Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.227
Filtrar
2.
Int J Mol Sci ; 22(17)2021 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-34502385

RESUMEN

Erythropoietin (Epo) is the critical hormone for erythropoiesis. In adults, Epo is mainly produced by a subset of interstitial fibroblasts in the kidney, with minor amounts being produced in the liver and the brain. In this study, we used the immortalized renal interstitial fibroblast cell line FAIK F3-5 to investigate the ability of the bioactive sphingolipid sphingosine 1-phosphate (S1P) to stimulate Epo production and to reveal the mechanism involved. Stimulation of cells with exogenous S1P under normoxic conditions (21% O2) led to a dose-dependent increase in Epo mRNA and protein levels and subsequent release of Epo into the medium. S1P also enhanced the stabilization of HIF-2α, a key transcription factor for Epo expression. S1P-stimulated Epo mRNA and protein expression was abolished by HIF-2α mRNA knockdown or by the HIF-2 inhibitor compound 2. Furthermore, the approved S1P receptor modulator FTY720, and its active form FTY720-phosphate, both exerted a similar effect on Epo expression as S1P. The effect of S1P on Epo was antagonized by the selective S1P1 and S1P3 antagonists NIBR-0213 and TY-52156, but not by the S1P2 antagonist JTE-013. Moreover, inhibitors of the classical MAPK/ERK, the p38-MAPK, and inhibitors of protein kinase (PK) C and D all blocked the effect of S1P on Epo expression. Finally, the S1P and FTY720 effects were recapitulated in the Epo-producing human neuroblastoma cell line Kelly, suggesting that S1P receptor-dependent Epo synthesis is of general relevance and not species-specific. In summary, these data suggest that, in renal interstitial fibroblasts, which are the primary source of plasma Epo, S1P1 and 3 receptor activation upregulates Epo under normoxic conditions. This may have a therapeutic impact on disease situations such as chronic kidney disease, where Epo production is impaired, causing anemia, but it may also have therapeutic value as Epo can mediate additional tissue-protective effects in various organs.


Asunto(s)
Eritropoyetina/metabolismo , Receptores de Esfingosina-1-Fosfato/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Línea Celular , Células Cultivadas , Eritropoyesis , Eritropoyetina/fisiología , Fibroblastos/metabolismo , Clorhidrato de Fingolimod/metabolismo , Humanos , Hipoxia/metabolismo , Hipoxia/fisiopatología , Riñón/metabolismo , Lisofosfolípidos/metabolismo , Ratones , Unión Proteica , Receptores de Lisoesfingolípidos/metabolismo , Insuficiencia Renal Crónica/metabolismo , Transducción de Señal , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato/fisiología
3.
Int J Mol Sci ; 22(13)2021 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-34281163

RESUMEN

Erythropoietin (EPO) acts on multiple tissues through its receptor EPOR, a member of a cytokine class I receptor superfamily with pleiotropic effects. The interaction of EPO and EPOR triggers the activation of several signaling pathways that induce erythropoiesis, including JAK2/STAT5, PI3K/AKT, and MAPK. The canonical EPOR/JAK2/STAT5 pathway is a known regulator of differentiation, proliferation, and cell survival of erythroid progenitors. In addition, its role in the protection of other cells, including cancer cells, is under intense investigation. The involvement of EPOR/JAK2/STAT5 in other processes such as mRNA splicing, cytoskeleton reorganization, and cell metabolism has been recently described. The transcriptomics, proteomics, and epigenetic studies reviewed in this article provide a detailed understanding of EPO signalization. Advances in this area of research may be useful for improving the efficacy of EPO therapy in hematologic disorders, as well as in cancer treatment.


Asunto(s)
Eritropoyetina/metabolismo , Factor de Transcripción STAT5/metabolismo , Factor de Transcripción STAT5/fisiología , Animales , Diferenciación Celular/efectos de los fármacos , Epigenómica/métodos , Eritropoyesis/efectos de los fármacos , Eritropoyetina/fisiología , Humanos , Janus Quinasa 2/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteómica/métodos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Eritropoyetina/metabolismo , Receptores de Eritropoyetina/fisiología , Factor de Transcripción STAT5/genética , Transducción de Señal/efectos de los fármacos , Transactivadores/metabolismo , Transcriptoma/genética
4.
Int J Mol Sci ; 22(14)2021 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-34299300

RESUMEN

Erythropoietin (EPO) is a glycoprotein cytokine known for its pleiotropic effects on various types of cells and tissues. EPO and its receptor EPOR trigger signaling cascades JAK2/STAT5, MAPK, and PI3K/AKT that are interconnected and irreplaceable for cell survival. In this article, we describe the role of the MAPK and PI3K/AKT signaling pathways during red blood cell formation as well as in non-hematopoietic tissues and tumor cells. Although the central framework of these pathways is similar for most of cell types, there are some stage-specific, tissue, and cell-lineage differences. We summarize the current state of research in this field, highlight the novel members of EPO-induced PI3K and MAPK signaling, and in this respect also the differences between erythroid and non-erythroid cells.


Asunto(s)
Eritropoyesis/fisiología , Eritropoyetina/fisiología , Animales , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Supervivencia Celular/fisiología , Humanos , Sistema de Señalización de MAP Quinasas , Modelos Biológicos , Neoplasias/fisiopatología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Eritropoyetina/fisiología , Transducción de Señal
5.
Transfus Apher Sci ; 60(4): 103160, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34217601

RESUMEN

BACKGROUND: COVID-19 virus has caused the world's deadliest pandemic. Early April 2020, the Delhi Government made it compulsory for people to wear face masks while going outdoors to curb disease spread. Prolonged use of surgical masks during the pandemic has been reported to cause many adverse effects. Intermittent hypoxia has been shown to activate erythropoietin (EPO leading to increased hemoglobin mass. AIM: To analyze whether face mask induced intermittent hypoxia has any effect on the hemoglobin levels of healthy blood donors. MATERIALS AND METHODS: We retrospectively analyzed donor data from 1st July 2019-31st December 2020 for hemoglobin distribution across hemoglobin ranges and donor deferral on basis of hemoglobin. Study population was divided into two cohorts Group 1- (1st July 2019-31 st March 2020): before implementation of mandatory face masks Group 2- (1st April 2020-31 st December 2020): after implementation of mandatory face masks RESULTS: Mean Hb of blood donors in Group 2 (15.01 ± 1.1 g/dl) was higher than Group1 (14.49 ± 1.15 g/dl), (p < 0.0001). 47.1 % group2 donors had Hb of 16.1-18 g/dl compared to group1 (38.4 %). 52.9 % group 2 donors had Hb between 12.5-15 g/dl compared to 61.6 % Group 1 (p < 0.05). Deferral due to anemia was lesser in group 2 compared to group 1 (p < 0.00001). Group 2 had significantly higher deferral due to high Hb (>18 gm/dl) was than Group 1 (p = 0.0039). CONCLUSION: This study including 19504 blood donors spanning over one and a half year shows that prolonged use of face mask by blood donors may lead to intermittent hypoxia and consequent increase in hemoglobin mass.


Asunto(s)
Donantes de Sangre , COVID-19/prevención & control , Eritropoyetina/fisiología , Hemoglobinas/análisis , Hipoxia/etiología , Máscaras/efectos adversos , Pandemias , SARS-CoV-2 , Adolescente , Adulto , Anciano , Estudios Transversales , Selección de Donante/normas , Femenino , Hemoglobinas/biosíntesis , Humanos , Hipoxia/sangre , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Adulto Joven
6.
BMC Nephrol ; 22(1): 203, 2021 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-34059008

RESUMEN

INTRODUCTION: The number of patients with end stage kidney disease (ESKD) are increasing world-side. While interstitial fibrosis (IF) is a common step for the progression to ESKD, therapeutic options for IF is still limited in clinical settings. We have reported that bone marrow-derived fibrotic cell, fibrocyte, is involved in the pathogenesis of kidney fibrosis. Also recent studies revealed that erythropoietin has protective effect on kidney diseases. However, it is unknown whether erythropoietin (EPO) inhibits fibrosis in progressive kidney injury. Therefore, we explored the impacts of EPO on kidney fibrosis with focusing on fibrocyte. METHOD: Fibrocyte was differentiated from peripheral mononuclear cells of healthy donor. Fibrocyte was stimulated with transforming growth factor beta (TGF)-ß with/without EPO treatment. Moreover, the therapeutic effect of EPO was evaluated in murine unilateral ureteral obstruction (UUO) model. RESULT: TGF-ß stimulation increased the expression of COL1 mRNA in fibrocyte. EPO signal reduced the expression of COL1 mRNA in dose dependent manner. EPO reduced mitochondrial oxidative stress and ameliorated mitochondrial membrane depolarization induced by TGF-ß stimulation. Moreover, EPO reduced the mRNA expression of mitochondria related molecules, TRAF6, in fibrocyte. In addition, the count of CD45+/αSMA + double-positive fibrocyte was decreased in the EPO-administered UUO kidneys. CONCLUSION: EPO signals function to prevent kidney fibrosis, particularly in fibrocyte. Regulating the renal accumulation of fibrocyte is a part of the anti-fibrotic functions of EPO.


Asunto(s)
Eritropoyetina/fisiología , Enfermedades Renales/metabolismo , Riñón/patología , Factor de Crecimiento Transformador beta/fisiología , Animales , Células de la Médula Ósea , Células Cultivadas , Colágeno/genética , Colágeno/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Eritropoyetina/uso terapéutico , Fibrosis/tratamiento farmacológico , Fibrosis/metabolismo , Humanos , Enfermedades Renales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/metabolismo
7.
Neuroendocrinology ; 111(11): 1029-1065, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33075777

RESUMEN

Stroke is a debilitating disease and has the ability to culminate in devastating clinical outcomes. Ischemic stroke followed by reperfusion entrains cerebral ischemia/reperfusion (I/R) injury, which is a complex pathological process and is associated with serious clinical manifestations. Therefore, the development of a robust and effective poststroke therapy is crucial. Granulocyte colony-stimulating factor (GCSF) and erythropoietin (EPO), originally discovered as hematopoietic growth factors, are versatile and have transcended beyond their traditional role of orchestrating the proliferation, differentiation, and survival of hematopoietic progenitors to one that fosters brain protection/neuroregeneration. The clinical indication regarding GCSF and EPO as an auspicious therapeutic strategy is conferred in a plethora of illnesses, including anemia and neutropenia. EPO and GCSF alleviate cerebral I/R injury through a multitude of mechanisms, involving antiapoptotic, anti-inflammatory, antioxidant, neurogenic, and angiogenic effects. Despite bolstering evidence from preclinical studies, the multiple brain protective modalities of GCSF and EPO failed to translate in clinical trials and thereby raises several questions. The present review comprehensively compiles and discusses key findings from in vitro, in vivo, and clinical data pertaining to the administration of EPO, GCSF, and other drugs, which alter levels of colony-stimulating factor (CSF) in the brain following cerebral I/R injury, and elaborates on the contributing factors, which led to the lost in translation of CSFs from bench to bedside. Any controversial findings are discussed to enable a clear overview of the role of EPO and GCSF as robust and effective candidates for poststroke therapy.


Asunto(s)
Factores Estimulantes de Colonias/fisiología , Eritropoyetina/fisiología , Accidente Cerebrovascular Isquémico , Daño por Reperfusión , Animales , Factores Estimulantes de Colonias/uso terapéutico , Eritropoyetina/uso terapéutico , Humanos , Accidente Cerebrovascular Isquémico/tratamiento farmacológico , Accidente Cerebrovascular Isquémico/inmunología , Accidente Cerebrovascular Isquémico/metabolismo , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/inmunología , Daño por Reperfusión/metabolismo
8.
Pediatr Res ; 90(3): 559-564, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33096541

RESUMEN

BACKGROUND: Apoptosis that occurs after hypoxia/reoxygenation (H/R) has an important role in the pathogenesis of necrotizing enterocolitis (NEC). Telomerase activity, showing the regeneration capacity, may also be important in the recovery process. Therefore, we aimed to investigate the effects of insulin-like growth factor-1 (IGF-1) and erythropoietin (EPO) on apoptosis and telomerase activity in an H/R model. METHODS: Young mice were divided into four groups each containing ten Balb/c mice. Group 1 (H/R) were exposed to H/R; group 2 and group 3 were pretreated with IGF-1 and EPO, respectively, for 7 days before H/R. Group 4 served as control. Intestinal injury was evaluated by histological scoring and assessment of apoptosis was performed by terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) test. Proapoptotic and antiapoptotic gene expressions and telomerase activity were analyzed by real-time PCR. RESULTS: IGF-1- and EPO-treated animals had decreased histological damage and apoptosis, confirmed by TUNEL test and caspase activity. Telomerase activity was increased in these animals in addition to increased expression of antiapoptotic genes. However, proapoptotic gene expressions were not statistically different. CONCLUSIONS: The protective effects of IGF-1 and EPO in H/R damage may be through increased expression of antiapoptotic genes and increased telomerase activity, especially for IGF-1. IMPACT: This is a comprehensive study measuring various variables, namely IGF-1, EPO, apoptosis, apoptotic and antiapoptotic genes, and telomerase activity in the NEC model. The intestinal protective effects of IGF-1 and EPO in H/R damage may occur through increased expression of antiapoptotic genes and increased telomerase activity. To the best of our knowledge, telomerase activity has not been investigated in the NEC model before. Regarding our results, novel strategies may be implemented for the early definitive diagnosis, robust preventive measures, and effective treatment modalities for NEC.


Asunto(s)
Apoptosis/fisiología , Enterocolitis Necrotizante/prevención & control , Eritropoyetina/fisiología , Factor I del Crecimiento Similar a la Insulina/fisiología , Telomerasa/metabolismo , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos BALB C
9.
J Integr Neurosci ; 19(3): 561-570, 2020 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-33070534

RESUMEN

Erythropoietin has been researched for its neuroprotective effects in ischemic stroke for over 30 years. Although erythropoietin can cause side effects that need to be controlled, it has been suggested to be effective in enhancing the prognosis of patients who are out of the therapeutic time window and have not received recombinant tissue plasminogen activator therapy. Studies on the mechanism of the function of erythropoietin have shown that it has various protective effects in ischemic brain injury after stroke, including promoting neurogenesis. In this review, we discuss the effects of erythropoietin on neurogenesis after ischemic brain injury and provide references for effective treatments for ischemic stroke, which is one of the leading causes of death worldwide.


Asunto(s)
Isquemia Encefálica/fisiopatología , Eritropoyetina/administración & dosificación , Eritropoyetina/fisiología , Accidente Cerebrovascular Isquémico/fisiopatología , Neurogénesis , Fármacos Neuroprotectores/administración & dosificación , Animales , Isquemia Encefálica/tratamiento farmacológico , Humanos , Accidente Cerebrovascular Isquémico/tratamiento farmacológico
10.
Expert Rev Hematol ; 13(11): 1175-1188, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33028115

RESUMEN

INTRODUCTION: Anemia has and will continue to be a central theme in medicine particularly as clinicians are treating a burgeoning population of complex multi-organ system processes. As a result of multiple randomized controlled trials (RCTs), meta-analyses, and societal recommendations overly restrictive paradigms and under-administration of erythropoiesis stimulating agents (ESAs) have likely been followed by clinicians among all specialties. AREAS COVERED: A review of anemia in the context of chronic kidney disease, hematologic malignancies, and cancer is presented with focus on the establishment of ESAs as integral in the treatment of anemia. Multiple RCTs and meta-analyses studying the use of ESAs are presented with focus upon their application to clinical practice. A 'compendium' is proffered describing the evolution, establishment, and implications of ESA administration initially among those with CKD with rapid subsequent application to the Hematology-Oncology population of patients. Literature search methodologies have included MEDLINE (1985-2020), PubMed (1996-2020), Cochrane Central Trials (1985-2020), EMBASE (2000-2020), and ClinicalTrials.gov (2000-2020). EXPERT OPINION: Upon evaluation of risks and benefits of ESAs focused opinion and commentary is made supporting more liberal use of these agents and strongly suggesting that the current underlying treatment 'pendulum' has perhaps shifted too far to the 'under-treatment' side in many cases.


Asunto(s)
Anemia/terapia , Neoplasias/complicaciones , Insuficiencia Renal Crónica/complicaciones , Anemia/tratamiento farmacológico , Anemia/etiología , Anemia/fisiopatología , Transfusión Sanguínea , COVID-19/epidemiología , Terapia Combinada , Epoetina alfa/efectos adversos , Epoetina alfa/uso terapéutico , Eritropoyetina/fisiología , Eritropoyetina/uso terapéutico , Testimonio de Experto , Predicción , Adhesión a Directriz , Hematínicos/administración & dosificación , Hematínicos/uso terapéutico , Neoplasias Hematológicas/complicaciones , Hematopoyesis , Humanos , Hierro/uso terapéutico , Medicina , Metaanálisis como Asunto , Estudios Multicéntricos como Asunto , Isquemia Miocárdica/complicaciones , Estudios Observacionales como Asunto , Pandemias , Guías de Práctica Clínica como Asunto , Pautas de la Práctica en Medicina , Ensayos Clínicos Controlados Aleatorios como Asunto , Receptores de Eritropoyetina/efectos de los fármacos , Diálisis Renal , Insuficiencia Renal Crónica/terapia , SARS-CoV-2 , Tromboembolia Venosa/inducido químicamente
11.
F1000Res ; 92020.
Artículo en Inglés | MEDLINE | ID: mdl-32983414

RESUMEN

More than 50 years of efforts to identify the major cytokine responsible for red blood cell (RBC) production (erythropoiesis) led to the identification of erythropoietin (EPO) in 1977 and its receptor (EPOR) in 1989, followed by three decades of rich scientific discovery. We now know that an elaborate oxygen-sensing mechanism regulates the production of EPO, which in turn promotes the maturation and survival of erythroid progenitors. Engagement of the EPOR by EPO activates three interconnected signaling pathways that drive RBC production via diverse downstream effectors and simultaneously trigger negative feedback loops to suppress signaling activity. Together, the finely tuned mechanisms that drive endogenous EPO production and facilitate its downstream activities have evolved to maintain RBC levels in a narrow physiological range and to respond rapidly to erythropoietic stresses such as hypoxia or blood loss. Examination of these pathways has elucidated the genetics of numerous inherited and acquired disorders associated with deficient or excessive RBC production and generated valuable drugs to treat anemia, including recombinant human EPO and more recently the prolyl hydroxylase inhibitors, which act partly by stimulating endogenous EPO synthesis. Ongoing structure-function studies of the EPOR and its essential partner, tyrosine kinase JAK2, suggest that it may be possible to generate new "designer" drugs that control selected subsets of cytokine receptor activities for therapeutic manipulation of hematopoiesis and treatment of blood cancers.


Asunto(s)
Eritrocitos/citología , Eritropoyesis , Eritropoyetina/fisiología , Receptores de Eritropoyetina/fisiología , Humanos , Janus Quinasa 2/fisiología , Transducción de Señal
12.
Rev Mal Respir ; 37(3): 193-196, 2020 Mar.
Artículo en Francés | MEDLINE | ID: mdl-32146057

RESUMEN

In addition to its role in erythropoiesis, erythropoietin (Epo) plays a role in tissue protection, which includes cardioprotective, nephroprotective and neuroprotective effects. The presence of Epo and its receptor (Epo-R) in pulmonary tissue also suggests a cytoprotective effect of Epo in the lung. Our project aims to document this role in a murine model under-expressing Epo. The obtained results will lead to a better understanding of the cytoprotective effects of Epo and will also give an appreciation of its beneficial effects in cases of lung injury.


Asunto(s)
Lesión Pulmonar Aguda/patología , Citoprotección , Eritropoyetina/farmacología , Eritropoyetina/fisiología , Riñón , Animales , Citoprotección/efectos de los fármacos , Citoprotección/genética , Modelos Animales de Enfermedad , Eritropoyetina/genética , Hematopoyesis/efectos de los fármacos , Hematopoyesis/genética , Humanos , Riñón/efectos de los fármacos , Riñón/patología , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/fisiología , Ratones
13.
Biochim Biophys Acta Mol Cell Res ; 1867(1): 118569, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31676353

RESUMEN

Water influx through aquaporin-1 (AQP-1) has been linked to the ability of different cell types to migrate, and therefore plays an important part in processes like metastasis and angiogenesis. Since the erythroid growth factor erythropoietin (Epo) is now recognized as an angiogenesis promoter, we investigated the participation of AQP-1 as a downstream effector of this cytokine in the migration of endothelial cells. Inhibition of AQP-1 with either mercury ions (Hg2+) or a specific siRNA led to an impaired migration of EA.hy926 endothelial cells exposed to Epo (wound-healing assays). Epo also induced the expression of AQP-1 at mRNA and protein levels, an effect which was dependent on the influx of extracellular calcium through L-type calcium channels as well as TRPC3 channels. The relationship between Epo and AQP-1 was further confirmed at shorter exposure times, as the cytokine was unable to trigger calcium influxes in cells where AQP-1 had previously been knocked down. Moreover, Epo promoted changes in the subcellular localization of AQP-1 as well as rearrangements in the actin cytoskeleton, which are consistent with a migratory phenotype. Worthy of note, carbamylated erythropoietin (cEpo), the non-erythropoietic and non-promigratory derivative of Epo, was incapable of AQP-1 modulation. The therapeutical implications of aquaporin targeting in angiogenesis-related diseases highlight the importance of the present results in the context of the relationship between AQP-1 and Epo.


Asunto(s)
Acuaporina 1/fisiología , Movimiento Celular/efectos de los fármacos , Eritropoyetina/farmacología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/fisiología , Células A549 , Acuaporina 1/antagonistas & inhibidores , Movimiento Celular/genética , Células Cultivadas , Eritropoyetina/fisiología , Humanos , ARN Interferente Pequeño/farmacología , Cicatrización de Heridas/efectos de los fármacos , Cicatrización de Heridas/genética
14.
World Neurosurg ; 131: 346-355, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31658577

RESUMEN

BACKGROUND: Erythropoietin (EPO) is a cytokine primarily involved in the regulation of erythropoiesis. In response to hypoxia-ischemia, hypoxia-inducible factor 1 induces EPO production, which, in turn, inhibits apoptosis of erythroid progenitor cells. By the same mechanism and acting through other signaling pathways, EPO exerts neuroprotective effects. Increased resistance to hypoxia and decreased apoptosis are thought to be important mechanisms for tumor progression, including malignant glioma. Because recent studies have demonstrated that EPO and its receptor (EPOR) are expressed in several tumors and can promote tumor growth, in the present study, we investigated EPO and EPOR expression in human glioma and the effect of EPO administration in a rat model of glioma implantation. METHODS: Using Western blotting and immunohistochemical analysis, we examined the expression of EPO, EPOR, platelet endothelial cell adhesion molecule, and Ki-67 in human glioma specimens and experimentally induced glioma in rats. In the experimental setting, a daily dose of recombinant human EPO (rHuEPO) or saline solution were administered for 21 days in Fischer rats subjected to 9L cell line implantation. RESULTS: In both human and animal specimens, we found an increase in EPOR expression as long as the lesion presented with an increasing malignant pattern. A significant direct correlation was found between the expression of EPOR and Ki-67 and EPOR and platelet endothelial cell adhesion molecule in low- and high-grade gliomas. The rats treated with rHuEPO presented with significantly larger tumor spread compared with the saline-treated rats. CONCLUSIONS: The results of our study have shown that the EPO/EPOR complex might play a significant role in the aggressive behavior of high-grade gliomas. The larger tumor spread in rHuEPO-treated rats suggests a feasible role for EPO in the aggressiveness and progression of malignant glioma.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Eritropoyetina/metabolismo , Glioma/metabolismo , Receptores de Eritropoyetina/metabolismo , Adulto , Anciano , Animales , Western Blotting , Neoplasias Encefálicas/etiología , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Eritropoyetina/farmacología , Eritropoyetina/fisiología , Femenino , Glioma/etiología , Glioma/patología , Humanos , Inmunohistoquímica , Antígeno Ki-67/metabolismo , Masculino , Persona de Mediana Edad , Trasplante de Neoplasias , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Ratas Endogámicas F344 , Receptores de Eritropoyetina/fisiología , Proteínas Recombinantes/farmacología , Carga Tumoral/efectos de los fármacos
15.
G Ital Nefrol ; 36(5)2019 Sep 24.
Artículo en Italiano | MEDLINE | ID: mdl-31580542

RESUMEN

Over the past two decades it has emerged that, in addition to erythropoietic activity, erythropoietin (EPO) has numerous other functions, including neuro-protective, anti-apoptotic, antioxidant, angiogenetic and immunomodulatory ones. EPO interacts with two different forms of its receptor (EPOR): a homodimer receptor, responsible for the erythropoietic effects, and a heterodimer receptor, responsible for the non-erythropoietic effects. The effects on the heterodimer receptor are responsible for EPO-induced prolongation of organ transplant survival in mice and humans. The development of new molecules that selectively target the heterodimer EPOR is allowing to test the effect of long-term treatments, without the possible complications related to the increased hematocrit.


Asunto(s)
Eritropoyesis/fisiología , Eritropoyetina/fisiología , Supervivencia de Injerto/fisiología , Receptores de Eritropoyetina/fisiología , Inmunidad Adaptativa , Anemia/tratamiento farmacológico , Anemia/etiología , Animales , Hipoxia de la Célula/fisiología , Eritropoyetina/genética , Eritropoyetina/farmacología , Supervivencia de Injerto/efectos de los fármacos , Corazón/efectos de los fármacos , Humanos , Inmunidad Celular , Inmunidad Innata , Riñón/efectos de los fármacos , Riñón/metabolismo , Ratones , Sistema Nervioso/metabolismo , Trasplante de Órganos , Ratas , Receptores del Factor Estimulante de Colonias/fisiología , Proteínas Recombinantes/uso terapéutico , Insuficiencia Renal Crónica/complicaciones , Retina/metabolismo
16.
Curr Opin Nephrol Hypertens ; 28(6): 600-606, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31567284

RESUMEN

PURPOSE OF REVIEW: Prolyl-hydroxylase inhibitors are a novel class of orally administered drugs that are under development for the treatment of anemia in patients with chronic kidney disease. This review discusses the biology of these drugs and their target - hypoxia-inducible factor and potential advantages and disadvantages of these therapies. Finally, we will discuss current trials in patients with both chronic kidney disease and end-stage renal disease. RECENT FINDINGS: Recent smaller studies have found that prolyl-hydroxylase are as effective as erythropoietin in treating anemia of chronic kidney disease. We do not yet know if they have the same cardiovascular and cancer-related risk profile and these questions will be answered by large phase III trials that are ongoing. SUMMARY: Although prolyl hydroxylase inhibitors have much potential, questions remain regarding their efficacy and safety. Should these concerns prove to be unfounded, the treatment of anemia in chronic kidney disease will likely be transformed over the next decade.


Asunto(s)
Anemia/tratamiento farmacológico , Inhibidores de Prolil-Hidroxilasa/uso terapéutico , Insuficiencia Renal Crónica/complicaciones , Administración Oral , Eritropoyesis , Eritropoyetina/fisiología , Humanos , Prolina Dioxigenasas del Factor Inducible por Hipoxia , Péptidos y Proteínas de Señalización Intracelular/fisiología , Fallo Renal Crónico/tratamiento farmacológico , Proteínas Mitocondriales/fisiología
17.
Vnitr Lek ; 65(7-8): 515-519, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31487995

RESUMEN

The cytokine erythropoietin is the main hemopoietic factor synthesized mainly by the kidney. However, erythro-poietin and its receptors are expressed in several tissues and exert pleiotropic activities also in nonhemopoietic tissues. Erythropoietin has an antiapoptotic activity and plays a potential neuroprotective, nefroprotective and cardioprotective role against ischemia and other type of injury. Erythropoietin is also involved in angiogenesis, neurogenesis, and the immune response. It can prevent metabolic alterations, vascular and neuronal degeneration, and inflammatory cell activation. Erythropoietin reduces hyperglycaemia and retards proliferative retinopathy in diabetic patients. Consequently, erythropoietin may be of therapeutic value for a variety of disorders. This short review provides an insight into the nonhemopoietic role of erythropoietin and its mechanisms of action. For elimination of polycythaemia after erythropoietin administration analogues without haematopoietic activity were prepared and tested in animals and in some cases also evaluated in clinical trials.


Asunto(s)
Eritropoyetina , Animales , Apoptosis , Eritropoyetina/farmacología , Eritropoyetina/fisiología , Humanos , Isquemia , Riñón , Neovascularización Patológica
18.
Curr Opin Nephrol Hypertens ; 28(4): 304-310, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31145704

RESUMEN

PURPOSE OF REVIEW: Recent research has revealed that regulation of the bone-secreted hormone fibroblast growth factor 23 (FGF23) is not limited to classical mineral factors. Specifically, bidirectional relationships have been described between FGF23 production and anemia, iron status, and inflammation. Here, we will review the latest published articles on the crosstalk between FGF23 and the aforementioned nonclassical factors. RECENT FINDINGS: It has been recently reported that erythropoietin, iron deficiency, and inflammation increase FGF23 production and metabolism. Moreover, FGF23 promotes anemia and regulates inflammatory responses. These findings are particularly important in the setting of chronic kidney disease which is characterized by elevated FGF23 levels and several associated comorbidities. SUMMARY: Regulation of FGF23 is complex and involves many bone and renal factors. More recently, erythropoietin, iron deficiency, and inflammation have been also shown to affect FGF23 transcription and cleavage. Importantly, FGF23 has emerged as a regulator of erythropoiesis, iron metabolism, and inflammation. These findings provide novel and important insights into the pathophysiologic mechanisms of chronic kidney disease and may present new opportunities for therapeutic clinical interventions.


Asunto(s)
Eritropoyetina/fisiología , Factores de Crecimiento de Fibroblastos/fisiología , Inflamación/complicaciones , Hierro/metabolismo , Insuficiencia Renal Crónica/etiología , Eritropoyesis , Factor-23 de Crecimiento de Fibroblastos , Humanos
19.
FEBS J ; 286(14): 2664-2669, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31095893

RESUMEN

Antidoping work is heavily based on scientific analyses of biological material, such as urine and blood. Because of the high stakes both for sports and for the athletes involved it is important that analyses are performed and interpreted in agreement with established scientific standards and professional norms. This is not always the case, as we document here. It is our experience that the antidoping movement does not appear willing to consider that errors can occur and should be corrected. The consequences of the lack of transparency and responsibility are carried by unlucky athletes. Scientific, ethical and legal considerations urge the antidoping movement to reform some of their rules and regulations and to include the possibility that the World Anti-Doping Agency position could, in some cases, be incorrect.


Asunto(s)
Doping en los Deportes/prevención & control , Doping en los Deportes/legislación & jurisprudencia , Eritropoyetina/fisiología , Humanos , Límite de Detección
20.
Acta Trop ; 195: 35-43, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31004564

RESUMEN

Toxoplasma gondii has evolved many successful strategies for immune evasion. However, the parasite-derived effectors involved in modulating NF-κB signalling pathway are largely unknown. T. gondii Cathepsin C1 (CPC1) is widely conserved among T. gondii strains and is important for T. gondii intracellular growth and proliferation. Our study showed that CPC1 protein could abrogate NF-κB activation after screening dense granule proteins. CPC1 suppressed NF-κB activation at or downstream of p65 and decreased the production of IL-1, IL-8, IL-6, IL-12, and TNF-α. Western blot analysis revealed that CPC1 inhibited phospho-p65 and CPC1 proteins primarily settled in cytoplasm. RNA sequencing analysis revealed that overexpression of CPC1 significantly upregulated erythropoietin (EPO), which can be induced by the hypoxia-inducible factor -1α (HIF-1α) during hypoxia. Furthermore, dual-luciferase reporter assays confirmed that CPC1 upregulated HIF-1α. Finally, both the knockdown of EPO and restriction of HIF-1α partially eliminated the suppression impact of CPC1 on the NF-κB signalling pathway. Our study identified a previously unrecognized role of CPC1 in the negative regulation of NF-κB activation through positive regulation of the HIF-1α/EPO axis. For the first time, CPC1 was shown to play an important role in immune evasion during T. gondii infection.


Asunto(s)
Catepsina C/fisiología , Eritropoyetina/fisiología , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , FN-kappa B/fisiología , Toxoplasma/inmunología , Células HEK293 , Humanos , Evasión Inmune , Transducción de Señal/fisiología , Toxoplasma/enzimología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA