Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 223
Filtrar
Más filtros

Medicinas Complementárias
Intervalo de año de publicación
2.
Dev Psychobiol ; 65(1): e22353, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36567653

RESUMEN

In this study, based on the excitatory/inhibitory imbalance theory of autism, the time window of GABA switch, the role of K-Cl co-transporter 2 (KCC2) in adjustment GABA switch, and brain permeability to erythropoietin (EPO), the effects of postnatal -EPO and- nano- erythropoietin (NEPO) have been evaluated in the valproic acid (VPA) rat model of autism. The VPA was administered for animal modeling of autism at gestational day (GD) 12.5 (600 mg/kg). Male offsprings were injected with EPO and NEPO in a clinically proper postnatal dosing regimen on postnatal days (PND) 1-5, and autistic-like behaviors were tested at the end of the first month. Then animals were sacrificed, and neuron morphology and KCC2 expression were examined by Nissl staining and Western blot. According to our findings, high-dose NEPO improved autism-associated phenotypes. Neuroprotective effects of EPO and NEPO have been shown in the hippocampus. Postnatal NEPO treatment reversed KCC2 expression abnormalities induced by prenatal VPA. Our results might support the role of KCC2 in ASD and the excitatory/inhibitory imbalance hypothesis. We suggested Nano- erythropoietin and other KCC2 interventions as a new approach to the early treatment and prevention of autism.


Asunto(s)
Trastorno Autístico , Eritropoyetina , Hipocampo , Simportadores , Animales , Femenino , Humanos , Masculino , Embarazo , Ratas , Trastorno Autístico/inducido químicamente , Trastorno Autístico/tratamiento farmacológico , Trastorno Autístico/metabolismo , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Modelos Animales de Enfermedad , Ácido gamma-Aminobutírico/metabolismo , Ácido gamma-Aminobutírico/farmacología , Ácido gamma-Aminobutírico/uso terapéutico , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Simportadores/metabolismo , Simportadores/farmacología , Simportadores/uso terapéutico , Ácido Valproico/farmacología , Eritropoyetina/farmacología , Eritropoyetina/uso terapéutico
3.
Life Sci ; 310: 121124, 2022 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-36306536

RESUMEN

AIMS: While elevated hepcidin levels with inflammation have been postulated as a putative mechanism hindering effective erythropoiesis after intravenous (IV) iron therapy in anemic patients undergoing surgery, little is known about the concomitant changes in other major regulators affecting erythropoiesis. This study investigated the activities of relevant regulators after iron replenishment in a rat model of iron deficiency anemia with inflammation. MAIN METHODS: Inflammation was induced by administration of complete Freund's adjuvant (CFA) in male Sprague-Dawley rats. After 2 weeks of CFA treatment, the rats received IV iron (CFA­iron) or saline (CFA-saline). The control group received saline instead of CFA and iron (saline-saline). At 1, 3, and 10 days after iron or saline treatment, inflammatory cytokines, oxidative markers, iron profiles, hepcidin, erythropoietin (EPO), erythroferrone (ERFE), fibroblast growth factor 23 (FGF 23), and expression of mRNA and proteins in the liver involved in hepcidin signaling pathways were measured. KEY FINDINGS: CFA treatment and iron restriction decreased hemoglobin and serum iron levels, significantly increasing inflammatory and oxidative markers. Iron supplementation did not restore hemoglobin levels despite improved iron profiles. CFA injections increased hepcidin and FGF 23 levels and decreased EPO and ERFE levels, which further intensified after iron supplementation with concomitantly elevated levels of oxidative stress and inflammatory markers. SIGNIFICANCE: Under inflammatory conditions, IV iron administration exacerbated inflammatory and oxidative stress and did not resolve anemia, even under iron deficiency conditions. Iron therapy exerted adverse influences on the changes in key regulators toward impeding erythropoiesis that was already impeded by inflammation.


Asunto(s)
Anemia Ferropénica , Anemia , Eritropoyetina , Deficiencias de Hierro , Masculino , Ratas , Animales , Hepcidinas/metabolismo , Eritropoyesis , Hierro/metabolismo , Ratas Sprague-Dawley , Eritropoyetina/farmacología , Anemia Ferropénica/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Inflamación/complicaciones , Hemoglobinas , Biomarcadores , Suplementos Dietéticos
4.
Bioengineered ; 13(5): 13021-13032, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35611764

RESUMEN

We investigated the mechanism of erythropoietin (EPO) in brain injury in premature mice based on Akt/mTOR/p70S6K signaling pathway. The brain injury model group of premature mice was obtained by intraperitoneal injection of lipopolysaccharide during pregnancy. Normal mice were taken as the control group. The model mice were divided into low-dose EPO (1,000 IU/kg, L-EPO), medium-dose EPO (2,500 IU/kg, M-EPO), and high-dose EPO groups (5,000 IU/kg, H-EPO) by intraperitoneal injection. The levels of malondialdehyde (MDA) and total superoxide dismutase (T-SOD) were detected. TUNEL staining and Western blotting were used to detect the differences in neuronal apoptosis index (AI), microglial polarization marker protein, and Akt/mTOR/p70S6K-related protein expression levels in each group. Compared with the control group, the protein levels of AI, MDA, Bax, and iNOS in the model, L-EPO, and M-EPO groups were significantly increased, while the T-SOD level and Bcl-2, ARG1, p-Akt, p-mTOR, and p-70S6K protein levels were significantly decreased (P < 0.05). Compared with the model group, AI, MAD levels and Bax, iNOS protein expression levels in L-EPO, M-EPO, and H-EPO groups were significantly decreased, while T-SOD level and Bcl-2, ARG1, p-Akt, p-mTOR, and p-70S6K protein levels were significantly increased. The changes were dose-dependent. In summary, EPO can activate microglia transformation from M1 to M2 through Akt/mTOR/p70S6K signaling pathway.


Asunto(s)
Lesiones Encefálicas , Eritropoyetina , Animales , Biopelículas , Eritropoyetina/farmacología , Ratones , Microglía/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Transducción de Señal , Superóxido Dismutasa/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteína X Asociada a bcl-2/metabolismo
5.
Eur J Pharmacol ; 912: 174583, 2021 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-34678238

RESUMEN

Anemia is a common feature and complication of chronic kidney disease (CKD). Erythropoiesis-stimulating agents (ESAs) and recombinant human erythropoietin have been used widely in renal anemia treatment. Recently, hypoxia-inducible factor-prolyl hydroxylase domain inhibitors (HIF-PHIs) that may improve the treatment of renal anemia patients were launched. Previous studies indicated that HIF-PHIs may decrease hepcidin levels and modulate iron metabolism, thereby increasing total iron-binding capacity and reducing the need for iron supplementation. Furthermore, HIF-PHIs can reduce inflammation and oxidative stress in CKD. Recombinant erythropoietin has become a routine treatment for patients with CKD and end-stage renal disease with relatively few adverse effects. However, higher doses of recombinant erythropoietin have been demonstrated to be an independent predictor of mortality in patients under hemodialysis. Phase III clinical trials of HIF-PHIs in patients with anemia and dialysis-dependent CKD have shown their efficacy and safety in both non-dialysis and dialysis CKD patients. However, HIFα binds to specific hypoxia-response elements in the vascular endothelial growth factor or retinoic acid-related orphan receptor gamma t (RORγt) promoter, which may be involved in the progression of cancer, psoriasis, and rheumatoid arthritis. In this paper, we have summarized the mechanism, clinical application, and clinical trials of HIF-PHIs in the treatment of renal anemia and aimed to provide an overview of the new drugs in clinical practice, as well as reconsider the advantages and disadvantages of HIF-PHIs and ESAs. Presently, there are not enough clinical studies examining the effects of long-term administration of HIF-PHIs. Therefore, further studies will be needed.


Asunto(s)
Anemia/tratamiento farmacológico , Anemia/metabolismo , Inhibidores Enzimáticos/farmacología , Hematínicos/farmacología , Prolina Dioxigenasas del Factor Inducible por Hipoxia/antagonistas & inhibidores , Insuficiencia Renal Crónica/tratamiento farmacológico , Insuficiencia Renal Crónica/metabolismo , Anemia/etiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/química , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Enfermedades Cardiovasculares/inducido químicamente , Inhibidores Enzimáticos/efectos adversos , Inhibidores Enzimáticos/uso terapéutico , Eritropoyetina/efectos adversos , Eritropoyetina/farmacología , Eritropoyetina/uso terapéutico , Hematínicos/efectos adversos , Hematínicos/uso terapéutico , Humanos , Insuficiencia Renal Crónica/complicaciones
6.
J Pediatr ; 238: 102-109.e8, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34324880

RESUMEN

OBJECTIVES: To test whether an increased iron dose is associated with improved neurodevelopment as assessed by the Bayley Scales of Infant Development, third edition (BSID-III) among infants enrolled in the Preterm Erythropoietin (Epo) Neuroprotection Trial (PENUT). STUDY DESIGN: This is a post hoc analysis of a randomized trial that enrolled infants born at 24-28 completed weeks of gestation. All infants in PENUT who were assessed with BSID-III at 2 years were included in this study. The associations between enteral iron dose at 60 and 90 days and BSID-III component scores were evaluated using generalized estimating equations models adjusted for potential confounders. RESULTS: In total, 692 infants were analyzed (355 placebo, 337 Epo). Enteral iron supplementation ranged from 0 to 14.7 mg/kg/d (IQR 2.1-5.8 mg/kg/d) at day 60, with a mean of 3.6 mg/kg/d in infants treated with placebo and 4.8 mg/kg/d in infants treated with Epo. A significant positive association was seen between BSID-III cognitive scores and iron dose at 60 days, with an effect size of 0.77 BSID points per 50 mg/kg increase in cumulative iron dose (P = .03). Greater iron doses were associated with greater motor and language scores but did not reach statistical significance. Results at 90 days were not significant. The effect size in the infants treated with Epo compared with placebo was consistently greater. CONCLUSIONS: A positive association was seen between iron dose at 60 days and cognitive outcomes. Our results suggest that increased iron supplementation in infants born preterm, at the doses administered in the PENUT Trial, may have positive neurodevelopmental effects, particularly in infants treated with Epo. TRIAL REGISTRATION: Clinicaltrials.gov: NCT01378273.


Asunto(s)
Hierro/administración & dosificación , Trastornos del Neurodesarrollo/prevención & control , Neuroprotección/efectos de los fármacos , Adulto , Nutrición Enteral , Eritropoyetina/administración & dosificación , Eritropoyetina/farmacología , Femenino , Humanos , Lactante , Recien Nacido Extremadamente Prematuro/crecimiento & desarrollo , Recién Nacido , Hierro/efectos adversos , Hierro/farmacología , Masculino , Embarazo , Estudios Prospectivos
7.
Biomed Pharmacother ; 139: 111558, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33894624

RESUMEN

Erythropoietin (EPO) is a hypoxia-induced hormone produced in adult kidneys with erythropoietic and non-erythropoietic effects. In vivo studies represent an important role to comprehend the efficacy and safety in the early phase of repurposing drugs. The aim is to evaluate the potential anti-inflammatory effect of EPO observed in animal models of disease. Following PRISMA statements, electronic database Medline via PubMed platform was used to search articles with the research expression ((erythropoietin [MeSH Terms]) AND (inflammation [MeSH Terms]) AND (disease models, animal [MeSH Terms])). The inclusion criteria were original articles, studies where EPO was administered, studies where inflammation was studied and/or evaluated, non-clinical studies in vivo with rodents, and articles published in English. Thirty-six articles met the criteria for qualitative analysis. Exogenous EPO was used in models of sepsis, traumatic brain injury, and autoimmune neuritis, with an average of 3000 IU/Kg for single and multiple doses, using mice and rats. Biomarkers such as immune-related effectors, cytokines, reactive oxygen species, prostaglandins, and other biomarkers were assessed. EPO has been recognized as a multifunctional cytokine with anti-inflammatory properties, showing its significant effect both in acute and chronic models of inflammation. Further non-clinical studies are suggested for the enlightenment of anti-inflammatory mechanisms of EPO in lower doses, allowing us to understand the translational data for humans.


Asunto(s)
Antiinflamatorios/farmacología , Eritropoyetina/farmacología , Inflamación/tratamiento farmacológico , Inflamación/fisiopatología , Animales , Biomarcadores , Evaluación Preclínica de Medicamentos , Humanos , Roedores
8.
Eur J Pharmacol ; 898: 173990, 2021 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-33657422

RESUMEN

Anemia with inflammation-induced defective iron utilization is a pathological condition observed in patients suffering from chronic kidney disease (CKD) or chronic inflammatory disease. There is no reasonable treatment for these conditions, because the effects of erythropoiesis stimulating agents (ESAs) or iron supplementation in the treatment of anemia are insufficient. JTZ-951 (enarodustat) has been characterized as a novel, orally bioavailable inhibitor of hypoxia-inducible factor prolyl hydroxylase (HIF-PH), and has been developed as a novel therapeutic agent for anemia with CKD. In this study, the effects of JTZ-951 on iron utilization during erythropoiesis and on anemia of inflammation were compared with those of recombinant human erythropoietin (rHuEPO) using normal rat and rat model of anemia of inflammation. In normal rats, under conditions in which JTZ-951 and rHuEPO showed similar erythropoietic effect, repeated doses of JTZ-951 induced erythropoiesis while retaining the hemoglobin content in red blood cells, while administration of rHuEPO resulted in decrease in some erythrocyte-related parameters. As for iron-related parameters during erythropoiesis, JTZ-951 exhibited more efficient iron utilization compared to rHuEPO. A single dose of JTZ-951 resulted in decrease in hepcidin expression observed within 24 h after administration, but a single dose of rHuEPO did not. In a rat model of anemia of inflammation (also known as a model with functional iron-deficiency), JTZ-951 showed erythropoietic effect, in contrast with rHuEPO. These results suggest that, unlike rHuEPO, JTZ-951 stimulates erythropoiesis by increasing iron utilization, and improves anemia of inflammation.


Asunto(s)
Anemia Ferropénica/tratamiento farmacológico , Inhibidores Enzimáticos/farmacología , Eritrocitos/efectos de los fármacos , Eritropoyesis/efectos de los fármacos , Eritropoyetina/farmacología , Hematínicos/farmacología , Prolina Dioxigenasas del Factor Inducible por Hipoxia/antagonistas & inhibidores , Hierro/sangre , Glicinas N-Sustituídas/farmacología , Piridinas/farmacología , Triazoles/farmacología , Anemia Ferropénica/sangre , Anemia Ferropénica/enzimología , Anemia Ferropénica/etiología , Animales , Artritis Experimental/complicaciones , Biomarcadores/sangre , Eritrocitos/enzimología , Femenino , Hepcidinas/genética , Hepcidinas/metabolismo , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Ratas Endogámicas Lew , Proteínas Recombinantes/farmacología
9.
Blood ; 136(9): 1080-1090, 2020 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-32438400

RESUMEN

Recombinant erythropoietin (EPO) and iron substitution are a standard of care for treatment of anemias associated with chronic inflammation, including anemia of chronic kidney disease. A black box warning for EPO therapy and concerns about negative side effects related to high-dose iron supplementation as well as the significant proportion of patients becoming EPO resistant over time explains the medical need to define novel strategies to ameliorate anemia of chronic disease (ACD). As hepcidin is central to the iron-restrictive phenotype in ACD, therapeutic approaches targeting hepcidin were recently developed. We herein report the therapeutic effects of a fully human anti-BMP6 antibody (KY1070) either as monotherapy or in combination with Darbepoetin alfa on iron metabolism and anemia resolution in 2 different, well-established, and clinically relevant rodent models of ACD. In addition to counteracting hepcidin-driven iron limitation for erythropoiesis, we found that the combination of KY1070 and recombinant human EPO improved the erythroid response compared with either monotherapy in a qualitative and quantitative manner. Consequently, the combination of KY1070 and Darbepoetin alfa resulted in an EPO-sparing effect. Moreover, we found that suppression of hepcidin via KY1070 modulates ferroportin expression on erythroid precursor cells, thereby lowering potentially toxic-free intracellular iron levels and by accelerating erythroid output as reflected by increased maturation of erythrocyte progenitors. In summary, we conclude that treatment of ACD, as a highly complex disease, becomes more effective by a multifactorial therapeutic approach upon mobilization of endogenous iron deposits and stimulation of erythropoiesis.


Asunto(s)
Anemia/terapia , Anticuerpos Monoclonales/uso terapéutico , Proteína Morfogenética Ósea 6/antagonistas & inhibidores , Darbepoetina alfa/uso terapéutico , Anemia/tratamiento farmacológico , Anemia/etiología , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/inmunología , Artritis/inducido químicamente , Artritis/complicaciones , Médula Ósea/metabolismo , Proteína Morfogenética Ósea 6/inmunología , Proteínas de Transporte de Catión/metabolismo , Citocinas/sangre , Darbepoetina alfa/administración & dosificación , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Eritropoyetina/farmacología , Eritropoyetina/uso terapéutico , Células Hep G2 , Humanos , Hierro/metabolismo , Ratones , Proteínas Musculares/sangre , Polisacáridos Bacterianos/toxicidad , Distribución Aleatoria , Proteínas Recombinantes/inmunología , Insuficiencia Renal Crónica/complicaciones
10.
Drug Test Anal ; 12(2): 261-267, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31655016

RESUMEN

Erythroferrone (ERFE) is a glycoprotein hormone secreted by erythroblasts in response to erythropoietin stimulation. ERFE suppresses the hepatic synthesis of the master iron-regulatory hormone, hepcidin. The impact of erythropoiesis stimulation on ERFE secretion in humans is poorly understood. This paucity of information is due in part to the lack of available means for ERFE quantification in serum samples. The present study tested a new sensitive sandwich immunoassay for human ERFE. This assay was used to demonstrate that injection of various erythropoiesis stimulating agents (ESAs) increased the blood ERFE levels in healthy volunteers. After exogenous stimulation of erythropoiesis, ERFE increased up to 8-fold with a detection window of 13 days. The impact of one unit of blood withdrawal on erythropoiesis stimulation of ERFE was also tested. ERFE significantly increased after blood withdrawal in subjects injected with both iron and saline solution, suggesting that iron supplementation did not mask the ERFE increase after blood withdrawal. The effects of exercise-induced muscle damage on ERFE was assessed by comparing ERFE levels with creatine kinase levels in samples from subjects with heavy exercise loads, and determined that this was not a confounder. The ERFE assay is a sensitive means to investigate the connection between iron metabolism and erythropoiesis in humans, and to detect ESA abuse in the antidoping field.


Asunto(s)
Eritropoyesis/efectos de los fármacos , Eritropoyetina/farmacología , Hematínicos/farmacología , Hormonas Peptídicas/sangre , Péptidos/farmacología , Detección de Abuso de Sustancias , Adulto , Biomarcadores/sangre , Eritropoyetina/administración & dosificación , Ejercicio Físico , Hematínicos/administración & dosificación , Humanos , Inyecciones , Hierro/administración & dosificación , Hierro/farmacología , Masculino , Péptidos/administración & dosificación , Detección de Abuso de Sustancias/métodos , Adulto Joven
11.
Int J Mol Sci ; 20(20)2019 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-31600973

RESUMEN

Anemia is a severe complication in patients with chronic kidney disease (CKD). Treatment with exogenous erythropoietin (EPO) can correct anemia in many with CKD. We produced 5/6-nephrectomized rats that became uremic and anemic at 25 days post surgery. Injection of the anemic 5/6-nephrectomized rats with 2.8 mg zinc/kg body weight raised their red blood cell (RBC) levels from approximately 85% of the control to 95% in one day and continued for 4 days. We compared the effect of ZnSO4 and recombinant human erythropoietin (rHuEPO) injections on relieving anemia in 5/6-nephrectomized rats. After three consecutive injections, both the ZnSO4 and rHuEPO groups had significantly higher RBC levels (98 ± 6% and 102 ± 6% of the control) than the saline group (90 ± 3% of the control). In vivo, zinc relieved anemia in 5/6-nephrectomized rats similar to rHuEPO. In vitro, we cultured rat bone marrow cells supplemented with ZnCl2, rHuEPO, or saline. In a 4-day suspension culture, we found that zinc induced erythropoiesis similar to rHuEPO. When rat bone marrow cells were supplement-cultured with zinc, we found that zinc stimulated the production of EPO in the culture medium and that the level of EPO produced was dependent on the concentration of zinc supplemented. The production of EPO via zinc supplementation was involved in the process of erythropoiesis.


Asunto(s)
Anemia/etiología , Suplementos Dietéticos , Eritropoyetina/farmacología , Proteínas Recombinantes/farmacología , Insuficiencia Renal Crónica/complicaciones , Zinc/administración & dosificación , Anemia/sangre , Anemia/tratamiento farmacológico , Animales , Biomarcadores , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Modelos Animales de Enfermedad , Índices de Eritrocitos/efectos de los fármacos , Eritropoyesis/efectos de los fármacos , Humanos , Ratas , Sulfato de Zinc/administración & dosificación
12.
Ann Clin Lab Sci ; 49(2): 257-264, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31028073

RESUMEN

The aim of this study was to explore the effects of erythropoietin (EPO) in acute lung injuries in rats with sepsis. 127 male SD rats were divided into 3 groups (n=8): group Sham, group ALI (sepsis-caused lung injury), group EPO (EPO intervention). The blood gas analysis, C-reactive protein level (CRP), tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6) and IL-10 were detected and compared. The water content fraction and myeloperoxidase (MPO) activities, apoptosis, and expression level of nuclear factor -κB (NF-κB p65) in lung tissues were also detected. Compared to group Sham, the oxygenation index of group ALI was decreased (140.66±12.82 vs. 194.7±16.36), CRP (peak value: 2.31±0.33 mg/L vs. 1.00±0.16 mg/L), IL-10 (peak value: 711.26±84.97 ng/L vs. 51.21±11.45 ng/L), TNF-a (63.69±6.85 ng/L vs. 12.92±0.91 ng/L), and IL-6 (peak value: 1768.93±195.11 ng/L vs. 68.71±11.48 ng/L) increased (P<0.01), and the lung cell apoptosis and expression of NF-κB p65 (302.75±51.38 vs. 121.46±11.79) were also increased (P<0.01), and there was a recovery of EPO group (P<0.01). Group ALI exhibited pulmonary interstitial and alveolar edema, hemorrhage, and lung collapse, but the EPO intervention significantly reduced the above lung pathological changes (the water content was 70.36±1.24% in Sham group, 84.26±3.07% in ALI group and 75.72±1.96% in EPO group, P<0.01; MPO: 2.23±0.67, 7.56±1.02, 5.73±0.86, P<0.01; apoptotic index: 3.73±0.39, 25.69±4.54, 9.59±2.59, P<0.01). EPO could inhibit the expression of NF-κB in lung tissues, inhibit the levels of proinflammatory cytokines TNF-a and IL-6, but improve the level of anti-inflammatory cytokine IL-10, thus reducing the pathological changes of lung tissues and producing protective effects towards lung tissues.


Asunto(s)
Lesión Pulmonar Aguda/tratamiento farmacológico , Eritropoyetina/uso terapéutico , Sustancias Protectoras/uso terapéutico , Sepsis/tratamiento farmacológico , Lesión Pulmonar Aguda/patología , Animales , Apoptosis/efectos de los fármacos , Análisis de los Gases de la Sangre , Ciego/patología , Eritropoyetina/farmacología , Mediadores de Inflamación/metabolismo , Ligadura , Pulmón/patología , Peroxidasa/metabolismo , Sustancias Protectoras/farmacología , Punciones , Ratas Sprague-Dawley , Factor de Transcripción ReIA/metabolismo , Agua/química
13.
J Alzheimers Dis ; 65(4): 1469-1483, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30175978

RESUMEN

Many experimental studies show that erythropoietin (EPO) has a neuroprotective action in the brain. EPO in acute and chronic neurological disorders, particularly in stroke, traumatic brain injury, Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis, has neuroprotective effects. We previously reported the neuroprotective effect of NeuroEPO, a low sialic form of EPO, against oxidative stress induced by glutamate excitotoxicity. In this paper, we analyze the effect of NeuroEPO against apoptosis induced by glutamate excitotoxicity in primary neuronal cultures obtained from the forebrains of Wistar rat embryos after 17 days of gestation. Excitotoxicity was induced after nine days of in vitro culture by treatment with a culture medium containing 100µM glutamate for 15 min. To withdraw glutamate, a new medium containing 100 ng NeuroEPO/mL was added. Apoptosis was analyzed after 24 h. Images obtained by phase contrast microscopy show that neurons treated with glutamate exhibit cell body shrinkage, loss of dendrites that do not make contact with neighboring cells, and that NeuroEPO was able to preserve the morphological characteristics of the control. Immunocytochemistry images show that the culture is essentially pure in neurons; that glutamate causes cell mortality, and that this is partially avoided when the culture medium is supplemented with NeuroEPO. Activation of intrinsic apoptotic pathways was analyzed. The decreases in Bcl-2/Bax ratio, increase in the release of cytochrome c, and in the expression and activity of caspase-3 observed in cells treated with glutamate, were restored by NeuroEPO. The results from this study show that NeuroEPO protects cortical neurons from glutamate-induced apoptosis via upregulation of Bcl-2 and inhibit glutamate-induced activation of caspase-3.


Asunto(s)
Eritropoyetina/farmacología , Agonistas de Aminoácidos Excitadores/farmacología , Ácido Glutámico/toxicidad , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Anilidas/farmacología , Animales , Caspasa 3/metabolismo , Células Cultivadas , Corteza Cerebral/citología , Citocromos c/metabolismo , Embrión de Mamíferos , Proteína Ácida Fibrilar de la Glía/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Oligopéptidos/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Ratas Wistar , Factores de Tiempo , Proteína X Asociada a bcl-2/metabolismo
14.
PLoS One ; 13(5): e0196684, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29738538

RESUMEN

OBJECTIVES: Anemia is a known driver for hypoxia inducible factor (HIF) which leads to increased renal erythropoietin (EPO) synthesis. Bone marrow (BM) EPO receptor (EPOR) signals are transduced through a JAK2-STAT5 pathway. The origins of anemia of chronic kidney disease (CKD) are multifactorial, including impairment of both renal EPO synthesis as well as intestinal iron absorption. We investigated the HIF- EPO- EPOR axis in kidney, BM and proximal tibia in anemic juvenile CKD rats. METHODS: CKD was induced by 5/6 nephrectomy in young (20 days old) male Sprague-Dawley rats while C group was sham operated. Rats were sacrificed 4 weeks after CKD induction and 5 minutes after a single bolus of IV recombinant human EPO. An additional control anemic (C-A) group was daily bled for 7 days. RESULTS: Hemoglobin levels were similarly reduced in CKD and C-A (11.4 ± 0.3 and 10.8±0.2 Vs 13.5±0.3 g/dL in C, p<0.0001). Liver hepcidin mRNA was decreased in CA but increased in CKD. Serum iron was unchanged while transferrin levels were mildly decreased in CKD. Kidney HIF2α protein was elevated in C-A but unchanged in CKD. Kidney EPO protein and mRNA levels were unchanged between groups. However, BM EPO protein (which reflects circulating EPO) was increased in C-A but remained unchanged in CKD. BM and proximal tibia EPOR were unchanged in C-A but decreased in CKD. Proximal tibial phospho-STAT5 increased after the EPO bolus in C but not in CKD. CONCLUSIONS: Compared to blood loss, anemia in young CKD rats is associated with inappropriate responses in the HIF-EPO-EPO-R axis: kidney HIF2α and renal EPO are not increased, BM and bone EPOR levels, as well as bone pSTAT5 response to EPO are reduced. Thus, anemia of CKD may be treated with additional therapeutic avenues beyond iron and EPO supplementation.


Asunto(s)
Anemia/etiología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Eritropoyetina/fisiología , Receptores de Eritropoyetina/fisiología , Insuficiencia Renal Crónica/complicaciones , Transducción de Señal/fisiología , Anemia/fisiopatología , Animales , Médula Ósea/metabolismo , Modelos Animales de Enfermedad , Eritropoyetina/biosíntesis , Eritropoyetina/genética , Eritropoyetina/farmacología , Hepcidinas/fisiología , Riñón/metabolismo , Hígado/metabolismo , Masculino , Nefrectomía/efectos adversos , ARN Mensajero/biosíntesis , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/farmacología , Insuficiencia Renal Crónica/sangre , Insuficiencia Renal Crónica/fisiopatología , Factor de Transcripción STAT5/fisiología
15.
J Hand Surg Am ; 43(4): 368-373, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29618417

RESUMEN

Current strategies for promoting faster and more effective peripheral nerve healing have utilized a wide variety of techniques and approaches. Nerve grafts, conduits, and stem cell therapy all have their respective advantages. However, there are still some difficulties in attaining complete functional recovery with a single treatment modality. The utilization of adjuvant treatments, in combination with current standard-of-care methods, offers the potential to improve patient outcomes. This paper highlights the current landscape of adjuvant treatments for enhancing peripheral nerve repair and regeneration.


Asunto(s)
Regeneración Nerviosa , Traumatismos de los Nervios Periféricos/terapia , Implantes Absorbibles , Aloinjertos , Autoinjertos , Bloqueadores de los Canales de Calcio/farmacología , Eritropoyetina/farmacología , Gabapentina/farmacología , Humanos , Inmunosupresores/farmacología , Compuestos de Litio/farmacología , Fármacos Neuroprotectores/farmacología , Procedimientos Neuroquirúrgicos/instrumentación , Nervios Periféricos/trasplante , Recuperación de la Función , Riluzol/farmacología , Trasplante de Células Madre , Ácido Valproico/farmacología , Venas/trasplante , Degeneración Walleriana/terapia
16.
Mol Med Rep ; 16(5): 5924-5930, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28849166

RESUMEN

Methylprednisolone sodium succinate (MPSS) has been suggested as a treatment for spinal cord injury (SCI), but its use has been limited due to its adverse effects. Erythropoietin (EPO) has been suggested as a promising candidate for limiting SCI in mammals. The aim of the present study was to investigate the effects of EPO in combination with MPSS on astrocytes following ischemic injury in vitro. Astrocytes were isolated from the cerebral cortex of postnatal day 3 Sprague­Dawley rats and cultured in vitro. Astrocyte ischemic injury was induced by oxygen and glucose deprivation for 4 h, and reperfusion was simulated by subsequent culture under normoxic conditions. The effects of EPO and MPSS on the expression of aquaporin­4 (AQP4) were investigated. Ischemic astrocytes were treated with EPO (10 U/ml), MPSS (10 µg/ml), or EPO (10 U/ml) in combination with MPSS (10 µg/ml) during reperfusion. The cell viability of astrocytes was assessed using an MTT assay. The mRNA and protein expression levels of AQP4 were determined using reverse transcription­quantitative polymerase chain reaction and western blot analysis, respectively. The role of the protein kinase C (PKC) signaling pathway in the molecular mechanisms underlying the effects of EPO and MPSS was also investigated. The present results demonstrated that following treatment with EPO and MPSS, the mRNA expression levels of AQP4 were upregulated and cell viability was enhanced. EPO and MPSS effectively inhibited the oxygen and glucose deprivation­mediated downregulation of AQP4 following reperfusion. In addition, the combined treatment with EPO and MPSS exhibited higher AQP4 expression levels and cell viability compared with each treatment alone. Finally, the effects of EPO and MPSS on AQP4 expression were partially reversed by pretreatment with the PKC inhibitor Ro 31­8220. The present study indicated that EPO and MPSS had a synergistic effect on AQP4 expression following reperfusion, and suggest that they may be combined in the treatment of SCI.


Asunto(s)
Acuaporina 4/genética , Astrocitos/efectos de los fármacos , Eritropoyetina/farmacología , Hemisuccinato de Metilprednisolona/farmacología , ARN Mensajero/genética , Animales , Animales Recién Nacidos , Acuaporina 4/agonistas , Acuaporina 4/metabolismo , Astrocitos/citología , Astrocitos/metabolismo , Hipoxia de la Célula/genética , Supervivencia Celular/efectos de los fármacos , Femenino , Regulación de la Expresión Génica , Glucosa/deficiencia , Glucosa/farmacología , Hipotálamo/citología , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Indoles/farmacología , Fármacos Neuroprotectores/farmacología , Oxígeno/farmacología , Cultivo Primario de Células , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/genética , Proteína Quinasa C/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , ARN Mensajero/agonistas , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/genética , Daño por Reperfusión/metabolismo , Transducción de Señal
17.
Pharm Biol ; 55(1): 108-113, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27925507

RESUMEN

CONTEXT: Angelica sinensis L. (Umbelliferae) has medicinal properties. OBJECTIVES: The present study evaluates the haematopoietic effects of A. sinensis polysaccharides (ASP) against lisinopril-induced anaemia. MATERIALS AND METHODS: Thirty healthy adult male albino rats were randomly divided into five groups (n = 6). Group I was control group. Group II was treated with angiotensin-converting enzyme inhibitor (ACEI, 20 mg/kg/day) to induce anaemia. In group III, erythropoietin (EPO, 100 IU/kg/each) was administered in combination with ACEI. Group IV was treated with ASP (1 g/kg/day), extracted from A. sinensis root caps. In Group V, ASP (1 g/kg/day) was treated with ACEI. After 28 days, blood and tissue samples were collected for haematological and histopathological analysis, respectively. RESULTS: The results showed that ACEI significantly reduced the haemoglobin (Hb, 10.0 g/dL), packed cell volume (PCV, 39.5%), red blood cells (RBCs, 6.2 million/mm3), mean corpuscular volume (MCV, 53.5 fL) and mean corpuscular haemoglobin (MCH, 16.2 pg/cell) values. In the group treated with ASP, the Hb (13.7 g/dL) and RBCs (7.8 million/mm3) increased significantly (p < 0.05). The combination of ASP and ACEI led to the significant (p < 0.05) reduction in Hb (10.7 g/dL), PCV (33.3%), RBCs (6.0 million/mm3), MCV (54.42 fL) and MCH (16.44 pg/cell) values. While histopathological examination of the liver and kidney cells showed a mild degree of toxicity in the ASP-treated group. CONCLUSION: ASP has a potentiating effect on haematological parameters when given alone. However, when administered simultaneously with lisinopril, it showed an unfavourable effect with more complicated anaemia so it should not be used with ACEIs.


Asunto(s)
Anemia/tratamiento farmacológico , Angelica sinensis/química , Eritrocitos/efectos de los fármacos , Hematínicos/farmacología , Hematopoyesis/efectos de los fármacos , Lisinopril , Extractos Vegetales/farmacología , Cápsula de Raíz de Planta/química , Polisacáridos/farmacología , Anemia/sangre , Anemia/inducido químicamente , Animales , Biomarcadores/sangre , Modelos Animales de Enfermedad , Índices de Eritrocitos , Eritrocitos/metabolismo , Eritropoyetina/farmacología , Hematínicos/aislamiento & purificación , Hematínicos/toxicidad , Hematócrito , Hemoglobinas/metabolismo , Interacciones de Hierba-Droga , Masculino , Fitoterapia , Extractos Vegetales/aislamiento & purificación , Extractos Vegetales/toxicidad , Plantas Medicinales , Polisacáridos/aislamiento & purificación , Polisacáridos/toxicidad , Ratas Wistar , Factores de Tiempo
18.
Am J Physiol Endocrinol Metab ; 311(5): E859-E868, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27624103

RESUMEN

Numerous compounds stimulate rodent ß-cell proliferation; however, translating these findings to human ß-cells remains a challenge. To examine human ß-cell proliferation in response to such compounds, we developed a medium-throughput in vitro method of quantifying adult human ß-cell proliferation markers. This method is based on high-content imaging of dispersed islet cells seeded in 384-well plates and automated cell counting that identifies fluorescently labeled ß-cells with high specificity using both nuclear and cytoplasmic markers. ß-Cells from each donor were assessed for their function and ability to enter the cell cycle by cotransduction with adenoviruses encoding cell cycle regulators cdk6 and cyclin D3. Using this approach, we tested 12 previously identified mitogens, including neurotransmitters, hormones, growth factors, and molecules, involved in adenosine and Tgf-1ß signaling. Each compound was tested in a wide concentration range either in the presence of basal (5 mM) or high (11 mM) glucose. Treatment with the control compound harmine, a Dyrk1a inhibitor, led to a significant increase in Ki-67+ ß-cells, whereas treatment with other compounds had limited to no effect on human ß-cell proliferation. This new scalable approach reduces the time and effort required for sensitive and specific evaluation of human ß-cell proliferation, thus allowing for increased testing of candidate human ß-cell mitogens.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Activinas/farmacología , Adenosina/análogos & derivados , Adenosina/farmacología , Agonistas del Receptor de Adenosina A2/farmacología , Adenosina-5'-(N-etilcarboxamida)/farmacología , Adulto , Automatización , Técnicas de Cultivo de Célula , Evaluación Preclínica de Medicamentos , Eritropoyetina/farmacología , Exenatida , Femenino , GABAérgicos/farmacología , Harmina/farmacología , Humanos , Incretinas/farmacología , Masculino , Persona de Mediana Edad , Inhibidores de la Monoaminooxidasa/farmacología , Miostatina/farmacología , Nucleósidos/farmacología , Péptidos/farmacología , Factor de Crecimiento Derivado de Plaquetas/farmacología , Prolactina/farmacología , Regeneración/efectos de los fármacos , Serotonina/farmacología , Agonistas de Receptores de Serotonina/farmacología , Vasodilatadores/farmacología , Ponzoñas/farmacología , Adulto Joven , Ácido gamma-Aminobutírico/farmacología
19.
Exp Hematol ; 44(10): 975-81, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27364565

RESUMEN

Differentiating erythroblasts are exposed to an oxidative environment. The dynamics of oxidative status during terminal erythropoiesis and how they affect cell differentiation in response to erythropoietin (Epo) are unclear. Here, we show that Epo induces reactive oxygen species (ROS) production in the early stages of terminal erythropoiesis. The levels of ROS correlate with CD71 surface expression and the uptake of iron and transferrin. ROS decreases in the late stages of terminal erythropoiesis, when the cells are preparing for enucleation. Consistently, treatment of erythroblasts with a low dose (5 mM) of N-acetyl-cysteine (NAC), a ROS scavenger, promotes enucleation. However, a high dose (20 mM) of NAC leads to significant cell death. Our study reveals an important function of Epo in regulating the dynamics of oxidative status and enucleation.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Eritroblastos/citología , Eritroblastos/efectos de los fármacos , Eritropoyesis/efectos de los fármacos , Eritropoyetina/farmacología , Animales , Apoptosis , Biomarcadores , Eritroblastos/metabolismo , Feto , Factores de Crecimiento de Célula Hematopoyética/farmacología , Hemoglobinas/metabolismo , Inmunofenotipificación , Hierro/metabolismo , Hígado/citología , Ratones , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Transferrina/metabolismo
20.
Crit Rev Oncol Hematol ; 104: 98-107, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27317353

RESUMEN

The World Health Organization defines a biosimilar as "a biotherapeutic product which is similar in terms of quality, safety and efficacy to an already licensed reference biotherapeutic product." Biosimilars are biologic medical products that are very distinct from small-molecule generics, as their active substance is a biological agent derived from a living organism. Approval processes are highly regulated, with guidance issued by the European Medicines Agency and US Food and Drug Administration. Approval requires a comparability exercise consisting of extensive analytical and preclinical in vitro and in vivo studies, and confirmatory clinical studies. Extrapolation of biosimilars from their original indication to another is a feasible but highly stringent process reliant on rigorous scientific justification. This review focuses on the processes involved in gaining biosimilar approval and extrapolation and details the comparability exercise undertaken in the European Union between originator erythropoietin-stimulating agent, Eprex(®), and biosimilar, Retacrit™.


Asunto(s)
Biosimilares Farmacéuticos/farmacología , Eritropoyetina/farmacología , Animales , Biosimilares Farmacéuticos/uso terapéutico , Evaluación Preclínica de Medicamentos , Eritropoyetina/uso terapéutico , Unión Europea , Humanos , Estados Unidos , United States Food and Drug Administration
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA