Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Synapse ; 78(1): e22282, 2024 01.
Article in English | MEDLINE | ID: mdl-37794768

ABSTRACT

Memory and learning allow animals to appropriate certain properties of nature with which they can navigate in it successfully. Memory is acquired slowly and consists of two major phases, a fragile early phase (short-term memory, <4 h) and a more robust and long-lasting late one (long-term memory, >4 h). Erythropoietin (EPO) prolongs memory from 24 to 72 h when animals are trained for 5 min in a place recognition task but not when training lasted 3 min (short-term memory). It is not known whether it promotes the formation of remote memory (≥21 days). We address whether the systemic administration of EPO can convert a short-term memory into a long-term remote memory, and the neural plasticity mechanisms involved. We evaluated the effect of training duration (3 or 5 min) on the expression of endogenous EPO and its receptor to shed light on the role of EPO in coordinating mechanisms of neural plasticity using a single-trial spatial learning test. We administered EPO 10 min post-training and evaluated memory after 24 h, 96 h, 15 days, or 21 days. We also determined the effect of EPO administered 10 min after training on the expression of arc and bdnf during retrieval at 24 h and 21 days. Data show that learning induces EPO/EPOr expression increase linked to memory extent, exogenous EPO prolongs memory up to 21 days; and prefrontal cortex bdnf expression at 24 h and in the hippocampus at 21 days, whereas arc expression increases at 21 days in the hippocampus and prefrontal cortex.


Subject(s)
Erythropoietin , Memory Consolidation , Animals , Brain-Derived Neurotrophic Factor/metabolism , Erythropoietin/pharmacology , Erythropoietin/metabolism , Receptors, Erythropoietin/metabolism , Brain/metabolism , Hippocampus/metabolism , Memory, Long-Term
2.
Exp Brain Res ; 240(5): 1303-1315, 2022 May.
Article in English | MEDLINE | ID: mdl-35234993

ABSTRACT

Erythropoietin is a cytokine that binds to the Erythropoietin receptor and regulates the formation of erythroid cells during erythropoiesis in the bone marrow. However, many other organs and tissues express Erythropoietin and its receptor, such as the Nervous System, which principally regulates tissue protection. In the Central Nervous System, Erythropoietin is principally expressed by astrocytes, while neurons mainly express Erythropoietin receptors. Moreover, Erythropoietin acts as a pleiotropic molecule with neuroprotective effects, and its mechanisms of signal transduction pathways are defined, and there is a growing interest in its therapeutic potential. This review focuses on the role of Erythropoietin and its relationship with HIF1, PI3/Akt, GSK3B, JAK/STAT, and MAPKs signaling pathways that leads to cell survival after injury in the Central Nervous System. Knowledge of these signaling systems comprehensively could better guide EPO treatment to restoring different SNC alterations mediated by different insults.


Subject(s)
Erythropoietin , Neuroprotective Agents , Erythropoietin/metabolism , Erythropoietin/pharmacology , Humans , Neuroprotection , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Receptors, Erythropoietin/metabolism , Signal Transduction
3.
J Neurochem ; 158(5): 1032-1057, 2021 09.
Article in English | MEDLINE | ID: mdl-34278579

ABSTRACT

Erythropoietin (Epo), the main erythropoiesis-stimulating factor widely prescribed to overcome anemia, is also known nowadays for its cytoprotective action on non-hematopoietic tissues. In this context, Epo showed not only its ability to cross the blood-brain barrier, but also its expression in the brain of mammals. In clinical trials, recombinant Epo treatment has been shown to stimulate neurogenesis; improve cognition; and activate antiapoptotic, antioxidant, and anti-inflammatory signaling pathways. These mechanisms, proposed to characterize a neuroprotective property, opened new perspectives on the Epo pharmacological potencies. However, many questions arise about a possible physiological role of Epo in the central nervous system (CNS) and the factors or environmental conditions that induce its expression. Although Epo may be considered a strong candidate to be used against neuronal damage, long-term treatments, particularly when high Epo doses are needed, may induce thromboembolic complications associated with increases in hematocrit and blood viscosity. To avoid these adverse effects, different Epo analogs without erythropoietic activity but maintaining neuroprotection ability are currently being investigated. Carbamylated erythropoietin, as well as alternative molecules like Epo fusion proteins and partial peptides of Epo, seems to match this profile. This review will focus on the discussion of experimental evidence reported in recent years linking erythropoietin and CNS function through investigations aimed at finding benefits in the treatment of neurodegenerative diseases. In addition, it will review the proposed mechanisms for novel derivatives which may clarify and, eventually, improve the neuroprotective action of Epo.


Subject(s)
Brain/metabolism , Erythropoietin/metabolism , Neurodegenerative Diseases/metabolism , Neuroprotection/physiology , Receptors, Erythropoietin/metabolism , Animals , Brain/drug effects , Erythropoietin/administration & dosage , Humans , Neurodegenerative Diseases/therapy , Neuroprotection/drug effects
4.
Mediators Inflamm ; 2020: 8937657, 2020.
Article in English | MEDLINE | ID: mdl-32184703

ABSTRACT

Chronic kidney disease (CKD) causes anemia by renal damage. In CKD, the kidney is submitted to hypoxia, persistent inflammation, leading to fibrosis and permanent loss of renal function. Human recombinant erythropoietin (rEPO) has been widely used to treat CKD-associated anemia and is known to possess organ-protective properties that are independent from its well-established hematopoietic effects. Nonhematopoietic effects of EPO are mediated by an alternative receptor that is proposed to consist of a heterocomplex between the erythropoietin receptor (EPOR) and the beta common receptor (ßcR). The present study explored the effects of rEPO to prevent renal fibrosis in adenine-induced chronic kidney disease (Ad-CKD) and their association with the expression of the heterodimer EPOR/ßcR. Male Wistar rats were randomized to control group (CTL), adenine-fed rats (Ad-CKD), and Ad-CKD with treatment of rEPO (1050 IU/kg, once weekly for 4 weeks). Ad-CKD rats exhibited anemia, uremia, decreased renal function, increased infiltration of inflammatory cells, tubular atrophy, and fibrosis. rEPO treatment not only corrected anemia but reduced uremia and partially improved renal function as well. In addition, we observed that rEPO diminishes tubular injury, prevents fibrosis deposition, and induces the EPOR/ßcR heteroreceptor. The findings may explain the extrahematopoietic effects of rEPO in CKD and provide new strategies for the treatment of renal fibrosis in CKD.


Subject(s)
Fibrosis/metabolism , Fibrosis/prevention & control , Renal Insufficiency, Chronic/chemically induced , Renal Insufficiency, Chronic/drug therapy , Animals , Blotting, Western , Erythropoietin/therapeutic use , Fluorescent Antibody Technique , Humans , Immunoprecipitation , Male , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Receptors, Erythropoietin/metabolism , Recombinant Proteins/therapeutic use
5.
Curr Pharm Des ; 26(12): 1356-1364, 2020.
Article in English | MEDLINE | ID: mdl-32072891

ABSTRACT

The most important activity of erythropoietin (EPO) is the regulation of erythrocyte production by activation of the erythropoietin receptor (EPO-R), which triggers the activation of anti-apoptotic and proliferative responses of erythroid progenitor cells. Additionally, to erythropoietic EPO activity, an antiapoptotic effect has been described in a wide spectrum of tissues. EPO low levels are found in the central nervous system (CNS), while EPO-R is expressed in most CNS cell types. In spite of EPO-R high levels expressed during the hypoxicischemic brain, insufficient production of endogenous cerebral EPO could be the cause of determined circuit alterations that lead to the loss of specific neuronal populations. In the heart, high EPO-R expression in cardiac progenitor cells appears to contribute to myocardial regeneration under EPO stimulation. Several lines of evidence have linked EPO to an antiapoptotic role in CNS and in heart tissue. In this review, an antiapoptotic role of EPO/EPO-R system in both brain and heart under hypoxic conditions, such as epilepsy and sudden death (SUDEP) has been resumed. Additionally, their protective effects could be a new field of research and a novel therapeutic strategy for the early treatment of these conditions and avoid SUDEP.


Subject(s)
Drug Resistant Epilepsy , Erythropoietin , Brain/metabolism , Cardiovascular System/pathology , Erythropoietin/metabolism , Humans , Receptors, Erythropoietin/metabolism , Sudden Unexpected Death in Epilepsy
6.
J Cardiovasc Pharmacol ; 73(5): 290-300, 2019 05.
Article in English | MEDLINE | ID: mdl-31082960

ABSTRACT

Erythropoietin (EPO) has been linked to cardioprotective effects. However, its effects during the aging process are little known. We investigated the effect of EPO administration on hemodynamic parameters, cardiac function, oxidative damage, and erythropoietin receptor (EPOR) expression pattern in the hypovolemic state. EPO was administered (1000 IU/kg/3 days) and then acute hemorrhage (20% blood loss) was induced in young and adult rats. There was no difference in plasmatic EPO in either age group. The hemodynamic basal condition was similar, without alterations in renal function and hematocrit, in both age groups. After bleeding, both EPO-treated age groups had increased blood pressure at the end of the experimental protocol, being greater in adult animals. EPO attenuated the tachycardic effect. Ejection fraction and fractional shortening were higher in adult EPO-treated rats subjected to hemorrhage. In the left ventricle, young and adult EPO-treated rats subjected to bleeding showed an increased EPOR expression. A different EPOR expression pattern was observed in the adult right atrial tissue, compared with young animals. EPO treatment decreased oxidative damage to lipids in both age groups. EPO treatment before acute hemorrhage improves cardiovascular function during the aging process, which is mediated by different EPOR pattern expression in the heart tissue.


Subject(s)
Cardiovascular System/drug effects , Epoetin Alfa/administration & dosage , Hematinics/administration & dosage , Hemodynamics/drug effects , Hemorrhage/drug therapy , Ventricular Function, Left/drug effects , Age Factors , Animals , Cardiovascular System/metabolism , Cardiovascular System/physiopathology , Disease Models, Animal , Hemorrhage/metabolism , Hemorrhage/physiopathology , Lipid Peroxidation/drug effects , Male , Myocardium/metabolism , Oxidative Stress/drug effects , Rats, Sprague-Dawley , Receptors, Erythropoietin/agonists , Receptors, Erythropoietin/metabolism
7.
Redox Biol ; 24: 101187, 2019 06.
Article in English | MEDLINE | ID: mdl-30965198

ABSTRACT

Erythropoietin is a glycoproteic hormone that regulates hematopoiesis by acting on its specific receptor (EpoR). The expression of EpoR in the central nervous system (CNS) suggests a role for this hormone in the brain. Recently, we developed a new Epo variant without hematopoietic activity called EpoL, which showed marked neuroprotective effects against oxidative stress in brain ischemia related models. In this study, we have evaluated the neuroprotective effects of EpoL against oxidative stress induced by chronic treatment with Aß. Our results show that EpoL was neuroprotective against Aß-induced toxicity by a mechanism that implicates EpoR, reduction in reactive oxygen species, and reduction in astrogliosis. Furthermore, EpoL treatment improved calcium handling and SV2 levels. Interestingly, the neuroprotective effect of EpoL against oxidative stress induced by chronic Aß treatment was achieved at a concentration 10 times lower than that of Epo. In conclusion, EpoL, a new variant of Epo without hematopoietic activity, is of potential interest for the treatment of diseases related to oxidative stress in the CNS such as Alzheimer disease.


Subject(s)
Erythropoietin/pharmacology , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Peptide Fragments/pharmacology , Recombinant Proteins/pharmacology , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/pharmacology , Animals , Calcium Signaling , Cell Line, Tumor , Cell Survival/drug effects , Erythropoietin/genetics , Erythropoietin/isolation & purification , Goats , Milk , Neuroprotective Agents/isolation & purification , Peptide Fragments/chemistry , Protein Multimerization , Reactive Oxygen Species/metabolism , Receptors, Erythropoietin/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification
8.
Kidney Int ; 93(5): 1131-1141, 2018 05.
Article in English | MEDLINE | ID: mdl-29395333

ABSTRACT

It is accepted that osteoblasts/osteocytes are the major source for circulating fibroblast growth factor 23 (FGF23). However, erythropoietic cells of bone marrow also express FGF23. The modulation of FGF23 expression in bone marrow and potential contribution to circulating FGF23 has not been well studied. Moreover, recent studies show that plasma FGF23 may increase early during acute kidney injury (AKI). Erythropoietin, a kidney-derived hormone that targets erythropoietic cells, increases in AKI. Here we tested whether an acute increase of plasma erythropoietin induces FGF23 expression in erythropoietic cells of bone marrow thereby contributing to the increase of circulating FGF23 in AKI. We found that erythroid progenitor cells of bone marrow express FGF23. Erythropoietin increased FGF23 expression in vivo and in bone marrow cell cultures via the homodimeric erythropoietin receptor. In experimental AKI secondary to hemorrhagic shock or sepsis in rodents, there was a rapid increase of plasma erythropoietin, and an induction of bone marrow FGF23 expression together with a rapid increase of circulating FGF23. Blockade of the erythropoietin receptor fully prevented the induction of bone marrow FGF23 and partially suppressed the increase of circulating FGF23. Finally, there was an early increase of both circulating FGF23 and erythropoietin in a cohort of patients with severe sepsis who developed AKI within 48 hours of admission. Thus, increases in plasma erythropoietin and erythropoietin receptor activation are mechanisms implicated in the increase of plasma FGF23 in AKI.


Subject(s)
Acute Kidney Injury/blood , Bone Marrow Cells/metabolism , Erythroid Precursor Cells/metabolism , Erythropoietin/blood , Fibroblast Growth Factors/blood , Acute Kidney Injury/etiology , Animals , Bone Marrow Cells/drug effects , Disease Models, Animal , Erythroid Precursor Cells/drug effects , Erythropoietin/pharmacology , Fibroblast Growth Factor-23 , Humans , Male , Mice, Inbred C57BL , Prospective Studies , Rats, Sprague-Dawley , Receptors, Erythropoietin/agonists , Receptors, Erythropoietin/metabolism , Recombinant Proteins/pharmacology , Sepsis/blood , Sepsis/complications , Shock, Hemorrhagic/blood , Shock, Hemorrhagic/complications , Time Factors , Up-Regulation
9.
Redox Biol ; 14: 285-294, 2018 04.
Article in English | MEDLINE | ID: mdl-28987867

ABSTRACT

Human erythropoietin is mainly recognized for its hematopoietic function; however, by binding to its receptor (EpoR), it can activate different signaling pathways as STAT, PI3K, MAPK and RAS to increase cellular differentiation or provide neuroprotective effects, among others. A recombinant human erythropoietin variant with low glycosylation and without hematopoietic effect (EpoL) was purified from skimmed goat milk. Recombinant human erythropoietin (Epo) was obtained from CHO cell line and used as control to compare EpoL effects. Neuroprotection studies were performed in PC12 cells and rat hippocampal slices. Cells were pretreated during 1h with EpoL or Epo and exposed to oxidative agents (H2O2 or FCCP); cell viability was assayed at the end of the experiment by the MTT method. Hippocampal slices were exposed to 15min of oxygen and glucose deprivation (OGD) and the neuroprotective drugs EpoL or Epo were incubated for 2h post-OGD in re-oxygenated medium. Cell cultures stressed with oxidative agents, and pretreated with EpoL, showed neuroprotective effects of 30% at a concentration 10 times lower than that of Epo. Moreover, similar differences were observed in OGD ex vivo assays. Neuroprotection elicited by EpoL was lost when an antibody against EpoR was present, indicating that its effect is EpoR-dependent. In conclusion, our results suggest that EpoL has a more potent neuroprotective profile than Epo against oxidative stress, mediated by activation of EpoR, thus EpoL represents an important target to develop a potential biopharmaceutical to treat different central nervous system pathologies related to oxidative stress such as stroke or neurodegenerative diseases.


Subject(s)
Erythropoietin/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Animals , CHO Cells , Cell Survival/drug effects , Cricetulus , Erythropoietin/genetics , Humans , Neurons/cytology , Neurons/metabolism , Neuroprotective Agents/metabolism , PC12 Cells , Rats , Receptors, Erythropoietin/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology
10.
Tumour Biol ; 37(10): 13581-13593, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27468719

ABSTRACT

Clear cell renal cell carcinoma (ccRCC) is the most common subtype of renal carcinomas. There is great interest to know the molecular basis of the tumor biology of ccRCC that might contribute to a better understanding of the aggressive biological behavior of this cancer and to identify early biomarkers of disease. This study describes the relationship among proliferation, survival, and apoptosis with the expression of key molecules related to tumoral hypoxia (hypoxia-inducible factor (HIF)-1α, erythropoietin (EPO), vascular endothelial growth factor (VEGF)), their receptors (EPO-R, VEGFR-2), and stearoyl desaturase-1 (SCD-1) in early stages of ccRCC. Tissue samples were obtained at the Urology Unit of the J.R. Vidal Hospital (Corrientes, Argentina), from patients who underwent radical nephrectomy for renal cancer between 2011 and 2014. Four experimental groups according to pathological stage and nuclear grade were organized: T1G1 (n = 6), T2G1 (n = 4), T1G2 (n = 7), and T2G2 (n = 7). The expression of HIF-1α, EPO, EPO-R, VEGF, VEGFR-2, Bcl-xL, and SCD-1 were evaluated by immunohistochemistry, Western blotting, and/or RT-PCR. Apoptosis was assessed by the TUNEL in situ assay, and tumor proliferation was determined by Ki-67 immunohistochemistry. Data revealed that HIF-1α, EPO, EPO-R, VEGF, and VEGF-R2 were overexpressed in most samples. The T1G1 group showed the highest EPO levels, approximately 200 % compared with distal renal tissue. Bcl-xL overexpression was concomitant with the enhancement of proliferative indexes. SCD-1 expression increased with the tumor size and nuclear grade. Moreover, the direct correlations observed between SCD-1/HIF-1α and SCD-1/Ki-67 increments suggest a link among these molecules, which would determine tumor progression in early stages of ccRCC. Our results demonstrate the relationship among proliferation, survival, and apoptosis with the expression of key molecules related to tumoral hypoxia (HIF-1α, EPO, VEGF), their receptors (EPO-R, VEGFR-2), and SCD-1 in early stages of ccRCC.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Renal Cell/pathology , Erythropoietin/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney Neoplasms/pathology , Receptors, Erythropoietin/metabolism , Stearoyl-CoA Desaturase/metabolism , Adult , Aged , Aged, 80 and over , Apoptosis , Biomarkers, Tumor/genetics , Blotting, Western , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/surgery , Cell Proliferation , Erythropoietin/genetics , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Immunoenzyme Techniques , Kidney Neoplasms/metabolism , Kidney Neoplasms/surgery , Male , Middle Aged , Neoplasm Grading , Neoplasm Staging , Prognosis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Erythropoietin/genetics , Reverse Transcriptase Polymerase Chain Reaction , Stearoyl-CoA Desaturase/genetics , Survival Rate , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , Young Adult
11.
Biomed Pharmacother ; 82: 606-13, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27470403

ABSTRACT

Sepsis remains the most important cause of acute kidney injury (AKI) and acute lung injury (ALI) in critically ill patients. The cecal ligation and puncture (CLP) model in experimental mice reproduces most of the clinical features of sepsis. Erythropoietin (EPO) is a well-known cytoprotective multifunctional hormone, which exerts anti-inflammatory, anti-oxidant, anti-apoptotic and pro-angiogenic effects in several tissues. The aim of this study was to evaluate the underlying mechanisms of EPO protection through the expression of the EPO/EPO receptor (EPO-R) and VEGF/VEF-R2 systems in kidneys and lungs of mice undergoing CLP-induced sepsis. Male inbred Balb/c mice were divided in three experimental groups: Sham, CLP, and CLP+EPO (3000IU/kg sc). Assessment of renal functional parameters, survival, histological examination, immunohistochemistry and/or Western blottings of EPO-R, VEGF and VEGF-R2 were performed at 18h post-surgery. Mice demonstrated AKI by elevation of serum creatinine and renal histologic damage. EPO treatment attenuates renal dysfunction and ameliorates kidney histopathologic changes. Additionally, EPO administration attenuates deleterious septic damage in renal cortex through the overexpression of EPO-R in tubular interstitial cells and the overexpression of the pair VEGF/VEGF-R2. Similarly CLP- induced ALI, as evidenced by parenchymal lung histopathologic alterations, was ameliorated through pulmonary EPO-R, VEGF and VEGF-R2 over expression suggesting and improvement in endothelial survival and functionality. This study demonstrates that EPO exerts protective effects in kidneys and lungs in mice with CLP-induced sepsis through the expression of EPO-R and the regulation of the VEGF/VEGF-R2 pair.


Subject(s)
Acute Kidney Injury/drug therapy , Acute Lung Injury/drug therapy , Erythropoietin/therapeutic use , Receptors, Erythropoietin/metabolism , Sepsis/microbiology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Acute Lung Injury/physiopathology , Animals , Blood Urea Nitrogen , Cecum/pathology , Creatinine/blood , Disease Models, Animal , Erythropoietin/administration & dosage , Erythropoietin/pharmacology , Kidney/drug effects , Kidney/pathology , Ligation , Male , Mice, Inbred BALB C , Punctures , Survival Analysis
12.
J Appl Physiol (1985) ; 121(1): 53-8, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27125843

ABSTRACT

Excessive erythrocytosis (EE) is the main sign of Chronic Mountain Sickness (CMS), a highly prevalent syndrome in Andean highlanders. Low pulse O2 saturation (SpO2) during sleep and serum androgens have been suggested to contribute to EE in CMS patients. However, whether these factors have a significant impact on the erythropoietin (Epo) system leading to EE is still unclear. We have recently shown that morning soluble Epo receptor (sEpoR), an endogenous Epo antagonist, is decreased in CMS patients suggesting increased Epo availability (increased Epo/sEpoR). The present study aimed to characterize the nocturnal concentration profile of sEpoR and Epo and their relationship with SpO2, Hct, and serum testosterone in healthy highlanders (HH) and CMS patients. Epo and sEpoR concentrations were evaluated every 4 h (6 PM to 6 AM) and nighttime SpO2 was continuously monitored (10 PM to 6 AM) in 39 male participants (CMS, n = 23; HH, n = 16) aged 21-65 yr from Cerro de Pasco, Peru (4,340 m). CMS patients showed higher serum Epo concentrations throughout the night and lower sEpoR from 10 PM to 6 AM. Consequently, Epo/sEpoR was significantly higher in the CMS group at every time point. Mean sleep-time SpO2 was lower in CMS patients compared with HH, while the percentage of sleep time spent with SpO2 < 80% was higher. Multiple-regression analysis showed mean sleep-time SpO2 and Epo/sEpoR as significant predictors of hematocrit corrected for potential confounders (age, body mass index, and testosterone). Testosterone levels were associated neither with Hct nor with erythropoietic factors. In conclusion, our results show sustained erythropoietic stimulus driven by the Epo system in CMS patients, further enhanced by a continuous exposure to accentuated nocturnal hypoxemia.


Subject(s)
Altitude Sickness/blood , Altitude Sickness/metabolism , Receptors, Erythropoietin/blood , Receptors, Erythropoietin/metabolism , Sleep/physiology , Adult , Aged , Altitude , Altitude Sickness/physiopathology , Androgens/blood , Chronic Disease , Hematocrit/methods , Humans , Hypoxia/blood , Hypoxia/metabolism , Hypoxia/physiopathology , Male , Middle Aged , Oxygen/metabolism , Peru , Polycythemia/metabolism , Polycythemia/physiopathology , Testosterone/blood , Young Adult
14.
Neurosci Lett ; 583: 16-20, 2014 Nov 07.
Article in English | MEDLINE | ID: mdl-25219375

ABSTRACT

Erythropoietin in the nervous system is a potential neuroprotective factor for cerebral ischemic damage due to specific-binding to the erythropoietin receptor, which is associated with survival mechanisms. However, the role of its receptor is unclear. Thus, this work assessed whether a low dose (500UI/Kg) of intranasal recombinant human erythropoietin administered 3h after ischemia induced changes in the activation of its receptor at the Tyr456-phosphorylated site in ischemic hippocampi in rats. The results showed that recombinant human erythropoietin after injury maintained cell survival and was associated with an increase in receptor phosphorylation at the Tyr456 site as an initial signaling step, which correlated with a neuroprotective effect.


Subject(s)
Brain Ischemia/metabolism , Erythropoietin/metabolism , Erythropoietin/pharmacology , Hippocampus/drug effects , Receptors, Erythropoietin/metabolism , Administration, Intranasal , Animals , Brain Ischemia/pathology , Cell Survival/drug effects , Cytoprotection , Erythropoietin/administration & dosage , Hippocampus/metabolism , Hippocampus/pathology , Humans , Male , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacology , Phosphorylation , Rats, Wistar , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Tyrosine/metabolism
15.
Mol Med Rep ; 9(5): 1895-902, 2014 May.
Article in English | MEDLINE | ID: mdl-24626629

ABSTRACT

The cytokines erythropoietin (Epo) and stem cell factor (SCF), coupled with the cooperation between their receptors (EpoR and c-Kit), are essential components of normal physiological erythropoiesis. In earlier studies, we demonstrated the expression of c-Kit and EpoR in cervical cancer cells. It was identified that SCF is a survival factor, whereas Epo promotes cell proliferation. Cooperation between EpoR and SCF in cervical cancer has rarely been studied, despite the fact that cell migration and anchorage independent growth are considered initial steps in metastasis. Thus, the aim of this study was to analyse the effect of SCF and Epo alone, or in combination, on the migration and anchorage independent growth of two cervical cancer-derived cell lines. First, we demonstrated the expression of EpoR and c-Kit in the cell lines. Next, we evaluated anchorage independent growth, and identified that Epo and SCF produced a modest number of colonies, whereas the combination Epo/SCF induced a significantly higher number of colonies. Migration was then evaluated in Boyden chambers. Co-stimulation with Epo/SCF induced a significantly higher number of migrating cells than either cytokine alone. SCF-, Epo- and Epo/SCF-induced migration was inhibited by blocking phosphorylation of Janus kinase 2 (JAK2). Accordingly, western blot analysis demonstrated that the JAK2/signal transducer and activator of transcription-5 (STAT5) axis was activated in all cases. By contrast, inhibition of extracellular signal-related kinase (ERK) 1/2 abrogated migration induced by SCF and Epo/SCF only. Concurrently, Epo induced a modest, transient activation of ERK1/2, whereas SCF and Epo/SCF prompted a strong, sustained phosphorylation of ERK1/2. The results from this study have revealed that co-stimulation with Epo/SCF promotes migration and anchorage independent cell growth, and that co-signalling from EpoR and c-Kit converge on JAK2/STAT5 activation. Furthermore, SCF- and Epo/SCF-induced migration depends on the sustained activation of ERK1/2. These results indicate that co-signalling from different cytokine receptors induces migration, and this suggests that migratory behaviour may be regulated by the cooperative activity of Epo and SCF in cells expressing their cognate receptors.


Subject(s)
Erythropoietin/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Stem Cell Factor/pharmacology , Uterine Cervical Neoplasms/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Janus Kinase 2/metabolism , Proto-Oncogene Proteins c-kit/genetics , Receptors, Erythropoietin/genetics , Receptors, Erythropoietin/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction/drug effects , Uterine Cervical Neoplasms/genetics
16.
Toxicology ; 318: 13-21, 2014 Apr 06.
Article in English | MEDLINE | ID: mdl-24561306

ABSTRACT

Sepsis remains the most important cause of acute kidney injury (AKI) in critically ill patients and is an independent predictor of poor outcome. The administration of lipopolysaccharide (LPS) to animals reproduces most of the clinical features of sepsis, including AKI, a condition associated with renal cellular dysfunction and apoptosis. Erythropoietin (EPO) is a well known cytoprotective multifunctional hormone, which exerts anti-inflammatory, anti-oxidant, anti-apoptotic and angiogenic effects in several tissues. The aim of this study was to evaluate the underlying mechanisms of EPO renoprotection through the expression of the EPO receptor (EPO-R) and the modulation of the intrinsic apoptotic pathway in LPS-induced AKI. Male inbred Balb/c mice were divided in four experimental groups: Control, LPS (8 mg/kg i.p.), EPO (3000 IU sc) and LPS+EPO. Assessment of renal function, histological examination, TUNEL in situ assay, immunohistochemistry and Western blottings of caspase-3, Bax, Bcl-xL, EPO-R and Cytochrome c were performed at 24h post treatment. LPS+EPO treatment significantly improved renal function and ameliorated histopathological injury when compared to the LPS treated group. Results showed that EPO treatment attenuates renal tubular apoptosis through: (a) the overexpression of EPO-R in tubular interstitial cells, (b) the reduction of Bax/Bcl-xL ratio, (c) the inhibition Cytochrome c release into the cytosol and (d) the decrease of the active caspase-3 expression. This study suggests that EPO exerts renoprotection on an experimental model of LPS-induced AKI. EPO induced renoprotection involves an anti-apoptotic effect through the expression of EPO-R and the regulation of the mitochondrial apoptotic pathway.


Subject(s)
Acute Kidney Injury/drug therapy , Acute Kidney Injury/physiopathology , Apoptosis/physiology , Cryoprotective Agents/pharmacology , Erythropoietin/pharmacology , Mitochondria/metabolism , Receptors, Erythropoietin/metabolism , Acute Kidney Injury/chemically induced , Animals , Apoptosis/drug effects , Disease Models, Animal , Humans , Kidney/metabolism , Lipopolysaccharides , Male , Mice , Mice, Inbred BALB C
17.
Biochim Biophys Acta ; 1833(8): 1960-8, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23602701

ABSTRACT

It is now recognized that in addition to its activity upon erythroid progenitor cells, erythropoietin (Epo) is capable of stimulating survival of different non-erythroid cells. Since stimulation of erythropoiesis is unwanted for neuroprotection, Epo-like compounds with a more selective action are under investigation. Although the carbamylated derivative of erythropoietin (cEpo) has demonstrated non-hematopoietic tissue protection without erythropoietic effect, little is known about differential mechanisms between Epo and cEpo. Therefore, we investigated signaling pathways which play a key role in Epo-induced proliferation. Here we show that cEpo blocked FOXO3a phosphorylation, allowing expression of downstream target p27(kip1) in UT-7 and TF-1 cells capable of erythroid differentiation. This is consistent with the involvement of cEpo in slowing down G1-to-S-phase progression compared with the effect of Epo upon cell cycle. In contrast, similar antiapoptotic actions of cEpo and Epo were observed in neuronal SH-SY5Y cells. Inhibition and competition assays suggest that Epo may act through both, the homodimeric (EpoR/EpoR) and the heterodimeric (EpoR/ßcR) receptors in neuronal SH-SY5Y cells and probably in the TF-1 cell type as well. Results also indicate that cEpo needs both the EpoR and ßcR subunits to prevent apoptosis of neuronal cells. Based on evidence suggesting that cell proliferation pathways were involved in the differential effect of Epo and cEpo, we went forward to studying downstream signals. Here we provide the first evidence that unlike Epo, cEpo failed to induce FOXO3a inactivation and subsequent p27(kip1) downregulation, which is clearly shown in the incapacity of cEpo to induce erythroid cell growth.


Subject(s)
Erythropoietin/analogs & derivatives , Erythropoietin/pharmacology , Signal Transduction/drug effects , Apoptosis/drug effects , Apoptosis/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Growth Processes/drug effects , Cell Growth Processes/genetics , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Erythroid Cells/drug effects , Erythroid Cells/metabolism , Erythropoiesis/drug effects , Erythropoiesis/genetics , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , G1 Phase/drug effects , G1 Phase/genetics , Humans , Neurons/drug effects , Neurons/metabolism , Phosphorylation , Receptors, Erythropoietin/genetics , Receptors, Erythropoietin/metabolism , S Phase/drug effects , S Phase/genetics , Signal Transduction/genetics
18.
FEBS J ; 280(7): 1630-42, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23384249

ABSTRACT

Inflammation is a physiological defense response, but may also represent a potential pathological process in neurological diseases. In this regard, microglia have a crucial role in either progression or amelioration of degenerative neuronal damage. Because of the role of hypoxia in pro-inflammatory mechanisms in the nervous system, and the potential anti-inflammatory protective effect of erythropoietin (Epo), we focused our investigation on the role of this factor on activation of microglia and neuroprotection. Activation of microglial cells (EOC-2) was achieved by chemical hypoxia induced by cobalt chloride (CoCl2 ) and characterized by increased levels of nitrite, tumor necrosis factor-α and reactive oxygen species production, as well as up-regulation of inducible nitric oxide synthase expression. Under these conditions, cell proliferation data and proliferating cell nuclear antigen (PCNA) staining demonstrated a mitogenic effect of chemical hypoxia. Even though pre-treatment with Epo did not prevent nitrite production, inducible nitric oxide synthase protein expression or tumor necrosis factor-α secretion, it prevented the oxidative stress induced by CoCl2 as well as cell proliferation. Neuronal cells (SH-SY5Y) cultured in the presence of conditioned medium from activated EOC-2 cells or macrophages (RAW 264.7) developed significant apoptosis, an effect that was abolished by Epo via Epo/Epo receptor activation. The results show that even though Epo did not exert a direct anti-inflammatory effect on microglia activation, it did increase the resistance of neurons to subsequent damage from pro-inflammatory agents. In addition to its anti-apoptotic ability, the Epo antioxidant effect may have an indirect influence on neuronal survival by modulation of the pro-inflammatory environment.


Subject(s)
Erythropoietin/metabolism , Microglia/metabolism , Microglia/pathology , Animals , Cell Hypoxia/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Cobalt/pharmacology , Culture Media, Conditioned/pharmacology , Erythropoietin/pharmacology , Humans , Inflammation/metabolism , Inflammation/pathology , Mice , Microglia/drug effects , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/pharmacology , Neurotoxicity Syndromes/pathology , Nitric Oxide Synthase Type II/metabolism , Nitrites/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Reactive Oxygen Species/metabolism , Receptors, Erythropoietin/metabolism , Tumor Necrosis Factor-alpha/metabolism
19.
PLoS One ; 7(1): e29893, 2012.
Article in English | MEDLINE | ID: mdl-22235348

ABSTRACT

BACKGROUND: Despite advances in supportive care, sepsis-related mortality remains high, especially in patients with acute kidney injury (AKI). Erythropoietin can protect organs against ischemia and sepsis. This effect has been linked to activation of intracellular survival pathways, although the mechanism remains unclear. Continuous erythropoietin receptor activator (CERA) is an erythropoietin with a unique pharmacologic profile and long half-life. We hypothesized that pretreatment with CERA would be renoprotective in the cecal ligation and puncture (CLP) model of sepsis-induced AKI. METHODS: RATS WERE RANDOMIZED INTO THREE GROUPS: control; CLP; and CLP+CERA (5 µg/kg body weight, i.p. administered 24 h before CLP). At 24 hours after CLP, we measured creatinine clearance, biochemical variables, and hemodynamic parameters. In kidney tissue, we performed immunoblotting--to quantify expression of the Na-K-2Cl cotransporter (NKCC2), aquaporin 2 (AQP2), Toll-like receptor 4 (TLR4), erythropoietin receptor (EpoR), and nuclear factor kappa B (NF-κB)--and immunohistochemical staining for CD68 (macrophage infiltration). Plasma interleukin (IL)-2, IL-1ß, IL-6, IL-10, interferon gamma, and tumor necrosis factor alpha were measured by multiplex detection. RESULTS: Pretreatment with CERA preserved creatinine clearance and tubular function, as well as the expression of NKCC2 and AQP2. In addition, CERA maintained plasma lactate at normal levels, as well as preserving plasma levels of transaminases and lactate dehydrogenase. Renal expression of TLR4 and NF-κB was lower in CLP+CERA rats than in CLP rats (p<0.05 and p<0.01, respectively), as were CD68-positive cell counts (p<0.01), whereas renal EpoR expression was higher (p<0.05). Plasma levels of all measured cytokines were lower in CLP+CERA rats than in CLP rats. CONCLUSION: CERA protects against sepsis-induced AKI. This protective effect is, in part, attributable to suppression of the inflammatory response.


Subject(s)
Acute Kidney Injury/etiology , Acute Kidney Injury/prevention & control , Erythropoietin/pharmacology , Multiple Organ Failure/etiology , Multiple Organ Failure/prevention & control , Polyethylene Glycols/pharmacology , Sepsis/complications , Animals , Cecum/surgery , Cytokines/metabolism , Gene Expression Regulation/drug effects , Hemodynamics/drug effects , Kidney Tubules/drug effects , Kidney Tubules/metabolism , Ligation/adverse effects , Macrophages/drug effects , Macrophages/immunology , Male , NF-kappa B/metabolism , Punctures/adverse effects , Rats , Rats, Wistar , Receptors, Erythropoietin/metabolism , Sepsis/etiology , Sepsis/immunology , Sepsis/metabolism , Toll-Like Receptor 4/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL