Abstract
Background
The mammalian circadian clock and its associated clock genes are increasingly be recognized as critical components for a number of physiological and disease processes that extend beyond hormone release, thermal regulation, and sleep-wake cycles. New evidence suggests that clinical behavior disruptions that involve prolonged shift work and even space travel may negatively impact circadian rhythm and lead to multi-system disease.
Methods
In light of the significant role circadian rhythm can hold over the body's normal physiology as well as disease processes, we examined and discussed the impact circadian rhythm and clock genes hold over lifespan, neurodegenerative disorders, and tumorigenesis.
Results
In experimental models, lifespan is significantly reduced with the introduction of arrhythmic mutants and leads to an increase in oxidative stress exposure. Interestingly, patients with Alzheimer's disease and Parkinson's disease may suffer disease onset or progression as a result of alterations in the DNA methylation of clock genes as well as prolonged pharmacological treatment for these disorders that may lead to impairment of circadian rhythm function. Tumorigenesis also can occur with the loss of a maintained circadian rhythm and lead to an increased risk for nasopharyngeal carcinoma, breast cancer, and metastatic colorectal cancer. Interestingly, the circadian clock system relies upon the regulation of the critical pathways of autophagy, the mechanistic target of rapamycin (mTOR), AMP activated protein kinase (AMPK), and silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae) (SIRT1) as well as proliferative mechanisms that involve the wingless pathway of Wnt/β-catenin pathway to foster cell survival during injury and block tumor cell growth.
Conclusions
Future targeting of the pathways of autophagy, mTOR, SIRT1, and Wnt that control mammalian circadian rhythm may hold the key for the development of novel and effective therapies against aging- related disorders, neurodegenerative disease, and tumorigenesis.
Keywords: aging, aging-related disorders, Alzheimer's disease, AMP activated protein kinase (AMPK), angiogenesis, apoptosis, autophagy, BMAL1, cardiovascular disease, β-catenin, circadian rhythm, CLOCK, clock genes, Cryptochrome, diabetes mellitus, hamartin (tuberous sclerosis 1)/tuberin (tuberous sclerosis 2) (TSC1/TSC2), Huntington's disease, mechanistic target of rapamycin (mTOR), mTOR Complex 1 (mTORC1), mTOR Complex 2 (mTORC2), metabolism, nerve growth factor, nicotinamide, nicotinamide adenine dinucleotide (NAD+), Parkinson's disease, period (PER), oxidative stress, programmed cell death, REV-ERBα, RORα, RORE, shift work, silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae) (SIRT1), sirtuin, space travel, stem cells, suprachiasmatic nucleus, wingless, Wnt
Circadian Rhythm and Clock Genes
The mammalian circadian clock is located in the suprachiasmatic nucleus (SCN) that lies above the optic chiasm to receive light input from photosensitive ganglion cells in the retina. The SCN relies upon the pineal gland, hypothalamic nuclei, and vasoactive intestinal peptide to control multiple process such as the release of hormones cortisol and melatonin, oxidative stress responses (1), and the regulation of body temperature in the circadian cycle (2-4).
The circadian clock relies upon cellular signals and light input to align itself with solar time and oscillate over a twenty-four hour period. This clock receives daily cues from external environmental sources that consist of daylight and darkness to drive circadian rhythm. Ultimately, the circadian rhythm controls behavior, normal physiology, and cellular biochemical transmission in an organism.
Members of the basic helix-loop-helix -PAS (Period-Arnt-Single-minded) transcription factor family, such as CLOCK and BMAL1 (5), oversee the expression of the genes Cryptochrome (Cry1 and Cry2) and Period (Per1, Per2, and Per3). Negative feedback is provided by PER:CRY heterodimers that can translocate to the nucleus to block the transcription of CLOCK:BMAL1 complexes. Additional regulatory loops consist of the activation by CLOCK:BMAL1 heterodimers of retinoic acid-related orphan nuclear receptors REV-ERBα and RORα. These receptors bind retinoic acid-related orphan receptor response elements (ROREs) present in the BMAL1 promoter to control transcription with RORs activating transcription and REV-ERBs repressing transcription to lead to a circadian oscillation of BMAL1 (6, 7).
Circadian Rhythm in Degenerative Disease and Cancer
Neurodegenerative diseases and decreased lifespan have been linked to the function of the mammalian circadian clock. In studies with Drosophila melanogaster, lifespan was reduced in three arrhythmic mutants involving ClkAR, cyc0 and tim0. In particular, ClkAR mutants had significant faster age-related locomotor deficits. Restoring Clk function was able to rescue Drosophila from the locomotor deficits. An increase in oxidative stress was noted with the mutant phenotypes, but deficits appeared to correlate best with loss of dopaminergic neurons (8). In patients with Alzheimer's disease, rhythmic methylation of BMAL1 has been found to be changed in the brains of patients with Alzheimer's disease, suggesting that alterations in the DNA methylation of clock genes may contribute to cognitive loss and behavior changes in individuals with Alzheimer's disease (9). Animal models of Parkinson's disease with 6-hydroxydopamine (6-OHDA) also show decreased BMAL1 and RORα that persisted with levodopa treatment, indication that long-term levodopa treatment may impair circadian rhythm function (10).
In regards to tumorigenesis and circadian rhythm involvement, TIMELESS, a mammalian homolog of a Drosophila circadian rhythm gene, has been found to be up-regulated in nasopharyngeal carcinoma and increased TIMELESS expression was associated with decreased overall survival. In addition, over-expression of TIMELESS led to resistance to cisplatin mediated apoptosis and activated the wingless pathway of Wnt/β-catenin pathway (11). Wnt proteins are cysteine-rich glycosylated proteins that oversee processes such as neuronal development, immunity, angiogenesis, fibrosis, stem cell proliferation, and tumorigenesis (12-14). Wnt and β-catenin signaling can block autophagy (15), apoptosis (16), affect sensory modalities (17), and lead to stem cell proliferation (18-20). However, these pathways can promote angiogenesis (21-23) and lead to tumor growth (24-29) that may align with the proliferative pathways of clock genes such as TIMELESS. On a clinical basis, disruption of circadian rhythms with shift work suggests that such duties also may increase the risk for developing cancer. Female nurses with long-term rotating night shift work had an increased risk for breast cancer (30). In addition, increased expression of the circadian gene hClock may contribute to tumorigenesis, such as the metastasis of colorectal cancer, through the enhanced expression of angiogenesis-related genes (31).
Circadian Rhythm and the Modulation of Autophagy
Autophagy is a process that recycles components of the cytoplasm in cells for tissue remodeling and eliminates non-functional organelles (32-36). The term macroautophagy refers to a classification of autophagy that recycles organelles and consists of the sequestration of cytoplasmic proteins and organelles into autophagosomes. These autophagosomes then combine with lysosomes for degradation and recycling (37, 38). Microautophagy describes the invagination of the lysosomal membranes for the sequestration and digestion of cytoplasmic components (39). Chaperone-mediated autophagy (40) uses cytosolic chaperones to transport cytoplasmic components across lysosomal membranes (41).
Autophagy can be involved in a number of degenerative disorders such as Alzheimer's disease (42-46), Parkinson's disease (41, 47, 48), Huntington's disease (49-51), and diabetes mellitus (12, 18, 33, 43, 52, 53). Importantly, autophagy also can impact cognitive decline (12, 54, 55) and aging processes (43, 56-60).
Interestingly, circadian rhythm dysfunction during cognitive loss and aging has been tied to the induction of autophagy (61). Studies with Drosophila show that the accumulation of neural aggregates observed with aging is associated with a reduction in the autophagy pathway. These neural aggregates lead to behavior impairments that can be resolved with the maintenance of autophagy pathways in neurons (62). In animal models of Alzheimer's disease, a basal circadian rhythm that controls macroautophagy may be necessary to limit cognitive decline and amyloid deposition (63). Even mild changes in the external environment that affect circadian rhythm may alter cognition. Chronic sleep fragmentation has been shown to affect autophagy proteins in the hippocampus (64) that may affect memory and cognition (44, 46, 55, 56, 65). In addition, autophagy in the hippocampus is depressed during the absence of the PER1 circadian clock protein that may worsen the pathology of cerebral ischemia (66).
Circadian Rhythm, mTOR and SIRT1
The potential of the mammalian circadian rhythm to control cell survival not only relies upon autophagy activity, but also upon other cellular signaling pathways that include the mechanistic target of rapamycin (mTOR) and the silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae) (SIRT1). mTOR, also known as the mammalian target of rapamycin and the FK506-binding protein 12-rapamycin complex-associated protein 1 (44), is a 289-kDa serine/threonine protein involved in multiple cellular processes that include autophagy. mTOR is the principal component of the protein complexes mTOR Complex 1 (mTORC1) and mTOR Complex 2 (mTORC2) (42, 44, 67, 68). mTORC1 contains the components Raptor, Deptor (DEP domain-containing mTOR interacting protein), the proline rich Akt substrate 40 kDa (PRAS40), and mammalian lethal with Sec13 protein 8, termed mLST8 (mLST8/GβL) (27). mTORC2 contains Rictor, Deptor, mLST8, the mammalian stress-activated protein kinase interacting protein (mSIN1), and the protein observed with Rictor-1 (Protor-1) (17, 42).
mTOR can oversee multiple physiologic and disease processes such as cellular metabolism (69, 70), bone formation (71-73), diabetes (35, 43, 74-77), neurodegenerative disorders (37, 78-83), dementia (12, 84-86), and cancer (25, 27, 87-91). In addition, mTOR has a significant role in the modulation of autophagy induction (92). Important in the signaling cascade of mTOR is AMP activated protein kinase (AMPK). AMPK can prevent mTORC1 activity through the activation of the hamartin (tuberous sclerosis 1)/tuberin (tuberous sclerosis 2) (TSC1/TSC2) complex and can lead to the induction of autophagy (46, 58, 93-95).
In light of the close association between autophagy and mTOR, it may come as no surprise that circadian pathways are intimately linked to mTOR pathways. Melatonin, a pineal hormone that controls circadian rhythm, also relies upon autophagy pathways and mTOR to control processes of aging and neurodegeneration (3). Loss of mTOR activation may be involved with altered circadian rhythm and cognitive decline during prolonged space flight (96). Cerebral ischemic infarction also may be influenced by alteration in circadian rhythm genes and fluctuations in mTOR activity (66, 97). In regards to cancer, some studies suggest that loss of mammalian circadian clock proteins such as period2 (Per2) can lead to enhanced mTOR activity and chemotherapy drug resistance (98).
Pathways of mTOR and AMPK are also linked to SIRT1 (12, 99, 100). SIRT1, a member of the sirtuin family, is a histone deacetylase (51, 55, 101-106). SIRT1 can transfer acetyl groups from ε-N-acetyl lysine amino acids on the histones of DNA to control transcription and is dependent upon nicotinamide adenine dinucleotide (NAD+) as a substrate (105, 107-110). Nicotinamide phosphoribosyltransferase (NAMPT) catalyzes the conversion of nicotinamide to nicotinamide mononucleotide through the salvage pathway of NAD+ synthesis (70, 73, 108). Nicotinamide mononucleotide is subsequently converted to NAD+ by enzymes in the nicotinamide/nicotinic acid mononucleotide adenylyltransferase (NMNAT) family.
SIRT1 is involved in multiple disease processes that include cancer (106, 111-113), vascular disease (39, 114-117), altered cellular metabolism (12, 102, 103, 118, 119), diabetes (18, 120-123), and neurodegenerative disorders (106, 124, 125). Many of these processes require the modulation of autophagy by SIRT1 (12, 40, 126, 127). SIRT1 controls stem cell survival by modulating autophagic flux (128) and SIRT1 activity is increased in conjunction with AMPK to lead to autophagy and cellular protection (129). Importantly, SIRT1 can have an inverse relationship with mTOR in embryonic stem cells (58, 70) and block mTOR to promote autophagy and protect embryonic stem cells during oxidative stress (130).
In regards to the control of circadian rhythm, SIRT1 may be involved with altered circadian rhythm function that affects the development of disorders such as Alzheimer's disease (131). Increased SIRT1 activity with a disruption in circadian rhythm also may result in increased susceptibility to mammary carcinogenesis (132). Yet, SIRT1 may be beneficial under specific circumstances to regulate circadian rhythm gene expression that can foster hepatocellular proliferation and liver regeneration following liver resection (133).
Conclusions and Future Perspectives
Located in the SCN, the mammalian circadian clock is emerging as a critical component for several disease processes that include aging related disorders, neurodegenerative diseases, and cancer. Relying upon cellular signals and light input to align itself with solar time, the circadian clock rhythm controls behavior, normal physiology, and biochemical cellular signal transduction. Neurodegenerative disorders such as Alzheimer's disease and Parkinson's disease may progress in the setting of altered circadian rhythm dysfunction. In a similar manner, loss of a proper circadian rhythm may lead to increased risk for nasopharyngeal carcinoma, breast cancer, and metastatic colorectal cancer. Clinical behavior disruptions that involve prolonged shift work and even space travel may negatively impact circadian rhythm. At the cellular level, regulation of the pathways of autophagy, mTOR, AMPK, and SIRT1 as well as proliferative mechanisms that involve Wnt may be vital for the normal physiologic regulation of the body's circadian rhythm. As our knowledge continues to expand in regards to the significant role circadian clock genes hold for disease states, future targeting of the underlying pathways that control mammalian circadian rhythm may hold the key for the development of novel therapies against aging-related disorders, neurodegenerative disease, and tumorigenesis.
Acknowledgments
This research was supported by the following grants to Kenneth Maiese: American Diabetes Association, American Heart Association, NIH NIEHS, NIH NIA, NIH NINDS, and NIH ARRA.
Footnotes
Competing Interests: There are no conflicts of interest to declare.
References
- 1.Patel SA, Velingkaar NS, Kondratov RV. Transcriptional Control of Antioxidant Defense by the Circadian Clock. Antioxid Redox Signal. 2014;20(18):2997–3006. doi: 10.1089/ars.2013.5671. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Hardeland R. Melatonin and the pathologies of weakened or dysregulated circadian oscillators. J Pineal Res. 2016 doi: 10.1111/jpi.12377. [DOI] [PubMed] [Google Scholar]
- 3.Jenwitheesuk A, Nopparat C, Mukda S, Wongchitrat P, Govitrapong P. Melatonin regulates aging and neurodegeneration through energy metabolism, epigenetics, autophagy and circadian rhythm pathways. International journal of molecular sciences. 2014;15(9):16848–84. doi: 10.3390/ijms150916848. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Xydous M, Prombona A, Sourlingas TG. The role of H3K4me3 and H3K9/14ac in the induction by dexamethasone of Per1 and Sgk1, two glucococorticoid early response genes that mediate the effects of acute stress in mammals. Biochim Biophys Acta. 2014;1839(9):866–72. doi: 10.1016/j.bbagrm.2014.07.011. [DOI] [PubMed] [Google Scholar]
- 5.Lin F, Chen Y, Li X, Zhao Q, Tan Z. Over-expression of circadian clock gene Bmal1 affects proliferation and the canonical Wnt pathway in NIH-3T3 cells. Cell Biochem Funct. 2013;31(2):166–72. doi: 10.1002/cbf.2871. [DOI] [PubMed] [Google Scholar]
- 6.Bunney BG, Li JZ, Walsh DM, Stein R, Vawter MP, Cartagena P, et al. Circadian dysregulation of clock genes: clues to rapid treatments in major depressive disorder. Mol Psychiatry. 2015;20(1):48–55. doi: 10.1038/mp.2014.138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Hood S, Amir S. Neurodegeneration and the Circadian Clock. Frontiers in aging neuroscience. 2017;9:170. doi: 10.3389/fnagi.2017.00170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Vaccaro A, Issa AR, Seugnet L, Birman S, Klarsfeld A. Drosophila Clock Is Required in Brain Pacemaker Neurons to Prevent Premature Locomotor Aging Independently of Its Circadian Function. PLoS Genet. 2017;13(1):e1006507. doi: 10.1371/journal.pgen.1006507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Cronin P, McCarthy MJ, Lim ASP, Salmon DP, Galasko D, Masliah E, et al. Circadian alterations during early stages of Alzheimer's disease are associated with aberrant cycles of DNA methylation in BMAL1. Alzheimer's & dementia : the journal of the Alzheimer's Association. 2017;13(6):689–700. doi: 10.1016/j.jalz.2016.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Li SY, Wang YL, Liu WW, Lyu DJ, Wang F, Mao CJ, et al. Long-term Levodopa Treatment Accelerates the Circadian Rhythm Dysfunction in a 6-hydroxydopamine Rat Model of Parkinson's Disease. Chin Med J (Engl) 2017;130(9):1085–92. doi: 10.4103/0366-6999.204920. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Liu SL, Lin HX, Lin CY, Sun XQ, Ye LP, Qiu F, et al. TIMELESS confers cisplatin resistance in nasopharyngeal carcinoma by activating the Wnt/beta-catenin signaling pathway and promoting the epithelial mesenchymal transition. Cancer Lett. 2017;402:117–30. doi: 10.1016/j.canlet.2017.05.022. [DOI] [PubMed] [Google Scholar]
- 12.Maiese K. Novel nervous and multi-system regenerative therapeutic strategies for diabetes mellitus with mTOR. Neural regeneration research. 2016;11(3):372–85. doi: 10.4103/1673-5374.179032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Maiese K. Disease onset and aging in the world of circular RNAs. J Transl Sci. 2016;2(6):327–9. doi: 10.15761/jts.1000158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Maiese K, Li F, Chong ZZ, Shang YC. The Wnt signaling pathway: Aging gracefully as a protectionist? Pharmacol Ther. 2008;118(1):58–81. doi: 10.1016/j.pharmthera.2008.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Ortiz-Masia D, Cosin-Roger J, Calatayud S, Hernandez C, Alos R, Hinojosa J, et al. Hypoxic macrophages impair autophagy in epithelial cells through Wnt1: relevance in IBD. Mucosal immunology. 2014;7(4):929–38. doi: 10.1038/mi.2013.108. [DOI] [PubMed] [Google Scholar]
- 16.Chong ZZ, Shang YC, Maiese K. Vascular injury during elevated glucose can be mitigated by erythropoietin and Wnt signaling. Curr Neurovasc Res. 2007;4(3):194–204. doi: 10.2174/156720207781387150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Maiese K. Warming Up to New Possibilities with the Capsaicin Receptor TRPV1: mTOR, AMPK, and Erythropoietin. Curr Neurovasc Res. 2017;14(2):184–9. doi: 10.2174/1567202614666170313105337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Maiese K. New Insights for Oxidative Stress and Diabetes Mellitus. Oxid Med Cell Longev. 2015;2015(2015:875961) doi: 10.1155/2015/875961. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Shah N, Morsi Y, Manasseh R. From mechanical stimulation to biological pathways in the regulation of stem cell fate. Cell Biochem Funct. 2014;32(4):309–25. doi: 10.1002/cbf.3027. [DOI] [PubMed] [Google Scholar]
- 20.Sun TJ, Tao R, Han YQ, Xu G, Liu J, Han YF. Therapeutic potential of umbilical cord mesenchymal stem cells with Wnt/beta-catenin signaling pathway pre-activated for the treatment of diabetic wounds. European review for medical and pharmacological sciences. 2014;18(17):2460–4. [PubMed] [Google Scholar]
- 21.Chen X, Wang CC, Song SM, Wei SY, Li JS, Zhao SL, et al. The administration of erythropoietin attenuates kidney injury induced by ischemia/reperfusion with increased activation of Wnt/beta-catenin signaling. Journal of the Formosan Medical Association = Taiwan yi zhi. 2015;114(5):430–7. doi: 10.1016/j.jfma.2015.01.007. [DOI] [PubMed] [Google Scholar]
- 22.Chong ZZ, Shang YC, Maiese K. Cardiovascular Disease and mTOR Signaling. Trends Cardiovasc Med. 2011;21(5):151–5. doi: 10.1016/j.tcm.2012.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Maiese K. WISP1: Clinical Insights for a Proliferative and Restorative Member of the CCN Family. Curr Neurovasc Res. 2014;11(4):378–89. doi: 10.2174/1567202611666140912115107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Jia S, Qu T, Feng M, Ji K, Li Z, Jiang W, et al. Association of Wnt1-inducible signaling pathway protein-1 with the proliferation, migration and invasion in gastric cancer cells. Tumour biology : the journal of the International Society for Oncodevelopmental Biology and Medicine. 2017;39(6):1010428317699755. doi: 10.1177/1010428317699755. [DOI] [PubMed] [Google Scholar]
- 25.Maiese K. Stem cell guidance through the mechanistic target of rapamycin. World J Stem Cells. 2015;7(7):999–1009. doi: 10.4252/wjsc.v7.i7.999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Maiese K. Non-coding RNAs: Cracking the Code for Clinical Disease. Curr Neurovasc Res. 2016 doi: 10.2174/1567202614999161206161215. [DOI] [PubMed] [Google Scholar]
- 27.Maiese K, Chong ZZ, Shang YC, Wang S. mTOR: on target for novel therapeutic strategies in the nervous system. Trends Mol Med. 2013;19(1):51–60. doi: 10.1016/j.molmed.2012.11.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Tsai HC, Tzeng HE, Huang CY, Huang YL, Tsai CH, Wang SW, et al. WISP-1 positively regulates angiogenesis by controlling VEGF-A expression in human osteosarcoma. Cell death & disease. 2017;8(4):e2750. doi: 10.1038/cddis.2016.421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Yang CM, Ji S, Li Y, Fu LY, Jiang T, Meng FD. beta-Catenin promotes cell proliferation, migration, and invasion but induces apoptosis in renal cell carcinoma. OncoTargets and therapy. 2017;10:711–24. doi: 10.2147/OTT.S117933. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Wegrzyn LR, Tamimi RM, Rosner BA, Brown SB, Stevens RG, Eliassen AH, et al. ROTATING NIGHT SHIFT WORK AND RISK OF BREAST CANCER IN THE NURSES' HEALTH STUDIES. Am J Epidemiol. 2017 doi: 10.1093/aje/kwx140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Wang Y, Sun N, Lu C, Bei Y, Qian R, Hua L. Upregulation of circadian gene ‘hClock’ contribution to metastasis of colorectal cancer. Int J Oncol. 2017;50(6):2191–9. doi: 10.3892/ijo.2017.3987. [DOI] [PubMed] [Google Scholar]
- 32.Chen C, Lu Y, Siu HM, Guan J, Zhu L, Zhang S, et al. Identification of Novel Vacuolin-1 Analogues as Autophagy Inhibitors by Virtual Drug Screening and Chemical Synthesis. Molecules. 2017;22(6) doi: 10.3390/molecules22060891. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Di Rosa M, Distefano G, Gagliano C, Rusciano D, Malaguarnera L. AUTOPHAGY IN DIABETIC RETINOPATHY. Current neuropharmacology. 2016 doi: 10.2174/1570159X14666160321122900. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Klionsky DJ, Abdelmohsen K, Abe A, Abedin MJ, Abeliovich H, Acevedo Arozena A, et al. Guidelines for the use and interpretation of assays for monitoring autophagy (3rd edition) Autophagy. 2016;12(1):1–222. doi: 10.1080/15548627.2015.1100356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Maiese K. mTOR: Driving apoptosis and autophagy for neurocardiac complications of diabetes mellitus. World J Diabetes. 2015;6(2):217–24. doi: 10.4239/wjd.v6.i2.217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Maiese K, Chong ZZ, Shang YC, Wang S. Targeting disease through novel pathways of apoptosis and autophagy. Expert opinion on therapeutic targets. 2012;16(12):1203–14. doi: 10.1517/14728222.2012.719499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Maiese K. Driving neural regeneration through the mammalian target of rapamycin. Neural regeneration research. 2014;9(15):1413–7. doi: 10.4103/1673-5374.139453. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.White CR, Datta G, Giordano S. High-Density Lipoprotein Regulation of Mitochondrial Function. Adv Exp Med Biol. 2017;982:407–29. doi: 10.1007/978-3-319-55330-6_22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Maiese K. Programming apoptosis and autophagy with novel approaches for diabetes mellitus. Curr Neurovasc Res. 2015;12(2):173–88. doi: 10.2174/1567202612666150305110929. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Maiese K. FoxO Transcription Factors and Regenerative Pathways in Diabetes Mellitus. Curr Neurovasc Res. 2015;12(4):404–13. doi: 10.2174/1567202612666150807112524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Moors TE, Hoozemans JJ, Ingrassia A, Beccari T, Parnetti L, Chartier-Harlin MC, et al. Therapeutic potential of autophagy-enhancing agents in Parkinson's disease. Molecular neurodegeneration. 2017;12(1):11. doi: 10.1186/s13024-017-0154-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Chong ZZ, Shang YC, Wang S, Maiese K. Shedding new light on neurodegenerative diseases through the mammalian target of rapamycin. Prog Neurobiol. 2012;99(2):128–48. doi: 10.1016/j.pneurobio.2012.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Li L. The Molecular Mechanism of Glucagon-Like Peptide-1 Therapy in Alzheimer's Disease, Based on a Mechanistic Target of Rapamycin Pathway. CNS drugs. 2017 doi: 10.1007/s40263-017-0431-2. [DOI] [PubMed] [Google Scholar]
- 44.Maiese K. Taking aim at Alzheimer's disease through the mammalian target of rapamycin. Ann Med. 2014;46(8):587–96. doi: 10.3109/07853890.2014.941921. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Murphy KE, Park JJ. Can Co-Activation of Nrf2 and Neurotrophic Signaling Pathway Slow Alzheimer's Disease? International journal of molecular sciences. 2017;18(6) doi: 10.3390/ijms18061168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Zhang ZH, Wu QY, Zheng R, Chen C, Chen Y, Liu Q, et al. Selenomethionine mitigates cognitive decline by targeting both tau hyperphosphorylation and autophagic clearance in an Alzheimer's disease mouse model. J Neurosci. 2017 doi: 10.1523/JNEUROSCI.3229-16.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Maiese K. Targeting molecules to medicine with mTOR, autophagy, and neurodegenerative disorders. Br J Clin Pharmacol. 2015 doi: 10.1111/bcp.12804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Zhou Q, Chen B, Wang X, Wu L, Yang Y, Cheng X, et al. Sulforaphane protects against rotenone-induced neurotoxicity in vivo: Involvement of the mTOR, Nrf2, and autophagy pathways. Scientific reports. 2016;6:32206. doi: 10.1038/srep32206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Hyrskyluoto A, Reijonen S, Kivinen J, Lindholm D, Korhonen L. GADD34 mediates cytoprotective autophagy in mutant huntingtin expressing cells via the mTOR pathway. Exp Cell Res. 2012;318(1):33–42. doi: 10.1016/j.yexcr.2011.08.020. [DOI] [PubMed] [Google Scholar]
- 50.Lee JH, Tecedor L, Chen YH, Monteys AM, Sowada MJ, Thompson LM, et al. Reinstating aberrant mTORC1 activity in Huntington's disease mice improves disease phenotypes. Neuron. 2015;85(2):303–15. doi: 10.1016/j.neuron.2014.12.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Maiese K. FoxO Proteins in the Nervous System. Anal Cell Pathol (Amst) 2015;2015:569392. doi: 10.1155/2015/569392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Gurlo T, Rivera JF, Butler AE, Cory M, Hoang J, Costes S, et al. CHOP Contributes to, But Is Not the Only Mediator of, IAPP Induced beta-Cell Apoptosis. Mol Endocrinol. 2016;30(4):446–54. doi: 10.1210/me.2015-1255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Qi Z, Xia J, Xue X, He Q, Ji L, Ding S. Long-term treatment with nicotinamide induces glucose intolerance and skeletal muscle lipotoxicity in normal chow-fed mice: compared to diet-induced obesity. The Journal of nutritional biochemistry. 2016;36:31–41. doi: 10.1016/j.jnutbio.2016.07.005. [DOI] [PubMed] [Google Scholar]
- 54.Crino PB. The mTOR signalling cascade: paving new roads to cure neurological disease. Nature reviews Neurology. 2016 doi: 10.1038/nrneurol.2016.81. [DOI] [PubMed] [Google Scholar]
- 55.Maiese K. Forkhead transcription factors: new considerations for alzheimer's disease and dementia. J Transl Sci. 2016;2(4):241–7. doi: 10.15761/JTS.1000146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Dong W, Wang R, Ma LN, Xu BL, Zhang JS, Zhao ZW, et al. Influence of age-related learning and memory capacity of mice: different effects of a high and low caloric diet. Aging Clin Exp Res. 2016;28(2):303–11. doi: 10.1007/s40520-015-0398-0. [DOI] [PubMed] [Google Scholar]
- 57.Maiese K. The bright side of reactive oxygen species: lifespan extension without cellular demise. J Transl Sci. 2016;2(3):185–7. doi: 10.15761/JTS.1000138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Maiese K. Erythropoietin and mTOR: A “One-Two Punch” for Aging-Related Disorders Accompanied by Enhanced Life Expectancy. Curr Neurovasc Res. 2016;13(4):329–40. doi: 10.2174/1567202613666160729164900. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Milisav I, Poljsak B, Ribaric S. Reduced risk of apoptosis: mechanisms of stress responses. Apoptosis. 2016 doi: 10.1007/s10495-016-1317-3. [DOI] [PubMed] [Google Scholar]
- 60.Vaiserman AM, Lushchak OV, Koliada AK. Anti-aging pharmacology: Promises and pitfalls. Ageing research reviews. 2016;31:9–35. doi: 10.1016/j.arr.2016.08.004. [DOI] [PubMed] [Google Scholar]
- 61.Bondy S, Maiese K. Aging and Age-Related Disorders. Springer Science; 2010. [Google Scholar]
- 62.Ratliff EP, Mauntz RE, Kotzebue RW, Gonzalez A, Achal M, Barekat A, et al. Aging and Autophagic Function Influences the Progressive Decline of Adult Drosophila Behaviors. PLoS One. 2015;10(7):e0132768. doi: 10.1371/journal.pone.0132768. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Chen X, Kondo K, Motoki K, Homma H, Okazawa H. Fasting activates macroautophagy in neurons of Alzheimer's disease mouse model but is insufficient to degrade amyloid-beta. Scientific reports. 2015;5:12115. doi: 10.1038/srep12115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.He Y, Cornelissen-Guillaume GG, He J, Kastin AJ, Harrison LM, Pan W. Circadian rhythm of autophagy proteins in hippocampus is blunted by sleep fragmentation. Chronobiology international. 2016;33(5):553–60. doi: 10.3109/07420528.2015.1137581. [DOI] [PubMed] [Google Scholar]
- 65.Min JJ, Huo XL, Xiang LY, Qin YQ, Chai KQ, Wu B, et al. Protective effect of Dl-3n-butylphthalide on learning and memory impairment induced by chronic intermittent hypoxia-hypercapnia exposure. Scientific reports. 2014;4:5555. doi: 10.1038/srep05555. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Rami A, Rawashdeh O. The hippocampal autophagic machinery is depressed in the absence of the circadian clock protein PER1 that may lead to vulnerability during cerebral ischemia. Curr Neurovasc Res. 2017 doi: 10.2174/1567202614666170619083239. (in press) [DOI] [PubMed] [Google Scholar]
- 67.Gulhati P, Bowen KA, Liu J, Stevens PD, Rychahou PG, Chen M, et al. mTORC1 and mTORC2 regulate EMT, motility, and metastasis of colorectal cancer via RhoA and Rac1 signaling pathways. Cancer Res. 2011;71(9):3246–56. doi: 10.1158/0008-5472.CAN-10-4058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Zoncu R, Efeyan A, Sabatini DM. mTOR: from growth signal integration to cancer, diabetes and ageing. Nat Rev Mol Cell Biol. 2011;12(1):21–35. doi: 10.1038/nrm3025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Lipina C, Hundal HS. Is REDD1 a Metabolic Eminence Grise? Trends Endocrinol Metab. 2016 doi: 10.1016/j.tem.2016.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Maiese K, Chong ZZ, Shang YC, Wang S. Novel directions for diabetes mellitus drug discovery. Expert opinion on drug discovery. 2013;8(1):35–48. doi: 10.1517/17460441.2013.736485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Joeng KS, Lee YC, Lim J, Chen Y, Jiang MM, Munivez E, et al. Osteocyte-specific WNT1 regulates osteoblast function during bone homeostasis. J Clin Invest. 2017 doi: 10.1172/JCI92617. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Ma J, Li M, Hock J, Yu X. Hyperactivation of mTOR critically regulates abnormal osteoclastogenesis in neurofibromatosis Type 1. J Orthop Res. 2012;30(1):144–52. doi: 10.1002/jor.21497. [DOI] [PubMed] [Google Scholar]
- 73.Maiese K. Picking a bone with WISP1 (CCN4): new strategies against degenerative joint disease. J Transl Sci. 2016;1(3):83–5. [PMC free article] [PubMed] [Google Scholar]
- 74.Chong ZZ, Maiese K. Mammalian Target of Rapamycin Signaling in Diabetic Cardiovascular Disease. Cardiovasc Diabetol. 2012;11(1):45. doi: 10.1186/1475-2840-11-45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Crespo MC, Tome-Carneiro J, Pintado C, Davalos A, Visioli F, Burgos-Ramos E. Hydroxytyrosol restores proper insulin signaling in an astrocytic model of Alzheimer's disease. BioFactors (Oxford, England) 2017 doi: 10.1002/biof.1356. [DOI] [PubMed] [Google Scholar]
- 76.Lin Z, Li X, Zhan X, Sun L, Gao J, Cao Y, et al. Construction of competitive endogenous RNA network reveals regulatory role of long non-coding RNAs in type 2 diabetes mellitus. J Cell Mol Med. 2017 doi: 10.1111/jcmm.13224. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Maiese K, Chong ZZ, Wang S, Shang YC. Oxidant Stress and Signal Transduction in the Nervous System with the PI 3-K, Akt, and mTOR Cascade. International journal of molecular sciences. 2013;13(11):13830–66. doi: 10.3390/ijms131113830. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Chong ZZ, Shang YC, Wang S, Maiese K. PRAS40 Is an Integral Regulatory Component of Erythropoietin mTOR Signaling and Cytoprotection. PLoS ONE. 2012;7(9):e45456. doi: 10.1371/journal.pone.0045456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Dijkstra AA, Ingrassia A, de Menezes RX, van Kesteren RE, Rozemuller AJ, Heutink P, et al. Evidence for Immune Response, Axonal Dysfunction and Reduced Endocytosis in the Substantia Nigra in Early Stage Parkinson's Disease. PLoS One. 2015;10(6):e0128651. doi: 10.1371/journal.pone.0128651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Park YS, Park JH, Ko J, Shin IC, Koh HC. mTOR inhibition by rapamycin protects against deltamethrin-induced apoptosis in PC12 Cells. Environmental toxicology. 2015 doi: 10.1002/tox.22216. [DOI] [PubMed] [Google Scholar]
- 81.Sasazawa Y, Sato N, Umezawa K, Simizu S. Conophylline protects cells in cellular models of neurodegenerative diseases by inducing mammalian target of rapamycin (mTOR)-independent autophagy. J Biol Chem. 2015;290(10):6168–78. doi: 10.1074/jbc.M114.606293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Shang YC, Chong ZZ, Wang S, Maiese K. WNT1 Inducible Signaling Pathway Protein 1 (WISP1) Targets PRAS40 to Govern beta-Amyloid Apoptotic Injury of Microglia. Curr Neurovasc Res. 2012;9(4):239–49. doi: 10.2174/156720212803530618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Tourette C, Li B, Bell R, O'Hare S, Kaltenbach LS, Mooney SD, et al. A Large-scale Huntingtin Protein Interaction Network Implicates Rho GTPase Signaling Pathways in Huntington's Disease. J Biol Chem. 2014;289(10):6709–26. doi: 10.1074/jbc.M113.523696. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Bellozi PM, Lima IV, Doria JG, Vieira EL, Campos AC, Candelario-Jalil E, et al. Neuroprotective effects of the anticancer drug NVP-BEZ235 (dactolisib) on amyloid-beta 1-42 induced neurotoxicity and memory impairment. Scientific reports. 2016;6:25226. doi: 10.1038/srep25226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Chen L, Zhang Y, Li D, Zhang N, Liu R, Han B, et al. Everolimus (RAD001) ameliorates vascular cognitive impairment by regulating microglial function via the mTORC1 signaling pathway. J Neuroimmunol. 2016;299:164–71. doi: 10.1016/j.jneuroim.2016.09.008. [DOI] [PubMed] [Google Scholar]
- 86.Park JA, Lee CH. Temporal changes in mammalian target of rapamycin (mTOR) and phosphorylated-mTOR expressions in the hippocampal CA1 region of rat with vascular dementia. Journal of veterinary science. 2017;18(1):11–6. doi: 10.4142/jvs.2017.18.1.11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Chong ZZ, Shang YC, Zhang L, Wang S, Maiese K. Mammalian target of rapamycin: hitting the bull's-eye for neurological disorders. Oxid Med Cell Longev. 2010;3(6):374–91. doi: 10.4161/oxim.3.6.14787. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Karthik GM, Ma R, Lovrot J, Kis LL, Lindh C, Blomquist L, et al. mTOR inhibitors counteract tamoxifen-induced activation of breast cancer stem cells. Cancer Lett. 2015;367(1):76–87. doi: 10.1016/j.canlet.2015.07.017. [DOI] [PubMed] [Google Scholar]
- 89.Kolev VN, Wright QG, Vidal CM, Ring JE, Shapiro IM, Ricono J, et al. PI3K/mTOR dual inhibitor VS-5584 preferentially targets cancer stem cells. Cancer Res. 2015;75(2):446–55. doi: 10.1158/0008-5472.CAN-14-1223. [DOI] [PubMed] [Google Scholar]
- 90.Malla R, Ashby CR, Jr, Narayanan NK, Narayanan B, Faridi JS, Tiwari AK. Proline-rich AKT substrate of 40-kDa (PRAS40) in the pathophysiology of cancer. Biochem Biophys Res Commun. 2015;463(3):161–6. doi: 10.1016/j.bbrc.2015.05.041. [DOI] [PubMed] [Google Scholar]
- 91.Tauasioudi K, Sakellariou S, Levidou G, Theodorou D, Muichalopoulos NV, Patsouris E, et al. Immunohistochemical and molecular analysis of PI3K/AKT/mTOR pathway in esophageal carcinoma. APMIS : acta pathologica, microbiologica, et immunologica Scandinavica. 2015 doi: 10.1111/apm.12398. [DOI] [PubMed] [Google Scholar]
- 92.Maiese K. Molecules to Medicine with mTOR: Translating Critical Pathways into Novel Therapeutic Strategies. Elsevier and Academic Press; 2016. [Google Scholar]
- 93.Duan P, Hu C, Quan C, Yu T, Zhou W, Yuan M, et al. 4-Nonylphenol induces apoptosis, autophagy and necrosis in Sertoli cells: Involvement of ROS-mediated AMPK/AKT-mTOR and JNK pathways. Toxicology. 2016 doi: 10.1016/j.tox.2016.01.004. [DOI] [PubMed] [Google Scholar]
- 94.Hughes MA, Downs RM, Webb GW, Crocker CL, Kinsey ST, Baumgarner BL. Acute high-caffeine exposure increases autophagic flux and reduces protein synthesis in C2C12 skeletal myotubes. Journal of muscle research and cell motility. 2017 doi: 10.1007/s10974-017-9473-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Zhang C, Li C, Chen S, Li Z, Ma L, Jia X, et al. Hormetic effect of panaxatriol saponins confers neuroprotection in PC12 cells and zebrafish through PI3K/AKT/mTOR and AMPK/SIRT1/FOXO3 pathways. Scientific reports. 2017;7:41082. doi: 10.1038/srep41082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Wu X, Li D, Liu J, Diao L, Ling S, Li Y, et al. Dammarane Sapogenins Ameliorates Neurocognitive Functional Impairment Induced by Simulated Long-Duration Spaceflight. Frontiers in pharmacology. 2017;8:315. doi: 10.3389/fphar.2017.00315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Beker MC, Caglayan B, Yalcin E, Caglayan AB, Turkseven S, Gurel B, et al. Time-of-Day Dependent Neuronal Injury After Ischemic Stroke: Implication of Circadian Clock Transcriptional Factor Bmal1 and Survival Kinase AKT. Mol Neurobiol. 2017 doi: 10.1007/s12035-017-0524-4. [DOI] [PubMed] [Google Scholar]
- 98.Chen B, Tan Y, Liang Y, Li Y, Chen L, Wu S, et al. Per2 participates in AKT-mediated drug resistance in A549/DDP lung adenocarcinoma cells. Oncology letters. 2017;13(1):423–8. doi: 10.3892/ol.2016.5430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Ma L, Dong W, Wang R, Li Y, Xu B, Zhang J, et al. Effect of caloric restriction on the SIRT1/mTOR signaling pathways in senile mice. Brain Res Bull. 2015;116:67–72. doi: 10.1016/j.brainresbull.2015.06.004. [DOI] [PubMed] [Google Scholar]
- 100.Maiese K. MicroRNAs and SIRT1: A Strategy for Stem Cell Renewal and Clinical Development? J Transl Sci. 2015;1(3):55–7. [PMC free article] [PubMed] [Google Scholar]
- 101.Charles S, Raj V, Arokiaraj J, Mala K. Caveolin1/protein arginine methyltransferase1/sirtuin1 axis as a potential target against endothelial dysfunction. Pharmacol Res. 2017;119:1–11. doi: 10.1016/j.phrs.2017.01.022. [DOI] [PubMed] [Google Scholar]
- 102.Chong ZZ, Shang YC, Wang S, Maiese K. SIRT1: New avenues of discovery for disorders of oxidative stress. Expert opinion on therapeutic targets. 2012;16(2):167–78. doi: 10.1517/14728222.2012.648926. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Cui L, Guo J, Zhang Q, Yin J, Li J, Zhou W, et al. Erythropoietin activates SIRT1 to protect human cardiomyocytes against doxorubicin-induced mitochondrial dysfunction and toxicity. Toxicol Lett. 2017;275:28–38. doi: 10.1016/j.toxlet.2017.04.018. [DOI] [PubMed] [Google Scholar]
- 104.Geng C, Xu H, Zhang Y, Gao Y, Li M, Liu X, et al. Retinoic acid ameliorates high-fat diet-induced liver steatosis through sirt1. Science China Life sciences. 2017 doi: 10.1007/s11427-016-9027-6. [DOI] [PubMed] [Google Scholar]
- 105.Hwang ES, Song SB. Nicotinamide is an inhibitor of SIRT1 in vitro, but can be a stimulator in cells. Cell Mol Life Sci. 2017 doi: 10.1007/s00018-017-2527-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Maiese K. SIRT1 and stem cells: In the forefront with cardiovascular disease, neurodegeneration and cancer. World J Stem Cells. 2015;7(2):235–42. doi: 10.4252/wjsc.v7.i2.235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Bruckbauer A, Banerjee J, Cao Q, Cui X, Jing J, Zha L, et al. Leucine-nicotinic acid synergy stimulates AMPK/Sirt1 signaling and regulates lipid metabolism and lifespan in Caenorhabditis elegans, and hyperlipidemia and atherosclerosis in mice. American journal of cardiovascular disease. 2017;7(2):33–47. [PMC free article] [PubMed] [Google Scholar]
- 108.Chong ZZ, Wang S, Shang YC, Maiese K. Targeting cardiovascular disease with novel SIRT1 pathways. Future Cardiol. 2012;8(1):89–100. doi: 10.2217/fca.11.76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Li WY, Ren JH, Tao NN, Ran LK, Chen X, Zhou HZ et al. The SIRT1 inhibitor, nicotinamide, inhibits hepatitis B virus replication in vitro and in vivo. Archives of virology. 2015 doi: 10.1007/s00705-015-2712-8. [DOI] [PubMed] [Google Scholar]
- 110.Maiese K, Chong ZZ, Shang YC, Wang S. Translating cell survival and cell longevity into treatment strategies with SIRT1. Rom J Morphol Embryol. 2011;52(4):1173–85. [PMC free article] [PubMed] [Google Scholar]
- 111.Lee JS, Park JR, Kwon OS, Lee TH, Nakano I, Miyoshi H, et al. SIRT1 is required for oncogenic transformation of neural stem cells and for the survival of “cancer cells with neural stemness” in a p53-dependent manner. Neuro-oncology. 2014;17(1):95–106. doi: 10.1093/neuonc/nou145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Lin L, Zheng X, Qiu C, Dongol S, Lv Q, Jiang J, et al. SIRT1 promotes endometrial tumor growth by targeting SREBP1 and lipogenesis. Oncology reports. 2014;32(6):2831–5. doi: 10.3892/or.2014.3521. [DOI] [PubMed] [Google Scholar]
- 113.Ziebarth AJ, Nowsheen S, Steg AD, Shah MM, Katre AA, Dobbin ZC, et al. Endoglin (CD105) contributes to platinum resistance and is a target for tumor-specific therapy in epithelial ovarian cancer. Clin Cancer Res. 2013;19(1):170–82. doi: 10.1158/1078-0432.CCR-12-1045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.D'Onofrio N, Vitiello M, Casale R, Servillo L, Giovane A, Balestrieri ML. Sirtuins in vascular diseases: Emerging roles and therapeutic potential. Biochim Biophys Acta. 2015;1852(7):1311–22. doi: 10.1016/j.bbadis.2015.03.001. [DOI] [PubMed] [Google Scholar]
- 115.Hung CH, Chan SH, Chu PM, Tsai KL. Quercetin is a potent anti-atherosclerotic compound by activation of SIRT1 signaling under oxLDL stimulation. Molecular nutrition & food research. 2015 doi: 10.1002/mnfr.201500144. [DOI] [PubMed] [Google Scholar]
- 116.Saboori S, Koohdani F, Nematipour E, Yousefi Rad E, Saboor-Yaraghi AA, Javanbakht MH, et al. Beneficial effects of omega-3 and vitamin E coadministration on gene expression of SIRT1 and PGC1alpha and serum antioxidant enzymes in patients with coronary artery disease. Nutrition, metabolism, and cardiovascular diseases : NMCD. 2016;26(6):489–94. doi: 10.1016/j.numecd.2015.11.013. [DOI] [PubMed] [Google Scholar]
- 117.Shao Y, Lv C, Wu C, Zhou Y, Wang Q. Mir-217 promotes inflammation and fibrosis in high glucose cultured rat glomerular mesangial cells via Sirt1/HIF-1alpha signaling pathway. Diabetes Metab Res Rev. 2016;32(6):534–43. doi: 10.1002/dmrr.2788. [DOI] [PubMed] [Google Scholar]
- 118.Guo P, Wang D, Wang X, Feng H, Tang Y, Sun R, et al. Effect and mechanism of fuzhisan and donepezil on the sirtuin 1 pathway and amyloid precursor protein metabolism in PC12 cells. Molecular medicine reports. 2016;13(4):3539–46. doi: 10.3892/mmr.2016.4957. [DOI] [PubMed] [Google Scholar]
- 119.Wang L, Teng R, Di L, Rogers H, Wu H, Kopp JB, et al. PPARalpha and Sirt1 mediate erythropoietin action in increasing metabolic activity and browning of white adipocytes to protect against obesity and metabolic disorders. Diabetes. 2013;62(12):4122–31. doi: 10.2337/db13-0518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Arunachalam G, Lakshmanan AP, Samuel SM, Triggle CR, Ding H. Molecular Interplay between microRNA-34a and Sirtuin1 in Hyperglycemia-Mediated Impaired Angiogenesis in Endothelial Cells: Effects of Metformin. J Pharmacol Exp Ther. 2016;356(2):314–23. doi: 10.1124/jpet.115.226894. [DOI] [PubMed] [Google Scholar]
- 121.Lamoke F, Shaw S, Yuan J, Ananth S, Duncan M, Martin P, et al. Increased Oxidative and Nitrative Stress Accelerates Aging of the Retinal Vasculature in the Diabetic Retina. PLoS One. 2015;10(10):e0139664. doi: 10.1371/journal.pone.0139664. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Poulose N, Raju R. Sirtuin regulation in aging and injury. Biochim Biophys Acta. 2015;1852(11):2442–55. doi: 10.1016/j.bbadis.2015.08.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Vikram A, Kim YR, Kumar S, Li Q, Kassan M, Jacobs JS, et al. Vascular microRNA-204 is remotely governed by the microbiome and impairs endothelium-dependent vasorelaxation by downregulating Sirtuin1. Nature communications. 2016;7:12565. doi: 10.1038/ncomms12565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Duan W. Sirtuins: from metabolic regulation to brain aging. Frontiers in aging neuroscience. 2013;5:36. doi: 10.3389/fnagi.2013.00036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Martin A, Tegla CA, Cudrici CD, Kruszewski AM, Azimzadeh P, Boodhoo D, et al. Role of SIRT1 in autoimmune demyelination and neurodegeneration. Immunologic research. 2015;61(3):187–97. doi: 10.1007/s12026-014-8557-5. [DOI] [PubMed] [Google Scholar]
- 126.Ma L, Fu R, Duan Z, Lu J, Gao J, Tian L, et al. Sirt1 is essential for resveratrol enhancement of hypoxia-induced autophagy in the type 2 diabetic nephropathy rat. Pathology, research and practice. 2016 doi: 10.1016/j.prp.2016.02.001. [DOI] [PubMed] [Google Scholar]
- 127.Ramalinga M, Roy A, Srivastava A, Bhattarai A, Harish V, Suy S, et al. MicroRNA-212 negatively regulates starvation induced autophagy in prostate cancer cells by inhibiting SIRT1 and is a modulator of angiogenesis and cellular senescence. Oncotarget. 2015 doi: 10.18632/oncotarget.5920. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Mazzoccoli G, Tevy MF, Borghesan M, Delle Vergini MR, Vinciguerra M. Caloric restriction and aging stem cells: the stick and the carrot? Exp Gerontol. 2014;50:137–48. doi: 10.1016/j.exger.2013.10.014. [DOI] [PubMed] [Google Scholar]
- 129.Jin X, Chen M, Yi L, Chang H, Zhang T, Wang L, et al. Delphinidin-3-glucoside protects human umbilical vein endothelial cells against oxidized low-density lipoprotein-induced injury by autophagy upregulation via the AMPK/SIRT1 signaling pathway. Molecular nutrition & food research. 2014;58(10):1941–51. doi: 10.1002/mnfr.201400161. [DOI] [PubMed] [Google Scholar]
- 130.Ou X, Lee MR, Huang X, Messina-Graham S, Broxmeyer HE. SIRT1 positively regulates autophagy and mitochondria function in embryonic stem cells under oxidative stress. Stem Cells. 2014;32(5):1183–94. doi: 10.1002/stem.1641. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Bellanti F, Iannelli G, Blonda M, Tamborra R, Villani R, Romano A, et al. Alterations of Clock Gene RNA Expression in Brain Regions of a Triple Transgenic Model of Alzheimer's Disease. J Alzheimers Dis. 2017 doi: 10.3233/JAD-160942. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Fang M, Ohman Strickland PA, Kang HG, Zarbl H. Uncoupling genotoxic stress responses from circadian control increases susceptibility to mammary carcinogenesis. Oncotarget. 2017;8(20):32752–68. doi: 10.18632/oncotarget.15678. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Bellet MM, Masri S, Astarita G, Sassone-Corsi P, Della Fazia MA, Servillo G. Histone Deacetylase SIRT1 Controls Proliferation, Circadian Rhythm, and Lipid Metabolism during Liver Regeneration in Mice. J Biol Chem. 2016;291(44):23318–29. doi: 10.1074/jbc.M116.737114. [DOI] [PMC free article] [PubMed] [Google Scholar]