Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
PeerJ ; 11: e15706, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37663284

RESUMO

Background: N6-methyladenosine (m6A) methylation epigenetically regulates normal hematopoiesis and plays a role in the pathogenesis of acute myeloid leukemia (AML). However, its potential value for prognosis remains elusive. Methods: Analysis of the datasets downloaded from The Cancer Genome Atlas and Genotype Tissue Expression databases revealed that the expression level of 20 regulators related to m6A RNA methylation differ between patients with AML and normal individuals. A prognostic risk model with three genes (YTHDF3, IGF2BP3, and HNRNPA2B1) was developed using univariate Cox regression and the least absolute shrinkage and selection operator Cox regression methods. Results: This established signature demonstrated good predictive efficacy with an area under the curve of 0.892 and 0.731 in the training cohort and the validation cohort, respectively. Patients with AML and an increased level of Insulin growth factor 2 mRNA binding protein 3 (IGF2BP3) expression exhibited a poor prognosis. IGF2BP3 knockdown significantly induced G0/G1 phase arrest and inhibited cell proliferation, apoptosis, and/or differentiation. Further, the JAK/STAT pathway may be involved in the regulation of EPOR expression by IGF2BP3-mediated m6A RNA methylation. Conclusion: These findings indicate that IGF2BP3 plays a carcinogenic role in AML, implying that it can predict patient survival and could be an effective strategy for AML therapy.


Assuntos
Insulina , Leucemia Mieloide Aguda , Humanos , Adenosina , Insulina Regular Humana , Peptídeos e Proteínas de Sinalização Intercelular , Janus Quinases , Leucemia Mieloide Aguda/genética , RNA Mensageiro/genética , Transdução de Sinais , Fatores de Transcrição STAT
2.
3.
Br J Haematol ; 198(1): 131-136, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35355248

RESUMO

Myeloproliferative neoplasms (MPN) are mainly sporadic but inherited variants have been associated with higher risk development. Here, we identified an EPOR variant (EPORP488S ) in a large family diagnosed with JAK2V617F -positive polycythaemia vera (PV) or essential thrombocytosis (ET). We investigated its functional impact on JAK2V617F clonal amplification in patients and found that the variant allele fraction (VAF) was low in PV progenitors but increase strongly in mature cells. Moreover, we observed that EPORP488S alone induced a constitutive phosphorylation of STAT5 in cell lines or primary cells. Overall, this study points for searching inherited-risk alleles affecting the JAK2/STAT pathway in MPN.


Assuntos
Transtornos Mieloproliferativos , Policitemia Vera , Receptores da Eritropoetina , Trombocitemia Essencial , Alelos , Mutação com Ganho de Função , Humanos , Janus Quinase 2/genética , Janus Quinase 2/metabolismo , Mutação , Transtornos Mieloproliferativos/diagnóstico , Transtornos Mieloproliferativos/genética , Policitemia Vera/genética , Receptores da Eritropoetina/genética , Trombocitemia Essencial/genética
4.
Stem Cell Rev Rep ; 18(4): 1408-1416, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34775556

RESUMO

In my previous study, I demonstrated that bone marrow-derived mononuclear cells (BM MNCs) secrete copious amounts of Transforming Growth Factor ß1 (TGFß1) in response to erythropoietin (EPO). In this study, I investigated the principal cell type involved in the process. I found that a large percentage of various marrow cells, but not their mature counterparts present in the peripheral blood, express EPO-receptors (EPO-R). Cell depletion experiments showed that depletion of Glycophorin positive erythroblasts and CD41+ megakaryocytes - the prime suspects - did not affect the EPO-mediated TGFß1 secretion by the BM MNCs. However, individual depletion of CD2+ T lymphocytes, CD14+ monocyte/macrophages, and CD19+ B cells affected the TGFß1 secretion by EPO-primed MNCs: depletion of CD2+ cells had the most striking effect. Unexpectedly, and most interestingly, depletion of CD15+ granulocytes led to a significant increase in the TGFß1 secretion by both naïve and EPO-primed BM MNCs, suggesting that these cells negatively regulate the process. Mechanistically, I show that the CD15+ cells exert this regulatory effect via secretion of both full-length and soluble EPO-R in the milieu. Overall my results, for the first time, unravel an in-built regulatory mechanism prevailing in the BM microenvironment that regulates the secretion of TGFß1 by controlling EPO-EPO-R interaction.My data could be relevant in understanding the pathophysiology of several conditions associated with deregulated production of TGFß1 in the marrow compartment.


Assuntos
Receptores da Eritropoetina , Fator de Crescimento Transformador beta1 , Medula Óssea , Granulócitos , Megacariócitos , Receptores da Eritropoetina/genética
5.
Front Immunol ; 12: 768435, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34925338

RESUMO

Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is a condition with an imbalanced inflammatory response and delayed resolution of inflammation. Macrophage polarization plays an important role in inflammation and resolution. However, the mechanism of macrophage polarization in ALI/ARDS is not fully understood. We found that mice with lipopolysaccharide administration developed lung injury with the accumulation of extracellular cold-inducible RNA-binding protein (eCIRP) in the lungs. eCIRP, as a damage-associated molecular pattern (DAMP), inhibited M2 macrophage polarization, thereby tipping the balance toward inflammation rather than resolution. Anti-CIRP antibodies reversed such phenotypes. The levels of macrophage erythropoietin (EPO) receptor (EPOR) were reduced after eCIRP treatment. Myeloid-specific EPOR-deficient mice displayed restrained M2 macrophage polarization and impaired inflammation resolution. Mechanistically, eCIRP impaired Rab26, a member of Ras superfamilies of small G proteins, and reduced the transportation of surface EPOR, which resulted in macrophage polarization toward the M1 phenotype. Moreover, EPO treatment hardly promotes M2 polarization in Rab26 knockout (KO) macrophages through EPOR. Collectively, macrophage EPOR signaling is impaired by eCIRP through Rab26 during ALI/ARDS, leading to the restrained M2 macrophage polarization and delayed inflammation resolution. These findings identify a mechanism of persistent inflammation and a potential therapy during ALI/ARDS.


Assuntos
Lesão Pulmonar Aguda/imunologia , Macrófagos/fisiologia , Proteínas de Ligação a RNA/fisiologia , Receptores da Eritropoetina/fisiologia , Proteínas rab de Ligação ao GTP/fisiologia , Animais , Polaridade Celular , Células Cultivadas , Inflamação/etiologia , Camundongos , Camundongos Endogâmicos C57BL , PPAR gama/fisiologia
6.
Folia Med (Plovdiv) ; 63(4): 475-480, 2021 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-35851166

RESUMO

INTRODUCTION: Erythropoietin (Epo) controls a variety of signal transduction pathways during oxidative stress. The main function of Epo and its receptor (EpoR) is the stimulation of erythropoiesis. AIM: The role of Epo and EpoR on non-hematopoietic normal and cancerous tissues is still poorly understood. This is the first report in which we aimed to investigate the role of Epo and EpoR systems at oxidative condition in human basal cell carcinoma (BCC), which is the most common tumour in the world.Materials and methods: Fresh normal and cancerous skin paired tissue was obtained from 63 patients who underwent curative BCC resection in Kahramanmaras, Turkey. Preliminary diagnosis of BCC was made in the dermatology clinic by excision and then the diagnosis was confirmed as histopathologic findings. Oxidative stress biomarkers such as superoxide dismutase (SOD) and catalase (CAT) activities, and malondialdehyde (MDA) levels in biopsy samples were measured spectrophotometrically, and also the levels of Epo and EpoR were measured by ELISA. RESULTS: While the levels of MDA in cancerous tissue of patients with skin BCC were significantly higher than normal neighbouring skin tissue (p<0.05), SOD and CAT activities decreased (p<0.05). Furthermore, a remarkable increase was found in the Epo level ofpatients with skin BCC in comparison with the normal neighbouring skin tissue (p<0.05). However, we found that EpoR levels decreased (p<0.05). CONCLUSIONS: Results indicate that there is an active oxidative process in BCC biopsies. The levels of increased Epo and decreased EpoR in oxidative condition due to hypoxia may aggravate tumour growth by its angiogenic activity.


Assuntos
Carcinoma , Eritropoetina , Eritropoetina/metabolismo , Humanos , Hipóxia , Estresse Oxidativo , Receptores da Eritropoetina/análise , Receptores da Eritropoetina/metabolismo , Transdução de Sinais , Superóxido Dismutase/metabolismo
7.
Mol Ther ; 29(1): 47-59, 2021 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-33010232

RESUMO

Many investigational adoptive immunotherapy regimens utilizing natural killer (NK) cells require the administration of interleukin-2 (IL-2) or IL-15, but these cytokines cause serious dose-dependent toxicities. To reduce or preclude the necessity for IL-2 use, we investigated whether genetic engineering of NK cells to express the erythropoietin (EPO) receptor (EPOR) or thrombopoietin (TPO) receptor (c-MPL) could be used as a method to improve NK cell survival and function. Viral transduction of NK-92 cells to express EPOR or c-MPL receptors conveyed signaling via appropriate pathways, protected cells from apoptosis, augmented cellular proliferation, and increased cell cytotoxic function in response to EPO or TPO ligands in vitro. In the presence of TPO, viral transduction of primary human NK cells to express c-MPL enhanced cellular proliferation and increased degranulation and cytokine production toward target cells in vitro. In contrast, transgenic expression of EPOR did not augment the proliferation of primary NK cells. In immunodeficient mice receiving TPO, in vivo persistence of primary human NK cells genetically modified to express c-MPL was higher compared with control NK cells. These data support the concept that genetic manipulation of NK cells to express hematopoietic growth factor receptors could be used as a strategy to augment NK cell proliferation and antitumor immunity.


Assuntos
Expressão Gênica , Imunomodulação/genética , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Neoplasias/imunologia , Neoplasias/terapia , Receptores da Eritropoetina/genética , Receptores de Trombopoetina/genética , Animais , Modelos Animais de Doenças , Engenharia Genética , Humanos , Imunoterapia/métodos , Camundongos , Transgenes
8.
Ann Palliat Med ; 9(6): 4010-4016, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33222459

RESUMO

BACKGROUND: Proliferative diabetic retinopathy (PDR) is a leading cause of blindness. This study aimed to analyze the expression of the erythropoietin receptor (EpoR) in patients with PDR and its correlation with postoperative visual prognosis. METHODS: Between May 2016 and May 2017, 89 patients who underwent vitreoretinal surgery (VRS) in Affiliated Hospital of North Sichuan Medical College were enrolled. Among them, 45 patients (52 eyes) with PDR and 44 patients (50 eyes) with retinal vein occlusion (RVO) made up the study group and the control group, respectively. All patients received 25G standard flattened three-channel lens-preserving VRS; all procedures were performed by the same physician. Recovery and change in best corrected visual acuity (BCVA) were observed. Subsequently, the patients were divided into group A (improved vision) and group B (no improvement in vision). The integrated optical density (IOD) value of EpoR expression among the groups were compared. Pearson's correlation analysis was used to analyze the correlation between the IOD values of EpoR and the change in visual acuity after surgery for PDR. RESULTS: The IOD value of EpoR in the study group was higher than that in the control group (P<0.05). The postoperative BCVA of the PDR patients was significantly higher than that before surgery (≥0.1 vs. ≤0.02, P<0.05). Of the 45 PDR patients (52 eyes), 34 patients (39 eyes) had improved visual acuity after surgery (group A), while 11 patients (13 eyes) experienced no improvement (group B). The IOD value of EpoR expression in group B was higher than that in group A (P<0.05). Pearson's correlation analysis showed that the IOD value of EpoR expression were positively correlated with the recovery. CONCLUSIONS: EpoR is expressed at a high level in PDR patients. The expression level of EpoR in the epiretinal membrane of PDR patients is positively correlated with the prognosis of postoperative vision. Therefore, EpoR has promise as a prognostic biomarker and a potential therapeutic target for PDR.


Assuntos
Diabetes Mellitus , Retinopatia Diabética , Receptores da Eritropoetina , Humanos , Prognóstico , Acuidade Visual
9.
Blood Cells Mol Dis ; 85: 102479, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32739800

RESUMO

Congenital erythrocytosis (CE) can be classified as primary and secondary and 82 consecutive patients of erythrocytosis who were JAK-2 mutation negative, were further investigated. The genomic DNA was extracted from all the patients and the EPO-R, VHL, EGLN1 and EPAS1 genes were PCR amplified and sequenced. The sequence analysis showed (28/82) 34.14% patients had mutations. Among them, (19/28) 67.86% patients had mutations in exon 8 of EPO-R gene, of which six were novel missense mutations, p.(Gly418Ala), p.(Gly390Ala), p.(Ala411Thr), p.(Gly475Val), p.(Glu490Asp), p.(Glu362Gln) and three were novel frameshift mutations, p.(Glu336*), p.(Pro327Hisfs*68), p.(Gly479Alafs*37). All these EPO-R patients were heterozygotes and were forming endogenous erythrocyte colonies (EEC). Some patients (8/28) 28.57% had mutations in VHL gene, out of which 3 novel homozygous missense mutations in exon 1 of VHL gene, p.Gly80Asp, p.Gln107Glu and p.Gln113Glu, were identified. In addition, (1/28) 3.5% patients had one reported heterozygous missense mutation in exon 12 of EPAS1 gene p.Gly537Arg and one novel frameshift mutation p.(Ala553Glyfs*58). Further, in silico analysis indicated most of the mutations, probably, were damaging the protein structures, causing the CE in these patients. In this study the mutations in EPO-R and EPAS1 genes were identified for the first time in India.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Policitemia/congênito , Receptores da Eritropoetina/genética , Proteína Supressora de Tumor Von Hippel-Lindau/genética , Adulto , Feminino , Mutação da Fase de Leitura , Humanos , Masculino , Pessoa de Meia-Idade , Mutação , Mutação de Sentido Incorreto , Mutação Puntual , Policitemia/genética , Adulto Jovem
12.
Saudi J Biol Sci ; 27(2): 653-658, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32210684

RESUMO

Erythropoietin receptors (EPORs) are present not only in erythrocyte precursors but also in non-hematopoietic cells including cancer cells. In this study, we determined the effect of fetal bovine serum (FBS) in culture medium on the EPOR expression and viability of the estrogen receptor (ER)-positive MCF-7 and ER-negative MDA-MB-231 breast cancer cells. Using flow cytometry, we showed that the inclusion of 10% FBS in the medium increased the EPOR expressions and viabilities of MDA-MB-231 and MCF-7 cells. The MDA-MB-231 showed greater EPOR expression than MCF-7 cells, suggesting that the presence of ERs on cells is associated with poor expression of EPOR. Culture medium containing 10% FBS also caused increased number of breast cancer cells entering the synthesis phase of the cell cycle. The study also showed that rHuEPO treatment did not affect viability of breast cancer cells. In conclusion, it was shown that the inclusion of FBS in culture medium increased expression of EPOR in breast cancer cells and rHuEPO treatment had no effect on the proliferation of these cancer cells.

13.
Cell Signal ; 69: 109554, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32027948

RESUMO

The formation of erythroid progenitor cells depends sharply upon erythropoietin (EPO), its cell surface receptor (erythropoietin receptor, EPOR), and Janus kinase 2 (JAK2). Clinically, recombinant human EPO (rhEPO) additionally is an important anti-anemia agent for chronic kidney disease (CKD), myelodysplastic syndrome (MDS) and chemotherapy, but induces hypertension, and can exert certain pro-tumorigenic effects. Cellular signals transduced by EPOR/JAK2 complexes, and the nature of EPO-modulated signal transduction factors, therefore are of significant interest. By employing phospho-tyrosine post-translational modification (p-Y PTM) proteomics and human EPO- dependent UT7epo cells, we have identified 22 novel kinases and phosphatases as novel EPO targets, together with their specific sites of p-Y modification. New kinases modified due to EPO include membrane palmitoylated protein 1 (MPP1) and guanylate kinase 1 (GUK1) guanylate kinases, together with the cytoskeleton remodeling kinases, pseudopodium enriched atypical kinase 1 (PEAK1) and AP2 associated kinase 1 (AAK1). Novel EPO- modified phosphatases include protein tyrosine phosphatase receptor type A (PTPRA), phosphohistidine phosphatase 1 (PHPT1), tensin 2 (TENC1), ubiquitin associated and SH3 domain containing B (UBASH3B) and protein tyrosine phosphatase non-receptor type 18 (PTPN18). Based on PTPN18's high expression in hematopoietic progenitors, its novel connection to JAK kinase signaling, and a unique EPO- regulated PTPN18-pY389 motif which is modulated by JAK2 inhibitors, PTPN18's actions in UT7epo cells were investigated. Upon ectopic expression, wt-PTPN18 promoted EPO dose-dependent cell proliferation, and survival. Mechanistically, PTPN18 sustained the EPO- induced activation of not only mitogen-activated protein kinases 1 and 3 (ERK1/2), AKT serine/threonine kinase 1-3 (AKT), and signal transducer and activator of transcription 5A and 5B (STAT5), but also JAK2. Each effect further proved to depend upon PTPN18's EPO- modulated (p)Y389 site. In analyses of the EPOR and the associated adaptor protein RHEX (regulator of hemoglobinization and erythroid cell expansion), wt-PTPN18 increased high molecular weight EPOR forms, while sharply inhibiting the EPO-induced phosphorylation of RHEX-pY141. Each effect likewise depended upon PTPN18-Y389. PTPN18 thus promotes signals for EPO-dependent hematopoietic cell growth, and may represent a new druggable target for myeloproliferative neoplasms.


Assuntos
Eritropoese , Eritropoetina/metabolismo , Janus Quinase 2/metabolismo , Fragmentos de Peptídeos/metabolismo , Proteínas Tirosina Fosfatases não Receptoras/fisiologia , Receptores da Eritropoetina/metabolismo , Linhagem Celular , Humanos , Proteômica , Transdução de Sinais
14.
Adv Exp Med Biol ; 1223: 17-30, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32030683

RESUMO

Erythropoietin (EPO), the primary cytokine of erythropoiesis, stimulates both proliferation and differentiation of erythroid progenitors and their maturation to red blood cells. Basal EPO levels maintain the optimum levels of circulating red blood cells. However, during hypoxia, EPO secretion and its expression is elevated drastically in renal interstitial fibroblasts, thereby increasing the number of erythroid progenitors and accelerating their differentiation to mature erythrocytes. A tight regulation of this pathway is therefore of paramount importance. The biological response to EPO is commenced through the involvement of its cognate receptor, EPOR. The receptor-ligand complex results in homodimerization and conformational changes, which trigger downstream signaling events and cause activation or inactivation of critical transcription factors that promote erythroid expansion. In recent years, recombinant human EPO (rEPO) has been widely used as a therapeutic tool to treat a number of anemias induced by infection, and chemotherapy for various cancers. However, several studies have uncovered a tumor promoting ability of EPO in man, which likely occurs through EPOR or alternative receptor(s). On the other hand, some studies have demonstrated a strong anticancer activity of EPO, although the mechanism still remains unclear. A thorough investigation of EPOR signaling could yield enhanced understanding of the pathobiology for a variety of disorders, as well as the potential novel therapeutic strategies. In this chapter, in addition to the clinical relevance of EPO/EPOR signaling, we review its anticancer efficacy within various tumor microenvironments.


Assuntos
Eritropoetina/metabolismo , Saúde , Neoplasias/metabolismo , Receptores da Eritropoetina/metabolismo , Transdução de Sinais , Microambiente Tumoral , Eritropoese , Humanos
15.
Saudi J Biol Sci ; 27(1): 407-411, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31889864

RESUMO

Tobacco smoking is a common risk factor of cardiovascular diseases, cancers and heart health problems. In Taif, the number of secondary polycythemia patients is increasing dramatically and most of those patients are heavy smokers. Therefore, this study is an attempt to understand the pathophysiological mechanism behind that problem. Whole blood and serum samples were collected from forty healthy people and forty tobacco smokers, voluntary for this study. Complete blood counts revealed a significant increase in the red blood cell count, hemoglobin concentrations, hematocrit and neutrophils with some elevations in total white blood cells, lymphocytes and monocytes. Moreover, serum analysis of both erythropoietin and interleukin-7 showed a significant reduction in their levels among smokers which were about 35% and 65% respectively. Gene expression study showed a significant upregulation of RAG-1, RAG-2 and EPOR-1 genes caused by tobacco smoking. In conclusion, data presented in the current study suggest that tobacco smoking might cause alveolar tissue inflammation and vascular injury causing an immune response that elevates the white blood cells count. Another suggestion is that tobacco smoking defects the pulmonary gaseous exchange mechanism leading to the secondary polycythemia indicated by the increase in red blood cell count, hemoglobin levels, hematocrit and by the low serum erythropoietin levels.

16.
Front Cell Dev Biol ; 8: 613885, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33644032

RESUMO

Erythroblastic islands (EBIs), discovered more than 60 years ago, are specialized microenvironments for erythropoiesis. This island consists of a central macrophage with surrounding developing erythroid cells. EBI macrophages have received intense interest in the verifications of the supporting erythropoiesis hypothesis. Most of these investigations have focused on the identification and functional analyses of EBI macrophages, yielding significant progresses in identifying and isolating EBI macrophages, as well as verifying the potential roles of EBI macrophages in erythropoiesis. EBI macrophages express erythropoietin receptor (Epor) both in mouse and human, and Epo acts on both erythroid cells and EBI macrophages simultaneously in the niche, thereby promoting erythropoiesis. Impaired Epor signaling in splenic niche macrophages significantly inhibit the differentiation of stress erythroid progenitors. Moreover, accumulating evidence suggests that EBI macrophage dysfunction may lead to certain erythroid hematological disorders. In this review, the heterogeneity, identification, and functions of EBI macrophages during erythropoiesis under both steady-state and stress conditions are outlined. By reviewing the historical data, we discuss the influence of EBI macrophages on erythroid hematopoietic disorders and propose a new hypothesis that erythroid hematopoietic disorders are driven by EBI macrophages.

17.
Cell Commun Signal ; 17(1): 152, 2019 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-31752873

RESUMO

BACKGROUND: Overexpression of erythropoietin (EPO) and EPO receptor (EPO-R) is associated with poor prognosis in non-small-cell lung carcinoma (NSCLC). Hypoxia, a potent EPO inducer, is a major stimulating factor in the growth of solid tumors. However, how EPO-R expression is regulated under hypoxia is largely unknown. METHODS: The role of EPO-R in NSCLC cell proliferation was assessed by RNA interference in vitro. Luciferase reporter assays were performed to map the promoter elements involved in the EPO-R mRNA transcription. Nuclear co-immunoprecipitation and chromatin immunoprecipitation were performed to assess the interaction among transcription factors HIF1α, SP1, and EGR1 in the regulation of EPO-R under hypoxia. The expression of key EPO-R transcription factors in clinical specimens were determined by immunohistochemistry. RESULTS: Hypoxia induced a dosage and time dependent EPO-R mRNA expression in NSCLC cells. Knockdown of EPO-R reduced NSCLC cell growth under hypoxia (P < 0.05). Mechanistically, a SP1-EGR1 overlapped DNA binding sequence was essential to the hypoxia induced EPO-R transcription. In the early phase of hypoxia, HIF1α interacted with EGR1 that negatively regulated EPO-R. With the exit of EGR1 in late phase, HIF1α positively regulated EPO-R expression through additive interaction with SP1. In clinical NSCLC specimen, SP1 was positively while EGR1 was negatively associated with active EPO-R expression (P < 0.05). CONCLUSIONS: HIF1α, SP1 and EGR1 mediated EPO-R expression played an essential role in hypoxia-induced NSCLC cell proliferation. Our study presents a novel mechanism of EPO-R regulation in the tumor cells, which may provide information support for NSCLC diagnosis and treatment.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Hipóxia/genética , Neoplasias Pulmonares/metabolismo , Receptores da Eritropoetina/genética , Fator de Transcrição Sp1/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular , Proliferação de Células , Humanos , Hipóxia/metabolismo , Neoplasias Pulmonares/patologia , Receptores da Eritropoetina/metabolismo , Transdução de Sinais
18.
G Ital Nefrol ; 36(5)2019 Sep 24.
Artigo em Italiano | MEDLINE | ID: mdl-31580542

RESUMO

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.


Assuntos
Eritropoese/fisiologia , Eritropoetina/fisiologia , Sobrevivência de Enxerto/fisiologia , Receptores da Eritropoetina/fisiologia , Imunidade Adaptativa , Anemia/tratamento farmacológico , Anemia/etiologia , Animais , Hipóxia Celular/fisiologia , Eritropoetina/genética , Eritropoetina/farmacologia , Sobrevivência de Enxerto/efeitos dos fármacos , Coração/efeitos dos fármacos , Humanos , Imunidade Celular , Imunidade Inata , Rim/efeitos dos fármacos , Rim/metabolismo , Camundongos , Sistema Nervoso/metabolismo , Transplante de Órgãos , Ratos , Receptores de Fator Estimulador de Colônias/fisiologia , Proteínas Recombinantes/uso terapêutico , Insuficiência Renal Crônica/complicações , Retina/metabolismo
19.
Blood Sci ; 1(1): 61-64, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35402789

RESUMO

Erythroblastic island (EBI), composed of a central macrophage surrounded by developing erythroid cells, is a structure found in hematopoietic tissues such as fetal liver and bone marrow. It is the first described hematopoietic niche that predominantly supports erythropoiesis. Although it is well accepted that EBIs and EBI macrophage play important roles during erythropoiesis, the mechanisms by which they support erythropoiesis remain largely unclear due to our inability to identify and isolate EBI macrophages. Earlier efforts to identify surface markers for EBI macrophages have focused on the adhesion molecules which are involved in macrophage's interaction with erythroblasts. These include EMP, Vcam1, CD169, CD163, and αV integrin. Findings from these earlier studies suggested that combination of Vcam1, CD169, and mouse macrophage surface marker F4/80 can be used to define mouse EBI macrophage. We found that not all F4/80+Vcam1+CD169+ macrophages are EBI macrophages. Instead, we discovered that EBI macrophages are characterized by the expression of Epor in both mouse and man. RNA-seq analyses of the newly identified EBI macrophages revealed that EBI macrophages have involved specialized function in supporting erythropoiesis. Our findings provide foundation for future studies. Here we will review current knowledge of EBI macrophages and discuss future perspectives.

20.
Neurotherapeutics ; 16(2): 404-415, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30361931

RESUMO

Reduced estradiol levels are associated with depression in women during the transition to and after menopause. A considerable number of studies focusing on the theme of treating depression through the activation of erythropoietin (EPO)-induced signaling pathways have been published. Venlafaxine is an approved antidepressant drug that inhibits both serotonin and norepinephrine transporters. The aim of the present study was to investigate the effects of venlafaxine on the depressive-like behaviors and serum estradiol levels in female rats following ovariectomy (OVX) and the possible roles of EPO-induced signaling pathways. Venlafaxine (10 mg/kg/day) was orally administered to OVX rats over a period of 4 weeks using two different treatment regimens: either starting 24 h or 2 weeks after OVX. Venlafaxine showed a superior efficacy in inducing antidepressant-like effects after an acute treatment (24 h post-OVX) than after the delayed treatment (2 weeks post-OVX) and was characterized by a decreased immobility time in the forced swimming test. In parallel, venlafaxine induced EPO and EPO receptor mRNA expression and increased levels of phospho-Janus kinase 2 (p-JAK2), phospho-signal transducer and activator of transcription 5, and phospho-extracellular signal-regulated kinase 1/2 in the hippocampus of OVX rats. Meanwhile, rats exhibited a marked reduction in the hippocampal Bax/Bcl2 ratio, caspase-3 activity, and tumor necrosis factor alpha levels after venlafaxine treatment. Venlafaxine also increased the hippocampal brain-derived neurotrophic factor and serum estradiol levels. Based on these findings, venlafaxine exerts a neuroprotective effect on OVX rats that is at least partially attributed to the activation of EPO/EPOR/JAK2 signaling pathways, anti-apoptotic activities, anti-inflammatory activities, and neurotrophic activities, as well as an increase in serum estradiol level. Graphical Abstract ᅟ.


Assuntos
Antidepressivos de Segunda Geração/farmacologia , Comportamento Animal/efeitos dos fármacos , Depressão/metabolismo , Estradiol/sangue , Transdução de Sinais/efeitos dos fármacos , Cloridrato de Venlafaxina/farmacologia , Animais , Eritropoetina/metabolismo , Feminino , Janus Quinase 2/metabolismo , Atividade Motora/efeitos dos fármacos , Ovariectomia , Ratos , Ratos Wistar , Receptores da Eritropoetina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA