Next Article in Journal
Genome-Wide Transcriptome Profiling of Mycobacterium smegmatis MC2 155 Cultivated in Minimal Media Supplemented with Cholesterol, Androstenedione or Glycerol
Previous Article in Journal
Varicella Skin Complications in Childhood: A Case Series and a Systematic Review of the Literature
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Erythropoietin Pathway: A Potential Target for the Treatment of Depression

School of Basic Medical Sciences, Beijing University of Chinese Medicine, School of Basic Medical Sciences, 11 Beisanhuandong Road, Chao Yang District, Beijing 100029, China
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2016, 17(5), 677; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms17050677
Submission received: 5 March 2016 / Revised: 5 April 2016 / Accepted: 27 April 2016 / Published: 6 May 2016
(This article belongs to the Section Biochemistry)

Abstract

:
During the past decade, accumulating evidence from both clinical and experimental studies has indicated that erythropoietin may have antidepressant effects. In addition to the kidney and liver, many organs have been identified as secretory tissues for erythropoietin, including the brain. Its receptor is expressed in cerebral and spinal cord neurons, the hypothalamus, hippocampus, neocortex, dorsal root ganglia, nerve axons, and Schwann cells. These findings may highlight new functions for erythropoietin, which was originally considered to play a crucial role in the progress of erythroid differentiation. Erythropoietin and its receptor signaling through JAK2 activate multiple downstream signaling pathways including STAT5, PI3K/Akt, NF-κB, and MAPK. These factors may play an important role in inflammation and neuroprogression in the nervous system. This is particularly true for the hippocampus, which is possibly related to learning, memory, neurocognitive deficits and mood alterations. Thus, the influence of erythropoietin on the downstream pathways known to be involved in the treatment of depression makes the erythropoietin-related pathway an attractive target for the development of new therapeutic approaches. Focusing on erythropoietin may help us understand the pathogenic mechanisms of depression and the molecular basis of its treatment.

Graphical Abstract

1. Introduction

Depression is the leading cause of psychiatric disability across the globe because of its chronic, treatment-resistant, and recurrent nature; high prevalence; and comorbidity with other chronic neurological and immune diseases [1]. Thus, depression is of major concern in terms of personal happiness and social welfare worldwide. Despite comprehensive biological research, the pathophysiology of depression remains largely unknown. The predominant hypothesis of the underlying mechanism generating depression rests on a low level of brain serotonin (5-hydroxytryptamine, 5-HT) and/or alterations of 5-HT receptors [2] in depressed patients. However, other hypotheses related to neuroinflammation and neuroplasticity are still being considered. For decades, traditional hypotheses of depression have underpinned research into the etiology of depression and in vitro testing archetypes; however, many patients continue to suffer from a number of psychiatric syndromes characterized by depressed mood symptoms and a loss of interest. Clinical data indicate that complete symptom remission occurs in only 30%–40% of patients whose treatment with first-line antidepressants is considered successful [3,4]. In addition, most available pharmacological treatment options that target causal factors of depression such as 5-HT and brain-derived neutrophic factor (BDNF) have a significant treatment-onset-response delay and fail to overturn neurocognitive dysfunction [5,6]. Because of these limitations, novel drugs or combinatorial treatments targeting different molecular pathways need to be developed.
In recent years, investigators have started to study inflammation and neuronal plasticity as significant processes underlying depression progression. A growing body of research suggests that depression is an inflammatory [7,8] and neuroprogressive [9,10,11,12] disorder, which could be accompanied by mitochondrial dysfunction [13] and induction of multiple oxidative and nitrosative pathways [14,15]. Both experimental and clinical evidence show that increased concentrations of pro-inflammatory cytokines and glucocorticoids, similar to those in chronically stressful situations and in depression, contribute to the behavioral changes associated with depression [16,17,18]. Targeting these pathways may have the potential to yield antidepressant outcomes.
The hematopoietic growth factor erythropoietin (EPO), known for its role in erythroid differentiation, was first defined as a humoral erythropoietic factor in parabiotic experiments [19] and in anemic plasma preparations [20] in 1950–1955. Since then, accumulating evidence has indicated that EPO has multiple targets and actions [21,22] other than those associated with its erythropoietic effects, similar to many other cytokines and growth factors. It is now widely accepted that EPO not only affects the hematopoietic system, but is also a multifunctional trophic factor that affects the general homoeostasis of the entire organism [23]. EPO has direct effects on immune cells [24], endothelial cells (ECs) [25], and bone marrow stromal cells [26], as well as cells of the heart, gastrointestinal tract, kidney, muscle, reproductive system [27], pancreas [28], and nervous system [29]. EPO is found to be produced in the central nervous system by neurons and astrocytes, where it exerts neurotrophic and neuroprotective effects by binding to EPO receptors (EPOR) in the brain [30,31]. Notably, we now know that in some kinds of acute and chronic neurodegenerative animal models, systemically delivered EPO is able to cross the blood brain barrier (BBB) and exhibits neuroprotective effects and promotion of neuroplasticity [32]. Further, accumulating evidence suggests that EPO has potential antidepressant effects. For this review, we present evidence that EPO-induced signaling pathways are involved in antidepressant activity or regression of depression, and describe the potential of EPO as a novel antidepressant. Ultimately, we hope that these data will lead to the development of EPO and/or its related signaling molecules as adjunct antidepressant therapies.

2. Expression of EPO and EPOR in the Nervous System

EPO is a 165-amino-acid protein and a member of the type I cytokine superfamily [33]. Several investigators detected the expression of EPO in other tissues, especially the central nervous system, presenting challenges of the common sense that only the kidney and the fetal liver were able to produce EPO [34]. In addition to the kidney, liver, and uterus, other tissues have been identified as EPO productive and secretory tissues, including peripheral endothelial cells, muscle cells, and insulin-producing cells [35].
Of all the newly identified EPO sites, the presence of EPO in the brain has generated the highest levels of interest and enthusiasm for further investigation. In the brain, the major sites of EPO production and secretion are the hippocampus, internal capsule, cortex, midbrain, cerebral endothelial cells and astrocytes [36,37]. When EPO was first discovered, it was thought that such a large protein could not cross the BBB, and several authors suggested that EPO had a paracrine and autocrine function in some kinds of cells, such as astrocytes [38,39]. However, recent research shows that EPO does indeed have the capacity to cross the BBB, which protects against a variety of potential brain injuries, including transient ischemia and reperfusion [40]. However, in the absence of injury to the BBB, EPO has limited access to the brain [41].
The EPOR, which was first characterized in the 1990s as a polypeptide with a single transmembrane domain and an extracellular domain containing a WSXWS motif [42], is expressed in progenitor cells from hematopoietic cells and ECs, skeletal muscle [43,44,45], and in the brain during the development stage and adulthood [46]. Studies have reported EPOR expression in parts of the nervous system, such as cerebral and spinal cord neurons, hypothalamus, hippocampus, neocortex, dorsal root ganglia, and nerve axons [47,48]. EPORs classically include two EPOR subunits, but may also associate with the β-common receptor (βcR, CD131) subunit [49,50]. This subunit is a crucial part of cytokine receptors such as interleukin (IL)-3, IL-5, and granulocyte-macrophage colony-stimulating factor (GM-CSF) [51], suggesting a potential role of βcR in EPO signaling pathways. Because βcR-knockout mice exhibit normal hematopoiesis [52], it has been suggested that a heteroreceptor complex comprising both EPOR and βcR could, at least partially, mediate the non-hematopoietic functions of EPO [53]. The receptors involved in tissue protection may differ from the hematopoietic EPORs, because some EPO derivatives, such as carbamylated EPO, were shown to mediate neuroprotection without stimulating erythropoiesis [54,55]. Accordingly, it has been suggested that the non-hematopoietic receptor may be a heteromer consisting of one hematopoietic EPOR together with one or more units of the βcR [56,57]. However, other studies detected little overlap of βcR and EPOR expression in the brain [58] and EPO-mediated protection has been demonstrated in cells that do not express βcR at detectable levels [59,60]. Another group of scientists identified ephrin-type B receptor 4 (EphB4) as an EPO receptor that triggers downstream signaling via STAT3 and promotes recombinant human EPO (rhEPO)-induced tumor growth and progression [61]. This receptor has been reported to be not only frequently amplified in some cancers [62], but also over-expressed in the brains of hypoxic-ischemic rats [63]. Through EphB2 signaling, it activates β-catenin in vitro and in vivo independently of Wnt-signaling and upregulates proneural transcription factors, and thus increases adult hippocampal neurogenesis [64]. Similarly, EphB4 is present and tends to colocalize with EPOR in a subset of cortical neurons [65]. These findings suggest that EPOR may consist of unidentified heterodimeric receptor subunits that may vary between different non-hematopoietic tissues.
In the adult human kidney and fetal liver, the release of EPO into the circulation depends upon tissue oxygen levels and transcription of the EPO gene is mediated via hypoxia-inducible factor (HIF)-2α [66]. In most tissues, including the brain, hypoxia-dependent expression of EPO and the EPOR is regulated principally by HIF-1, an a,b-heterodimeric protein which is activated by a collection of stimulators, such as hypoxia [67,68]. Each member of the HIF family, including HIF-1α, HIF-1β, and HIF-3α, appears to play an important role in the regulation of EPO and EPOR expression to protect against hypoxic cell injury [69]. Other cellular disturbances, such as hypoglycemia, increased levels of intracellular Ca2+, or intense neuronal depolarization generated by mitochondrial reactive oxygen species (ROS), can increase cerebral EPO expression via HIF activation [70,71,72]. However, the HIF family is not the only factor that can modify the expression of EPO and the EPOR. The GATA transcription factors, key regulators of hematopoiesis, such as GATA1 and GATA2, also contribute to EPO gene regulation [73]. Anemic stress, insulin release and cytokines including insulin-like growth factor, tumor necrosis factor-α [74], interleukin-1β and interleukin-6 [75,76] can also lead to increased expression of EPO and the EPOR in both neuronal and non-neuronal cell populations. Interestingly, a recent study found that hippocampal expression of EPO was decreased in mice by chronic unpredictable stress, and 5-HT could increase EPO expression in the hippocampus [77], which is possible related to verbal and visual learning and memory and spectrum of neurocognitive deficits and mood alterations [78,79]. This finding may highlight EPO as a potential target in the treatment of depression.

3. EPO-Induced Intracellular Signaling Pathways

The existence of the EPO/EPOR signaling pathway has recently been detected in a diversity of systems, but its precise role and function in neurobiology are still controversial (Figure 1). EPO acts via binding to its cell surface receptor, which consists of two EPOR molecules [80]. In non-neuronal cells, in a similar but more complex manner than in the hematopoietic system, EPO induces tyrosine phosphorylation of the EPOR and its associated kinase, Janus kinase 2 (JAK2); in fact, a comparable system has been addressed in neurons [81]. Endogenous and exogenous EPO can bind and stimulate the EPOR to induce phosphorylation of JAK2 [82,83]. Different receptors are involved in each tissue type and multiple neuroprotective signaling pathways are activated downstream of EPOR/JAK2 in the nervous system. Activated JAK2 induces various signaling pathways via several adaptor proteins such as phosphoinositide 3-kinase (PI3K), signal transducer and activator of transcription 5 (STAT5), nuclear factor kappa B (NF-κB) [84,85,86] and p42/44 mitogen-activated protein kinase (MAPK) [87]. All of these signaling pathways are known for promoting not only red blood cell proliferation, but also vasodilation [88], insulin-sensitization [89], and for having antithrombotic [90], anti-inflammatory and anti-apoptotic actions [91,92]. In particular, STAT5 and NFκB translocate into the nucleus and serve as transcription factors for Bcl-2 [93] and Bcl-xL [94], which are antiapoptotic genes. Both components of the signal transduction pathways (e.g., Akt/PKB) and gene products regulated by activated transcription factors (e.g., Bcl-2 and BclX) have been demonstrated to interfere with apoptotic processes in the nervous system [95].
The occurrence of EPOR splice variants [96] and the possible involvement of the EPOR-βcR heterodimer have received limited consideration [97]. Whereas homodimeric EPORs have been extensively studied, the existence of the heterodimeric complex is controversial and requires further study. Interestingly, similar signal transduction events, including activation of STAT5, are mediated via the EPOR-βcR hetero-receptor complex, which requires high local concentrations of EPO to be activated [98].

3.1. JAK2

JAK2 is a non-receptor tyrosine kinase involved in receptor signaling and hematopoiesis [99]. Both hematopoietic and non-hematopoietic effects are initiated by two tyrosine kinases of the JAK2 type, leading to trans- and EPOR-phosphorylation after receptor activation [100,101]. All dominant signaling pathways activated by EPO in erythropoiesis have also been implicated in the regulation of gene expression leading to neuroprotection [27]. Activated JAK2 induces a variety of signaling pathways that are known to affect the gene transcription involved in neuronal survival related to EPO [102,103].

3.2. STAT5

JAK2-mediated EPOR phosphorylation typically enables phosphorylation and dimerization of STAT transcription factors including STAT1, STAT3, and STAT5a/b [104,105], which translocate to the nucleus and activate regulated genes. These gene products can then interfere with apoptotic processes [56,106]. The family of mammalian STAT transcription factors regulates diverse functions implicated in developmental and homeostatic processes including apoptosis, growth, migration, proliferation, and differentiation [107,108].
In particular, STAT5, which mediates cellular responses to cytokines, growth factors, and hormones [109], has been implicated in EPO-stimulated erythropoiesis as well as protective mechanisms in various non-hematopoietic mammalian tissues including the nervous system [110]. It has been implicated in the control of neuronal cell fate decisions such as differentiation, proliferation, and apoptosis. Notably, EPO-mediated activation of JAK2/STAT5 leads to up-regulation of the anti-apoptotic Bcl-XL and Bcl-2 genes, thereby protecting proerythroblasts from apoptosis [111,112].

3.3. NF-κB

NF-κB consists of homo- and hetero-dimers of five members of the Rel family: NF-κB1 (p50/p105), NF-κB2 (p52/p100), RelA (p65), RelB (I-REL), and c-Rel [113]. NF-κB can be activated by the phosphorylation of a tyrosine residue of the p50 subunit which then translocates into the nucleus after the release of IκB, and is known to be crucial factor in the differentiation of neuronal cells [114,115]. As NF-κB is known to be a downstream regulator of tumor necrosis factor (TNF)-α, it is particularly important in the neuroinflammatory processes involved in depression [116]. Given its major role in mediating inflammatory processes, many researchers have suggested that NF-κB is not only present in various immune cells but also on the surface of the BBB [117]. In the brain, proinflammatory cytokines activate both neuronal and non-neuronal cells (e.g., microglia, astrocytes, and oligodendroglia) via the NF-κB cascade in a similar manner to that occurring in the peripheral inflammatory response [118]. NF-κB activation is regulated by glucocorticoids which inhibit NF-κB activity, and decreases the activation of some pro-inflammatory cytokines in turn [119]. NF-κB is crucial for mediating the stress-induced inhibition of neurogenesis and at least some depressive behavior [120]. EPO-related production of forebrain neural stem cells (NSCs) [121] and prevention of neuronal apoptosis [122,123] require activation of NF-κB.

3.4. PI3K/Akt

PI3K/Akt signaling has been identified as the major transduction pathway for EPO-mediated cell protection in various mammalian non-hematopoietic tissues [124,125]. Previous studies have used the PI3K inhibitor LY294002 to prevent Akt phosphorylation and abrogate the protective effects of EPO [126,127,128]. The PI3K/Akt signal transduction pathway is known to play an important role in regulating major cellular processes, such as cell growth [129], proliferation and survival [130], cell metabolism, and autophagy [131]. There is also evidence that EPO can promote axonal growth and branching via activation of the PI3K/Akt pathway in polarized hippocampal neurons [132,133]. However other scientists who have studied mammalian neuroprotective and neuroregenerative signal transduction pathways for their contribution to rhEPO-mediated neuroprotection in locust brain neurons have demonstrated an involvement of JAK and STAT, but not of PI3K, in beneficial mechanisms that interfere with apoptotic processes [110]. This finding suggests that the pathways affected by EPO and its derivatives may be slightly different across species.
EPO markedly enhances the oxidative stress-sensitive activity of Akt and prevents the activation of microglia, which was one of the most important cellular components of neuroinflammation [134,135,136]. Since the inhibition of Akt phosphorylation blocks the cellular protective effects induced by EPO, Akt activity appears to be vital for EPO-mediated tissue protection [137]. Akt can also inactivate Bad, a member of pro-apoptotic Bcl-2 family, through phosphorylation of its serine residues [138]. EPO is linked to Bad through the anti-apoptotic Bcl-2 family member Bcl-xL. Studies have suggested that EPO was able to prevent cellular injury through maintaining the expression of Bcl-2 and Bcl-xL and altering the Bcl:Bax ratio [139]. EPO-induced activation of Akt also activated by phosphorylation of endothelial nitric oxide synthase (eNOS) [140]. Interestingly, Akt can significantly increase NF-κB and HIF-1 activation, resulting in increased EPO expression [141].

3.5. ERK/MAPK

The mitogen-activated proteins kinases (MAPKs) are a family of evolutionarily conserved molecules which play crucial roles in cellular signaling pathways and gene expression, consisting of three major members: Extracellular signal-regulated kinase (ERK), p38, and c-Jun N-terminal kinase [142], which represent three different intracellular-signaling cascades. Phosphorylation activates MAPKs, which transduce a broad range of extracellular stimuli into various intracellular responses through both transcriptional and non-transcriptional regulation [143]. Initiation of the ERK/MAPK cascade involves activation of three kinases, Ras, Raf, and MAPK/ERK kinase [144], and the ERK/MAPK pathway is customarily thought to play important roles in cell proliferation and differentiation [145]. Long-lasting activation of MAPK activity is known as a key mediator of cell differentiation [146], invariably involving translocation of ERK from the cytoplasm to the nucleus [147].
It is interesting to note that the MAPK family is involved in differentiation of neuronal cells [148,149] and astrocytes [150], and has been indicated to produce EPO, stimulating neuron or oligodendrocyte differentiation and accelerating the proliferation of astrocytes. MAPKs enter the nucleus and induce transcription of target genes involved mainly with inhibition of apoptosis and cell proliferation. In contrast with erythroid cell types, the EPO-mediated signaling pathways are less well characterized for non-erythroid tissues and, therefore, limited information on the mechanisms underlying the EPO-induced antidepressant effects is available.

4. EPO in the Treatment of Depression

The relationship of EPO and depression has been investigated in a number of studies. According to a systematic review, beneficial effects of EPO on hippocampus-dependent memory function and on depression-relevant behavior were observed in some animal and clinical studies, thus highlighting EPO as a candidate agent to manage cognitive dysfunction and mood symptoms in the future [151]. In some animal studies, EPO treatment can have some antidepressant effects, discriminable both morphologically and behaviorally [152]. Various behavioral tests, such as the forced swim test, novelty-induced hypophagia (NIH) test and novel object recognition test, proved useful in demonstrating improved cognitive function in rodent models (rat and mouse) following treatment with EPO [153]. Another study indicated that there was no effect on memory and depression- or anxiety-like behaviors three days after a single administration of EPO, but there was improvement of spatial and object recognition memory [154]. In additive, EPO in the brain can be induced by electroconvulsive seizures (ECS) and independently exhibits antidepressant-like effects according to the forced swim and NIH tests. Finally, analysis for gene expression profiles revealed that EPO alters the expression of neurotrophic genes such as BDNF [155,156].
In humans, beneficial effects of EPO on cognitive functions have been recognized as early as around the time of introduction of EPO for the clinical treatment of renal anemia [157,158,159]. In healthy volunteers, a single high dose of EPO reduced the neuronal response to fear one week after administration without evoking any erythropoietic alterations [160]. Following EPO administration, along with increased neural and cognitive processing of facial expressions, a short-term effect of improved mood symptoms was reported in the first three days, which is similar to the neuro-behavioral effects obtained in acute administration of selective serotonin reuptake inhibitor (SSRI) antidepressants [161]. Three days post-administration of EPO also showed decreased neural responses to negative vs. positive pictures in a network of sites including the hippocampus [162]. However, another study showed that ARA290, an EPO-derived peptide, tended to lower the recognition of facial expressions of happiness and disgust and had no effects on mood and affective symptoms [163]. A double-blind study comprising 19 patients with acute depression provided evidence that EPO was found to reduce left amygdala-hippocampal response to fearful stimuli [164]. Some randomized controlled studies indicated EPO may provide a therapeutic option for patients with mood disorders [164,165,166,167]. Recently, clinical evidence of the procognitive potential of EPO has been proved by a randomized controlled trial (RCT) involving moderately depressed patients with treatment-resistant depression (N = 40) [168]. EPO also down-regulated plasma BDNF levels in patients with treatment-resistant depression (TRD; N = 40) [169]. Taken together, these seven clinical findings (Table 1) suggested that EPO may provide a therapeutic option for patients with depression. Nonetheless, all of these clinical studies have some limitations, such as the small sample sizes of patients, concurrent use of antidepressant medications in many patients, incomplete examination of mood and arousal changes, and unclear baseline data.
The mechanism of depression is very complex. A shrinkage of the hippocampal volume in depressive patients [170] and a decreased number of astrocytes and neurons in the prefrontal cortex and striatum [171] associated with depression were observed in some clinical and experimental studies. Scientists have examined and advanced the theory that depression is an inflammatory disorder [172,173] and is related to neuroprogression, especially in the hippocampus [174]. Indeed, one hypothesis that has recently gained traction suggests that depression is caused by a breakdown in neural plasticity arising from on-going inflammatory processes and an overactive stress-response system [175,176], which leads to structural and functional abnormalities in the fronto-limbic brain circuitry [177]. Based on the pharmacological effects of EPO in the nervous system, it might, for example, attenuate neuroinflammatory processes [178], and improve hippocampal neurogenesis [179]. Consistent with the above theory of depression, there is reason to believe that EPO pathways could be a potential target for the treatment of depression.

4.1. Inflammation in EPO-Related Treatments

A recent meta-analysis study indicated that depression is characterized by increased levels of IL-6 and TNF-α in plasma, which is interpreted to indicate that depression may be considered as an inflammatory disorder [180]. It was also shown that systems related to the mitigation of the inflammatory response may be potential therapeutic targets for mood disorders [181]. Non-steroidal anti-inflammatory drugs (NSAIDs), such as acetylsalicylic acid and celecoxib, have an adjunctive effect when combined with SSRIs in the treatment of clinical and experimental depression [182].
EPO is considered to have potential anti-inflammatory capacities, especially as evidenced by its successful application in a number of animal models of chronic inflammation [183,184]. EPO impairs the formation of pro-inflammatory factors such as TNF-α, IL-6, IL-12/IL-23 subunits and nitric oxide (NO) via induction of inducible NO synthase (iNOS) in macrophages [185]. It also has anti-inflammatory effects by reducing reactive astrocytosis and microglia activation and the number of immune cells in the injured sites [186,187]. The mechanisms of these anti-inflammatory effects have not been investigated widely; however, a recent report showed that inhibition of the NF-κB p65 subunit is likely to be essential [188].

4.2. Neuroprogression in EPO-Related Treatment

Neuroprogression is defined as the progression of neurodegeneration, apoptosis, and reduced neurogenesis, and structural, functional, molecular, and cellular modifications and neuronal plasticity; together, these phenomena most likely result from inflammation and other factors [189]. Depressive disorder is related to some structural brain changes, such as decreased hippocampal volume [190], which might come from a stressors-caused decrease in neurogenesis [191,192].
EPO has been implicated in the accommodation of neuroprogression and may play an important antidepressant role in the progress of depression. According to some experiments, EPO improved antidepressant and anti-anxiety-like effects in the forced swim test, which related to significantly increased hippocampal neurogenesis [193]; however, no evidence of a general EPO-related increase in mobility was observed in the open field test [155]. Systemically administered EPO crosses the BBB in therapeutically effective concentrations [194] and exerts neuroprotective and neurotrophic effects [195] in traumatic, hypoxic-ischemic, excitotoxic, and inflammatory brain damage [196,197], and in neurodegenerative and neuropsychiatric conditions [198,199,200]. These morphological effects of EPO are caused by direct action on neurons through EPO-EPOR pathways and are strongly correlated with brain-derived neurotrophic factor (BDNF), which plays a crucial role in neuronal survival and proliferation [201]. BDNF and EPO share a common set of intracellular signaling pathways including the PI3K and MAPK cascades [202,203,204]. EPO was reported to induce BDNF expression, inducing potential neuroplastic effects [154].

5. Conclusions and Perspective

Depression is a global issue and the leading cause of burden and disability worldwide. This very complex psychosocial and biological phenomenon contains intricate neurophysiological, behavioral, psychosocial, and affective constituents. The underlying mechanisms of depression have been difficult to illuminate because of the heterogeneous nature and the different etiologies of the disease. One potentially valuable theory states that not only the alterations in the volume of the hippocampus, prefrontal cortex, thalamus, and basal ganglia, but also inflammatory conditions are related to the causative mechanisms of depression. These findings suggest that targeting several pathophysiologic mechanisms rather than neurotransmitter systems specifically holds promise for developing innovative therapeutic strategies.
During the past decade, accumulating evidence has indicated that EPO may have potential as a treatment for depression, suggesting that endogenous cytokines may play an important role in the pathogenesis of depression. In addition to the kidney, liver, and uterus, many organs have been identified as secretory tissues for EPO, including the brain. The EPOR is expressed in cerebral and spinal cord neurons, and in the hypothalamus, hippocampus, neocortex, dorsal root ganglia and nerve axons. The discovery of EPO and EPOR in the nervous system highlights new functions for EPO, which was only considered to play a crucial role in the progress of erythroid differentiation. Mounting experimental evidence suggests that EPO treatment, which has clearly shown antidepressant and procognitive effects, may alleviate inflammation and neuroprogression in depression models. Therefore, exploring EPO-EPOR and their downstream signaling pathways may greatly improve our understanding of the pathogenic mechanisms that underlie depression and the molecular basis of its treatment. EPO-EPOR signaling through JAK2 activates multiple downstream signaling pathways including STAT5, PI3K/Akt, NF-κB, and MAPK. These factors may play an important role in inflammation and neuroprogression in the nervous system, particularly in the hippocampus, which is heavily involved in the development of depression. Unfortunately, to date there have been few reports about the relationship between EPO and depression. Thus, the role of the EPO-EPOR pathway in the treatment of depression makes it an attractive target for the development of new approaches to treating depression. This will also help to identify new targets for pharmacological intervention.
However, with the numerous effects attributed to EPO, several aspects of potential EPO treatment for depression must be clarified.
First, in the coming years, it will be crucial to evaluate the level of the expression of EPO or EPOR and to inhibit their downstream effectors to unequivocally define the role of EPO-EPOR pathways in depression processes. EPO antibodies and EPOR antagonists should be used in studies of the relationship between depression and EPO-EPOR pathways. Further research is necessary to determine the exact role of EPO-EPOR pathways in the progress of depression, including activation of EPO and specific subtypes of its signal transduction. In fact, this will likely be an active area of research for many years to come. As has been reported, derivatives such as carbamylated erythropoietin (CEPO) and asialo-EPO may also have neuroprotective functions. These derivatives need to be examined in greater detail. Development of new therapeutics to treat depression provides significant evidence of our new understanding of its homeostasis and pathophysiological features.
Second, EPOR is widely expressed in several tissues, including the muscle, liver, heart, and spinal cord, where it might be involved in physiological and pathophysiological processes, including tissue protection and immunomodulation. As we know, depression is associated with many neurological disorders and other chronic physical health conditions, such as diabetes, chronic liver disease, heart disease, and cardiovascular disease. It is, therefore, possible that studying EPO-EPOR pathways will help to explain the connection between depression and other disabilities.
Third, it is unclear whether the EPO-triggered signaling cascades differ across tissue, whether one cell type expresses both types of EPORs, and how these types of EPORs might differentially affect EPO-induced cellular and intracellular pathways and effects. Whether some other cytokines such as interleukin-3 (IL-3), granulocyte-macrophage colony-stimulating factor (GM-CSF) or IL-5 can signal through the EPOR-βcR complex, interfere with, or modify EPO signaling pathways remains to be unknown. In the future, animal models, such as gene knock-in and knock-outs of EPOR or βcR should be developed to study these issues.
Finally, despite the promising evidence for EPO as an additional treatment for mood disorders, it is important to acknowledge one major limitation of EPO. The hematopoietic action of EPO with repeated administration would necessitate close monitoring of hematocrit and thrombocyte levels and, potentially, blood-letting in non-anemic patient populations. Some studies show that exogenous EPO-treated cancer patients have been associated with tumor progression [205,206,207,208], which suggests the potential risk of the use of EPO. However, with further research discerning between the mechanisms of its antidepressant and hematopoietic effects, we may be able to develop EPO derivatives with antidepressant effects that lack a hematopoietic function.
In conclusion, research to identify compounds and therapeutic strategies targeting EPO pathways may be essential for the treatment of depression. Furthermore, extensive clinical trials are required to evaluate more effective and safer drugs related to EPO pathways in humans.

Acknowledgments

The authors thank Elsevier Language Services for providing language assistance and for proofreading the manuscript. This work was supported by the National Basic Research Program of China (973 Program,Number:2011CB505100) and the Classical Prescription Basic Research Team of the Beijing University of Chinese Medicine.

Author Contributions

Chongyang Ma, Fafeng Cheng, Xueqian Wang, Changming Zhai, Wenchao Yue, Yajun Lian, Qingguo Wang contributed to writing the review.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

EPOErythropoietin
EPORErythropoietin receptor
βcR/CD131β-common receptor
5-HT5-hydroxytryptamine
BDNFBrain-derived neutrophic factor
ECsEndothelial cells
BBBBlood brain barrier
GM-CSFgranulocyte-macrophage colony-stimulating factor
IL-3Interleukin 3
IL-5Interleukin 5
EphB4Ephrin-type B receptor 4
RhEPORecombinant human EPO
CEPOCarbamylated erythropoietin
HIFHypoxia-inducible factor
ROSReactive oxygen species
JAK2Janus kinase 2
PI3KPhosphoinositide 3-kinase
NF-κBNuclear factor kappa B
STAT5Transducer and activator of transcription 5
AktProtein Ser/Thr kinase
MAPKP42/44 mitogen-activated protein kinase
SSRISelective serotonin reuptake inhibitor
NSAIDsNon-steroidal anti-inflammatory drugs
NONitric oxide
iNOSInducible NO synthase

References

  1. Moussavi, S.; Chatterji, S.; Verdes, E.; Tandon, A.; Patel, V.; Ustun, B. Depression, chronic diseases, and decrements in health: Results from the World Health Surveys. Lancet 2007, 370, 851–858. [Google Scholar] [CrossRef]
  2. Maes, M.; Meltzer, H. The serotonin hypothesis of major depression. Psychopharmacol. Fourth Gener. Prog. 1995, 10, 933–934. [Google Scholar]
  3. Rush, A.J.; Trivedi, M.H.; Stewart, J.W.; Nierenberg, A.A.; Fava, M.; Kurian, B.T.; Warden, D.; Morris, D.W.; Luther, J.F.; Husain, M.M. Combining medications to enhance depression outcomes (CO-MED): Acute and long-term outcomes of a single-blind randomized study. Am. J. Psychiatry 2011, 168, 689–701. [Google Scholar] [CrossRef] [PubMed]
  4. John Rush, A.; Trivedi, M.H.; Wisniewski, S.R.; Nierenberg, A.A.; Stewart, J.W.; Warden, D.; Niederehe, G.; Thase, M.E.; Lavori, P.W.; Lebowitz, B.D. Acute and longer-term outcomes in depressed outpatients requiring one or several treatment steps: A STAR* D report. Am. J. Psychiatry 2006, 163, 1905. [Google Scholar] [CrossRef] [PubMed]
  5. Behnken, A.; Schöning, S.; Gerß, J.; Konrad, C.; de Jong-Meyer, R.; Zwanzger, P.; Arolt, V. Persistent non-verbal memory impairment in remitted major depression—Caused by encoding deficits? J. Affect. Disord. 2010, 122, 144–148. [Google Scholar] [CrossRef] [PubMed]
  6. Preiss, M.; Kucerova, H.; Lukavsky, J.; Stepankova, H.; Sos, P.; Kawaciukova, R. Cognitive deficits in the euthymic phase of unipolar depression. Psychiatry Res. 2009, 169, 235–239. [Google Scholar] [CrossRef] [PubMed]
  7. Sperner-Unterweger, B.; Kohl, C.; Fuchs, D. Immune changes and neurotransmitters: Possible interactions in depression? Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2014, 48, 268–276. [Google Scholar] [CrossRef] [PubMed]
  8. Sharma, A. Systems genomics support for immune and inflammation hypothesis of depression. Curr. Neuropharmacol. 2016, 14, 1. [Google Scholar] [CrossRef]
  9. Vaidya, V.A.; Duman, R.S. Depression–emerging insights from neurobiology. Br. Med. Bull. 2001, 57, 61–79. [Google Scholar] [CrossRef] [PubMed]
  10. Maes, M.; Leonard, B.; Fernandez, A.; Kubera, M.; Nowak, G.; Veerhuis, R.; Gardner, A.; Ruckoanich, P.; Geffard, M.; Altamura, C. (Neuro) inflammation and neuroprogression as new pathways and drug targets in depression: From antioxidants to kinase inhibitors. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2011, 35, 659–663. [Google Scholar] [CrossRef] [PubMed]
  11. Vaváková, M.; Ďuračková, Z.; Trebatická, J. Markers of oxidative stress and neuroprogression in depression disorder. Oxid. Med. Cell. Longev. 2015, 2015, 1–12. [Google Scholar] [CrossRef] [PubMed]
  12. Gałecki, P.; Talarowska, M.; Anderson, G.; Berk, M.; Maes, M. Mechanisms underlying neurocognitive dysfunctions in recurrent major depression. Med. Sci. Monit. Int. Med. J. Exp. Clin. Res. 2015, 21, 1535. [Google Scholar]
  13. Leonard, B.; Maes, M. Mechanistic explanations how cell-mediated immune activation, inflammation and oxidative and nitrosative stress pathways and their sequels and concomitants play a role in the pathophysiology of unipolar depression. Neurosci. Biobehav. Rev. 2012, 36, 764–785. [Google Scholar] [CrossRef] [PubMed]
  14. Anderson, G.; Berk, M.; Dean, O.; Moylan, S.; Maes, M. Role of immune-inflammatory and oxidative and nitrosative stress pathways in the etiology of depression: Therapeutic implications. CNS Drugs 2014, 28, 1–10. [Google Scholar] [CrossRef] [PubMed]
  15. Moylan, S.; Berk, M.; Dean, O.M.; Samuni, Y.; Williams, L.J.; O’Neil, A.; Hayley, A.C.; Pasco, J.A.; Anderson, G.; Jacka, F.N. Oxidative & nitrosative stress in depression: Why so much stress? Neurosci. Biobehav. Rev. 2014, 45, 46–62. [Google Scholar] [PubMed]
  16. Kim, Y.-K.; Na, K.-S.; Myint, A.-M.; Leonard, B.E. The role of pro-inflammatory cytokines in neuroinflammation, neurogenesis and the neuroendocrine system in major depression. Prog. Neuro Psychopharmacol. Biol. Psychiatry 2016, 64, 277–284. [Google Scholar] [CrossRef] [PubMed]
  17. Horowitz, M.A.; Zunszain, P.A. Neuroimmune and neuroendocrine abnormalities in depression: Two sides of the same coin. Ann. N. Y. Acad. Sci. 2015, 1351, 68–79. [Google Scholar] [CrossRef] [PubMed]
  18. Guan, X.T.; Lin, W.J.; Tang, M.M. Comparison of stress-induced and LPS-induced depressive-like behaviors and the alterations of central proinflammatory cytokines mRNA in rats. PsyCh J. 2015, 4, 113–122. [Google Scholar] [CrossRef] [PubMed]
  19. Reissmann, K.R. Studies on the mechanism of erythropoietic stimulation in parabiotic rats during hypoxia. Blood 1950, 5, 372–380. [Google Scholar] [PubMed]
  20. Plzak, L.; Fried, W.; Jacobson, L.; Bethard, W. Demonstration of stimulation of erythropoiesis by plasma from anemic rats using Fe59. J. Lab. Clin. Med. 1955, 46, 671–678. [Google Scholar] [PubMed]
  21. Nairz, M.; Sonnweber, T.; Schroll, A.; Theurl, I.; Weiss, G. The pleiotropic effects of erythropoietin in infection and inflammation. Microbes Infect. 2012, 14, 238–246. [Google Scholar] [CrossRef] [PubMed]
  22. Shaheen, M.; Broxmeyer, H. The humoral regulation of hematopoiesis. Hematol. Basic Princ. Pract. 2005, 5, 253–275. [Google Scholar]
  23. Buemi, M.; Cavallaro, E.; Floccari, F.; Sturiale, A.; Aloisi, C.; Trimarchi, M.; Grasso, G.; Corica, F.; Frisina, N. Erythropoietin and the brain: From neurodevelopment to neuroprotection. Clin. Sci. 2002, 103, 275–282. [Google Scholar] [CrossRef] [PubMed]
  24. Yuan, R.; Wang, B.; Lu, W.; Maeda, Y. A distinct region in erythropoietin that induces immuno/inflammatory modulation and tissue protection. Neurotherapeutics 2015, 12, 850–861. [Google Scholar] [CrossRef] [PubMed]
  25. Hand, C.C.; Brines, M. Promises and pitfalls in erythopoietin-mediated tissue protection: Are nonerythropoietic derivatives a way forward? J. Investig. Med. Off. Publ. Am. Fed. Clin. Res. 2011, 59, 1073. [Google Scholar]
  26. McGee, S.; Havens, A.; Shiozawa, Y.; Jung, Y.; Taichman, R. Effects of erythropoietin on the bone microenvironment. Growth Factors 2012, 30, 22–28. [Google Scholar] [CrossRef] [PubMed]
  27. Brines, M.; Cerami, A. Discovering erythropoietin’s extra-hematopoietic functions: Biology and clinical promise. Kidney Int. 2006, 70, 246–250. [Google Scholar] [CrossRef] [PubMed]
  28. Choi, D.; Schroer, S.A.; Lu, S.Y.; Wang, L.; Wu, X.; Liu, Y.; Zhang, Y.; Gaisano, H.Y.; Wagner, K.-U.; Wu, H. Erythropoietin protects against diabetes through direct effects on pancreatic β cells. J. Exp. Med. 2010, 207, 2831–2842. [Google Scholar] [CrossRef] [PubMed]
  29. Sytkowski, A.J. The neurobiology of erythropoietin. Cell. Mol. Neurobiol. 2011, 31, 931–937. [Google Scholar] [CrossRef] [PubMed]
  30. Masuda, S.; Chikuma, M.; Sasaki, R. Insulin-like growth factors and insulin stimulate erythropoietin production in primary cultured astrocytes. Brain Res. 1997, 746, 63–70. [Google Scholar] [CrossRef]
  31. Genc, S.; Kuralay, F.; Genc, K.; Akhisaroglu, M.; Fadiloglu, S.; Yorukoglu, K.; Fadiloğlu, M.; Gure, A. Erythropoietin exerts neuroprotection in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated C57/BL mice via increasing nitric oxide production. Neurosci. Lett. 2001, 298, 139–141. [Google Scholar] [CrossRef]
  32. Chu, H.; Ding, H.; Tang, Y.; Dong, Q. Erythropoietin protects against hemorrhagic blood–brain barrier disruption through the effects of aquaporin-4. Lab. Investig. 2014, 94, 1042–1053. [Google Scholar] [CrossRef] [PubMed]
  33. Brines, M.; Cerami, A. Erythropoietin and engineered innate repair activators. Methods Mol. Biol. 2013, 982, 1–11. [Google Scholar] [PubMed]
  34. Juul, S. Erythropoietin in the central nervous system, and its use to prevent hypoxic-ischemic brain damage. Acta Paediatr. 2002, 91, 36–42. [Google Scholar] [CrossRef]
  35. Chong, Z.; Kang, J.-Q.; Maiese, K. Erythropoietin: Cytoprotection in vascular and neuronal cells. Curr. Drug Targets-Cardiovasc. Hematol. Disord. 2003, 3, 141–154. [Google Scholar] [CrossRef]
  36. Genc, S.; Koroglu, T.F.; Genc, K. Erythropoietin as a novel neuroprotectant. Restor. Neurol. Neurosci. 2003, 22, 105–119. [Google Scholar]
  37. Marti, H.H. Erythropoietin and the hypoxic brain. J. Exp. Biol. 2004, 207, 3233–3242. [Google Scholar] [CrossRef] [PubMed]
  38. Masuda, S.; Okano, M.; Yamagishi, K.; Nagao, M.; Ueda, M.; Sasaki, R. A novel site of erythropoietin production. Oxygen-dependent production in cultured rat astrocytes. J. Biol. Chem. 1994, 269, 19488–19493. [Google Scholar] [PubMed]
  39. Lappin, T.R.; Maxwell, A.P.; Johnston, P.G. EPO’s alter ego: Erythropoietin has multiple actions. Stem Cells 2002, 20, 485–492. [Google Scholar] [CrossRef] [PubMed]
  40. Brines, M.L.; Ghezzi, P.; Keenan, S.; Agnello, D.; De Lanerolle, N.C.; Cerami, C.; Itri, L.M.; Cerami, A. Erythropoietin crosses the blood–brain barrier to protect against experimental brain injury. Proc. Natl. Acad. Sci. USA 2000, 97, 10526–10531. [Google Scholar] [CrossRef] [PubMed]
  41. Upadhyay, R.K. Drug delivery systems, CNS protection, and the blood brain barrier. BioMed Res. Int. 2014, 2014, 1–37. [Google Scholar] [CrossRef] [PubMed]
  42. Youssoufian, H.; Longmore, G.; Neumann, D.; Yoshimura, A.; Lodish, H. Structure, function, and activation of the erythropoietin receptor. Blood 1993, 81, 2223–2223. [Google Scholar] [PubMed]
  43. Anagnostou, A.; Liu, Z.; Steiner, M.; Chin, K.; Lee, E.S.; Kessimian, N.; Noguchi, C.T. Erythropoietin receptor mRNA expression in human endothelial cells. Proc. Natl. Acad. Sci. USA 1994, 91, 3974–3978. [Google Scholar] [CrossRef] [PubMed]
  44. Ogilvie, M.; Yu, X.; Nicolas-Metral, V.; Pulido, S.M.; Liu, C.; Ruegg, U.T.; Noguchi, C.T. Erythropoietin stimulates proliferation and interferes with differentiation of myoblasts. J. Biol. Chem. 2000, 275, 39754–39761. [Google Scholar] [CrossRef] [PubMed]
  45. Kuhrt, D.; Wojchowski, D.M. Emerging EPO and EPO receptor regulators, and signal transducers. Blood 2015, 125, 3536–3541. [Google Scholar] [CrossRef] [PubMed]
  46. Juul, S.E.; Anderson, D.K.; Li, Y.; Christensen, R.D. Erythropoietin and erythropoietin receptor in the developing human central nervous system. Pediatr. Res. 1998, 43, 40–49. [Google Scholar] [CrossRef] [PubMed]
  47. Li, X.; Gonias, S.L.; Campana, W.M. Schwann cells express erythropoietin receptor and represent a major target for Epo in peripheral nerve injury. Glia 2005, 51, 254–265. [Google Scholar] [CrossRef] [PubMed]
  48. Ott, C.; Martens, H.; Hassouna, I.; Oliveira, B.; Erck, C.; Zafeiriou, M.; Peteri, U.; Hesse, D.; Gerhart, S.; Altas, B. Widespread expression of erythropoietin receptor in brain and its induction by injury. Mol. Med. 2015. [Google Scholar] [CrossRef] [PubMed]
  49. Blake, T.J.; Jenkins, B.J.; D’Andrea, R.J.; Gonda, T.J. Functional cross-talk between cytokine receptors revealed by activating mutations in the extracellular domain of the β-subunit of the GM-CSF receptor. J. Leukoc. Biol. 2002, 72, 1246–1255. [Google Scholar] [PubMed]
  50. Bond, W.S.; Rex, T.S. Evidence that erythropoietin modulates neuroinflammation through differential action on neurons, astrocytes, and microglia. Front. Immunol. 2014, 5. [Google Scholar] [CrossRef] [PubMed]
  51. Lee, T.-S.; Lu, K.-Y.; Yu, Y.-B.; Lee, H.-T.; Tsai, F.-C. β Common receptor mediates erythropoietin-conferred protection on OxLDL-induced lipid accumulation and inflammation in macrophages. Med. Inflamm. 2015, 2015, 1–13. [Google Scholar]
  52. Elliott, S.; Sinclair, A.M. The effect of erythropoietin on normal and neoplastic cells. Biol. Targets Ther. 2012, 6, 163. [Google Scholar]
  53. Zhang, Y.; Wang, L.; Dey, S.; Alnaeeli, M.; Suresh, S.; Rogers, H.; Teng, R.; Noguchi, C.T. Erythropoietin action in stress response, tissue maintenance and metabolism. Int. J. Mol. Sci. 2014, 15, 10296–10333. [Google Scholar] [CrossRef] [PubMed]
  54. Leist, M.; Ghezzi, P.; Grasso, G.; Bianchi, R.; Villa, P.; Fratelli, M.; Savino, C.; Bianchi, M.; Nielsen, J.; Gerwien, J. Derivatives of erythropoietin that are tissue protective but not erythropoietic. Science 2004, 305, 239–242. [Google Scholar] [CrossRef] [PubMed]
  55. Dumont, F.; Bischoff, P. Non-erythropoietic tissue-protective peptides derived from erythropoietin: WO2009094172. Expert Opin. Ther. Pat. 2010, 20, 715–723. [Google Scholar] [CrossRef] [PubMed]
  56. Brines, M.; Cerami, A. Emerging biological roles for erythropoietin in the nervous system. Nat. Rev. Neurosci. 2005, 6, 484–494. [Google Scholar] [CrossRef] [PubMed]
  57. Brines, M.; Grasso, G.; Fiordaliso, F.; Sfacteria, A.; Ghezzi, P.; Fratelli, M.; Latini, R.; Xie, Q.-W.; Smart, J.; Su-Rick, C.-J. Erythropoietin mediates tissue protection through an erythropoietin and common β-subunit heteroreceptor. Proc. Natl. Acad. Sci. USA 2004, 101, 14907–14912. [Google Scholar] [CrossRef] [PubMed]
  58. Sanchez, P.E.; Navarro, F.P.; Fares, R.P.; Nadam, J.; Georges, B.; Moulin, C.; Le Cavorsin, M.; Bonnet, C.; Ryvlin, P.; Belmeguenai, A. Erythropoietin receptor expression is concordant with erythropoietin but not with common β chain expression in the rat brain throughout the life span. J. Comp. Neurol. 2009, 514, 403–414. [Google Scholar] [CrossRef] [PubMed]
  59. Um, M.; Lodish, H.F. Antiapoptotic effects of erythropoietin in differentiated neuroblastoma SH-SY5Y cells require activation of both the STAT5 and AKT signaling pathways. J. Biol. Chem. 2006, 281, 5648–5656. [Google Scholar] [CrossRef] [PubMed]
  60. Nadam, J.; Navarro, F.; Sanchez, P.; Moulin, C.; Georges, B.; Laglaine, A.; Pequignot, J.-M.; Morales, A.; Ryvlin, P.; Bezin, L. Neuroprotective effects of erythropoietin in the rat hippocampus after pilocarpine-induced status epilepticus. Neurobiol. Dis. 2007, 25, 412–426. [Google Scholar] [CrossRef] [PubMed]
  61. Pradeep, S.; Huang, J.; Mora, E.M.; Nick, A.M.; Cho, M.S.; Wu, S.Y.; Noh, K.; Pecot, C.V.; Rupaimoole, R.; Stein, M.A. Erythropoietin stimulates tumor growth via EphB4. Cancer Cell 2015, 28, 610–622. [Google Scholar] [CrossRef] [PubMed]
  62. Chen, T.; Liu, X.; Yi, S.; Zhang, J.; Ge, J.; Liu, Z. EphB4 is overexpressed in gliomas and promotes the growth of glioma cells. Tumor Biol. 2013, 34, 379–385. [Google Scholar] [CrossRef] [PubMed]
  63. Zhu, L.; Qian, L.; Wang, S.; Wang, T.; Jiang, L. Expression of ephrinB2 and EphB4 in a neonatal rat model of periventricular white matter damage. J. Perinat. Med. 2015, 43, 367–371. [Google Scholar] [CrossRef] [PubMed]
  64. Ashton, R.S.; Conway, A.; Pangarkar, C.; Bergen, J.; Lim, K.-I.; Shah, P.; Bissell, M.; Schaffer, D.V. Astrocytes regulate adult hippocampal neurogenesis through ephrin-B signaling. Nat. Neurosci. 2012, 15, 1399–1406. [Google Scholar] [CrossRef] [PubMed]
  65. Uhlén, M.; Fagerberg, L.; Hallström, B.M.; Lindskog, C.; Oksvold, P.; Mardinoglu, A.; Sivertsson, Å.; Kampf, C.; Sjöstedt, E.; Asplund, A. Tissue-based map of the human proteome. Science 2015, 347, 1260419. [Google Scholar] [CrossRef] [PubMed]
  66. Noguchi, C.T.; Wang, L.; Rogers, H.M.; Teng, R.; Jia, Y. Survival and proliferative roles of erythropoietin beyond the erythroid lineage. Expert Rev. Mol. Med. 2008, 10, e36. [Google Scholar] [CrossRef] [PubMed]
  67. Cataldi, A. Cell responses to oxidative stressors. Curr. Pharm. Des. 2010, 16, 1387–1395. [Google Scholar] [CrossRef] [PubMed]
  68. Chu, H.X.; Jones, N.M. Changes in Hypoxia-Inducible Factor-1 (HIF-1) and Regulatory Prolyl Hydroxylase (PHD) Enzymes Following Hypoxic–Ischemic Injury in the Neonatal Rat. Neurochem. Res. 2015, 41, 515–522. [Google Scholar] [CrossRef] [PubMed]
  69. Heidbreder, M.; Fröhlich, F.; Jöhren, O.; Dendorfer, A.; Qadri, F.; Dominiak, P. Hypoxia rapidly activates HIF-3α mRNA expression. FASEB J. 2003, 17, 1541–1543. [Google Scholar] [PubMed]
  70. Kaelin, W.G.; Ratcliffe, P.J. Oxygen sensing by metazoans: The central role of the HIF hydroxylase pathway. Mol. Cell 2008, 30, 393–402. [Google Scholar] [CrossRef] [PubMed]
  71. Marzo, F.; Lavorgna, A.; Coluzzi, G.; Santucci, E.; Tarantino, F.; Rio, T.; Conti, E.; Autore, C.; Agati, L.; Andreotti, F. Erythropoietin in heart and vessels: Focus on transcription and signaling pathways. J. Thromb. Thrombolysis 2008, 26, 183–187. [Google Scholar] [CrossRef] [PubMed]
  72. Obara, N.; Imagawa, S.; Nakano, Y.; Suzuki, N.; Yamamoto, M.; Nagasawa, T. Suppression of erythropoietin gene expression by cadmium depends on inhibition of HIF-1, not stimulation of GATA-2. Arch. Toxicol. 2003, 77, 267–273. [Google Scholar] [PubMed]
  73. Obara, N.; Suzuki, N.; Kim, K.; Nagasawa, T.; Imagawa, S.; Yamamoto, M. Repression via the GATA box is essential for tissue-specific erythropoietin gene expression. Blood 2008, 111, 5223–5232. [Google Scholar] [CrossRef] [PubMed]
  74. Li, C.; Jiang, J.; Fan, Y.; Fu, G.; Wang, J.; Fan, W. Knockout of the tumor necrosis factor α receptor 1 gene can up-regulate erythropoietin receptor during myocardial ischemia-reperfusion injury in mice. Chin. Med. J. 2009, 122, 566–570. [Google Scholar] [PubMed]
  75. Erbaş, O.; Çınar, B.P.; Solmaz, V.; Çavuşoğlu, T.; Ateş, U. The neuroprotective effect of erythropoietin on experimental Parkinson model in rats. Neuropeptides 2015, 49, 1–5. [Google Scholar] [CrossRef] [PubMed]
  76. Nagai, A.; Nakagawa, E.; Choi, H.B.; Hatori, K.; Kobayashi, S.; Kim, S.U. Erythropoietin and erythropoietin receptors in human CNS neurons, astrocytes, microglia, and oligodendrocytes grown in culture. J. Neuropathol. Exp. Neurol. 2001, 60, 386–392. [Google Scholar] [CrossRef] [PubMed]
  77. Choi, M.; Son, H. Effects of serotonin on erythropoietin expression in mouse hippocampus. Exp. Neurobiol. 2013, 22, 45–50. [Google Scholar] [CrossRef] [PubMed]
  78. Cruz-Gomez, A.; Belenguer-Benavides, A.; Martinez-Bronchal, B.; Fittipaldi-Marquez, M.; Forn, C. Structural and functional changes of the hippocampus in patients with multiple sclerosis and their relationship with memory processes. Rev. Neurol. 2016, 62, 6–12. [Google Scholar] [PubMed]
  79. Dietrich, J.; Prust, M.; Kaiser, J. Chemotherapy, cognitive impairment and hippocampal toxicity. Neuroscience 2015, 309, 224–232. [Google Scholar] [CrossRef] [PubMed]
  80. Constantinescu, S.N.; Ghaffari, S.; Lodish, H.F. The erythropoietin receptor: Structure, activation and intracellular signal transduction. Trends Endocrinol. Metab. 1999, 10, 18–23. [Google Scholar] [CrossRef]
  81. Digicaylioglu, M.; Lipton, S.A. Erythropoietin-mediated neuroprotection involves cross-talk between Jak2 and NF-κB signaling cascades. Nature 2001, 412, 641–647. [Google Scholar] [CrossRef] [PubMed]
  82. Noguchi, C.T.; Asavaritikrai, P.; Teng, R.; Jia, Y. Role of erythropoietin in the brain. Crit. Rev. Oncol./Hematol. 2007, 64, 159–171. [Google Scholar] [CrossRef] [PubMed]
  83. Maxwell, P.; Melendez-Rodríguez, F.; Matchett, K.B.; Aragones, J.; Ben-Califa, N.; Jaekel, H.; Hengst, L.; Lindner, H.; Bernardini, A.; Brockmeier, U. Novel antibodies directed against the human erythropoietin receptor: Creating a basis for clinical implementation. Br. J. Haematol. 2015, 168, 429–442. [Google Scholar] [CrossRef] [PubMed]
  84. Carter-Su, C.; Schwartz, J.; Argetsinger, L.S. Growth Hormone Signaling Pathways. Growth Horm. IGF Res. 2015, 28, 11–15. [Google Scholar] [CrossRef] [PubMed]
  85. Li, B.; Zhang, G.; Li, R.; Duan, C. Phosphoinositide 3-kinase/Akt Pathway Mediates Fip1-like1-platelet-derived Growth Factor Receptor α-induced Cell Infiltration and Activation: Possible Molecular Mechanism for the Malignant Phenotype of Chronic Eosinophilic Leukemia. Cancer Transl. Med. 2015, 1, 31. [Google Scholar]
  86. van der Kooij, M.A.; Groenendaal, F.; Kavelaars, A.; Heijnen, C.J.; van Bel, F. Neuroprotective properties and mechanisms of erythropoietin in in vitro and in vivo experimental models for hypoxia/ischemia. Brain Res. Rev. 2008, 59, 22–33. [Google Scholar] [CrossRef] [PubMed]
  87. Kwon, M.-S.; Kim, M.-H.; Kim, S.-H.; Park, K.-D.; Yoo, S.-H.; Oh, I.-U.; Pak, S.; Seo, Y.-J. Erythropoietin exerts cell protective effect by activating PI3K/Akt and MAPK pathways in C6 Cells. Neurol. Res. 2014, 36, 215–223. [Google Scholar] [CrossRef] [PubMed]
  88. Serizawa, K.; Yogo, K.; Tashiro, Y.; Aizawa, K.; Kawasaki, R.; Hirata, M.; Endo, K. Epoetin beta pegol prevents endothelial dysfunction as evaluated by flow-mediated dilation in chronic kidney disease rats. Eur. J. Pharmacol. 2015, 767, 10–16. [Google Scholar] [CrossRef] [PubMed]
  89. Alliouachene, S.; Bilanges, B.; Chicanne, G.; Anderson, K.E.; Pearce, W.; Ali, K.; Valet, C.; Posor, Y.; Low, P.C.; Chaussade, C. Inactivation of the class II PI3K-C2β potentiates insulin signaling and sensitivity. Cell Rep. 2015, 13, 1881–1894. [Google Scholar] [CrossRef] [PubMed]
  90. Bouvard, C.; Galy-Fauroux, I.; Grelac, F.; Carpentier, W.; Lokajczyk, A.; Gandrille, S.; Colliec-Jouault, S.; Fischer, A.-M.; Helley, D. Low-Molecular-Weight Fucoidan Induces Endothelial Cell Migration via the PI3K/AKT Pathway and Modulates the Transcription of Genes Involved in Angiogenesis. Mar. Drugs 2015, 13, 7446–7462. [Google Scholar] [CrossRef] [PubMed]
  91. Hellweg, C.E. The nuclear factor κB pathway: A link to the immune system in the radiation response. Cancer Lett. 2015, 368, 275–289. [Google Scholar] [CrossRef] [PubMed]
  92. Lanzillotta, A.; Porrini, V.; Bellucci, A.; Benarese, M.; Branca, C.; Parrella, E.; Spano, P.F.; Pizzi, M. NF-κB in innate neuroprotection and age-related neurodegenerative diseases. Front. Neurol. 2015, 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Correia, C.; Lee, S.-H.; Meng, X.W.; Vincelette, N.D.; Knorr, K.L.; Ding, H.; Nowakowski, G.S.; Dai, H.; Kaufmann, S.H. Emerging understanding of Bcl-2 biology: Implications for neoplastic progression and treatment. Biochim. Biophys. Acta Mol. Cell Res. 2015, 1853, 1658–1671. [Google Scholar] [CrossRef] [PubMed]
  94. Schwartz, C.; Willebrand, R.; Huber, S.; Rupec, R.A.; Wu, D.; Locksley, R.; Voehringer, D. Eosinophil-specific deletion of IκBα in mice reveals a critical role of NF-κ-induced Bcl-xL for inhibition of apoptosis. Blood 2015, 125, 3896–3904. [Google Scholar] [CrossRef] [PubMed]
  95. Villa, P.; Bigini, P.; Mennini, T.; Agnello, D.; Laragione, T.; Cagnotto, A.; Viviani, B.; Marinovich, M.; Cerami, A.; Coleman, T.R. Erythropoietin selectively attenuates cytokine production and inflammation in cerebral ischemia by targeting neuronal apoptosis. J. Exp. Med. 2003, 198, 971–975. [Google Scholar] [CrossRef] [PubMed]
  96. Arcasoy, M.O.; Jiang, X.; Haroon, Z.A. Expression of erythropoietin receptor splice variants in human cancer. Biochem. Biophys. Res. Commun. 2003, 307, 999–1007. [Google Scholar] [CrossRef]
  97. Broxmeyer, H.E. Erythropoietin: Multiple targets, actions, and modifying influences for biological and clinical consideration. J. Exp. Med. 2013, 210, 205–208. [Google Scholar] [CrossRef] [PubMed]
  98. Haq, R.; Halupa, A.; Beattie, B.K.; Mason, J.M.; Zanke, B.W.; Barber, D.L. Regulation of erythropoietin-induced STAT serine phosphorylation by distinct mitogen-activated protein kinases. J. Biol. Chem. 2002, 277, 17359–17366. [Google Scholar] [CrossRef] [PubMed]
  99. Zou, H.; Yan, D.; Mohi, G. Differential biological activity of disease-associated JAK2 mutants. FEBS Lett. 2011, 585, 1007–1013. [Google Scholar] [CrossRef] [PubMed]
  100. Koulnis, M.; Porpiglia, E.; Hidalgo, D.; Socolovsky, M. Erythropoiesis: From Molecular Pathways to System Properties. In A Systems Biology Approach to Blood; Springer: New York, NY, USA, 2014; pp. 37–58. [Google Scholar]
  101. Li, K.; Cao, R.-J.; Zhu, X.-J.; Liu, X.-Y.; Li, L.-Y.; Cui, S.-S. Erythropoietin Attenuates the Apoptosis of Adult Neurons After Brachial Plexus Root Avulsion by Downregulating JNK Phosphorylation and c-Jun Expression and Inhibiting c-PARP Cleavage. J. Mol. Neurosci. 2015, 56, 917–925. [Google Scholar] [CrossRef] [PubMed]
  102. Tong, W.; Zhang, J.; Lodish, H.F. Lnk inhibits erythropoiesis and Epo-dependent JAK2 activation and downstream signaling pathways. Blood 2005, 105, 4604–4612. [Google Scholar] [CrossRef] [PubMed]
  103. Kong, D.; Zhuo, L.; Gao, C.; Shi, S.; Wang, N.; Huang, Z.; Li, W.; Hao, L. Erythropoietin protects against cisplatin-induced nephrotoxicity by attenuating endoplasmic reticulum stress-induced apoptosis. J. Nephrol. 2013, 26, 219–227. [Google Scholar] [CrossRef] [PubMed]
  104. Imagawa, S.; Yamamoto, M.; Miura, Y. Negative regulation of the erythropoietin gene expression by the GATA transcription factors. Blood 1997, 89, 1430–1439. [Google Scholar] [PubMed]
  105. Wojchowski, D.M.; Gregory, R.C.; Miller, C.P.; Pandit, A.K.; Pircher, T.J. Signal transduction in the erythropoietin receptor system. Exp. Cell Res. 1999, 253, 143–156. [Google Scholar] [CrossRef] [PubMed]
  106. Nguyen, A.Q.; Cherry, B.H.; Scott, G.F.; Ryou, M.-G.; Mallet, R.T. Erythropoietin: Powerful protection of ischemic and post-ischemic brain. Exp. Biol. Med. 2014, 239, 1461–1475. [Google Scholar] [CrossRef] [PubMed]
  107. Nicholson, S.E.; Murray, P.J. Regulation of Macrophage Polarization by the STAT–SOCS Signaling Axis. In Macrophages: Biology and Role in the Pathology of Diseases; Springer: New York, NY, USA, 2014; pp. 497–508. [Google Scholar]
  108. Rawlings, J.S.; Rosler, K.M.; Harrison, D.A. The JAK/STAT signaling pathway. J. Cell Sci. 2004, 117, 1281–1283. [Google Scholar] [CrossRef] [PubMed]
  109. Ihle, J.N. The Stat family in cytokine signaling. Curr. Opin. Cell Biol. 2001, 13, 211–217. [Google Scholar] [CrossRef]
  110. Miljus, N.; Heibeck, S.; Jarrar, M.; Micke, M.; Ostrowski, D.; Ehrenreich, H.; Heinrich, R. Erythropoietin-mediated protection of insect brain neurons involves JAK and STAT but not PI3K transduction pathways. Neuroscience 2014, 258, 218–227. [Google Scholar] [CrossRef] [PubMed]
  111. Silva, M.; Grillot, D.; Benito, A.; Richard, C.; Nunez, G.; Fernandez-Luna, J.L. Erythropoietin can promote erythroid progenitor survival by repressing apoptosis through Bcl-XL and Bcl-2. Blood 1996, 88, 1576–1582. [Google Scholar] [PubMed]
  112. Wei, L.; Han, B.H.; Li, Y.; Keogh, C.L.; Holtzman, D.M.; Yu, S.P. Cell death mechanism and protective effect of erythropoietin after focal ischemia in the whisker-barrel cortex of neonatal rats. J. Pharmacol. Exp. Ther. 2006, 317, 109–116. [Google Scholar] [CrossRef] [PubMed]
  113. Crampton, S.J.; O’keeffe, G.W. NF-κB: Emerging roles in hippocampal development and function. Int. J. Biochem. Cell Biol. 2013, 45, 1821–1824. [Google Scholar] [CrossRef] [PubMed]
  114. Lee, S.M.; Nga, N.T.H.; Park, M.H.; Kim, K.S.; Cho, K.J.; Moon, D.C.; Kim, H.Y.; Hong, J.T. EPO receptor-mediated ERK kinase and NF-κB activation in erythropoietin-promoted differentiation of astrocytes. Biochem. Biophys. Res. Commun. 2004, 320, 1087–1095. [Google Scholar] [CrossRef] [PubMed]
  115. Gutierrez, H.; Davies, A.M. Regulation of neural process growth, elaboration and structural plasticity by NF-κB. Trends Neurosci. 2011, 34, 316–325. [Google Scholar] [CrossRef] [PubMed]
  116. Wang, K.; Wu, H.; Chi, M.; Zhang, J.; Wang, G.; Li, H. Electroacupuncture inhibits apoptosis of splenic lymphocytes in traumatized rats through modulation of the TNF-α/NF-κB signaling pathway. Mol. Med. Rep. 2015, 11, 237–241. [Google Scholar] [PubMed]
  117. Coelho-Santos, V.; Leitão, R.A.; Cardoso, F.L.; Palmela, I.; Rito, M.; Barbosa, M.; Brito, M.A.; Fontes-Ribeiro, C.A.; Silva, A.P. The TNF-α/NF-κB signaling pathway has a key role in methamphetamine-induced blood-brain barrier dysfunction. J. Cereb. Blood Flow Metab. 2015, 35, 1260–1271. [Google Scholar] [CrossRef] [PubMed]
  118. Anisman, H.; Merali, Z.; Hayley, S. Neurotransmitter, peptide and cytokine processes in relation to depressive disorder: Comorbidity between depression and neurodegenerative disorders. Prog. Neurobiol. 2008, 85, 1–74. [Google Scholar] [CrossRef] [PubMed]
  119. Padgett, D.A.; Glaser, R. How stress influences the immune response. Trends Immunol. 2003, 24, 444–448. [Google Scholar] [CrossRef]
  120. Koo, J.W.; Russo, S.J.; Ferguson, D.; Nestler, E.J.; Duman, R.S. Nuclear factor-κB is a critical mediator of stress-impaired neurogenesis and depressive behavior. Proc. Natl. Acad. Sci. USA 2010, 107, 2669–2674. [Google Scholar] [CrossRef] [PubMed]
  121. Wang, L.; Zhang, Z.; Wang, Y.; Zhang, R.; Chopp, M. Treatment of stroke with erythropoietin enhances neurogenesis and angiogenesis and improves neurological function in rats. Stroke 2004, 35, 1732–1737. [Google Scholar] [CrossRef] [PubMed]
  122. Shingo, T.; Sorokan, S.T.; Shimazaki, T.; Weiss, S. Erythropoietin regulates the in vitro and in vivo production of neuronal progenitors by mammalian forebrain neural stem cells. J. Neurosci. 2001, 21, 9733–9743. [Google Scholar] [PubMed]
  123. Matsushita, H.; Johnston, M.V.; Lange, M.S.; Wilson, M.A. Protective effect of erythropoietin in neonatal hypoxic ischemia in mice. Neuroreport 2003, 14, 1757–1761. [Google Scholar] [CrossRef] [PubMed]
  124. Chateauvieux, S.; Grigorakaki, C.; Morceau, F.; Dicato, M.; Diederich, M. Erythropoietin, erythropoiesis and beyond. Biochem. Pharmacol. 2011, 82, 1291–1303. [Google Scholar] [CrossRef] [PubMed]
  125. Zhang, J.; Zou, Y.R.; Zhong, X.; Deng, H.D.F.; Pu, L.; Peng, K.; Wang, L. Erythropoietin pretreatment ameliorates renal ischaemia-reperfusion injury by activating PI3K/Akt signaling. Nephrology 2015, 20, 266–272. [Google Scholar] [CrossRef] [PubMed]
  126. Trincavelli, M.L.; da Pozzo, E.; Ciampi, O.; Cuboni, S.; Daniele, S.; Abbracchio, M.P.; Martini, C. Regulation of erythropoietin receptor activity in endothelial cells by different erythropoietin (EPO) derivatives: An in vitro study. Int. J. Mol. Sci. 2013, 14, 2258–2281. [Google Scholar] [CrossRef] [PubMed]
  127. Miki, T.; Miura, T.; Yano, T.; Takahashi, A.; Sakamoto, J.; Tanno, M.; Kobayashi, H.; Ikeda, Y.; Nishihara, M.; Naitoh, K. Alteration in erythropoietin-induced cardioprotective signaling by postinfarct ventricular remodeling. J. Pharmacol. Exp. Ther. 2006, 317, 68–75. [Google Scholar] [CrossRef] [PubMed]
  128. Jia, Y.; Mo, S.-J.; Feng, Q.-Q.; Zhan, M.-L.; OuYang, L.-S.; Chen, J.-C.; Ma, Y.-X.; Wu, J.-J.; Lei, W.-L. EPO-dependent activation of PI3K/Akt/FoxO3a signaling mediates neuroprotection in in vitro and in vivo models of Parkinson’s disease. J. Mol. Neurosci. 2014, 53, 117–124. [Google Scholar] [CrossRef] [PubMed]
  129. Fingar, D.C.; Salama, S.; Tsou, C.; Harlow, E.; Blenis, J. Mammalian cell size is controlled by mTOR and its downstream targets S6K1 and 4EBP1/eIF4E. Genes Dev. 2002, 16, 1472–1487. [Google Scholar] [CrossRef] [PubMed]
  130. Fingar, D.C.; Blenis, J. Target of rapamycin (TOR): An integrator of nutrient and growth factor signals and coordinator of cell growth and cell cycle progression. Oncogene 2004, 23, 3151–3171. [Google Scholar] [CrossRef] [PubMed]
  131. Kumar, D.; Shankar, S.; Srivastava, R.K. Rottlerin induces autophagy and apoptosis in prostate cancer stem cells via PI3K/Akt/mTOR signaling pathway. Cancer Lett. 2014, 343, 179–189. [Google Scholar] [CrossRef] [PubMed]
  132. Ransome, M.I.; Turnley, A.M. Erythropoietin promotes axonal growth in a model of neuronal polarization. Mol. Cell. Neurosci. 2008, 38, 537–547. [Google Scholar] [CrossRef] [PubMed]
  133. Wang, T.; Yuan, W.; Liu, Y.; Zhang, Y.; Wang, Z.; Zhou, X.; Ning, G.; Zhang, L.; Yao, L.; Feng, S. The role of the JAK-STAT pathway in neural stem cells, neural progenitor cells and reactive astrocytes after spinal cord injury (Review). Biomed. Rep. 2015, 3, 141–146. [Google Scholar] [PubMed]
  134. Chong, Z.Z.; Kang, J.-Q.; 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, 320–330. [Google Scholar] [CrossRef] [PubMed]
  135. Chong, Z.Z.; Lin, S.H.; Kang, J.Q.; 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, 659–669. [Google Scholar] [CrossRef] [PubMed]
  136. Chong, Z.Z.; Kang, J.Q.; 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, 1107–1118. [Google Scholar] [CrossRef] [PubMed]
  137. Sirén, A.-L.; Faßhauer, T.; Bartels, C.; Ehrenreich, H. Therapeutic potential of erythropoietin and its structural or functional variants in the nervous system. Neurotherapeutics 2009, 6, 108–127. [Google Scholar] [CrossRef] [PubMed]
  138. Singh, S.S.; Yap, W.N.; Arfuso, F.; Kar, S.; Wang, C.; Cai, W.; Dharmarajan, A.M.; Sethi, G.; Kumar, A.P. Targeting the PI3K/Akt signaling pathway in gastric carcinoma: A reality for personalized medicine? World J. Gastroenterol. 2015, 21, 12261. [Google Scholar] [CrossRef] [PubMed]
  139. Vairano, M.; Russo, C.D.; Pozzoli, G.; Battaglia, A.; Scambia, G.; Tringali, G.; Aloe-Spiriti, M.A.; Preziosi, P.; Navarra, P. Erythropoietin exerts anti-apoptotic effects on rat microglial cells in vitro. Eur. J. Neurosci. 2002, 16, 584–592. [Google Scholar] [CrossRef] [PubMed]
  140. Urao, N.; Okigaki, M.; Yamada, H.; Aadachi, Y.; Matsuno, K.; Matsui, A.; Matsunaga, S.; Tateishi, K.; Nomura, T.; Takahashi, T. Erythropoietin-mobilized endothelial progenitors enhance reendothelialization via Akt-endothelial nitric oxide synthase activation and prevent neointimal hyperplasia. Circ. Res. 2006, 98, 1405–1413. [Google Scholar] [CrossRef] [PubMed]
  141. Figueroa, Y.G.; Chan, A.K.; Ibrahim, R.; Tang, Y.; Burow, M.E.; Alam, J.; Scandurro, A.B.; Beckman, B.S. NF-κB plays a key role in hypoxia-inducible factor-1-regulated erythropoietin gene expression. Exp. Hematol. 2002, 30, 1419–1427. [Google Scholar] [CrossRef]
  142. Honda, T.; Kataoka, T.R.; Ueshima, C.; Miyachi, Y.; Kabashima, K. A Case of Noonan Syndrome with Multiple Subcutaneous Tumours with MAPK-ERK/p38 Activation. Acta Derm. Venereol. 2016, 96, 130–131. [Google Scholar] [CrossRef] [PubMed]
  143. Qi, C.; Xu, M.; Gan, J.; Yang, X.; Wu, N.; Song, L.; Yuan, W.; Liu, Z. Erythropoietin improves neurobehavior by reducing dopaminergic neuron loss in a 6-hydroxydopamine-induced rat model. Int. J. Mol. Med. 2014, 34, 440–450. [Google Scholar] [PubMed]
  144. New, L.; Li, Y.; Ge, B.; Zhong, H.; Mansbridge, J.; Liu, K.; Han, J. SB203580 promote EGF-stimulated early morphological differentiation in PC12 cell through activating ERK pathway. J. Cell. Biochem. 2001, 83, 585–596. [Google Scholar] [CrossRef] [PubMed]
  145. Strack, S. Overexpression of the protein phosphatase 2A regulatory subunit Bγ promotes neuronal differentiation by activating the MAP kinase (MAPK) cascade. J. Biol. Chem. 2002, 277, 41525–41532. [Google Scholar] [CrossRef] [PubMed]
  146. Robinson, M.J.; Stippec, S.A.; Goldsmith, E.; White, M.A.; Cobb, M.H. A constitutively active and nuclear form of the MAP kinase ERK2 is sufficient for neurite outgrowth and cell transformation. Curr. Biol. 1998, 8, 1141–1152. [Google Scholar] [CrossRef]
  147. Rosenblum, K.; Futter, M.; Voss, K.; Erent, M.; Skehel, P.A.; French, P.; Obosi, L.; Jones, M.W.; Bliss, T.V. The role of extracellular regulated kinases I/II in late-phase long-term potentiation. J. Neurosci. 2002, 22, 5432–5441. [Google Scholar]
  148. Jung, K.M.; Park, K.S.; Oh, J.H.; Jung, S.Y.; Yang, K.H.; Song, Y.S.; Son, D.J.; Park, Y.H.; Yun, Y.P.; Lee, M.K. Activation of p38 mitogen-activated protein kinase and activator protein-1 during the promotion of neurite extension of PC-12 cells by 15-deoxy-Δ12, 14-prostaglandin J2. Mol. Pharmacol. 2003, 63, 607–616. [Google Scholar] [CrossRef] [PubMed]
  149. Park, K.S.; Da Lee, R.; Kang, S.-K.; Han, S.Y.; Park, K.L.; Yang, K.H.; Song, Y.S.; Park, H.J.; Lee, Y.M.; Yun, Y.P. Neuronal differentiation of embryonic midbrain cells by upregulation of peroxisome proliferator-activated receptor-γ via the JNK-dependent pathway. Exp. Cell Res. 2004, 297, 424–433. [Google Scholar] [CrossRef] [PubMed]
  150. Sugawa, M.; Sakurai, Y.; Ishikawa-Ieda, Y.; Suzuki, H.; Asou, H. Effects of erythropoietin on glial cell development; oligodendrocyte maturation and astrocyte proliferation. Neurosci. Res. 2002, 44, 391–403. [Google Scholar] [CrossRef]
  151. Miskowiak, K.W.; Vinberg, M.; Harmer, C.J.; Ehrenreich, H.; Kessing, L.V. Erythropoietin: A candidate treatment for mood symptoms and memory dysfunction in depression. Psychopharmacology 2012, 219, 687–698. [Google Scholar] [CrossRef] [PubMed]
  152. Miu, A.C.; Olteanu, A.I.; Chiş, I.; Heilman, R.M. Have no fear, erythropoietin is here: Erythropoietin protects fear conditioning performances after functional inactivation of the amygdala. Behav. Brain Res. 2004, 155, 223–229. [Google Scholar] [CrossRef] [PubMed]
  153. Duman, C.H.; Newton, S.S. Evaluating effects of EPO in rodent behavioral assays related to depression. Methods Mol. Biol. 2013, 127–140. [Google Scholar] [CrossRef]
  154. Leconte, C.; Bihel, E.; Lepelletier, F.-X.; Bouët, V.; Saulnier, R.; Petit, E.; Boulouard, M.; Bernaudin, M.; Schumann-Bard, P. Comparison of the effects of erythropoietin and its carbamylated derivative on behavior and hippocampal neurogenesis in mice. Neuropharmacology 2011, 60, 354–364. [Google Scholar] [CrossRef] [PubMed]
  155. Girgenti, M.J.; Hunsberger, J.; Duman, C.H.; Sathyanesan, M.; Terwilliger, R.; Newton, S.S. Erythropoietin induction by electroconvulsive seizure, gene regulation, and antidepressant-like behavioral effects. Biol. Psychiatry 2009, 66, 267–274. [Google Scholar] [CrossRef] [PubMed]
  156. Viviani, B.; Bartesaghi, S.; Corsini, E.; Villa, P.; Ghezzi, P.; Garau, A.; Galli, C.L.; Marinovich, M. Erythropoietin protects primary hippocampal neurons increasing the expression of brain-derived neurotrophic factor. J. Neurochem. 2005, 93, 412–421. [Google Scholar] [CrossRef] [PubMed]
  157. Jelkmann, W. Erythropoietin: Structure, control of production, and function. Physiol Rev. 1992, 72, 449–489. [Google Scholar] [PubMed]
  158. Kramer, L.; Madl, C.; Stockenhuber, F.; Yeganehfar, W.; Eisenhuber, E.; Derfler, K.; Lenz, K.; Schneider, B.; Grimm, G. Beneficial effect of renal transplantation on cognitive brain function. Kidney Int. 1996, 49, 833–838. [Google Scholar] [CrossRef] [PubMed]
  159. Pickett, J.L.; Theberge, D.C.; Brown, W.S.; Schweitzer, S.U.; Nissenson, A.R. Normalizing hematocrit in dialysis patients improves brain function. Am. J. Kidney Dis. 1999, 33, 1122–1130. [Google Scholar] [CrossRef]
  160. Miskowiak, K.; O’sullivan, U.; Harmer, C.J. Erythropoietin reduces neural and cognitive processing of fear in human models of antidepressant drug action. Biol. Psychiatry 2007, 62, 1244–1250. [Google Scholar] [CrossRef] [PubMed]
  161. Miskowiak, K.; Inkster, B.; Selvaraj, S.; Wise, R.; Goodwin, G.M.; Harmer, C.J. Erythropoietin improves mood and modulates the cognitive and neural processing of emotion 3 days post administration. Neuropsychopharmacology 2008, 33, 611–618. [Google Scholar] [CrossRef] [PubMed]
  162. Miskowiak, K.W.; Favaron, E.; Hafizi, S.; Inkster, B.; Goodwin, G.M.; Cowen, P.J.; Harmer, C.J. Effects of erythropoietin on emotional processing biases in patients with major depression: An exploratory fMRI study. Psychopharmacology 2009, 207, 133–142. [Google Scholar] [CrossRef] [PubMed]
  163. Cerit, H.; Veer, I.M.; Dahan, A.; Niesters, M.; Harmer, C.J.; Miskowiak, K.W.; Rombouts, S.A.; van der Does, W. Testing the antidepressant properties of the peptide ARA290 in a human neuropsychological model of drug action. Eur. Neuropsychopharmacol. 2015, 25, 2289–2299. [Google Scholar] [CrossRef] [PubMed]
  164. Miskowiak, K.W.; Favaron, E.; Hafizi, S.; Inkster, B.; Goodwin, G.M.; Cowen, P.J.; Harmer, C.J. Erythropoietin modulates neural and cognitive processing of emotional information in biomarker models of antidepressant drug action in depressed patients. Psychopharmacology 2010, 210, 419–428. [Google Scholar] [CrossRef] [PubMed]
  165. Falkai, P.; Schmitt, A. Erythropoietin as an innovative add-on therapy for depression. Biol. Psychiatry 2015, 78, 222. [Google Scholar] [CrossRef] [PubMed]
  166. Miskowiak, K.W.; Vinberg, M.; Macoveanu, J.; Ehrenreich, H.; Køster, N.; Inkster, B.; Paulson, O.B.; Kessing, L.V.; Skimminge, A.; Siebner, H.R. Effects of erythropoietin on hippocampal volume and memory in mood disorders. Biol. Psychiatry 2015, 78, 270–277. [Google Scholar] [CrossRef] [PubMed]
  167. Miskowiak, K.W.; Vinberg, M.; Harmer, C.J.; Ehrenreich, H.; Knudsen, G.M.; Macoveanu, J.; Hansen, A.R.; Paulson, O.B.; Siebner, H.R.; Kessing, L.V. Effects of erythropoietin on depressive symptoms and neurocognitive deficits in depression and bipolar disorder. Trials 2010, 11, 97. [Google Scholar] [PubMed]
  168. Miskowiak, K.W.; Vinberg, M.; Christensen, E.M.; Bukh, J.D.; Harmer, C.J.; Ehrenreich, H.; Kessing, L.V. Recombinant human erythropoietin for treating treatment-resistant depression: A double-blind, randomized, placebo-controlled phase 2 trial. Neuropsychopharmacology 2014, 39, 1399. [Google Scholar] [CrossRef] [PubMed]
  169. Vinberg, M.; Miskowiak, K.; Hoejman, P.; Pedersen, M.; Kessing, L.V. The effect of recombinant erythropoietin on plasma brain derived neurotrophic factor levels in patients with affective disorders: A randomised controlled study. PLoS ONE 2015, 10, e0127629. [Google Scholar] [CrossRef] [PubMed]
  170. Ongür, D.; Drevets, W.C.; Price, J.L. Glial reduction in the subgenual prefrontal cortex in mood disorders. Proc. Natl. Acad. Sci. USA 1998, 95, 13290–13295. [Google Scholar] [CrossRef] [PubMed]
  171. Rajkowska, G.; Miguel-Hidalgo, J.J.; Wei, J.; Dilley, G.; Pittman, S.D.; Meltzer, H.Y.; Overholser, J.C.; Roth, B.L.; Stockmeier, C.A. Morphometric evidence for neuronal and glial prefrontal cell pathology in major depression. Biol. Psychiatry 1999, 45, 1085–1098. [Google Scholar] [CrossRef]
  172. Maes, M.; Meltzer, H.Y.; Bosmans, E.; Bergmans, R.; Vandoolaeghe, E.; Ranjan, R.; Desnyder, R. Increased plasma concentrations of interleukin-6, soluble interleukin-6, soluble interleukin-2 and transferrin receptor in major depression. J. Affect. Disord. 1995, 34, 301–309. [Google Scholar] [CrossRef]
  173. Maes, M. Depression is an inflammatory disease, but cell-mediated immune activation is the key component of depression. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2011, 35, 664–675. [Google Scholar] [CrossRef] [PubMed]
  174. Maes, M.; Galecki, P.; Chang, Y.S.; Berk, M. A review on the oxidative and nitrosative stress (O&NS) pathways in major depression and their possible contribution to the (neuro) degenerative processes in that illness. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2011, 35, 676–692. [Google Scholar]
  175. Duman, R.S.; Monteggia, L.M. A neurotrophic model for stress-related mood disorders. Biol. Psychiatry 2006, 59, 1116–1127. [Google Scholar] [CrossRef] [PubMed]
  176. Miller, A.H.; Maletic, V.; Raison, C.L. Inflammation and its discontents: The role of cytokines in the pathophysiology of major depression. Biol. Psychiatry 2009, 65, 732–741. [Google Scholar] [CrossRef] [PubMed]
  177. Berton, O.; Nestler, E.J. New approaches to antidepressant drug discovery: Beyond monoamines. Nat. Rev. Neurosci. 2006, 7, 137–151. [Google Scholar] [CrossRef] [PubMed]
  178. Markoulaki, D.; Kostikas, K.; Papatheodorou, G.; Koutsokera, A.; Alchanatis, M.; Bakakos, P.; Gourgoulianis, K.I.; Roussos, C.; Koulouris, N.G.; Loukides, S. Hemoglobin, erythropoietin and systemic inflammation in exacerbations of chronic obstructive pulmonary disease. Eur. J. Intern. Med. 2011, 22, 103–107. [Google Scholar] [CrossRef] [PubMed]
  179. Xiong, Y.; Mahmood, A.; Zhang, Y.; Meng, Y.; Zhang, Z.G.; Qu, C.; Sager, T.N.; Chopp, M. Effects of posttraumatic carbamylated erythropoietin therapy on reducing lesion volume and hippocampal cell loss, enhancing angiogenesis and neurogenesis, and improving functional outcome in rats following traumatic brain injury. J. Neurosurg. 2011, 114, 549. [Google Scholar] [CrossRef] [PubMed]
  180. Dowlati, Y.; Herrmann, N.; Swardfager, W.; Liu, H.; Sham, L.; Reim, E.K.; Lanctôt, K.L. A meta-analysis of cytokines in major depression. Biol. Psychiatry 2010, 67, 446–457. [Google Scholar] [CrossRef] [PubMed]
  181. Furtado, M.; Katzman, M.A. Neuroinflammatory pathways in anxiety, posttraumatic stress, and obsessive compulsive disorders. Psychiatry Res. 2015, 229, 37–48. [Google Scholar] [CrossRef] [PubMed]
  182. Wang, Y.; Yang, F.; Liu, Y.-F.; Gao, F.; Jiang, W. Acetylsalicylic acid as an augmentation agent in fluoxetine treatment resistant depressive rats. Neurosci. Lett. 2011, 499, 74–79. [Google Scholar] [CrossRef] [PubMed]
  183. Meng, R.; Zhu, D.; Bi, Y.; Yang, D.; Wang, Y. Erythropoietin inhibits gluconeogenesis and inflammation in the liver and improves glucose intolerance in high-fat diet-fed mice. PLoS ONE 2013, 8, e53557. [Google Scholar] [CrossRef] [PubMed]
  184. Yuan, R.; Maeda, Y.; Li, W.; Lu, W.; Cook, S.; Dowling, P. Erythropoietin: A potent inducer of peripheral immuno/inflammatory modulation in autoimmune EAE. PLoS ONE 2008, 3, e1924. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Alnaeeli, M.; Noguchi, C.T. Erythropoietin and obesity-induced white adipose tissue inflammation: Redefining the boundaries of the immunometabolism territory. Adipocyte 2015, 4, 153–157. [Google Scholar] [CrossRef] [PubMed]
  186. Sun, Y.; Calvert, J.W.; Zhang, J.H. Neonatal hypoxia/ischemia is associated with decreased inflammatory mediators after erythropoietin administration. Stroke 2005, 36, 1672–1678. [Google Scholar] [CrossRef] [PubMed]
  187. Xiong, Y.; Mahmood, A.; Qu, C.; Kazmi, H.; Zhang, Z.G.; Noguchi, C.T.; Schallert, T.; Chopp, M. Erythropoietin improves histological and functional outcomes after traumatic brain injury in mice in the absence of the neural erythropoietin receptor. J. Neurotrauma 2010, 27, 205–215. [Google Scholar] [CrossRef] [PubMed]
  188. Nairz, M.; Schroll, A.; Moschen, A.R.; Sonnweber, T.; Theurl, M.; Theurl, I.; Taub, N.; Jamnig, C.; Neurauter, D.; Huber, L.A. Erythropoietin contrastingly affects bacterial infection and experimental colitis by inhibiting nuclear factor-κB-inducible immune pathways. Immunity 2011, 34, 61–74. [Google Scholar] [CrossRef] [PubMed]
  189. Cardoso, T.; Bauer, I.E.; Meyer, T.D.; Kapczinski, F.; Soares, J.C. Neuroprogression and cognitive functioning in bipolar disorder: A systematic review. Curr. Psychiatry Rep. 2015, 17, 1–24. [Google Scholar] [CrossRef] [PubMed]
  190. Campbell, S.; Marriott, M.; Nahmias, C.; MacQueen, G.M. Lower hippocampal volume in patients suffering from depression: A meta-analysis. Am. J. Psychiatry 2004, 161, 598–607. [Google Scholar] [CrossRef] [PubMed]
  191. Mcewen, B.S. Allostasis and allostatic load: Implications for neuropsychopharmacology. Neuropsychopharmacology 2000, 22, 108–124. [Google Scholar] [CrossRef]
  192. Dranovsky, A.; Hen, R. Hippocampal neurogenesis: Regulation by stress and antidepressants. Biol. Psychiatry 2006, 59, 1136–1143. [Google Scholar] [CrossRef] [PubMed]
  193. Osborn, M.; Rustom, N.; Clarke, M.; Litteljohn, D.; Rudyk, C.; Anisman, H.; Hayley, S. Antidepressant-like effects of erythropoietin: A focus on behavioral and hippocampal processes. PLoS ONE 2013, 8, e72813. [Google Scholar] [CrossRef] [PubMed]
  194. Ehrenreich, H.; Aust, C.; Krampe, H.; Jahn, H.; Jacob, S.; Herrmann, M.; Sirén, A.-L. Erythropoietin: Novel approaches to neuroprotection in human brain disease. Metab. Brain Dis. 2004, 19, 195–206. [Google Scholar] [CrossRef] [PubMed]
  195. Matsuyama, T.; Tanaka, T.; Tatsumi, K.; Daijo, H.; Kai, S.; Harada, H.; Fukuda, K. Midazolam inhibits the hypoxia-induced up-regulation of erythropoietin in the central nervous system. Eur. J. Pharmacol. 2015, 761, 189–198. [Google Scholar] [CrossRef] [PubMed]
  196. Wu, Q.; Chen, W.; Sinha, B.; Tu, Y.; Manning, S.; Thomas, N.; Zhou, S.; Jiang, H.; Ma, H.; Kroessler, D.A. Neuroprotective agents for neonatal hypoxic-ischemic brain injury. Drug Discov. Today 2015, 20, 1372–1381. [Google Scholar] [CrossRef] [PubMed]
  197. Wu, Y.W.; Gonzalez, F.F. Erythropoietin: A novel therapy for hypoxic–ischaemic encephalopathy? Dev. Med. Child Neurol. 2015, 57, 34–39. [Google Scholar] [CrossRef] [PubMed]
  198. Agnello, D.; Bigini, P.; Villa, P.; Mennini, T.; Cerami, A.; Brines, M.L.; Ghezzi, P. Erythropoietin exerts an anti-inflammatory effect on the CNS in a model of experimental autoimmune encephalomyelitis. Brain Res. 2002, 952, 128–134. [Google Scholar] [CrossRef]
  199. Sättler, M.; Merkler, D.; Maier, K.; Stadelmann, C.; Ehrenreich, H.; Bähr, M.; Diem, R. Neuroprotective effects and intracellular signaling pathways of erythropoietin in a rat model of multiple sclerosis. Cell Death Differ. 2004, 11, S181–S192. [Google Scholar] [CrossRef] [PubMed]
  200. Li, W.; Maeda, Y.; Yuan, R.R.; Elkabes, S.; Cook, S.; Dowling, P. Beneficial effect of erythropoietin on experimental allergic encephalomyelitis. Ann. Neurol. 2004, 56, 767–777. [Google Scholar] [CrossRef] [PubMed]
  201. Berk, M.; Kapczinski, F.; Andreazza, A.; Dean, O.; Giorlando, F.; Maes, M.; Yücel, M.; Gama, C.; Dodd, S.; Dean, B. Pathways underlying neuroprogression in bipolar disorder: Focus on inflammation, oxidative stress and neurotrophic factors. Neurosci. Biobehav. Rev. 2011, 35, 804–817. [Google Scholar] [CrossRef] [PubMed]
  202. Luo, L.; Liu, X.-L.; Li, J.; Mu, R.-H.; Liu, Q.; Yi, L.-T.; Geng, D. Macranthol promotes hippocampal neuronal proliferation in mice via BDNF–TrkB–PI3K/Akt signaling pathway. Eur. J. Pharmacol. 2015, 762, 357–363. [Google Scholar] [CrossRef] [PubMed]
  203. Mao, X.-Y.; Cao, Y.-G.; Ji, Z.; Zhou, H.-H.; Liu, Z.-Q.; Sun, H.-L. Topiramate protects against glutamate excitotoxicity via activating BDNF/TrkB-dependent ERK pathway in rodent hippocampal neurons. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2015, 60, 11–17. [Google Scholar] [CrossRef] [PubMed]
  204. Criscuolo, C.; Fabiani, C.; Bonadonna, C.; Origlia, N.; Domenici, L. BDNF prevents amyloid-dependent impairment of LTP in the entorhinal cortex by attenuating p38 MAPK phosphorylation. Neurobiol. Aging 2015, 36, 1303–1309. [Google Scholar] [CrossRef] [PubMed]
  205. Kumar, S.M.; Zhang, G.; Bastian, B.C.; Arcasoy, M.O.; Karande, P.; Pushparajan, A.; Acs, G.; Xu, X. Erythropoietin receptor contributes to melanoma cell survival in vivo. Oncogene 2012, 31, 1649–1660. [Google Scholar] [CrossRef] [PubMed]
  206. Wang, L.; Li, H.-G.; Xia, Z.-S.; Wen, J.-M.; Lv, J. Prognostic significance of erythropoietin and erythropoietin receptor in gastric adenocarcinoma. World J. Gastroenterol. 2011, 17, 3933–3940. [Google Scholar] [CrossRef] [PubMed]
  207. Rózsás, A.; Berta, J.; Rojkó, L.; Horváth, L.Z.; Keszthelyi, M.; Kenessey, I.; László, V.; Berger, W.; Grusch, M.; Hoda, M.A. Erythropoietin receptor expression is a potential prognostic factor in human lung adenocarcinoma. PLoS ONE 2013, 8, e77459. [Google Scholar]
  208. Fuge, F.; Doleschel, D.; Rix, A.; Gremse, F.; Wessner, A.; Winz, O.; Mottaghy, F.; Lederle, W.; Kiessling, F. In-vivo detection of the erythropoietin receptor in tumours using positron emission tomography. Eur. Radiol. 2015, 25, 472–479. [Google Scholar] [CrossRef] [PubMed]
Figure 1. EPO-induced intracellular signaling pathways Erythropoietin and its receptor signaling through JAK2 activate multiple downstream signaling molecules including STAT5, PI3K/Akt, NF-κB, and MAPK. These factors may play an important role in inflammation and neuroprogression in the nervous system.
Figure 1. EPO-induced intracellular signaling pathways Erythropoietin and its receptor signaling through JAK2 activate multiple downstream signaling molecules including STAT5, PI3K/Akt, NF-κB, and MAPK. These factors may play an important role in inflammation and neuroprogression in the nervous system.
Ijms 17 00677 g001
Table 1. Clinical study on the antidepressant effect of EPO.
Table 1. Clinical study on the antidepressant effect of EPO.
AuthorEPO FormSubjectDrug AdministrationRandomizedDouble-BlindMain FindingLimitationsSafety
Kamilla W. Miskowiak et al. (2008) [161]Erythropoietin (40,000 International Unit, IU)healthy volunteers (N = 24)injection onceYYDuring faces processing EPO enhanced activation in the left amygdala and right precuneus to happy and fearful expressions. This was paired with improved recognition of all facial expressions, in particular of low intensity happiness and fear. This is similar to behavioral effects observed with acute administration of serotonergic antidepressants.1, pharmacological fMRI studies in general is the possibility that drug effects on neural response may be confounded by non-specific effects on neural coupling and cerebral hemodynamics. 2, more detailed examination of the mood and arousal changes seen following EPO and their relation to changes in emotional processing observed three days post-administration should be performed. 3, the clinical effect in patients suffering from depression is unknown.Blood pressure and subjective state were monitored for 2 h following the injection.
Hilâl Cerit et al. (2008) [163]ARA290 (2 mg)healthy volunteers (N = 36)injection, onceYYARA290-treated individuals displayed lower neural responses to happy faces in the fusiform gyrus. ARA290 tended to lower the recognition of happy and disgust facial expressions. Although ARA290 was not associated with a better memory for positive words, it was associated with faster categorization of positive vs. negative words. Finally, ARA290 increased attention towards positive emotional pictures. No effects were observed on mood and affective symptoms.1, the limited clinical potential of EPO to treat depressive symptoms in non-anemic patients, due to the hematopoietic actions of EPO with repeated administration. 2, the human proof-of-concept studies were conducted in relatively small samples.After administration, the participant was monitored for 10 min. Dose selection was based on previous studies in humans in which no safety concerns were reported.
Kamilla W. Miskowiak et al. (2009) [162]Erythropoietin (40,000 IU)depressed patients (N = 17)injection onceYYEPO reduced neural response to negative vs. positive pictures three days post-administration in a network of areas including the hippocampus, ventromedial prefrontal and parietal cortex. After the scan, EPO-treated patients showed improved memory compared with those that were given placebo. The effects occurred in the absence of changes in mood or hematological parameters, suggesting that they originated from direct neurobiological actions of EPO.1, an exploratory study in a small patient sample. 2, The majority of patientswere also taking antidepressant medicationBlood pressure, well-being and subjective state was monitored for 2 h following the injection.
Kamilla W. Miskowiak et al. (2010) [164]Erythropoietin(40,000 IU) depressed patients (N = 19)injection onceYYEPO reduced neural response to fearful vs. happy faces in the amygdala and hippocampus, and to fearful faces vs. baseline in superior temporal and occipitoparietal regions three days after administration in acutely depressed patients. This was accompanied by a specific reduction in the recognition of fear in EPO-treated patients after the scan similar to the effects on face recognition seen with antidepressant drug treatment.1, an exploratory study in a small patient sample. 2, the majority of patients were taking antidepressant medication. 3, the current study used a between-groups design, and it is unknown whether baseline differences existed between the two groups. 4, the application of EPO in the treatment of neuropsychiatric disorder may have some undesirable effects.Following EPO/saline administration, blood pressure, well-being, and subjective state were monitored for 2 h.
Kamilla W. Miskowiak et al. (2014) [168]Erythropoietin(40,000 IU)depressed patients (N = 40)injection weekly (8 weeks)YYHDRS-17, GAF, and remission rates showed no effects of EPO over saline at week 9. However, EPO improved BDI and WHOQOL-BREF, and this was maintained at follow-up week 14. EPO enhanced verbal recall and recognition, which was sustained at follow-up. Exploratory analysis in patients fulfilling depression severity criteria at trial start revealed ameliorated HDRS-17 in EPO vs. saline groups, which was sustained at week 14. Exploratory analysis in the complete cohort showed that EPO reduced depression composite at weeks 9 and 14.1, the extensive exclusion criteria may limit the ability to generalize our findings to clinical practice. 2, the EPO-associated increase in red blood cell levels could confound the interpretation of the effects of EPO as neural in origin. 3, they did not screen for or exclude co-morbid axis II disorder as this would have resulted in a subsample of patients who were not representative for the target population of treatment-resistant patients. 4, their study may not have been adequately powered to detect a significant effect on primary outcome measure, although a positive signal was apparent on the additional depression-relevant outcomes and explorative score of depressive syndrome severity. 5, patients had been treated for years without any improvement, and that a treatment period of eight weeks is very short in such chronic, recurrent condition.Weekly monitoring of blood tests and any side effects was performed by a physician not involved in outcome measure assessments.
Kamilla W. Miskowiak et al. (2015) [166]Erythropoietin(40,000 IU)BD/TRD patients (N = 69 )injection weekly (8 weeks)YYCompared with saline, EPO was associated with mood-independent memory improvement and reversal of brain matter loss in the left hippocampalcornu ammonis 1 to cornu ammonis 3 and subiculum. Using the entire sample, memory improvement was associated with subfield hippocampal volume increase independent of mood change.1, their cohort included both patients with TRD and BD, since these mood disorders may involve differential, although partially overlapping, pathogenic processes. 2, three complementary methods to capture different aspects of hippocampal volume changes have their own limitations, and reflect different structural measures.Blood tests were taken on a weekly basis from baseline to week 10 (two weeks after treatment completion) and again in week 14.
Maj Vinberg et al. (2015) [169]Erythropoietin(40,000 IU)BD/TRD patients (N = 83 )injection weekly (8 weeks)YYEPO down-regulated plasma BDNF levels in patients with TRD (mean reduction at week 9 (95% CI): EPO 10.94 ng/L (4.51–21.41 ng/L); mean increase at week 9: Saline 0.52 ng/L, p = 0.04 (−5.88–4.48 ng/L) p = 0.04, partial η2 = 0.12). No significant effects were found on BDNF levels in partially remitted patients with BD (p = 0.35).1, they did not register daily physical exercise level, and since EPO is well known for its potential doping capacity, the change in BDNF levels could be due to increased exercise levels in the intervention group. 2, the relatively few participants is a limitation. 3, patients received weekly intravenous infusions of either EPO or saline for eight weeks (weeks 1–8) in addition to their current antidepressant medication.Blood tests were taken on a weekly basis from baseline to week 10.

Share and Cite

MDPI and ACS Style

Ma, C.; Cheng, F.; Wang, X.; Zhai, C.; Yue, W.; Lian, Y.; Wang, Q. Erythropoietin Pathway: A Potential Target for the Treatment of Depression. Int. J. Mol. Sci. 2016, 17, 677. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms17050677

AMA Style

Ma C, Cheng F, Wang X, Zhai C, Yue W, Lian Y, Wang Q. Erythropoietin Pathway: A Potential Target for the Treatment of Depression. International Journal of Molecular Sciences. 2016; 17(5):677. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms17050677

Chicago/Turabian Style

Ma, Chongyang, Fafeng Cheng, Xueqian Wang, Changming Zhai, Wenchao Yue, Yajun Lian, and Qingguo Wang. 2016. "Erythropoietin Pathway: A Potential Target for the Treatment of Depression" International Journal of Molecular Sciences 17, no. 5: 677. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms17050677

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop