Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Oct 25.
Published in final edited form as: Curr Neurovasc Res. 2016;13(4):329–340. doi: 10.2174/1567202613666160729164900

Erythropoietin and mTOR: A “One-Two Punch” for Aging-Related Disorders Accompanied by Enhanced Life Expectancy

Kenneth Maiese 1,*
PMCID: PMC5079807  NIHMSID: NIHMS807750  PMID: 27488211

Abstract

Life expectancy continues to increase throughout the world, but is accompanied by a rise in the incidence of non-communicable diseases. As a result, the benefits of an increased lifespan can be limited by aging-related disorders that necessitate new directives for the development of effective and safe treatment modalities. With this objective, the mechanistic target of rapamycin (mTOR), a 289-kDa serine/threonine protein, and its related pathways of mTOR Complex 1 (mTORC1), mTOR Complex 2 (mTORC2), proline rich Akt substrate 40 kDa (PRAS40), AMP activated protein kinase (AMPK), Wnt signaling, and silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae) (SIRT1), have generated significant excitement for furthering novel therapies applicable to multiple systems of the body. Yet, the biological and clinical outcome of these pathways can be complex especially with oversight of cell death mechanisms that involve apoptosis and autophagy. Growth factors, and in particular erythropoietin (EPO), are one avenue under consideration to implement control over cell death pathways since EPO can offer potential treatment for multiple disease entities and is intimately dependent upon mTOR signaling. In experimental and clinical studies, EPO appears to have significant efficacy in treating several disorders including those involving the developing brain. However, in mature populations that are affected by aging-related disorders, the direction for the use of EPO to treat clinical disease is less clear that may be dependent upon a number of factors including the understanding of mTOR signaling. Continued focus upon the regulatory elements that control EPO and mTOR signaling could generate critical insights for targeting a broad range of clinical maladies.

Keywords: Akt, aging, aging-related disorders, Alzheimer’s disease, AMP activated protein kinase (AMPK), apoptosis, autophagy, cardiovascular disease, caspase, diabetes mellitus, epidermal growth factor, erythropoietin, hamartin (tuberous sclerosis 1)/tuberin (tuberous sclerosis 2) (TSC1/TSC2), Huntington’s disease, hypoxia-inducible factor 1, insulin, lifespan, mechanistic target of rapamycin (mTOR), mTOR Complex 1 (mTORC1), mTOR Complex 2 (mTORC2), nicotinamide, nicotinamide adenine dinucleotide (NAD+), non-communicable diseases, oxidative stress, phosphoinositide 3–kinase (PI 3-K), programmed cell death, proline rich Akt substrate 40 kDa (PRAS40), silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae) (SIRT1), sirtuin, stem cells, wingless, Wnt, Wnt1 inducible signaling pathway protein 1 (WISP1)

Increased Lifespan, Degenerative Disorders, and Cell Injury

Throughout the globe, both lifespan and the incidence of non-communicable diseases (NCDs) are increasing (1). The world’s population is becoming older (2) and the number of individuals over the age of sixty-five has more than doubled during the previous fifty years. Life expectancy is approaching eighty years of age and has been marked by a one percent decrease in the age-adjusted death rate from the years 2000 through 2011 (3). In addition, the incidence of non-communicable diseases (NCDs) is rising and according to the World Health Organization more than sixty percent of the fifty-seven million global deaths are attributable to NCDs (4). This rise in NCDs closely follows the increase in life expectancy of the world’s population (Table 1).

Table 1.

Highlights for Designing Novel Treatments with Erythropoietin and mTOR

  • Both life expectancy and the incidence of non-communicable diseases are increasing throughout the globe that necessitates new strategies to treat aging-related disorders.

  • The mechanistic target of rapamycin (mTOR), a 289-kDa serine/threonine protein, and its related pathways of mTOR Complex 1 (mTORC1), mTOR Complex 2 (mTORC2), proline rich Akt substrate 40 kDa (PRAS40), AMP activated protein kinase (AMPK), Wnt signaling, and silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae) (SIRT1), offer exciting prospects for the design of therapies for multiple disorders.

  • Closely coupled to the ability of mTOR signaling to govern cell survival is the oversight of the programmed death pathways of apoptosis and autophagy.

  • A fine control of mTOR is necessary over apoptotic and autophagic pathways to achieve desired biological and clinical outcomes.

  • Erythropoietin (EPO) offers an exciting prospect to couple with the benefits of mTOR signaling, since EPO relies upon mTOR pathways and can implement control over apoptosis and autophagy to foster potential treatment for multiple disease entities.

  • Although EPO is gaining clinical acceptance in cell and animal studies as well as clinical studies for the developing brain, applications of EPO for mature and aging adults require further clarification that can benefit from continued elucidation the regulatory elements that control EPO and mTOR signaling.

With increasing age, various systems of the body can become affected by disease. Accumulation of senescent cells with increased age is believed to contribute to loss of tissue and organ function (5, 6). Oxidative, metabolic, and mechanical stressors also increase with age that can promote tissue injury (711). Systems of the body may become affected with age-related disorders disorders, such as lung disease and increased inflammation, which can become detrimental to overall lifespan (12). Cardiovascular disease (13, 14), musculoskeletal disease (15, 16), and psychiatric disorders (17) also can progress with aging. Equally important is the impact of aging on the nervous system (18). Learning and memory capacity can decline with loss of hippocampal and cortical neurons (19, 20) and may ultimately result in progressive neurodegenerative disease (2125).

As a result of progressive cellular and system insults in the body, clinical disease can ensue. For example, infections of prion agents lead to progressive and fatal neurodegenerative diseases (26). Cardiovascular disease and diabetes mellitus (DM) can lead to acute neurodegenerative diseases such as stroke (27, 28). In regards to neurodegenerative disorders (29, 30), 800,000 strokes occur per year in the United States (US) at an annual cost of 75 billion US dollars (29). Traumatic brain injury (TBI) (31, 32) can have multiple effects resulting in acute injury to the nervous system as well as subsequent chronic aging-related impairment (3335). Approximately 50,000 individuals die every year as a result of TBI and more than 100,000 individuals suffer with chronic disability (36). Furthermore, at least ten percent of the global population over the age of sixty-five are affected with sporadic Alzheimer’s disease (AD) (37, 38). In contrast, familial cases of AD represent less than 2 percent of all presentations (24), usually occur prior to age 55 (39), and represent an autosomal dominant form of a mutated amyloid precursor protein (APP) gene as well as mutations in the presenilin 1 or 2 genes (40).

Closely tied to the progression of clinical disease with advanced age are metabolic disorders (1, 10, 11, 41, 42) that includes DM (8, 43). In particular, DM is increasing in incidence throughout the world (28, 41) and it is predicted that greater than 350 million individuals currently have DM (4448). Approximately an additional eight million individuals are considered to suffer from metabolic disorders but are yet to be identified as having DM (4951). Overall, per reports by the Centers for Medicare and Medicaid Services (CMS), the care for patients with DM consumes 17 percent of the Gross Domestic Product in the US as reported by (52).

Disorders such as DM affect all systems of the body (10, 11, 5360). For example, DM can negatively impact the immune system, musculoskeletal function, hepatic metabolism, and renal clearance (50, 53, 55, 59, 6164). In the cardiovascular system, DM can lead to impaired angiogenesis, platelet dysfunction, atherosclerosis, endothelial progenitor cell injury, cardiac impairment, and loss of vascular cells (8, 14, 27, 62, 6568). In the nervous system, DM results in cortical injury and stroke (62, 67, 6972), retinal disease (50, 7375), dementia (76), AD (51, 61, 71, 77), peripheral neuropathy (78, 79), and psychiatric disorders (80, 81).

The mechanistic target of rapamycin (mTOR)

Multiple cellular targets may be at the root of degenerative disorders that occur with increased lifespan and advanced aging, but it is the mechanistic target of rapamycin (mTOR), a 289-kDa serine/threonine protein, that has generated considerable excitement for developing novel therapies to avert cellular dysfunction and cell death (8286) (Table 1). mTOR is considered a vital component that affects multiple cellular pathways during the aging process (6, 19, 8790). mTOR also is known as the mammalian target of rapamycin and the FK506-binding protein 12-rapamycin complex-associated protein 1 (37). The target of rapamycin (TOR) was first documented in Saccharomyces cerevisiae with the genes TOR1 and TOR2 (90). TOR1 and TOR2 were found to encode the Tor1 and Tor2 isoforms in yeast through the use of rapamycin-resistant TOR mutants. Both TOR and mTOR activity can be blocked by rapamycin, a macrolide antibiotic in that exists in Streptomyces hygroscopicus.

Encoded by a single gene FRAP1, mTOR is the principal component of the protein complexes mTOR Complex 1 (mTORC1) and mTOR Complex 2 (mTORC2) (37, 87, 91, 92). mTORC1 contains the components Raptor, Deptor (DEP domain-containing mTOR interacting protein), the proline rich Akt substrate 40 kDa (PRAS40), and mammalian lethal with Sec13 protein 8, termed mLST8 (mLST8/GβL) (90). As noted, rapamacyin can block mTOR activity. Without the presence of rapamaycin, mTOR fosters Raptor activity. This involves the binding of p70 ribosomal S6 kinase (p70S6K) and the eukaryotic initiation factor 4E (eIF4E)-binding protein 1 (4EBP1) to Raptor. mLST8 also promotes mTOR kinase activity. In contrast, Deptor is inhibitory and blocks mTORC1 activity by binding to the FAT (FKBP12 -rapamycin-associated protein (FRAP), ataxia-telangiectasia (ATM), and the transactivation/transformation domain-associated protein) domain of mTOR. In addition, PRAS40 blocks mTORC1 activity by preventing the association of p70S6K and 4EBP1 with Raptor (93, 94). mTORC1 becomes active once PRAS40 is phosphorylated by protein kinase B (Akt) to release PRAS40 from Raptor and sequester PRAS40 with the cytoplasmic docking protein 14-3-3 (9599). mTORC2 contains Rictor, Deptor, mLST8, the mammalian stress-activated protein kinase interacting protein (mSIN1), and the protein observed with Rictor-1 (Protor-1) (87, 100). mTORC2 activates protein kinases, such as glucocorticoid induced protein kinase 1 (SGK1), a member of the protein kinase A/protein kinase G/protein kinase C (AGC) family of protein kinases. Protor-1, a Rictor-binding subunit of mTORC2, activates SGK1 (101, 102). The kinase domain of mTOR phosphorylates mSIN1 to prevent lysosomal degradation of this protein. Rictor (103) and mSIN1 (104) promote cellular survival by phosphorylating Akt at serine473 and leading to threonine308 phosphorylation by phosphoinositide-dependent kinase 1 (PDK1).

Vital in the signaling cascade of mTOR and cell survival is AMP activated protein kinase (AMPK) (1, 105, 106). Under some conditions, AMPK may be protective for cell survival and also regulate cellular metabolism through its inhibition of mTOR (106, 107). Metformin, an agent that controls hyperglycemia in DM, blocks mTOR activity through AMPK and leads to the induction of autophagy. As a result, cardiomyopathy is limited in experimental models of DM (108) and endothelial cell senescence is reduced during applications of metformin (109). AMPK maintains metabolic function of cells, prevents atherosclerosis, modulates immune system activity, and can prevent β-amyloid (Aβ) accumulation (25, 40, 110113). AMPK activation also may improve memory retention in models of AD and DM (114). With silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae) (SIRT1) (13, 62), AMPK can protect endothelial cells through the induction of autophagy against oxidized low-density lipoproteins (115). Yet, limited AMPK activity also may be required for cellular protection. For example, reduced AMPK activity can lead to the protection of pancreatic islet cells in mice (116), limit Aβ toxicity in cerebral microglial cells (117), and prevent inflammation in the nervous system (118).

AMPK blocks mTORC1 activity through the activation of the hamartin (tuberous sclerosis 1)/tuberin (tuberous sclerosis 2) (TSC1/TSC2) complex. TSC2 is a GTPase-activating protein (GAP) converting G protein Rheb (Rheb-GTP) into the inactive GDP-bound form (Rheb-GDP). Once Rheb-GTP is active, Rheb-GTP combines with Raptor to control the binding of 4EBP1 to mTORC1 and increase mTORC1 activity (119). AMPK phosphorylates TSC2 to increase GAP activity to change Rheb-GTP into the inactive Rheb-GDP and to inhibit mTORC1 activity (120). As noted above, AMPK also functions with SIRT1 (13, 62). In addition, AMPK can increase nicotinamide phosphoribosyltransferase (NAMPT) activity to convert nicotinamide to nicotinamide mononucleotide (57, 121, 122). This conversion of nicotinamide increases nicotinamide adenine dinucleotide (NAD+) levels, decreases levels of the SIRT1 inhibitor nicotinamide (46, 121), and promotes SIRT1 transcription (28, 123, 124).

In addition to AMPK, phosphoinositide 3-kinase (PI 3-K) and Akt also play significant roles in the mTOR signaling pathway (9, 21, 85). The terminal domains of mTOR oversee the catalytic activity, binding, and phosphorylation of mTOR (90). The C-terminal domain of mTOR has a sequence homology to the catalytic domain of the PI 3-K family and contains several phosphorylation sites that regulate mTOR. Downstream from PI 3-K, Akt can block activity of the TSC1/TSC2 complex that inhibits mTORC1 (125128). Akt controls TSC1/TSC2 complex and its phosphorylation of TSC2 (29). Other pathways, such as extracellular signal-regulated kinases (ERKs), protein p90 ribosomal S6 kinase 1 (RSK1), and glycogen synthase kinase -3β (GSK-3β) also can modulate the activity TSC1/TSC2 complex. It should be noted that under some conditions that promote cell survival, a limited activity of TSC2 and AMPK is necessary since complete knockdown of TSC2 can result in cell death (117). Although the TSC1/TSC2 complex inhibits mTORC1 activity, mTORC2 activity is increased during activation of the TSC1/TSC2 complex through the amino (N)-terminal region of TSC2 and the C-terminal region of Rictor (129).

Targeting cell death through mTOR

mTOR oversees pathways of cell death through apoptosis and autophagy, two mechanisms that can determine cell survival through programmed cell death (105, 130, 131) (Table 1). Apoptosis results in the activation of nucleases and proteases involving caspases (132). These processes impact both the early phase of apoptosis with the loss of plasma membrane phosphatidylserine (PS) asymmetry and a later phase that leads to genomic DNA degradation (133135). Loss of membrane PS asymmetry activates inflammatory cells to engulf and remove injured cells (136139). However, if the engulfment of inflammatory cells can be prevented, functional cells expressing membrane PS residues can rescued (50, 140142). In contrast, the destruction of cellular DNA, once it occurs, is usually not considered to be completely reversible (143).

Activation of mTOR usually blocks apoptotic cell death in the nervous system (87, 144). Loss of mTOR activity (105) or components of this pathway, such as mTORC2, can result in apoptotic cell death (130). Activation of either mTOR or downstream pathways that involve p70S6K can protect against Aβ toxicity (21, 25, 97, 117, 145148), cerebral ischemia (149, 150), and oxidative stress exposure (151, 152). During metabolic disease such as DM (28), mTOR activation can prevent the development of atherosclerosis (153) and through glucagon-like peptide-1 agonists can protect pancreatic β-cells from cholesterol mediated apoptotic cell injury (152). In addition, mTOR activation can prevent neural apoptotic cell loss during DM through the epidermal growth factor receptor (154) and foster pancreatic β-cell proliferation (155)

Yet, in some cases, limiting the activity of mTOR may be necessary to block apoptotic cell death (156). In such cases, induction of autophagy during mTOR inhibition leads to the prevention of cell death. At least 33 autophagic related genes (Atg) have been identified in yeast with TOR and can affect multiple disorders (57, 157). Atg1, Atg13 (also known as Apg13), and Atg17 are associated with the PI 3-K, Akt, and TOR pathways (158). Autophagy recycles components of the cell cytoplasm for tissue remodeling and to remove non-functional organelles (30, 131, 159, 160). Macroautophagy is the classification of autophagy that recycles organelles and consists of the sequestration of cytoplasmic proteins and organelles into autophagosomes that combine with lysosomes for degradation and recycling (93, 161, 162). Other categories of autophagy involve microautophagy that uses the invagination of the lysosomal membrane for the sequestration and digestion of cytoplasmic components (131). Chaperone-mediated autophagy uses cytosolic chaperones to transport cytoplasmic components across lysosomal membranes (163).

Autophagy with the inhibition of mTOR activity can be protective for cells. Inhibition of mTOR with the induction of autophagy can increase cell survival in neonatal models of ischemia (164) and during excitotoxicity (165). Autophagy protects cells during prion protein disease (166) and in models of Huntington’s disease (HD)(90, 167). Autophagy activation can reduce Aβ production and improve memory function in animal models of AD (168). In a similar manner, loss of autophagy may be detrimental. In experimental models of AD, disease progression and duration increases with dysfunctional autophagic processes and reduction in mTOR activity (159). Vascular cell injury occurs during cerebral ischemia if autophagy is blocked (169). In regards to metabolic disease, consumption of a high calorie diet can block autophagy and facilitate hippocampal neuronal loss in mice (20). In addition, autophagy haploinsufficiency with deletion of an essential Atg7 gene in mouse models of obesity promotes increased insulin resistance with elevated lipids and inflammation (170). Similar to disorders such as HD, autophagy also may be necessary for the removal of misfolded proteins and to eliminate non-functioning mitochondria to maintain β-cell function and prevent the onset of DM (171). Autophagy also can improve insulin sensitivity during high fat diets in mice (172).

Induction of autophagy may be regulated not only through mTOR, but also through SIRT1 (173, 174). SIRT1 increases lifespan in higher organisms and provides protection against oxidative stress (175). SIRT1 is protective, such as with erythropoietin (EPO) growth factor protection (46, 176180), and has an inverse relationship with mTOR (62). SIRT1 inhibits mTOR pathways and promotes autophagy to protect human embryonic stem cells against oxidative stress (181). SIRT1 blocks mTOR signaling to promote neuronal growth (182) and can oversee cellular metabolism during caloric restriction (183). In endothelial cells exposed to oxidized low density lipoproteins that can lead to atherosclerosis, SIRT1 up-regulation in combination with AMPK activity and mTOR inhibition results in autophagy that is necessary for cell protection (115).

Yet, autophagy is not always beneficial and may lead to cell injury. During Wnt1 inducible signaling pathway protein 1 (WISP1) signaling, autophagy may be a component of cell death in addition to apoptosis (184). Increased activity of autophagy can lead to loss of cardiac and liver tissue in diabetic rats during attempts to achieve glycemic control through diet modification (185). During periods of elevated glucose, advanced glycation end products (AGEs), agents that can result in complications during DM, can result in the induction of autophagy and vascular smooth muscle proliferation with atherosclerosis progression (186) as well as cardiomyopathy (187). With periods of elevated glucose exposure, autophagy can injure endothelial progenitor cells, lead to mitochondrial oxidative stress (188), and block angiogenesis (189). Furthermore, a reduction in autophagy with the activation of mTOR in animal models of traumatic spinal cord injury leads to improvement in function and increased survival of motor neurons (190). Inhibition of autophagy also reduces infarct size and protects cerebral neurons during experimental stroke (191). Blockade of autophagy and activation of mTOR protects dopaminergic neurons during oxidative stress exposure (192, 193). In tri-cultures of neurons, astrocytes, and microglia that are exposed to inflammatory stressors and Aβ, cell injury is worse during the induction of autophagy (194).

Erythropoietin and the modulation of mTOR

It is evident that a fine balance is required between apoptotic and autophagic pathways to achieve optimal cell survival and disease reduction. Growth factors are one avenue to consider to achieve fine control over cell injury pathways of apoptosis and autophagy. Growth factors are advocated for the treatment of both acute and chronic disorders (71, 195199). In particular, the growth factor EPO is considered to offer an exciting therapeutic strategy for the treatment of disorders that result in cell injury and cell death especially since it is intimately involved with mTOR signaling (Table 1) (85, 200207). EPO has the capacity to offer protection against a number of disease entities (208212) as well as enhanced biological activity (213, 214). For example, EPO has been reported to improve clinical outcome during development (215), neurodegenerative disorders (216), stroke (217222), aging (223), TBI (32, 224), vascular disease (217222), depression (208, 225), and metabolic disturbances (63, 211, 226, 227).

Structure and Biological Activity of Erythropoietin

Currently, erythropoiesis-stimulating agents (ESAs), which include EPO, are approved for the treatment of anemia that results from chronic kidney failure, human immunodeficiency virus, and chemotherapy. In addition, EPO can be administered to reduce blood transfusions for surgery (228, 229). The EPO gene is located on chromosome 7 and is a single copy in a 5.4 kb region of the genomic DNA. The gene encodes for a polypeptide chain protein that has initially 193 amino acids (230). EPO is then processed and cleaved of a 27 amino acid hydrophobic secretory leader at the amino-terminal to result in a 166 amino acid peptide protein (231). With the removal of a carboxy-terminal arginine166 in the mature human and recombinant human EPO (rhEPO), a protein of 165 amino acids with a molecular weight of 30.4 kDa is subsequently generated (226, 227, 232, 233).

EPO has four glycosylated chains that include three N-linked and one O-linked acidic oligosaccharide side chains (71). The N-linked glycosylation sites exist at aspartyl24, aspartyl38, and aspartyl83. The O-linked glycosylation site occurs at serine126 (209). It is postulated that the N- and O-linked chains are for the production and secretion of the mature EPO (234). In addition, the carbohydrates are needed for the clearance of EPO, since EPO molecules with high sialic acid content can be easily cleared by the body through the liver (235).

In regards to the biological activity of EPO, the glycosylated chains protect EPO from free radical oxygen degradation (71). The oligosaccharides in EPO can offer protection from free radical activity as well (236) and the carbohydrate chains can stabilize the EPO protein (237). The disulfide bonds determine EPO activity since reduction of the two disulfide bonds formed between cysteine7 and cysteine160 and between cysteine29 and cysteine33 leads to functional loss of EPO. Alkylation of the sulfhydryl groups results in irreversible loss of the activity of EPO. Re-oxidization of EPO after reduction by guanidine restores almost 85 percent of the biological activity of EPO (238).

Expression and Production of Erythropoietin

EPO is present in the brain, uterus, and liver (239243), but the primary site for the production and secretion of EPO is the kidney peritubular interstitial cells (243). Production of EPO and the EPO receptor (EPOR) are changed during development (233). EPO production and EPOR expression in gestation are increased, but later EPO and EPOR are reduced following birth with EPO regulated by the tissue oxygen supply.

EPO expression is controlled by changes in oxygen tension and not by the concentration of red blood cells (50, 239, 244). Hypoxia-inducible factor 1 (HIF-1) regulates the expression of EPO and EPOR to increase the production of EPO as required (230, 239, 245, 246). After HIF-1 activation, gene transcription of EPO and EPOR occurs and is controlled through the transcription enhancer region in the 3′-flanking region of the EPO gene that binds to HIF-1 (230, 233).

Interestingly, EPO production can be controlled through other mechanisms not directly tied to hypoxia (247). For example, agents that limit inflammation in cerebral microglia have been demonstrated to lead to the release of EPO (248). Infections. such as malaria, also can result in significant serum levels of EPO (249). EPO serum concentrations are elevated during xenon anesthesia in cardiac surgery (250). Cytokines, including insulin-like growth factor, tumor necrosis factor-α (TNF-α) (251), interleukin-1β (IL-1β), and interleukin-6 (IL-6) can raise EPO and EPOR expressions (230, 242, 252). Other factors, such as cadmium exposure, intracellular calcium, and neuronal depolarizations, can alter EPO expression (71, 217, 240).

Cellular protection with Erythropoietin and mTOR

Several cell survival pathways that EPO oversees are intimately tied to mTOR signaling (205207, 209, 247, 253, 254). For stem cell development and the maturation of cells, EPO relies upon mTOR for the differentiation of neural precursor cells to achieve a neuronal phenotype (254). EPO also has been shown to promote adult hippocampal neurogenesis that is considered to be important for effective antidepressant treatment and requires mTOR activity to resolve affective disorders (225). EPO also plays a role in osteoblastogenesis and osteoclastogenesis that is dependent upon pathways of mTOR (255).

In regards to cellular protection, retinal progenitor cells are protected from oxidative stress by EPO and mTOR pathways (205). EPO requires mTOR activation for prevention of cognitive loss in models of sepsis-induced encephalopathy (256). EPO oversees mTOR and down-stream signaling pathways that involve PRAS40 to increase neuronal survival during oxygen-glucose deprivation (95). EPO also increases mTOR activity during hypoxia-reoxygenation stress to protect hippocampus-derived neuronal cells (204).

As part of the mTOR pathway, AMPK is closely associated with the protective capacity of EPO. EPO controls inflammation in the nervous system through AMPK (248). Yet, it is necessary for EPO to control a specific level of AMPK and mTOR activity to protect cells under some conditions of oxidative stress (117). In addition, EPO can rely upon AMPK pathways for anti-oxidant gene expression (122).

In regards to cellular protection with EPO and mTOR during programmed cell death, EPO blocks apoptotic cell death through the activation of mTOR (46, 57, 205207, 257). EPO can block Aβ toxicity through Wnt signaling and mTOR pathways to prevent caspase activation and apoptosis (97). EPO also fosters microglial survival during oxidative stress through mTOR (206). Given that Wnt signaling and WISP1 are also involved in metabolic pathways (60, 244, 258261), EPO also provides cellular protection during metabolic disorders through Wnt signaling (262265).

In relation to autophagy, EPO inhibits autophagy through the activation of mTOR (20, 57, 266). Activation of mTOR blocks autophagy by phosphorylating autophagic related genes (Atg) and proteins that include Atg13 and ULKs to inhibit the UNC like kinase complex ULK-Atg13-FIP200 (37). EPO can promote cellular protection during hypoxia and oxidative stress in retinal progenitor cells by limiting the induction of autophagy (205). EPO can prevent excessive autophagy that precedes apoptosis during experimental neonatal necrotizing enterocolitis (207). EPO also can modify the activity of autophagy and limit neonatal brain damage in the developing rodent during hyperoxia exposure and oxygen toxicity (267). In some cases, EPO also relies upon the beneficial effects of autophagy. In neuronal cell line models, EPO can suppress apoptotic cell injury through the increased activity of AMPK and increasing autophagy-related signaling pathways (257). In addition, the concentration of EPO may affect mTOR signaling and apoptotic cell death. Elevated concentrations of EPO can lead to cellular damage and lessen the activity of mTOR (253).

Future Perspectives

The interest generated with EPO has led to the initiation of multiple clinical trials for EPO. Yet, the role mTOR may play with EPO in treating clinical disease requires further investigation. Currently, EPO appears to have efficacy in treating conditions that involve the developing brain. Elevated EPO concentrations during infant maturation have been correlated with increased Mental Development Index scores (268). In a randomized, double-blind placebo-controlled study involving preterm infants, EPO was demonstrated to improve white matter development assessed by diffusion tensor imaging and tract-based spatial statistics (269). High-dose erythropoietin treatment within forty-two hours after birth in preterm infants has been associated with reduced risk of brain injury documented through magnetic resonance imaging (215, 270). Hypothermia combined with high-dose erythropoietin also appears to be protective against hypoxic-ischemic encephalopathy with improved one-year motor function (271).

In mature populations that are affected by aging-related disorders, the conclusion for the use of EPO to treat clinical disease is less clear. In a small study with twenty-six Parkinson’s disease patients, recombinant EPO administration improved cardiovascular autonomic dysfunction and cognition, but did not alter motor function (216). Increased expression of the EPO receptor in temporal cortical and hippocampal astrocytes in sporadic AD patients has been observed and considered to be an early neuroprotective pathway (272). The biosimilar epoetin α (Binocrit) administered in elderly patients with myelodysplastic syndromes also have experienced improved cognitive function that may be related to resolution of anemia (273). Yet, in relation to TBI, neither the administration of EPO or maintaining hemoglobin concentration above 10 g/dL led to improvement in neurological outcome at six months (274). In a large clinical trial with close to six hundred patients that experienced brain injury, EPO did not significantly affect six-month mortality, reduce severe neurological dysfunction, or increase the occurrence of deep venous thrombosis of the lower limbs (275). In addition, administration of human choriogonadotropin alfa followed by EPO did not show improvement in neurological recovery in patients with ischemic stroke (276). Similar to prior studies with cardiovascular disease (229, 250, 277279), additional work suggests that high concentrations of EPO may not be effective for cardiac protection. Recent studies with out-of-hospital cardiac arrest demonstrate that EPO did not confer a benefit and was associated with a higher thrombotic complication rate (280, 281).

It is possible that lower doses of EPO or different times of administration may be more effective for clinical disease especially since new work suggests a role for EPO and EPOR autoantibodies for disease progression (282, 283). Additional studies suggest that low concentrations of EPO may be beneficial to the cardiovascular system (284286) and also may benefit neurological function. However, only modulating the concentration or timing of administration of EPO may not offer the greatest gains to translate experimental in vitro and in vivo studies with EPO and mTOR into clinical success. Targeting the ability of EPO to govern specific cellular pathways such as mTOR signaling pathways would increase our understanding substantially and potentially yield new treatment modalities for multiple clinical disorders (Table 1). mTOR oversees critical pathways of programmed cell death that involve apoptosis and autophagy that can markedly impact clinical disease and aging-related disorders. For example, increased mTOR signaling may be required to regulate the β-site amyloid precursor protein (APP)-cleaving enzyme 1 (β-secretase, BACE1) that promotes Aβ accumulation in AD. Elevated mTORC1 activity reduces BACE1 and limits Aβ generation (145). However, additional studies suggest that some degree of inhibition of mTOR may be necessary to enhance Aβ clearance and improve spatial learning through the activation of autophagy (287). Greater emphasis upon the regulatory pathways controlling EPO and mTOR signaling could offer significant fruits for targeting several clinical disorders.

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 conflict of interest.

References

  • 1.Maiese K. Novel nervous and multi-system regenerative therapeutic strategies for diabetes mellitus with mTOR. Neural regeneration research. 2016;11(3):372–85. doi: 10.4103/1673-5374.179032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Kudryavtseva AV, Krasnov GS, Dmitriev AA, Alekseev BY, Kardymon OL, Sadritdinova AF, et al. Mitochondrial dysfunction and oxidative stress in aging and cancer. Oncotarget. 2016 doi: 10.18632/oncotarget.9821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Minino AM. Death in the United States, 2011. NCHS data brief. 2013;(115):1–8. [PubMed] [Google Scholar]
  • 4.Organization WH. Description of the global burden of NCDs, their risk factors and determinants. Global status report on noncommunicable diseases 2010. 2011 Apr;:1–176. [Google Scholar]
  • 5.Okada M, Kim HW, Matsu-Ura K, Wang YG, Xu M, Ashraf M. Abrogation of Age-Induced MicroRNA-195 Rejuvenates the Senescent Mesenchymal Stem Cells by Reactivating Telomerase. Stem Cells. 2015 doi: 10.1002/stem.2211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Walters HE, Deneka-Hannemann S, Cox LS. Reversal of phenotypes of cellular senescence by pan-mTOR inhibition. Aging (Albany NY) 2016 doi: 10.18632/aging.100872. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Lan AP, Chen J, Chai ZF, Hu Y. The neurotoxicity of iron, copper and cobalt in Parkinson’s disease through ROS-mediated mechanisms. Biometals : an international journal on the role of metal ions in biology, biochemistry, and medicine. 2016 doi: 10.1007/s10534-016-9942-4. [DOI] [PubMed] [Google Scholar]
  • 8.Lawler JM, Rodriguez DA, Hord JM. Mitochondria in the middle: Exercise preconditioning protection of striated muscle. J Physiol. 2016 doi: 10.1113/JP270656. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Maiese K. Picking a bone with WISP1 (CCN4): new strategies against degenerative joint disease. J Transl Sci. 2016;1(3):83–5. [PMC free article] [PubMed] [Google Scholar]
  • 10.Marrif HI, Al-Sunousi SI. Pancreatic beta Cell Mass Death. Frontiers in pharmacology. 2016;7:83. doi: 10.3389/fphar.2016.00083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Maulucci G, Daniel B, Cohen O, Avrahami Y, Sasson S. Hormetic and regulatory effects of lipid peroxidation mediators in pancreatic beta cells. Mol Aspects Med. 2016 doi: 10.1016/j.mam.2016.03.001. [DOI] [PubMed] [Google Scholar]
  • 12.Curjuric I, Imboden M, Bridevaux PO, Gerbase MW, Haun M, Keidel D, et al. Common SIRT1 variants modify the effect of abdominal adipose tissue on aging-related lung function decline. Age (Dordr) 2016;38(3):52. doi: 10.1007/s11357-016-9917-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Favero G, Franceschetti L, Rodella LF, Rezzani R. Sirtuins, aging, and cardiovascular risks. Age (Dordr) 2015;37(4):9804. doi: 10.1007/s11357-015-9804-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.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]
  • 15.Gabay O, Clouse KA. Epigenetics of cartilage diseases. Joint, bone, spine : revue du rhumatisme. 2015 doi: 10.1016/j.jbspin.2015.10.004. [DOI] [PubMed] [Google Scholar]
  • 16.Palmer KT, Goodson N. Ageing, musculoskeletal health and work. Best practice & research Clinical rheumatology. 2015;29(3):391–404. doi: 10.1016/j.berh.2015.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Ignacio ZM, Reus GZ, Arent CO, Abelaira HM, Pitcher MR, Quevedo J. New perspectives on the involvement of mTOR in depression as well as in the action of antidepressant drugs. Br J Clin Pharmacol. 2015 doi: 10.1111/bcp.12845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Bondy S, Maiese K. Aging and Age-Related Disorders. Springer Science; 2010. [Google Scholar]
  • 19.Choi HS, Ahn JH, Park JH, Won MH, Lee CH. Age-dependent changes in the protein expression levels of Redd1 and mTOR in the gerbil hippocampus during normal aging. Molecular medicine reports. 2016;13(3):2409–14. doi: 10.3892/mmr.2016.4835. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Dong W, Wang R, Ma LN, Xu BL, Zhang JS, Zhao ZW, et al. Influence of age-related learning and memory capacity of mice: different effects of a high and low caloric diet. Aging Clin Exp Res. 2016;28(2):303–11. doi: 10.1007/s40520-015-0398-0. [DOI] [PubMed] [Google Scholar]
  • 21.Bellozi PM, Lima IV, Doria JG, Vieira EL, Campos AC, Candelario-Jalil E, et al. Neuroprotective effects of the anticancer drug NVP-BEZ235 (dactolisib) on amyloid-beta 1-42 induced neurotoxicity and memory impairment. Scientific reports. 2016;6:25226. doi: 10.1038/srep25226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Chong ZZ, Li F, Maiese K. Oxidative stress in the brain: Novel cellular targets that govern survival during neurodegenerative disease. Prog Neurobiol. 2005;75(3):207–46. doi: 10.1016/j.pneurobio.2005.02.004. [DOI] [PubMed] [Google Scholar]
  • 23.Guo P, Wang D, Wang X, Feng H, Tang Y, Sun R, et al. Effect and mechanism of fuzhisan and donepezil on the sirtuin 1 pathway and amyloid precursor protein metabolism in PC12 cells. Molecular medicine reports. 2016;13(4):3539–46. doi: 10.3892/mmr.2016.4957. [DOI] [PubMed] [Google Scholar]
  • 24.Maiese K, Chong ZZ, Hou J, Shang YC. New strategies for Alzheimer’s disease and cognitive impairment. Oxid Med Cell Longev. 2009;2(5):279–89. doi: 10.4161/oxim.2.5.9990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Zhao H, Wang ZC, Wang KF, Chen XY. Abeta peptide secretion is reduced by Radix Polygalaeinduced autophagy via activation of the AMPK/mTOR pathway. Molecular medicine reports. 2015;12(2):2771–6. doi: 10.3892/mmr.2015.3781. [DOI] [PubMed] [Google Scholar]
  • 26.Chen LN, Sun J, Yang XD, Xiao K, Lv Y, Zhang BY, et al. The Brain NO Levels and NOS Activities Ascended in the Early and Middle Stages and Descended in the Terminal Stage in Scrapie-Infected Animal Models. Mol Neurobiol. 2016 doi: 10.1007/s12035-016-9755-z. [DOI] [PubMed] [Google Scholar]
  • 27.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;24(3):283–307. doi: 10.1517/13543784.2015.974804. [DOI] [PubMed] [Google Scholar]
  • 28.Maiese K. New Insights for Oxidative Stress and Diabetes Mellitus. Oxid Med Cell Longev. 2015 doi: 10.1155/2015/875961. 2015(2015:875961) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Maiese K. Cutting through the Complexities of mTOR for the Treatment of Stroke. Curr Neurovasc Res. 2014;11(2):177–86. doi: 10.2174/1567202611666140408104831. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.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 doi: 10.1007/s12035-014-9029-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.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 doi: 10.1111/jnc.13082. [DOI] [PubMed] [Google Scholar]
  • 32.Maiese K. Charting a course for erythropoietin in traumatic brain injury. J Transl Sci. 2016;2(2):140–4. doi: 10.15761/jts.1000131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Lee J, Gu X, Wei L, Wei Z, Dix TA, Yu SP. Therapeutic Effects of Pharmacologically induced Hypothermia against Traumatic Brain Injury in Mice. J Neurotrauma. 2014 doi: 10.1089/neu.2013.3251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Maiese K. The Merck Manual. 19. 2011. Coma and impaired consciousness. Professional Edition. [Google Scholar]
  • 35.Wang JW, Wang HD, Cong ZX, Zhou XM, Xu JG, Jia Y, et al. Puerarin ameliorates oxidative stress in a rodent model of traumatic brain injury. J Surg Res. 2014;186(1):328–37. doi: 10.1016/j.jss.2013.08.027. [DOI] [PubMed] [Google Scholar]
  • 36.Kumar PR, Essa MM, Al-Adawi S, Dradekh G, Memon MA, Akbar M, et al. Omega-3 Fatty acids could alleviate the risks of traumatic brain injury - a mini review. Journal of traditional and complementary medicine. 2014;4(2):89–92. doi: 10.4103/2225-4110.130374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Maiese K. Taking aim at Alzheimer’s disease through the mammalian target of rapamycin. Ann Med. 2014;46(8):587–96. doi: 10.3109/07853890.2014.941921. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Maiese K. Forkhead transcription factors: new considerations for alzheimer’s disease and dementia. J Transl Sci. 2016;2(4):241–7. doi: 10.15761/JTS.1000146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Agis-Torres A, Solhuber M, Fernandez M, Sanchez-Montero JM. Multi-Target-Directed Ligands and other Therapeutic Strategies in the Search of a Real Solution for Alzheimer’s Disease. Current neuropharmacology. 2014;12(1):2–36. doi: 10.2174/1570159X113116660047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Maiese K. Targeting molecules to medicine with mTOR, autophagy, and neurodegenerative disorders. Br J Clin Pharmacol. 2015 doi: 10.1111/bcp.12804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Abuhammad A, Taha MO. QSAR studies in the discovery of novel type-II diabetic therapies. Expert opinion on drug discovery. 2016;11(2):197–214. doi: 10.1517/17460441.2016.1118046. [DOI] [PubMed] [Google Scholar]
  • 42.Wahl D, Cogger VC, Solon-Biet SM, Waern RV, Gokarn R, Pulpitel T, et al. Nutritional strategies to optimise cognitive function in the aging brain. Ageing research reviews. 2016 doi: 10.1016/j.arr.2016.06.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Prattichizzo F, De Nigris V, La Sala L, Procopio AD, Olivieri F, Ceriello A. “Inflammaging” as a Druggable Target: A Senescence-Associated Secretory Phenotype-Centered View of Type 2 Diabetes. Oxid Med Cell Longev. 2016;2016:1810327. doi: 10.1155/2016/1810327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.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 doi: 10.1152/ajpregu.00262.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Maiese K, Chong ZZ, Shang YC, Hou J. Novel Avenues of Drug Discovery and Biomarkers for Diabetes Mellitus. Journal of clinical pharmacology. 2011;51(2):128–52. doi: 10.1177/0091270010362904. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Maiese K, Chong ZZ, Shang YC, Wang S. Novel directions for diabetes mellitus drug discovery. Expert opinion on drug discovery. 2013;8(1):35–48. doi: 10.1517/17460441.2013.736485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Rutter MK, Massaro JM, Hoffmann U, O’Donnell CJ, Fox CS. Fasting Glucose, Obesity, and Coronary Artery Calcification in Community-Based People Without Diabetes. Diabetes Care. 2012 doi: 10.2337/dc11-1950. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Xu E, Schwab M, Marette A. Role of protein tyrosine phosphatases in the modulation of insulin signaling and their implication in the pathogenesis of obesity-linked insulin resistance. Rev Endocr Metab Disord. 2014;15(1):79–97. doi: 10.1007/s11154-013-9282-4. [DOI] [PubMed] [Google Scholar]
  • 49.Harris MI, Eastman RC. Early detection of undiagnosed diabetes mellitus: a US perspective. Diabetes Metab Res Rev. 2000;16(4):230–6. doi: 10.1002/1520-7560(2000)9999:9999<::aid-dmrr122>3.0.co;2-w. [DOI] [PubMed] [Google Scholar]
  • 50.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]
  • 51.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]
  • 52.Centers for Medicare and Medicaid Services. National Health Expenditure Projections 2012–2022. 2013 wwwcmsgov.
  • 53.Folwarczna J, Janas A, Pytlik M, Cegiela U, Sliwinski L, Krivosikova Z, et al. Effects of Trigonelline, an Alkaloid Present in Coffee, on Diabetes-Induced Disorders in the Rat Skeletal System. Nutrients. 2016;8(3) doi: 10.3390/nu8030133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Gurlo T, Rivera JF, Butler AE, Cory M, Hoang J, Costes S, et al. CHOP Contributes to, But Is Not the Only Mediator of, IAPP Induced beta-Cell Apoptosis. Mol Endocrinol. 2016;30(4):446–54. doi: 10.1210/me.2015-1255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Kumar P, Swain MM, Pal A. Hyperglycemia-induced inflammation caused down-regulation of 8-oxoG-DNA glycosylase levels in murine macrophages is mediated by oxidative-nitrosative stress-dependent pathways. Int J Biochem Cell Biol. 2016;73:82–98. doi: 10.1016/j.biocel.2016.02.006. [DOI] [PubMed] [Google Scholar]
  • 56.Ma L, Fu R, Duan Z, Lu J, Gao J, Tian L, et al. Sirt1 is essential for resveratrol enhancement of hypoxia-induced autophagy in the type 2 diabetic nephropathy rat. Pathology, research and practice. 2016 doi: 10.1016/j.prp.2016.02.001. [DOI] [PubMed] [Google Scholar]
  • 57.Maiese K. mTOR: Driving apoptosis and autophagy for neurocardiac complications of diabetes mellitus. World J Diabetes. 2015;6(2):217–24. doi: 10.4239/wjd.v6.i2.217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.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]
  • 59.Motawi TK, Darwish HA, Hamed MA, El-Rigal NS, Naser AF. A Therapeutic Insight of Niacin and Coenzyme Q10 Against Diabetic Encephalopathy in Rats. Mol Neurobiol. 2016 doi: 10.1007/s12035-016-9765-x. [DOI] [PubMed] [Google Scholar]
  • 60.Sahin Ersoy G, Altun Ensari T, Subas S, Giray B, Simsek EE, Cevik O. WISP1 is a novel adipokine linked to metabolic parameters in gestational diabetes mellitus. J Matern Fetal Neonatal Med. 2016:1–5. doi: 10.1080/14767058.2016.1192118. [DOI] [PubMed] [Google Scholar]
  • 61.Di Rosa M, Malaguarnera L. Chitotriosidase: A New Inflammatory Marker in Diabetic Complications. Pathobiology. 2016;83(4):211–9. doi: 10.1159/000443932. [DOI] [PubMed] [Google Scholar]
  • 62.Maiese K. SIRT1 and stem cells: In the forefront with cardiovascular disease, neurodegeneration and cancer. World J Stem Cells. 2015;7(2):235–42. doi: 10.4252/wjsc.v7.i2.235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Maiese K. Erythropoietin and diabetes mellitus. World J Diabetes. 2015;6(14):1259–73. doi: 10.4239/wjd.v6.i14.1259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Pektas MB, Sadi G, Koca HB, Yuksel Y, Vurmaz A, Koca T, et al. Resveratrol Ameliorates the Components of Hepatic Inflammation and Apoptosis in a Rat Model of Streptozotocin-Induced Diabetes. Drug development research. 2016 doi: 10.1002/ddr.21287. [DOI] [PubMed] [Google Scholar]
  • 65.Chong ZZ, Maiese K. Mammalian Target of Rapamycin Signaling in Diabetic Cardiovascular Disease. Cardiovasc Diabetol. 2012;11(1):45. doi: 10.1186/1475-2840-11-45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Tang PC, Ng YF, Ho S, Gyda M, Chan SW. Resveratrol and cardiovascular health--promising therapeutic or hopeless illusion? Pharmacol Res. 2014;90:88–115. doi: 10.1016/j.phrs.2014.08.001. [DOI] [PubMed] [Google Scholar]
  • 67.Xu YJ, Tappia PS, Neki NS, Dhalla NS. Prevention of diabetes-induced cardiovascular complications upon treatment with antioxidants. Heart failure reviews. 2014;19(1):113–21. doi: 10.1007/s10741-013-9379-6. [DOI] [PubMed] [Google Scholar]
  • 68.Yu L, Liang H, Dong X, Zhao G, Jin Z, Zhai M, et al. Reduced silent information regulator 1 signaling exacerbates myocardial ischemia-reperfusion injury in type 2 diabetic rats and the protective effect of melatonin. J Pineal Res. 2015;59(3):376–90. doi: 10.1111/jpi.12269. [DOI] [PubMed] [Google Scholar]
  • 69.Alexandru N, Popov D, Georgescu A. Platelet dysfunction in vascular pathologies and how can it be treated. Thromb Res. 2012;129(2):116–26. doi: 10.1016/j.thromres.2011.09.026. [DOI] [PubMed] [Google Scholar]
  • 70.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;171(13):3146–57. doi: 10.1111/bph.12655. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Maiese K, Chong ZZ, Hou J, Shang YC. Erythropoietin and oxidative stress. Curr Neurovasc Res. 2008;5(2):125–42. doi: 10.2174/156720208784310231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.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]
  • 73.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]
  • 74.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;18(10):1141–53. doi: 10.1089/ars.2012.4583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Wang P, Xing Y, Chen C, Chen Z, Qian Z. Advanced glycation end-product (AGE) induces apoptosis in human retinal ARPE-19 cells via promoting mitochondrial dysfunction and activating the Fas-FasL signaling. Biosci Biotechnol Biochem. 2015:1–7. doi: 10.1080/09168451.2015.1095065. [DOI] [PubMed] [Google Scholar]
  • 76.Deshmukh R, Kaundal M, Bansal V, Samardeep Caffeic acid attenuates oxidative stress, learning and memory deficit in intra-cerebroventricular streptozotocin induced experimental dementia in rats. Biomed Pharmacother. 2016;81:56–62. doi: 10.1016/j.biopha.2016.03.017. [DOI] [PubMed] [Google Scholar]
  • 77.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 doi: 10.1096/fj.14-262048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.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]
  • 79.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]
  • 80.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;531(2):176–81. doi: 10.1016/j.neulet.2012.10.045. [DOI] [PubMed] [Google Scholar]
  • 81.Reagan LP. Diabetes as a chronic metabolic stressor: causes, consequences and clinical complications. Exp Neurol. 2012;233(1):68–78. doi: 10.1016/j.expneurol.2011.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Berry M, Ahmed Z, Morgan-Warren P, Fulton D, Logan A. Prospects for mTOR-mediated functional repair after central nervous system trauma. Neurobiol Dis. 2016;85:99–110. doi: 10.1016/j.nbd.2015.10.002. [DOI] [PubMed] [Google Scholar]
  • 83.Citraro R, Leo A, Constanti A, Russo E, De Sarro G. mTOR pathway inhibition as a new therapeutic strategy in epilepsy and epileptogenesis. Pharmacol Res. 2016;107:333–43. doi: 10.1016/j.phrs.2016.03.039. [DOI] [PubMed] [Google Scholar]
  • 84.Crino PB. The mTOR signalling cascade: paving new roads to cure neurological disease. Nature reviews Neurology. 2016 doi: 10.1038/nrneurol.2016.81. [DOI] [PubMed] [Google Scholar]
  • 85.Lee HJ, Koh SH, Song KM, Seol IJ, Park HK. The Akt/mTOR/p70S6K Pathway Is Involved in the Neuroprotective Effect of Erythropoietin on Hypoxic/Ischemic Brain Injury in a Neonatal Rat Model. Neonatology. 2016;110(2):93–100. doi: 10.1159/000444360. [DOI] [PubMed] [Google Scholar]
  • 86.Maiese K. Molecules to Medicine with mTOR: Translating Critical Pathways into Novel Therapeutic Strategies. Elsevier and Academic Press; 2016. [Google Scholar]
  • 87.Chong ZZ, Shang YC, Wang S, Maiese K. Shedding new light on neurodegenerative diseases through the mammalian target of rapamycin. Prog Neurobiol. 2012;99(2):128–48. doi: 10.1016/j.pneurobio.2012.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.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]
  • 89.Johnson SC, Sangesland M, Kaeberlein M, Rabinovitch PS. Modulating mTOR in aging and health. Interdisciplinary topics in gerontology. 2015;40:107–27. doi: 10.1159/000364974. [DOI] [PubMed] [Google Scholar]
  • 90.Maiese K, Chong ZZ, Shang YC, Wang S. mTOR: on target for novel therapeutic strategies in the nervous system. Trends Mol Med. 2013;19(1):51–60. doi: 10.1016/j.molmed.2012.11.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Gulhati P, Bowen KA, Liu J, Stevens PD, Rychahou PG, Chen M, et al. mTORC1 and mTORC2 regulate EMT, motility, and metastasis of colorectal cancer via RhoA and Rac1 signaling pathways. Cancer Res. 2011;71(9):3246–56. doi: 10.1158/0008-5472.CAN-10-4058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Zoncu R, Efeyan A, Sabatini DM. mTOR: from growth signal integration to cancer, diabetes and ageing. Nat Rev Mol Cell Biol. 2011;12(1):21–35. doi: 10.1038/nrm3025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Maiese K. Driving neural regeneration through the mammalian target of rapamycin. Neural regeneration research. 2014;9(15):1413–7. doi: 10.4103/1673-5374.139453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Malla R, Ashby CR, Jr, Narayanan NK, Narayanan B, Faridi JS, Tiwari AK. Proline-rich AKT substrate of 40-kDa (PRAS40) in the pathophysiology of cancer. Biochem Biophys Res Commun. 2015;463(3):161–6. doi: 10.1016/j.bbrc.2015.05.041. [DOI] [PubMed] [Google Scholar]
  • 95.Chong ZZ, Shang YC, Wang S, Maiese K. PRAS40 Is an Integral Regulatory Component of Erythropoietin mTOR Signaling and Cytoprotection. PLoS ONE. 2012;7(9):e45456. doi: 10.1371/journal.pone.0045456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Fonseca BD, Smith EM, Lee VH, MacKintosh C, Proud CG. PRAS40 is a target for mammalian target of rapamycin complex 1 and is required for signaling downstream of this complex. J Biol Chem. 2007;282(34):24514–24. doi: 10.1074/jbc.M704406200. [DOI] [PubMed] [Google Scholar]
  • 97.Shang YC, Chong ZZ, Wang S, Maiese K. WNT1 Inducible Signaling Pathway Protein 1 (WISP1) Targets PRAS40 to Govern beta-Amyloid Apoptotic Injury of Microglia. Curr Neurovasc Res. 2012;9(4):239–49. doi: 10.2174/156720212803530618. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Wang H, Zhang Q, Wen Q, Zheng Y, Philip L, Jiang H, et al. Proline-rich Akt substrate of 40kDa (PRAS40): a novel downstream target of PI3k/Akt signaling pathway. Cell Signal. 2012;24(1):17–24. doi: 10.1016/j.cellsig.2011.08.010. [DOI] [PubMed] [Google Scholar]
  • 99.Xiong X, Xie R, Zhang H, Gu L, Xie W, Cheng M, et al. PRAS40 plays a pivotal role in protecting against stroke by linking the Akt and mTOR pathways. Neurobiol Dis. 2014;66:43–52. doi: 10.1016/j.nbd.2014.02.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Cai Z, Yan LJ. Rapamycin, Autophagy, and Alzheimer’s Disease. Journal of biochemical and pharmacological research. 2013;1(2):84–90. [PMC free article] [PubMed] [Google Scholar]
  • 101.Garcia-Martinez JM, Alessi DR. mTOR complex 2 (mTORC2) controls hydrophobic motif phosphorylation and activation of serum- and glucocorticoid-induced protein kinase 1 (SGK1) Biochem J. 2008;416(3):375–85. doi: 10.1042/BJ20081668. [DOI] [PubMed] [Google Scholar]
  • 102.Pearce LR, Sommer EM, Sakamoto K, Wullschleger S, Alessi DR. Protor-1 is required for efficient mTORC2-mediated activation of SGK1 in the kidney. Biochem J. 2011;436(1):169–79. doi: 10.1042/BJ20102103. [DOI] [PubMed] [Google Scholar]
  • 103.Sarbassov DD, Guertin DA, Ali SM, Sabatini DM. Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex. Science. 2005;307(5712):1098–101. doi: 10.1126/science.1106148. [DOI] [PubMed] [Google Scholar]
  • 104.Frias MA, Thoreen CC, Jaffe JD, Schroder W, Sculley T, Carr SA, et al. mSin1 is necessary for Akt/PKB phosphorylation, and its isoforms define three distinct mTORC2s. Curr Biol. 2006;16(18):1865–70. doi: 10.1016/j.cub.2006.08.001. [DOI] [PubMed] [Google Scholar]
  • 105.Duan P, Hu C, Quan C, Yu T, Zhou W, Yuan M, et al. 4-Nonylphenol induces apoptosis, autophagy and necrosis in Sertoli cells: Involvement of ROS-mediated AMPK/AKT-mTOR and JNK pathways. Toxicology. 2016 doi: 10.1016/j.tox.2016.01.004. [DOI] [PubMed] [Google Scholar]
  • 106.Hung CH, Chan SH, Chu PM, Lin HC, Tsai KL. Metformin regulates oxLDL-facilitated endothelial dysfunction by modulation of SIRT1 through repressing LOX-1-modulated oxidative signaling. Oncotarget. 2016 doi: 10.18632/oncotarget.7387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Jin D, Zhao T, Feng WW, Mao GH, Zou Y, Wang W, et al. Schisandra polysaccharide increased glucose consumption by up-regulating the expression of GLUT-4. International journal of biological macromolecules. 2016;87:555–62. doi: 10.1016/j.ijbiomac.2016.03.028. [DOI] [PubMed] [Google Scholar]
  • 108.Xie Z, Lau K, Eby B, Lozano P, He C, Pennington B, et al. Improvement of cardiac functions by chronic metformin treatment is associated with enhanced cardiac autophagy in diabetic OVE26 mice. Diabetes. 2011;60(6):1770–8. doi: 10.2337/db10-0351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Arunachalam G, Samuel SM, Marei I, Ding H, Triggle CR. Metformin modulates hyperglycaemia-induced endothelial senescence and apoptosis through SIRT1. Br J Pharmacol. 2014;171(2):523–35. doi: 10.1111/bph.12496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Hung CH, Chan SH, Chu PM, Tsai KL. Quercetin is a potent anti-atherosclerotic compound by activation of SIRT1 signaling under oxLDL stimulation. Molecular nutrition & food research. 2015 doi: 10.1002/mnfr.201500144. [DOI] [PubMed] [Google Scholar]
  • 111.Kai Y, Kawano Y, Yamamoto H, Narahara H. A possible role for AMP-activated protein kinase activated by metformin and AICAR in human granulosa cells. Reproductive biology and endocrinology : RB&E. 2015;13(1):27. doi: 10.1186/s12958-015-0023-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Kurdi A, De Meyer GR, Martinet W. Potential therapeutic effects of mTOR inhibition in atherosclerosis. Br J Clin Pharmacol. 2015 doi: 10.1111/bcp.12820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Zhao Y, Sun Y, Ding Y, Wang X, Zhou Y, Li W, et al. GL-V9, a new synthetic flavonoid derivative, ameliorates DSS-induced colitis against oxidative stress by up-regulating Trx-1 expression via activation of AMPK/FOXO3a pathway. Oncotarget. 2015;6(28):26291–307. doi: 10.18632/oncotarget.4657. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.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 doi: 10.3233/JAD-140564. [DOI] [PubMed] [Google Scholar]
  • 115.Jin X, Chen M, Yi L, Chang H, Zhang T, Wang L, et al. Delphinidin-3-glucoside protects human umbilical vein endothelial cells against oxidized low-density lipoprotein-induced injury by autophagy upregulation via the AMPK/SIRT1 signaling pathway. Molecular nutrition & food research. 2014;58(10):1941–51. doi: 10.1002/mnfr.201400161. [DOI] [PubMed] [Google Scholar]
  • 116.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;107(1–2):42–9. doi: 10.1016/j.lfs.2014.04.034. [DOI] [PubMed] [Google Scholar]
  • 117.Shang YC, Chong ZZ, Wang S, Maiese K. Tuberous sclerosis protein 2 (TSC2) modulates CCN4 cytoprotection during apoptotic amyloid toxicity in microglia. Curr Neurovasc Res. 2013;10(1):29–38. doi: 10.2174/156720213804806007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Russo E, Andreozzi F, Iuliano R, Dattilo V, Procopio T, Fiume G, et al. Early molecular and behavioral response to lipopolysaccharide in the WAG/Rij rat model of absence epilepsy and depressive-like behavior, involves interplay between AMPK, AKT/mTOR pathways and neuroinflammatory cytokine release. Brain Behav Immun. 2014;42:157–68. doi: 10.1016/j.bbi.2014.06.016. [DOI] [PubMed] [Google Scholar]
  • 119.Sato T, Nakashima A, Guo L, Tamanoi F. Specific activation of mTORC1 by Rheb G-protein in vitro involves enhanced recruitment of its substrate protein. J Biol Chem. 2009;284(19):12783–91. doi: 10.1074/jbc.M809207200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Inoki K, Zhu T, Guan KL. TSC2 mediates cellular energy response to control cell growth and survival. Cell. 2003;115(5):577–90. doi: 10.1016/s0092-8674(03)00929-2. [DOI] [PubMed] [Google Scholar]
  • 121.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]
  • 122.Wang L, Di L, Noguchi CT. AMPK is involved in mediation of erythropoietin influence on metabolic activity and reactive oxygen species production in white adipocytes. Int J Biochem Cell Biol. 2014;54:1–9. doi: 10.1016/j.biocel.2014.06.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Chong ZZ, Shang YC, Wang S, Maiese K. SIRT1: New avenues of discovery for disorders of oxidative stress. Expert opinion on therapeutic targets. 2012;16(2):167–78. doi: 10.1517/14728222.2012.648926. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Wang Y, Liang Y, Vanhoutte PM. SIRT1 and AMPK in regulating mammalian senescence: a critical review and a working model. FEBS Lett. 2011;585(7):986–94. doi: 10.1016/j.febslet.2010.11.047. [DOI] [PubMed] [Google Scholar]
  • 125.Chong ZZ, Shang YC, Wang S, Maiese K. A Critical Kinase Cascade in Neurological Disorders: PI 3-K, Akt, and mTOR. Future Neurol. 2012;7(6):733–48. doi: 10.2217/fnl.12.72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Janku F, Wheler JJ, Westin SN, Moulder SL, Naing A, Tsimberidou AM, et al. PI3K/AKT/mTOR inhibitors in patients with breast and gynecologic malignancies harboring PIK3CA mutations. J Clin Oncol. 2012;30(8):777–82. doi: 10.1200/JCO.2011.36.1196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Maiese K. Therapeutic targets for cancer: current concepts with PI 3-K, Akt, & mTOR. The Indian journal of medical research. 2013;137(2):243–6. [PMC free article] [PubMed] [Google Scholar]
  • 128.Morgan-Warren PJ, Berry M, Ahmed Z, Scott RA, Logan A. Exploiting mTOR signaling: a novel translatable treatment strategy for traumatic optic neuropathy? Invest Ophthalmol Vis Sci. 2013;54(10):6903–16. doi: 10.1167/iovs.13-12803. [DOI] [PubMed] [Google Scholar]
  • 129.Huang J, Dibble CC, Matsuzaki M, Manning BD. The TSC1-TSC2 complex is required for proper activation of mTOR complex 2. Mol Cell Biol. 2008;28(12):4104–15. doi: 10.1128/MCB.00289-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Feehan RP, Shantz LM. Negative regulation of the FOXO3a transcription factor by mTORC2 induces a pro-survival response following exposure to ultraviolet-B irradiation. Cell Signal. 2016 doi: 10.1016/j.cellsig.2016.03.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Maiese K, Chong ZZ, Shang YC, Wang S. Targeting disease through novel pathways of apoptosis and autophagy. Expert opinion on therapeutic targets. 2012;16(12):1203–14. doi: 10.1517/14728222.2012.719499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Maiese K, Chong ZZ, Hou J, Shang YC. Oxidative stress: Biomarkers and novel therapeutic pathways. Exp Gerontol. 2010;45(3):217–34. doi: 10.1016/j.exger.2010.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Shang YC, Chong ZZ, Hou J, Maiese K. Wnt1, FoxO3a, and NF-kappaB oversee microglial integrity and activation during oxidant stress. Cell Signal. 2010;22(9):1317–29. doi: 10.1016/j.cellsig.2010.04.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Viola G, Bortolozzi R, Hamel E, Moro S, Brun P, Castagliuolo I, et al. MG-2477, a new tubulin inhibitor, induces autophagy through inhibition of the Akt/mTOR pathway and delayed apoptosis in A549 cells. Biochem Pharmacol. 2012;83(1):16–26. doi: 10.1016/j.bcp.2011.09.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Wong DZ, Kadir HA, Lee CL, Goh BH. Neuroprotective properties of Loranthus parasiticus aqueous fraction against oxidative stress-induced damage in NG108-15 cells. J Nat Med. 2012;66(3):544–51. doi: 10.1007/s11418-011-0622-y. [DOI] [PubMed] [Google Scholar]
  • 136.Bailey TJ, Fossum SL, Fimbel SM, Montgomery JE, Hyde DR. The inhibitor of phagocytosis, O-phospho-L-serine, suppresses Muller glia proliferation and cone cell regeneration in the light-damaged zebrafish retina. Exp Eye Res. 2010;91(5):601–12. doi: 10.1016/j.exer.2010.07.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.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;7(2):95–112. doi: 10.2174/156720210791184899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.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;6(4):223–38. doi: 10.2174/156720209789630302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.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;49(1):105–15. doi: 10.1007/s12031-012-9900-8. [DOI] [PubMed] [Google Scholar]
  • 140.Kim S, Kang IH, Nam JB, Cho Y, Chung DY, Kim SH, et al. Ameliorating the Effect of Astragaloside IV on Learning and Memory Deficit after Chronic Cerebral Hypoperfusion in Rats. Molecules. 2015;20(2):1904–21. doi: 10.3390/molecules20021904. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Xin YJ, Yuan B, Yu B, Wang YQ, Wu JJ, Zhou WH, et al. Tet1-mediated DNA demethylation regulates neuronal cell death induced by oxidative stress. Scientific reports. 2015;5:7645. doi: 10.1038/srep07645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Yu T, Li L, Chen T, Liu Z, Liu H, Li Z. Erythropoietin attenuates advanced glycation endproducts-induced toxicity of schwann cells in vitro. Neurochem Res. 2015;40(4):698–712. doi: 10.1007/s11064-015-1516-2. [DOI] [PubMed] [Google Scholar]
  • 143.Maiese K. FoxO Proteins in the Nervous System. Anal Cell Pathol (Amst) 2015;2015:569392. doi: 10.1155/2015/569392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.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;143(1):81–96. doi: 10.1093/toxsci/kfu211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Shahani N, Pryor W, Swarnkar S, Kholodilov N, Thinakaran G, Burke RE, et al. Rheb GTPase regulates beta-secretase levels and amyloid beta generation. J Biol Chem. 2014;289(9):5799–808. doi: 10.1074/jbc.M113.532713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Shang YC, Chong ZZ, Wang S, Maiese K. Prevention of beta-amyloid degeneration of microglia by erythropoietin depends on Wnt1, the PI 3-K/mTOR pathway, Bad, and Bcl-xL. Aging (Albany NY) 2012;4(3):187–201. doi: 10.18632/aging.100440. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Wang Y, Wang YX, Liu T, Law PY, Loh HH, Qiu Y, et al. mu-Opioid receptor attenuates Abeta oligomers-induced neurotoxicity through mTOR signaling. CNS Neurosci Ther. 2015;21(1):8–14. doi: 10.1111/cns.12316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Xue Z, Guo Y, Zhang S, Huang L, He Y, Fang R, et al. Beta-asarone attenuates amyloid beta-induced autophagy via Akt/mTOR pathway in PC12 cells. Eur J Pharmacol. 2014;741:195–204. doi: 10.1016/j.ejphar.2014.08.006. [DOI] [PubMed] [Google Scholar]
  • 149.Gubern C, Camos S, Hurtado O, Rodriguez R, Romera VG, Sobrado M, et al. Characterization of Gcf2/Lrrfip1 in experimental cerebral ischemia and its role as a modulator of Akt, mTOR and beta-catenin signaling pathways. Neuroscience. 2014;268C:48–65. doi: 10.1016/j.neuroscience.2014.02.051. [DOI] [PubMed] [Google Scholar]
  • 150.Koh PO. Ferulic acid attenuates focal cerebral ischemia-induced decreases in p70S6 kinase and S6 phosphorylation. Neurosci Lett. 2013;555:7–11. doi: 10.1016/j.neulet.2013.09.001. [DOI] [PubMed] [Google Scholar]
  • 151.Zhong X, Lin R, Li Z, Mao J, Chen L. Effects of Salidroside on Cobalt Chloride-Induced Hypoxia Damage and mTOR Signaling Repression in PC12 Cells. Biol Pharm Bull. 2014;37(7):1199–206. doi: 10.1248/bpb.b14-00100. [DOI] [PubMed] [Google Scholar]
  • 152.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;7(2):231–9. doi: 10.1111/1753-0407.12177. [DOI] [PubMed] [Google Scholar]
  • 153.Peng N, Meng N, Wang S, Zhao F, Zhao J, Su L, et al. An activator of mTOR inhibits oxLDL-induced autophagy and apoptosis in vascular endothelial cells and restricts atherosclerosis in apolipoprotein E(−/−) mice. Scientific reports. 2014;4:5519. doi: 10.1038/srep05519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Kimura R, Okouchi M, Kato T, Imaeda K, Okayama N, Asai K, et al. Epidermal growth factor receptor transactivation is necessary for glucagon-like peptide-1 to protect PC12 cells from apoptosis. Neuroendocrinology. 2013;97(4):300–8. doi: 10.1159/000345529. [DOI] [PubMed] [Google Scholar]
  • 155.Miao XY, Gu ZY, Liu P, Hu Y, Li L, Gong YP, et al. The human glucagon-like peptide-1 analogue liraglutide regulates pancreatic beta-cell proliferation and apoptosis via an AMPK/mTOR/P70S6K signaling pathway. Peptides. 2013;39:71–9. doi: 10.1016/j.peptides.2012.10.006. [DOI] [PubMed] [Google Scholar]
  • 156.Park YS, Park JH, Ko J, Shin IC, Koh HC. mTOR inhibition by rapamycin protects against deltamethrin-induced apoptosis in PC12 Cells. Environmental toxicology. 2015 doi: 10.1002/tox.22216. [DOI] [PubMed] [Google Scholar]
  • 157.Weckman A, Di Ieva A, Rotondo F, Syro LV, Ortiz LD, Kovacs K, et al. Autophagy in the endocrine glands. Journal of molecular endocrinology. 2014;52(2):R151–63. doi: 10.1530/JME-13-0241. [DOI] [PubMed] [Google Scholar]
  • 158.Klionsky DJ, Abdelmohsen K, Abe A, Abedin MJ, Abeliovich H, Acevedo Arozena A, et al. Guidelines for the use and interpretation of assays for monitoring autophagy (3rd edition) Autophagy. 2016;12(1):1–222. doi: 10.1080/15548627.2015.1100356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Francois A, Rioux-Bilan A, Quellard N, Fernandez B, Janet T, Chassaing D, et al. Longitudinal follow-up of autophagy and inflammation in brain of APPswePS1dE9 transgenic mice. J Neuroinflammation. 2014;11(1):139. doi: 10.1186/s12974-014-0139-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Vakifahmetoglu-Norberg H, Xia HG, Yuan J. Pharmacologic agents targeting autophagy. J Clin Invest. 2015;125(1):5–13. doi: 10.1172/JCI73937. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Frederick C, Ando K, Leroy K, Heraud C, Suain V, Buee L, et al. Rapamycin ester analog CCI-779/Temsirolimus alleviates tau pathology and improves motor deficit in mutant tau transgenic mice. J Alzheimers Dis. 2015;44(4):1145–56. doi: 10.3233/JAD-142097. [DOI] [PubMed] [Google Scholar]
  • 162.Sasazawa Y, Sato N, Umezawa K, Simizu S. Conophylline protects cells in cellular models of neurodegenerative diseases by inducing mammalian target of rapamycin (mTOR)-independent autophagy. J Biol Chem. 2015;290(10):6168–78. doi: 10.1074/jbc.M114.606293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Maiese K. FoxO Transcription Factors and Regenerative Pathways in Diabetes Mellitus. Curr Neurovasc Res. 2015;12(4):404–13. doi: 10.2174/1567202612666150807112524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Balduini W, Carloni S, Buonocore G. Autophagy in hypoxia-ischemia induced brain injury. J Matern Fetal Neonatal Med. 2012;25(Suppl 1):30–4. doi: 10.3109/14767058.2012.663176. [DOI] [PubMed] [Google Scholar]
  • 165.Kulbe JR, Mulcahy Levy JM, Coultrap SJ, Thorburn A, Bayern KU. Excitotoxic glutamate insults block autophagic flux in hippocampal neurons. Brain Res. 2014;1542:12–9. doi: 10.1016/j.brainres.2013.10.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Jeong JK, Moon MH, Bae BC, Lee YJ, Seol JW, Kang HS, et al. Autophagy induced by resveratrol prevents human prion protein-mediated neurotoxicity. Neurosci Res. 2012;73(2):99–105. doi: 10.1016/j.neures.2012.03.005. [DOI] [PubMed] [Google Scholar]
  • 167.Chan TM, Chen JY, Ho LI, Lin HP, Hsueh KW, Liu DD, et al. ADSC therapy in neurodegenerative disorders. Cell Transplant. 2014;23(4–5):549–57. doi: 10.3727/096368914X678445. [DOI] [PubMed] [Google Scholar]
  • 168.Zhu Z, Yan J, Jiang W, Yao XG, Chen J, Chen L, et al. Arctigenin effectively ameliorates memory impairment in Alzheimer’s disease model mice targeting both beta-amyloid production and clearance. J Neurosci. 2013;33(32):13138–49. doi: 10.1523/JNEUROSCI.4790-12.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Urbanek T, Kuczmik W, Basta-Kaim A, Gabryel B. Rapamycin induces of protective autophagy in vascular endothelial cells exposed to oxygen-glucose deprivation. Brain Res. 2014;1553:1–11. doi: 10.1016/j.brainres.2014.01.017. [DOI] [PubMed] [Google Scholar]
  • 170.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]
  • 171.Liu Z, Stanojevic V, Brindamour LJ, Habener JF. GLP1-derived nonapeptide GLP1(28–36)amide protects pancreatic beta-cells from glucolipotoxicity. J Endocrinol. 2012;213(2):143–54. doi: 10.1530/JOE-11-0328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.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;64(1):36–48. doi: 10.2337/db14-0267. [DOI] [PubMed] [Google Scholar]
  • 173.Maiese K. Stem cell guidance through the mechanistic target of rapamycin. World J Stem Cells. 2015;7(7):999–1009. doi: 10.4252/wjsc.v7.i7.999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Maiese K. MicroRNAs and SIRT1: A Strategy for Stem Cell Renewal and Clinical Development? J Transl Sci. 2015;1(3):55–7. [PMC free article] [PubMed] [Google Scholar]
  • 175.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;283(41):27810–9. doi: 10.1074/jbc.M804681200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.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;8(3):220–35. doi: 10.2174/156720211796558069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.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]
  • 178.Wang L, Jia Y, Rogers H, Wu YP, Huang S, Noguchi CT. GATA-binding protein 4 (GATA-4) and T-cell acute leukemia 1 (TAL1) regulate myogenic differentiation and erythropoietin response via cross-talk with Sirtuin1 (Sirt1) J Biol Chem. 2012;287(36):30157–69. doi: 10.1074/jbc.M112.376640. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Wang L, Teng R, Di L, Rogers H, Wu H, Kopp JB, et al. PPARalpha and Sirt1 mediate erythropoietin action in increasing metabolic activity and browning of white adipocytes to protect against obesity and metabolic disorders. Diabetes. 2013;62(12):4122–31. doi: 10.2337/db13-0518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Wu H, Wang H, Zhang W, Wei X, Zhao J, Yan P, et al. rhEPO affects apoptosis in hippocampus of aging rats by upregulating SIRT1. Int J Clin Exp Pathol. 2015;8(6):6870–80. [PMC free article] [PubMed] [Google Scholar]
  • 181.Ou X, Lee MR, Huang X, Messina-Graham S, Broxmeyer HE. SIRT1 positively regulates autophagy and mitochondria function in embryonic stem cells under oxidative stress. Stem Cells. 2014;32(5):1183–94. doi: 10.1002/stem.1641. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Guo W, Qian L, Zhang J, Zhang W, Morrison A, Hayes P, et al. Sirt1 overexpression in neurons promotes neurite outgrowth and cell survival through inhibition of the mTOR signaling. J Neurosci Res. 2011;89(11):1723–36. doi: 10.1002/jnr.22725. [DOI] [PubMed] [Google Scholar]
  • 183.Ma L, Dong W, Wang R, Li Y, Xu B, Zhang J, et al. Effect of caloric restriction on the SIRT1/mTOR signaling pathways in senile mice. Brain Res Bull. 2015;116:67–72. doi: 10.1016/j.brainresbull.2015.06.004. [DOI] [PubMed] [Google Scholar]
  • 184.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;9(2):89–99. doi: 10.2174/156720212800410858. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.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]
  • 186.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;29(4):613–8. doi: 10.3892/ijmm.2012.891. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Lee Y, Hong Y, Lee SR, Chang KT. Autophagy contributes to retardation of cardiac growth in diabetic rats. Lab Anim Res. 2012;28(2):99–107. doi: 10.5625/lar.2012.28.2.99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.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]
  • 189.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]
  • 190.Walker CL, Walker MJ, Liu NK, Risberg EC, Gao X, Chen J, et al. Systemic bisperoxovanadium activates Akt/mTOR, reduces autophagy, and enhances recovery following cervical spinal cord injury. PLoS One. 2012;7(1):e30012. doi: 10.1371/journal.pone.0030012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Yin B, Liang H, Chen Y, Chu K, Huang L, Fang L, et al. EGB1212 post-treatment ameliorates hippocampal CA1 neuronal death and memory impairment induced by transient global cerebral ischemia/reperfusion. Am J Chin Med. 2013;41(6):1329–41. doi: 10.1142/S0192415X13500894. [DOI] [PubMed] [Google Scholar]
  • 192.Choi KC, Kim SH, Ha JY, Kim ST, Son JH. A novel mTOR activating protein protects dopamine neurons against oxidative stress by repressing autophagy related cell death. J Neurochem. 2010;112(2):366–76. doi: 10.1111/j.1471-4159.2009.06463.x. [DOI] [PubMed] [Google Scholar]
  • 193.Lan AP, Chen J, Zhao Y, Chai Z, Hu Y. mTOR Signaling in Parkinson’s Disease. Neuromolecular Med. 2016 doi: 10.1007/s12017-016-8417-7. [DOI] [PubMed] [Google Scholar]
  • 194.Francois A, Terro F, Janet T, Bilan AR, Paccalin M, Page G. Involvement of interleukin-1beta in the autophagic process of microglia: relevance to Alzheimer’s disease. J Neuroinflammation. 2013;10:151. doi: 10.1186/1742-2094-10-151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Benedict A, Bansal N, Senina S, Hooper I, Lundberg L, de la Fuente C, et al. Repurposing FDA-approved drugs as therapeutics to treat Rift Valley fever virus infection. Frontiers in microbiology. 2015;6:676. doi: 10.3389/fmicb.2015.00676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Engels MC, Rajarajan K, Feistritzer R, Sharma A, Nielsen UB, Schalij MJ, et al. Insulin-like growth factor promotes cardiac lineage induction in vitro by selective expansion of early mesoderm. Stem Cells. 2014;32(6):1493–502. doi: 10.1002/stem.1660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Geldman A, Pallen CJ. Protein tyrosine phosphatases in mast cell signaling. Methods in molecular biology (Clifton, NJ) 2015;1220:269–86. doi: 10.1007/978-1-4939-1568-2_17. [DOI] [PubMed] [Google Scholar]
  • 198.Lauzon MA, Daviau A, Marcos B, Faucheux N. Growth factor treatment to overcome Alzheimer’s dysfunctional signaling. Cell Signal. 2015;27(6):1025–38. doi: 10.1016/j.cellsig.2015.02.018. [DOI] [PubMed] [Google Scholar]
  • 199.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]
  • 200.Chong ZZ, Kang JQ, Maiese K. Erythropoietin is a novel vascular protectant through activation of Akt1 and mitochondrial modulation of cysteine proteases. Circulation. 2002;106(23):2973–9. doi: 10.1161/01.cir.0000039103.58920.1f. [DOI] [PubMed] [Google Scholar]
  • 201.Chong ZZ, Kang JQ, Maiese K. Apaf-1, Bcl-xL, Cytochrome c, and Caspase-9 Form the Critical Elements for Cerebral Vascular Protection by Erythropoietin. J Cereb Blood Flow Metab. 2003;23(3):320–30. doi: 10.1097/01.WCB.0000050061.57184.AE. [DOI] [PubMed] [Google Scholar]
  • 202.Chong ZZ, Kang JQ, Maiese K. Erythropoietin fosters both intrinsic and extrinsic neuronal protection through modulation of microglia, Akt1, Bad, and caspase-mediated pathways. Br J Pharmacol. 2003;138(6):1107–18. doi: 10.1038/sj.bjp.0705161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Chong ZZ, Lin SH, Kang JQ, Maiese K. Erythropoietin prevents early and late neuronal demise through modulation of Akt1 and induction of caspase 1, 3, and 8. J Neurosci Res. 2003;71(5):659–69. doi: 10.1002/jnr.10528. [DOI] [PubMed] [Google Scholar]
  • 204.Ryou MG, Choudhury GR, Li W, Winters A, Yuan F, Liu R, et al. Methylene blue-induced neuronal protective mechanism against hypoxia-reoxygenation stress. Neuroscience. 2015;301:193–203. doi: 10.1016/j.neuroscience.2015.05.064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Sanghera KP, Mathalone N, Baigi R, Panov E, Wang D, Zhao X, et al. The PI3K/Akt/mTOR pathway mediates retinal progenitor cell survival under hypoxic and superoxide stress. Mol Cell Neurosci. 2011;47(2):145–53. doi: 10.1016/j.mcn.2011.03.010. [DOI] [PubMed] [Google Scholar]
  • 206.Shang YC, Chong ZZ, Wang S, Maiese K. Erythropoietin and Wnt1 Govern Pathways of mTOR, Apaf-1, and XIAP in Inflammatory Microglia. Curr Neurovasc Res. 2011;8(4):270–85. doi: 10.2174/156720211798120990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Yu Y, Shiou SR, Guo Y, Lu L, Westerhoff M, Sun J, et al. Erythropoietin protects epithelial cells from excessive autophagy and apoptosis in experimental neonatal necrotizing enterocolitis. PLoS One. 2013;8(7):e69620. doi: 10.1371/journal.pone.0069620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Ma C, Cheng F, Wang X, Zhai C, Yue W, Lian Y, et al. Erythropoietin Pathway: A Potential Target for the Treatment of Depression. International journal of molecular sciences. 2016;17(5) doi: 10.3390/ijms17050677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Maiese K. Regeneration in the nervous system with erythropoietin. Frontiers in bioscience (Landmark edition) 2016;21:561–96. doi: 10.2741/4408. [DOI] [PubMed] [Google Scholar]
  • 210.Maiese K, Chong ZZ, Li F, Shang YC. Erythropoietin: elucidating new cellular targets that broaden therapeutic strategies. Prog Neurobiol. 2008;85(2):194–213. doi: 10.1016/j.pneurobio.2008.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Niu HS, Chang CH, Niu CS, Cheng JT, Lee KS. Erythropoietin ameliorates hyperglycemia in type 1-like diabetic rats. Drug design, development and therapy. 2016;10:1877–84. doi: 10.2147/DDDT.S105867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Samy DM, Ismail CA, Nassra RA, Zeitoun TM, Nomair AM. Downstream modulation of extrinsic apoptotic pathway in streptozotocin-induced Alzheimer’s dementia in rats: Erythropoietin versus curcumin. Eur J Pharmacol. 2016;770:52–60. doi: 10.1016/j.ejphar.2015.11.046. [DOI] [PubMed] [Google Scholar]
  • 213.Stoppe C, Ney J, Brenke M, Goetzenich A, Emontzpohl C, Schalte G, et al. Sub-anesthetic Xenon Increases Erythropoietin Levels in Humans: A Randomized Controlled Trial. Sports Med. 2016 doi: 10.1007/s40279-016-0505-1. [DOI] [PubMed] [Google Scholar]
  • 214.Tsitsimpikou C, Kouretas D, Tsarouhas K, Fitch K, Spandidos DA, Tsatsakis A. Applications and biomonitoring issues of recombinant erythropoietins for doping control. Ther Drug Monit. 2011;33(1):3–13. doi: 10.1097/FTD.0b013e31820032c4. [DOI] [PubMed] [Google Scholar]
  • 215.Leuchter RH, Gui L, Poncet A, Hagmann C, Lodygensky GA, Martin E, et al. Association between early administration of high-dose erythropoietin in preterm infants and brain MRI abnormality at term-equivalent age. JAMA. 2014;312(8):817–24. doi: 10.1001/jama.2014.9645. [DOI] [PubMed] [Google Scholar]
  • 216.Jang W, Park J, Shin KJ, Kim JS, Kim JS, Youn J, et al. Safety and efficacy of recombinant human erythropoietin treatment of non-motor symptoms in Parkinson’s disease. J Neurol Sci. 2014;337(1–2):47–54. doi: 10.1016/j.jns.2013.11.015. [DOI] [PubMed] [Google Scholar]
  • 217.Chong ZZ, Kang JQ, Maiese K. Erythropoietin: cytoprotection in vascular and neuronal cells. Curr Drug Targets Cardiovasc Haematol Disord. 2003;3(2):141–54. doi: 10.2174/1568006033481483. [DOI] [PubMed] [Google Scholar]
  • 218.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;150(7):839–50. doi: 10.1038/sj.bjp.0707161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Gammella E, Leuenberger C, Gassmann M, Ostergaard L. Evidence of synergistic/additive effects of sildenafil and erythropoietin in enhancing survival and migration of hypoxic endothelial cells. Am J Physiol Lung Cell Mol Physiol. 2013;304(4):L230–9. doi: 10.1152/ajplung.00112.2012. [DOI] [PubMed] [Google Scholar]
  • 220.Gut N, Piecha G, Aldebssi F, Schaefer S, Bekeredjian R, Schirmacher P, et al. Erythropoietin combined with ACE inhibitor prevents heart remodeling in 5/6 nephrectomized rats independently of blood pressure and kidney function. Am J Nephrol. 2013;38(2):124–35. doi: 10.1159/000353106. [DOI] [PubMed] [Google Scholar]
  • 221.Kang J, Yun JY, Hur J, Kang JA, Choi JI, Ko SB, et al. Erythropoietin priming improves the vasculogenic potential of G-CSF mobilized human peripheral blood mononuclear cells. Cardiovasc Res. 2014;104(1):171–82. doi: 10.1093/cvr/cvu180. [DOI] [PubMed] [Google Scholar]
  • 222.Szygula Z, Lubkowska A, Giemza C, Skrzek A, Bryczkowska I, Dolegowska B. Hematological Parameters, and Hematopoietic Growth Factors: Epo and IL-3 in Response to Whole-Body Cryostimulation (WBC) in Military Academy Students. PLoS One. 2014;9(4):e93096. doi: 10.1371/journal.pone.0093096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Lourhmati A, Buniatian GH, Paul C, Verleysdonk S, Buecheler R, Buadze M, et al. Age-dependent astroglial vulnerability to hypoxia and glutamate: the role for erythropoietin. PLoS One. 2013;8(10):e77182. doi: 10.1371/journal.pone.0077182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Millet A, Bouzat P, Trouve-Buisson T, Batandier C, Pernet-Gallay K, Gaide-Chevronnay L, et al. Erythropoietin and Its Derivates Modulate Mitochondrial Dysfunction after Diffuse Traumatic Brain Injury. J Neurotrauma. 2016 doi: 10.1089/neu.2015.4160. [DOI] [PubMed] [Google Scholar]
  • 225.Osborn M, Rustom N, Clarke M, Litteljohn D, Rudyk C, Anisman H, et al. Antidepressant-like effects of erythropoietin: a focus on behavioural and hippocampal processes. PLoS One. 2013;8(9):e72813. doi: 10.1371/journal.pone.0072813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.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]
  • 227.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]
  • 228.Li F, Chong ZZ, Maiese K. Erythropoietin on a Tightrope: Balancing Neuronal and Vascular Protection between Intrinsic and Extrinsic Pathways. Neurosignals. 2004;13(6):265–89. doi: 10.1159/000081963. [DOI] [PubMed] [Google Scholar]
  • 229.Palazzuoli A, Ruocco G, Pellegrini M, De Gori C, Del Castillo G, Giordano N, et al. The role of erythropoietin stimulating agents in anemic patients with heart failure: solved and unresolved questions. Ther Clin Risk Manag. 2014;10:641–50. doi: 10.2147/TCRM.S61551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Maiese K, Li F, Chong ZZ. New avenues of exploration for erythropoietin. Jama. 2005;293(1):90–5. doi: 10.1001/jama.293.1.90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Imai N, Kawamura A, Higuchi M, Oh-eda M, Orita T, Kawaguchi T, et al. Physicochemical and biological comparison of recombinant human erythropoietin with human urinary erythropoietin. J Biochem (Tokyo) 1990;107(3):352–9. doi: 10.1093/oxfordjournals.jbchem.a123050. [DOI] [PubMed] [Google Scholar]
  • 232.Castaneda-Arellano R, Beas-Zarate C, Feria-Velasco AI, Bitar-Alatorre EW, Rivera-Cervantes MC. From neurogenesis to neuroprotection in the epilepsy: signalling by erythropoietin. Frontiers in bioscience (Landmark edition) 2014;19:1445–55. doi: 10.2741/4295. [DOI] [PubMed] [Google Scholar]
  • 233.Maiese K, Chong ZZ, Shang YC. Raves and risks for erythropoietin. Cytokine Growth Factor Rev. 2008;19(2):145–55. doi: 10.1016/j.cytogfr.2008.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Krantz SB. Erythropoietin. Blood. 1991;77(3):419–34. [PubMed] [Google Scholar]
  • 235.Tsuda E, Kawanishi G, Ueda M, Masuda S, Sasaki R. The role of carbohydrate in recombinant human erythropoietin. Eur J Biochem. 1990;188(2):405–11. doi: 10.1111/j.1432-1033.1990.tb15417.x. [DOI] [PubMed] [Google Scholar]
  • 236.Uchida E, Morimoto K, Kawasaki N, Izaki Y, Abdu Said A, Hayakawa T. Effect of active oxygen radicals on protein and carbohydrate moieties of recombinant human erythropoietin. Free Radic Res. 1997;27(3):311–23. doi: 10.3109/10715769709065769. [DOI] [PubMed] [Google Scholar]
  • 237.Toyoda T, Itai T, Arakawa T, Aoki KH, Yamaguchi H. Stabilization of human recombinant erythropoietin through interactions with the highly branched N-glycans. J Biochem (Tokyo) 2000;128(5):731–7. doi: 10.1093/oxfordjournals.jbchem.a022809. [DOI] [PubMed] [Google Scholar]
  • 238.Wang FF, Kung CK, Goldwasser E. Some chemical properties of human erythropoietin. Endocrinology. 1985;116(6):2286–92. doi: 10.1210/endo-116-6-2286. [DOI] [PubMed] [Google Scholar]
  • 239.Caprara C, Grimm C. From oxygen to erythropoietin: relevance of hypoxia for retinal development, health and disease. Prog Retin Eye Res. 2012;31(1):89–119. doi: 10.1016/j.preteyeres.2011.11.003. [DOI] [PubMed] [Google Scholar]
  • 240.Chong ZZ, Kang JQ, Maiese K. Angiogenesis and plasticity: role of erythropoietin in vascular systems. J Hematother Stem Cell Res. 2002;11(6):863–71. doi: 10.1089/152581602321080529. [DOI] [PubMed] [Google Scholar]
  • 241.Kato S, Aoyama M, Kakita H, Hida H, Kato I, Ito T, et al. Endogenous erythropoietin from astrocyte protects the oligodendrocyte precursor cell against hypoxic and reoxygenation injury. J Neurosci Res. 2011;89(10):1566–74. doi: 10.1002/jnr.22702. [DOI] [PubMed] [Google Scholar]
  • 242.Maiese K, Li F, Chong ZZ. Erythropoietin in the brain: can the promise to protect be fulfilled? Trends Pharmacol Sci. 2004;25(11):577–83. doi: 10.1016/j.tips.2004.09.006. [DOI] [PubMed] [Google Scholar]
  • 243.Moore EM, Bellomo R, Nichol AD. Erythropoietin as a novel brain and kidney protective agent. Anaesth Intensive Care. 2011;39(3):356–72. doi: 10.1177/0310057X1103900306. [DOI] [PubMed] [Google Scholar]
  • 244.Maiese K. Triple play: Promoting neurovascular longevity with nicotinamide, WNT, and erythropoietin in diabetes mellitus. Biomed Pharmacother. 2008;62(4):218–32. doi: 10.1016/j.biopha.2008.01.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245.Guven Bagla A, Ercan E, Asgun HF, Ickin M, Ercan F, Yavuz O, et al. Experimental acute myocardial infarction in rats: HIF-1alpha, caspase-3, erythropoietin and erythropoietin receptor expression and the cardioprotective effects of two different erythropoietin doses. Acta histochemica. 2013;115(7):658–68. doi: 10.1016/j.acthis.2013.01.005. [DOI] [PubMed] [Google Scholar]
  • 246.Nishimura K, Tokida M, Katsuyama H, Nakagawa H, Matsuo S. The effect of hemin-induced oxidative stress on erythropoietin production in HepG2 cells. Cell Biol Int. 2014 doi: 10.1002/cbin.10329. [DOI] [PubMed] [Google Scholar]
  • 247.Maiese K, Chong ZZ, Shang YC, Wang S. Erythropoietin: new directions for the nervous system. International journal of molecular sciences. 2012;13(9):11102–29. doi: 10.3390/ijms130911102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248.Tsai CF, Kuo YH, Yeh WL, Wu CY, Lin HY, Lai SW, et al. Regulatory Effects of Caffeic Acid Phenethyl Ester on Neuroinflammation in Microglial Cells. International journal of molecular sciences. 2015;16(3):5572–89. doi: 10.3390/ijms16035572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249.Diez-Padrisa N, Aguilar R, Machevo S, Morais L, Nhampossa T, O’Callaghan-Gordo C, et al. Erythropoietin levels are not independently associated with malaria-attributable severe disease in mozambican children. PLoS ONE. 2011;6(8):e24090. doi: 10.1371/journal.pone.0024090. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 250.Stoppe C, Coburn M, Fahlenkamp A, Ney J, Kraemer S, Rossaint R, et al. Elevated serum concentrations of erythropoietin after xenon anaesthesia in cardiac surgery: secondary analysis of a randomized controlled trial. British journal of anaesthesia. 2015;114(4):701–3. doi: 10.1093/bja/aev060. [DOI] [PubMed] [Google Scholar]
  • 251.Li CL, Jiang J, Fan YQ, Fu GS, Wang JA, Fan WM. Knockout of the tumor necrosis factor a receptor 1 gene can up-regulate erythropoietin receptor during myocardial ischemia-reperfusion injury in mice. Chin Med J (Engl) 2009;122(5):566–70. [PubMed] [Google Scholar]
  • 252.Chong ZZ, Kang JQ, Maiese K. Hematopoietic factor erythropoietin fosters neuroprotection through novel signal transduction cascades. J Cereb Blood Flow Metab. 2002;22(5):503–14. doi: 10.1097/00004647-200205000-00001. [DOI] [PubMed] [Google Scholar]
  • 253.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;42(4):554–61. doi: 10.1111/j.1365-2184.2009.00617.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254.Marfia G, Madaschi L, Marra F, Menarini M, Bottai D, Formenti A, et al. Adult neural precursors isolated from post mortem brain yield mostly neurons: an erythropoietin-dependent process. Neurobiol Dis. 2011;43(1):86–98. doi: 10.1016/j.nbd.2011.02.004. [DOI] [PubMed] [Google Scholar]
  • 255.Kim J, Jung Y, Sun H, Joseph J, Mishra A, Shiozawa Y, et al. Erythropoietin mediated bone formation is regulated by mTOR signaling. J Cell Biochem. 2012;113(1):220–8. doi: 10.1002/jcb.23347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 256.Wang GB, Ni YL, Zhou XP, Zhang WF. The AKT/mTOR pathway mediates neuronal protective effects of erythropoietin in sepsis. Mol Cell Biochem. 2014;385(1–2):125–32. doi: 10.1007/s11010-013-1821-5. [DOI] [PubMed] [Google Scholar]
  • 257.Jang W, Kim HJ, Li H, Jo KD, Lee MK, Yang HO. The Neuroprotective Effect of Erythropoietin on Rotenone-Induced Neurotoxicity in SH-SY5Y Cells Through the Induction of Autophagy. Mol Neurobiol. 2015 doi: 10.1007/s12035-015-9316-x. [DOI] [PubMed] [Google Scholar]
  • 258.Li FQ, Zeng DK, Jia CL, Zhou P, Yin L, Zhang B, et al. The effects of sodium tanshinone IIa sulfonate pretreatment on high glucose-induced expression of fractalkine and apoptosis in human umbilical vein endothelial cells. International journal of clinical and experimental medicine. 2015;8(4):5279–86. [PMC free article] [PubMed] [Google Scholar]
  • 259.Maiese K, Li F, Chong ZZ, Shang YC. The Wnt signaling pathway: Aging gracefully as a protectionist? Pharmacol Ther. 2008;118(1):58–81. doi: 10.1016/j.pharmthera.2008.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 260.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. 2015;64(3):856–66. doi: 10.2337/db14-0444. [DOI] [PubMed] [Google Scholar]
  • 261.Tulsulkar J, Nada SE, Slotterbeck BD, McInerney MF, Shah ZA. Obesity and hyperglycemia lead to impaired post-ischemic recovery after permanent ischemia in mice. Obesity (Silver Spring, Md. 2015 doi: 10.1002/oby.21388. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262.Chen X, Wang CC, Song SM, Wei SY, Li JS, Zhao SL, et al. The administration of erythropoietin attenuates kidney injury induced by ischemia/reperfusion with increased activation of Wnt/beta-catenin signaling. Journal of the Formosan Medical Association = Taiwan yi zhi. 2015;114(5):430–7. doi: 10.1016/j.jfma.2015.01.007. [DOI] [PubMed] [Google Scholar]
  • 263.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;8(2):103–20. doi: 10.2174/156720211795495402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 264.Chong ZZ, Shang YC, Maiese K. Vascular injury during elevated glucose can be mitigated by erythropoietin and Wnt signaling. Curr Neurovasc Res. 2007;4(3):194–204. doi: 10.2174/156720207781387150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 265.Danielyan L, Schafer R, Schulz A, Ladewig T, Lourhmati A, Buadze M, et al. Survival, neuron-like differentiation and functionality of mesenchymal stem cells in neurotoxic environment: the critical role of erythropoietin. Cell Death Differ. 2009;16(12):1599–614. doi: 10.1038/cdd.2009.95. [DOI] [PubMed] [Google Scholar]
  • 266.Bargiela A, Cerro-Herreros E, Fernandez-Costa JM, Vilchez JJ, Llamusi B, Artero R. Increased autophagy and apoptosis contribute to muscle atrophy in a myotonic dystrophy type 1 Drosophila model. Dis Model Mech. 2015;8(7):679–90. doi: 10.1242/dmm.018127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 267.Bendix I, Schulze C, Haefen C, Gellhaus A, Endesfelder S, Heumann R, et al. Erythropoietin modulates autophagy signaling in the developing rat brain in an in vivo model of oxygen-toxicity. International journal of molecular sciences. 2012;13(10):12939–51. doi: 10.3390/ijms131012939. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 268.Bierer R, Peceny MC, Hartenberger CH, Ohls RK. Erythropoietin concentrations and neurodevelopmental outcome in preterm infants. Pediatrics. 2006;118(3):e635–40. doi: 10.1542/peds.2005-3186. [DOI] [PubMed] [Google Scholar]
  • 269.O’Gorman RL, Bucher HU, Held U, Koller BM, Huppi PS, Hagmann CF. Tract-based spatial statistics to assess the neuroprotective effect of early erythropoietin on white matter development in preterm infants. Brain. 2015;138(Pt 2):388–97. doi: 10.1093/brain/awu363. [DOI] [PubMed] [Google Scholar]
  • 270.Messier AM, Ohls RK. Neuroprotective effects of erythropoiesis-stimulating agents in term and preterm neonates. Curr Opin Pediatr. 2014 doi: 10.1097/MOP.0000000000000077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 271.Wu YW, Mathur AM, Chang T, McKinstry RC, Mulkey SB, Mayock DE, et al. High-Dose Erythropoietin and Hypothermia for Hypoxic-Ischemic Encephalopathy: A Phase II Trial. Pediatrics. 2016;137(6) doi: 10.1542/peds.2016-0191. [DOI] [PubMed] [Google Scholar]
  • 272.Assaraf MI, Diaz Z, Liberman A, Miller WH, Jr, Arvanitakis Z, Li Y, et al. Brain erythropoietin receptor expression in Alzheimer disease and mild cognitive impairment. J Neuropathol Exp Neurol. 2007;66(5):389–98. doi: 10.1097/nen.0b013e3180517b28. [DOI] [PubMed] [Google Scholar]
  • 273.Castelli R, Deliliers GL, Colombo R, Moreo G, Gallipoli P, Pantaleo G. Biosimilar epoetin in elderly patients with low-risk myelodysplastic syndromes improves anemia, quality of life, and brain function. Ann Hematol. 2014;93(9):1523–9. doi: 10.1007/s00277-014-2070-8. [DOI] [PubMed] [Google Scholar]
  • 274.Robertson CS, Hannay HJ, Yamal JM, Gopinath S, Goodman JC, Tilley BC, et al. Effect of erythropoietin and transfusion threshold on neurological recovery after traumatic brain injury: a randomized clinical trial. JAMA. 2014;312(1):36–47. doi: 10.1001/jama.2014.6490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 275.Nichol A, French C, Little L, Haddad S, Presneill J, Arabi Y, et al. Erythropoietin in traumatic brain injury (EPO-TBI): a double-blind randomised controlled trial. Lancet. 2015;386(10012):2499–506. doi: 10.1016/S0140-6736(15)00386-4. [DOI] [PubMed] [Google Scholar]
  • 276.Cramer SC, Hill MD. Human choriogonadotropin and epoetin alfa in acute ischemic stroke patients (REGENESIS-LED trial) International journal of stroke : official journal of the International Stroke Society. 2014;9(3):321–7. doi: 10.1111/ijs.12260. [DOI] [PubMed] [Google Scholar]
  • 277.Najjar SS, Rao SV, Melloni C, Raman SV, Povsic TJ, Melton L, et al. Intravenous erythropoietin in patients with ST-segment elevation myocardial infarction: REVEAL: a randomized controlled trial. Jama. 2011;305(18):1863–72. doi: 10.1001/jama.2011.592. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 278.Taniguchi N, Nakamura T, Sawada T, Matsubara K, Furukawa K, Hadase M, et al. Erythropoietin prevention trial of coronary restenosis and cardiac remodeling after ST-elevated acute myocardial infarction (EPOC-AMI): a pilot, randomized, placebo-controlled study. Circ J. 2010;74(11):2365–71. doi: 10.1253/circj.cj-10-0267. [DOI] [PubMed] [Google Scholar]
  • 279.Wen Y, Xu J, Ma X, Gao Q. High-dose erythropoietin in acute ST-segment elevation myocardial infarction: a meta-analysis of randomized controlled trials. American journal of cardiovascular drugs : drugs, devices, and other interventions. 2013;13(6):435–42. doi: 10.1007/s40256-013-0042-3. [DOI] [PubMed] [Google Scholar]
  • 280.Cariou A, Deye N, Vivien B, Richard O, Pichon N, Bourg A, et al. Early High-Dose Erythropoietin Therapy After Out-of-Hospital Cardiac Arrest: A Multicenter, Randomized Controlled Trial. J Am Coll Cardiol. 2016;68(1):40–9. doi: 10.1016/j.jacc.2016.04.040. [DOI] [PubMed] [Google Scholar]
  • 281.Kudenchuk PJ. Erythropoietin for Out-of-Hospital Cardiac Arrest: Growing Together or Apart? J Am Coll Cardiol. 2016;68(1):50–2. doi: 10.1016/j.jacc.2016.03.598. [DOI] [PubMed] [Google Scholar]
  • 282.Casadevall N, Nataf J, Viron B, Kolta A, Kiladjian J-J, Martin-Dupont P, et al. Pure Red-Cell Aplasia and Antierythropoietin Antibodies in Patients Treated with Recombinant Erythropoietin. N Engl J Med. 2002;346(7):469–75. doi: 10.1056/NEJMoa011931. [DOI] [PubMed] [Google Scholar]
  • 283.Hara A, Furuichi K, Yamahana J, Yasuda H, Iwata Y, Sakai N, et al. Effect of Autoantibodies to Erythropoietin Receptor in Systemic Lupus Erythematosus with Biopsy-proven Lupus Nephritis. The Journal of rheumatology. 2016 doi: 10.3899/jrheum.151430. [DOI] [PubMed] [Google Scholar]
  • 284.Bergmann MW, Haufe S, von Knobelsdorff-Brenkenhoff F, Mehling H, Wassmuth R, Munch I, et al. A pilot study of chronic, low-dose epoetin-{beta} following percutaneous coronary intervention suggests safety, feasibility, and efficacy in patients with symptomatic ischaemic heart failure. Eur J Heart Fail. 2011;13(5):560–8. doi: 10.1093/eurjhf/hfr002. [DOI] [PubMed] [Google Scholar]
  • 285.Parvin A, Pranap R, Shalini U, Devendran A, Baker JE, Dhanasekaran A. Erythropoietin protects cardiomyocytes from cell death during hypoxia/reperfusion injury through activation of survival signaling pathways. PLoS One. 2014;9(9):e107453. doi: 10.1371/journal.pone.0107453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 286.Zhang X, Dong S, Qin Y, Bian X. Protective effect of erythropoietin against myocardial injury in rats with sepsis and its underlying mechanisms. Molecular medicine reports. 2015;11(5):3317–29. doi: 10.3892/mmr.2015.3155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 287.Jiang T, Yu JT, Zhu XC, Tan MS, Wang HF, Cao L, et al. Temsirolimus promotes autophagic clearance of amyloid-beta and provides protective effects in cellular and animal models of Alzheimer’s disease. Pharmacol Res. 2014;81C:54–63. doi: 10.1016/j.phrs.2014.02.008. [DOI] [PubMed] [Google Scholar]

RESOURCES