Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 5.159
Filtrar
1.
PeerJ ; 12: e17123, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38560469

RESUMO

Background: The purpose of the present study was to investigate the effect of erythropoietin (EPO) on lung ischemia-reperfusion injury (LIRI). Methods: Sprague Dawley rats and BEAS-2B cells were employed to construct an ischemia-reperfusion (I/R)-induced model in vivo and in vitro, respectively. Afterward, I/R rats and tert-butyl hydroperoxide (TBHP)-induced cells were treated with different concentrations of EPO. Furthermore, 40 patients with LIRI and healthy controls were enrolled in the study. Results: It was observed that lung tissue damage, cell apoptosis and the expression of BAX and caspase-3 were higher in the LIRI model in vivo and in vitro than in the control group, nevertheless, the Bcl-2, FGF23 and FGFR4 expression level was lower than in the control group. EPO administration significantly reduced lung tissue damage and cell apoptosis while also up-regulating the expression of FGF23 and FGFR4. Rescue experiments indicated that EPO exerted a protective role associated with the FGF23/FGFR4/p-ERK1/2 signal pathway. Notably, the expression of serum EPO, FGF23, FGFR4 and Bcl-2 was decreased in patients with LIRI, while the expression of caspase-3 and BAX was higher. Conclusion: EPO could effectively improve LIRI, which might be related to the activation of the FGF23/FGFR4/p-ERK1/2 signaling pathway.


Assuntos
Eritropoetina , Traumatismo por Reperfusão , Animais , Humanos , Ratos , Proteína X Associada a bcl-2/metabolismo , Caspase 3/genética , Epoetina alfa/metabolismo , Eritropoetina/farmacologia , Isquemia , Pulmão/metabolismo , Sistema de Sinalização das MAP Quinases , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos Sprague-Dawley , Receptor Tipo 4 de Fator de Crescimento de Fibroblastos/genética , Traumatismo por Reperfusão/tratamento farmacológico , Transdução de Sinais
2.
BMC Oral Health ; 24(1): 391, 2024 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-38539151

RESUMO

BACKGROUND: Tongue is complex muscular organ that may be affected by recurrent or chronic ulcerations and malignances that require effective treatment to enhance healing and tissue regeneration. So, this study aimed to evaluate the efficiency of erythropoietin (EPO) hydrogel as an anti-inflammatory and an inducer of neovascularization during healing of induced rats' tongue defects. METHODS: Thirty six rats were divided into three groups; Group I (negative control): tongues were left without ulceration and received no treatment, Group II (positive control): tongue defects were prepared on the tongues' dorsal surfaces, measuring (5 mm × 2 mm) using a tissue punch rotary drill for standardization, and left untreated, Group III (EPO group): tongue defects were prepared as in group II, then injected circumferentially around wound margins with a single high dose of EPO hydrogel of 5000 U/kg on the day of defect preparation. Animals were euthanized on seventh and fourteenth days after treatment, tongue specimens were collected, and paraffin blocks were prepared and processed for histological assessment by hematoxylin and eosin stain and immunohistochemical evaluation of anti-iNOS and anti-VEGF followed by histomorphometrical analysis and the relevant statistical tests. RESULTS: At both time points, the EPO treated group showed significantly enhanced tissue regeneration marked by the histologically better regenerated tissue with well developed, thick walled and well-organized blood vessels and significant reduction in defect depth compared to positive control group. EPO group also showed significant decrease in iNOS and significant increase in VEGF antibodies indicating its anti-inflammatory and neovascularization effects respectively. CONCLUSION: EPO treatment can significantly accelerate regeneration and filling of tongue defects by reducing tissue inflammation and enhancing neovascularization. Therefore, EPO could be a potential therapeutic strategy for accelerating healing of tongue ulcers. However, further investigations are required to optimize the dose and unravel any potential side effects before its clinical application.


Assuntos
Eritropoetina , Hidrogéis , Ratos , Animais , Hidrogéis/farmacologia , Eritropoetina/farmacologia , Eritropoetina/uso terapêutico , Cicatrização , Anti-Inflamatórios/farmacologia , Língua
3.
J Psychopharmacol ; 38(4): 362-374, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38519416

RESUMO

BACKGROUND: Persistent cognitive impairment is frequent across bipolar disorder (BD) and major depressive disorder (MDD), highlighting an urgent need for pro-cognitive treatments. AIM: This study investigated effects of erythropoietin (EPO) on cognitive impairment and dorsal prefrontal cortex (dPFC) activity in affective disorders. METHODS: In this randomized, double-blinded, placebo-controlled trial, cognitively impaired patients with remitted BD or MDD received 1 weekly recombinant human EPO (40,000 IU/mL) or saline infusion for a 12-week period. Assessments were conducted at baseline, after 2 weeks of treatment (week 3), immediately after treatment (week 13) and at 6-months follow-up. Participants underwent functional MRI during performance on a n-back working memory (WM) task at baseline and week 3, and for a subgroup 6 weeks post-treatment (week 18). The primary outcome was a cognitive composite score at week 13, whereas secondary outcomes comprised sustained attention and functioning. WM-related dPFC activity was a tertiary outcome. RESULTS: Data were analysed for 101 of the 103 included patients (EPO, n = 58; saline, n = 43). There were no effects of EPO over saline on any cognitive or functional outcomes or on WM-related dPFC activity. CONCLUSIONS: The absence of treatment-related changes in cognition and neural activity was unexpected and contrasts with multiple previous preclinical and clinical studies. It is possible that the lack of effects resulted from a recent change in the manufacturing process for EPO. Nevertheless, the findings support the validity of dPFC target engagement as a biomarker model for pro-cognitive effects, according to which treatments that do not improve cognition should not modulate dPFC activity. TRIAL REGISTRATIONS: EudraCT no.: 2016-004023-24; ClinicalTrials.gov identifier: NCT03315897.


Assuntos
Disfunção Cognitiva , Transtorno Depressivo Maior , Eritropoetina , Humanos , Transtorno Depressivo Maior/tratamento farmacológico , Transtorno Depressivo Maior/psicologia , Transtornos do Humor/tratamento farmacológico , Eritropoetina/farmacologia , Eritropoetina/uso terapêutico , Disfunção Cognitiva/tratamento farmacológico , Cognição , Córtex Pré-Frontal , Resultado do Tratamento , Método Duplo-Cego
4.
J Neuropathol Exp Neurol ; 83(3): 161-167, 2024 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-38263262

RESUMO

Recombinant human erythropoietin (rh-EPO) has been shown to stimulate neurogenesis and angiogenesis, both of which play crucial roles in the repair of brain injuries. Previously, we observed that rh-EPO treatment effectively reduced brain damage and enhanced angiogenesis in a neonatal rat model of periventricular white matter damage (PWMD). The objective of this research is to investigate the specific mechanism through which rh-EPO regulates angiogenesis following PWMD in premature neonates. We conducted experiments utilizing a neonatal PWMD model. Following rh-EPO treatment, the levels of erythropoietin receptor (EPOR) were found to be increased in the damaged brain of rats. Although the total amount of extracellular signal-regulated kinase (ERK), a downstream protein in the EPO signaling pathway, remained unchanged, there was clear upregulation of phosphorylated ERK1 (p-ERK1) levels. The increase in levels of p-ERK1 was inhibited by an ERK kinase inhibitor, while the total amount of ERK remained unchanged. Conversely, the levels of EPOR were not affected by the inhibitor. Notably, the introduction of rh-EPO led to a significant increase in the frequency of angiogenesis-related cells and the expression levels of angiogenic factors. However, these effects were nullified when the ERK pathway was blocked. These findings indicate that rh-EPO enhances angiogenic responses through the EPOR-ERK1 pathway in a neonatal PWMD model.


Assuntos
Eritropoetina , Substância Branca , Ratos , Animais , Humanos , Animais Recém-Nascidos , Substância Branca/metabolismo , Ratos Sprague-Dawley , Eritropoetina/farmacologia , Eritropoetina/metabolismo , Transdução de Sinais/fisiologia , Receptores da Eritropoetina/metabolismo
5.
Int J Mol Sci ; 25(2)2024 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-38279224

RESUMO

Many large-scale studies show that exogenous erythropoietin, erythropoiesis-stimulating agents, lack any renoprotective effects. We investigated the effects of endogenous erythropoietin on renal function in kidney ischemic reperfusion injury (IRI) using the prolyl hydroxylase domain (PHD) inhibitor, Roxadustat (ROX). Four h of hypoxia (7% O2) and 4 h treatment by ROX prior to IRI did not improve renal function. In contrast, 24-72 h pretreatment by ROX significantly improved the decline of renal function caused by IRI. Hypoxia and 4 h ROX increased interstitial cells-derived Epo production by 75- and 6-fold, respectively, before IRI, and worked similarly to exogenous Epo. ROX treatment for 24-72 h increased Epo production during IRI by 9-fold. Immunohistochemistry revealed that 24 h ROX treatment induced Epo production in proximal and distal tubules and worked similarly to endogenous Epo. Our data show that tubular endogenous Epo production induced by 24-72 h ROX treatment results in renoprotection but peritubular exogenous Epo production by interstitial cells induced by hypoxia and 4 h ROX treatment did not. Stimulation of tubular, but not peritubular, Epo production may link to renoprotection.


Assuntos
Eritropoetina , Inibidores de Prolil-Hidrolase , Traumatismo por Reperfusão , Humanos , Eritropoetina/farmacologia , Rim , Epoetina alfa/farmacologia , Inibidores de Prolil-Hidrolase/farmacologia , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/prevenção & controle , Hipóxia
6.
Synapse ; 78(1): e22282, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37794768

RESUMO

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


Assuntos
Eritropoetina , Consolidação da Memória , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Eritropoetina/farmacologia , Eritropoetina/metabolismo , Receptores da Eritropoetina/metabolismo , Encéfalo/metabolismo , Hipocampo/metabolismo , Memória de Longo Prazo
7.
Hum Cell ; 37(1): 214-228, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37968533

RESUMO

Pulmonary arterial hypertension (PAH) is a chronic disease thatultimately progresses to right-sided heart failure and death. Erythropoietin (EPO) has been shown to have therapeutic potential in cardiovascular diseases, including PAH. In this study, we aimed to investigate the improvement effect of EPO pretreated bone marrow mesenchymal stem cells (BMSCs) on PAH. BMSCs were obtained from the bone marrow of male SD rats. Female rats were randomly divided into six groups, including control group, monocrotaline (MCT)-induced group, and four groups with different doses of EPO pretreated BMSCs. Lung tissue was taken for testing at 2 weeks of treatment. Our results showed EPO promoted homing and endothelial cell differentiation of BMSCs in the lung tissues of PAH rats. EPO and BMSCs treatment attenuated pulmonary arterial pressure, polycythemia, and pulmonary artery structural remodeling. Furthermore, BMSCs inhibited pulmonary vascular endothelial-to-mesenchymal transition (EndoMT) in PAH rats, which was further suppressed by EPO in a concentration-dependent manner. Meanwhile, EPO and BMSC treatment elevated pulmonary angiogenesis in PAH rats. BMSCs inhibited TNF-α, IL-1ß, IL-6, and MCP-1 in lung tissues of PAH rats, which was further decreased by EPO in a concentration-dependent manner. Thus, EPO improved pulmonary hypertension (PH) by promoting the homing and differentiation of BMSCs in lung tissue.


Assuntos
Eritropoetina , Hipertensão Pulmonar , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Ratos , Feminino , Masculino , Animais , Hipertensão Pulmonar/terapia , Hipertensão Pulmonar/induzido quimicamente , Ratos Sprague-Dawley , Transplante de Células-Tronco Mesenquimais/métodos , Pulmão , Eritropoetina/farmacologia , Diferenciação Celular , Remodelação Vascular , Células da Medula Óssea
8.
Inflammopharmacology ; 32(1): 523-536, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37578618

RESUMO

BACKGROUND: Neuroinflammation plays an important role in spinal cord injury (SCI), and an increasing number of studies have focused on the role of astrocytes in neuroinflammation. Pyroptosis is an inflammation-related form of programmed cell death, and neuroinflammation induced by astrocytes in the form of pyroptosis has been widely reported in many central nervous system diseases. Recent studies have found that erythropoietin has significant anti-inflammatory and neuroprotective effects in SCI; however, it has not been reported whether erythropoietin can reduce neuroinflammation by inhibiting neural cell pyroptosis in SCI. METHODS: A GEO dataset (GSE153720) was used to analyse the expression of pyroptosis-related genes in sham astrocytes and astrocytes 7 days, 1 month and 3 months after SCI. TargetScan and miRDB databases were used to predict the miRNA that could bind to the 3'UTR of rat Gsdmd. Primary rat spinal astrocytes were used for in vitro experiments, and the modified version of Allen's method was used to establish the rat SCI model. Western blotting, quantitative real-time polymerase chain reaction, flow cytometry, immunofluorescence, lactate dehydrogenase release assay and propidium iodide staining were used to detect the pyroptosis phenotype. A dual luciferase reporter gene assay was used to verify that miR-325-3p can bind to the 3'UTR of Gsdmd. RESULTS: We found that pyroptosis-related genes mediated by the canonical NLRP3 inflammasome were highly expressed in astrocytes in an SCI animal model by bioinformatic analysis. We also observed that erythropoietin could reduce astrocyte pyroptosis in vivo and in vitro. In addition, we predicted miRNAs that regulate Gsdmd, the pyroptosis executor, and verified that erythropoietin inhibits astrocyte pyroptosis in SCI through the miR-325-3p/Gsdmd axis. CONCLUSIONS: We demonstrated that erythropoietin can inhibit astrocyte pyroptosis through the miR-325-3p/Gsdmd axis. This study is expected to provide a new mechanism for erythropoietin in the treatment of SCI and a more reliable theoretical basis for clinical research.


Assuntos
Eritropoetina , Traumatismos da Medula Espinal , Animais , Ratos , Astrócitos , Piroptose , Regiões 3' não Traduzidas , Doenças Neuroinflamatórias , Eritropoetina/farmacologia , Traumatismos da Medula Espinal/tratamento farmacológico
9.
Eur Neuropsychopharmacol ; 79: 38-48, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38128460

RESUMO

Electroconvulsive therapy (ECT) is one of the most effective and rapid-acting treatment for severe depression but is associated with cognitive side-effects. Identification of add-on treatments that counteract these side-effects would be very helpful. This randomized, double-blinded, placebo-controlled, parallel-group study investigated the effects of four add-on erythropoietin (EPO; 40,000 IU/ml) or saline (placebo) infusions over 2.5 weeks of ECT (eight ECT sessions) in severely depressed patients with unipolar or bipolar depression. Neuropsychological assessments were conducted pre-ECT, three days after the eighth ECT (week 4), and at a 3-month follow-up. Further, functional magnetic resonance imaging (fMRI) was conducted after the eighth ECT. The primary outcome was change from pre- to post-ECT in a 'speed of complex cognitive processing' composite. Secondary outcomes were verbal and autobiographical memory. Of sixty randomized patients, one dropped out before baseline. Data were thus analysed for 59 patients (EPO, n = 33; saline, n = 26), of whom 28 had fMRI data. No ECT-related decline occurred in the primary global cognition measure (ps≥0.1), and no effect of EPO versus saline was observed on this outcome (ps≥0.3). However post-ECT, EPO-treated patients exhibited faster autobiographical memory recall than saline-treated patients (p = 0.02), which was accompanied by lower memory-related parietal cortex activity. The absence of global cognition changes with ECT and EPO, coupled with the specific impact of EPO on autobiographical memory recall speed and memory-related parietal cortex activity, suggests that assessing autobiographical memory may provide increased sensitivity in evaluating and potentially preventing cognitive side-effects of ECT. TRIAL REGISTRATIONS: ClinicalTrials.gov: NCT03339596, EudraCT no.: 2016-002326-36.


Assuntos
Eletroconvulsoterapia , Eritropoetina , Humanos , Eletroconvulsoterapia/efeitos adversos , Eletroconvulsoterapia/métodos , Depressão , Resultado do Tratamento , Eritropoetina/uso terapêutico , Eritropoetina/farmacologia , Epoetina alfa , Cognição , Método Duplo-Cego
10.
Biochem Pharmacol ; 220: 116008, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38154543

RESUMO

Recombinant human erythropoietin (rHuEPO) is a prevalent treatment for anemia in patients with chronic kidney disease. However, up to 10% of these patients exhibit EPO resistance or hyporesponsiveness, which may be caused by the depletion of erythroid progenitor cells. Thrombopoietin (TPO) has the potential to promote the growth of early progenitor cells and correct the depletion. In this study, we investigate the efficacy and the underlying mechanism of the combination therapy of TPO and EPO to EPO resistance. First, the in vivo studies suggested that intensive EPO treatment induced progenitor cell depletion in the bone marrow, where the depletion was corrected by TPO. Then, colony assays showed that EPO and TPO synergistically enhanced the burst-forming unit-erythroid (BFU-E) production but antagonistically boosted the colony-forming units of megakaryocytes (CFU-MK) production. Also, we found TPO promoted hematopoietic stem and progenitor cells (HSPCs) production, while EPO drove HSPCs toward the erythroid lineage. Additionally, EPO induced more megakaryocytic-erythroid progenitors (MEPs) toward the erythroid output. Model-based simulations indicate the efficacy of this combination therapy for treating EPO-resistant anemia in rats. In conclusion, our study demonstrated the efficacy of combination therapy in addressing EPO-resistant anemia by correcting EPO-induced erythroid progenitor depletion.


Assuntos
Anemia , Eritropoetina , Animais , Humanos , Ratos , Células Precursoras Eritroides , Eritropoetina/farmacologia , Eritropoetina/uso terapêutico , Células-Tronco Hematopoéticas , Megacariócitos , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Trombopoetina/farmacologia , Trombopoetina/uso terapêutico
11.
Eur Rev Med Pharmacol Sci ; 27(22): 10958-10967, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-38039026

RESUMO

OBJECTIVE: The aim of this study was to investigate the protective effect of recombinant erythropoietin at different doses on brain injury in premature infants and the related effects on blood routine, liver function, intellectual development, mental development index (MDI), psychomotor development index (PDI), etc. PATIENTS AND METHODS: A total of 120 premature infants were divided into four groups, including experimental group A (n=30), experimental group B (n=30), experimental group C (n=30) and control group (n=30). The experimental group was treated with different doses of recombinant erythropoietin for brain injury protection of premature infants, while the control group with conventional methods. RESULTS: There was no statistical significance in all test indicators of the four groups of patients before the intervention. After the intervention experiment, the S-100B index was p<0.05, and the erythropoietin (EPO) index was p<0.05. In the comparison of IL-6 indicators, the indicators of the experimental group were reduced after the comparison experiment, and there were significant differences, p<0.05. In neonatal behavior evaluation, there was a statistical difference between groups A and B and the control group (p<0.05), and no statistical significance was shown between group C and the control group (p>0.05). In the intelligence test comparison, the F value of the experimental group was 3.113 three months after treatment. After six months, the F value was 3.654. After nine months, the F value was 3.392 with p<0.05. In the comparison of blood routine indicators, the p-values of four indicators between groups were more than 0.05. In the comparison of liver function indexes, the indexes of groups A, B, and C were significantly changed before and after treatment, and the data after treatment were significantly different from those before treatment, p<0.05. In the comparison of development, there were no significant differences observed in the p-values of the two indicators of vigorous exercise and language in the experimental group. CONCLUSIONS: Recombinant erythropoietin has a protective effect on infants with brain injury and can improve the intellectual development of premature infants, but has no significant effect on blood routine indicators. It can effectively improve the MDI, PDI, and related cytokines of premature infants, and has certain significance for the treatment of brain injury.


Assuntos
Lesões Encefálicas , Eritropoetina , Recém-Nascido , Lactente , Humanos , Recém-Nascido Prematuro , Eritropoetina/farmacologia , Eritropoetina/uso terapêutico , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/prevenção & controle , Proteínas Recombinantes/uso terapêutico
12.
Urologiia ; (6): 64-67, 2023 Dec.
Artigo em Russo | MEDLINE | ID: mdl-38156685

RESUMO

AIM: To study the efficiency of pre-administration of a peptide mimicking the spatial structure of erythropoietin -chain B in modeling experimental post-contrast acute kidney injury. MATERIALS AND METHODS: In this study, an experimental model of post-contrast acute kidney injury was created using a non-steroidal anti-inflammatory drug, a nitric oxide synthase inhibitor, and injection of iopromide to mature male mice. After 48 hours, a comprehensive assessment of the concentration of creatinine, urea, glomerular filtration rate, the ratio of urea/albumin in the serum, as well as the level of proteinuria and interleukin 6 in the urine was carried out. RESULTS: A peptide mimicking the spatial structure of erythropoietin -chain B, administered at a dose of 100 g/kg 30 minutes before modeling of pathologic process, contributes to a significant decrease in creatinine and urea concentrations by 2.5 and 1.8 times, respectively, with an increase in glomerular filtration rate 4.4 times. In addition, in the group with pharmacological correction, there was a significant decrease in the ratio of urea/albumin by 2.2 times, a decrease in the level of proteinuria by 61.9% and a decrease in the concentration of pro-inflammatory interleukin-6 in the urine by 2.1 times. CONCLUSION: Thus, the preliminary administration of a peptide that mimics the spatial structure of the erythropoietin -chain B helps to reduce the severity of post-contrast acute kidney injury in the experiment, due to anti-inflammatory properties.


Assuntos
Injúria Renal Aguda , Eritropoetina , Masculino , Camundongos , Animais , Creatinina/uso terapêutico , Eritropoetina/farmacologia , Eritropoetina/uso terapêutico , Injúria Renal Aguda/induzido quimicamente , Peptídeos/uso terapêutico , Proteinúria/patologia , Ureia/uso terapêutico , Albuminas/uso terapêutico , Anti-Inflamatórios/uso terapêutico , Rim/patologia
13.
Cell Mol Biol (Noisy-le-grand) ; 69(10): 239-243, 2023 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-37953557

RESUMO

This study aimed to investigate the influence of recombinant human erythropoietin (rHuEPO) on pentylenetetrazol (PTZ)-induced neuronal apoptosis in epilepsy rats, and to explore the signaling pathways related to the action. Healthy Sprague-Dawley rats aged 8 weeks old were randomly divided into 5 groups, namely, control group, PTZ model group, PTZ + rHuEPO intervention group, PTZ + SB431542 + rHuEPO intervention group and PTZ + SB431542 (TGF-ß/Smad inhibitor) intervention group. The expressions of apoptotic proteins [tumor necrosis factor receptor 1 (TNFR1) and caspase-3] and the transforming growth factor-beta (TGF-ß)/Smad signaling pathway-related proteins [phosphorylated smad3 (p-smad3) and TGF-ß1] in the brain tissues were determined via Western blotting (WB). Epilepsy was successfully induced by PTZ in the rats. The results of the TUNEL assay showed that the intervention with rHuEPO could remarkably reduce the number of PTZ-induced apoptotic neurons in the hippocampus, while SB431542 inhibitor could attenuate the protective effect of rHuEPO against neuronal apoptosis (P<0.05). In addition, the intraperitoneal injection of 50 µg/kg rHuEPO could activate the TGF-ß/Smad signaling pathway, markedly up-regulate the expressions of TGF-ß1 and p-smad3 (P<0.05), down-regulate the expressions of apoptotic proteins TNFR1 and caspase-3 (P<0.01) and reduce neuronal apoptosis. Moreover, SB431542 was able to notably repress the protective effect of rHuEPO against neuronal apoptosis, and down-regulate the expressions of p-smad3 and TGF-ß1 (P<0.01). In conclusion, the inhibitory effect of rHuEPO on nerve cell apoptosis in epilepsy rats may be realized by activating the TGF-ß/Smad signaling pathway, thus relieving neuronal apoptosis and ameliorating the symptoms of epilepsy.


Assuntos
Epilepsia , Eritropoetina , Animais , Humanos , Ratos , Apoptose , Caspase 3/metabolismo , Epilepsia/induzido quimicamente , Epilepsia/tratamento farmacológico , Eritropoetina/farmacologia , Ratos Sprague-Dawley , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta , Fator de Crescimento Transformador beta1/metabolismo
14.
BMC Musculoskelet Disord ; 24(1): 894, 2023 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-37978375

RESUMO

BACKGROUND: Steroid-induced avascular necrosis of the femoral head (SANFH) is characterized by osteoblast apoptosis, leading to a loss of bone structure and impaired hip joint function. It has been demonstrated that erythropoietin (EPO) performs a number of biological roles. OBJECTIVE: We examined the effects of EPO on SANFH and its regulation of the STAT1-caspase 3 signaling pathway. METHOD: In vitro, osteoblasts were treated with dexamethasone (Dex) or EPO. We identified the cytotoxicity of EPO by CCK-8, the protein expression of P-STAT1, cleaved-caspase9, cleaved-caspase3, Bcl-2, BAX, and cytochrome c by Western blotting, and evaluated the apoptosis of osteoblasts by flow cytometry. In vivo, we analyzed the protective effect of EPO against SANFH by hematoxylin and eosin (H&E), Immunohistochemical staining, and Micro-computed tomography (CT). RESULTS: In vitro, EPO had no apparent toxic effect on osteoblasts. In Dex-stimulated cells, EPO therapy lowered the protein expression of BAX, cytochrome c, p-STAT1, cleaved-caspase9, and cleaved-caspase3 while increasing the expression of Bcl-2. EPO can alleviate the apoptosis induced by Dex. In vivo, EPO can lower the percentage of empty bone lacunae in SANFH rats. CONCLUSION: The present study shows that EPO conferred beneficial effects in rats with SANFH by inhibiting STAT1-caspase 3 signaling, suggesting that EPO may be developed as a treatment for SANFH.


Assuntos
Eritropoetina , Necrose da Cabeça do Fêmur , Ratos , Animais , Caspase 3/metabolismo , Proteína X Associada a bcl-2/metabolismo , Necrose da Cabeça do Fêmur/induzido quimicamente , Necrose da Cabeça do Fêmur/tratamento farmacológico , Necrose da Cabeça do Fêmur/metabolismo , Citocromos c/metabolismo , Citocromos c/farmacologia , Microtomografia por Raio-X , Apoptose , Transdução de Sinais , Osteoblastos/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Eritropoetina/farmacologia , Esteroides/efeitos adversos
15.
Sci Rep ; 13(1): 19522, 2023 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-37945644

RESUMO

Myelination is a process tightly regulated by a variety of neurotrophic factors. Here, we show-by analyzing two transgenic mouse lines, one overexpressing EPO selectively in the brain Tg21(PDGFB-rhEPO) and another with targeted removal of EPO receptors (EPORs) from oligodendrocyte progenitor cells (OPC)s (Sox10-cre;EpoRfx/fx mice)-a key function for EPO in regulating developmental brain myelination. Overexpression of EPO resulted in faster postnatal brain growth and myelination, an increased number of myelinating oligodendrocytes, faster axonal myelin ensheathment, and improved motor coordination. Conversely, targeted ablation of EPORs from OPCs reduced the number of mature oligodendrocytes and impaired motor coordination during the second postnatal week. Furthermore, we found that EPORs are transiently expressed in the subventricular zone (SVZ) during the second postnatal week and EPO increases the postnatal expression of essential oligodendrocyte pro-differentiation and pro-maturation (Nkx6.2 and Myrf) transcripts, and the Nfatc2/calcineurin pathway. In contrast, ablation of EPORs from OPCs inactivated the Erk1/2 pathway and reduced the postnatal expression of the transcripts. Our results reveal developmental time windows in which EPO therapies could be highly effective for stimulating oligodendrocyte maturation and myelination.


Assuntos
Eritropoetina , Oligodendroglia , Camundongos , Animais , Oligodendroglia/metabolismo , Bainha de Mielina/metabolismo , Encéfalo/metabolismo , Camundongos Transgênicos , Eritropoetina/genética , Eritropoetina/farmacologia , Eritropoetina/metabolismo , Diferenciação Celular/fisiologia
16.
Bioconjug Chem ; 34(12): 2205-2214, 2023 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-38032892

RESUMO

In mammalian cells, growth factor-induced intracellular signaling and protein synthesis play a critical role in cellular physiology and homeostasis. In the brain's glymphatic system (GS), the water-conducting activity of aquaporin-4 (AQPN-4) membrane channels (expressed in polarized fashion on astrocyte end-feet) mediates the clearance of wastes through the convective transport of fluid and solutes through the perivascular space. The glycoprotein erythropoietin (EPO) has been shown to induce the astrocyte expression of AQPN-4 via signaling through the EPO receptor and the JAK/STAT signaling pathway. Here, we self-assemble EPO in a multivalent fashion onto the surface of semiconductor quantum dots (QDs) (driven by polyhistidine-based self-assembly) to drive the interaction of the bioconjugates with EPOR on human astrocytes (HA). This results in a 2-fold augmentation of JAK/STAT signaling activity and a 1.8-fold enhancement in the expression of AQPN-4 in cultured primary HA compared to free EPO. This translates into a 2-fold increase in the water transport rate in HA cells as measured by the calcein AM water transport assay. Importantly, EPO-QD-induced augmented AQPN-4 expression does not elicit any deleterious effect on the astrocyte viability. We discuss our results in the context of the implications of EPO-nanoparticle (NP) bioconjugates for use as research tools to understand the GS and their potential as therapeutics for the modulation of GS function. More generally, our results illustrate the utility of NP bioconjugates for the controlled modulation of growth factor-induced intracellular signaling.


Assuntos
Aquaporinas , Eritropoetina , Pontos Quânticos , Animais , Humanos , Astrócitos/metabolismo , Receptores da Eritropoetina/metabolismo , Eritropoetina/metabolismo , Eritropoetina/farmacologia , Água/metabolismo , Aquaporinas/metabolismo , Aquaporinas/farmacologia , Mamíferos/metabolismo
17.
Allergol Immunopathol (Madr) ; 51(6): 60-66, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37937497

RESUMO

BACKGROUND: Severe pneumonia is a kind of disease that develops from lung inflammation, and new drugs are still required to treat the same. Erythropoietin (EPO) is widely used to treat anemia in patients. However, there are fewer studies on the role of EPO in neutrophil extracellular trappings (NETs) and pneumonia, and the mechanism is unclear. OBJECTIVE: To investigate the possible effects of EPO on the formation of NETs and progression of pneumonia. METHODS: Mice pneumonia model was induced by tracheal lipopolysaccharide (LPS) administration. Hematoxylin and eosin (H&E) staining and automatic blood cell analysis were performed in this model to confirm the effects of EPO on lung injury. Flow cytometry, enzyme-linked immunosorbent serological assay, and immunostaining assay were conducted to detect the effects of EPO on the inflammation as well as formation of NETs in mice. Immunoblot was further conducted to confirm the mechanism. RESULTS: EPO alleviated LPS-induced lung injury. EPO reduced the release of inflammatory factors induced by LPS. In addition, EPO inhibited the formation of NETs. Mechanically, EPO inhibited tumor necrosis factor (TNF) receptor associated factor 2 (TRAF2)/nuclear factor kappa-B (NF-κB) activity in mouse models, and therefore suppressed the progression of pneumonia. CONCLUSION: EPO inhibited formation of NETs to ameliorate lung injury in a pneumonia model, and could serve as a drug of pneumonia.


Assuntos
Lesão Pulmonar Aguda , Eritropoetina , Armadilhas Extracelulares , Pneumonia , Humanos , Camundongos , Animais , Lipopolissacarídeos/efeitos adversos , Pneumonia/induzido quimicamente , Eritropoetina/uso terapêutico , Eritropoetina/farmacologia , Lesão Pulmonar Aguda/tratamento farmacológico
18.
Neuroreport ; 34(17): 801-810, 2023 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-37938927

RESUMO

To investigate the neuroprotection of recombinant human erythropoietin (rhEPO) against hypoxic/ischemic (HI) insult in three-day-old rats. Postnatal day 3 (PD3) rats were randomly divided into three groups: Sham group, HI group and HI+rhEPO group. Ligation of the right common carotid artery and hypoxia to induce HI brain injury. After HI insult, the rats received intraperitoneal injection of rhEPO (5000 IU/Kg, qod) in HI+rhEPO group or equal saline in other groups. On PD10, damage of brain tissue was examined by hematoxylin-eosin (HE) staining, observation of neuronal apoptosis in the hippocampus and cortex using immunofluorescence assay (marker: TUNEL). Immunohistochemical staining or western blotting was performed to detect the expression of cyclooxygenase-2 (COX-2), Caspase-3 and phosphorylated Akt (p-Akt) protein. On PD28, cognitive ability of rats was assessed by Morris water maze test. HI injury causes brain pathological morphology and cognitive function damage in PD3 rats, which can be alleviated by rhEPO intervention. Compared with the HI group, the HI+rhEPO group showed an increase in platform discovery rate and cross platform frequency, while the search platform time was shortened (P < 0.05). The proportion of TUNEL positive neurons and the expression of COX-2 and Caspase-3 proteins in brain tissue in the hippocampus and cortex was decreased, while the expression of p-Akt protein was upregulated (P < 0.05). RhEPO could protect against the pathological and cognitive impairment of immature brain induced by HI insult. This neuroprotective activity may involve in inhibiting inflammatory and apoptosis by activation of PI3K/Akt signaling pathway.


Assuntos
Eritropoetina , Hipóxia-Isquemia Encefálica , Fármacos Neuroprotetores , Humanos , Ratos , Animais , Caspase 3/metabolismo , Ratos Sprague-Dawley , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ciclo-Oxigenase 2/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Encéfalo/metabolismo , Hipóxia/metabolismo , Hipóxia-Isquemia Encefálica/complicações , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Hipóxia-Isquemia Encefálica/metabolismo , Eritropoetina/farmacologia , Eritropoetina/uso terapêutico , Isquemia/metabolismo , Animais Recém-Nascidos , Fármacos Neuroprotetores/farmacologia
19.
Aging (Albany NY) ; 15(21): 12264-12274, 2023 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-37934569

RESUMO

PURPOSE: To investigate the influences and mechanism of erythropoietin (EPO) on the cognitive function of vascular dementia (VD) rats. METHODS: 1) Spatial memory capacity was assessed by Morris water maze test; 2) Pathological conditions of brain tissues were detected by hematoxylin-eosin (HE) staining; 3) The effect of treatment on apoptosis was observed by terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) staining; 4) Western blotting was used to examine the protein expression in hippocampal neurons. RESULTS: The escape latency and swimming distance in the EPO group were much shorter than those in the Model group on the fifth day. In the spatial exploration test, the time spent in the target quadrant was longer, the number of platform crossings was larger and the swimming speed was higher in the Sham group and EPO group than those in the Model group. The results of HE staining showed that the cells in the hippocampal CA1 region were arranged closely in the Sham group, loosely and disorderly in the Model group but significantly better in the EPO group. Compared with that in the Model group, the number of apoptotic cells in the EPO group was obviously smaller. The results of Western blotting revealed that the expressions of EPO, p-EPOR, p-SHP2, p-TrKB, p-PI3K, p-ERK1/2 and Bcl-2 rose, while the expressions of P22, P47, Caspase-3, Caspase-9 and Bax significantly declined in the EPO group. CONCLUSIONS: EPO can effectively ameliorate the cognitive dysfunction induced by chronic hypoperfusion in VD rats by mediating oxidative stress-related pathways.


Assuntos
Demência Vascular , Eritropoetina , Ratos , Animais , Demência Vascular/tratamento farmacológico , Demência Vascular/metabolismo , Ratos Sprague-Dawley , Eritropoetina/farmacologia , Hipocampo/metabolismo , Apoptose , Cognição
20.
J Mol Endocrinol ; 71(3)2023 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-37924639

RESUMO

Several studies provide evidence that erythropoietin (EPO) could play an important role in the recovery of the heart subjected to ischemia-reperfusion. In this regard, it has been suggested that EPO could be involved in protein kinase B (Akt) activation as a cell survival protein. The aim of the present study was to investigate the effects of EPO on the Akt/glycogen synthase kinase 3 beta (GSK-3ß) pathway in the presence or absence of wortmannin (W, Akt inhibitor) and its relationship with mitochondrial morphology and function preservation in ischemic-reperfused rat hearts. EPO improved the functional recovery of the heart subjected to ischemia-reperfusion, reduced the release of CK and the infarct size, and promoted preservation of the mitochondrial structure. Moreover, it reduced tissue lactate content and preserved glycogen in order to prevent ischemia. The results showed greater Akt activation, accompanied by preservation of swelling and mitochondrial calcium retention capacity, as well as an increase in ATP synthesis capacity. These results were accompanied by an inhibition of GSK-3ß, suggesting regulation of Akt on the opening of the mitochondrial permeability transition pore. All these beneficial effects exerted by acute treatment with EPO were prevented by W. The present study provided novel evidence that EPO not only enhances intrinsic activation of Akt during myocardial ischemia-reperfusion but also promotes GSK-3ß inhibition, contributing to mitochondrial structure and function preservation.


Assuntos
Cardiotônicos , Eritropoetina , Coração , Proteínas Proto-Oncogênicas c-akt , Traumatismo por Reperfusão , Animais , Ratos , Eritropoetina/farmacologia , Glicogênio Sintase Quinase 3 beta/metabolismo , Isquemia , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Cardiotônicos/farmacologia , Coração/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...