Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Sep 11.
Published in final edited form as: Curr Neurovasc Res. 2015;12(4):404–413. doi: 10.2174/1567202612666150807112524

FoxO Transcription Factors and Regenerative Pathways in Diabetes Mellitus

Kenneth Maiese 1,*
PMCID: PMC4567483  NIHMSID: NIHMS714257  PMID: 26256004

Abstract

Mammalian forkhead transcription factors of the O class (FoxO) are exciting targets under consideration for the development of new clinical entities to treat metabolic disorders and diabetes mellitus (DM). DM, a disorder that currently affects greater than 350 million individuals globally, can become a devastating disease that leads to cellular injury through oxidative stress pathways and affects multiple systems of the body. FoxO proteins can regulate insulin signaling, gluconeogenesis, insulin resistance, immune cell migration, and cell senescence. FoxO proteins also control cell fate through oxidative stress and pathways of autophagy and apoptosis that either lead to tissue regeneration or cell demise. Furthermore, FoxO signaling can be dependent upon signal transduction pathways that include silent mating type information regulation 2 homolog 1 (S. cerevisiae) (SIRT1), Wnt, and Wnt1 inducible signaling pathway protein 1 (WISP1). Cellular metabolic pathways driven by FoxO proteins are complex, can lead to variable clinical outcomes, and require in-depth analysis of the epigenetic and post-translation protein modifications that drive FoxO protein activation and degradation.

Keywords: Akt, apoptosis, autophagy, β-catenin, caspase, CCN, diabetes mellitus, epigenetic, erythropoietin, forkhead, FoxO, metabolism, nicotinamide, oxidative stress, sirtuins, SgK, SIRT1, stem cells, WISP, Wnt

1. Diabetes Mellitus: Clinical Implications

Diabetes mellitus (DM) is a devastating disease that can involve any system of the body (1, 2). In the nervous system, DM can result in cerebral ischemia stroke (3-8), retinal disease (9-12), peripheral nerve disorders (13, 14), dementia such as Alzheimer's disease (15-18), and psychiatric disorders (19, 20). Vascular disease can be mediated by DM and result in endothelial cell senescence (21), injury to endothelial cells (22-28), cardiovascular disease (29-37), atherosclerosis (8, 38-40), platelet dysfunction (3, 41), loss of endothelial progenitor cells (42-47), and impaired angiogenesis (27, 35, 48). Given the broad disorders that can be a result of DM in the nervous and cardiovascular systems, it comes as no surprise that DM can have profound negative effects on the immune system (36, 49-55), renal function (56-60), hepatic metabolism (13, 61-66), and musculoskeletal integrity (38, 67-70).

The incidence of DM is increasing throughout the world (71). Approximately 350 million individuals currently have DM, but the World Health Organization estimates that DM will be the seventh leading cause of death by the year 2030 (72). In developed countries such as the United States that have an estimated 21 million individuals with DM (73), it is surprising to learn that another 8 million individuals are believed to suffer from metabolic disorders but remain undiagnosed (12, 74, 75). Costs to care for individuals with DM are significant. Almost $9,000 USD are spent in the United States for each individual with DM per year and overall care for patients with DM consumes 17% of the Gross Domestic Product (76).

2. Cellular Injury with Diabetes Mellitus

At the cellular level, a significant mediator that leads to injury during DM involves oxidative stress and the release of reactive oxygen species (ROS) (8, 13, 77-80). Several genetic polymorphisms in oxidative stress pathways have recently been linked to the complications of DM (81). Agents that can promote the complications of DM, such as advanced glycation end products (AGEs) (30), result in the release of ROS, caspase activation (47, 82-84), and loss of anti-oxidant levels (85). In the nervous system, oxidative stress during DM can affect cognition (86), behavior (19), retinal nerve function (25), and brain mitochondria (52, 87-91). In the cardiac system, oxidative stress during DM can be responsible for cardiomyopathy (37, 92), myocardial infarction and re-perfusion injury (93), cardiomyocyte injury (94), and fibrosis (37). Vascular cell and endothelial cell dysfunction occurs during DM through oxidative stress pathways that involve impairment of stem cell and progenitor cell mobilization (42, 44), endothelial senescence (21), inhibition of angiogenesis (48), vascular aging (95), and endothelial cell injury (22-24, 46, 96-98). Pancreatic human islet cells also are susceptible to oxidative stress injury during DM (99, 100) and high lipid exposure (101). In clinical studies, poor glycemic control in DM patients can lead to the depression of endothelial progenitor cell levels (43). Patients with DM have serum markers of oxidative stress with ischemia-modified albumin (102).

During oxidative stress with DM, cell death can result from programmed cell death pathways of autophagy and apoptosis. Apoptosis has both an early phase that involves the externalization of plasma membrane lipid phosphatidylserine (PS) residues and a subsequent phase that consists of genomic DNA degradation (103-105). Macrophages and microglia can be prevented from engulfing otherwise functional cells that are tagged with PS residues by blocking membrane PS externalization (2, 106, 107). The later phase of apoptosis with DNA destruction is not readily reversible (108-113). In experimental studies with diabetic rats, elevated glucose can lead to oxidative stress and apoptotic injury in areas of the prefrontal cortex of the brain (19). Hyperglycemia not only leads to endothelial senescence, but also results in the presence of apoptotic makers and cell death on endothelial cells (21, 23, 96, 97). Elevated glucose in diabetic mice can injure pancreatic islet cells and lead to apoptotic cell death (99).

In reference to autophagy, this is a process that removes non-functional organelles and recycles cytoplasmic components to remodel tissue (105, 114-117). During macroautophagy, cytoplasmic proteins and organelles are sequestered into autophagosomes and then combined with lysosomes for degradation and are recycled for future cellular processes (32, 46, 71, 118, 119). Under some circumstances, autophagy may protect cells during DM by eliminating non-functional organelles and may be less of a significant mediator of cell injury (120). Autophagy removes misfolded proteins and eliminates non-functioning mitochondria to prevent β-cell dysfunction and the onset of DM (121). Autophagy has been reported to improve insulin sensitivity during high fat diets in mice (69). Exercise in murine models can foster the induction of autophagy and regulate glucose homeostasis (122). Loss of autophagy with haploinsufficiency of an essential Atg7 gene in murine models of obesity results in increased insulin resistance with elevated lipids and inflammation (118). It is important to note that autophagy and apoptosis are closely tied in DM to influence cell survival. Autophagy may protect cardiomyocytes from apoptotic cell death during DM (32).

Autophagy has a detrimental side during DM (123). Increased activity of autophagy can result in the loss of cardiac and liver tissue in diabetic rats during diet modification in attempts to achieve glycemic control (64). AGEs lead to atherosclerosis (38) and cardiomyopathy (92) through the induction of autophagy. Autophagy has been reported to injure endothelial progenitor cells, promote mitochondrial oxidative and endoplasmic reticulum stress (124), and block angiogenesis (46) during exposure to elevated glucose.

3. Targeting Forkhead Transcription Factors of the “O” Class

Mammalian forkhead transcription factors assigned to the O class represent a novel target for drug development to treat metabolic disorders and DM (125, 126) (Table 1). Mammalian FOXO proteins, FOXO1, FOXO3, FOXO4, and FOXO6 (127), have a butterfly-like appearance on X-ray crystallography (128) and nuclear magnetic resonance (129). The forkhead box (FOX) family of genes has a conserved forkhead domain described as a “winged helix” (130). In regards to nomenclature, all letters are capitalized for human Fox proteins (131). Only the initial letter is listed as uppercase for the mouse and for all other chordates the initial and subclass letters are in uppercase (34, 113, 132, 133).

Table 1. FoxO Transcription Factors and Diabetes Mellitus.

FoxO Proteins Biology
Expression and Structure FoxO proteins possess a butterfly-like appearance on X-ray crystallography and nuclear magnetic resonance
FoxO proteins are present throughout the body and have a conserved forkhead domain described as a “winged helix”
Post -Translational Modification FoxO proteins are modified by phosphorylation, acetylation, and ubiquitylation through pathways involving Akt, SgK, SIRT1, and Wnt signaling with WISP1
Akt and SgK phosphorylate FoxO proteins at different sites affording various pathways to control FoxO protein activity
Programmed Cell Death Under some conditions, the activation of FoxO proteins may prevent apoptotic cellular injury during oxidative stress
FoxO protein activation can lead to the induction of autophagy and increase cell survival, such as during the clearance of toxic mHtt protein in neurons and during starvation to maintain cardiac function
However, activation of FoxO proteins usually leads to apoptotic membrane PS externalization and DNA degradation. In some circumstances, a reduction in autophagy is required to block oxidative stress injury
Cellular Signaling Wnt signaling can promote cellular protection against apoptotic cell death through the inactivation of FoxO proteins
WISP1 can limit FoxO3a activity, block caspase 1 and 3 activity, and promote SIRT1 nuclear trafficking
FoxO proteins can control SIRT1 transcription and increase SIRT1 expression. In addition, SIRT1 promotes FoxO-driven SIRT1 autotranscription through the activation and deacetylation of FoxO proteins
Agents that are protective during DM, such as EPO and nicotinamide, can phosphorylate and inactive FoxO proteins to lead to cellular protection
Outcomes with Diabetes Mellitus Some clinical studies suggest a detrimental effect associated with FoxO proteins and DM
Inactivation of FoxO proteins during DM may protect pancreatic β-cells, retinal cells, renal cells, and glucose homeostasis
In some clinical populations, FoxO protein activation may be protective during metabolic disorders for the immune system, assist with insulin signaling, and maintain energy reserves

Akt: protein kinase B; DM: diabetes mellitus; DNA: deoxyribonucleic acid; EPO: erythropoietin; FoxO: mammalian forkhead transcription factors of the O class; mHtt: mutant Huntingtin; PS: phosphatidylserine; SgK: serum- and glucocorticoid-inducible protein kinase; SIRT1: silent mating type information regulation 2 homolog 1 (S. cerevisiae); WISP1: wnt1 inducible signaling pathway protein 1; Wnt: proteins derived from the Drosophila Wingless (Wg) and the mouse Int-1 genes

FoxO proteins are found throughout the body. In regards to metabolic signaling, FoxO proteins are conserved among multiple species that include Caenorhabditis elegans, Drosophila melanogaster, and mammals. FoxO proteins are homologous to the transcription factor DAuer Formation-16 (DAF-16) in the worm Caenorhabditis elegans. DAF-16 can determine metabolic insulin signaling and lead to lifespan extension (134, 135).

Both epigenetic (7, 136, 137) and post-translation protein modifications regulate the function of FoxO proteins. FoxO proteins are modified by phosphorylation (22, 97, 138-147), acetylation (137, 138, 148), and ubiquitylation (24, 52, 149, 150) (Table 1). Phosphorylation of FoxOs can occur through the serine-threonine kinase protein kinase B (Akt) (54, 75, 151-155) and the serum- and glucocorticoid-inducible protein kinase (SgK) (156). Akt and SgK phosphorylate FoxO proteins at different sites affording various pathways to control FoxO protein activity (157). Kinases such as Akt phosphorylate FoxO proteins to foster binding to 14-3-3 proteins in the cell cytoplasm, prevent nuclear translocation of FoxOs, and then block the transcription of target genes that promote apoptosis (141, 145, 158, 159). Yet, the phosphorylation site of FoxO proteins by specific protein kinases can be important in determining the activity of forkhead transcription factors. For example, mammalian sterile 20-like kinase-1 can phosphorylate FOXO proteins, disrupt the binding to 14-3-3, and allow nuclear translocation of FOXO to promote apoptotic cell death (160).

Similar to post-translational phosphorylation of FoxO proteins, ubiquitylation and acetylation are also vital counterparts to modulate activity of FoxO proteins (35, 161). Ubiquitination and the degradation of FoxO proteins can be fostered through the silent mating type information regulation 2 homolog 1 (S. cerevisiae) (SIRT1) (148, 162, 163). SIRT1 can oversee stem cell development (5, 164) and protect cells through the inhibition of FoxO activity (53, 148, 165-167). FoxOs can bind to the SIRT1 promoter region that contains a cluster of five putative FoxO core binding repeat motifs (5 x insulin receptor substrate (IRS-1)) and a forkhead-like consensus-binding site (FKHD-L) to promote SIRT1 transcription (168). FoxO proteins can then regulate SIRT1 transcription and increase SIRT1 expression (168). SIRT1 promotes FoxO-driven SIRT1 autotranscription through the activation and deacetylation of FoxOs (169). Akt also results in the ubiquitination and degradation of FoxOs through the 26S proteasome of FoxO proteins (170, 171). In relation to acetylation, FoxO proteins are acetylated by histone acetyltransferases that include p300, the CREB-binding protein (CBP), and the CBP-associated factor (172). Once acetylated such as by CBP, FoxO proteins translocate to the cell nucleus but have diminished activity since acetylation of lysine residues on FoxO proteins can limit the ability of FoxO proteins to bind to DNA (173). Acetylation can increase phosphorylation of FoxO proteins mediated through Akt (173). FoxO proteins are deacetylated by histone deacetylases, such as SIRT1 (170, 174-176). Histone deacetylase 2 (HDAC2) also forms a physical complex with FoxO3a to affect FoxO3a-dependent gene transcription and oxidative stress-induced cell death (137).

4. FoxO proteins, Oxidative Stress, Apoptosis, and Autophagy

FoxO proteins play a critical role in cell survival and regeneration (Table 1). FoxO proteins may have different effects upon cell survival during oxidative stress with FOXO1 preventing oxidative stress damage and FOXO3a promoting oxidative cell death in systems that involve the maternal decidua (177). Under some conditions, the activation of FoxO proteins may prevent apoptotic cell injury during oxidative stress such as in chondrocytes (178). The conditional deletion of FoxO1, FoxO3a, and FoxO4 in mouse hematopoietic stem cells may be detrimental and lead to an increase in ROS (179). In addition, FoxO3a may be necessary with other pathways for rejuvenating the function of mesenchymal stem cells (180).

FoxO proteins can lead to the induction of autophagy and increase cell survival (Table 1). In experimental models of full-length mutant Huntingtin (mHtt) transgenic mice, ectopic expression of FoxO1 leads to autophagy and the clearance of toxic mHtt protein in neurons (181). Expression of a constitutively active form of FoxO3 increases human articular chondrocyte cell viability and the expression of autophagy related proteins (178). SIRT1-mediated deacetylation of FoxO1 leads to starvation-induced increases in autophagic flux that maintain left ventricular function during periods of starvation (182). Cardiac expression of constitutively active FoxO3 reverses heart atrophy through the activation of autophagic pathways (183). Yet, FoxO cell protection may not always be directly tied to the induction of autophagy. Up-regulation of FoxO3 and SIRT1 with a reduction in autophagy occurs in human bronchial epithelial cells exposed to cigarette smoke condensates in the presence of the anti-oxidant Amurensis H (Vam3), a dimeric derivative of resveratrol, that can reduce oxidative stress (184).

With apoptotic cell death, FoxO proteins promote membrane PS externalization and DNA degradation to lead to cell injury (185, 186). Endothelial cell dysfunction occurs with a reduction in SIRT1 expression and an increase in FoxO1 expression during exposure to elevated glucose (21). Several studies support a detrimental role for FoxO proteins that can lead to cell injury. For example, inhibition or gene knockdown of FoxO1 or FoxO3a leads to protection against microglial cell injury during oxidative stress (187) and Aβ exposure (185), increased neuronal cell survival through nicotinamide adenine dinucleotide (NAD+) precursors (140), growth factor protection with erythropoietin (EPO) (22, 139, 141, 162) and neurotrophins (188-190), protection against cerebral ischemia (159), and increased cell survival with metabotropic glutamate receptor activation (191). EPO can phosphorylate FoxO proteins, inactivate these proteins (75, 192), and lead to cellular protection in models of experimental DM and oxidative stress (22, 141, 193).

FoxO cell death pathways are dependent, in part, upon Wnt signaling (Table 1). Wnt signaling, including Wnt1 inducible signaling pathway protein 1 (WISP1), also known as CCN4 (194, 195), regulates cell development, vascular growth, immunity, cancer, stem cell proliferation (196-200) and cellular metabolism (25, 195, 198, 200). Wnt signaling may assist in wound healing during DM (200) and can prevent vascular injury during experimental DM (22, 23). Wnt signaling promotes cellular protection against apoptotic cell death through the inactivation of FoxO proteins. Phosphorylation and inhibition of FoxO3a activity by Wnt signaling β-catenin protects hepatocytes from apoptosis (143). Osteoblastic differentiation can be preserved in the presence of oxidative stress through the increased expression of Wnt signaling pathways and the inhibition of FoxO3a (201). Wnt signaling blocks apoptosis through the inhibition of FoxO3a activity to prevent cytochrome c release, Bad phosphorylation, and activation of caspases (186). WISP1 also can increase cell survival by limiting FoxO3a activity, blocking caspase 1 and 3 activation, and fostering SIRT1 nuclear trafficking (145). Growth factors such as EPO use Wnt signaling during experimental DM to block FoxO3a activity and increase endothelial survival (22).

Of note, FoxO regulation of Wnt signaling may be beneficial under other conditions. FoxO proteins can inhibit prostate cell malignant phenotypes by down-regulating Wnt signaling and β-catenin (202). The absence or loss of FoxO activity, such as through microRNA activity, may indicate an increased risk for cancer development (203) and promote tumor growth (204).

5. Pursuing Regenerative Pathways with FoxO for Diabetes Mellitus

In addition to impacting cellular survival and regeneration, FoxOs are important in regulating cellular metabolism and DM (34, 113, 125) (Table 1). Several studies suggest that inactivation of FoxO proteins may foster cytoprotection during DM. Insulin resistance may be resolved with the genetic deletion of hepatic FoxO1 (205). During exposure to high glucose in endothelial cells, FoxO1 is acetylated and leads to premature senescence that can be detrimental to vascular function (21). Activation of FoxO3a occurs during elevated glucose exposure that results in mitochondrial membrane depolarization, cytochrome c release, and caspase activation with subsequent apoptotic cell death (22, 24, 97). In mice, FoxO6 depletion can prevent diet-induced glucose intolerance and insulin resistance through inhibition of hepatic gluconeogenesis and limiting macrophage infiltration in the liver and adipose tissues (206). FoxO proteins can influence pancreatic β-cell survival as well. Thioredoxin-interacting protein (TXNIP) is necessary for β-cell survival. FoxO1 can bind to the TXNIP promoter, decreases TXNIP expression, and prevents TXNIP expression during the exposure to glucose (207). Retinal disease is prevented during therapies tied to FoxO3a inhibition in murine models of DM (208). In studies that examine the loss of SIRT1 with concurrent increased activity of Foxo1 in mice, insulin sensitivity is lost and overproduction of hepatic glucose leads to chronic hyperglycemia and increased ROS production (209). Models of diabetic nephropathy show that transforming growth factor-beta results in the nuclear exclusion of FoxO3a, blocks FoxO3a transcriptional activity, and protects renal mesangial cells from apoptosis (210). Enteric neurons can be protected from hyperglycemia by glial cell line-derived neurotrophic factor that activates Akt and inhibits FoxO3a activation (188). Mice overexpressing Foxo1 in skeletal muscle have impaired glycemic control and impaired skeletal muscle function (211).

Some clinical studies suggest a detrimental effect associated with forkhead transcription factors, body weight, and DM. A single nucleotide polymorphism in the 5′ flanking region of FOXO3a was associated with the greatest body mass index in individuals who were homozygous for the major allele of FOXO3a (212). Additional studies report that haplotype analyses of FOXO1a revealed that carriers of a specific haplotype had higher HbA1c levels with increased mortality risk attributable to death from DM (213). Analyses with FOXO3a haplotypes demonstrate no differences in metabolic profile, fertility or fecundity, but an increased risk of stroke is present for specific Foxo3a haplotypes (213). Agents that control FoxO protein activity during metabolic disturbances may offer treatment for patients with DM. One potential agent to consider for the maintenance of cellular metabolism in DM is nicotinamide (52, 214-223). Nicotinamide can improve metabolic control in combination therapy (224), can be protective for pancreatic β-cell function (225), and reverses disease during diabetic peripheral neuropathy in animal models (226). Oral nicotinamide administration can protect β-cell function and prevent clinical disease in islet-cell antibody-positive first-degree relatives of Type 1 DM (225). Nicotinamide may be protective through the post-translational modification of FoxO3a. Nicotinamide can inhibit FoxO3a activity and preserve the integrity of the FoxO3a protein to block FoxO3a proteolysis that can yield pro-apoptotic amino-terminal fragments (140). It is important to discuss that prolonged exposure to nicotinamide in some studies has been reported to impair β-cell function and reduce cell growth (227, 228). As a result, studies have shown a benefit with reduction in nicotinamide activity through nicotinamidases that degrade nicotinamide and lead to increased lifespan (215, 229-231).

However, in some sub-populations with specific haplotypes for FoxO1 and FoxO3, increased metabolic risk and disease from FoxO proteins in cardiovascular disorders may not occur (232). In fact, FoxO proteins may exert a beneficial effect during metabolic disorders under some conditions. FoxO proteins are homologous to DAF-16 that can regulate cellular metabolism and lifespan (149, 230, 233). FoxO proteins can stimulate the insulin-like growth factor binding protein-1 (IGFBP1) promoter by binding to a conserved insulin response sequence (234). Insulin and insulin-like growth factor-1 (IGF-1) can control and suppress this activity through activation of Akt (234, 235). Interferon-gamma driven expression of tryptophan catabolism by cytotoxic T lymphocyte antigen 4 can activate Foxo3a to protect dendritic cells from ROS generation in non-obese diabetic mice (236). In caloric restricted mice that have decreased energy reserves, mRNA expression in rat skeletal muscles is increased for specific Foxo proteins, suggesting that Foxo may have a protective function during metabolic disorders (237). In addition, FoxO1 expression leads to increased insulin signaling to regulate cellular metabolism in Drosophila and mammalian cells (238).

6. Future Considerations

DM affects multiple systems throughout the body leading to significant disability as well as death. New development of strategies targeting FoxO proteins may offer the opportunity to effectively provide clinical treatments for disorders of cellular metabolism and the clinical complications of DM. However, a number of hurdles must be overcome in consideration of the clinical utility of FoxO proteins. Further understanding of the pathways that determine injury in DM and the biological role of FoxO proteins are necessary to move forward. For example, how can cell death pathways of autophagy and apoptosis determine tissue injury during FoxO activation? Oxidative stress is a significant mediator of cell injury during DM. Under some conditions, autophagy can improve insulin sensitivity and regulate glucose homeostasis. FoxO proteins can lead to the induction of autophagy and enhance cell survival such that FoxO proteins reduce oxidative stress and assist with metabolic homeostasis. Yet, increased autophagy during DM may lead to the loss of tissue, may injure endothelial progenitor cells, and promote oxidative stress. Activation of FoxO proteins may prevent apoptotic cell injury during oxidative stress in some cell types. In other scenarios, it is the inhibition of FoxO protein activity that leads to cytoprotection that may require the activation of Wnt signaling pathways as well as SIRT1. Control of FoxO activity may be dependent upon a number of factors including the phosphorylation site of FoxO proteins by specific protein kinases that can determine whether FoxO proteins foster cell survival or conversely lead to cell death.

Furthermore, what are the factors that influence the ability of FoxO proteins to affect cellular metabolism? A number of studies suggest that inactivation of FoxO proteins may foster cytoprotection during DM, block insulin resistance, assist with pancreatic β-cell survival, and prevent immune cell tissue infiltration. Some clinical studies support a role to limit FoxO activity and decrease the risk of mortality from DM. In contrast, other clinical work indicates that there is little or no association between increased metabolic risk and FoxO proteins. Other experimental studies suggest that increased FoxO protein expression is beneficial for insulin signaling and maintaining energy reserves. Epigenetic as well as post-translational modification of FoxO proteins during impairments in cellular metabolism may play an essential role in impacting the variable clinical outcome modulated by FoxO proteins. Only through the initiation and progression of future investigations can these questions be addressed to both safely and effectively develop FoxO signal transduction pathways into viable clinical treatments for metabolic disorders and DM.

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

Conflict of Interest: The authors declare no conflicts of interest.

References

  • 1.Esser N, Paquot N, Scheen AJ. Anti-inflammatory agents to treat or prevent type 2 diabetes, metabolic syndrome and cardiovascular disease. Expert opinion on investigational drugs. 2015 Mar;24(3):283–307. doi: 10.1517/13543784.2015.974804. [DOI] [PubMed] [Google Scholar]
  • 2.Maiese K. New Insights for Oxidative Stress and Diabetes Mellitus. Oxid Med Cell Longev. 2015;2015(Article ID 875961) doi: 10.1155/2015/875961. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Alexandru N, Popov D, Georgescu A. Platelet dysfunction in vascular pathologies and how can it be treated. Thromb Res. 2012 Feb;129(2):116–26. doi: 10.1016/j.thromres.2011.09.026. [DOI] [PubMed] [Google Scholar]
  • 4.Jiang T, Yu JT, Zhu XC, Wang HF, Tan MS, Cao L, et al. Acute metformin preconditioning confers neuroprotection against focal cerebral ischaemia by pre-activation of AMPK-dependent autophagy. Br J Pharmacol. 2014 Jul;171(13):3146–57. doi: 10.1111/bph.12655. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Maiese K. SIRT1 and stem cells: In the forefront with cardiovascular disease, neurodegeneration and cancer. World J Stem Cells. 2015 Mar 26;7(2):235–42. doi: 10.4252/wjsc.v7.i2.235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Maiese K, Chong ZZ, Hou J, Shang YC. Erythropoietin and oxidative stress. Curr Neurovasc Res. 2008 May;5(2):125–42. doi: 10.2174/156720208784310231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Xiao FH, He YH, Li QG, Wu H, Luo LH, Kong QP. A genome-wide scan reveals important roles of DNA methylation in human longevity by regulating age-related disease genes. PLoS One. 2015;10(3):e0120388. doi: 10.1371/journal.pone.0120388. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Xu YJ, Tappia PS, Neki NS, Dhalla NS. Prevention of diabetes-induced cardiovascular complications upon treatment with antioxidants. Heart failure reviews. 2014 Jan;19(1):113–21. doi: 10.1007/s10741-013-9379-6. [DOI] [PubMed] [Google Scholar]
  • 9.Busch S, Kannt A, Kolibabka M, Schlotterer A, Wang Q, Lin J, et al. Systemic treatment with erythropoietin protects the neurovascular unit in a rat model of retinal neurodegeneration. PLoS One. 2014;9(7):e102013. doi: 10.1371/journal.pone.0102013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Fu D, Wu M, Zhang J, Du M, Yang S, Hammad SM, et al. Mechanisms of modified LDL-induced pericyte loss and retinal injury in diabetic retinopathy. Diabetologia. 2012 Nov;55(11):3128–40. doi: 10.1007/s00125-012-2692-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Lee K, Hu Y, Ding L, Chen Y, Takahashi Y, Mott R, et al. Therapeutic potential of a monoclonal antibody blocking the Wnt pathway in diabetic retinopathy. Diabetes. 2012 Nov;61(11):2948–57. doi: 10.2337/db11-0300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Maiese K. Novel applications of trophic factors, Wnt and WISP for neuronal repair and regeneration in metabolic disease. Neural regeneration research. 2015;10(4):518–28. doi: 10.4103/1673-5374.155427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Gomes MB, Negrato CA. Alpha-lipoic acid as a pleiotropic compound with potential therapeutic use in diabetes and other chronic diseases. Diabetology & metabolic syndrome. 2014;6(1):80. doi: 10.1186/1758-5996-6-80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Gomez-Brouchet A, Blaes N, Mouledous L, Fourcade O, Tack I, Frances B, et al. Beneficial effects of levobupivacaine regional anaesthesia on postoperative opioid induced hyperalgesia in diabetic mice. Journal of translational medicine. 2015;13(1):208. doi: 10.1186/s12967-015-0575-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Du LL, Chai DM, Zhao LN, Li XH, Zhang FC, Zhang HB, et al. AMPK Activation Ameliorates Alzheimer's Disease-Like Pathology and Spatial Memory Impairment in a Streptozotocin-Induced Alzheimer's Disease Model in Rats. J Alzheimers Dis. 2014 Aug 11; doi: 10.3233/JAD-140564. [DOI] [PubMed] [Google Scholar]
  • 16.Kapogiannis D, Boxer A, Schwartz JB, Abner EL, Biragyn A, Masharani U, et al. Dysfunctionally phosphorylated type 1 insulin receptor substrate in neural-derived blood exosomes of preclinical Alzheimer's disease. FASEB J. 2014 Oct 23; doi: 10.1096/fj.14-262048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Maiese K, Chong ZZ, Shang YC. Raves and risks for erythropoietin. Cytokine Growth Factor Rev. 2008 Apr;19(2):145–55. doi: 10.1016/j.cytogfr.2008.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.White MF. IRS2 integrates insulin/IGF1 signalling with metabolism, neurodegeneration and longevity. Diabetes Obes Metab. 2014 Sep;16(Suppl 1):4–15. doi: 10.1111/dom.12347. [DOI] [PubMed] [Google Scholar]
  • 19.Aksu I, Ates M, Baykara B, Kiray M, Sisman AR, Buyuk E, et al. Anxiety correlates to decreased blood and prefrontal cortex IGF-1 levels in streptozotocin induced diabetes. Neurosci Lett. 2012 Dec 7;531(2):176–81. doi: 10.1016/j.neulet.2012.10.045. [DOI] [PubMed] [Google Scholar]
  • 20.Reagan LP. Diabetes as a chronic metabolic stressor: causes, consequences and clinical complications. Exp Neurol. 2012 Jan;233(1):68–78. doi: 10.1016/j.expneurol.2011.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Arunachalam G, Samuel SM, Marei I, Ding H, Triggle CR. Metformin modulates hyperglycaemia-induced endothelial senescence and apoptosis through SIRT1. Br J Pharmacol. 2014 Jan;171(2):523–35. doi: 10.1111/bph.12496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Chong ZZ, Hou J, Shang YC, Wang S, Maiese K. EPO Relies upon Novel Signaling of Wnt1 that Requires Akt1, FoxO3a, GSK-3beta, and beta-Catenin to Foster Vascular Integrity During Experimental Diabetes. Curr Neurovasc Res. 2011 May 1;8(2):103–20. doi: 10.2174/156720211795495402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Chong ZZ, Shang YC, Maiese K. Vascular injury during elevated glucose can be mitigated by erythropoietin and Wnt signaling. Curr Neurovasc Res. 2007 Aug;4(3):194–204. doi: 10.2174/156720207781387150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Hou J, Chong ZZ, Shang YC, Maiese K. FoxO3a governs early and late apoptotic endothelial programs during elevated glucose through mitochondrial and caspase signaling. Mol Cell Endocrinol. 2010 Mar 4;321(2):194–206. doi: 10.1016/j.mce.2010.02.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Liu Q, Li J, Cheng R, Chen Y, Lee K, Hu Y, et al. Nitrosative stress plays an important role in wnt pathway activation in diabetic retinopathy. Antioxid Redox Signal. 2013 Apr 1;18(10):1141–53. doi: 10.1089/ars.2012.4583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Schaffer SW, Jong CJ, Mozaffari M. Role of oxidative stress in diabetes-mediated vascular dysfunction: unifying hypothesis of diabetes revisited. Vascul Pharmacol. 2012 Nov-Dec;57(5-6):139–49. doi: 10.1016/j.vph.2012.03.005. [DOI] [PubMed] [Google Scholar]
  • 27.Wang L, Di L, Noguchi CT. Erythropoietin, a novel versatile player regulating energy metabolism beyond the erythroid system. Int J Biol Sci. 2014;10(8):921–39. doi: 10.7150/ijbs.9518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Zhang Y, Wang L, Dey S, Alnaeeli M, Suresh S, Rogers H, et al. Erythropoietin action in stress response, tissue maintenance and metabolism. International journal of molecular sciences. 2014;15(6):10296–333. doi: 10.3390/ijms150610296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Aragno M, Mastrocola R, Ghe C, Arnoletti E, Bassino E, Alloatti G, et al. Obestatin induced recovery of myocardial dysfunction in type 1 diabetic rats: underlying mechanisms. Cardiovasc Diabetol. 2012;11:129. doi: 10.1186/1475-2840-11-129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Chong ZZ, Maiese K. Mammalian Target of Rapamycin Signaling in Diabetic Cardiovascular Disease. Cardiovasc Diabetol. 2012 Apr 30;11(1):45. doi: 10.1186/1475-2840-11-45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Das A, Durrant D, Koka S, Salloum FN, Xi L, Kukreja RC. Mammalian target of rapamycin (mTOR) inhibition with rapamycin improves cardiac function in type 2 diabetic mice: potential role of attenuated oxidative stress and altered contractile protein expression. J Biol Chem. 2014 Feb 14;289(7):4145–60. doi: 10.1074/jbc.M113.521062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.He C, Zhu H, Li H, Zou MH, Xie Z. Dissociation of Bcl-2-Beclin1 complex by activated AMPK enhances cardiac autophagy and protects against cardiomyocyte apoptosis in diabetes. Diabetes. 2013 Apr;62(4):1270–81. doi: 10.2337/db12-0533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Ling S, Birnbaum Y, Nanhwan MK, Thomas B, Bajaj M, Li Y, et al. Dickkopf-1 (DKK1) phosphatase and tensin homolog on chromosome 10 (PTEN) crosstalk via microRNA interference in the diabetic heart. Basic Res Cardiol. 2013;108(3):352. doi: 10.1007/s00395-013-0352-2. [DOI] [PubMed] [Google Scholar]
  • 34.Maiese K, Chong ZZ, Shang YC, Hou J. FoxO proteins: cunning concepts and considerations for the cardiovascular system. Clin Sci (Lond) 2009 Feb;116(3):191–203. doi: 10.1042/CS20080113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Maiese K, Hou J, Chong ZZ, Shang YC. A fork in the path: Developing therapeutic inroads with FoxO proteins. Oxid Med Cell Longev. 2009 Jul;2(3):119–29. doi: 10.4161/oxim.2.3.8916. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Portbury AL, Ronnebaum SM, Zungu M, Patterson C, Willis MS. Back to your heart: ubiquitin proteasome system-regulated signal transduction. J Mol Cell Cardiol. 2012 Mar;52(3):526–37. doi: 10.1016/j.yjmcc.2011.10.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Zhang C, Zhang L, Chen S, Feng B, Lu X, Bai Y, et al. The prevention of diabetic cardiomyopathy by non-mitogenic acidic fibroblast growth factor is probably mediated by the suppression of oxidative stress and damage. PLoS One. 2013;8(12):e82287. doi: 10.1371/journal.pone.0082287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Hu P, Lai D, Lu P, Gao J, He H. ERK and Akt signaling pathways are involved in advanced glycation end product-induced autophagy in rat vascular smooth muscle cells. Int J Mol Med. 2012 Apr;29(4):613–8. doi: 10.3892/ijmm.2012.891. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Maiese K. mTOR: Driving apoptosis and autophagy for neurocardiac complications of diabetes mellitus. World J Diabetes. 2015 Mar 15;6(2):217–24. doi: 10.4239/wjd.v6.i2.217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Wang F, Ma X, Zhou M, Pan X, Ni J, Gao M, et al. Serum pigment epithelium-derived factor levels are independently correlated with the presence of coronary artery disease. Cardiovasc Diabetol. 2013;12:56. doi: 10.1186/1475-2840-12-56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Razmara M, Hjemdahl P, Ostenson CG, Li N. Platelet hyperprocoagulant activity in Type 2 diabetes mellitus: attenuation by glycoprotein IIb/IIIa inhibition. J Thromb Haemost. 2008 Dec;6(12):2186–92. doi: 10.1111/j.1538-7836.2008.03185.x. [DOI] [PubMed] [Google Scholar]
  • 42.Albiero M, Poncina N, Tjwa M, Ciciliot S, Menegazzo L, Ceolotto G, et al. Diabetes causes bone marrow autonomic neuropathy and impairs stem cell mobilization via dysregulated p66Shc and Sirt1. Diabetes. 2014 Apr;63(4):1353–65. doi: 10.2337/db13-0894. [DOI] [PubMed] [Google Scholar]
  • 43.Balestrieri ML, Servillo L, Esposito A, D'Onofrio N, Giovane A, Casale R, et al. Poor glycaemic control in type 2 diabetes patients reduces endothelial progenitor cell number by influencing SIRT1 signalling via platelet-activating factor receptor activation. Diabetologia. 2013 Jan;56(1):162–72. doi: 10.1007/s00125-012-2749-0. [DOI] [PubMed] [Google Scholar]
  • 44.Barthelmes D, Zhu L, Shen W, Gillies MC, Irhimeh MR. Differential gene expression in Lin-/VEGF-R2+ bone marrow-derived endothelial progenitor cells isolated from diabetic mice. Cardiovasc Diabetol. 2014;13(1):42. doi: 10.1186/1475-2840-13-42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Chong ZZ, Maiese K. The Src homology 2 domain tyrosine phosphatases SHP-1 and SHP-2: diversified control of cell growth, inflammation, and injury. Histol Histopathol. 2007 Nov;22(11):1251–67. doi: 10.14670/hh-22.1251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Kim KA, Shin YJ, Akram M, Kim ES, Choi KW, Suh H, et al. High glucose condition induces autophagy in endothelial progenitor cells contributing to angiogenic impairment. Biol Pharm Bull. 2014;37(7):1248–52. doi: 10.1248/bpb.b14-00172. [DOI] [PubMed] [Google Scholar]
  • 47.Weinberg E, Maymon T, Weinreb M. AGEs induce caspase-mediated apoptosis of rat BMSCs via TNFalpha production and oxidative stress. Journal of molecular endocrinology. 2014 Feb;52(1):67–76. doi: 10.1530/JME-13-0229. [DOI] [PubMed] [Google Scholar]
  • 48.Chen JX, Tuo Q, Liao DF, Zeng H. Inhibition of Protein Tyrosine Phosphatase Improves Angiogenesis via Enhancing Ang-1/Tie-2 Signaling in Diabetes. Exp Diabetes Res. 2012;2012:836759. doi: 10.1155/2012/836759. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.da Rosa LC, Chiuso-Minicucci F, Zorzella-Pezavento SF, Franca TG, Ishikawa LL, Colavite PM, et al. Bacille Calmette-Guerin/DNAhsp65 prime-boost is protective against diabetes in non-obese diabetic mice but not in the streptozotocin model of type 1 diabetes. Clin Exp Immunol. 2013 Sep;173(3):430–7. doi: 10.1111/cei.12140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Hamed S, Bennett CL, Demiot C, Ullmann Y, Teot L, Desmouliere A. Erythropoietin, a novel repurposed drug: an innovative treatment for wound healing in patients with diabetes mellitus. Wound repair and regeneration : official publication of the Wound Healing Society [and] the European Tissue Repair Society. 2014 Jan-Feb;22(1):23–33. doi: 10.1111/wrr.12135. [DOI] [PubMed] [Google Scholar]
  • 51.Jia G, Aroor AR, Martinez-Lemus LA, Sowers JR. Invited Review: Over-nutrition, mTOR Signaling and Cardiovascular Diseases. Am J Physiol Regul Integr Comp Physiol. 2014 Sep 24; doi: 10.1152/ajpregu.00262.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Maiese K, Chong ZZ, Hou J, Shang YC. The vitamin nicotinamide: translating nutrition into clinical care. Molecules. 2009;14(9):3446–85. doi: 10.3390/molecules14093446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.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]
  • 54.Puthanveetil P, Wan A, Rodrigues B. FoxO1 is crucial for sustaining cardiomyocyte metabolism and cell survival. Cardiovasc Res. 2013 Mar 1;97(3):393–403. doi: 10.1093/cvr/cvs426. [DOI] [PubMed] [Google Scholar]
  • 55.Zhao Y, Scott NA, Fynch S, Elkerbout L, Wong WW, Mason KD, et al. Autoreactive T cells induce necrosis and not BCL-2-regulated or death receptor-mediated apoptosis or RIPK3-dependent necroptosis of transplanted islets in a mouse model of type 1 diabetes. Diabetologia. 2014 Oct 10; doi: 10.1007/s00125-014-3407-5. [DOI] [PubMed] [Google Scholar]
  • 56.Hao J, Li F, Liu W, Liu Q, Liu S, Li H, et al. Phosphorylation of PRAS40-Thr246 Involves in Renal Lipid Accumulation of Diabetes. J Cell Physiol. 2013 Dec 18; doi: 10.1002/jcp.24533. [DOI] [PubMed] [Google Scholar]
  • 57.Hao J, Zhu L, Li F, Liu Q, Zhao X, Liu S, et al. Phospho-mTOR: a novel target in regulation of renal lipid metabolism abnormality of diabetes. Exp Cell Res. 2013 Aug 15;319(14):2296–306. doi: 10.1016/j.yexcr.2013.06.013. [DOI] [PubMed] [Google Scholar]
  • 58.Nakazawa J, Isshiki K, Sugimoto T, Araki S, Kume S, Yokomaku Y, et al. Renoprotective effects of asialoerythropoietin in diabetic mice against ischaemia-reperfusion-induced acute kidney injury. Nephrology (Carlton) 2010 Feb;15(1):93–101. doi: 10.1111/j.1440-1797.2009.01170.x. [DOI] [PubMed] [Google Scholar]
  • 59.Pandya KG, Budhram R, Clark GJ, Lau-Cam CA. Taurine can enhance the protective actions of metformin against diabetes-induced alterations adversely affecting renal function. Adv Exp Med Biol. 2015;803:227–50. doi: 10.1007/978-3-319-15126-7_20. [DOI] [PubMed] [Google Scholar]
  • 60.Perez-Gallardo RV, Noriega-Cisneros R, Esquivel-Gutierrez E, Calderon-Cortes E, Cortes-Rojo C, Manzo-Avalos S, et al. Effects of diabetes on oxidative and nitrosative stress in kidney mitochondria from aged rats. Journal of bioenergetics and biomembranes. 2014 Nov 26; doi: 10.1007/s10863-014-9594-4. [DOI] [PubMed] [Google Scholar]
  • 61.Caron AZ, He X, Mottawea W, Seifert EL, Jardine K, Dewar-Darch D, et al. The SIRT1 deacetylase protects mice against the symptoms of metabolic syndrome. FASEB J. 2014 Mar;28(3):1306–16. doi: 10.1096/fj.13-243568. [DOI] [PubMed] [Google Scholar]
  • 62.Castano D, Larequi E, Belza I, Astudillo AM, Martinez-Anso E, Balsinde J, et al. Cardiotrophin-1 eliminates hepatic steatosis in obese mice by mechanisms involving AMPK activation. Journal of hepatology. 2014 May;60(5):1017–25. doi: 10.1016/j.jhep.2013.12.012. [DOI] [PubMed] [Google Scholar]
  • 63.Gong H, Pang J, Han Y, Dai Y, Dai D, Cai J, et al. Age-dependent tissue expression patterns of Sirt1 in senescence-accelerated mice. Molecular medicine reports. 2014 Oct 15;10(6):3296–302. doi: 10.3892/mmr.2014.2648. [DOI] [PubMed] [Google Scholar]
  • 64.Lee JH, Lee JH, Jin M, Han SD, Chon GR, Kim IH, et al. Diet control to achieve euglycemia induces significant loss of heart and liver weight via increased autophagy compared with ad libitum diet in diabetic rats. Exp Mol Med. 2014;46:e111. doi: 10.1038/emm.2014.52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Maiese K, Chong ZZ, Shang YC, Wang S. Novel directions for diabetes mellitus drug discovery. Expert opinion on drug discovery. 2013 Jan;8(1):35–48. doi: 10.1517/17460441.2013.736485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Malla R, Wang Y, Chan WK, Tiwari AK, Faridi JS. Genetic ablation of PRAS40 improves glucose homeostasis via linking the AKT and mTOR pathways. Biochem Pharmacol. 2015 Apr 27; doi: 10.1016/j.bcp.2015.04.016. [DOI] [PubMed] [Google Scholar]
  • 67.Deblon N, Bourgoin L, Veyrat-Durebex C, Peyrou M, Vinciguerra M, Caillon A, et al. Chronic mTOR inhibition by rapamycin induces muscle insulin resistance despite weight loss in rats. Br J Pharmacol. 2012 Apr;165(7):2325–40. doi: 10.1111/j.1476-5381.2011.01716.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Gao J, Li J, An Y, Liu X, Qian Q, Wu Y, et al. Increasing effect of Tangzhiqing formula on IRS-1-dependent PI3K/AKT signaling in muscle. BMC complementary and alternative medicine. 2014;14:198. doi: 10.1186/1472-6882-14-198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Liu Y, Palanivel R, Rai E, Park M, Gabor TV, Scheid MP, et al. Adiponectin stimulates autophagy and reduces oxidative stress to enhance insulin sensitivity during high fat diet feeding in mice. Diabetes. 2014 Jul 28;64(1):36–48. doi: 10.2337/db14-0267. [DOI] [PubMed] [Google Scholar]
  • 70.Zhang T, Fang M, Fu ZM, Du HC, Yuan H, Xia GY, et al. Expression of PI3-K, PKB and GSK-3 beta in the skeletal muscle tissue of gestational diabetes mellitus. Asian Pacific journal of tropical medicine. 2014 Apr;7(4):309–12. doi: 10.1016/S1995-7645(14)60045-6. [DOI] [PubMed] [Google Scholar]
  • 71.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]
  • 72.World Health Organization. Global status report on noncommunicable diseases 2010. Geneva: 2011. [Google Scholar]
  • 73.Centers for Disease Control and Prevention. National Diabetes Statistics Report: Estimates of Diabetes and Its Burden in the United States. US Department of Health and Human Services; Atlanta, GA: 2014. [Google Scholar]
  • 74.Harris MI, Eastman RC. Early detection of undiagnosed diabetes mellitus: a US perspective. Diabetes Metab Res Rev. 2000 Jul-Aug;16(4):230–6. doi: 10.1002/1520-7560(2000)9999:9999<::aid-dmrr122>3.0.co;2-w. [DOI] [PubMed] [Google Scholar]
  • 75.Maiese K, Chong ZZ, Shang YC. Mechanistic insights into diabetes mellitus and oxidative stress. Curr Med Chem. 2007;14(16):1729–38. doi: 10.2174/092986707781058968. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Centers for Medicare and Medicaid Services. National Health Expenditure Projections 2012-2022. 2013 www.cms.gov.
  • 77.Bagul PK, Banerjee SK. Insulin resistance, oxidative stress and cardiovascular complications: role of sirtuins. Curr Pharm Des. 2013;19(32):5663–77. doi: 10.2174/13816128113199990372. [DOI] [PubMed] [Google Scholar]
  • 78.Kashihara N, Haruna Y, Kondeti VK, Kanwar YS. Oxidative stress in diabetic nephropathy. Curr Med Chem. 2010;17(34):4256–69. doi: 10.2174/092986710793348581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Maiese K, Chong ZZ, Hou J, Shang YC. Oxidative stress: Biomarkers and novel therapeutic pathways. Exp Gerontol. 2010 Mar;45(3):217–34. doi: 10.1016/j.exger.2010.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Maiese K, Chong ZZ, Shang YC, Hou J. Novel Avenues of Drug Discovery and Biomarkers for Diabetes Mellitus. Journal of clinical pharmacology. 2011 Mar 10;51(2):128–52. doi: 10.1177/0091270010362904. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Haldar SR, Chakrabarty A, Chowdhury S, Haldar A, Sengupta S, Bhattacharyya M. Oxidative Stress-Related Genes in Type 2 Diabetes: Association Analysis and Their Clinical Impact. Biochemical genetics. 2015 May 20; doi: 10.1007/s10528-015-9675-z. [DOI] [PubMed] [Google Scholar]
  • 82.Jiang YL, Ning Y, Ma XL, Liu YY, Wang Y, Zhang Z, et al. Alteration of the proteome profile of the pancreas in diabetic rats induced by streptozotocin. Int J Mol Med. 2011 Aug;28(2):153–60. doi: 10.3892/ijmm.2011.696. [DOI] [PubMed] [Google Scholar]
  • 83.Maiese K, Shang YC, Chong ZZ, Hou J. Diabetes mellitus: channeling care through cellular discovery. Curr Neurovasc Res. 2010 Feb 1;7(1):59–64. doi: 10.2174/156720210790820217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Yang H, Jin X, Kei Lam CW, Yan SK. Oxidative stress and diabetes mellitus. Clin Chem Lab Med. 2011 Nov;49(11):1773–82. doi: 10.1515/CCLM.2011.250. [DOI] [PubMed] [Google Scholar]
  • 85.Kumar D, Rizvi SI. Black tea extract improves anti-oxidant profile in experimental diabetic rats. Archives of physiology and biochemistry. 2015 Jun 29;:1–7. doi: 10.3109/13813455.2015.1043308. [DOI] [PubMed] [Google Scholar]
  • 86.Mao XY, Cao DF, Li X, Yin JY, Wang ZB, Zhang Y, et al. Huperzine A ameliorates cognitive deficits in streptozotocin-induced diabetic rats. International journal of molecular sciences. 2014;15(5):7667–83. doi: 10.3390/ijms15057667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Harish G, Mahadevan A, Pruthi N, Sreenivasamurthy SK, Puttamallesh VN, Keshava Prasad TS, et al. Characterization of traumatic brain injury in human brains reveals distinct cellular and molecular changes in contusion and pericontusion. J Neurochem. 2015 Feb 25; doi: 10.1111/jnc.13082. [DOI] [PubMed] [Google Scholar]
  • 88.Kwon SH, Hong SI, Ma SX, Lee SY, Jang CG. 3′,4′,7-trihydroxyflavone prevents apoptotic cell death in neuronal cells from hydrogen peroxide-induced oxidative stress. Food Chem Toxicol. 2015 Feb 23; doi: 10.1016/j.fct.2015.02.014. [DOI] [PubMed] [Google Scholar]
  • 89.Stefano GB, Snyder C, Kream RM. Mitochondria, Chloroplasts in Animal and Plant Cells: Significance of Conformational Matching. Med Sci Monit. 2015;21:2073–8. doi: 10.12659/MSM.894758. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Wei L, Ding L, Mo MS, Lei M, Zhang L, Chen K, et al. Wnt3a protects SH-SY5Y cells against 6-hydroxydopamine toxicity by restoration of mitochondria function. Translational neurodegeneration. 2015;4:11. doi: 10.1186/s40035-015-0033-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Zhou Q, Liu C, Liu W, Zhang H, Zhang R, Liu J, et al. Rotenone induction of hydrogen peroxide inhibits mTOR-mediated S6K1 and 4E-BP1/eIF4E pathways, leading to neuronal apoptosis. Toxicol Sci. 2015 Jan;143(1):81–96. doi: 10.1093/toxsci/kfu211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Lee Y, Hong Y, Lee SR, Chang KT. Autophagy contributes to retardation of cardiac growth in diabetic rats. Lab Anim Res. 2012 Jun;28(2):99–107. doi: 10.5625/lar.2012.28.2.99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Paiva MA, Rutter-Locher Z, Goncalves LM, Providencia LA, Davidson SM, Yellon DM, et al. Enhancing AMPK activation during ischemia protects the diabetic heart against reperfusion injury. Am J Physiol Heart Circ Physiol. 2011 Jun;300(6):H2123–34. doi: 10.1152/ajpheart.00707.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Kui L, Weiwei Z, Ling L, Daikun H, Guoming Z, Linuo Z, et al. Ghrelin inhibits apoptosis induced by high glucose and sodium palmitate in adult rat cardiomyocytes through the PI3K-Akt signaling pathway. Regul Pept. 2009 Jun 5;155(1-3):62–9. doi: 10.1016/j.regpep.2009.03.003. [DOI] [PubMed] [Google Scholar]
  • 95.Mikhed Y, Daiber A, Steven S. Mitochondrial Oxidative Stress, Mitochondrial DNA Damage and Their Role in Age-Related Vascular Dysfunction. International journal of molecular sciences. 2015;16(7):15918–53. doi: 10.3390/ijms160715918. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Gao W, Ferguson G, Connell P, Walshe T, O'Brien C, Redmond EM, et al. Glucose attenuates hypoxia-induced changes in endothelial cell growth by inhibiting HIF-1alpha expression. Diabetes & vascular disease research : official journal of the International Society of Diabetes and Vascular Disease. 2014 May 22;11(4):270–80. doi: 10.1177/1479164114533356. [DOI] [PubMed] [Google Scholar]
  • 97.Hou J, Chong ZZ, Shang YC, Maiese K. Early apoptotic vascular signaling is determined by Sirt1 through nuclear shuttling, forkhead trafficking, bad, and mitochondrial caspase activation. Curr Neurovasc Res. 2010 May;7(2):95–112. doi: 10.2174/156720210791184899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Orimo M, Minamino T, Miyauchi H, Tateno K, Okada S, Moriya J, et al. Protective role of SIRT1 in diabetic vascular dysfunction. Arterioscler Thromb Vasc Biol. 2009 Jun;29(6):889–94. doi: 10.1161/ATVBAHA.109.185694. [DOI] [PubMed] [Google Scholar]
  • 99.Guan FY, Gu J, Li W, Zhang M, Ji Y, Li J, et al. Compound K protects pancreatic islet cells against apoptosis through inhibition of the AMPK/JNK pathway in type 2 diabetic mice and in MIN6 beta-cells. Life Sci. 2014 Jun 27;107(1-2):42–9. doi: 10.1016/j.lfs.2014.04.034. [DOI] [PubMed] [Google Scholar]
  • 100.Liu Z, Stanojevic V, Brindamour LJ, Habener JF. GLP1-derived nonapeptide GLP1(28-36)amide protects pancreatic beta-cells from glucolipotoxicity. J Endocrinol. 2012 May;213(2):143–54. doi: 10.1530/JOE-11-0328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Zhou J, Wu J, Zheng F, Jin M, Li H. Glucagon-like peptide-1 analog-mediated protection against cholesterol-induced apoptosis via mammalian target of rapamycin activation in pancreatic betaTC-6 cells -1mTORbetaTC-6. Journal of diabetes. 2015 Mar;7(2):231–9. doi: 10.1111/1753-0407.12177. [DOI] [PubMed] [Google Scholar]
  • 102.Kurban S, Mehmetoglu I, Yerlikaya HF, Gonen S, Erdem S. Effect of chronic regular exercise on serum ischemia-modified albumin levels and oxidative stress in type 2 diabetes mellitus. Endocr Res. 2011;36(3):116–23. doi: 10.3109/07435800.2011.566236. [DOI] [PubMed] [Google Scholar]
  • 103.Fong Y, Lin YC, Wu CY, Wang HM, Lin LL, Chou HL, et al. The antiproliferative and apoptotic effects of sirtinol, a sirtuin inhibitor on human lung cancer cells by modulating Akt/beta-catenin-Foxo3a axis. ScientificWorldJournal. 2014;2014:937051. doi: 10.1155/2014/937051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Guo X, Chen Y, Liu Q, Wu J, Wang L, Tang X, et al. Ac-cel, a novel antioxidant, protects against hydrogen peroxide-induced injury in PC12 cells via attenuation of mitochondrial dysfunction. J Mol Neurosci. 2013 Jul;50(3):453–61. doi: 10.1007/s12031-013-9955-1. [DOI] [PubMed] [Google Scholar]
  • 105.Maiese K, Chong ZZ, Shang YC, Wang S. Targeting disease through novel pathways of apoptosis and autophagy. Expert opinion on therapeutic targets. 2012 Dec;16(12):1203–14. doi: 10.1517/14728222.2012.719499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Schutters K, Reutelingsperger C. Phosphatidylserine targeting for diagnosis and treatment of human diseases. Apoptosis. 2010 Sep;15(9):1072–82. doi: 10.1007/s10495-010-0503-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Wei L, Sun C, Lei M, Li G, Yi L, Luo F, et al. Activation of Wnt/beta-catenin Pathway by Exogenous Wnt1 Protects SH-SY5Y Cells Against 6-Hydroxydopamine Toxicity. J Mol Neurosci. 2013 Jan;49(1):105–15. doi: 10.1007/s12031-012-9900-8. [DOI] [PubMed] [Google Scholar]
  • 108.Chong ZZ, Li F, Maiese K. Oxidative stress in the brain: Novel cellular targets that govern survival during neurodegenerative disease. Prog Neurobiol. 2005 Feb;75(3):207–46. doi: 10.1016/j.pneurobio.2005.02.004. [DOI] [PubMed] [Google Scholar]
  • 109.Chong ZZ, Li F, Maiese K. Attempted Cell Cycle Induction in Post-Mitotic Neurons Occurs in Early and Late Apoptotic Programs Through Rb, E2F1, and Caspase 3. Curr Neurovasc Res. 2006 Feb;3(1):25–39. doi: 10.2174/156720206775541741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Favaloro B, Allocati N, Graziano V, Di Ilio C, De Laurenzi V. Role of Apoptosis in disease. Aging (Albany NY) 2012 May;4(5):330–49. doi: 10.18632/aging.100459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Folch J, Junyent F, Verdaguer E, Auladell C, Pizarro JG, Beas-Zarate C, et al. Role of cell cycle re-entry in neurons: a common apoptotic mechanism of neuronal cell death. Neurotox Res. 2012 Oct;22(3):195–207. doi: 10.1007/s12640-011-9277-4. [DOI] [PubMed] [Google Scholar]
  • 112.Nguyen AQ, Cherry BH, Scott GF, Ryou MG, Mallet RT. Erythropoietin: Powerful protection of ischemic and post-ischemic brain. Exp Biol Med (Maywood) 2014 Mar 4; doi: 10.1177/1535370214523703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Shao S, Yang Y, Yuan G, Zhang M, Yu X. Signaling molecules involved in lipid-induced pancreatic Beta-cell dysfunction. Dna Cell Biol. 2013 Feb;32(2):41–9. doi: 10.1089/dna.2012.1874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Cai Z, Yan LJ. Rapamycin, Autophagy, and Alzheimer's Disease. Journal of biochemical and pharmacological research. 2013 Jun;1(2):84–90. [PMC free article] [PubMed] [Google Scholar]
  • 115.Chen W, Sun Y, Liu K, Sun X. Autophagy: a double-edged sword for neuronal survival after cerebral ischemia. Neural regeneration research. 2014 Jun 15;9(12):1210–6. doi: 10.4103/1673-5374.135329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Chen Y, Liu XR, Yin YQ, Lee CJ, Wang FT, Liu HQ, et al. Unravelling the multifaceted roles of Atg proteins to improve cancer therapy. Cell Prolif. 2014 Apr;47(2):105–12. doi: 10.1111/cpr.12095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Nakka VP, Prakash-Babu P, Vemuganti R. Crosstalk Between Endoplasmic Reticulum Stress, Oxidative Stress, and Autophagy: Potential Therapeutic Targets for Acute CNS Injuries. Mol Neurobiol. 2014 Dec 9; doi: 10.1007/s12035-014-9029-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Lim YM, Lim H, Hur KY, Quan W, Lee HY, Cheon H, et al. Systemic autophagy insufficiency compromises adaptation to metabolic stress and facilitates progression from obesity to diabetes. Nature communications. 2014;5:4934. doi: 10.1038/ncomms5934. [DOI] [PubMed] [Google Scholar]
  • 119.Vakifahmetoglu-Norberg H, Xia HG, Yuan J. Pharmacologic agents targeting autophagy. J Clin Invest. 2015 Jan;125(1):5–13. doi: 10.1172/JCI73937. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Wang S, Chong ZZ, Shang YC, Maiese K. WISP1 (CCN4) autoregulates its expression and nuclear trafficking of beta-catenin during oxidant stress with limited effects upon neuronal autophagy. Curr Neurovasc Res. 2012 Apr 4;9(2):89–99. doi: 10.2174/156720212800410858. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Liu Y, Shi S, Gu Z, Du Y, Liu M, Yan S, et al. Impaired autophagic function in rat islets with aging. Age (Dordr) 2013 Jul 28;35(5):1531–44. doi: 10.1007/s11357-012-9456-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.He C, Bassik MC, Moresi V, Sun K, Wei Y, Zou Z, et al. Exercise-induced BCL2-regulated autophagy is required for muscle glucose homeostasis. Nature. 2012 Jan 26;481(7382):511–5. doi: 10.1038/nature10758. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.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]
  • 124.Martino L, Masini M, Novelli M, Beffy P, Bugliani M, Marselli L, et al. Palmitate activates autophagy in INS-1E beta-cells and in isolated rat and human pancreatic islets. PLoS ONE. 2012;7(5):e36188. doi: 10.1371/journal.pone.0036188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Kousteni S. FoxO1, the transcriptional chief of staff of energy metabolism. Bone. 2012 Feb;50(2):437–43. doi: 10.1016/j.bone.2011.06.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Maiese K, Chong ZZ, Shang YC. OutFOXOing disease and disability: the therapeutic potential of targeting FoxO proteins. Trends Mol Med. 2008 May;14(5):219–27. doi: 10.1016/j.molmed.2008.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Maiese K. Forkhead Transcription Factors: Vital Elements in Biology and Medicine. Advances in Experimental Medicine and Biology, Springer Science and Business Media. 2010;665 [PubMed] [Google Scholar]
  • 128.Clark KL, Halay ED, Lai E, Burley SK. Co-crystal structure of the HNF-3/fork head DNA-recognition motif resembles histone H5. Nature. 1993 Jul 29;364(6436):412–20. doi: 10.1038/364412a0. [DOI] [PubMed] [Google Scholar]
  • 129.Jin C, Marsden I, Chen X, Liao X. Sequence specific collective motions in a winged helix DNA binding domain detected by 15N relaxation NMR. Biochemistry. 1998 Apr 28;37(17):6179–87. doi: 10.1021/bi980031v. [DOI] [PubMed] [Google Scholar]
  • 130.Maiese K, Chong ZZ, Shang YC, Hou J. A “FOXO” in sight: targeting Foxo proteins from conception to cancer. Med Res Rev. 2009 May;29(3):395–418. doi: 10.1002/med.20139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Maiese K. FoxO proteins in the nervous system. Anal Cell Pathol (Amst) 2015;2015(Article ID 569392) doi: 10.1155/2015/569392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Cheng Z, White MF. Targeting Forkhead box O1 from the concept to metabolic diseases: lessons from mouse models. Antioxid Redox Signal. 2011 Feb 15;14(4):649–61. doi: 10.1089/ars.2010.3370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Kaestner KH, Knochel W, Martinez DE. Unified nomenclature for the winged helix/forkhead transcription factors. Genes Dev. 2000 Jan 15;14(2):142–6. [PubMed] [Google Scholar]
  • 134.Lin K, Dorman JB, Rodan A, Kenyon C. daf-16: An HNF-3/forkhead family member that can function to double the life-span of Caenorhabditis elegans. Science. 1997 Nov 14;278(5341):1319–22. doi: 10.1126/science.278.5341.1319. [DOI] [PubMed] [Google Scholar]
  • 135.Ogg S, Paradis S, Gottlieb S, Patterson GI, Lee L, Tissenbaum HA, et al. The Fork head transcription factor DAF-16 transduces insulin-like metabolic and longevity signals in C. elegans. Nature. 1997 Oct 30;389(6654):994–9. doi: 10.1038/40194. [DOI] [PubMed] [Google Scholar]
  • 136.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]
  • 137.Peng S, Zhao S, Yan F, Cheng J, Huang L, Chen H, et al. HDAC2 selectively regulates FOXO3a-mediated gene transcription during oxidative stress-induced neuronal cell death. J Neurosci. 2015 Jan 21;35(3):1250–9. doi: 10.1523/JNEUROSCI.2444-14.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Carbajo-Pescador S, Mauriz JL, Garcia-Palomo A, Gonzalez-Gallego J. FoxO proteins: regulation and molecular targets in liver cancer. Curr Med Chem. 2014 Apr;21(10):1231–46. doi: 10.2174/0929867321666131228205703. [DOI] [PubMed] [Google Scholar]
  • 139.Chamorro ME, Wenker SD, Vota DM, Vittori DC, Nesse AB. Signaling pathways of cell proliferation are involved in the differential effect of erythropoietin and its carbamylated derivative. Biochim Biophys Acta. 2013 Aug;1833(8):1960–8. doi: 10.1016/j.bbamcr.2013.04.006. [DOI] [PubMed] [Google Scholar]
  • 140.Chong ZZ, Lin SH, Maiese K. The NAD+ precursor nicotinamide governs neuronal survival during oxidative stress through protein kinase B coupled to FOXO3a and mitochondrial membrane potential. J Cereb Blood Flow Metab. 2004 Jul;24(7):728–43. doi: 10.1097/01.WCB.0000122746.72175.0E. [DOI] [PubMed] [Google Scholar]
  • 141.Chong ZZ, Maiese K. Erythropoietin involves the phosphatidylinositol 3-kinase pathway, 14-3-3 protein and FOXO3a nuclear trafficking to preserve endothelial cell integrity. Br J Pharmacol. 2007 Apr;150(7):839–50. doi: 10.1038/sj.bjp.0707161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Scodelaro Bilbao P, Boland R. Extracellular ATP regulates FoxO family of transcription factors and cell cycle progression through PI3K/Akt in MCF-7 cells. Biochim Biophys Acta. 2013 Oct;1830(10):4456–69. doi: 10.1016/j.bbagen.2013.05.034. [DOI] [PubMed] [Google Scholar]
  • 143.Tao GZ, Lehwald N, Jang KY, Baek J, Xu B, Omary MB, et al. Wnt/beta-catenin signaling protects mouse liver against oxidative stress-induced apoptosis through the inhibition of forkhead transcription factor FoxO3. J Biol Chem. 2013 Jun 14;288(24):17214–24. doi: 10.1074/jbc.M112.445965. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Uranga RM, Katz S, Salvador GA. Enhanced phosphatidylinositol 3-kinase (PI3K)/Akt signaling has pleiotropic targets in hippocampal neurons exposed to iron-induced oxidative stress. J Biol Chem. 2013 Jul 5;288(27):19773–84. doi: 10.1074/jbc.M113.457622. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Wang S, Chong ZZ, Shang YC, Maiese K. WISP1 neuroprotection requires FoxO3a post-translational modulation with autoregulatory control of SIRT1. Curr Neurovasc Res. 2013 Nov 12;10(1):54–60. doi: 10.2174/156720213804805945. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Wong CP, Kaneda T, Hadi AH, Morita H. Ceramicine B, a limonoid with anti-lipid droplets accumulation activity from Chisocheton ceramicus. J Nat Med. 2014 Jan;68(1):22–30. doi: 10.1007/s11418-013-0755-2. [DOI] [PubMed] [Google Scholar]
  • 147.Zeldich E, Chen CD, Colvin TA, Bove-Fenderson EA, Liang J, Tucker Zhou TB, et al. The neuroprotective effect of Klotho is mediated via regulation of members of the redox system. J Biol Chem. 2014 Aug 29;289(35):24700–15. doi: 10.1074/jbc.M114.567321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Wang W, Yan C, Zhang J, Lin R, Lin Q, Yang L, et al. SIRT1 inhibits TNF-alpha-induced apoptosis of vascular adventitial fibroblasts partly through the deacetylation of FoxO1. Apoptosis. 2013 Jun;18(6):689–701. doi: 10.1007/s10495-013-0833-7. [DOI] [PubMed] [Google Scholar]
  • 149.Huang H, Tindall DJ. Dynamic FoxO transcription factors. J Cell Sci. 2007 Aug 1;120(Pt 15):2479–87. doi: 10.1242/jcs.001222. [DOI] [PubMed] [Google Scholar]
  • 150.Tanaka T, Iino M. Knockdown of Sec8 promotes cell-cycle arrest at G /S phase by inducing p21 via control of FOXO proteins. The FEBS journal. 2013 Dec 3; doi: 10.1111/febs.12669. [DOI] [PubMed] [Google Scholar]
  • 151.Chong ZZ, Li F, Maiese K. Activating Akt and the brain's resources to drive cellular survival and prevent inflammatory injury. Histol Histopathol. 2005 Jan;20(1):299–315. doi: 10.14670/hh-20.299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Huang CY, Chan CY, Chou IT, Lien CH, Hung HC, Lee MF. Quercetin induces growth arrest through activation of FOXO1 transcription factor in EGFR-overexpressing oral cancer cells. The Journal of nutritional biochemistry. 2013 Sep;24(9):1596–603. doi: 10.1016/j.jnutbio.2013.01.010. [DOI] [PubMed] [Google Scholar]
  • 153.Maiese K. Taking aim at Alzheimer's disease through the mammalian target of rapamycin. Ann Med. 2014 Aug 8;46(8):587–96. doi: 10.3109/07853890.2014.941921. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Park J, Ko YS, Yoon J, Kim MA, Park JW, Kim WH, et al. The forkhead transcription factor FOXO1 mediates cisplatin resistance in gastric cancer cells by activating phosphoinositide 3-kinase/Akt pathways. Gastric cancer : official journal of the International Gastric Cancer Association and the Japanese Gastric Cancer Association. 2013 Nov 8; doi: 10.1007/s10120-013-0314-2. [DOI] [PubMed] [Google Scholar]
  • 155.Qi XF, Li YJ, Chen ZY, Kim SK, Lee KJ, Cai DQ. Involvement of the FoxO3a pathway in the ischemia/reperfusion injury of cardiac microvascular endothelial cells. Experimental and molecular pathology. 2013 Oct;95(2):242–7. doi: 10.1016/j.yexmp.2013.08.003. [DOI] [PubMed] [Google Scholar]
  • 156.Leong ML, Maiyar AC, Kim B, O'Keeffe BA, Firestone GL. Expression of the serum- and glucocorticoid-inducible protein kinase, Sgk, is a cell survival response to multiple types of environmental stress stimuli in mammary epithelial cells. J Biol Chem. 2003 Feb 21;278(8):5871–82. doi: 10.1074/jbc.M211649200. [DOI] [PubMed] [Google Scholar]
  • 157.Maiese K, Chong ZZ, Shang YC, Hou J. Clever cancer strategies with FoxO transcription factors. Cell Cycle. 2008 Dec 15;7(24):3829–39. doi: 10.4161/cc.7.24.7231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Arimoto-Ishida E, Ohmichi M, Mabuchi S, Takahashi T, Ohshima C, Hayakawa J, et al. Inhibition of phosphorylation of a forkhead transcription factor sensitizes human ovarian cancer cells to cisplatin. Endocrinology. 2004 Apr;145(4):2014–22. doi: 10.1210/en.2003-1199. [DOI] [PubMed] [Google Scholar]
  • 159.Won CK, Ji HH, Koh PO. Estradiol prevents the focal cerebral ischemic injury-induced decrease of forkhead transcription factors phosphorylation. Neurosci Lett. 2006 May 1;398(1-2):39–43. doi: 10.1016/j.neulet.2005.12.060. [DOI] [PubMed] [Google Scholar]
  • 160.Lehtinen MK, Yuan Z, Boag PR, Yang Y, Villen J, Becker EB, et al. A conserved MST-FOXO signaling pathway mediates oxidative-stress responses and extends life span. Cell. 2006 Jun 2;125(5):987–1001. doi: 10.1016/j.cell.2006.03.046. [DOI] [PubMed] [Google Scholar]
  • 161.Chong ZZ, Shang YC, Wang S, Maiese K. Shedding new light on neurodegenerative diseases through the mammalian target of rapamycin. Prog Neurobiol. 2012 Aug 15;99(2):128–48. doi: 10.1016/j.pneurobio.2012.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Hou J, Wang S, Shang YC, Chong ZZ, Maiese K. Erythropoietin Employs Cell Longevity Pathways of SIRT1 to Foster Endothelial Vascular Integrity During Oxidant Stress. Curr Neurovasc Res. 2011 Aug 1;8(3):220–35. doi: 10.2174/156720211796558069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Wang F, Chan CH, Chen K, Guan X, Lin HK, Tong Q. Deacetylation of FOXO3 by SIRT1 or SIRT2 leads to Skp2-mediated FOXO3 ubiquitination and degradation. Oncogene. 2012 Mar 22;31(12):1546–57. doi: 10.1038/onc.2011.347. [DOI] [PubMed] [Google Scholar]
  • 164.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 Jul;1852(7):1311–22. doi: 10.1016/j.bbadis.2015.03.001. [DOI] [PubMed] [Google Scholar]
  • 165.Chong ZZ, Shang YC, Wang S, Maiese K. SIRT1: New avenues of discovery for disorders of oxidative stress. Expert opinion on therapeutic targets. 2012 Feb;16(2):167–78. doi: 10.1517/14728222.2012.648926. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Chong ZZ, Wang S, Shang YC, Maiese K. Targeting cardiovascular disease with novel SIRT1 pathways. Future Cardiol. 2012 Jan;8(1):89–100. doi: 10.2217/fca.11.76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Paraiso AF, Mendes KL, Santos SH. Brain activation of SIRT1: role in neuropathology. Mol Neurobiol. 2013 Dec;48(3):681–9. doi: 10.1007/s12035-013-8459-x. [DOI] [PubMed] [Google Scholar]
  • 168.Xiong S, Salazar G, Patrushev N, Alexander RW. FoxO1 Mediates an Autofeedback Loop Regulating SIRT1 Expression. J Biol Chem. 2011 Feb 18;286(7):5289–99. doi: 10.1074/jbc.M110.163667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Kobayashi Y, Furukawa-Hibi Y, Chen C, Horio Y, Isobe K, Ikeda K, et al. SIRT1 is critical regulator of FOXO-mediated transcription in response to oxidative stress. Int J Mol Med. 2005 Aug;16(2):237–43. [PubMed] [Google Scholar]
  • 170.Jagani Z, Singh A, Khosravi-Far R. FoxO tumor suppressors and BCR-ABL-induced leukemia: a matter of evasion of apoptosis. Biochim Biophys Acta. 2008 Jan;1785(1):63–84. doi: 10.1016/j.bbcan.2007.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Plas DR, Thompson CB. Akt activation promotes degradation of tuberin and FOXO3a via the proteasome. J Biol Chem. 2003 Apr 4;278(14):12361–6. doi: 10.1074/jbc.M213069200. [DOI] [PubMed] [Google Scholar]
  • 172.Maiese K, Chong ZZ, Hou J, Shang YC. The “O” class: crafting clinical care with FoxO transcription factors. Adv Exp Med Biol. 2009;665:242–60. doi: 10.1007/978-1-4419-1599-3_18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Matsuzaki H, Daitoku H, Hatta M, Aoyama H, Yoshimochi K, Fukamizu A. Acetylation of Foxo1 alters its DNA-binding ability and sensitivity to phosphorylation. Proc Natl Acad Sci U S A. 2005 Aug 9;102(32):11278–83. doi: 10.1073/pnas.0502738102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Maiese K, Chong ZZ, Shang YC. “Sly as a FOXO”: New paths with Forkhead signaling in the brain. Curr Neurovasc Res. 2007 Nov;4(4):295–302. doi: 10.2174/156720207782446306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Myatt SS, Lam EW. The emerging roles of forkhead box (Fox) proteins in cancer. Nat Rev Cancer. 2007 Nov;7(11):847–59. doi: 10.1038/nrc2223. [DOI] [PubMed] [Google Scholar]
  • 176.van der Horst A, Burgering BM. Stressing the role of FoxO proteins in lifespan and disease. Nat Rev Mol Cell Biol. 2007 Jun;8(6):440–50. doi: 10.1038/nrm2190. [DOI] [PubMed] [Google Scholar]
  • 177.Kajihara T, Jones M, Fusi L, Takano M, Feroze-Zaidi F, Pirianov G, et al. Differential expression of FOXO1 and FOXO3a confers resistance to oxidative cell death upon endometrial decidualization. Mol Endocrinol. 2006 Oct;20(10):2444–55. doi: 10.1210/me.2006-0118. [DOI] [PubMed] [Google Scholar]
  • 178.Akasaki Y, Alvarez-Garcia O, Saito M, Carames B, Iwamoto Y, Lotz MK. FOXO transcription factors support oxidative stress resistance in human chondrocytes. Arthritis & rheumatology (Hoboken, NJ) 2014 Sep 3;66(12):3349–58. doi: 10.1002/art.38868. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Tothova Z, Kollipara R, Huntly BJ, Lee BH, Castrillon DH, Cullen DE, et al. FoxOs are critical mediators of hematopoietic stem cell resistance to physiologic oxidative stress. Cell. 2007 Jan 26;128(2):325–39. doi: 10.1016/j.cell.2007.01.003. [DOI] [PubMed] [Google Scholar]
  • 180.Xia W, Zhang F, Xie C, Jiang M, Hou M. Macrophage migration inhibitory factor confers resistance to senescence through CD74-dependent AMPK-FOXO3a signaling in mesenchymal stem cells. Stem cell research & therapy. 2015 Apr 22;6(1):82. doi: 10.1186/s13287-015-0076-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Vidal RL, Figueroa A, Court FA, Thielen P, Molina C, Wirth C, et al. Targeting the UPR transcription factor XBP1 protects against Huntington's disease through the regulation of FoxO1 and autophagy. Hum Mol Genet. 2012 May 15;21(10):2245–62. doi: 10.1093/hmg/dds040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Hariharan N, Maejima Y, Nakae J, Paik J, Depinho RA, Sadoshima J. Deacetylation of FoxO by Sirt1 Plays an Essential Role in Mediating Starvation-Induced Autophagy in Cardiac Myocytes. Circ Res. 2010 Dec 10;107(12):1470–82. doi: 10.1161/CIRCRESAHA.110.227371. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Schips TG, Wietelmann A, Hohn K, Schimanski S, Walther P, Braun T, et al. FoxO3 induces reversible cardiac atrophy and autophagy in a transgenic mouse model. Cardiovasc Res. 2011 Sep 1;91(4):587–97. doi: 10.1093/cvr/cvr144. [DOI] [PubMed] [Google Scholar]
  • 184.Shi J, Yin N, Xuan LL, Yao CS, Meng AM, Hou Q. Vam3, a derivative of resveratrol, attenuates cigarette smoke-induced autophagy. Acta Pharmacol Sin. 2012 Jul;33(7):888–96. doi: 10.1038/aps.2012.73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Shang YC, Chong ZZ, Hou J, Maiese K. The forkhead transcription factor FoxO3a controls microglial inflammatory activation and eventual apoptotic injury through caspase 3. Curr Neurovasc Res. 2009 Feb;6(1):20–31. doi: 10.2174/156720209787466064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Shang YC, Chong ZZ, Hou J, Maiese K. Wnt1, FoxO3a, and NF-kappaB oversee microglial integrity and activation during oxidant stress. Cell Signal. 2010 Sep;22(9):1317–29. doi: 10.1016/j.cellsig.2010.04.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Shang YC, Chong ZZ, Hou J, Maiese K. FoxO3a governs early microglial proliferation and employs mitochondrial depolarization with caspase 3, 8, and 9 cleavage during oxidant induced apoptosis. Curr Neurovasc Res. 2009 Nov;6(4):223–38. doi: 10.2174/156720209789630302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Anitha M, Gondha C, Sutliff R, Parsadanian A, Mwangi S, Sitaraman SV, et al. GDNF rescues hyperglycemia-induced diabetic enteric neuropathy through activation of the PI3K/Akt pathway. J Clin Invest. 2006 Feb;116(2):344–56. doi: 10.1172/JCI26295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Zheng WH, Kar S, Quirion R. FKHRL1 and its homologs are new targets of nerve growth factor Trk receptor signaling. J Neurochem. 2002 Mar;80(6):1049–61. doi: 10.1046/j.0022-3042.2002.00783.x. [DOI] [PubMed] [Google Scholar]
  • 190.Zhu W, Bijur GN, Styles NA, Li X. Regulation of FOXO3a by brain-derived neurotrophic factor in differentiated human SH-SY5Y neuroblastoma cells. Brain Res Mol Brain Res. 2004 Jul 5;126(1):45–56. doi: 10.1016/j.molbrainres.2004.03.019. [DOI] [PubMed] [Google Scholar]
  • 191.Chong ZZ, Li F, Maiese K. Group I Metabotropic Receptor Neuroprotection Requires Akt and Its Substrates that Govern FOXO3a, Bim, and beta-Catenin During Oxidative Stress. Curr Neurovasc Res. 2006 May;3(2):107–17. doi: 10.2174/156720206776875830. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Kashii Y, Uchida M, Kirito K, Tanaka M, Nishijima K, Toshima M, et al. A member of Forkhead family transcription factor, FKHRL1, is one of the downstream molecules of phosphatidylinositol 3-kinase-Akt activation pathway in erythropoietin signal transduction. Blood. 2000;96(3):941–9. [PubMed] [Google Scholar]
  • 193.Andreucci M, Fuiano G, Presta P, Lucisano G, Leone F, Fuiano L, et al. Downregulation of cell survival signalling pathways and increased cell damage in hydrogen peroxide-treated human renal proximal tubular cells by alpha-erythropoietin. Cell Prolif. 2009 Aug;42(4):554–61. doi: 10.1111/j.1365-2184.2009.00617.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Maiese K. WISP1: Clinical Insights for a Proliferative and Restorative Member of the CCN Family. Curr Neurovasc Res. 2014 Sep 12;11(4):378–89. doi: 10.2174/1567202611666140912115107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Murahovschi V, Pivovarova O, Ilkavets I, Dmitrieva RM, Docke S, Keyhani-Nejad F, et al. WISP1 is a novel adipokine linked to inflammation in obesity. Diabetes. 2014 Oct 3; doi: 10.2337/db14-0444. [DOI] [PubMed] [Google Scholar]
  • 196.Bayod S, Menella I, Sanchez-Roige S, Lalanza JF, Escorihuela RM, Camins A, et al. Wnt pathway regulation by long-term moderate exercise in rat hippocampus. Brain Res. 2014 Jan 16;1543:38–48. doi: 10.1016/j.brainres.2013.10.048. [DOI] [PubMed] [Google Scholar]
  • 197.Berwick DC, Harvey K. The regulation and deregulation of Wnt signaling by PARK genes in health and disease. J Mol Cell Biol. 2014 Feb;6(1):3–12. doi: 10.1093/jmcb/mjt037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Maiese K, Li F, Chong ZZ, Shang YC. The Wnt signaling pathway: Aging gracefully as a protectionist? Pharmacol Ther. 2008 Apr;118(1):58–81. doi: 10.1016/j.pharmthera.2008.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Marchetti B, Pluchino S. Wnt your brain be inflamed? Yes, it Wnt! Trends Mol Med. 2013 Mar;19(3):144–56. doi: 10.1016/j.molmed.2012.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.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]
  • 201.Yang Y, Su Y, Wang D, Chen Y, Wu T, Li G, et al. Tanshinol attenuates the deleterious effects of oxidative stress on osteoblastic differentiation via Wnt/FoxO3a signaling. Oxid Med Cell Longev. 2013;2013:351895. doi: 10.1155/2013/351895. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Liu H, Yin J, Wang H, Jiang G, Deng M, Zhang G, et al. FOXO3a modulates WNT/beta-catenin signaling and suppresses epithelial-to-mesenchymal transition in prostate cancer cells. Cell Signal. 2015 Mar;27(3):510–8. doi: 10.1016/j.cellsig.2015.01.001. [DOI] [PubMed] [Google Scholar]
  • 203.Tan C, Liu S, Tan S, Zeng X, Yu H, Li A, et al. Polymorphisms in MicroRNA Target Sites of Forkhead Box O Genes Are Associated with Hepatocellular Carcinoma. PLoS One. 2015;10(3):e0119210. doi: 10.1371/journal.pone.0119210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Wang GJ, Liu GH, Ye YW, Fu Y, Zhang XF. The role of microRNA-1274a in the tumorigenesis of gastric cancer: Accelerating cancer cell proliferation and migration via directly targeting FOXO4. Biochem Biophys Res Commun. 2015 Apr 17;459(4):629–35. doi: 10.1016/j.bbrc.2015.02.160. [DOI] [PubMed] [Google Scholar]
  • 205.I OS, Zhang W, Wasserman DH, Liew CW, Liu J, Paik J, et al. FoxO1 integrates direct and indirect effects of insulin on hepatic glucose production and glucose utilization. Nature communications. 2015;6:7079. doi: 10.1038/ncomms8079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Calabuig-Navarro V, Yamauchi J, Lee S, Zhang T, Liu YZ, Sadlek K, et al. FoxO6 Depletion Attenuates Hepatic Gluconeogenesis and Protects Against Fat-Induced Glucose Disorder in Mice. J Biol Chem. 2015 May 5; doi: 10.1074/jbc.M115.650994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Kibbe C, Chen J, Xu G, Jing G, Shalev A. FOXO1 competes with carbohydrate response element-binding protein (ChREBP) and inhibits thioredoxin-interacting protein (TXNIP) transcription in pancreatic beta cells. J Biol Chem. 2013 Aug 9;288(32):23194–202. doi: 10.1074/jbc.M113.473082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Tang L, Zhang Y, Jiang Y, Willard L, Ortiz E, Wark L, et al. Dietary wolfberry ameliorates retinal structure abnormalities in db/db mice at the early stage of diabetes. Exp Biol Med (Maywood) 2011 Sep 1;236(9):1051–63. doi: 10.1258/ebm.2011.010400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Wang RH, Kim HS, Xiao C, Xu X, Gavrilova O, Deng CX. Hepatic Sirt1 deficiency in mice impairs mTorc2/Akt signaling and results in hyperglycemia, oxidative damage, and insulin resistance. J Clin Invest. 2011 Nov;121(11):4477–90. doi: 10.1172/JCI46243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Kato M, Yuan H, Xu ZG, Lanting L, Li SL, Wang M, et al. Role of the Akt/FoxO3a pathway in TGF-beta1-mediated mesangial cell dysfunction: a novel mechanism related to diabetic kidney disease. J Am Soc Nephrol. 2006 Dec;17(12):3325–35. doi: 10.1681/ASN.2006070754. [DOI] [PubMed] [Google Scholar]
  • 211.Kamei Y, Miura S, Suzuki M, Kai Y, Mizukami J, Taniguchi T, et al. Skeletal muscle FOXO1 (FKHR) transgenic mice have less skeletal muscle mass, down-regulated Type I (slow twitch/red muscle) fiber genes, and impaired glycemic control. J Biol Chem. 2004 Sep 24;279(39):41114–23. doi: 10.1074/jbc.M400674200. [DOI] [PubMed] [Google Scholar]
  • 212.Kim JR, Jung HS, Bae SW, Kim JH, Park BL, Choi YH, et al. Polymorphisms in FOXO gene family and association analysis with BMI. Obesity (Silver Spring, Md. 2006 Feb;14(2):188–93. doi: 10.1038/oby.2006.24. [DOI] [PubMed] [Google Scholar]
  • 213.Kuningas M, Magi R, Westendorp RG, Slagboom PE, Remm M, van Heemst D. Haplotypes in the human Foxo1a and Foxo3a genes; impact on disease and mortality at old age. Eur J Hum Genet. 2007 Mar;15(3):294–301. doi: 10.1038/sj.ejhg.5201766. [DOI] [PubMed] [Google Scholar]
  • 214.Chong ZZ, Lin SH, Li F, Maiese K. The sirtuin inhibitor nicotinamide enhances neuronal cell survival during acute anoxic injury through Akt, Bad, PARP, and mitochondrial associated “anti-apoptotic” pathways. Curr Neurovasc Res. 2005 Oct;2(4):271–85. doi: 10.2174/156720205774322584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Chong ZZ, Maiese K. Enhanced Tolerance against Early and Late Apoptotic Oxidative Stress in Mammalian Neurons through Nicotinamidase and Sirtuin Mediated Pathways. Curr Neurovasc Res. 2008 Aug;5(3):159–70. doi: 10.2174/156720208785425666. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Guo JM, Dong WZ, Liu AJ, Cheng MH, Su DF. Nicotinamide postpones stroke in stroke-prone spontaneously hypertensive rats. CNS Neurosci Ther. 2012 Mar;18(3):267–8. doi: 10.1111/j.1755-5949.2011.00287.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Itzhaki O, Greenberg E, Shalmon B, Kubi A, Treves AJ, Shapira-Frommer R, et al. Nicotinamide inhibits vasculogenic mimicry, an alternative vascularization pathway observed in highly aggressive melanoma. PLoS One. 2013;8(2):e57160. doi: 10.1371/journal.pone.0057160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Kwak JY, Ham HJ, Kim CM, Hwang ES. Nicotinamide exerts antioxidative effects on senescent cells. Mol Cells. 2015 Mar;38(3):229–35. doi: 10.14348/molcells.2015.2253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Maiese K, Chong ZZ. Nicotinamide: necessary nutrient emerges as a novel cytoprotectant for the brain. Trends Pharmacol Sci. 2003 May;24(5):228–32. doi: 10.1016/S0165-6147(03)00078-6. [DOI] [PubMed] [Google Scholar]
  • 220.Oblong JE. The evolving role of the NAD+/nicotinamide metabolome in skin homeostasis, cellular bioenergetics, and aging. DNA Repair (Amst) 2014 Nov;23:59–63. doi: 10.1016/j.dnarep.2014.04.005. [DOI] [PubMed] [Google Scholar]
  • 221.Peterson TC, Hoane MR, McConomy K, Farin F, Bammler T, MacDonald J, et al. A Combination Therapy of Nicotinamide and Progesterone Improves Functional Recovery Following Traumatic Brain Injury. J Neurotrauma. 2014 Oct 14; doi: 10.1089/neu.2014.3530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Vonder Haar C, Maass WR, Jacobs EA, Hoane MR. Deficits in discrimination after experimental frontal brain injury are mediated by motivation and can be improved by nicotinamide administration. J Neurotrauma. 2014 Oct 15;31(20):1711–20. doi: 10.1089/neu.2014.3459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Wang L, Ding D, Salvi R, Roth JA. Nicotinamide adenine dinucleotide prevents neuroaxonal degeneration induced by manganese in cochlear organotypic cultures. Neurotoxicology. 2014 Jan;40:65–74. doi: 10.1016/j.neuro.2013.11.007. [DOI] [PubMed] [Google Scholar]
  • 224.Crino A, Schiaffini R, Ciampalini P, Suraci MC, Manfrini S, Visalli N, et al. A two year observational study of nicotinamide and intensive insulin therapy in patients with recent onset type 1 diabetes mellitus. J Pediatr Endocrinol Metab. 2005 Aug;18(8):749–54. doi: 10.1515/jpem.2005.18.8.749. [DOI] [PubMed] [Google Scholar]
  • 225.Olmos PR, Hodgson MI, Maiz A, Manrique M, De Valdes MD, Foncea R, et al. Nicotinamide protected first-phase insulin response (FPIR) and prevented clinical disease in first-degree relatives of type-1 diabetics. Diabetes Res Clin Pract. 2006 Mar;71(3):320–33. doi: 10.1016/j.diabres.2005.07.009. [DOI] [PubMed] [Google Scholar]
  • 226.Stevens MJ, Li F, Drel VR, Abatan OI, Kim H, Burnett D, et al. Nicotinamide reverses neurological and neurovascular deficits in streptozotocin diabetic rats. J Pharmacol Exp Ther. 2007 Jan;320(1):458–64. doi: 10.1124/jpet.106.109702. [DOI] [PubMed] [Google Scholar]
  • 227.Liu HK, Green BD, Flatt PR, McClenaghan NH, McCluskey JT. Effects of long-term exposure to nicotinamide and sodium butyrate on growth, viability, and the function of clonal insulin secreting cells. Endocr Res. 2004 Feb;30(1):61–8. doi: 10.1081/erc-120028485. [DOI] [PubMed] [Google Scholar]
  • 228.Reddy S, Salari-Lak N, Sandler S. Long-term effects of nicotinamide-induced inhibition of poly(adenosine diphosphate-ribose) polymerase activity in rat pancreatic islets exposed to interleukin-1 beta. Endocrinology. 1995;136(5):1907–12. doi: 10.1210/endo.136.5.7720637. [DOI] [PubMed] [Google Scholar]
  • 229.Balan V, Miller GS, Kaplun L, Balan K, Chong ZZ, Li F, et al. Life span extension and neuronal cell protection by Drosophila nicotinamidase. J Biol Chem. 2008 Oct 10;283(41):27810–9. doi: 10.1074/jbc.M804681200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Moroz N, Carmona JJ, Anderson E, Hart AC, Sinclair DA, Blackwell TK. Dietary restriction involves NAD -dependent mechanisms and a shift toward oxidative metabolism. Aging Cell. 2014 Sep 25;13(6):1075–85. doi: 10.1111/acel.12273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Sanchez-Carron G, Garcia-Garcia MI, Zapata-Perez R, Takami H, Garcia-Carmona F, Sanchez-Ferrer A. Biochemical and mutational analysis of a novel nicotinamidase from Oceanobacillus iheyensis HTE831. PLoS One. 2013;8(2):e56727. doi: 10.1371/journal.pone.0056727. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Zhao Y, Yu Y, Tian X, Yang X, Li X, Jiang F, et al. Association Study to Evaluate FoxO1 and FoxO3 Gene in CHD in Han Chinese. PLoS One. 2014;9(1):e86252. doi: 10.1371/journal.pone.0086252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Maiese K, Hou J, Chong ZZ, Shang YC. Erythropoietin, forkhead proteins, and oxidative injury: biomarkers and biology. ScientificWorldJournal. 2009;9:1072–104. doi: 10.1100/tsw.2009.121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Guo S, Rena G, Cichy S, He X, Cohen P, Unterman T. Phosphorylation of serine 256 by protein kinase B disrupts transactivation by FKHR and mediates effects of insulin on insulin-like growth factor-binding protein-1 promoter activity through a conserved insulin response sequence. J Biol Chem. 1999 Jun 11;274(24):17184–92. doi: 10.1074/jbc.274.24.17184. [DOI] [PubMed] [Google Scholar]
  • 235.Nakae J, Park BC, Accili D. Insulin stimulates phosphorylation of the forkhead transcription factor FKHR on serine 253 through a Wortmannin-sensitive pathway. J Biol Chem. 1999 Jun 4;274(23):15982–5. doi: 10.1074/jbc.274.23.15982. [DOI] [PubMed] [Google Scholar]
  • 236.Fallarino F, Bianchi R, Orabona C, Vacca C, Belladonna ML, Fioretti MC, et al. CTLA-4-Ig activates forkhead transcription factors and protects dendritic cells from oxidative stress in nonobese diabetic mice. J Exp Med. 2004 Oct 18;200(8):1051–62. doi: 10.1084/jem.20040942. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Furuyama T, Yamashita H, Kitayama K, Higami Y, Shimokawa I, Mori N. Effects of aging and caloric restriction on the gene expression of Foxo1, 3, and 4 (FKHR, FKHRL1, and AFX) in the rat skeletal muscles. Microscopy research and technique. 2002 Nov 15;59(4):331–4. doi: 10.1002/jemt.10213. [DOI] [PubMed] [Google Scholar]
  • 238.Puig O, Tjian R. Transcriptional feedback control of insulin receptor by dFOXO/FOXO1. Genes Dev. 2005 Oct 15;19(20):2435–46. doi: 10.1101/gad.1340505. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES