Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Med Rev (2021) ; 4(3): 235-238, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38919399

RESUMEN

The protein, N-myc downstream-regulated gene 2 (NDRG2), a tumor suppressor, is significantly decreased or absent in many types of cancer. There is a significant negative correlation between the levels of NDRG2 and the development and progression of cancer tumor recurrence and tumor invasion, in different cancers. In contrast, the in vitro and in vivo overexpression of the NDRG2 protein decreases the proliferation, growth, adhesion and migration of many types of cancer cells. The in vitro overexpression of NDRG2 increases the efficacy of certain anticancer drugs in specific types of cancer cells. We hypothesize that the delivery of the mRNA of the NDRG2 protein, encapsulated by lipid nanoparticles, could represent a potential treatment of metastatic and drug-resistant cancers. This would be accomplished using a self-amplifying mRNA that encodes the NDRG2 protein and an RNA-dependent-RNA polymerase, obtained from an in vitrotranscribed (IVT) mRNA. The IVT mRNA would be encapsulated in a lipid nanoformulation. The efficacy of the nanoformulation would be determined in cultured cancer cells and if the results are positive, nude mice transplanted with either drug-resistant or metastatic drug-resistant cancer cells, would be treated with the nano- formulation and monitored for efficacy and adverse effects. If the appropriate preclinical studies indicate this formulation is efficacious and safe, it is possible it could be evaluated in clinical trials.

2.
Reprod Sci ; 29(10): 2894-2907, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35349119

RESUMEN

Preterm birth accounts for the majority of perinatal mortality worldwide, and there remains no FDA-approved drug to prevent it. Recently, we discovered that the common drug excipient, N,N-dimethylacetamide (DMA), delays inflammation-induced preterm birth in mice by inhibiting NF-κB. Since we reported this finding, it has come to light that a group of widely used, structurally related aprotic solvents, including DMA, N-methyl-2-pyrrolidone (NMP) and dimethylformamide (DMF), have anti-inflammatory efficacy. We show here that DMF suppresses LPS-induced TNFα secretion from RAW 264.7 cells and IL-6 and IL-8 secretion from HTR-8 cells at concentrations that do not significantly affect cell viability. Like DMA, DMF protects IκBα from degradation and prevents the p65 subunit of NF-κB from translocating to the nucleus. In vivo, DMF decreases LPS-induced inflammatory cell infiltration and expression of TNFα and IL-6 in the placental labyrinth, all to near baseline levels. Finally, DMF decreases the rate of preterm birth in LPS-induced pregnant mice (P<.0001) and the rate at which pups are spontaneously aborted (P<.0001). In summary, DMF, a widely used solvent structurally related to DMA and NMP, delays LPS-induced preterm birth in a murine model without overt toxic effects. Re-purposing the DMA/DMF/NMP family of small molecules as anti-inflammatory drugs is a promising new approach to delaying or reducing the incidence of inflammation-induced preterm birth and potentially attenuating other inflammatory disorders as well.


Asunto(s)
Dimetilformamida , Nacimiento Prematuro , Acetamidas , Animales , Antiinflamatorios/farmacología , Dimetilformamida/efectos adversos , Modelos Animales de Enfermedad , Excipientes/efectos adversos , Femenino , Humanos , Recién Nacido , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Inflamación/prevención & control , Interleucina-6 , Interleucina-8 , Lipopolisacáridos/farmacología , Ratones , Inhibidor NF-kappaB alfa , FN-kappa B/metabolismo , Placenta/metabolismo , Embarazo , Nacimiento Prematuro/inducido químicamente , Nacimiento Prematuro/prevención & control , Solventes/efectos adversos , Factor de Necrosis Tumoral alfa
3.
Pharmaceutics ; 13(7)2021 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-34371697

RESUMEN

A novel treatment strategy by co-targeting c-Myc and tumor stroma was explored in vemurafenib-resistant melanoma. BRD4 proteolysis targeting chimera (ARV-825) and nintedanib co-loaded PEGylated nanoliposomes (ARNIPL) were developed to incorporate a synergistic cytotoxic ratio. Both the molecules have extremely poor aqueous solubility. A modified hydration method with citric acid was used to improve the loading of both the molecules in liposomes. ARNIPL with mean particle size 111.1 ± 6.55 nm exhibited more than 90% encapsulation efficiency for both the drugs and was found to be physically stable for a month at 4 °C. Both the molecules and ARNIPL showed significantly higher cytotoxicity, apoptosis and down-regulation of target proteins BRD4 and c-Myc in vemurafenib-resistant cell line (A375R). Vasculogenic mimicry and clonogenic potential of A375R were significantly inhibited by ARNIPL. Tumor growth inhibition in 3D spheroids with reduction of TGF-ß1 was observed with ARNIPL treatment. Therefore, ARNIPL could be a promising therapeutic approach for the treatment of vemurafenib-resistant melanoma.

4.
Cancers (Basel) ; 12(9)2020 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-32899268

RESUMEN

Multidrug resistance (MDR) associated with the overexpression of ABC transporters is one of the key causes of chemotherapy failure. Various compounds blocking the function and/or downregulating the expression of these transporters have been developed over the last few decades. However, their potency and toxicity have always been a concern. In this report, we found that BMS-599626 is a highly potent inhibitor of the ABCG2 transporter, inhibiting its efflux function at 300 nM. Our study repositioned BMS-599626, a highly selective pan-HER kinase inhibitor, as a chemosensitizer in ABCG2-overexpressing cell lines. As shown by the cytotoxicity assay results, BMS-599626, at noncytotoxic concentrations, sensitizes ABCG2-overexpressing cells to topotecan and mitoxantrone, two well-known substrates of ABCG2. The results of our radioactive drug accumulation experiment show that the ABCG2-overexpressing cells, treated with BMS-599626, had an increase in the accumulation of substrate chemotherapeutic drugs, as compared to their parental subline cells. Moreover, BMS-599626 did not change the protein expression or cell surface localization of ABCG2 and inhibited its ATPase activity. Our in-silico docking study also supports the interaction of BMS-599626 with the substrate-binding site of ABCG2. Taken together, these results suggest that administration of chemotherapeutic drugs, along with nanomolar concentrations (300 nM) of BMS-599626, may be effective against ABCG2-mediated MDR in clinical settings.

5.
Diabetes ; 69(8): 1650-1661, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32444367

RESUMEN

An adverse maternal in utero and lactation environment can program offspring for increased risk for metabolic disease. The aim of this study was to determine whether N-acetylcysteine (NAC), an anti-inflammatory antioxidant, attenuates programmed susceptibility to obesity and insulin resistance in offspring of mothers on a high-fat diet (HFD) during pregnancy. CD1 female mice were acutely fed a standard breeding chow or HFD. NAC was added to the drinking water (1 g/kg) of the treatment cohorts from embryonic day 0.5 until the end of lactation. NAC treatment normalized HFD-induced maternal weight gain and oxidative stress, improved the maternal lipidome, and prevented maternal leptin resistance. These favorable changes in the in utero environment normalized postnatal growth, decreased white adipose tissue (WAT) and hepatic fat, improved glucose and insulin tolerance and antioxidant capacity, reduced leptin and insulin, and increased adiponectin in HFD offspring. The lifelong metabolic improvements in the offspring were accompanied by reductions in proinflammatory gene expression in liver and WAT and increased thermogenic gene expression in brown adipose tissue. These results, for the first time, provide a mechanistic rationale for how NAC can prevent the onset of metabolic disease in the offspring of mothers who consume a typical Western HFD.


Asunto(s)
Acetilcisteína/uso terapéutico , Dieta Alta en Grasa/efectos adversos , Enfermedades Metabólicas/tratamiento farmacológico , Enfermedades Metabólicas/metabolismo , Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/efectos de los fármacos , Tejido Adiposo Blanco/metabolismo , Adiposidad/efectos de los fármacos , Animales , Antioxidantes/metabolismo , Temperatura Corporal , Calorimetría Indirecta , Femenino , Prueba de Tolerancia a la Glucosa , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Inyecciones Intraperitoneales , Resistencia a la Insulina , Masculino , Ratones , Aumento de Peso/efectos de los fármacos
6.
Am J Reprod Immunol ; 83(4): e13221, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31943498

RESUMEN

PROBLEM: As more women join the skilled-trade workforce, the effects of workplace exposures on pregnancy need to be explored. This study aims to identify the effects of mild steel and stainless steel welding fume exposures on cultured placental trophoblast cells. METHOD OF STUDY: Welding fumes (mild steel and stainless steel) were generously donated by Lincoln Electric. Electron microscopy was used to characterize welding fume particle size and the ability of particles to enter extravillous trophoblast cells (HTR-8/SVneo). Cellular viability, free radical production, cytokine production, and ability of cells to maintain invasive properties were analyzed, respectively, by WST-1, electron paramagnetic resonance, DCFH-DA, V-plex MULTI-SPOT assay system, and a matrix gel invasion assay. RESULTS: For all three welding fume types, average particle size was <210 nm. HTR-8/SVneo cells internalized welding particles, and nuclear condensation was observed. Cellular viability was significantly decreased at the high dose of 100 µg/mL for all three welding fumes, and stainless steel generated the greatest production of the hydroxyl radical, and intracellular reactive oxygen species. Production of the cytokines IL-1ß and TNFα were not observed in response to welding fume exposure, but IL-6 and IL-8 were. Finally, the invasive capability of cells was decreased upon exposure to both mild steel and stainless steel welding fumes. CONCLUSION: Welding fumes are cytotoxic to extravillous trophoblasts, as is evident by the production of free radicals, pro-inflammatory cytokines, and the observed decrease in invasive capabilities.


Asunto(s)
Contaminantes Atmosféricos/efectos adversos , Exposición Profesional/efectos adversos , Trofoblastos/patología , Supervivencia Celular , Células Cultivadas , Citocinas/metabolismo , Femenino , Humanos , Mediadores de Inflamación/metabolismo , Estrés Oxidativo , Embarazo , Primer Trimestre del Embarazo , Especies Reactivas de Oxígeno/metabolismo , Acero Inoxidable , Soldadura
7.
Am J Pathol ; 189(11): 2246-2257, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31430466

RESUMEN

The mechanism by which poor maternal nutrition can affect the long-term health of offspring is poorly understood. In mice, we previously found that maternal high-fat diet (HFD) exposure results in reduced fetal growth regardless of maternal genotype. We tested our hypothesis that maternal HFD-induced inflammation contributes to metabolic disease susceptibility of the offspring via alterations in the placenta. The effect of maternal genotype, diet, and treatment with the anti-inflammatory compound N-acetylcysteine (NAC) on placental morphologic features was investigated. Placentas from wild-type dams maintained on a HFD but not those heterozygous (+/-) for Glut4 (Slc2a4) on the same diet had an increase in decidual inflammation and vasculopathy occurring together. NAC administration resulted in amelioration of HFD-induced decidual vasculopathy independent of offspring genotype and sex. Consistent with these morphologic improvements, placentas from HFD dams treated with NAC had decreased mRNA and immunostaining of IL-1ß and monocyte chemoattractant protein-1, decreased mRNA of inflammatory genes, and increased mRNA of Vegfa. These results strongly suggest consumption of an HFD results in vascular changes in placenta reflected by alterations in expression of pivotal vascular developmental markers and inflammatory genes all of which are ameliorated by NAC. These placental changes play a key role in the increased programed metabolic disease of HFD-exposed offspring.


Asunto(s)
Acetilcisteína/uso terapéutico , Dieta Alta en Grasa/efectos adversos , Inflamación/prevención & control , Placenta/efectos de los fármacos , Complicaciones del Embarazo/prevención & control , Enfermedades Vasculares/prevención & control , Animales , Modelos Animales de Enfermedad , Femenino , Inflamación/complicaciones , Inflamación/patología , Masculino , Fenómenos Fisiologicos Nutricionales Maternos/fisiología , Ratones , Ratones Transgénicos , Placenta/patología , Embarazo , Complicaciones del Embarazo/etiología , Enfermedades Vasculares/complicaciones , Enfermedades Vasculares/patología
8.
Drug Discov Today ; 24(10): 2086-2095, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31173912

RESUMEN

The mammalian target of rapamycin (mTOR) is a highly conserved serine/threonine kinase that regulates a variety of cellular processes, influencing diverse pathological conditions including a variety of cancers. Accordingly, therapies that target mTOR as anticancer agents benefit patients in various clinical settings. It is therefore important to fully investigate mTOR signaling at a molecular level and corresponding mTOR inhibitors to identify additional clinical opportunities of targeting mTOR in cancers. In this review, we introduce the function and regulation of the mTOR signaling pathway and organize and summarize the different roles of mTOR in cancers and a variety of mTOR inhibitors that can be used as anticancer agents. This article aims to enlighten and guide the development of mTOR-targeted anticancer agents in the future.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Humanos , Neoplasias/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/fisiología
9.
Cancer Lett ; 442: 91-103, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30391357

RESUMEN

Phosphoinositide 3-kinase gamma isoform (PI3Kγ) plays a critical role in myeloid-derived cells of the immunosuppressive tumor microenvironment. IPI-549, a recently discovered small molecule selective PI3Kγ inhibitor, is currently under immuno-oncology clinical trials in combination with nivolumab, an anti-PD-1 monoclonal antibody immune checkpoint blocker. The purpose of this study is to investigate whether IPI-549 could reverse P-glycoprotein (P-gp)-mediated MDR when combined with chemotherapeutic substrates of P-gp. Cytotoxicity assays showed that IPI-549 reverses P-gp-mediated MDR in SW620/Ad300 and LLC-PK-MDR1 cells. IPI-549 increases the amount of intracellular paclitaxel and inhibits the efflux of paclitaxel out of SW620/Ad300 cells. ABCB1-ATPase assay showed that IPI-549 stimulates the activity of ABCB1-ATPase. IPI-549 does not alter the expression and does not affect the subcellular localization of P-gp in SW620/Ad300 cells. The combination of IPI-549 with paclitaxel showed that IPI-549 potentiates the anti-tumor effects of paclitaxel in P-gp-overexpressing MDR SW620/Ad300 xenograft tumors. With clinical trials beginning to add newly approved immune checkpoint-based immunotherapy into standard-of-care immunogenic chemotherapy to improve patient outcomes, our findings support the rationale of adding IPI-549 to both the chemotherapeutic and immunotherapeutic aspects of cancer combination treatment strategies.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias del Colon/tratamiento farmacológico , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Paclitaxel/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/farmacología , Subfamilia B de Transportador de Casetes de Unión a ATP/agonistas , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Animales , Antineoplásicos/química , Antineoplásicos/metabolismo , Sitios de Unión , Fosfatidilinositol 3-Quinasa Clase Ib/química , Fosfatidilinositol 3-Quinasa Clase Ib/metabolismo , Neoplasias del Colon/enzimología , Neoplasias del Colon/inmunología , Neoplasias del Colon/patología , Humanos , Células LLC-PK1 , Masculino , Ratones Desnudos , Simulación del Acoplamiento Molecular , Proteínas de Neoplasias/metabolismo , Paclitaxel/metabolismo , Unión Proteica , Conformación Proteica , Inhibidores de Proteínas Quinasas/química , Transducción de Señal/efectos de los fármacos , Porcinos , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Mol Med ; 22: 747-758, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27782292

RESUMEN

Previously, we have shown that N,N-dimethylacetamide (DMA) prevents inflammation-induced preterm birth in a murine model, inhibits LPS-induced increases in placental pro-inflammatory cytokines and up-regulates the anti-inflammatory cytokine Interleukin-10 (IL-10). However, DMA's mechanism of action remains to be elucidated. In the current study we investigate how DMA produces its anti-inflammatory effect. Using in vitro and ex vivo models, we show that DMA suppresses secretion of pro-inflammatory cytokines in lipopolysaccharide (LPS)-induced RAW 264.7 cells, TNFα-challenged JEG-3 cells and LPS-stimulated human placental explants. DMA significantly attenuated the secretion of TNFα, IL-6, IL-10, and granulocyte macrophage colony stimulating factor (GM-CSF) from LPS-stimulated RAW 264.7 cells, IL-6 secretion from TNFα-stimulated JEG-3 cells and TNFα, IL-6, IL-10, GM-CSF and Interleukin-8 (IL-8) from LPS-stimulated human placental explants. We further investigated if DMA's effect on cytokine expression involves the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and mitogen-activated protein kinase (MAPK) pathways. DMA (10 mM) significantly inhibited nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, alpha (IκBα) degradation in LPS-stimulated RAW 264.7 cells, but there was no significant change in the expression of phosphorylated or native forms of downstream proteins in the MAPK pathway. In addition, DMA significantly attenuated luciferase activity in cells co-transfected with NF-κB-Luc reporter plasmid, but not with AP-1-Luc or CEBP-Luc reporters. Overall, our findings suggest that the anti-inflammatory activity of DMA is mediated by inhibition of the NF-κB pathway via decreased IκBα degradation.

11.
Am J Pathol ; 185(3): 862-9, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25579843

RESUMEN

Premature delivery occurs in 12% of all births, and accounts for nearly half of long-term neurological morbidity, and 60% to 80% of perinatal mortality. Despite advances in obstetrics and neonatology, the rate of premature delivery has increased approximately 12% since 1990. The single most common cause of spontaneous preterm birth is infection. Several lines of evidence have demonstrated the role of endothelin-1 as both a constrictor of uterine myometrial smooth muscle and a proinflammatory mediator. Endothelin-1 activates the phospholipase C pathway, leading to activation of protein kinase C and, in turn, sphingosine kinase (SphK). The inhibition of SphK has been recently shown to control the proinflammatory response associated with sepsis. We show herein, for the first time, that SphK inhibition prevents inflammation-associated preterm birth in a murine model. Rescue of pups from premature abortion with an SphK inhibitor occurs by suppression of the proinflammatory cytokines tumor necrosis factor α, Il-1ß, and Il-6 and attenuation of polymorphonuclear inflammatory cells into the placental labyrinth. Moreover, we postulate that inhibition of SphK leads to suppression of endothelin-converting enzyme-1 expression, indicating the presence of an endothelin-converting enzyme 1/endothelin 1-SphK positive feedback loop. This work introduces a novel approach for the control of infection-triggered preterm labor, a condition for which there is no effective treatment.


Asunto(s)
Citocinas/metabolismo , Inhibidores Enzimáticos/uso terapéutico , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Placenta/metabolismo , Nacimiento Prematuro/prevención & control , Animales , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Femenino , Inflamación/metabolismo , Lipopolisacáridos , Ratones , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Placenta/efectos de los fármacos , Embarazo , Nacimiento Prematuro/inducido químicamente , Nacimiento Prematuro/metabolismo
12.
Am J Pathol ; 183(2): 422-30, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23770347

RESUMEN

The proinflammatory response leads to various types of pathologic pathways, including the development of preterm birth. Preterm birth occurs in 12% of deliveries in the United States and causes more than 70% of perinatal morbidity and mortality. The most common cause of spontaneous preterm birth is intrauterine infection in the mother. There is accumulating evidence indicating that the release of proinflammatory cytokines plays a critical role in the pathogenesis of inflammation-associated premature delivery. We found that the common organic solvent, N,N-dimethylacetamide (DMA), prevents endotoxin-induced preterm birth in timed pregnant C57BL/6 embryonic day (E)15.5 mice and rescues their pups from spontaneous abortion at doses many-fold lower than those currently used clinically and in a dose-dependent fashion. We also provide histologic evidence that DMA suppresses the endotoxin-triggered proinflammatory response by significantly attenuating inflammatory cell infiltration of placental tissue. Furthermore, immunoblotting analysis of placental tissue harvested from our murine models revealed DMA-mediated regulation of expression of the proinflammatory cytokines IL-1ß, tumor necrosis factor α, and IL-6, and increased expression of the regulatory inflammatory cytokine IL-10. By using in vitro studies, we provide evidence that DMA suppresses macrophage function and that this small molecule prevents nuclear translocation of nuclear factor-kB. These results suggest that DMA represents a newly discovered, nontoxic therapy for a broad range of inflammatory disorders.


Asunto(s)
Acetamidas/farmacología , Antiinflamatorios/farmacología , Citocinas/efectos de los fármacos , Endotoxinas/toxicidad , Nacimiento Prematuro/prevención & control , Animales , Femenino , Lipopolisacáridos/toxicidad , Ratones , Ratones Endogámicos C57BL , FN-kappa B/efectos de los fármacos , Embarazo , Nacimiento Prematuro/inducido químicamente
13.
PLoS One ; 7(10): e44117, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23082110

RESUMEN

Neurological and cognitive impairment persist in more than 20% of cerebral malaria (CM) patients long after successful anti-parasitic treatment. We recently reported that long term memory and motor coordination deficits are also present in our experimental cerebral malaria model (ECM). We also documented, in a murine model, a lack of obvious pathology or inflammation after parasite elimination, suggesting that the long-term negative neurological outcomes result from potentially reversible biochemical and physiological changes in brains of ECM mice, subsequent to acute ischemic and inflammatory processes. Here, we demonstrate for the first time that acute ECM results in significantly reduced activation of protein kinase B (PKB or Akt) leading to decreased Akt phosphorylation and inhibition of the glycogen kinase synthase (GSK3ß) in the brains of mice infected with Plasmodium berghei ANKA (PbA) compared to uninfected controls and to mice infected with the non-neurotrophic P. berghei NK65 (PbN). Though Akt activation improved to control levels after chloroquine treatment in PbA-infected mice, the addition of lithium chloride, a compound which inhibits GSK3ß activity and stimulates Akt activation, induced a modest, but significant activation of Akt in the brains of infected mice when compared to uninfected controls treated with chloroquine with and without lithium. In addition, lithium significantly reversed the long-term spatial and visual memory impairment as well as the motor coordination deficits which persisted after successful anti-parasitic treatment. GSK3ß inhibition was significantly increased after chloroquine treatment, both in lithium and non-lithium treated PbA-infected mice. These data indicate that acute ECM is associated with abnormalities in cell survival pathways that result in neuronal damage. Regulation of Akt/GSK3ß with lithium reduces neuronal degeneration and may have neuroprotective effects in ECM. Aberrant regulation of Akt/GSK3ß signaling likely underlies long-term neurological sequelae observed in ECM and may yield adjunctive therapeutic targets for the management of CM.


Asunto(s)
Cognición/efectos de los fármacos , Litio/farmacología , Malaria Cerebral/tratamiento farmacológico , Malaria Cerebral/enzimología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Enfermedad Aguda , Animales , Cloroquina/farmacología , Cloroquina/uso terapéutico , Femenino , Técnica del Anticuerpo Fluorescente , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Immunoblotting , Litio/uso terapéutico , Malaria Cerebral/parasitología , Malaria Cerebral/fisiopatología , Ratones , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Parasitemia/tratamiento farmacológico , Parasitemia/enzimología , Parasitemia/parasitología , Parasitemia/fisiopatología , Fosforilación/efectos de los fármacos , Proteínas tau/metabolismo
14.
Am J Physiol Endocrinol Metab ; 302(5): E522-31, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22167521

RESUMEN

Alterations in insulin signaling as well as insulin action predispose to infertility as well as adverse pregnancy outcomes; however, little is known about the role of glucagon signaling in reproduction. The glucagon receptor knockout (Gcgr(-/-)) mouse created by our laboratory was used to define the role of glucagon signaling in maintaining normal reproduction. In this mouse model, lack of glucagon signaling did not alter the hypothalamic-pituitary-ovarian axis. Pregnant Gcgr(-/-) female mice displayed persistent hypoglycemia and hyperglucagonemia. Gcgr(-/-) pregnancies were associated with decreased fetal weight, increased late-gestation fetal demise, and significant abnormalities of placentation. Gcgr(-/-) placentas contained areas of extensive mineralization, fibrinoid necrosis, narrowing of the vascular channels, and a thickened interstitium associated with trophoblast hyperplasia. Absent glucagon signaling did not alter glycogen content in Gcgr(-/-) placentas but significantly downregulated genes that control growth, adrenergic signaling, vascularization, oxidative stress, and G protein-coupled receptors. Our data suggest that, similarly to insulin, glucagon action contributes to normal female reproductive function.


Asunto(s)
Enfermedades Fetales/etiología , Glucagón/fisiología , Hipoglucemia/etiología , Enfermedades Placentarias/etiología , Embarazo/fisiología , Receptores de Glucagón/fisiología , Animales , Femenino , Muerte Fetal/etiología , Enfermedades Fetales/metabolismo , Retardo del Crecimiento Fetal/etiología , Regulación del Desarrollo de la Expresión Génica , Glucagón/sangre , Heterocigoto , Hipoglucemia/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ovario/efectos de los fármacos , Ovario/fisiología , Adenohipófisis/metabolismo , Adenohipófisis/patología , Placenta/metabolismo , Placenta/patología , Enfermedades Placentarias/metabolismo , Enfermedades Placentarias/patología , Placentación , Proteínas Gestacionales/genética , Proteínas Gestacionales/metabolismo , Receptores de Glucagón/genética , Transducción de Señal , Superovulación/efectos de los fármacos
15.
Exp Lung Res ; 34(3): 141-54, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18307123

RESUMEN

Phosphoramidon blocks the formation of endothelin-1 (ET-1), a proinflammatory mediator implicated in the pathogenesis of a variety of lung diseases. To determine whether phosphoramidon can ameliorate pulmonary inflammation, our laboratory undertook a series of experiments involving treatment of hamsters with either intraperitoneal (i.p.) or aerosolized phosphoramidon prior to induction of acute lung injury by intratracheal administration of lipopolysaccharide (LPS). The results indicate that phosphoramidon significantly reduces LPS-induced pulmonary inflammation as measured by lung histology, neutrophil content of bronchoalveolar lavage (BAL) fluid, percent tumor necrosis factor receptor 1 (TNFR1)-labeled BAL macrophages, and alveolar septal cell apoptosis. In additional experiments, i.p. administration of a novel endothelin A receptor anatgonist (HJP272) similarly decreased BAL neutrophils, whereas i.p. administration of either ET-1, or its precursor peptide, "big" ET-1, had the opposite effect. These findings support further evaluation of phosphoramidon and other ET-1 suppressors as potential treatments for human inflammatory lung disease.


Asunto(s)
Glicopéptidos/uso terapéutico , Lipopolisacáridos/toxicidad , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Animales , Apoptosis , Cricetinae , Antagonistas de los Receptores de la Endotelina A , Endotelina-1/farmacología , Pulmón/efectos de los fármacos , Pulmón/patología , Mesocricetus , Receptores Tipo I de Factores de Necrosis Tumoral/análisis , Síndrome de Dificultad Respiratoria/inducido químicamente
16.
Brain Res Gene Expr Patterns ; 1(2): 135-40, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15018810

RESUMEN

ProSAAS (encoded by mouse gene Pcsk1n) is a recently described neuroendocrine secretory pathway protein that is cleaved into smaller peptides that may function in cell-cell signalling. ProSAAS and its processing intermediates are also potent inhibitors of prohormone convertase 1 (PC1), which is encoded by mouse gene Pcsk1. In order to gain insight into the function of proSAAS, we have examined the distribution of several proSAAS-derived peptides and PC1 by immunohistochemistry throughout mouse development. The distribution patterns of both SAAS and PC1 are broad from E9 to E11, with some enrichment in neural tube-derived tissues. By E15, the expression of SAAS is largely restricted to neuroendocrine tissues known to produce bioactive peptides. In general, the expression pattern of PC1 overlaps with that of SAAS and other proSAAS-derived peptides, consistent with the hypothesis that proSAAS functions as an endogenous PC1 inhibitor.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Proteínas del Tejido Nervioso/biosíntesis , Proproteína Convertasa 1/biosíntesis , Animales , Encéfalo/embriología , Carboxipeptidasa H/química , Cromograninas/metabolismo , Inhibidores Enzimáticos/farmacología , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Neuropéptidos/química , Péptidos/química , Transducción de Señal , Factores de Tiempo , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA