Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Proc Natl Acad Sci U S A ; 115(29): E6920-E6926, 2018 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-29967151

RESUMEN

Isoxazolines are oral insecticidal drugs currently licensed for ectoparasite control in companion animals. Here we propose their use in humans for the reduction of vector-borne disease incidence. Fluralaner and afoxolaner rapidly killed Anopheles, Aedes, and Culex mosquitoes and Phlebotomus sand flies after feeding on a drug-supplemented blood meal, with IC50 values ranging from 33 to 575 nM, and were fully active against strains with preexisting resistance to common insecticides. Based on allometric scaling of preclinical pharmacokinetics data, we predict that a single human median dose of 260 mg (IQR, 177-407 mg) for afoxolaner, or 410 mg (IQR, 278-648 mg) for fluralaner, could provide an insecticidal effect lasting 50-90 days against mosquitoes and Phlebotomus sand flies. Computational modeling showed that seasonal mass drug administration of such a single dose to a fraction of a regional population would dramatically reduce clinical cases of Zika and malaria in endemic settings. Isoxazolines therefore represent a promising new component of drug-based vector control.


Asunto(s)
Control de Enfermedades Transmisibles/métodos , Culicidae/crecimiento & desarrollo , Insecticidas/farmacología , Control de Mosquitos/métodos , Mosquitos Vectores/crecimiento & desarrollo , Psychodidae/crecimiento & desarrollo , Animales , Humanos
2.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1862(5): 541-551, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28214558

RESUMEN

Small heterodimer partner (SHP) is an atypical nuclear receptor expressed in heart that has been shown to inhibit the hypertrophic response. Here, we assessed the role of SHP in cardiac metabolism and inflammation. Mice fed a high-fat diet (HFD) displayed glucose intolerance accompanied by increased cardiac mRNA levels of Shp. In HL-1 cardiomyocytes, SHP overexpression inhibited both basal and insulin-stimulated glucose uptake and impaired the insulin signalling pathway (evidenced by reduced AKT and AS160 phosphorylation), similar to insulin resistant cells generated by high palmitate/high insulin treatment (HP/HI; 500µM/100nM). In addition, SHP overexpression increased Socs3 mRNA and reduced IRS-1 protein levels. SHP overexpression also induced Cd36 expression (~6.2 fold; p<0.001) linking to the observed intramyocellular lipid accumulation. SHP overexpressing cells further showed altered expression of genes involved in lipid metabolism, i.e., Acaca, Acadvl or Ucp3, augmented NF-κB DNA-binding activity and induced transcripts of inflammatory genes, i.e., Il6 and Tnf mRNA (~4-fold induction, p<0.01). Alterations in metabolism and inflammation found in SHP overexpressing cells were associated with changes in the mRNA levels of Ppara (79% reduction, p<0.001) and Pparg (~58-fold induction, p<0.001). Finally, co-immunoprecipitation studies showed that SHP overexpression strongly reduced the physical interaction between PPARα and the p65 subunit of NF-κB, suggesting that dissociation of these two proteins is one of the mechanisms by which SHP initiates the inflammatory response in cardiac cells. Overall, our results suggest that SHP upregulation upon high-fat feeding leads to lipid accumulation, insulin resistance and inflammation in cardiomyocytes.


Asunto(s)
Inflamación/metabolismo , Resistencia a la Insulina/genética , Metabolismo de los Lípidos/genética , Miocardio/metabolismo , Receptores Citoplasmáticos y Nucleares/biosíntesis , Animales , Regulación de la Expresión Génica , Glucosa/metabolismo , Humanos , Inflamación/patología , Insulina/metabolismo , Ratones , Miocardio/patología , Miocitos Cardíacos/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/metabolismo , Receptores Citoplasmáticos y Nucleares/genética , Transducción de Señal
3.
Biochem J ; 473(7): 937-47, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26831516

RESUMEN

AMP-activated protein kinase (AMPK) is a metabolic stress-sensing kinase. We previously showed that glucose deprivation induces autophosphorylation of AMPKß at Thr-148, which prevents the binding of AMPK to glycogen. Furthermore, in MIN6 cells, AMPKß1 binds to R6 (PPP1R3D), a glycogen-targeting subunit of protein phosphatase type 1 (PP1), thereby regulating the glucose-induced inactivation of AMPK. In the present study, we further investigated the interaction of R6 with AMPKß and the possible dependency on Thr-148 phosphorylation status. Yeast two-hybrid (Y2H) analyses and co-immunoprecipitation (IP) of the overexpressed proteins in human embryonic kidney (HEK) 293T) cells revealed that both AMPKß1 and AMPK-ß2 wild-type (WT) isoforms bind to R6. The AMPKß-R6 interaction was stronger with the muscle-specific AMPKß2-WT and required association with the substrate-binding motif of R6. When HEK293T cells or C2C12 myotubes were cultured in high-glucose medium, AMPKß2-WT and R6 weakly interacted. In contrast, glycogen depletion significantly enhanced this protein interaction. Mutation of AMPKß2 Thr-148 prevented the interaction with R6 irrespective of the intracellular glycogen content. Treatment with the AMPK activator oligomycin enhanced the AMPKß2-R6 interaction in conjunction with increased Thr-148 phosphorylation in cells grown in low-glucose medium. These data are in accordance with R6 binding directly to AMPKß2 when both proteins detach from the diminishing glycogen particle, which is simultaneous with increased AMPKß2 Thr-148 autophosphorylation. Such a model points to a possible control of AMPK by PP1-R6 upon glycogen depletion in muscle.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Glucógeno/metabolismo , Mutación Missense , Proteína Fosfatasa 1/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Sustitución de Aminoácidos , Glucógeno/genética , Células HEK293 , Humanos , Fosforilación , Unión Proteica , Proteína Fosfatasa 1/genética
4.
J Biol Chem ; 290(18): 11715-28, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25792737

RESUMEN

The mammalian AMP-activated protein kinase (AMPK) is an obligatory αßγ heterotrimeric complex carrying a carbohydrate-binding module (CBM) in the ß-subunit (AMPKß) capable of attaching AMPK to glycogen. Nonetheless, AMPK localizes at many different cellular compartments, implying the existence of mechanisms that prevent AMPK from glycogen binding. Cell-free carbohydrate binding assays revealed that AMPK autophosphorylation abolished its carbohydrate-binding capacity. X-ray structural data of the CBM displays the central positioning of threonine 148 within the binding pocket. Substitution of Thr-148 for a phospho-mimicking aspartate (T148D) prevents AMPK from binding to carbohydrate. Overexpression of isolated CBM or ß1-containing AMPK in cellular models revealed that wild type (WT) localizes to glycogen particles, whereas T148D shows a diffuse pattern. Pharmacological AMPK activation and glycogen degradation by glucose deprivation but not forskolin enhanced cellular Thr-148 phosphorylation. Cellular glycogen content was higher if pharmacological AMPK activation was combined with overexpression of T148D mutant relative to WT AMPK. In summary, these data show that glycogen-binding capacity of AMPKß is regulated by Thr-148 autophosphorylation with likely implications in the regulation of glycogen turnover. The findings further raise the possibility of regulated carbohydrate-binding function in a wider variety of CBM-containing proteins.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Glucógeno/metabolismo , Proteínas Quinasas Activadas por AMP/química , Proteínas Quinasas Activadas por AMP/genética , Activación Enzimática , Células HEK293 , Células Hep G2 , Humanos , Modelos Moleculares , Mutación , Fosforilación , Unión Proteica , Conformación Proteica , Transporte de Proteínas , Treonina
5.
Commun Biol ; 6(1): 216, 2023 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-36823266

RESUMEN

The sporozoite stages of malaria parasites are the primary cause of infection of the vertebrate host and are targeted by (experimental) vaccines. Yet, little is known about their susceptibility to chemical intervention. Phenotypic high-throughput screens have not been feasible due to a lack of in vitro systems. Here we tested 78 marketed and experimental antimalarial compounds in miniaturized assays addressing sporozoite viability, gliding motility, hepatocyte traversal, and intrahepatocytic schizogony. None potently interfered with sporozoite viability or motility but ten compounds acted at the level of schizogony with IC50s < 100 nM. To identify compounds directly targeting sporozoites, we screened 81,000 compounds from the Global Health Diversity and reFRAME libraries in a sporozoite viability assay using a parasite expressing a luciferase reporter driven by the circumsporozoite promoter. The ionophore gramicidin emerged as the single hit from this screening campaign. Its effect on sporozoite viability translated into reduced gliding motility and an inability of sporozoites to invade human primary hepatocytes and develop into hepatic schizonts. While providing proof of concept for a small molecule sporontocidal mode of action, our combined data indicate that liver schizogony is more accessible to chemical intervention by (candidate) antimalarials.


Asunto(s)
Antimaláricos , Malaria , Animales , Humanos , Esporozoítos , Ensayos Analíticos de Alto Rendimiento , Malaria/tratamiento farmacológico , Malaria/parasitología , Antimaláricos/farmacología , Antimaláricos/uso terapéutico , Hígado
6.
Nat Cancer ; 2(11): 1152-1169, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-35122061

RESUMEN

The nongenetic mechanisms required to sustain malignant tumor state are poorly understood. During the transition from benign tumors to malignant carcinoma, tumor cells need to repress differentiation and acquire invasive features. Using transcriptional profiling of cancer stem cells from benign tumors and malignant skin squamous cell carcinoma (SCC), we identified the nuclear receptor NR2F2 as uniquely expressed in malignant SCC. Using genetic gain of function and loss of function in vivo, we show that NR2F2 is essential for promoting the malignant tumor state by controlling tumor stemness and maintenance in mouse and human SCC. We demonstrate that NR2F2 promotes tumor cell proliferation, epithelial-mesenchymal transition and invasive features, while repressing tumor differentiation and immune cell infiltration by regulating a common transcriptional program in mouse and human SCCs. Altogether, we identify NR2F2 as a key regulator of malignant cancer stem cell functions that promotes tumor renewal and restricts differentiation to sustain a malignant tumor state.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias Cutáneas , Animales , Carcinoma de Células Escamosas/genética , Diferenciación Celular/genética , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica , Ratones , Procesos Neoplásicos , Neoplasias Cutáneas/genética
7.
J Med Chem ; 59(7): 2879-93, 2016 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-26510622

RESUMEN

As a central regulator of metabolism, the AMP-activated protein kinase (AMPK) is an established therapeutic target for metabolic diseases. Beyond the metabolic area, the number of medical fields that involve AMPK grows continuously, expanding the potential applications for AMPK modulators. Even though indirect AMPK activators are used in the clinics for their beneficial metabolic outcome, the few described direct agonists all failed to reach the market to date, which leaves options open for novel targeting methods. As AMPK is not actually a single molecule and has different roles depending on its isoform composition, the opportunity for isoform-specific targeting has notably come forward, but the currently available modulators fall short of expectations. In this review, we argue that with the amount of available structural and ligand data, computer-based drug design offers a number of opportunities to undertake novel and isoform-specific targeting of AMPK.


Asunto(s)
Proteínas Quinasas Activadas por AMP/química , Proteínas Quinasas Activadas por AMP/metabolismo , Diseño de Fármacos , Terapia Molecular Dirigida/métodos , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Diseño Asistido por Computadora , Activación Enzimática/efectos de los fármacos , Humanos , Resistencia a la Insulina , Ligandos , Neoplasias/metabolismo , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA