Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
1.
Drug Test Anal ; 14(8): 1377-1386, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35322582

RESUMEN

The World Anti-Doping Agency (WADA) has recently implemented dried blood spots (DBSs) as a matrix for doping control. However, specifications regarding the analysis of the class of prohibited substances called erythropoietin (EPO) receptor agonists (ERAs) from DBSs are not yet described. The aim of this study was to find optimal conditions (sample volume and storage) to sensitively detect endogenous erythropoietin (hEPO) and prohibited ERAs from DBSs and compare detection limits to WADA-stipulated minimum required performance levels (MRPLs) for ERAs in serum/plasma samples. Venous whole blood was spotted onto Whatman 903 DBS cards with primarily 60 µl of blood, but various volumes from 20 to75 µl were tested. All samples were immunopurified with MAIIA EPO Purification Gel kit (EPGK) and analysed with sodium N-lauroylsarcosinate polyacrylamide gel electrophoresis (SAR-PAGE) and Western blot. Sixty-microliter DBSs allowed the detection of the four main ERAs (BRP, NESP, CERA and EPO-Fc) at concentrations close to WADA's MRPLs described for 500 µl of serum/plasma. Different storage temperatures, from -20°C to 37°C, were evaluated and did not affect ERA detection. A comparison of the detection of endogenous EPO from the different anti-doping matrices (urine, serum and DBSs produced from upper arm capillary blood) from five participants for 6 weeks was performed. Endogenous EPO extracted from DBSs showed intra-individual variations in male and female subjects, but less than in urine. Doping controls would benefit from the stability of ERAs on DBSs: It can be a complementary matrix for ERA analysis, particularly in the absence of EPO signals in urine.


Asunto(s)
Doping en los Deportes , Hematínicos , Receptores de Eritropoyetina , Detección de Abuso de Sustancias , Pruebas con Sangre Seca , Electroforesis en Gel de Poliacrilamida , Femenino , Hematínicos/análisis , Humanos , Masculino , Receptores de Eritropoyetina/agonistas , Detección de Abuso de Sustancias/métodos
2.
Cell Death Dis ; 12(2): 177, 2021 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-33579907

RESUMEN

Erythropoietin (EPO) is not only an erythropoiesis hormone but also an immune-regulatory cytokine. The receptors of EPO (EPOR)2 and tissue-protective receptor (TPR), mediate EPO's immune regulation. Our group firstly reported a non-erythropoietic peptide derivant of EPO, cyclic helix B peptide (CHBP), which could inhibit macrophages inflammation and dendritic cells (DCs) maturation. As a kind of innate immune regulatory cell, myeloid-derived suppressor cells (MDSCs) share a common myeloid progenitor with macrophages and DCs. In this study, we investigated the effects on MDSCs differentiation and immunosuppressive function via CHBP induction. CHBP promoted MDSCs differentiate toward M-MDSCs with enhanced immunosuppressive capability. Infusion of CHBP-induced M-MDSCs significantly prolonged murine skin allograft survival compared to its counterpart without CHBP stimulation. In addition, we found CHBP increased the proportion of CD11b+Ly6G-Ly6Chigh CD127+ M-MDSCs, which exerted a stronger immunosuppressive function compared to CD11b+Ly6G-Ly6Chigh CD127- M-MDSCs. In CHBP induced M-MDSCs, we found that EPOR downstream signal proteins Jak2 and STAT3 were upregulated, which had a strong relationship with MDSC function. In addition, CHBP upregulated GATA-binding protein 3 (GATA-3) protein translation level, which was an upstream signal of CD127 and regulator of STAT3. These effects of CHBP could be reversed if Epor was deficient. Our novel findings identified a new subset of M-MDSCs with better immunosuppressive capability, which was induced by the EPOR-mediated Jak2/GATA3/STAT3 pathway. These results are beneficial for CHBP clinical translation and MDSC cell therapy in the future.


Asunto(s)
Rechazo de Injerto/prevención & control , Supervivencia de Injerto/efectos de los fármacos , Inmunosupresores/farmacología , Subunidad alfa del Receptor de Interleucina-7/metabolismo , Células Supresoras de Origen Mieloide/efectos de los fármacos , Células Supresoras de Origen Mieloide/trasplante , Receptores de Eritropoyetina/agonistas , Trasplante de Piel , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular/efectos de los fármacos , Factor de Transcripción GATA3/metabolismo , Rechazo de Injerto/inmunología , Rechazo de Injerto/metabolismo , Janus Quinasa 2/metabolismo , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Células Supresoras de Origen Mieloide/inmunología , Células Supresoras de Origen Mieloide/metabolismo , Fenotipo , Receptores de Eritropoyetina/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Trasplante de Piel/efectos adversos , Factores de Tiempo
3.
J Cardiovasc Pharmacol ; 73(5): 290-300, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31082960

RESUMEN

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.


Asunto(s)
Sistema Cardiovascular/efectos de los fármacos , Epoetina alfa/administración & dosificación , Hematínicos/administración & dosificación , Hemodinámica/efectos de los fármacos , Hemorragia/tratamiento farmacológico , Función Ventricular Izquierda/efectos de los fármacos , Factores de Edad , Animales , Sistema Cardiovascular/metabolismo , Sistema Cardiovascular/fisiopatología , Modelos Animales de Enfermedad , Hemorragia/metabolismo , Hemorragia/fisiopatología , Peroxidación de Lípido/efectos de los fármacos , Masculino , Miocardio/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ratas Sprague-Dawley , Receptores de Eritropoyetina/agonistas , Receptores de Eritropoyetina/metabolismo
4.
Nephron ; 140(1): 24-30, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29945128

RESUMEN

BACKGROUND: Clinicians providing dialysis care have numerous erythropoiesis-stimulating agents (ESAs) available for treating anemia. We sought to provide a contemporary description of ESA types used in hemodialysis (HD) settings in nine European countries. METHODS: Our study uses Dialysis Outcomes and Practice Patterns Study phase 5 (2012-2015) data from nine European countries (Belgium, France, -Germany, Italy, Russia, Spain, Sweden, Turkey, and the United Kingdom). A total of 164 facilities and 3,281 patients contributed cross-sectional data. ESA types captured included short-acting epoetins (e.g., epoetin alfa, beta, etc., including biosimilars), darbepoetin alfa, and continuous erythropoietin receptor agonist (CERA; methoxy polyethylene glycol-epoetin beta). RESULTS: We observed broad variability across countries in prescription of ESA types: prescription of epoetin alfa or epoetin beta ranged from 22% (France) to 78% (Russia), darbepoetin alfa prescription ranged from 13% (Russia) to 53% (UK), and CERA prescription ranged from <3% (Sweden) to 26% (France). Prescription of different ESA types varied substantially within some European countries from 2012-2015 but not across all countries in aggregate. Number of ESA types prescribed by a facility varied from 1, 2, 3, or 4 different ESA types in 32, 40, 21, and 8% of facilities, respectively. No differences were seen in the unadjusted distributions of achieved hemoglobin values by ESA type. CONCLUSION: A variety of short- and long-acting ESAs are commonly used in European HD facilities to maintain hemoglobin at remarkably similar levels with each ESA type. The availability of numerous ESA options for managing anemia has allowed European providers to optimize anemia management according to the particular circumstances of each patient.


Asunto(s)
Eritropoyetina/uso terapéutico , Hematínicos/uso terapéutico , Diálisis Renal , Anemia/tratamiento farmacológico , Anemia/etiología , Biosimilares Farmacéuticos , Estudios de Cohortes , Estudios Transversales , Eritropoyetina/administración & dosificación , Eritropoyetina/análogos & derivados , Europa (Continente) , Hemoglobinas/análisis , Humanos , Fallo Renal Crónico/complicaciones , Fallo Renal Crónico/terapia , Pautas de la Práctica en Medicina , Estudios Prospectivos , Receptores de Eritropoyetina/agonistas , Diálisis Renal/efectos adversos , Resultado del Tratamiento
5.
Am J Physiol Regul Integr Comp Physiol ; 314(5): R700-R708, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29443550

RESUMEN

Recent studies indicate that erythropoietin (EPO) is present in many areas of the brain and is active in the restoration of impaired neurons. In this study, we examined the presence of EPO and its role in bulbospinal neurons in the rostral ventrolateral medulla (RVLM). Hypoxia is often accompanied by a high blood pressure (BP). We hypothesized that EPO is produced in response to hypoxia in RVLM neurons and then activates them. To investigate whether RVLM neurons are sensitive to EPO, we examined the changes in the membrane potentials (MPs) of bulbospinal RVLM neurons using the whole cell patch-clamp technique during superfusion with EPO. A brainstem-spinal cord preparation was used for the experiments. EPO depolarized the RVLM neurons, and soluble erythropoietin receptor (SEPOR), an antagonist of EPO, hyperpolarized them. Furthermore, hypoxia-depolarized RVLM neurons were significantly hyperpolarized by SEPOR. In histological examinations, the EPO-depolarized RVLM neurons showed the presence of EPO receptor (EPOR). The RVLM neurons that possessed EPORs showed the presence of EPO and hypoxia-inducible factor (HIF)-2α. We also examined the levels of HIF-2α and EPO messenger RNA (mRNA) in the ventral sites of the medullas (containing RVLM areas) in response to hypoxia. The levels of HIF-2α and EPO mRNA in the hypoxia group were significantly greater than those in the control group. These results suggest that EPO is produced in response to hypoxia in RVLM neurons and causes a high BP via the stimulation of those neurons. EPO may be one of the neurotransmitters produced by RVLM neurons during hypoxia.


Asunto(s)
Eritropoyetina/metabolismo , Bulbo Raquídeo/metabolismo , Neuronas/metabolismo , Potenciales de Acción , Animales , Animales Recién Nacidos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Presión Sanguínea , Hipoxia de la Célula , Eritropoyetina/genética , Eritropoyetina/farmacología , Hipertensión/etiología , Hipertensión/metabolismo , Hipertensión/fisiopatología , Hipoxia/complicaciones , Hipoxia/metabolismo , Hipoxia/fisiopatología , Técnicas In Vitro , Bulbo Raquídeo/efectos de los fármacos , Bulbo Raquídeo/fisiopatología , Neuronas/efectos de los fármacos , Ratas Wistar , Receptores de Eritropoyetina/agonistas , Receptores de Eritropoyetina/metabolismo , Regulación hacia Arriba
6.
Kidney Int ; 93(5): 1131-1141, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29395333

RESUMEN

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.


Asunto(s)
Lesión Renal Aguda/sangre , Células de la Médula Ósea/metabolismo , Células Precursoras Eritroides/metabolismo , Eritropoyetina/sangre , Factores de Crecimiento de Fibroblastos/sangre , Lesión Renal Aguda/etiología , Animales , Células de la Médula Ósea/efectos de los fármacos , Modelos Animales de Enfermedad , Células Precursoras Eritroides/efectos de los fármacos , Eritropoyetina/farmacología , Factor-23 de Crecimiento de Fibroblastos , Humanos , Masculino , Ratones Endogámicos C57BL , Estudios Prospectivos , Ratas Sprague-Dawley , Receptores de Eritropoyetina/agonistas , Receptores de Eritropoyetina/metabolismo , Proteínas Recombinantes/farmacología , Sepsis/sangre , Sepsis/complicaciones , Choque Hemorrágico/sangre , Choque Hemorrágico/complicaciones , Factores de Tiempo , Regulación hacia Arriba
7.
Vitam Horm ; 105: 19-37, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28629518

RESUMEN

Amgen solved the high-resolution cocrystal structure of erythropoietin (EPO) bound to the extracellular part of the receptor (EPOR) in 1998, which reveals that the EPO-EPOR interaction surface is formed by 11 salt bridges, 17 H-bonds, and 2 hydrophobic clusters centered at a pair of crucial phenylalanines (F93). The EPOR has two domains, one that penetrates the membrane and a second extracellular domain that forms one arm of the binding site for the EPO ligand. The complete competent receptor-binding site is a homodimer of EPOR with the two arms forming a funnel-shaped cup where EPO binds. The two binding arms of the EPOR dimer meet at the membrane at a 120 degree angle, which Amgen characterizes as, "erythropoietin imposes a unique angular relationship and orientation that is responsible for optimal signaling." They come to this conclusion, because the EPOR cocrystallized with 2 equivalents of a 20 residue EPO mimetic peptide created at Robert Wood Johnson (RWJ) activates the receptor with a 3 order of magnitude reduction in potency, and the binding arms are forced to meet at the membrane with an angle of 180 degrees. The vast interaction surface between EPO and EPOR forms a singularly important three-dimensional structure responsible for hematopoietic stem cell proliferation and differentiation-this is Amgen's conclusion. This goal of this work is to present experimental and computational evidence that the Amgen structure is a postsignaling off-state and that the RWJ structure with the partially active peptide mimetics is an on-state. A detailed side-by-side comparison of the two structures will be presented along with literature evidence that calls into question the Amgen claim that their structure is a unique on-state. A computational fragment-based drug discovery method applied to the RWJ structure was used to locate and characterize a new predicted small molecule binding site and a fragment analysis was performed based on theories of asymmetry to create a proposed agonist with MW<300. When this molecule was experimentally tested, it displaced radiolabeled EPO with nanomolar potency and transformed human hematopoietic stem cells into red blood cells with subnanomolar potency. Obviously, this small molecule makes none of the EPO-EPOR interactions that Amgen stated were essential for fully turning on the receptor and provides strong evidence that stabilizing receptor asymmetry, not specific interactions, is the critical factor needed for activating signal transduction. Finally, when the agonist was altered to remove the asymmetric component, it still was able to displace radiolabeled EPO in competition binding experiments, but it no longer activated the receptor.


Asunto(s)
Diseño de Fármacos , Eritropoyetina/química , Receptores de Eritropoyetina/agonistas , Receptores de Eritropoyetina/química , Animales , Sitios de Unión , Humanos , Unión Proteica , Conformación Proteica , Receptores de Eritropoyetina/metabolismo , Transducción de Señal/fisiología
8.
Neuropharmacology ; 123: 34-45, 2017 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-28504123

RESUMEN

Friedreich's ataxia (FA) is a progressive neurodegenerative disease caused by reduced levels of the mitochondrial protein frataxin (FXN). Recombinant human erythropoietin (rhEPO) increased FXN protein in vitro and in early clinical studies, while no published reports evaluate rhEPO in animal models of FA. STS-E412 and STS-E424 are novel small molecule agonists of the tissue-protective, but not the erythropoietic EPO receptor. We find that rhEPO, STS-E412 and STS-E424 increase FXN expression in vitro and in vivo. RhEPO, STS-E412 and STS-E424 increase FXN by up to 2-fold in primary human cortical cells and in retinoic-acid differentiated murine P19 cells. In primary human cortical cells, the increase in FXN protein was accompanied by an increase in FXN mRNA, detectable within 4 h. RhEPO and low nanomolar concentrations of STS-E412 and STS-E424 also increase FXN in normal and FA patient-derived PBMC by 20%-40% within 24 h, an effect that was comparable to that by HDAC inhibitor 4b. In vivo, STS-E412 increased Fxn mRNA and protein in wild-type C57BL6/j mice. RhEPO, STS-E412, and STS-E424 increase FXN expression in the heart of FXN-deficient KIKO mice. In contrast, FXN expression in the brains of KIKO mice increased following treatment with STS-E412 and STS-E424, but not following treatment with rhEPO. Unexpectedly, rhEPO-treated KIKO mice developed severe splenomegaly, while no splenomegaly was observed in STS-E412- or STS-E424-treated mice. RhEPO, STS-E412 and STS-E424 upregulate FXN expression in vitro at equal efficacy, however, the effects of the small molecules on FXN expression in the CNS are superior to rhEPO in vivo.


Asunto(s)
Eritropoyetina/farmacología , Fármacos Neuroprotectores/farmacología , Pirimidinas/farmacología , Receptores de Eritropoyetina/agonistas , Proteínas Recombinantes/farmacología , Triazoles/farmacología , Adulto , Animales , Células Cultivadas , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Femenino , Ataxia de Friedreich/tratamiento farmacológico , Ataxia de Friedreich/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Proteínas de Unión a Hierro/genética , Proteínas de Unión a Hierro/metabolismo , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Persona de Mediana Edad , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Receptores de Eritropoyetina/metabolismo , Adulto Joven , Frataxina
9.
J Neurochem ; 141(1): 63-74, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28142212

RESUMEN

Erythropoietin (Epo) plays a dual role as an erythropoiesis-stimulating hormone and a locally produced cytoprotectant in various vertebrate tissues. Splice variants and engineered derivatives of Epo that mediate neuroprotection but do not stimulate erythropoiesis suggest that alternative receptors, different from the 'classical' homodimeric receptor involved in haematopoiesis, mediate neuroprotective Epo functions. Previous studies on grasshoppers demonstrated neuroprotective and neuroregenerative effects of Epo that involved similar transduction pathways as in mammals. To advance the characterization of yet unidentified neuroprotective Epo receptors, we studied the neuroprotective potency of the human non-erythropoietic Epo splice variant EV-3 in primary cultured locust brain neurons. We demonstrate that EV-3, like Epo, protects locust neurons from hypoxia-induced apoptotic death through activation of the Janus kinase/signal transducer and activator of transcription transduction pathway. Using the fluorescent dye FM1-43 to quantify endocytotic activity we show that both Epo and EV-3 increase the number of fluorescently labelled endocytotic vesicles. This reveals that binding of Epo to its neuroprotective receptor induces endocytosis, as it has been described for the mammalian homodimeric Epo-receptor expressed by erythroid progenitors. Reduction in Epo-stimulated endocytotic activity following pre-exposure to EV-3 indicated that both Epo and its splice variant bind to the same receptor on locust neurons. The shared neuroprotective potency of Epo and EV-3 in insect and mammalian neurons, in the absence of erythropoietic effects of EV-3 in mammals, suggests a greater similarity of the unidentified nervous Epo receptors (or receptor complexes) across phyla than between mammalian haematopoietic and neuroprotective receptors. Insects may serve as suitable models to evaluate the specific protective mechanisms mediated by Epo and its variants in non-erythropoietic mammalian tissues.


Asunto(s)
Encéfalo/metabolismo , Endocitosis/fisiología , Neuroprotección/fisiología , Receptores de Eritropoyetina/metabolismo , Animales , Encéfalo/efectos de los fármacos , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Endocitosis/efectos de los fármacos , Eritropoyetina/metabolismo , Eritropoyetina/farmacología , Femenino , Humanos , Insectos , Locusta migratoria , Masculino , Neuroprotección/efectos de los fármacos , Receptores de Eritropoyetina/agonistas , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología
10.
Nefrologia ; 36(3): 232-6, 2016.
Artículo en Inglés, Español | MEDLINE | ID: mdl-27137103
11.
Shock ; 46(4): 390-7, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27172159

RESUMEN

BACKGROUND: Alternate erythropoietin (EPO)-mediated signaling via the EPOR/CD131 heteromeric receptor exerts the tissue-protective actions of EPO in a wide spectrum of injuries, especially ischemic diseases. Circulating endothelial progenitor cells contribute to endothelial repair and post-natal angiogenesis after chronic ischemic injury. This work aims to investigate the effects of ARA290, a specific agonist of EPOR/CD131 complex, on a subpopulation of endothelial progenitor cells named endothelial colony-forming cells (ECFCs) and to characterize its contribution to ECFCs-induced angiogenesis after peripheral ischemia. METHODS: ARA290 effects on ECFCs properties were studied using cell cultures in vitro. We injected ARA290 to mice undergoing chronic hindlimb ischemia (CLI) in combination with ECFC transplantation. The homing of transplanted ECFC to ischemic tissue in vivo was assessed by SPECT/CT imaging. RESULTS: In vitro, ARA290 enhanced the proliferation, migration, and resistance to H2O2-induced apoptosis of ECFCs. After ECFC transplantation to mice with CLI, a single ARA290 injection enhanced the ischemic/non-ischemic ratio of hindlimb blood flow and capillary density after 28 days and the homing of radiolabeled transplanted cells to the ischemic leg 4 h after transplantation. Prior neutralization of platelet-endothelial cell adhesion molecule-1 (CD31) expressed by the transplanted cells inhibited ARA290-induced improvement of homing. DISCUSSION: ARA290 induces specific improvement of the biological activity of ECFCs. ARA290 administration in combination with ECFCs has a synergistic effect on post-ischemic angiogenesis in vivo. This potentiation appears to rely, at least in part, on a CD31-dependent increase in homing of the transplanted cells to the ischemic tissue.


Asunto(s)
Subunidad beta Común de los Receptores de Citocinas/metabolismo , Oligopéptidos/farmacología , Receptores de Eritropoyetina/metabolismo , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Células Cultivadas , Subunidad beta Común de los Receptores de Citocinas/antagonistas & inhibidores , Células Progenitoras Endoteliales/efectos de los fármacos , Células Progenitoras Endoteliales/metabolismo , Miembro Posterior/metabolismo , Miembro Posterior/patología , Humanos , Peróxido de Hidrógeno/farmacología , Isquemia/metabolismo , Isquemia/patología , Ratones , Estrés Oxidativo/efectos de los fármacos , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Receptores de Eritropoyetina/agonistas , Transducción de Señal/efectos de los fármacos , Tomografía Computarizada por Tomografía Computarizada de Emisión de Fotón Único
13.
Mol Pharmacol ; 88(2): 357-67, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26018904

RESUMEN

Erythropoietin (EPO) and its receptor are expressed in a wide variety of tissues, including the central nervous system. Local expression of both EPO and its receptor is upregulated upon injury or stress and plays a role in tissue homeostasis and cytoprotection. High-dose systemic administration or local injection of recombinant human EPO has demonstrated encouraging results in several models of tissue protection and organ injury, while poor tissue availability of the protein limits its efficacy. Here, we describe the discovery and characterization of the nonpeptidyl compound STS-E412 (2-[2-(4-chlorophenoxy)ethoxy]-5,7-dimethyl-[1,2,4]triazolo[1,5-a]pyrimidine), which selectively activates the tissue-protective EPO receptor, comprising an EPO receptor subunit (EPOR) and the common ß-chain (CD131). STS-E412 triggered EPO receptor phosphorylation in human neuronal cells. STS-E412 also increased phosphorylation of EPOR, CD131, and the EPO-associated signaling molecules JAK2 and AKT in HEK293 transfectants expressing EPOR and CD131. At low nanomolar concentrations, STS-E412 provided EPO-like cytoprotective effects in primary neuronal cells and renal proximal tubular epithelial cells. The receptor selectivity of STS-E412 was confirmed by a lack of phosphorylation of the EPOR/EPOR homodimer, lack of activity in off-target selectivity screening, and lack of functional effects in erythroleukemia cell line TF-1 and CD34(+) progenitor cells. Permeability through artificial membranes and Caco-2 cell monolayers in vitro and penetrance across the blood-brain barrier in vivo suggest potential for central nervous system availability of the compound. To our knowledge, STS-E412 is the first nonpeptidyl, selective activator of the tissue-protective EPOR/CD131 receptor. Further evaluation of the potential of STS-E412 in central nervous system diseases and organ protection is warranted.


Asunto(s)
Encéfalo/embriología , Eritropoyetina/metabolismo , Neuronas/metabolismo , Pirimidinas/farmacología , Pirimidinas/farmacocinética , Receptores de Eritropoyetina/agonistas , Triazoles/farmacología , Triazoles/farmacocinética , Animales , Disponibilidad Biológica , Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/citología , Células CACO-2 , Células Cultivadas , Subunidad beta Común de los Receptores de Citocinas/metabolismo , Células HEK293 , Humanos , Ratas , Transducción de Señal/efectos de los fármacos
14.
Curr Top Med Chem ; 15(10): 955-69, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25832721

RESUMEN

Parkinson's disease (PD) is a neurodegenerative disease affecting more than a million people in the USA alone. While there are effective symptomatic treatments for PD, there is an urgent need for new therapies that slow or halt the progressive death of dopaminergic neurons. Significant progress has been made in understanding the pathophysiology of PD, which has substantially facilitated the discovery efforts to identify novel drugs. The tissue-protective erythropoietin (EPO) receptor, EPOR/CD131, has emerged as one promising target for disease-modifying therapies. Recombinant human EPO (rhEPO), several variants of EPO, EPO-mimetic peptides, cell-based therapies using cells incubated with or expressing EPO, gene therapy vectors encoding EPO, and small molecule EPO mimetic compounds all show potential as therapeutic candidates. Agonists of the EPOR/CD131 receptor demonstrate potent anti-apoptotic, antioxidant, and anti-inflammatory effects and protect neurons, including dopaminergic neurons, from diverse insults in vitro and in vivo. When delivered directly to the striatum, rhEPO protects dopaminergic neurons in animal models of PD. Early-stage clinical trials testing systemic rhEPO have provided encouraging results, while additional controlled studies are required to fully assess the potential of the treatment. Poor CNS availability of proteins and challenges related to invasive delivery limit delivery of EPO protein. Several variants of EPO and small molecule agonists of the EPO receptors are making progress in preclinical studies and may offer solutions to these challenges. While EPO was initially discovered as the primary modulator of erythropoiesis, the discovery and characterization of the tissue-protective EPOR/CD131 receptor offer an opportunity to selectively target the neuroprotective receptor as an approach to identify disease-modifying treatments for PD.


Asunto(s)
Antiparkinsonianos/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Enfermedad de Parkinson/tratamiento farmacológico , Receptores de Eritropoyetina/agonistas , Animales , Humanos
15.
Cell ; 160(6): 1196-208, 2015 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-25728669

RESUMEN

Most cell-surface receptors for cytokines and growth factors signal as dimers, but it is unclear whether remodeling receptor dimer topology is a viable strategy to "tune" signaling output. We utilized diabodies (DA) as surrogate ligands in a prototypical dimeric receptor-ligand system, the cytokine Erythropoietin (EPO) and its receptor (EpoR), to dimerize EpoR ectodomains in non-native architectures. Diabody-induced signaling amplitudes varied from full to minimal agonism, and structures of these DA/EpoR complexes differed in EpoR dimer orientation and proximity. Diabodies also elicited biased or differential activation of signaling pathways and gene expression profiles compared to EPO. Non-signaling diabodies inhibited proliferation of erythroid precursors from patients with a myeloproliferative neoplasm due to a constitutively active JAK2V617F mutation. Thus, intracellular oncogenic mutations causing ligand-independent receptor activation can be counteracted by extracellular ligands that re-orient receptors into inactive dimer topologies. This approach has broad applications for tuning signaling output for many dimeric receptor systems.


Asunto(s)
Receptores de Eritropoyetina/química , Receptores de Eritropoyetina/metabolismo , Transducción de Señal , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/metabolismo , Línea Celular , Cristalografía por Rayos X , Dimerización , Eritropoyetina/metabolismo , Humanos , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Ratones , Modelos Moleculares , Simulación de Dinámica Molecular , Datos de Secuencia Molecular , Mutación Puntual , Ingeniería de Proteínas , Receptores de Eritropoyetina/agonistas , Receptores de Eritropoyetina/antagonistas & inhibidores , Alineación de Secuencia
16.
Nephrology (Carlton) ; 20(4): 266-72, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25581532

RESUMEN

AIM: Renal ischaemia-reperfusion (I/R) injury, a primary cause of acute renal failure, can induce high morbidity and mortality. This study aimed to explore the effect of erythropoietin on renal I/R injury and its underlying mechanism. METHODS: Fifty male Sprague-Dawley rats were randomly allocated to three groups (n = 10): the sham group, the renal ischaemia-reperfusion-saline (IRI) group, and the IRI+-Erythropoietin (EPO) group. Erythropoietin (250, 500, 1000 U/kg) was intraperitoneally injected 30 min before inducing I/R. Renal I/R injury were induced by clamping the left renal artery for 30 min followed by reperfusion, along with a contralateral nephrectomy. Renal function and histological damage were determined after 24 h reperfusion. The expression of pro-inflammatory cytokines interleukin-6 (IL-6), interleukin-1 ß (IL-1ß), and tumour necrosis factor-α (TNF-α) in the serum and renal tissue were evaluated by enzyme linked immunosorbent assay (ELISA) and reverse transcription-polymerase chain reaction (RT-PCR), respectively. Further, the effects of erythropoietin on PI3K/Akt signalling, erythropoietin receptor (EPOR) and nuclear factor (NF)-κB activation were measured by Western blotting. RESULTS: Erythropoietin pretreatment can significantly decrease the level of renal dysfunction in a dose-dependent manner, attenuated the renal histological changes, the expression of TNF-α, IL-1ß, and IL-6, the levels of reactive oxygen species (ROS) production and NF-κB p65 phosphorylation in renal tissue upon IRI. Moreover, erythropoietin pretreatment could further activate the PI3K/Akt signalling and induced EPOR activity. CONCLUSIONS: Erythropoietin pretreatment could attenuate renal I/R injury by suppressing inflammation, which was associated with activating PI3K/Akt signalling though EPOR activation. Our findings suggest that erythropoietin may be a novel practical strategy to prevent renal I/R injury.


Asunto(s)
Lesión Renal Aguda/prevención & control , Antiinflamatorios/administración & dosificación , Eritropoyetina/administración & dosificación , Riñón/efectos de los fármacos , Fosfatidilinositol 3-Quinasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Daño por Reperfusión/prevención & control , Transducción de Señal/efectos de los fármacos , Lesión Renal Aguda/sangre , Lesión Renal Aguda/enzimología , Lesión Renal Aguda/patología , Animales , Biomarcadores/sangre , Citoprotección , Modelos Animales de Enfermedad , Activación Enzimática , Mediadores de Inflamación/sangre , Interleucina-1beta/sangre , Interleucina-6/sangre , Riñón/enzimología , Riñón/patología , Masculino , Fosforilación , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Receptores de Eritropoyetina/agonistas , Receptores de Eritropoyetina/metabolismo , Daño por Reperfusión/sangre , Daño por Reperfusión/enzimología , Daño por Reperfusión/patología , Factor de Transcripción ReIA/metabolismo , Factor de Necrosis Tumoral alfa/sangre
17.
J Pharm Biomed Anal ; 100: 316-321, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25194345

RESUMEN

Accurate determination of in vitro biological activity of therapeutic erythropoietin is essential in quality control of recombinant human erythropoietin (rHuEPO) pharmaceutical products. However, most of currently-used methods leave much to be desired so that a simpler, quicker and more accurate method is urgently needed. The bioassay described here utilizes a sub clone of UT-7/epo cell line stably transfected with luciferase gene under the control of sis inducible element and interferon γ-activated sequence element promoter. Active erythropoietin could induce the expression of luciferase by signaling through the erythropoietin receptor and the dose-response curve showed good linearity, yielding a coefficient of determination of 0.99 or higher. The optimized assay was simpler with the operation completed within 24h and more sensitive with EC50 being 0.077IU/mL. The accuracy estimates ranged from 81.7% to 102.4%, and both intra-assay and inter-assay precision was below 15.0%. The robustness of the assay was demonstrated by no effect of passage levels of the cells on the performance of the assay (p values: 0.772 for sample 1 and 0.943 for sample 2). Besides, Bland-Altman analysis showed a high consistency of the new assay with in vivo reticulocyte assay in results. These results suggested that the new reporter gene assay can be a viable supplement to the traditional reticulocyte assay and employed in potency determination of rHuEPO pharmaceutical products.


Asunto(s)
Bioensayo/métodos , Eritropoyetina/farmacología , Genes Reporteros , Luciferasas/biosíntesis , Animales , Bioensayo/normas , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Luciferasas/genética , Ratones Endogámicos BALB C , Regiones Promotoras Genéticas/efectos de los fármacos , Receptores de Eritropoyetina/agonistas , Receptores de Eritropoyetina/metabolismo , Proteínas Recombinantes/farmacología , Reproducibilidad de los Resultados , Reticulocitos/efectos de los fármacos , Reticulocitos/metabolismo , Transfección
18.
Exp Clin Transplant ; 12(3): 220-6, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24907722

RESUMEN

OBJECTIVES: Posttransplant anemia is associated with an increased risk of congestive heart failure, left ventricular hypertrophy, and death. The purpose of this study was to evaluate the effect of long-acting erythropoietin-stimulating agents on anemia after kidney transplant. MATERIALS AND METHODS: In 2306 kidney transplant recipients, 250 anemic patients (11%) with stable graft function were followed at the Hamed Al-Essa Organ Transplant Centre (Kuwait) and were assessed for anemia. We enrolled 120 patients into this open-label study in 2 groups: group 1 had treatment with darbepoetin alfa (86 patients) and group 2 had continuous erythropoietin receptor activator (34 patients). RESULTS: Patient age correlated negatively with serum iron level. Serum ferritin correlated negatively with hemoglobin level 6 months after transplant but not at time of transplant. Most patients were women who received their grafts from male donors. The 2 groups were comparable in frequency of rejection and mean hemoglobin and serum albumin levels at 3, 6, 9, and 12 months after transplant. There was no difference between the 2 groups in renal function (estimated glomerular filtration rate); posttransplant complications such as new-onset diabetes after transplant, hypertension, serious bacterial infections, or patient and graft outcomes. CONCLUSIONS: Anemia is an important problem after kidney transplant, and iron use is suboptimal in kidney transplant recipients. Darbepoetin alfa and continuous erythropoietin receptor activator had comparable positive results.


Asunto(s)
Anemia/tratamiento farmacológico , Eritropoyetina/análogos & derivados , Hematínicos/uso terapéutico , Trasplante de Riñón/efectos adversos , Polietilenglicoles/uso terapéutico , Receptores de Eritropoyetina/agonistas , Adulto , Anemia/sangre , Anemia/diagnóstico , Anemia/etiología , Biomarcadores/sangre , Darbepoetina alfa , Eritropoyetina/uso terapéutico , Femenino , Humanos , Kuwait , Masculino , Persona de Mediana Edad , Receptores de Eritropoyetina/metabolismo , Factores de Tiempo , Resultado del Tratamiento , Adulto Joven
19.
Biochem J ; 459(3): 455-66, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24552351

RESUMEN

Erythroid homoeostasis is primarily controlled by Epo (erythropoietin) receptor signalling; however, the Lyn tyrosine kinase plays an important subsidiary role in regulating the erythroid compartment. Nonetheless, specific erythroid pathways that require Lyn activity and their biological significance remain unclear. To address this, we asked what consequence loss of Lyn had on the ex vivo expansion and maturation of splenic erythroid progenitors and Epo receptor signalling. Pharmacological inhibition of Lyn with PP2 inhibited the survival of terminally differentiated erythroblasts. Less committed erythroid progenitors expanded well, whereas early splenic Lyn(-/-) erythroblasts had attenuated ex vivo expansion, and late stage Lyn(-/-) erythroblasts were retarded in completing morphological maturation ex vivo. Furthermore, immortalized Lyn(-/-) erythroblasts were slower growing, less viable and inhibited in their differentiation. Signalling studies showed that Lyn was required for both positive GAB2/Akt/FoxO3 (forkhead box O3) survival signals as well as negative feedback of JAK2 (Janus kinase 2)/STAT5 (signal transducer and activator of transcription 5) and ERK1/2 (extracellular-signal-regulated kinase 1/2) signals via SHP-1 (Src homology 2 domain-containing protein tyrosine phosphatase 1). During differentiation, Lyn controls survival and cell cycle exit as demonstrated by reduced STAT5 and FoxO3/GSKα/ß (glycogen synthase kinase α/ß) phosphorylation and diminished p27(Kip1) induction in Lyn-deficient erythroblasts. Lyn deficiency alters the balance of pro- and anti-apoptotic molecules (BAD and BclXL), thereby reducing survival and preventing cell cycle exit. Consequently, Lyn facilitates normal erythrocyte production by influencing different stages of erythroid progenitor expansion, and mature cell development and survival signalling.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Eritroblastos/metabolismo , Células Precursoras Eritroides/metabolismo , Eritropoyesis , Receptores de Eritropoyetina/metabolismo , Transducción de Señal , Familia-src Quinasas/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/agonistas , Proteínas Reguladoras de la Apoptosis/antagonistas & inhibidores , Diferenciación Celular/efectos de los fármacos , Línea Celular Transformada , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Embrión de Mamíferos/citología , Eritroblastos/citología , Eritroblastos/efectos de los fármacos , Células Precursoras Eritroides/citología , Células Precursoras Eritroides/efectos de los fármacos , Eritropoyesis/efectos de los fármacos , Eritropoyetina/farmacología , Hematínicos/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Receptores de Eritropoyetina/agonistas , Transducción de Señal/efectos de los fármacos , Bazo/citología , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/genética
20.
J Nephrol ; 26(5): 829-35, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24052463

RESUMEN

Erythropoietin (Epo) has long been known to be the principal hematopoietic growth factor that regulates cellular proliferation and differentiation of the erythroid lineage. Recombinant Epo (rEpo) is frequently used in the prevention and treatment of anemia of chronic kidney disease (CKD) and in the treatment of anemia associated with cancer. The expression of the Epo receptor (EpoR) in certain cancer cells has raised the possibility that exogenous rEpo may have direct effects on tumor cells with potential for stimulation of proliferation and resistance to chemotherapy and possibly metastasis. With recent clinical trials reporting negative outcomes with rEpo in the treatment of cancer-related anemia, with increased mortality in those who received rEpo, it has become necessary for nephrologists to analyze and review the effect of use of rEpo in treatment of anemia of CKD in patients who may have current or previously treated malignancy. It is known that CKD patients have an increased prevalence of certain malignancies (Wong G et al. Association of CKD and cancer risk in older people. J Am Soc Nephrol. 2009;20(6):1341-1350). If a CKD patient develops cancer or has a previous history of cancer, there is a dilemma which faces the treating nephrologist with respect to the use of rEpo in this situation. So far this issue has been addressed infrequently in the nephrology literature, and the guidelines are unclear. This review describes Epo and EpoR biology, reviews use of rEpo for treatment of cancer-related anemia and makes a case to the nephrology community for the careful use of rEpo in CKD patients with cancer.


Asunto(s)
Anemia/tratamiento farmacológico , Eritropoyetina/uso terapéutico , Hematínicos/uso terapéutico , Neoplasias/complicaciones , Insuficiencia Renal Crónica/terapia , Anemia/etiología , Anemia/metabolismo , Anemia/mortalidad , Proliferación Celular/efectos de los fármacos , Eritropoyetina/efectos adversos , Hematínicos/efectos adversos , Humanos , Neoplasias/metabolismo , Neoplasias/mortalidad , Neoplasias/patología , Pronóstico , Receptores de Eritropoyetina/agonistas , Receptores de Eritropoyetina/metabolismo , Insuficiencia Renal Crónica/complicaciones , Insuficiencia Renal Crónica/diagnóstico , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/mortalidad , Factores de Riesgo , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA