caubet, S. et al. Proteolytic processing of OPA1 links mitochondrial dysfunction to alterations in mitochondrial morphology. J Biol Chem 281, 37972-37979 (2006).
171. Adaniya, S.M., J, O.U., Cypress, M.W., Kusakari, Y. & Jhun, B.S. Posttranslational modifications of mitochondrial fission and fusion proteins in cardiac physiology and pathophysiology. Am J Physiol Cell Physiol 316, C583-C604 (2019).
172. Gardner, S.E., Humphry, M., Bennett, M.R. & Clarke, M.C. Senescent Vascular Smooth Muscle Cells Drive Inflammation Through an Interleukin-1alpha-Dependent Senescence-Associated Secretory Phenotype. Arterioscler Thromb Vasc Biol 35, 1963-1974 (2015).
173. Aquino-Martinez, R. et al. Senescent cells exacerbate chronic inflammation and contribute to periodontal disease progression in old mice. J Periodontol 92, 1483-1495 (2021).
174. Gao, L.B. et al. Identification of a small molecule SR9009 that activates NRF2 to counteract cellular senescence. Aging Cell, e13483 (2021).
175. Hara, H. et al. Mitochondrial fragmentation in cigarette smoke-induced bronchial epithelial cell senescence. Am J Physiol Lung Cell Mol Physiol 305, L737-746 (2013).
176. Kamogashira, T., Hayashi, K., Fujimoto, C., Iwasaki, S. & Yamasoba, T. Functionally and morphologically damaged mitochondria observed in auditory cells under senescence-inducing stress. NPJ Aging Mech Dis 3, 2 (2017).
177. Weir, H.J. et al. Dietary Restriction and AMPK Increase Lifespan via Mitochondrial Network and Peroxisome Remodeling. Cell Metab 26, 884-896 e885 (2017).
178. McCann, K.J. et al. IFNgamma regulates NAD+ metabolism to promote the respiratory burst in human monocytes. Blood Adv 6, 3821-3834 (2022).
179. Shi, L. et al. Cellular senescence induced by S100A9 in mesenchymal stromal cells through NLRP3 inflammasome activation. Aging (Albany NY) 11, 9626-9642 (2019).
180. Kornienko, J.S. et al. High doses of synthetic antioxidants induce premature senescence in cultivated mesenchymal stem cells. Sci Rep 9, 1296 (2019).
Part II:
1. Behringer, R.R., Mathews, L.S., Palmiter, R.D. & Brinster, R.L. Dwarf mice produced by genetic ablation of growth hormone-expressing cells. Genes Dev 2, 453-461 (1988).
2. Dehkhoda, F., Lee, C.M.M., Medina, J. & Brooks, A.J. The Growth Hormone Receptor: Mechanism of Receptor Activation, Cell Signaling, and Physiological Aspects. Front Endocrinol (Lausanne) 9, 35 (2018).
3. Yakar, S. et al. Serum complexes of insulin-like growth factor-1 modulate skeletal integrity and carbohydrate metabolism. FASEB J 23, 709-719 (2009).
4. Able, A.A., Burrell, J.A. & Stephens, J.M. STAT5-Interacting Proteins: A Synopsis of Proteins that Regulate STAT5 Activity. Biology (Basel) 6 (2017).
5. Yu, C.L., Jin, Y.J. & Burakoff, S.J. Cytosolic tyrosine dephosphorylation of STAT5. Potential role of SHP-2 in STAT5 regulation. J Biol Chem 275, 599-604 (2000).
6. Hennighausen, L. & Robinson, G.W. Interpretation of cytokine signaling through the transcription factors STAT5A and STAT5B. Genes Dev 22, 711-721 (2008).
7. Zhou, Y. et al. A mammalian model for Laron syndrome produced by targeted disruption of the mouse growth hormone receptor/binding protein gene (the Laron mouse). Proc Natl Acad Sci U S A 94, 13215-13220 (1997).
8. Fan, Y. et al. Liver-specific deletion of the growth hormone receptor reveals essential role of growth hormone signaling in hepatic lipid metabolism. J Biol Chem 284, 19937-19944 (2009).
9. List, E.O. et al. Liver-specific GH receptor gene-disrupted (LiGHRKO) mice have decreased endocrine IGF-I, increased local IGF-I, and altered body size, body composition, and adipokine profiles. Endocrinology 155, 1793-1805 (2014).
10. Cordoba-Chacon, J. et al. Growth Hormone Inhibits Hepatic De Novo Lipogenesis in Adult Mice. Diabetes 64, 3093-3103 (2015).
11. Fang, F., Shi, X., Brown, M.S., Goldstein, J.L. & Liang, G. Growth hormone acts on liver to stimulate autophagy, support glucose production, and preserve blood glucose in chronically starved mice. Proc Natl Acad Sci U S A 116, 7449-7454 (2019).
12. Seguy, D., Vahedi, K., Kapel, N., Souberbielle, J.C. & Messing, B. Low-dose growth hormone in adult home parenteral nutrition-dependent short bowel syndrome patients: a positive study. Gastroenterology 124, 293-302 (2003).
13. Tavakkolizadeh, A. et al. Effect of growth hormone on intestinal Na+/glucose cotransporter activity. JPEN J Parenter Enteral Nutr 25, 18-22 (2001).
14. Denson, L.A. et al. A randomized controlled trial of growth hormone in active pediatric Crohn disease. J Pediatr Gastroenterol Nutr 51, 130-139 (2010).
15. Slonim, A.E. et al. A preliminary study of growth hormone therapy for Crohn's disease. N Engl J Med 342, 1633-1637 (2000).
16. Young, J.A. et al. Characterization of an intestine-specific GH receptor knockout (IntGHRKO) mouse. Growth Horm IGF Res 46-47, 5-15 (2019).
17. Egecioglu, E. et al. Growth hormone receptor deficiency results in blunted ghrelin feeding response, obesity, and hypolipidemia in mice. Am J Physiol Endocrinol Metab 290, E317-325 (2006).
18. Bonkowski, M.S. et al. Long-lived growth hormone receptor knockout mice show a delay in age-related changes of body composition and bone characteristics. J Gerontol A Biol Sci Med Sci 61, 562-567 (2006).
19. List, E.O. et al. The role of GH in adipose tissue: lessons from adipose-specific GH receptor gene-disrupted mice. Mol Endocrinol 27, 524-535 (2013).
20. List, E.O. et al. Adipocyte-Specific GH Receptor-Null (AdGHRKO) Mice Have Enhanced Insulin Sensitivity With Reduced Liver Triglycerides. Endocrinology 160, 68-80 (2019).
21. Ran, L. et al. Loss of Adipose Growth Hormone Receptor in Mice Enhances Local Fatty Acid Trapping and Impairs Brown Adipose Tissue Thermogenesis. iScience 16, 106-121 (2019).
22. Castro, J.R. et al. Expression of growth hormone receptor in the human brain. Neurosci Lett 281, 147-150 (2000).
23. Furigo, I.C., Metzger, M., Teixeira, P.D., Soares, C.R. & Donato, J., Jr. Distribution of growth hormone-responsive cells in the mouse brain. Brain Struct Funct 222, 341-363 (2017).
24. Caputo, M. et al. Regulation of GH and GH Signaling by Nutrients. Cells 10 (2021).
25. Zhao, T.J. et al. Ghrelin O-acyltransferase (GOAT) is essential for growth hormone-mediated survival of calorie-restricted mice. Proc Natl Acad Sci U S A 107, 7467-7472 (2010).
26. Furigo, I.C. et al. Growth hormone regulates neuroendocrine responses to weight loss via AgRP neurons. Nat Commun 10, 662 (2019).
27. List, E.O. et al. Removal of growth hormone receptor (GHR) in muscle of male mice replicates some of the health benefits seen in global GHR-/- mice. Aging (Albany NY) 7, 500-512 (2015).
28. Lupu, F., Terwilliger, J.D., Lee, K., Segre, G.V. & Efstratiadis, A. Roles of growth hormone and insulin-like growth factor 1 in mouse postnatal growth. Dev Biol 229, 141-162 (2001).
29. Holmes, S.J., Economou, G., Whitehouse, R.W., Adams, J.E. & Shalet, S.M. Reduced bone mineral density in patients with adult onset growth hormone deficiency. J Clin Endocrinol Metab 78, 669-674 (1994).
30. Singhal, V., Goh, B.C., Bouxsein, M.L., Faugere, M.C. & DiGirolamo, D.J. Osteoblast-restricted Disruption of the Growth Hormone Receptor in Mice Results in Sexually Dimorphic Skeletal Phenotypes. Bone Res 1, 85-97 (2013).
31. Liu, Z. et al. DMP-1-mediated Ghr gene recombination compromises skeletal development and impairs skeletal response to intermittent PTH. FASEB J 30, 635-652 (2016).
32. Kim, H. et al. Structural basis for assembly and disassembly of the IGF/IGFBP/ALS ternary complex. Nat Commun 13, 4434 (2022).
33. Svegliati-Baroni, G. et al. Insulin and insulin-like growth factor-1 stimulate proliferation and type I collagen accumulation by human hepatic stellate cells: differential effects on signal transduction pathways. Hepatology 29, 1743-1751 (1999).
34. Huat, T.J. et al. IGF-1 enhances cell proliferation and survival during early differentiation of mesenchymal stem cells to neural progenitor-like cells. BMC Neurosci 15, 91 (2014).
35. Hakuno, F. & Takahashi, S.I. IGF1 receptor signaling pathways. J Mol Endocrinol 61, T69-T86 (2018).
36. Liu, J.P., Baker, J., Perkins, A.S., Robertson, E.J. & Efstratiadis, A. Mice carrying null mutations of the genes encoding insulin-like growth factor I (Igf-1) and type 1 IGF receptor (Igf1r). Cell 75, 59-72 (1993).
37. Powell-Braxton, L. et al. IGF-I is required for normal embryonic growth in mice. Genes Dev 7, 2609-2617 (1993).
38. Sjogren, K. et al. Liver-derived insulin-like growth factor I (IGF-I) is the principal source of IGF-I in blood but is not required for postnatal body growth in mice. Proc Natl Acad Sci U S A 96, 7088-7092 (1999).
39. Yakar, S. et al. Normal growth and development in the absence of hepatic insulin-like growth factor I. Proc Natl Acad Sci U S A 96, 7324-7329 (1999).
40. Stratikopoulos, E., Szabolcs, M., Dragatsis, I., Klinakis, A. & Efstratiadis, A. The hormonal action of IGF1 in postnatal mouse growth. Proc Natl Acad Sci U S A 105, 19378-19383 (2008).
41. Holzenberger, M. et al. IGF-1 receptor regulates lifespan and resistance to oxidative stress in mice. Nature 421, 182-187 (2003).
42. Boucher, J., Tseng, Y.H. & Kahn, C.R. Insulin and insulin-like growth factor-1 receptors act as ligand-specific amplitude modulators of a common pathway regulating gene transcription. J Biol Chem 285, 17235-17245 (2010).
43. O'Neill, B.T. et al. Differential Role of Insulin/IGF-1 Receptor Signaling in Muscle Growth and Glucose Homeostasis. Cell Rep 11, 1220-1235 (2015).
44. Goldstein, J.L. et al. Surviving starvation: essential role of the ghrelin-growth hormone axis. Cold Spring Harb Symp Quant Biol 76, 121-127 (2011).
45. Gahete, M.D. et al. Insulin and IGF-I inhibit GH synthesis and release in vitro and in vivo by separate mechanisms. Endocrinology 154, 2410-2420 (2013).
46. Inagaki, T. et al. Inhibition of growth hormone signaling by the fasting-induced hormone FGF21. Cell Metab 8, 77-83 (2008).
47. Yamamoto, M. et al. SIRT1 regulates adaptive response of the growth hormone--insulin-like growth factor-I axis under fasting conditions in liver. Proc Natl Acad Sci U S A 110, 14948-14953 (2013).
48. Nakagawa, Y. et al. Starvation-induced transcription factor CREBH negatively governs body growth by controlling GH signaling. FASEB J 35, e21663 (2021).
49. Wan, X. et al. Dietary protein-induced hepatic IGF-1 secretion mediated by PPARgamma activation. PLoS One 12, e0173174 (2017).
50. Tsugawa, Y., Handa, H. & Imai, T. Arginine induces IGF-1 secretion from the endoplasmic reticulum. Biochem Biophys Res Commun 514, 1128-1132 (2019).
51. Chen, L., Chen, Q., Rong, P., Wang, H.Y. & Chen, S. The energy sensing LKB1-AMPKalpha1 pathway regulates IGF1 secretion and consequent activation of the IGF1R-PKB pathway in primary hepatocytes. FEBS J 284, 2096-2109 (2017).
52. Zhang, W., Wang, Q., Song, P. & Zou, M.H. Liver kinase b1 is required for white adipose tissue growth and differentiation. Diabetes 62, 2347-2358 (2013).
53. Guo, L. et al. Cell non-autonomous regulation of hepatic IGF-1 and neonatal growth by Kinase Suppressor of Ras 2 (KSR2). Sci Rep 6, 32093 (2016).
54. Wang, T. et al. GDF15 is a heart-derived hormone that regulates body growth. EMBO Mol Med (2017).
55. Nutzenadel, W. Failure to thrive in childhood. Dtsch Arztebl Int 108, 642-649 (2011).
56. Homan, G.J. Failure to Thrive: A Practical Guide. Am Fam Physician 94, 295-299 (2016).
57. Fazeli, P.K. & Klibanski, A. Determinants of GH resistance in malnutrition. J Endocrinol 220, R57-65 (2014).
58. Mullis, P.E. Genetics of isolated growth hormone deficiency. J Clin Res Pediatr Endocrinol 2, 52-62 (2010).
59. Losa, M., Gatti, E., Rossini, A. & Lanzi, R. Replacement therapy with growth hormone and pituitary tumor recurrence: the relevance of the problem. J Endocrinol Invest 31, 75-78 (2008).
60. Darzy, K.H. & Shalet, S.M. Radiation-induced growth hormone deficiency. Horm Res 59 Suppl 1, 1-11 (2003).
61. Grimberg, A. & Allen, D.B. Growth hormone treatment for growth hormone deficiency and idiopathic short stature: new guidelines shaped by the presence and absence of evidence. Curr Opin Pediatr 29, 466-471 (2017).
62. Rivkees, S.A., Bode, H.H. & Crawford, J.D. Long-term growth in juvenile acquired hypothyroidism: the failure to achieve normal adult stature. N Engl J Med 318, 599-602 (1988).
63. Underwood, L.E. Growth retardation in chronic diseases: possible mechanisms. Acta Paediatr Suppl 88, 93-96 (1999).
64. Anneren, G., Gustavson, K.H., Sara, V.R. & Tuvemo, T. Growth retardation in Down syndrome in relation to insulin-like growth factors and growth hormone. Am J Med Genet Suppl 7, 59-62 (1990).
65. Aycan, Z. & Bas, V.N. Prader-Willi syndrome and growth hormone deficiency. J Clin Res Pediatr Endocrinol 6, 62-67 (2014).
66. Haverkamp, F. et al. Growth retardation in Turner syndrome: aneuploidy, rather than specific gene loss, may explain growth failure. J Clin Endocrinol Metab 84, 4578-4582 (1999).
67. Braun, L.R. & Marino, R. Disorders of Growth and Stature. Pediatr Rev 38, 293-304 (2017).
68. Lee, R.C., Feinbaum, R.L. & Ambros, V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 75, 843-854 (1993).
69. Ha, M. & Kim, V.N. Regulation of microRNA biogenesis. Nat Rev Mol Cell Biol 15, 509-524 (2014).
70. Kim, V.N., Han, J. & Siomi, M.C. Biogenesis of small RNAs in animals. Nat Rev Mol Cell Biol 10, 126-139 (2009).
71. Lin, S. & Gregory, R.I. MicroRNA biogenesis pathways in cancer. Nat Rev Cancer 15, 321-333 (2015).
72. Tarantino, C. et al. miRNA 34a, 100, and 137 modulate differentiation of mouse embryonic stem cells. FASEB J 24, 3255-3263 (2010).
73. Chen, J. et al. MicroRNA-30a ameliorates hepatic fibrosis by inhibiting Beclin1-mediated autophagy. J Cell Mol Med 21, 3679-3692 (2017).
74. Zhang, Z.W. et al. MicroRNA-122 regulates caspase-8 and promotes the apoptosis of mouse cardiomyocytes. Braz J Med Biol Res 50, e5760 (2017).
75. Peng, Y. & Croce, C.M. The role of MicroRNAs in human cancer. Signal Transduct Target Ther 1, 15004 (2016).
76. Geekiyanage, H. & Chan, C. MicroRNA-137/181c regulates serine palmitoyltransferase and in turn amyloid beta, novel targets in sporadic Alzheimer's disease. J Neurosci 31, 14820-14830 (2011).
77. Lai, C.Y. et al. Modulated expression of human peripheral blood microRNAs from infancy to adulthood and its role in aging. Aging Cell 13, 679-689 (2014).
78. Zhang, Z., Florez, S., Gutierrez-Hartmann, A., Martin, J.F. & Amendt, B.A. MicroRNAs regulate pituitary development, and microRNA 26b specifically targets lymphoid enhancer factor 1 (Lef-1), which modulates pituitary transcription factor 1 (Pit-1) expression. J Biol Chem 285, 34718-34728 (2010).
79. Yu, Z.W. et al. Roles of differential expression of miR-543-5p in GH regulation in rat anterior pituitary cells and GH3 cells. PLoS One 14, e0222340 (2019).
80. Cheng, Y. et al. Pituitary miRNAs target GHRHR splice variants to regulate GH synthesis by mediating different intracellular signalling pathways. RNA Biol 17, 1754-1766 (2020).
81. Elzein, S. & Goodyer, C.G. Regulation of human growth hormone receptor expression by microRNAs. Mol Endocrinol 28, 1448-1459 (2014).
82. Kamran, F. et al. Evidence That Up-Regulation of MicroRNA-29 Contributes to Postnatal Body Growth Deceleration. Mol Endocrinol 29, 921-932 (2015).
83. Cushing, L. et al. Disruption of miR-29 Leads to Aberrant Differentiation of Smooth Muscle Cells Selectively Associated with Distal Lung Vasculature. PLoS Genet 11, e1005238 (2015).
84. Luikenhuis, S., Giacometti, E., Beard, C.F. & Jaenisch, R. Expression of MeCP2 in postmitotic neurons rescues Rett syndrome in mice. Proc Natl Acad Sci U S A 101, 6033-6038 (2004).
85. Mellios, N. et al. beta2-Adrenergic receptor agonist ameliorates phenotypes and corrects microRNA-mediated IGF1 deficits in a mouse model of Rett syndrome. Proc Natl Acad Sci U S A 111, 9947-9952 (2014).
86. Papaioannou, G., Inloes, J.B., Nakamura, Y., Paltrinieri, E. & Kobayashi, T. let-7 and miR-140 microRNAs coordinately regulate skeletal development. Proc Natl Acad Sci U S A 110, E3291-3300 (2013).
87. Yu, S.L. et al. MicroRNA signature predicts survival and relapse in lung cancer. Cancer Cell 13, 48-57 (2008).
88. Smrt, R.D. et al. MicroRNA miR-137 regulates neuronal maturation by targeting ubiquitin ligase mind bomb-1. Stem Cells 28, 1060-1070 (2010).
89. Sun, G. et al. miR-137 forms a regulatory loop with nuclear receptor TLX and LSD1 in neural stem cells. Nat Commun 2, 529 (2011).
90. Chen, L. et al. miR-137 is frequently down-regulated in glioblastoma and is a negative regulator of Cox-2. Eur J Cancer 48, 3104-3111 (2012).
91. Verma, P., Augustine, G.J., Ammar, M.R., Tashiro, A. & Cohen, S.M. A neuroprotective role for microRNA miR-1000 mediated by limiting glutamate excitotoxicity. Nat Neurosci 18, 379-385 (2015).
92. Wright, C., Turner, J.A., Calhoun, V.D. & Perrone-Bizzozero, N. Potential Impact of miR-137 and Its Targets in Schizophrenia. Front Genet 4, 58 (2013).
93. Liu, M. et al. miR-137 targets Cdc42 expression, induces cell cycle G1 arrest and inhibits invasion in colorectal cancer cells. Int J Cancer 128, 1269-1279 (2011).
94. Zhu, X. et al. miR-137 inhibits the proliferation of lung cancer cells by targeting Cdc42 and Cdk6. FEBS Lett 587, 73-81 (2013).
95. Su, T.J. et al. Oncogenic miR-137 contributes to cisplatin resistance via repressing CASP3 in lung adenocarcinoma. Am J Cancer Res 6, 1317-1330 (2016).
96. Chang, J.C. et al. Adaptive adipose tissue stromal plasticity in response to cold stress and antibody-based metabolic therapy. Sci Rep 9, 8833 (2019).
97. Devanna, P. & Vernes, S.C. A direct molecular link between the autism candidate gene RORa and the schizophrenia candidate MIR137. Sci Rep 4, 3994 (2014).
98. Schizophrenia Psychiatric Genome-Wide Association Study, C. Genome-wide association study identifies five new schizophrenia loci. Nat Genet 43, 969-976 (2011).
99. Cheng, Y. et al. Partial loss of psychiatric risk gene Mir137 in mice causes repetitive behavior and impairs sociability and learning via increased Pde10a. Nat Neurosci (2018).
100. Jing, X. et al. Crosstalk of humoral and cell-cell contact-mediated signals in postnatal body growth. Cell Rep 2, 652-665 (2012).
101. Laron, Z. Laron syndrome (primary growth hormone resistance or insensitivity): the personal experience 1958-2003. J Clin Endocrinol Metab 89, 1031-1044 (2004).
102. Amselem, S. et al. Laron dwarfism and mutations of the growth hormone-receptor gene. N Engl J Med 321, 989-995 (1989).
103. Zhao, Y., Xiao, X., Frank, S.J., Lin, H.Y. & Xia, Y. Distinct mechanisms of induction of hepatic growth hormone resistance by endogenous IL-6, TNF-alpha, and IL-1beta. Am J Physiol Endocrinol Metab 307, E186-198 (2014).
104. Yan, H.L. et al. MiR-137 Deficiency Causes Anxiety-Like Behaviors in Mice. Front Mol Neurosci 12, 260 (2019).
105. Siegert, S. et al. The schizophrenia risk gene product miR-137 alters presynaptic plasticity. Nat Neurosci 18, 1008-1016 (2015).
106. Tarquinio, D.C. et al. Growth failure and outcome in Rett syndrome: specific growth references. Neurology 79, 1653-1661 (2012).
107. Amir, R.E. et al. Rett syndrome is caused by mutations in X-linked MECP2, encoding methyl-CpG-binding protein 2. Nat Genet 23, 185-188 (1999).
108. Castro, J. et al. Functional recovery with recombinant human IGF1 treatment in a mouse model of Rett Syndrome. Proc Natl Acad Sci U S A 111, 9941-9946 (2014).
109. Maimaiti, S. et al. Neuron-specific Mafb knockout causes growth retardation accompanied by an impaired GH/IGF-I axis. Exp Anim (2019).
110. Nielsen, H. Predicting Secretory Proteins with SignalP. Methods Mol Biol 1611, 59-73 (2017).
111. Sticht, C., De La Torre, C., Parveen, A. & Gretz, N. miRWalk: An online resource for prediction of microRNA binding sites. PLoS One 13, e0206239 (2018).
-
dc.identifier.urihttp://tdr.lib.ntu.edu.tw/jspui/handle/123456789/87364-
dc.description.abstract在第一部分,老化相關的健康問題隨人口逐漸老化而日漸重要,因此開發抗老化藥物是目前重要的課題,過去粒線體已經被證明參與細胞老化的過程,但針對此特徵的抗老藥物卻鮮少被探索,因此在本研究,我們證明二苯碘鎓 (Diphenyleneiodonium, DPI)可以顯著促進粒線體分裂與抑制其呼吸,在不同的細胞老化模型的評估下,二苯碘鎓減少了各項老化的標誌,同時抑制了老化造成的高呼吸率,此外,利用老化老鼠來測試二苯碘鎓抗老化的能力,發現其不僅可以減少老化標誌,也可以減緩老化相關的肝臟纖維化及免疫細胞浸潤,最後,二苯碘鎓也可以改善老化所造成的生理功能退化。總而言之,二苯碘鎓是一個具有抗老化能力藥物,未來可以單獨或合併其他抗老藥物來改善老化相關的疾病或狀態。第二部分, 微小核糖核酸 (MicroRNA) 可以透過影響訊息核糖核酸來調控基因表達,進而參與在許多細胞及生理功能,其中microRNA-137 (miR-137)不僅在不同物種中具有保守性且在神經系統中高度表達,並且調控神經發育與功能,另外也參與在各種癌症的病程,然而miR-137是否參與其他生理功能及癌症生成的角色尚未闡明,因此本研究主要透過在小鼠上剔除Mir137來探索其在活體之功能。有趣的是,Mir137 剔除鼠出現許多表現型包括成長遲緩、出生後死亡、骨質疏鬆、脂肪萎縮、低血糖、低體溫。進一步探索Mir137缺失導致成長遲緩的原因,我們發現Growth hormone (GH)/Insulin-like growth factor 1 (IGF1) 軸被破壞,血清的IGF1降低並伴隨GH的上升,透過體外及體內的實驗發現,Mir137缺失會導致非細胞自主性的GH阻抗,最後利用Cre/LoxP 系統確認了小鼠腦部Mir137的缺失會導致此現象。總結來說,腦部miR-137可以透過影響GH/IGF1途徑來調控身體成長。zh_TW
dc.description.abstractIn the part I: As the population ages, health issues with aging are becoming increasingly important, making the development of anti-aging drugs a current topic of interest. Previous research has shown that mitochondria play a role in the aging process, but the use of anti-aging drugs targeting this feature has been rare. In this study, we found that Diphenyleneiodonium (DPI) significantly increased mitochondrial fission and suppressed its respiration. When evaluated DPI effect on various models of cell senescence, DPI reduced various senescence markers and decreased the senescence-induced hyper oxygen consumption rate. Additionally, when tested on aging mice, DPI was found to not only reduce senescence markers, but also slow down aging-related liver fibrosis and infiltration of immune cells. It also improved the deterioration of physiological functions caused by aging. Overall, DPI is an effective anti-aging drug that can be used alone or in combination with other anti-aging drugs to improve aging-related diseases or conditions in the future. In the part II: MicroRNA can regulate gene expression by affecting messenger RNA and is involved in many cellular and physiological functions. Mir137 is high conservation in various species and highly expressed in neuron system. It plays a role in neurodevelopment and function, as well as various cancers. However, the role of miR-137 in physiological functions and cancer development is not yet fully clarification. In this study, we explored the function of miR-137 in mice by eliminating it. Interestingly, Mir137 knockout mice displayed several phenotypes including growth retardation, postnatal death, osteoporosis, lipoatrophy, hypoglycemia, and hypothermia. To further understand the cause of the growth retardation caused by the loss of Mir137, we found that Growth hormone (GH)/Insulin-like growth factor (IGF1) axis was disrupted and serum IGF1 decreased with an increase in GH. Through in vitro and in vivo experiments, we discovered that the loss of Mir137 led to non-cell-autonomous GH resistance. Finally, using the Cre/LoxP system, we confirmed that the loss of Mir137 in the mouse brain led to this phenomenon. In summary, brain miR-137 can regulate body growth by affecting the GH/IGF1 pathway.en
dc.description.provenanceSubmitted by admin ntu (admin@lib.ntu.edu.tw) on 2023-05-18T17:18:10Z
No. of bitstreams: 0
en
dc.description.provenanceMade available in DSpace on 2023-05-18T17:18:10Z (GMT). No. of bitstreams: 0en
dc.description.tableofcontentsCONTENTS
致謝 I
摘要 II
ABSTRACT IV
CONTENTS VI
LIST OF FIGURES XXII
LIST OF TABLES XXX
PART I: SENOMORPHIC EFFECT OF DIPHENYLENEIODONIUM THROUGH AMPK/MFF/DRP1 MEDIATED MITOCHONDRIAL FISSION. 1
二苯碘鎓透過AMPK/MFF/DRP1主導粒線體分裂以抗老化 1
摘要 2
Abstract 3
A. Introduction: 5
1. Aging 5
2. Cellular senescence 5
2.1 Cell-cycle arrest 6
2.1.1 The p53/p21 pathway 6
2.1.2 p16/pRB pathway 7
2.2 Senescence-associated secretory phenotype 7
2.3 Morphological changes 8
2.3.1 Cell size 8
2.3.2 Cellular granularity 8
2.4 Senescence-associated β-galactosidase 9
3. Stress-induced cellular senescence 9
3.1 Telomere dysfunction and DNA damage 9
3.2 Oncogene activation 10
3.3 Oxidative stress 10
4. Mitochondria and cellular senescence 11
4.1 Mitochondrial morphology dynamics 12
4.1.1 Mitochondrial fusion proteins 13
4.1.1.1 Mitofusin1/2 (MFN1/2) 13
4.1.1.2 Optic atrophy 1 (OPA1) 13
4.1.2 Mitochondrial fission proteins 14
4.1.2.1 DRP1 14
4.1.2.1 Phosphorylation 15
4.1.2.2 SUMOylation 16
4.1.2.3 Ubiquitination 16
4.1.2.4 Palmitoylation 17
4.1.2.5 S-nitrosylation 18
4.1.2.6 O-GlcNAcylation 19
4.1.2.7 DRP1 recruitment factors 19
4.2 Mitochondrial morphology dysregulation and aging 20
4.2.1 Saccharomyces cerevisiae 20
4.2.2 Caenorhabditis elegans 21
4.2.3 Drosophila melanogaster 22
4.2.4 Mus musculus and Homo sapiens 23
5. Elimination of senescent cells 24
5.1 Senolytic drugs 24
5.2 Senomorphic drug 24
5.3 Cell therapy 25
6. Targeting mitochondria to clear senescent cells 25
7. Cell and animal models for aging research 26
7.1 Cell models of senescence 26
7.1.1 Replicative senescence 26
7.1.2 Stress-induced senescence 27
7.1.3 Oncogene-induced senescence 27
7.2 Mouse models of aging 27
7.2.1 Aging mice model 27
7.2.2 Lung senescence and fibrosis model 28
7.2.3 Senescent-cell transplantation model 28
8. Diphenyleneiodonium chloride (DPI) 29
9. Specific aim: 29
B. Material and Method: 30
1. Cell culture and treatments 30
2. Animal experiments 31
3. Aged mice model 31
4. Bleomycin-induced lung fibrosis model 31
5. Immunofluorescence staining 32
6. Cell viability assay 33
7. Mitochondrial respiration detection by Seahorse analysis 33
8. Tetramethylrhodamine (TMRM) staining 34
9. SA-β galactosidase (SA-β-gal) staining 34
10. Western blots and antibodies 35
11. Reverse transcription (RT)-qPCR 37
12. Cell size analysis by flow cytometry 37
13. Reactive oxygen species (ROS) detection by flow cytometry 37
14. Masson’s trichrome staining 38
15. Immunohistochemistry staining and antibodies 38
16. Physical function test 39
17. Lung function test 39
18. Statistical analysis 40
C. Result: 41
1. Potential anti-senescence compound was identified by mitochondrial morphology regulation. 41
2. Low dosage DPI repressed mitochondrial respiration without cytotoxicity. 42
3. DPI attenuates senescence in both stress- and replication-induced cell models 44
4. DPI-induced mitochondrial fragmentation was mediated by AMPK/MFF/DRP1 axis. 46
5. DPI effect of anti-senescence is contributed by DRP1-mediated mitochondrial fission. 48
6. Senescence markers is reduced in aged mice by DPI treatment 49
7. DPI treatment improves age-related histopathological changes and restore physical functions in aged mice 50
8. DPI improves bleomycin-induced lung fibrosis 51
D. Discussion: 52
E. Figure: 57
Figure 1. Mitochondrial morphology changes of senescent MEF cells. 57
Figure 2. Cell viability test of cortisone, TBBz, DPI. 58
Figure 3. DPI dramatically down regulated mitochondrial oxygen consumption rate in NIH3T3 cells. 59
Figure 4. DPI dose-dependent decreased mitochondrial oxygen consumption rate and mitochondrial membrane potential in NIH3T3 cells. 60
Figure 5. Low concentration of DPI showed without cytotoxicity in short- and long-term treatment. 62
Figure 6. BrdU treatment induced cellular senescence dose-dependently. 63
Figure 7. Expression of cell cycle arrest genes and SASP factors were evaluated after BrdU treatment. 65
Figure 8. DPI reduced SA-β gal activity and increased proliferation marker Ki-67 in BrdU-induced NIH3T3 senescence model. 67
Figure 9. DPI reduced senescence markers in BrdU-induced NIH3T3 senescence model. 68
Figure 10. DPI treatment improved mitochondrial elongation and reduced the level of ROS in BrdU-induced senescent NIH3T3 cells. 69
Figure 11. DPI reversed mitochondrial respiration change in BrdU-induced NIH3T3 senescent cells. 71
Figure 12. Replicative cellular senescence was presented of MEFs after serial cell culture. 72
Figure 13. Expression of cell cycle arrest genes and SASP genes in senescent MEF cells. 73
Figure 14. DPI treatment reduced SA-β gal activity and increased proliferation marker Ki-67 in MEFs. 74
Figure 15. DPI reduced senescence markers in senescent MEFs. 75
Figure 16. DPI reversed mitochondrial respiration change in senescent MEFs. 77
Figure 17. Irradiation (IR) induced mitochondrial elongation. 78
Figure 18. DPI attenuate BrdU- and IR-induced cellular senescence in IMR90 cells. 79
Figure 19. DPI showed that did not specifically reduce senescent cells. 81
Figure 20. Translocation of DRP1 to mitochondria was increased by DPI treatment. 82
Figure 21. DPI treatment did not alter the expression of DRP1 recruitment proteins and DRP1 phosphorylation. 83
Figure 22. DPI promoted MFF phosphorylation via AMPK pathway. 85
Figure 23. Inhibition of Drp1 reversed DPI induced mitochondrial fragmentation. 86
Figure 24. Inhibition of DRP1 activity reversed DPI-induced mitochondrial respiration change. 87
Figure 25. DRP1 activity inhibition by Mdivi-1 reversed DPI anti-senescence effect in MEFs. 88
Figure 26. Mdivi-1 treatment reversed DPI effect on reduction of senescence markers. 89
Figure 27. Mdivi-1 reversed DPI-induced mitochondrial respiration change in senescent MEFs. 90
Figure 28. DPI enhanced mitochondria fragmentation in senescent MEFs through the action of DRP1 but failed to inhibit ROS production in the short term. 91
Figure 29. DPI treatment promoted DRP1 mitochondrial translocation in senescent MEFs and did not affect mitochondrial dynamic protein expression. 93
Figure 30. Mdivi-1 reversed DPI induced mitochondrial fragmentation and ROS change in senescent MEFs in long-term treatment. 94
Figure 31. DPI toxicity was tested in mice. 96
Figure 32. DPI reduced SA-β gal activity in kidney without liver toxicity in mice. 97
Figure 33. DPI reduced cell cycle arrest genes in liver and kidney of mice. 98
Figure 34. DPI reduced SASP genes in liver and kidney of mice with DPI treatment. 99
Figure 35. Mice treated with DPI that showed without body weight change and liver toxicity. 100
Figure 36. Treatment with DPI resulted in a reduction in the number of SA-β positive cells in the liver and kidney of aged mice. 102
Figure 37. Expression of cell cycle arrest genes Cdkn1a and Cdkn2a mRNA in the livers and kidneys of aged mice was assessed following DPI treatment. 104
Figure 38. The expression pattern of SASP factors in the liver and kidney of aged mice was examined after DPI treatment. 105
Figure 39. The DPI treatment leads to a reduction in immune cell infiltration and an improvement in liver fibrosis in aged mice. 106
Figure 40. DPI treatment enhances the physiological capabilities of aged mice. 108
Figure 41. DPI treatment improved lung fibrosis and reduced cell cycle arrest gene expression in bleomycin-induced mice. 109
Figure 42. DPI treatment improved SASP expression in lung tissue of bleomycin-induced mice. 111
Figure 43. DRP1 phosphorylation was not altered by ROS. 112
Figure 44. Summary. 113
F. Table: 114
Table 1. Primer list of QPCR 114
G. Reference 116
H. Appendix 140
PART II: INVESTIGATION OF MIR-137 ROLE IN PHYSIOLOGICAL FUNCTIONS: FOCUS ON GROWTH RETARDATION. 151
探究MIR-137在生理功能上所扮演的角色: 專注於成長遲緩 151
摘要 152
Abstract 154
A. Introduction: 156
1. The growth hormone and insulin-like growth factor 1 axis (GH/IGF1 axis) 156
1.1 Growth hormone: 156
1.1.1 Growth hormone signal transduction: 157
1.1.2 Whole-body Ghr KO mice: 158
1.1.3 Liver-specific KO mice: 158
1.1.4 Intestinal epithelial Ghr KO mice: 159
1.1.5 Adipocyte Ghr KO mice: 160
1.1.6 Brain Ghr KO mice: 161
1.1.7 Muscle Ghr KO mice: 162
1.1.8 Bone Ghr KO mice 163
1.2 Insulin-like growth factor 1 (IGF1): 163
1.2.1 IGF1 signal transduction: 164
1.2.2 Igf1 whole-body KO mice: 164
1.2.3 Liver Igf1 KO mice: 165
1.2.4 Igf1r KO mice: 166
1.3 Regulation of the GH/IGF1 Axis: 166
1.3.1 Regulation of GH expression and secretion: 166
1.3.2 Regulation of IGF1 expression and secretion: 167
1.3.3 Non-cell-autonomous regulation of the GH/IGF1 axis for body growth control 168
2. Body growth disorders: 169
2.1 Malnutrition 170
2.2 Growth hormone deficiency (GHD) 170
2.3 Hypothyroidism 170
2.4 Chronic diseases 170
2.5 Genetic syndromes 171
3. MicroRNA (miRNA): 171
3.1. MicroRNA in body growth regulation: 172
3.2. MicroRNA-137: 174
4. Specific aim: 175
B. Materials and Methods: 176
1. Production of mice with tissue-specific Mir137 deletion: 176
2. Polymerase chain reaction (PCR): 176
3. Real-time quantitative PCR (QPCR): 176
4. Western blot: 177
5. Complete blood count (CBC): 178
6. Hematoxylin and eosin staining (H&E staining): 178
7. Trabecular bone (TB) and bone mineral density (BMD) detection by micro-tomography (micro-CT): 179
8. Body temperature measurement via infrared camera: 179
9. Blood glucose detection: 179
10. Enzyme-linked immunosorbent assay (ELISA): 180
11. Recombinant protein treatment: 180
12. Primary hepatocyte isolation and GH pathway analysis: 180
13. cDNA microarray and RNA sequencing: 181
14. Statistical analysis 182
C. Results: 183
1. Successful creation of Mir137 knockout mice 183
2. Expression profiling of miR-137 in brain areas and the effect on brain cell population 183
3. Mir137 KO mice showed severe postnatal growth retardation and early death 184
4. Abnormalities in bone and adipose tissues were found in Mir137 KO mice 185
5. Hepatic IGF1 production was disrupted in Mir137 KO mice 187
6. Hepatic IGF1 suppression was regulated via non-cell-autonomous pathway 189
7. miR-137 deficiency led to GH resistance 190
8. Growth retardation in Mir137 KO mice did not occur through the starvation pathway 191
9. Brain-specific deletion of miR-137 led to growth retardation 192
10. MiR-137 deficiency did not affect GDF15 expression in tissues 194
11. Analysis of liver and brain gene expression 194
D. Discussion: 196
E. Figure: 203
Figure 1. Targeting strategy for Mir137 knockout by homologous recombination. 203
Figure 2. Expression of Mir137 in organs was measured via Taqman real-time polymerase chain reaction (RT-PCR) analysis. 204
Figure 3. Expression profiling of Mir137 in several brain areas. 206
Figure 4. Highest expression of Mir137 in P14 brain. 207
Figure 5. miR-137 deficiency did not affect cell populations in the brain. 208
Figure 6. Mir137 KO mice showed severe growth retardation. 209
Figure 7. Major organs were smaller in miR-137-deficient mice. 210
Figure 8. There was no difference in body size between WT (+/+) and KO (−/−) mice at P1. 211
Figure 9. Mir137 knockout mice showed early postnatal death. 212
Figure 10. Complete blood count (CBC) for miR-137-deficient mice. 213
Figure 11. Histology analysis of miR-137-deficient mice. 215
Figure 12. Osteoporosis was found in Mir137 knockout mice. 216
Figure 13. Brown and white fatty tissues were smaller in extent in miR-137-deficient mice. 217
Figure 14. miR-137 deficiency led to adipose tissue atrophy. 219
Figure 15. Expression pattern of markers of browning adipocytes in white fatty tissue. 220
Figure 16. miR-137 deficiency caused hypoglycemia. 221
Figure 17. Mir137 knockout led to hypothermia. 222
Figure 18. Expression of hepatic Igf1 and Igf1-associated genes was disrupted in Mir137 knockout mice. 223
Figure 19. miR-137 deficiency did not affect Igf1 mRNA expression in other organs, but it induced Igf1r mRNA expression. 224
Figure 20. Positive correlation of serum IGF1 with body weight. 225
Figure 21. AKT and ERK phosphorylation downregulated in miR-137-deficient tissues. 226
Figure 22. Liver p-STAT5 expression was downregulated in Mir137 KO mice. 227
Figure 23. miR-137 deficiency impaired GH-downstream processes only in the liver. 228
Figure 24. miR-137 deficiency did not affect hepatocyte GH signaling under GH stimulation. 229
Figure 25. Analysis of GH expression in pituitary gland and serum. 230
Figure 26. Exogenous growth hormone (GH) injection induced rapid phosphorylation of STAT5. 231
Figure 27. miR-137 deficiency suppressed phosphorylation of STAT5 under exogenous GH stimulation. 232
Figure 28. Exogenous ghrelin facilitated endogenous GH secretion and hepatic STAT5 phosphorylation. 233
Figure 29. Phosphorylation of STAT5 was suppressed in miR-137-deficient mice under ghrelin treatment. 234
Figure 30. GH receptor (GHR) mRNA expression was significantly reduced in liver and brain of miR-137-deficient mice. 235
Figure 31. Liver starvation gene expression. 236
Figure 32. Expression of starvation markers was monitored in mouse liver and serum. 237
Figure 33. Autophagy as a starvation marker could be used to monitor the status of mice. 238
Figure 34. miR-137 deficiency did not lead to autophagy activation. 239
Figure 35. Tissue-specific knockout of Mir137 was obtained using the Cre-LoxP system. 240
Figure 36. Specific knockout of Mir137 in the brain led to growth retardation. 241
Figure 37. Expression of GH signaling pathway and GH-downstream genes was dramatically reduced in brain-specific KO mice. 242
Figure 38. STAT5 phosphorylation levels were lower under KO serum treatment. 243
Figure 39. miR-137 was highly expressed in the right atrium. 244
Figure 40. Deletion of Mir137 did not affect Gdf15 expression in the heart or other organs. 245
Figure 41. Gene-expression analysis of miR-137-deficient mice. 246
Figure 42. Quality control of liver microarray. 247
Figure 43. Pathway map and Go process analysis for liver genes through MetaCore. 248
Figure 44. Selected genes from brain next-generation sequencing data were validated via QPCR and ELISA. 249
Figure 45. Deletion of miR-137 in the brain resulted in impaired function of the GH/IGF1 axis and postnatal growth retardation via the endocrine pathway. 250
F. Table 251
Table 1. Primer for genotyping. 251
Table 2. Primers for QPCR 252
Table 3. The Statistic of genotype in offspring. 255
Table 4. Gene list: Partial of genes dose-dependent up-regulated in brain of Mir137 deficiency mice 256
G. Reference: 259
H. Appendix 274
 
LIST OF FIGURES
Part I:
Figure 1. Mitochondrial morphology changes of senescent MEF cells. 57
Figure 2. Cell viability test of cortisone, TBBz, DPI. 58
Figure 3. DPI dramatically down regulated mitochondrial oxygen consumption rate in NIH3T3 cells. 59
Figure 4. DPI dose-dependent decreased mitochondrial oxygen consumption rate and mitochondrial membrane potential in NIH3T3 cells. 60
Figure 5. Low concentration of DPI showed without cytotoxicity in short- and long-term treatment. 62
Figure 6. BrdU treatment induced cellular senescence dose-dependently. 63
Figure 7. Expression of cell cycle arrest genes and SASP factors were evaluated after BrdU treatment. 65
Figure 8. DPI reduced SA-β gal activity and increased proliferation marker Ki-67 in BrdU-induced NIH3T3 senescence model. 67
Figure 9. DPI reduced senescence markers in BrdU-induced NIH3T3 senescence model. 68
Figure 10. DPI treatment improved mitochondrial elongation and reduced the level of ROS in BrdU-induced senescent NIH3T3 cells. 69
Figure 11. DPI reversed mitochondrial respiration change in BrdU-induced NIH3T3 senescent cells. 71
Figure 12. Replicative cellular senescence was presented of MEFs after serial cell culture. 72
Figure 13. Expression of cell cycle arrest genes and SASP genes in senescent MEF cells. 73
Figure 14. DPI treatment reduced SA-β gal activity and increased proliferation marker Ki-67 in MEFs. 74
Figure 15. DPI reduced senescence markers in senescent MEFs. 75
Figure 16. DPI reversed mitochondrial respiration change in senescent MEFs. 77
Figure 17. Irradiation (IR) induced mitochondrial elongation. 78
Figure 18. DPI attenuate BrdU- and IR-induced cellular senescence in IMR90 cells. 79
Figure 19. DPI showed that did not specifically reduce senescent cells. 81
Figure 20. Translocation of DRP1 to mitochondria was increased by DPI treatment. 82
Figure 21. DPI treatment did not alter the expression of DRP1 recruitment proteins and DRP1 phosphorylation. 83
Figure 22. DPI promoted MFF phosphorylation via AMPK pathway. 85
Figure 23. Inhibition of Drp1 reversed DPI induced mitochondrial fragmentation. 86
Figure 24. Inhibition of DRP1 activity reversed DPI-induced mitochondrial respiration change. 87
Figure 25. DRP1 activity inhibition by Mdivi-1 reversed DPI anti-senescence effect in MEFs. 88
Figure 26. Mdivi-1 treatment reversed DPI effect on reduction of senescence markers. 89
Figure 27. Mdivi-1 reversed DPI-induced mitochondrial respiration change in senescent MEFs. 90
Figure 28. DPI enhanced mitochondria fragmentation in senescent MEFs through the action of DRP1 but failed to inhibit ROS production in the short term. 91
Figure 29. DPI treatment promoted DRP1 mitochondrial translocation in senescent MEFs and did not affect mitochondrial dynamic protein expression. 93
Figure 30. Mdivi-1 reversed DPI induced mitochondrial fragmentation and ROS change in senescent MEFs in long-term treatment. 94
Figure 31. DPI toxicity was tested in mice. 96
Figure 32. DPI reduced SA-β gal activity in kidney without liver toxicity in mice. 97
Figure 33. DPI reduced cell cycle arrest genes in liver and kidney of mice. 98
Figure 34. DPI reduced SASP genes in liver and kidney of mice with DPI treatment. 99
Figure 35. Mice treated with DPI that showed without body weight change and liver toxicity. 100
Figure 36. Treatment with DPI resulted in a reduction in the number of SA-β positive cells in the liver and kidney of aged mice. 102
Figure 37. Expression of cell cycle arrest genes Cdkn1a and Cdkn2a mRNA in the livers and kidneys of aged mice was assessed following DPI treatment. 104
Figure 38. The expression pattern of SASP factors in the liver and kidney of aged mice was examined after DPI treatment. 105
Figure 39. The DPI treatment leads to a reduction in immune cell infiltration and an improvement in liver fibrosis in aged mice. 106
Figure 40. DPI treatment enhances the physiological capabilities of aged mice. 108
Figure 41. DPI treatment improved lung fibrosis and reduced cell cycle arrest gene expression in bleomycin-induced mice. 109
Figure 42. DPI treatment improved SASP expression in lung tissue of bleomycin-induced mice. 111
Figure 43. DRP1 phosphorylation was not altered by ROS. 112
Figure 44. Summary. 113
Part II
Figure 1. Targeting strategy for Mir137 knockout by homologous recombination. 203
Figure 2. Expression of Mir137 in organs was measured via Taqman real-time polymerase chain reaction (RT-PCR) analysis. 204
Figure 3. Expression profiling of Mir137 in several brain areas. 206
Figure 4. Highest expression of Mir137 in P14 brain. 207
Figure 5. miR-137 deficiency did not affect cell populations in the brain. 208
Figure 6. Mir137 KO mice showed severe growth retardation. 209
Figure 7. Major organs were smaller in miR-137-deficient mice. 210
Figure 8. There was no difference in body size between WT (+/+) and KO (−/−) mice at P1. 211
Figure 9. Mir137 knockout mice showed early postnatal death. 212
Figure 10. Complete blood count (CBC) for miR-137-deficient mice. 213
Figure 11. Histology analysis of miR-137-deficient mice. 215
Figure 12. Osteoporosis was found in Mir137 knockout mice. 216
Figure 13. Brown and white fatty tissues were smaller in extent in miR-137-deficient mice. 217
Figure 14. miR-137 deficiency led to adipose tissue atrophy. 219
Figure 15. Expression pattern of markers of browning adipocytes in white fatty tissue. 220
Figure 16. miR-137 deficiency caused hypoglycemia. 221
Figure 17. Mir137 knockout led to hypothermia. 222
Figure 18. Expression of hepatic Igf1 and Igf1-associated genes was disrupted in Mir137 knockout mice. 223
Figure 19. miR-137 deficiency did not affect Igf1 mRNA expression in other organs, but it induced Igf1r mRNA expression. 224
Figure 20. Positive correlation of serum IGF1 with body weight. 225
Figure 21. AKT and ERK phosphorylation downregulated in miR-137-deficient tissues. 226
Figure 22. Liver p-STAT5 expression was downregulated in Mir137 KO mice. 227
Figure 23. miR-137 deficiency impaired GH-downstream processes only in the liver. 228
Figure 24. miR-137 deficiency did not affect hepatocyte GH signaling under GH stimulation. 229
Figure 25. Analysis of GH expression in pituitary gland and serum. 230
Figure 26. Exogenous growth hormone (GH) injection induced rapid phosphorylation of STAT5. 231
Figure 27. miR-137 deficiency suppressed phosphorylation of STAT5 under exogenous GH stimulation. 232
Figure 28. Exogenous ghrelin facilitated endogenous GH secretion and hepatic STAT5 phosphorylation. 233
Figure 29. Phosphorylation of STAT5 was suppressed in miR-137-deficient mice under ghrelin treatment. 234
Figure 30. GH receptor (GHR) mRNA expression was significantly reduced in liver and brain of miR-137-deficient mice. 235
Figure 31. Liver starvation gene expression. 236
Figure 32. Expression of starvation markers was monitored in mouse liver and serum. 237
Figure 33. Autophagy as a starvation marker could be used to monitor the status of mice. 238
Figure 34. miR-137 deficiency did not lead to autophagy activation. 239
Figure 35. Tissue-specific knockout of Mir137 was obtained using the Cre-LoxP system. 240
Figure 36. Specific knockout of Mir137 in the brain led to growth retardation. 241
Figure 37. Expression of GH signaling pathway and GH-downstream genes was dramatically reduced in brain-specific KO mice. 242
Figure 38. STAT5 phosphorylation levels were lower under KO serum treatment. 243
Figure 39. miR-137 was highly expressed in the right atrium. 244
Figure 40. Deletion of Mir137 did not affect Gdf15 expression in the heart or other organs. 245
Figure 41. Gene-expression analysis of miR-137-deficient mice. 246
Figure 42. Quality control of liver microarray. 247
Figure 43. Pathway map and Go process analysis for liver genes through MetaCore. 248
Figure 44. Selected genes from brain next-generation sequencing data were validated via QPCR and ELISA. 249
Figure 45. Deletion of miR-137 in the brain resulted in impaired function of the GH/IGF1 axis and postnatal growth retardation via the endocrine pathway. 250

LIST OF TABLES
PartI:
Table 1. Primer list of QPCR 114
Part II
Table 1. Primer for genotyping. 251
Table 2. Primers for QPCR 252
Table 3. The Statistic of genotype in offspring. 255
Table 4. Gene list: Partial of genes dose-dependent up-regulated in brain of Mir137 deficiency mice 256
-
dc.language.isoen-
dc.subject微小核糖核酸-137zh_TW
dc.subject成長賀爾蒙阻抗zh_TW
dc.subject成長遲緩zh_TW
dc.subject老化zh_TW
dc.subject二苯碘鎓zh_TW
dc.subject粒線體zh_TW
dc.subjectGH resistanceen
dc.subjectMiR-137en
dc.subjectGrowth retardationen
dc.subjectDiphenyleneiodoniumen
dc.subjectAgingen
dc.subjectMitochondriaen
dc.titlePart I: 二苯碘鎓透過AMPK/MFF/DRP1主導粒線體分裂以抗老化; Part II: 探究miR-137在生理功能上所扮演的角色: 專注於成長遲緩zh_TW
dc.titlePart I: Senomorphic Effect of Diphenyleneiodonium through AMPK/MFF/DRP1 Mediated Mitochondrial Fission; Part II: Investigation of miR-137 Role in Physiological Functions: Focus on Growth Retardationen
dc.typeThesis-
dc.date.schoolyear111-1-
dc.description.degree博士-
dc.contributor.oralexamcommittee楊泮池;周玉山;郭靜穎;張以承zh_TW
dc.contributor.oralexamcommitteePan-Chyr Yang;Yuh-Shan Jou;Ching-Ying Kuo;Yi-Cheng Changen
dc.subject.keyword老化,二苯碘鎓,粒線體,微小核糖核酸-137,成長遲緩,成長賀爾蒙阻抗,zh_TW
dc.subject.keywordAging,Diphenyleneiodonium,Mitochondria,MiR-137,Growth retardation,GH resistance,en
dc.relation.page284-
dc.identifier.doi10.6342/NTU202300422-
dc.rights.note同意授權(限校園內公開)-
dc.date.accepted2023-02-15-
dc.contributor.author-college生命科學院-
dc.contributor.author-dept基因體與系統生物學學位學程-
dc.date.embargo-lift2025-07-31-
顯示於系所單位:基因體與系統生物學學位學程

文件中的檔案:
檔案 大小格式 
ntu-111-1.pdf
授權僅限NTU校內IP使用(校園外請利用VPN校外連線服務)
15.83 MBAdobe PDF
顯示文件簡單紀錄


系統中的文件,除了特別指名其著作權條款之外,均受到著作權保護,並且保留所有的權利。

社群連結
聯絡資訊
10617臺北市大安區羅斯福路四段1號
No.1 Sec.4, Roosevelt Rd., Taipei, Taiwan, R.O.C. 106
Tel: (02)33662353
Email: ntuetds@ntu.edu.tw
意見箱
相關連結
館藏目錄
國內圖書館整合查詢 MetaCat
臺大學術典藏 NTU Scholars
臺大圖書館數位典藏館
本站聲明
© NTU Library All Rights Reserved