Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Reprod Biol Endocrinol ; 19(1): 96, 2021 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-34176510

RESUMEN

BACKGROUND: Successful human embryo implantation requires the differentiation of endometrial stromal cells (ESCs) into decidual cells during a process called decidualization. ESCs express specific markers of decidualization, including prolactin, insulin-like growth factor-binding protein-1 (IGFBP-1), and connexin-43. Decidual cells also control of trophoblast invasion by secreting various factors, such as matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases. Preimplantation factor (PIF) is a recently identified, embryo-derived peptide with activities at the fetal-maternal interface. It creates a favorable pro-inflammatory environment in human endometrium and directly controls placental development by increasing the human trophoblastic cells' ability to invade the endometrium. We hypothesized that PIF's effects on the endometrium counteract its pro-invasive effects. METHODS: We tested sPIF effect on the expression of three decidualization markers by RT-qPCR and/or immunochemiluminescence assay. We examined sPIF effect on human ESC migration by performing an in vitro wound healing assay. We analyzed sPIF effect on endometrial control of human trophoblast invasion by performing a zymography and an invasion assay. RESULTS: Firstly, we found that a synthetic analog of PIF (sPIF) significantly upregulates the mRNA expression of IGFBP-1 and connexin-43, and prolactin secretion in ESCs - suggesting a pro-differentiation effect. Secondly, we showed that the HTR-8/SVneo trophoblastic cell line's invasive ability was low in the presence of conditioned media from ESCs cultured with sPIF. Thirdly, this PIF's anti-invasive action was associated with a specifically decrease in MMP-9 activity. CONCLUSION: Taken as a whole, our results suggest that PIF accentuates the decidualization process and the production of endometrial factors that limit trophoblast invasion. By controlling both trophoblast and endometrial cells, PIF therefore appears to be a pivotal player in the human embryo implantation process.


Asunto(s)
Decidua/citología , Decidua/efectos de los fármacos , Endometrio/citología , Endometrio/efectos de los fármacos , Proteínas Gestacionales/administración & dosificación , Trofoblastos/efectos de los fármacos , Adulto , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Células Cultivadas , Decidua/fisiología , Endometrio/fisiología , Femenino , Humanos , Células del Estroma/efectos de los fármacos , Células del Estroma/fisiología , Trofoblastos/fisiología
2.
Animal ; 13(11): 2569-2575, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-30968808

RESUMEN

Expression of estrus (EST) near the time of fixed-time artificial insemination (FTAI) increases pregnancy success in beef females. This outcome has been associated with improved pregnancy establishment and maintenance, although research is still warranted to validate this theory. Hence, this experiment compared ovarian, uterine and conceptus factors associated with pregnancy establishment in Bos indicus beef cows according to estrous expression during a FTAI protocol. One hundred lactating multiparous Nelore cows received a 2 mg injection of estradiol benzoate and an intravaginal progesterone (P4) releasing device on day -11, a 12.5 mg injection of prostaglandin F2α on day -4, P4 device removal in addition to 0.6 mg injection of estradiol cypionate and 300 IU injection of equine chorionic gonadotropin on day -2, and FTAI on day 0. An estrous detection patch was attached to the tailhead of each cow on day -2, and estrous expression was defined as removal of >50% of the rub-off coating from the patch at FTAI. Overall, 39 cows expressed EST, 55 did not express EST (NOEST), and six cows lost their patch and were discarded from the experiment. Ovarian ultrasonography was performed at FTAI, and on days 7 and 15 of the experiment. Blood samples were also collected on days 7 and 15. Only cows without a corpus luteum (CL) on day 0, and with a CL on days 7 and 15 remained in the experiment (EST, n=36; NOEST, n=48). On day 15, cows were randomly selected within each group (EST, n=29; NOEST, n=30) for conceptus collection via transcervical flushing, followed by endometrial biopsy in the uterine horn ipsilateral to the CL. Within cows not assigned to conceptus collection, blood samples were collected for whole blood RNA extraction (day 20) and pregnancy status was verified by transrectal ultrasonography (day 30). Diameter of dominant follicle on day 0 and plasma P4 concentrations on day 7 were greater (P⩽0.02) in EST v. NOEST cows. Conceptus length and messenger RNA (mRNA) expression of prostaglandin E synthase and interferon-tau were greater (P⩽0.04) in EST v. NOEST cows. Moreover, EST cows diagnosed as pregnant on day 30 had greater (P<0.01) blood mRNA expression of myxovirus resistance 2 on day 20 compared with NOEST. In summary, estrous expression near the time of FTAI enhanced pregnancy establishment factors in B. indicus cows, including conceptus development and mRNA expression of interferon-tau.


Asunto(s)
Bovinos/fisiología , Estro/fisiología , Inseminación Artificial/veterinaria , Interferón Tipo I/administración & dosificación , Proteínas Gestacionales/administración & dosificación , ARN Mensajero/metabolismo , Animales , Gonadotropina Coriónica/administración & dosificación , Cuerpo Lúteo , Dinoprost/administración & dosificación , Estradiol/administración & dosificación , Estradiol/análogos & derivados , Sincronización del Estro , Femenino , Interferón Tipo I/genética , Lactancia , Folículo Ovárico/metabolismo , Ovario/metabolismo , Embarazo , Proteínas Gestacionales/genética , Índice de Embarazo , Progesterona/administración & dosificación , Progesterona/sangre
3.
Pol J Vet Sci ; 22(1): 75-82, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30997764

RESUMEN

In the current study, twenty lambs, aged 4 months, half male and half female, were classified into four groups, with five in each group. The experimental three groups of lambs were given intravenous (IV), intramuscular (IM) and subcutaneous (SC) administrations of recombinant ovine interferon-τ (roIFN-τ). The fourth group (normal control) of lambs was given normal saline injections in the same way. After administrations, blood samples were collected from the tested animals at different time points post injection, and the serum titers of roIFN-τ were measured using cytopathic effect (CPE) inhibition bioassay. The results of calculating pharmacokinetic (PK) parameters using DAS software showed that the PK characteristics of roIFN-τ through IV injection conformed to the two-compartment open model, whose half-life of distribution phases (T1/2α) was 0.33±0.034 h and the elimination half-life(T1/2ß) was 5.01±0.24 h. However, the PK features of IM injection and SC injection of roIFN-τ conformed to the one compartment open model, whose Tmax were 3.11±0.26 h and 4.83±0.43 h, respectively, together with an elimination half life(T1/2ß) of 9.11±0.76 h and 7. 43±0.58 h, and an absorption half-life (T1/2k(a)) of 1.13±0.31 h and 1.85±0.40 h, respectively. The bioavailability of roIFN-τ after IM administration reaches 73.57%, which is greater than that of SC administration (53.43%). These results indicate that the drug administration effect can be preferably obtained following a single dose IM administration of the roIFN-τ aqueous preparation. This study will facilitate the clinical application of roIFN-τ as a potential antiviral agent in future work.


Asunto(s)
Antivirales/farmacocinética , Interferón Tipo I/farmacocinética , Proteínas Gestacionales/farmacocinética , Ovinos/sangre , Animales , Antivirales/sangre , Área Bajo la Curva , Femenino , Semivida , Inyecciones Intravenosas , Interferón Tipo I/administración & dosificación , Masculino , Proteínas Gestacionales/administración & dosificación , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacocinética , Ovinos/metabolismo
4.
Drug Des Devel Ther ; 12: 1977-1983, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30013317

RESUMEN

INTRODUCTION: Human placental protein 13 (PP13) is a galectin predominantly expressed by the placenta. Low serum concentrations of PP13 in early pregnancy indicate a higher risk of developing preeclampsia. METHODS: The pharmacokinetic disposition and bioavailability of PP13 were determined by single intravenous and subcutaneous administration to 12 healthy New Zealand White rabbits. The serum pharmacokinetic values were determined by enzyme-linked immunosorbent assay, and are best described by a two-compartment model. RESULTS: Both volume of distribution and the area under the curve were dose dependent for the intravenous group (p<0.01). PP13 elimination half-life was also found to be different between the groups (p<0.01). The bioavailability of PP13 following subcutaneous administration was found to be 57%. CONCLUSION: This study shows that the concentration of total PP13 released into the maternal circulation during pregnancy might be much higher than previously estimated.


Asunto(s)
Galectinas/administración & dosificación , Galectinas/farmacocinética , Proteínas Gestacionales/administración & dosificación , Proteínas Gestacionales/farmacocinética , Administración Intravenosa , Animales , Relación Dosis-Respuesta a Droga , Femenino , Galectinas/sangre , Inyecciones Subcutáneas , Proteínas Gestacionales/sangre , Conejos , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/sangre , Proteínas Recombinantes/farmacocinética
5.
Biomed Res Int ; 2017: 2350482, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28331850

RESUMEN

The aim of the present study was to determine the anti-inflammatory effect of IFN-τ on endometritis using a mouse model of S. aureus-induced endometritis and to elucidate the mechanism of action underlying these effects. In the present study, the effect of IFN-τ on S. aureus growth was monitored by turbidimeter at 600 nm. IFN-τ did not affect S. aureus growth. The histopathological changes indicated that IFN-τ had a protective effect on uterus tissues with S. aureus infection. The ELISA and qPCR results showed the production of the proinflammatory cytokines TNF-α, IL-1ß, and IL-6 was decreased with IFN-τ treatment. In contrast, the level of the anti-inflammatory cytokine IL-10 was increased. We further studied the signaling pathway associated with these observations, and the qPCR results showed that the expression of TLR2 was repressed by IFN-τ. Furthermore, the western blotting results showed the phosphorylation of IκB, NF-κB p65, and MAPKs (p38, JNK, and ERK) was inhibited by IFN-τ treatment. The results suggested that IFN-τ may be a potential drug for the treatment of uterine infection due to S. aureus or other infectious inflammatory diseases.


Asunto(s)
Endometritis/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Interferón Tipo I/administración & dosificación , Proteínas Gestacionales/administración & dosificación , Infecciones Estafilocócicas/tratamiento farmacológico , Animales , Antiinflamatorios/administración & dosificación , Modelos Animales de Enfermedad , Endometritis/microbiología , Endometritis/patología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inflamación/microbiología , Inflamación/patología , Interferón Tipo I/metabolismo , Ratones , FN-kappa B/biosíntesis , Proteínas Gestacionales/metabolismo , Infecciones Estafilocócicas/genética , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/patogenicidad , Receptor Toll-Like 2/biosíntesis , Factor de Transcripción ReIA/biosíntesis , Útero/efectos de los fármacos , Útero/microbiología , Útero/patología
6.
Theriogenology ; 92: 111-120, 2017 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-28237325

RESUMEN

In cows, lysophosphatidic acid (LPA), which acts in an auto/paracrine manner, serves as a luteotropic factor during early pregnancy by stimulating progesterone and prostaglandin E2 secretion, thus protecting the bovine corpus luteum and early embryo development. Our hypothesis was that LPA exerted some local effects on the bovine endometrium prior to early embryo-maternal interactions and that interferon tau (IFNτ), the pregnancy recognition signal, modulated this action. In the present study, we applied an in vitro model involving whole-transcriptomic profiling to examine the effects of LPA on gene expression in bovine endometrial cells. Microarray analyses revealed 36, 269 and 284 differentially expressed transcripts in bovine endometrial cells in the control vs. LPA, control vs. LPA + IFNτ and LPA vs. LPA + IFNτ groups, respectively. The expression of matrix metalloproteinase 13 (MMP13) and radical S-adenosyl methionine domain containing 2 (RSAD2) was increased in the LPA-treated endometrial cells. Among the transcripts differentially regulated by LPA together with IFNτ, many of the genes were classical- or novel-type I IFN-stimulated genes (ISGs). The results indicated that 10 of the 16 analyzed genes showed a positive correlation with their corresponding microarray data upon real-time PCR validation, indicating a considerable consistency between both techniques. In summary, these transcriptional profiling studies identified a number of genes that were regulated by LPA alone and LPA together with IFNτ in endometrial cells from the bovine uterus. Available studies support the idea that LPA, which acts in an auto/paracrine manner on the endometrium, alters the expression of genes that are probably important for uterine receptivity, maternal immune tolerance to the embryo and conceptus growth and development during early pregnancy. Moreover, the differentially expressed genes (DEGs) that increased in the LPA + IFNτ-treated endometrial cells are largely in response to IFNτ actions and are possibly associated with crucial biological processes during the peri-implantation period of pregnancy.


Asunto(s)
Bovinos/fisiología , Endometrio/citología , Regulación de la Expresión Génica/efectos de los fármacos , Interferón Tipo I/farmacología , Lisofosfolípidos/farmacología , Proteínas Gestacionales/farmacología , Transcriptoma/fisiología , Animales , Femenino , Interferón Tipo I/administración & dosificación , Lisofosfolípidos/administración & dosificación , Proteínas Gestacionales/administración & dosificación
7.
Antiviral Res ; 143: 134-141, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28137623

RESUMEN

Foot-and-mouth disease (FMD) is an economically important disease in most parts of the world and new therapeutic agents are needed to protect the animals before vaccination can trigger the host immune response. Although several interferons have been used for their antiviral activities against Foot-and-mouth disease virus (FMDV), ovine interferon tau 4 (OvIFN-τ4), with a broad-spectrum of action, cross-species antiviral activity, and lower incidence of toxicity in comparison to other type І interferons, has not yet been evaluated for this indication. This is the first study to evaluate the antiviral activity of OvIFN-τ4 against various strains of FMDV. The effective anti-cytopathic concentration of OvIFN-τ4 and its effectiveness pre- and post-infection with FMDV were tested in vitro in LFBK cells. In vivo activity of OvIFN-τ4 was then confirmed in a mouse model of infection. OvIFN-τ4 at a concentration of 500 ng, protected mice until 5days post-FMDV challenge and provided 90% protection for 10 days following FMDV challenge. These results suggest that OvIFN-τ4 could be used as an alternative to other interferons or antiviral agents at the time of FMD outbreak.


Asunto(s)
Antivirales/farmacología , Virus de la Fiebre Aftosa/efectos de los fármacos , Fiebre Aftosa/prevención & control , Interferón Tipo I/farmacología , Proteínas Gestacionales/farmacología , Animales , Línea Celular , Clonación Molecular , Virus de la Fiebre Aftosa/patogenicidad , Expresión Génica , Interferón Tipo I/administración & dosificación , Interferón Tipo I/genética , Dosificación Letal Mediana , Ratones , Proteínas Gestacionales/administración & dosificación , Proteínas Gestacionales/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Ovinos , Vacunación
8.
BMC Cardiovasc Disord ; 14: 53, 2014 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-24742302

RESUMEN

BACKGROUND: To comparatively evaluate the cardioprotective activity of placental growth factor (PGF) delivered through direct injection and a nanoparticle-based system respectively and to study the underlying mechanisms in a rat model of acute myocardial infarction (AMI). METHODS: Poly lactic-co-glycolic acid (PLGA)-based PGF-carrying nanoparticles (PGF-PLGANPs) were created. The mean size and morphology of particles were analyzed with particle size analyzer and transmission electronic microscopy (TEM). Encapsulation efficiency and sustained-release dose curve were analyzed by ELISA. Sprague-Dawley rats were randomized into four groups (n = 10). While animals in the first group were left untreated as controls, those in the other 3 groups underwent surgical induction of AMI, followed by treatment with physiological saline, PGF, and PGF-PLGANPs, respectively. Cardiac function was evaluated by transthoracic echocardiography at 4 weeks after treatment. At 6 weeks, rats were sacrificed, infarction size was analyzed with Masson trichrome staining, and protein contents of TIMP-2, MT1-MMP and MMP-2 at the infarction border were determined by immunohistochemistry and western blotting analysis. RESULTS: PGF was released for at least 15 days, showing successful preparation of PGF-PLGANPs. Coronary artery ligation successfully induced AMI. Compared to physiological saline control, PGF, injected to the myocardium either as a nude molecule or in a form of nanoparticles, significantly reduced infarction size, improved cardiac function, and elevated myocardial expression of TIMP-2, MT1-MMP, and MMP-2 (P < 0.05). The effect of PGF-PLGANPs was more pronounced than that of non-encapsulated PGF (P < 0.05). CONCLUSION: Target PGF delivery to myocardium may improve cardiac function after AMI in rats. PLGA-based nanoparticles appear to be a better approach to delivery PGF. PGF exerts its cardioprotective effect at least partially through regulating metalloproteinase-mediated myocardial tissue remodeling.


Asunto(s)
Cardiotónicos/administración & dosificación , Portadores de Fármacos , Ácido Láctico/química , Infarto del Miocardio/tratamiento farmacológico , Nanopartículas , Ácido Poliglicólico/química , Proteínas Gestacionales/administración & dosificación , Animales , Cardiotónicos/química , Química Farmacéutica , Preparaciones de Acción Retardada , Modelos Animales de Enfermedad , Humanos , Inyecciones Intralesiones , Metaloproteinasa 14 de la Matriz/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocardio/metabolismo , Miocardio/patología , Factor de Crecimiento Placentario , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Proteínas Gestacionales/química , Ratas Sprague-Dawley , Proteínas Recombinantes/administración & dosificación , Solubilidad , Volumen Sistólico/efectos de los fármacos , Factores de Tiempo , Inhibidor Tisular de Metaloproteinasa-2/metabolismo , Función Ventricular Izquierda/efectos de los fármacos , Remodelación Ventricular/efectos de los fármacos
9.
PLoS One ; 8(12): e80128, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24324591

RESUMEN

Multiple sclerosis (MS) is a complex multifactorial disease of the central nervous system (CNS) for which animal models have mainly addressed downstream immunopathology but not potential inducers of autoimmunity. In the absence of a pathogen known to cause neuroinflammation in MS, Mycobacterial lysate is commonly used in the form of complete Freund's adjuvant to induce autoimmunity to myelin proteins in Experimental Allergic Encephalomyelitis (EAE), an animal model for MS. The present study demonstrates that a protein from the human endogenous retrovirus HERV-W family (MSRV-Env) can be used instead of mycobacterial lysate to induce autoimmunity and EAE in mice injected with MOG, with typical anti-myelin response and CNS lesions normally seen in this model. MSRV-Env was shown to induce proinflammatory response in human macrophage cells through TLR4 activation pathway. The present results demonstrate a similar activation of murine dendritic cells and show the ability of MSRV-Env to trigger EAE in mice. In previous studies, MSRV-Env protein was reproducibly detected in MS brain lesions within microglia and perivascular macrophages. The present results are therefore likely to provide a model for MS, in which the upstream adjuvant triggering neuroinflammation is the one detected in MS active lesions. This model now allows pre-clinical studies with therapeutic agents targeting this endogenous retroviral protein in MS.


Asunto(s)
Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/patología , Productos del Gen env/administración & dosificación , Inmunidad Innata/efectos de los fármacos , Ratones , Proteínas Gestacionales/administración & dosificación , Adyuvantes Inmunológicos/administración & dosificación , Animales , Células Cultivadas , Sistema Nervioso Central , Células Dendríticas , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Expresión Génica , Productos del Gen env/inmunología , Humanos , Receptores de Lipopolisacáridos/genética , Receptores de Lipopolisacáridos/inmunología , Ratones Endogámicos C57BL , Ratones Noqueados , Esclerosis Múltiple/genética , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/patología , Glicoproteína Mielina-Oligodendrócito/administración & dosificación , Fragmentos de Péptidos/administración & dosificación , Proteínas Gestacionales/inmunología , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/inmunología , Receptor Toll-Like 4/deficiencia , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología
10.
Biofactors ; 39(5): 552-63, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23804503

RESUMEN

Male Zucker diabetic fatty (ZDF) rats were used to study effects of oral administration of interferon tau (IFNT) in reducing obesity. Eighteen ZDF rats (28 days of age) were assigned randomly to receive 0, 4, or 8 µg IFNT/kg body weight (BW) per day (n = 6/group) for 8 weeks. Water consumption was measured every two days. Food intake and BW were recorded weekly. Energy expenditure in 4-, 6-, 8-, and 10-week-old rats was determined using indirect calorimetry. Starting at 7 weeks of age, urinary glucose, and ketone bodies were tested daily. Rates of glucose and oleate oxidation in liver, brown adipose tissue, and abdominal adipose tissue, as well as leucine catabolism in skeletal muscle, and lipolysis in white and brown adipose tissues were greater for rats treated with 8 µg IFNT/kg BW/day in comparison with control rats. Treatment with 8 µg IFNT/kg BW/day increased heat production, reduced BW gain and adiposity, ameliorated fatty liver syndrome, delayed the onset of diabetes, and decreased concentrations of glucose, free fatty acids, triacylglycerol, cholesterol, and branched-chain amino acids in plasma, compared with control rats. Oral administration of 8 µg IFNT/kg BW/day ameliorated oxidative stress in skeletal muscle, liver, and adipose tissue, as indicated by decreased ratios of oxidized glutathione to reduced glutathione and increased concentrations of tetrahydrobiopterin. These results indicate that IFNT stimulates oxidation of energy substrates and reduces obesity in ZDF rats and may have broad important implications for preventing and treating obesity-related diseases in mammals.


Asunto(s)
Adiposidad/efectos de los fármacos , Fármacos Antiobesidad/administración & dosificación , Diabetes Mellitus Tipo 2/prevención & control , Interferón Tipo I/administración & dosificación , Obesidad/tratamiento farmacológico , Proteínas Gestacionales/administración & dosificación , Adiponectina/sangre , Tejido Adiposo Blanco/efectos de los fármacos , Tejido Adiposo Blanco/patología , Administración Oral , Aminoácidos/sangre , Animales , Glucemia , Diabetes Mellitus Tipo 2/sangre , Evaluación Preclínica de Medicamentos , Metabolismo Energético/efectos de los fármacos , Glicerol/metabolismo , Insulina/sangre , Leptina/sangre , Leucina Transaminasa/metabolismo , Lípidos/sangre , Hígado/efectos de los fármacos , Hígado/patología , Masculino , Obesidad/sangre , Obesidad/patología , Tamaño de los Órganos , Oxidación-Reducción , Estrés Oxidativo , Ratas , Ratas Zucker
11.
Fetal Diagn Ther ; 33(4): 257-64, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23406577

RESUMEN

OBJECTIVES: Here, we performed a pathophysiological examination of the vascular function of rodent in the presence of placental protein 13 (PP13) and its implication to regulate the development of preeclampsia. METHODS: Single i.v. injection and prolonged in vivo exposure to PP13 via osmotic pumps were performed in gravid and non-gravid rats to examine the influence of PP13 on blood pressure and heart rate in animals. The effect of PP13 was also examined in isolated uterine and mesenteric arteries, along with the examination of placental blood supply. RESULTS: Human PP13 has a major impact on the maternal cardiovascular system of rodents by reducing blood pressure, either at single or prolonged exposure, and causing significant vasodilatation in isolated arteries. Prolonged exposure was followed by increased elaboration and angiogenesis of the uteroplacental arteries supplying the placenta. CONCLUSION: This is the first study describing effects of PP13 on vasodilatation and uterine artery remodeling. The results imply that PP13 may have a physiological role in improving uteroplacental blood flow. The findings of this study make it tempting to speculate that keeping PP13 levels within a certain 'therapeutic window' during pregnancy may facilitate proper adaptation of the maternal vasculature to pregnancy.


Asunto(s)
Galectinas/metabolismo , Arterias Mesentéricas/metabolismo , Placenta/irrigación sanguínea , Proteínas Gestacionales/metabolismo , Arteria Uterina/metabolismo , Útero/irrigación sanguínea , Administración Intravenosa , Animales , Presión Sanguínea/efectos de los fármacos , Femenino , Galectinas/administración & dosificación , Galectinas/genética , Galectinas/farmacología , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Técnicas In Vitro , Infusiones Subcutáneas , Arterias Mesentéricas/efectos de los fármacos , Placenta/efectos de los fármacos , Placenta/metabolismo , Circulación Placentaria/efectos de los fármacos , Preeclampsia/sangre , Preeclampsia/metabolismo , Preeclampsia/prevención & control , Embarazo , Proteínas Gestacionales/administración & dosificación , Proteínas Gestacionales/genética , Proteínas Gestacionales/farmacología , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/sangre , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Arteria Uterina/efectos de los fármacos , Útero/efectos de los fármacos , Útero/metabolismo , Vasodilatación/efectos de los fármacos
12.
Reprod Biol Endocrinol ; 10: 21, 2012 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-22439976

RESUMEN

BACKGROUND: Interferon tau (IFNT), which is secreted into the uterine cavity during the maternal recognition period (MRP), is a key factor for establishment of pregnancy. The present study aims to clarify the relationship between the ability of a bovine conceptus to produce IFNT during the MRP and the conceptus's ability to establish pregnancy. METHODS: In the first experiment, IFNT (0, 500, or 1000 micrograms) was administered into the uterine horn ipsilateral to the CL 16 or 17 d after standing estrus, and mRNA levels of IFN-stimulated gene 15-kDa protein (ISG15) and Mx2 in peripheral blood mononuclear cells (PBMCs) were determined. In the second experiment, we investigated ISG15 mRNA expression in PBMCs during the MRP in cattle after either artificial insemination (AI) or embryo transfer (ET). RESULTS: Intrauterine administration of IFNT stimulated ISG15 and Mx2 gene expressions in PBMCs in cattle, and there was a positive correlation between the expressions of peripheral markers and the quantity of IFNT administered. In pregnant and normal interestrous interval (< 25 d) cattle (nIEI cattle), expression levels of the ISG15 gene showed similar patterns after AI and ET, and ISG15 mRNA expression was increased in pregnant cattle but unchanged in nIEI cattle. In contrast, ISG15 gene expression in extended interestrous interval (greater than or equal to 25 d) cattle (eIEI cattle) differed after ET compared with AI. In eIEI cattle after ET, ISG15 gene expression increased, such that the value on day 18 was intermediate between those of pregnant and nIEI cattle. In eIEI cattle after AI, ISG15 gene expression did not increase throughout the observation period. CONCLUSIONS: The results of the current study indicate that the quantity of conceptus-derived IFNT can be estimated by measuring ISG15 mRNA levels in PBMCs from cattle. Using this approach, we demonstrate that ISG15 gene expression during the MRP in eIEI cattle differed after ET compared with AI. In addition, the modest increase in ISG15 gene expression in eIEI cattle after ET suggests that late embryo losses were due to delayed or insufficient growth of the conceptus during the MRP in cattle.


Asunto(s)
Bovinos/embriología , Pérdida del Embrión , Expresión Génica/efectos de los fármacos , Interferón Tipo I/administración & dosificación , Interferones/farmacología , Leucocitos Mononucleares/metabolismo , Proteínas Gestacionales/administración & dosificación , Animales , Transferencia de Embrión/veterinaria , Femenino , Inseminación Artificial/veterinaria , Interferón Tipo I/fisiología , Leucocitos Mononucleares/química , Embarazo , Proteínas Gestacionales/fisiología , ARN Mensajero/análisis , Útero/efectos de los fármacos
13.
Res Vet Sci ; 93(1): 381-5, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21652051

RESUMEN

This study tested the effect of recombinant bovine interferon-tau (rboIFN-τ) on the length of estrous cycle, luteal lifespan and side effects of rboIFN-τ in the cow. A normal estrous cycle in six non-lactating cycling Holstein cows was observed (non-treated cycle), and either 2.0 mg of liposomalized rboIFN-τ (treated cycle) or bovine serum albumin (BSA; placebo cycle) was infused in the uterus on day 7 of the estrous cycle (day 0=day of ovulation). Rectal temperature, heart rate and respiratory rate were recorded and blood samples were collected before and after the treatments. The length of the estrous cycle and corpus luteum lifespan in rboIFN-τ treated cycles were not significantly different from those of the non-treated and placebo cycles. In contrast, the rboIFN-τ treatment caused a transient increase in rectal temperature and a decrease in the number of peripheral lymphocytes and neutrophils after the treatment.


Asunto(s)
Ciclo Estral/efectos de los fármacos , Interferón Tipo I/farmacología , Fase Luteínica/efectos de los fármacos , Proteínas Gestacionales/farmacología , Animales , Temperatura Corporal/efectos de los fármacos , Bovinos/sangre , Bovinos/fisiología , Cuerpo Lúteo/efectos de los fármacos , Ciclo Estral/sangre , Femenino , Frecuencia Cardíaca/efectos de los fármacos , Interferón Tipo I/administración & dosificación , Recuento de Leucocitos/veterinaria , Fase Luteínica/sangre , Recuento de Linfocitos/veterinaria , Neutrófilos/efectos de los fármacos , Proteínas Gestacionales/administración & dosificación , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacología , Frecuencia Respiratoria/efectos de los fármacos , Factores de Tiempo
14.
Int J Nanomedicine ; 6: 2667-78, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22114497

RESUMEN

BACKGROUND: Acute myocardial ischemia results in scar formation with ventricular dilatation and eventually heart failure. Placental growth factor (PlGF) is reported to stimulate angiogenesis and improve cardiac function. In this study, it was hypothesized that intramyocardial injection of PlGF contained in nanoparticles can be released at the site of action for an extended time period as a sustained slow-release protective mechanism that accelerates myocardial recovery in a rat model of ischemic cardiomyopathy. METHODS: PlGF-loaded chitosan-alginate nanoparticles were injected into an acute myocardial infarction model in rats (n = 10 per group). Transthoracic echocardiography was performed at different time intervals. Enzyme-linked immunosorbent assay was used to measure the serum cytokines levels at 8 weeks. Hearts were stained with Masson's trichrome for scar area analysis. Immunofluorostaining was performed to evaluate the extent of myocardial angiogenesis at the infarction border. PlGF enzyme-linked immunosorbent assay was used to measure the in vitro release kinetics of PlGF-loaded nanoparticles. RESULTS: At 8 weeks after coronary ligation, hearts that were treated with PlGF-loaded chitosan-alginate nanoparticles had significant increases in left-ventricular function (P < 0.01), vascular density (P < 0.01), and in the serum level of the anti-inflammatory cytokine interleukin-10 (P < 0.05). There was significant decrease in scar area formation (P < 0.05) and in serum levels of the proinflammatory cytokines tumor necrosis factor-alpha and interleukin-6 (P < 0.01). In vitro PlGF-release kinetic studies showed a sustained release of PlGF from the particles over a 120-hour period. CONCLUSION: The use of nanoparticles as a vehicle for PlGF delivery, as opposed to the direct injection of the growth factor after acute myocardial infarction, can provide sustained slow-release PlGF therapy, enhancing the positive effects of the growth factor in the setting of acute myocardial ischemia.


Asunto(s)
Infarto del Miocardio/tratamiento farmacológico , Nanopartículas/administración & dosificación , Proteínas Gestacionales/administración & dosificación , Alginatos/química , Análisis de Varianza , Animales , Quitosano/química , Citocinas/sangre , Electrocardiografía , Femenino , Ácido Glucurónico/química , Ácidos Hexurónicos/química , Inmunohistoquímica , Nanopartículas/química , Neovascularización Fisiológica/efectos de los fármacos , Tamaño de la Partícula , Vehículos Farmacéuticos/administración & dosificación , Vehículos Farmacéuticos/química , Factor de Crecimiento Placentario , Proteínas Gestacionales/farmacocinética , Ratas , Ratas Endogámicas Lew , Volumen Sistólico/efectos de los fármacos , Remodelación Ventricular/efectos de los fármacos
15.
Cardiovasc Res ; 89(2): 410-8, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-20935166

RESUMEN

AIMS: Inadequate capillary growth in pressure-overload hypertrophy impairs myocardial perfusion and substrate delivery, contributing to progression to failure. Capillary growth is tightly regulated by angiogenesis growth factors like vascular endothelial growth factor (VEGF) and endogenous inhibitors such as the splice variant of VEGF receptor-1, sVEGFR-1. We hypothesized that inadequate expression of VEGF and up-regulation of VEGFR-1 and its soluble splice variant, sVEGFR-1, restrict capillary growth in pressure-overload hypertrophy. METHODS AND RESULTS: Neonatal New Zealand White rabbits underwent aortic banding. mRNA (qRT-PCR) and protein levels (immunoblotting) were determined in hypertrophied and control myocardium (7/group) for total VEGF, VEGFR-1, sVEGFR-1, VEGFR-2, and phospho-VEGFR-1 and -R-2. Free VEGF was determined by enzyme-linked immunoassay (ELISA) in hypertrophied myocardium, controls, and hypertrophied hearts following inhibition of sVEGFR-1 with placental growth factor (PlGF). VEGFR-1 and sVEGFR-1 mRNA (seven-fold up-regulation, P = 0.001) and protein levels were significantly up-regulated in hypertrophied hearts vs. controls (VEGFR-1: 44 ± 8 vs. 23 ± 1, P = 0.031; sVEGFR-1: 71 ± 13 vs. 31 ± 3, P = 0.016). There was no change in VEGF and VEGFR-2 mRNA or protein levels in hypertrophied compared with controls hearts. A significant decline in free, unbound VEGF was found in hypertrophied myocardium which was reversed following inhibition of sVEGFR-1 with PlGF, which was accompanied by phosphorylation of VEGFR-1 and VEGFR-2. CONCLUSION: Up-regulation of the soluble VEGFR-1 in pressure-loaded myocardium prevents capillary growth by trapping VEGF. Inhibition of sVEGFR-1 released sufficient VEGF to induce angiogenesis and preserved contractile function. These data suggest sVEGFR-1 as possible therapeutic targets to prevent heart failure.


Asunto(s)
Capilares/metabolismo , Cardiomegalia/metabolismo , Miocardio/metabolismo , Neovascularización Fisiológica , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Envejecimiento , Animales , Animales Recién Nacidos , Aorta/cirugía , Western Blotting , Capilares/efectos de los fármacos , Capilares/fisiopatología , Cardiomegalia/diagnóstico por imagen , Cardiomegalia/genética , Cardiomegalia/fisiopatología , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Ligadura , Neovascularización Fisiológica/efectos de los fármacos , Fosforilación , Factor de Crecimiento Placentario , Proteínas Gestacionales/administración & dosificación , ARN Mensajero/metabolismo , Conejos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ultrasonografía , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
16.
Biol Reprod ; 82(4): 725-35, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20042537

RESUMEN

Interferon tau (IFNT) from the ovine conceptus has paracrine actions on the endometrium that alter release of prostaglandin F(2alpha) (PGF) and protect the corpus luteum (CL). Antiviral activity in uterine vein blood and expression of interferon-stimulated genes (ISGs) in CL is greater in pregnant than in nonpregnant ewes. We hypothesized that IFNT contributes to antiviral activity in uterine vein blood and has endocrine actions on the CL. Preadsorption of IFNT with antiserum against recombinant ovine (ro) IFNT revealed that antiviral activity in uterine vein blood from pregnant ewes was mediated by IFNT. Endocrine actions of IFNT were examined after infusing either roIFNT or bovine serum albumin (BSA; 200 microg/24 h; mini-osmotic pump) into the uterine vein of nonpregnant ewes from Day 10 to Day 11 postestrus. The abundance of ISG15 mRNA and protein was greater in CL (P < 0.05) from ewes receiving 24-h roIFNT infusion compared to that from ewes receiving 24-h BSA infusion. Injection of PGF at 12 h following insertion of mini-osmotic pumps resulted in a decline in serum progesterone concentrations 6 through 12 h later in BSA-infused ewes; however, in roIFNT-infused ewes, a similar decline in progesterone concentrations at 6 h was followed by recovery to control values at 12 h. Ewes then received infusions (200 microg/day) of either roIFNT or BSA for 7 days beginning on Day 10 of the estrous cycle. All BSA-infused ewes returned to estrus by Day 19, whereas 80% of roIFNT-infused ewes maintained luteal-phase concentrations of progesterone through Day 32. In conclusion, IFNT is released from the uterus into the uterine vein and acts through an endocrine mechanism to induce ISGs in the CL and delay luteolysis.


Asunto(s)
Interferón Tipo I/administración & dosificación , Fase Luteínica/efectos de los fármacos , Proteínas Gestacionales/administración & dosificación , Preñez , Ovinos , Útero/irrigación sanguínea , Animales , Antivirales/administración & dosificación , Bovinos , Células Cultivadas , Implantación del Embrión/efectos de los fármacos , Implantación del Embrión/genética , Femenino , Bombas de Infusión Implantables , Infusiones Intravenosas , Fase Luteínica/fisiología , Modelos Biológicos , Ovario/irrigación sanguínea , Ovario/efectos de los fármacos , Embarazo , Factores de Tiempo , Útero/efectos de los fármacos
17.
Circ J ; 73(9): 1674-82, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19602778

RESUMEN

BACKGROUND: Placental growth factor (PlGF), a homolog of vascular endothelial growth factor, is reported to stimulate angiogenesis and arteriogenesis in pathological conditions. It was recently demonstrated that PlGF is rapidly produced in myocardial tissue during acute myocardial infarction (MI). However, the effects of exogenous PlGF administration on the healing process after MI are not fully understood. The purpose of the present study was to examine whether PlGF treatment has therapeutic potential in MI. METHODS AND RESULTS: Recombinant human PlGF (rhPlGF: 10 microg) was administered continuously for 3 days in a mouse model of acute MI. rhPlGF treatment significantly improved survival rate after MI and preserved cardiac function relative to control mice. The numbers of CD31-positive cells and alpha-smooth muscle actin-positive vessels in the infarct area were significantly increased in the rhPlGF group. Endothelial progenitor cells (Flk-1(+)Sca-1(+) cells) were mobilized by rhPlGF into the peripheral circulation. Furthermore, rhPlGF promoted the recruitment of GFP-labeled bone marrow cells to the infarct area, but only a few of those migrating cells differentiated into endothelial cells. CONCLUSIONS: Exogenous PlGF plays an important role in healing processes by improving cardiac function and stimulating angiogenesis following MI. It can be considered as a new therapeutic molecule.


Asunto(s)
Inductores de la Angiogénesis/administración & dosificación , Vasos Coronarios/efectos de los fármacos , Infarto del Miocardio/tratamiento farmacológico , Neovascularización Fisiológica/efectos de los fármacos , Proteínas Gestacionales/administración & dosificación , Actinas/metabolismo , Animales , Antígenos Ly/metabolismo , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Vasos Coronarios/metabolismo , Vasos Coronarios/fisiopatología , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Humanos , Bombas de Infusión Implantables , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Infarto del Miocardio/metabolismo , Infarto del Miocardio/fisiopatología , Factor de Crecimiento Placentario , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Proteínas Recombinantes/administración & dosificación , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Factores de Tiempo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/administración & dosificación , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Función Ventricular Izquierda/efectos de los fármacos , Remodelación Ventricular/efectos de los fármacos
18.
Autoimmunity ; 41(7): 543-53, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18608174

RESUMEN

Interferon-alpha (IFN-alpha) inhibits the development of diabetes in animal models of autoimmune diabetes. However, the mechanism of the action is not fully understood and drug toxicity could limit its potential clinical utility. Interferon-tau (IFN-tau) is another type 1 interferon, which has less toxicity but may have different biologic activity than IFN-alpha. This study explores the effect of IFN-tau on the diabetic process in non-obese diabetic (NOD) mice. IFN-tau by intraperitoneal, subcutaneous, or oral routes of administration decreased the development of spontaneous diabetes in NOD mice. Islet inflammation was decreased 50%. IFN-tau administration to recipient mice prevented the development of passively transferred and cyclophosphamide accelerated diabetes. IFN-tau treatment also decreased anti-islet effector activity of NOD splenic cells. Immunoregulatory activity of splenic cells was augmented by IFN-tau administration as was the number of splenic CD25+CD4+ cells. Concanavalin A (Con A)-induced release of IFN-gamma was decreased in spleen cells from IFN-tau treated mice. In conclusion, IFN-tau inhibits spontaneous autoimmune diabetes and passively transferred diabetes in the NOD mouse. This diabetes sparing activity may be due to an induction of regulatory cells, possibly CD25+CD4+ T cells, which in turn inhibit anti-islet effector cell activity and the development of insulitis and diabetes. Due to the lower drug toxicity, IFN-tau could be a better drug candidate than IFN-alpha for experimental clinical trials.


Asunto(s)
Células Dendríticas/inmunología , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Interferón Tipo I/uso terapéutico , Proteínas Gestacionales/uso terapéutico , Linfocitos T Reguladores/inmunología , Traslado Adoptivo , Animales , Ciclofosfamida/farmacología , Células Dendríticas/metabolismo , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/prevención & control , Modelos Animales de Enfermedad , Femenino , Inmunosupresores/farmacología , Interferón Tipo I/administración & dosificación , Interferón Tipo I/farmacología , Interferón gamma/biosíntesis , Islotes Pancreáticos/inmunología , Ratones , Ratones Endogámicos NOD , Proteínas Gestacionales/administración & dosificación , Proteínas Gestacionales/farmacología , Bazo/efectos de los fármacos , Bazo/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/metabolismo
19.
J Card Surg ; 21(6): 559-64, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17073953

RESUMEN

BACKGROUND: Heart failure occurs predominantly due to coronary artery disease and may be amenable to novel revascularization therapies. This study evaluated the effects of placental growth factor (PlGF), a potent angiogenic agent, in a rat model of ischemic cardiomyopathy. METHODS: Wistar rats underwent high proximal ligation of the left anterior descending coronary artery and direct injection of PlGF (n = 10) or saline as a control (n = 10) into the myocardium bordering the ischemic area. After 2 weeks, the following parameters were evaluated: ventricular function with an aortic flow probe and a pressure/volume conductance catheter, left ventricular (LV) geometry by histology, and angiogenesis by immunofluorescence. RESULTS: PlGF animals had increased angiogenesis compared to controls (22.8 +/- 3.5 vs. 12.4 +/- 3.2 endothelial cells/high-powered field, p < 0.03). PlGF animals had less ventricular cavity dilation (LV diameter 8.4 +/- 0.2 vs. 9.2 +/- 0.2 mm, p < 0.03) and increased border zone wall thickness (1.85 +/- 0.1 vs. 1.38 +/- 0.2 mm, p < 0.03). PlGF animals had improved cardiac function as measured by maximum LV pressure (95.7 +/- 4 vs. 73.7 +/- 2 mmHg, p = 0.001), maximum dP/dt (4206 +/- 362 vs. 2978 +/- 236 mmHg/sec, p = 0.007), and ejection fraction (25.7 +/- 2 vs. 18.6 +/- 1%, p = 0.02). CONCLUSIONS: Intramyocardial delivery of PlGF following a large myocardial infarction enhanced border zone angiogenesis, attenuated adverse ventricular remodeling, and preserved cardiac function. This therapy may be useful as an adjunct or alternative to standard revascularization techniques in patients with ischemic heart failure.


Asunto(s)
Inductores de la Angiogénesis/uso terapéutico , Cardiomiopatías/tratamiento farmacológico , Isquemia Miocárdica/tratamiento farmacológico , Proteínas Gestacionales/uso terapéutico , Inductores de la Angiogénesis/administración & dosificación , Animales , Gasto Cardíaco , Cardiomiopatías/fisiopatología , Inyecciones , Masculino , Isquemia Miocárdica/fisiopatología , Miocardio , Neovascularización Fisiológica/efectos de los fármacos , Factor de Crecimiento Placentario , Proteínas Gestacionales/administración & dosificación , Ratas , Ratas Wistar , Función Ventricular/efectos de los fármacos
20.
Fertil Steril ; 85(5): 1553-6, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16647384

RESUMEN

Glycodelin modulates vascular endothelial growth factor (VEGF) production in cumulus cells in vitro. Patients with normal gonadotropin responses who were undergoing IVF demonstrated increased VEGF production to glycodelin, whereas poor responders had a decreased response to glycodelin.


Asunto(s)
Glicoproteínas/administración & dosificación , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Proteínas Gestacionales/administración & dosificación , Factor A de Crecimiento Endotelial Vascular/metabolismo , Adulto , Estudios de Casos y Controles , Células Cultivadas , Relación Dosis-Respuesta a Droga , Femenino , Glicodelina , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...