Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Asian Nat Prod Res ; : 1-9, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38860546

RESUMEN

Pegmolesatide, a synthetic, polyethylene-glycolylated, peptide-based erythropoiesis-stimulating agent (ESA), has been recently approved in China. Pegmolesatide is derived from the structure of endogenous erythropoietin (EPO), a natural product in mammals. This study compared the in vitro effects and selectivity of pegmolesatide to those of recombinant EPO and carbamylated EPO (CEPO) through computer-aided analyses and biological tests. The findings indicate that pegmolesatide exhibited the same stimulating effect on erythropoiesis as EPO with fewer side effects than EPO and CEPO.

2.
Protein Eng Des Sel ; 342021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-34725710

RESUMEN

Erythropoietin enhances oxygen delivery and reduces hypoxia-induced cell death, but its pro-thrombotic activity is problematic for use of erythropoietin in treating hypoxia. We constructed a fusion protein that stimulates red blood cell production and neuroprotection without triggering platelet production, a marker for thrombosis. The protein consists of an anti-glycophorin A nanobody and an erythropoietin mutant (L108A). The mutation reduces activation of erythropoietin receptor homodimers that induce erythropoiesis and thrombosis, but maintains the tissue-protective signaling. The binding of the nanobody element to glycophorin A rescues homodimeric erythropoietin receptor activation on red blood cell precursors. In a cell proliferation assay, the fusion protein is active at 10-14 M, allowing an estimate of the number of receptor-ligand complexes needed for signaling. This fusion protein stimulates erythroid cell proliferation in vitro and in mice, and shows neuroprotective activity in vitro. Our erythropoietin fusion protein presents a novel molecule for treating hypoxia.


Asunto(s)
Eritropoyetina , Animales , Eritropoyesis , Eritropoyetina/genética , Eritropoyetina/metabolismo , Hipoxia , Ratones , Unión Proteica , Receptores de Eritropoyetina/genética , Receptores de Eritropoyetina/metabolismo
3.
Int J Mol Sci ; 23(1)2021 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-35008482

RESUMEN

The two erythropoietin (EPO) receptor forms mediate different cellular responses to erythropoietin. While hematopoiesis is mediated via the homodimeric EPO receptor (EPOR), tissue protection is conferred via a heteromer composed of EPOR and CD131. In the skeletal system, EPO stimulates osteoclast precursors and induces bone loss. However, the underlying molecular mechanisms are still elusive. Here, we evaluated the role of the heteromeric complex in bone metabolism in vivo and in vitro by using Cibinetide (CIB), a non-erythropoietic EPO analogue that exclusively binds the heteromeric receptor. CIB is administered either alone or in combination with EPO. One month of CIB treatment significantly increased the cortical (~5.8%) and trabecular (~5.2%) bone mineral density in C57BL/6J WT female mice. Similarly, administration of CIB for five consecutive days to female mice that concurrently received EPO on days one and four, reduced the number of osteoclast progenitors, defined by flow cytometry as Lin-CD11b-Ly6Chi CD115+, by 42.8% compared to treatment with EPO alone. In addition, CIB alone or in combination with EPO inhibited osteoclastogenesis in vitro. Our findings introduce CIB either as a stand-alone treatment, or in combination with EPO, as an appealing candidate for the treatment of the bone loss that accompanies EPO treatment.


Asunto(s)
Densidad Ósea/efectos de los fármacos , Eritropoyetina/metabolismo , Oligopéptidos/farmacología , Osteogénesis/efectos de los fármacos , Animales , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Femenino , Hematopoyesis/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo
4.
Physiol Rep ; 6(12): e13751, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29939494

RESUMEN

Clinically, erythropoietin (EPO) is known to increase systemic vascular resistance and arterial blood pressure. However, EPO stimulates the production of the potent vasodilator, nitric oxide (NO), in culture endothelial cells. The mechanism by which EPO causes vasoconstriction despite stimulating NO production may be dependent on its ability to activate two receptor complexes, the homodimeric EPO (EPOR2 ) and the heterodimeric EPOR/ß-common receptor (ßCR). The purpose of this study was to investigate the contribution of each receptor to the vasoactive properties of EPO. First-order, mesenteric arteries were isolated from 16-week-old male C57BL/6 mice, and arterial function was studied in pressure arteriographs. To determine the contribution of each receptor complex, EPO-stimulating peptide (ESP), which binds and activates the heterodimeric EPOR/ßCR complex, and EPO, which activates both receptors, were added to the arteriograph chamber 20 min prior to evaluation of endothelium-dependent (acetylcholine, bradykinin, A23187) and endothelium-independent (sodium nitroprusside) vasodilator responses. Only ACh-induced vasodilation was impaired in arteries pretreated with EPO or ESP. EPO and ESP pretreatment abolished ACh-induced vasodilation by 100% and 60%, respectively. EPO and ESP did not affect endothelium-independent vasodilation by SNP. Additionally, a novel ßCR inhibitory peptide (ßIP), which was computationally developed, prevented the impairment of acetylcholine-induced vasodilation by EPO and ESP, further implicating the EPOR/ßCR complex. Last, pretreatment with either EPO or ESP did not affect vasoconstriction by phenylephrine and KCl. Taken together, these findings suggest that acute activation of the heterodimeric EPOR/ßCR in endothelial cells leads to a selective impairment of ACh-mediated vasodilator response in mouse mesenteric resistance arteries.


Asunto(s)
Acetilcolina/antagonistas & inhibidores , Subunidad beta Común de los Receptores de Citocinas/efectos de los fármacos , Eritropoyetina/farmacología , Arterias Mesentéricas/fisiología , Receptores de Eritropoyetina/efectos de los fármacos , Vasodilatación/fisiología , Acetilcolina/farmacología , Animales , Arteriolas/fisiología , Subunidad beta Común de los Receptores de Citocinas/fisiología , Endotelio Vascular/fisiología , Masculino , Arterias Mesentéricas/efectos de los fármacos , Ratones Endogámicos C57BL , Nitroprusiato/farmacología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Receptores de Eritropoyetina/fisiología , Proteínas Recombinantes/farmacología , Vasodilatación/efectos de los fármacos , Vasodilatadores/antagonistas & inhibidores , Vasodilatadores/farmacología
5.
Peptides ; 104: 1-6, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29635062

RESUMEN

In short-term animal models of ischemia, erythropoietin (EPO) signaling through the heterodimeric EPO receptor (EPOR)/ß-common receptor (ßCR) is believed to elicit tissue protective effects. However, large, randomized, controlled trials demonstrate that targeting a higher hemoglobin level by administering higher doses of EPO, which are more likely to activate the heterodimeric EPOR/ßCR, is associated with an increase in adverse cardiovascular events. Thus, inhibition of long-term activation of the ßCR may have therapeutic implications. This study aimed to design and evaluate the efficacy of novel computationally designed ßCR inhibitory peptides (ßIP). These novel ßIPs were designed based on a truncated portion of Helix-A from EPO, specifically residues 11-26 (VLERYLLEAKEAEKIT). Seven novel peptides (P1 to P7) were designed. Peptide 7 (P7), VLERYLHEAKHAEKIT, demonstrated the most robust inhibitory activity. We also report here the ability of P7 to inhibit ßCR-induced nitric oxide (NO) production and angiogenesis in human umbilical vein endothelial cells (HUVECs). Specifically, we found that P7 ßIP completely abolished EPO-induced NO production. The inhibitory effect could be overcome with super physiological doses of EPO, suggesting a competitive inhibition. ßCR-induced angiogenesis in HUVEC's was also abolished with treatment of P7 ßIP, but P7 ßIP did not inhibit vascular endothelial growth factor (VEGF)-induced angiogenesis. In addition, we demonstrate that the novel P7 ßIP does not inhibit EPO-induced erythropoiesis with use of peripheral blood mononuclear cells (PBMCs). These results, for the first time, describe a novel, potent ßCR peptide inhibitor that inhibit the actions of the ßCR without affecting erythropoiesis.


Asunto(s)
Subunidad beta Común de los Receptores de Citocinas/metabolismo , Eritropoyetina/farmacología , Células Madre de Sangre Periférica/metabolismo , Transducción de Señal/fisiología , Secuencia de Aminoácidos , Células Cultivadas , Biología Computacional , Células Endoteliales de la Vena Umbilical Humana , Humanos , Datos de Secuencia Molecular , Óxido Nítrico/metabolismo , Células Madre de Sangre Periférica/efectos de los fármacos , Estructura Secundaria de Proteína , Transducción de Señal/genética
6.
Mol Neurobiol ; 54(7): 5051-5060, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-27541284

RESUMEN

Carbamylated erythropoietin (CEPO), an EPO derivative, is attracting widespread interest due to neuroprotective effects without erythropoiesis. However, little is known about molecular mechanisms behind CEPO-mediated neuroprotection. In primary neurons with oxygen-glucose deprivation (OGD) and mice with hypoxia-reoxygenation, the neuroprotection and possible molecular mechanism of CEPO were performed by immunohistochemistry and immunocytochemistry, Western blot, RT-PCR, and ELISA. The comparisons were analyzed by ANOVA followed by unpaired two-tailed Student's t test. Both CEPO and EPO showed the neuroprotective effects in OGD model and hypoxic brain. CEPO did not trigger JAK-2 but activated AKT through glial cell line-derived neurotrophic factor (GDNF). It has been shown that CEPO acts upon a heteroreceptor complex comprising both the EPO receptor and the common ß receptor subunit (ßcR, also known as CD131). The blockage of CD131 reduced CEPO-mediated GDNF production, while GFR receptor blockage and GDNF neutralization inhibited CEPO-induced neurogenesis. Addition of GDNF to cultured neurons increased phosphorylation of AKT. CEPO protects neurons possible through the CD131/GDNF/AKT pathway.


Asunto(s)
Hipoxia/tratamiento farmacológico , Neuronas/metabolismo , Transducción de Señal , Animales , Subunidad beta Común de los Receptores de Citocinas/metabolismo , Eritropoyetina/análogos & derivados , Eritropoyetina/farmacología , Femenino , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Hipoxia/metabolismo , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Neuroprotección/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos
7.
Pain Rep ; 1(1): e566, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29392190

RESUMEN

The innate repair receptor (IRR) is a heteromer of the erythropoietin receptor and the ß-common (CD131) receptor, which simultaneously activates anti-inflammatory and tissue repair pathways. Experimental data suggest that after peripheral nerve injury, the IRR is upregulated in the spinal cord and modulates the neurogenic inflammatory response. The recently introduced selective IRR agonist ARA290 is an 11-amino acid peptide initially tested in animal models of neuropathy. After sciatic nerve injury, ARA290 produced a rapid and long-term relief of mechanical and cold allodynia in normal mice, but not in animals with a ß-common receptor knockout phenotype. In humans, ARA290 has been evaluated in patients with small fiber neuropathy associated with sarcoidosis or type 2 diabetes (T2D) mellitus. In patients with sarcoidosis, ARA290 significantly improved neuropathic and autonomic symptoms, as well as quality of life as assessed by the small fiber neuropathy screening list questionnaire. In addition, ARA290 treatment for 28 days initiated a regrowth of small nerve fibers in the cornea, but not in the epidermis. In patients with T2D, the results were similar to those observed in patients with sarcoidosis along with an improved metabolic profile. In both populations, ARA290 lacked significant adverse effects. These experimental and clinical studies show that ARA290 effectively reprograms a proinflammatory, tissue-damaging milieu into one of healing and tissue repair. Further clinical trials with long-term treatment and follow-up are needed to assess the full potential of IRR activation by ARA290 as a disease-modifying therapy in neuropathy of various etiologies.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA