Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 13(1): 7690, 2022 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-36509758

RESUMEN

The brain is a major sanctuary site for metastatic cancer cells that evade systemic therapies. Through pre-clinical pharmacological, biological, and molecular studies, we characterize the functional link between drug resistance and central nervous system (CNS) relapse in Epidermal Growth Factor Receptor- (EGFR-) mutant non-small cell lung cancer, which can progress in the brain when treated with the CNS-penetrant EGFR inhibitor osimertinib. Despite widespread osimertinib distribution in vivo, the brain microvascular tumor microenvironment (TME) is associated with the persistence of malignant cell sub-populations, which are poised to proliferate in the brain as osimertinib-resistant lesions over time. Cellular and molecular features of this poised state are regulated through a Ras homolog family member A (RhoA) and Serum Responsive Factor (SRF) gene expression program. RhoA potentiates the outgrowth of disseminated tumor cells on osimertinib treatment, preferentially in response to extracellular laminin and in the brain. Thus, we identify pre-existing and adaptive features of metastatic and drug-resistant cancer cells, which are enhanced by RhoA/SRF signaling and the brain TME during the evolution of osimertinib-resistant disease.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Proteína de Unión al GTP rhoA/genética , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Receptores ErbB/genética , Compuestos de Anilina/farmacología , Compuestos de Anilina/uso terapéutico , Encéfalo/patología , Mutación , Resistencia a Antineoplásicos/genética , Microambiente Tumoral
2.
Nutrition ; 89: 111291, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34111672

RESUMEN

OBJECTIVE: The aim of this study was to quantify the bioaccessibility of phosphorus from amino acid-based formulas (AAFs) under different digestive conditions. METHODS: We developed in-vitro batch digestion models with stomach digestion at different pH mimicking the normal digestive condition and conditions representing use of acid-suppressive medication. To validate bioaccessibility findings, we devised a low phosphorus murine model to test phosphorus bioavailability under compromised digestive conditions using proton pump inhibitors (PPIs) to neutralize stomach pH. RESULTS: In vitro phosphorus bioaccessibility of AAFs Neocate® Infant and Neocate Junior ranged between 57% and 65% under normal digestive conditions for infants (stomach pH 3.5) and between 38% and 46% under conditions that simulated bypass of stomach acidification, which is comparable to control diet and two EleCare® AAFs. In vivo bioavailability analysis showed that both Neocate formulas were able to normalize plasma phosphorus levels when administered to low phosphorus mice along with PPIs (control diet + PPI 8 ± 0.4; Neocate Infant 10.1 ± 0.9; Neocate Junior 9.2 ± 0.6; EleCare Infant 8.6 ± 0.4; EleCare Junior 8.7 ± 0.5; n = 8-10; P < 0.0001 versus baseline 3.4 ± 0.2 mg/dL). In comparison, plasma phosphorus levels remained lower on the low phosphorus diet (5.7 ± 0.2 mg/dL). Furthermore, urinary phosphorus/creatinine and intact fibroblast growth factor 23 were significantly lowered by low phosphorus diet. In contrast, intact parathyroid hormone and 1,25-dihydroxy vitamin D decreased and increased, respectively, and these parameters likewise normalized in mice administered AAFs. CONCLUSION: The present findings indicated that phosphorus bioaccessibility in the in-vitro batch digestion model translates well into phosphorus bioavailability in mice even under compromised digestive conditions that bypass gastric acidification.


Asunto(s)
Fósforo , Estómago , Aminoácidos , Animales , Disponibilidad Biológica , Digestión , Ratones
3.
Sci Rep ; 10(1): 3069, 2020 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-32080237

RESUMEN

Low blood phosphate (Pi) reduces muscle function in hypophosphatemic disorders. Which Pi transporters are required and whether hormonal changes due to hypophosphatemia contribute to muscle function is unknown. To address these questions we generated a series of conditional knockout mice lacking one or both house-keeping Pi transporters Pit1 and Pit2 in skeletal muscle (sm), using the postnatally expressed human skeletal actin-cre. Simultaneous conditional deletion of both transporters caused skeletal muscle atrophy, resulting in death by postnatal day P13. smPit1-/-, smPit2-/- and three allele mutants are fertile and have normal body weights, suggesting a high degree of redundance for the two transporters in skeletal muscle. However, these mice show a gene-dose dependent reduction in running activity also seen in another hypophosphatemic model (Hyp mice). In contrast to Hyp mice, grip strength is preserved. Further evaluation of the mechanism shows reduced ERK1/2 activation and stimulation of AMP kinase in skeletal muscle from smPit1-/-; smPit2-/- mice consistent with energy-stress. Similarly, C2C12 myoblasts show a reduced oxygen consumption rate mediated by Pi transport-dependent and ERK1/2-dependent metabolic Pi sensing pathways. In conclusion, we here show that Pit1 and Pit2 are essential for normal myofiber function and survival, insights which may improve management of hypophosphatemic myopathy.


Asunto(s)
Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo III/metabolismo , Factor de Transcripción Pit-1/metabolismo , Alelos , Animales , Línea Celular , Supervivencia Celular , Transporte de Electrón , Metabolismo Energético , Fuerza de la Mano , Ratones Noqueados , Modelos Biológicos , Células Musculares/metabolismo , Necrosis , Consumo de Oxígeno , Fosfatos/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo III/deficiencia , Factor de Transcripción Pit-1/deficiencia
4.
PLoS One ; 14(10): e0223052, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31613887

RESUMEN

To further investigate the role of the phosphate (Pi) transporter PIT1 in Pi homeostasis and tissue mineralization, we developed a transgenic mouse expressing the C-terminal influenza hemagglutinin (HA) epitope-tagged human PIT1 transporter under control of the cytomegalovirus/chicken beta actin/rabbit beta-globin gene (CAG) promotor and a loxP-stop-loxP (LSL) cassette permitting conditional activation of transgene expression (LSL-HA-hPITtg/+). For an initial characterization of this conditional mouse model, germline excision of the LSL cassette was performed to induce expression of the transgene in all mouse tissues (HA-hPIT1tg/+). Recombination was confirmed using genomic DNA obtained from blood samples of these mice. Furthermore, expression of HA-hPIT1 was found to be at least 10-fold above endogenous mouse Pit1 in total RNA isolated from multiple tissues and from cultured primary calvaria osteoblasts (PCOB) estimated by semi-quantitative RT-PCR. Robust expression of the HA-hPIT1 protein was also observed upon immunoblot analysis in most tissues and permits HA-mediated immunoprecipitation of the transporter. Characterization of the phenotype of HA-hPIT1tg/+ mice at 80 days of age when fed a standard chow (0.7% Pi and 1% calcium) showed elevated plasma Pi, but normal plasma iPTH, iFGF23, serum calcium, BUN, 1,25-dihydroxy vitamin D levels and urine Pi, calcium and protein excretion when compared to WT littermates. Likewise, no change in bone mineral density was observed upon uCT analysis of the distal femur obtained from these mice. In conclusion, heterozygous overexpression of HA-hPIT1 is compatible with life and causes hyperphosphatemia while bone and mineral metabolism of these mice are otherwise normal.


Asunto(s)
Efecto Fundador , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Ratones Transgénicos/genética , Fosfatos/metabolismo , Factor de Transcripción Pit-1/genética , Transgenes , Actinas/genética , Actinas/metabolismo , Animales , Transporte Biológico , Densidad Ósea , Calcitriol/sangre , Pollos , Citomegalovirus/genética , Citomegalovirus/metabolismo , Femenino , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Humanos , Masculino , Ratones , Ratones Transgénicos/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo , Hormona Paratiroidea/genética , Hormona Paratiroidea/metabolismo , Cultivo Primario de Células , Regiones Promotoras Genéticas , Conejos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Cráneo/citología , Cráneo/metabolismo , Factor de Transcripción Pit-1/metabolismo , Globinas beta/genética , Globinas beta/metabolismo
5.
Nat Rev Endocrinol ; 14(11): 637-655, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30218014

RESUMEN

Inorganic phosphate (Pi) is essential for signal transduction and cell metabolism, and is also an essential structural component of the extracellular matrix of the skeleton. Pi is sensed in bacteria and yeast at the plasma membrane, which activates intracellular signal transduction to control the expression of Pi transporters and other genes that control intracellular Pi levels. In multicellular organisms, Pi homeostasis must be maintained in the organism and at the cellular level, requiring an endocrine and metabolic Pi-sensing mechanism, about which little is currently known. This Review will discuss the metabolic effects of Pi, which are mediated by Pi transporters, inositol pyrophosphates and SYG1-Pho81-XPR1 (SPX)-domain proteins to maintain cellular phosphate homeostasis in the musculoskeletal system. In addition, we will discuss how Pi is sensed by the human body to regulate the production of fibroblast growth factor 23 (FGF23), parathyroid hormone and calcitriol to maintain serum levels of Pi in a narrow range. New findings on the crosstalk between iron and Pi homeostasis in the regulation of FGF23 expression will also be outlined. Mutations in components of these metabolic and endocrine phosphate sensors result in genetic disorders of phosphate homeostasis, cardiomyopathy and familial basal ganglial calcifications, highlighting the importance of this newly emerging area of research.


Asunto(s)
Huesos/metabolismo , Factores de Crecimiento de Fibroblastos/genética , Homeostasis/genética , Minerales/metabolismo , Fosfatos/metabolismo , Animales , Factor-23 de Crecimiento de Fibroblastos , Marcadores Genéticos/genética , Humanos , Ratones , Sistema Musculoesquelético/metabolismo , Sensibilidad y Especificidad , Transducción de Señal/genética , Receptor de Retrovirus Xenotrópico y Politrópico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...