Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Sci Rep ; 11(1): 18015, 2021 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-34504172

RESUMEN

Obesity and high-fat diet (HFD) consumption result in hypothalamic inflammation and metabolic dysfunction. While the TLR4 activation by dietary fats is a well-characterized pathway involved in the neuronal and glial inflammation, the role of its accessory proteins in diet-induced hypothalamic inflammation remains unknown. Here, we demonstrate that the knockdown of TLR4-interactor with leucine-rich repeats (Tril), a functional component of TLR4, resulted in reduced hypothalamic inflammation, increased whole-body energy expenditure, improved the systemic glucose tolerance and protection from diet-induced obesity. The POMC-specific knockdown of Tril resulted in decreased body fat, decreased white adipose tissue inflammation and a trend toward increased leptin signaling in POMC neurons. Thus, Tril was identified as a new component of the complex mechanisms that promote hypothalamic dysfunction in experimental obesity and its inhibition in the hypothalamus may represent a novel target for obesity treatment.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/genética , Proteínas de la Membrana/genética , Neuronas/metabolismo , Obesidad/etiología , Proopiomelanocortina/genética , Receptor Toll-Like 4/genética , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Animales , Dieta Alta en Grasa/efectos adversos , Metabolismo Energético/genética , Regulación de la Expresión Génica , Prueba de Tolerancia a la Glucosa , Hipotálamo/patología , Inflamación , Péptidos y Proteínas de Señalización Intercelular/deficiencia , Masculino , Proteínas de la Membrana/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/patología , Obesidad/metabolismo , Obesidad/patología , Proopiomelanocortina/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Receptor Toll-Like 4/metabolismo
2.
Mol Brain ; 14(1): 65, 2021 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-33823883

RESUMEN

Palmitate is a saturated fatty acid that is well known to induce endoplasmic reticulum (ER) stress and autophagy. A high-fat diet increases the palmitate level in the hypothalamus, the main region of the brain regulating energy metabolism. Interestingly, hypothalamic palmitate level is also increased under starvation, urging the study to distinguish the effects of elevated hypothalamic palmitate level under different nutrient conditions. Herein, we show that ER-phagy (ER-targeted selective autophagy) is required for progress of ER stress and that palmitate decreases ER stress by inhibiting ER-phagy in hypothalamic cells under starvation. Palmitate inhibited starvation-induced ER-phagy by increasing the level of B-cell lymphoma 2 (Bcl-2) protein, which inhibits autophagy initiation. These findings suggest that, unlike the induction of ER stress under nutrient-rich conditions, palmitate protects hypothalamic cells from starvation-induced stress by inhibiting ER-phagy.


Asunto(s)
Proteínas Relacionadas con la Autofagia/metabolismo , Estrés del Retículo Endoplásmico/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Palmitatos/farmacología , Animales , Autofagosomas/metabolismo , Línea Celular Transformada , Medios de Cultivo/farmacología , Técnicas de Silenciamiento del Gen , Genes bcl-2 , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Interferencia de ARN , ARN Interferente Pequeño/genética , Inanición
3.
Int J Mol Sci ; 22(4)2021 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-33672787

RESUMEN

Altered lipid metabolic pathways including hydrolysis of triglycerides are key players in the pathogenesis of nonalcoholic fatty liver disease (NAFLD). Whether adiponutrin (patatin-like phospholipase domain containing protein-3-PNPLA3) and monoacylglycerol lipase (MGL) synergistically contribute to disease progression remains unclear. We generated double knockout (DKO) mice lacking both Mgl and Pnpla3; DKO mice were compared to Mgl-/- after a challenge by high-fat diet (HFD) for 12 weeks to induce steatosis. Serum biochemistry, liver transaminases as well as histology were analyzed. Fatty acid (FA) profiling was assessed in liver and adipose tissue by gas chromatography. Markers of inflammation and lipid metabolism were analyzed. Bone marrow derived macrophages (BMDMs) were isolated and treated with oleic acid. Combined deficiency of Mgl and Pnpla3 resulted in weight gain on a chow diet; when challenged by HFD, DKO mice showed increased hepatic FA synthesis and diminished beta-oxidation compared to Mgl-/-.DKO mice exhibited more pronounced hepatic steatosis with inflammation and recruitment of immune cells to the liver associated with accumulation of saturated FAs. Primary BMDMs isolated from the DKO mice showed increased inflammatory activities, which could be reversed by oleic acid supplementation. Pnpla3 deficiency aggravates the effects of Mgl deletion on steatosis and inflammation in the liver under HFD challenge.


Asunto(s)
Proteínas de la Membrana/deficiencia , Monoacilglicerol Lipasas/deficiencia , Enfermedad del Hígado Graso no Alcohólico/enzimología , Enfermedad del Hígado Graso no Alcohólico/patología , Aumento de Peso , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Animales , Células Cultivadas , Ácidos Grasos/metabolismo , Humanos , Inflamación/patología , Metabolismo de los Lípidos , Hígado/patología , Macrófagos/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Monoacilglicerol Lipasas/metabolismo , Ácido Oléico , Fenotipo , Células U937
4.
Sci Rep ; 10(1): 16947, 2020 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-33046791

RESUMEN

Protein 4.1N, a member of the protein 4.1 family, is highly expressed in the brain. But its function remains to be fully defined. Using 4.1N-/- mice, we explored the function of 4.1N in vivo. We show that 4.1N-/- mice were born at a significantly reduced Mendelian ratio and exhibited high mortality between 3 to 5 weeks of age. Live 4.1N-/- mice were smaller than 4.1N+/+ mice. Notably, while there were no significant differences in organ/body weight ratio for most of the organs, the testis/body and ovary/body ratio were dramatically decreased in 4.1N-/- mice, demonstrating selective effects of 4.1N deficiency on the development of the reproductive systems. Histopathology of the reproductive organs showed atrophy of both testis and ovary. Specifically, in the testis there is a lack of spermatogenesis, lack of leydig cells and lack of mature sperm. Similarly, in the ovary there is a lack of follicular development and lack of corpora lutea formation, as well as lack of secretory changes in the endometrium. Examination of pituitary glands revealed that the secretory granules were significantly decreased in pituitary glands of 4.1N-/- compared to 4.1N+/+. Moreover, while GnRH was expressed in both neuronal cell body and axons in the hypothalamus of 4.1N+/+ mice, it was only expressed in the cell body but not the axons of 4.1N-/- mice. Our findings uncover a novel role for 4.1N in the axis of hypothalamus-pituitary gland-reproductive system.


Asunto(s)
Proteínas del Citoesqueleto/deficiencia , Proteínas del Citoesqueleto/fisiología , Genitales/metabolismo , Genitales/patología , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/fisiología , Neuropéptidos/deficiencia , Neuropéptidos/fisiología , Sistemas Neurosecretores/metabolismo , Sistemas Neurosecretores/patología , Animales , Proteínas del Citoesqueleto/genética , Femenino , Hormona Liberadora de Gonadotropina/genética , Hormona Liberadora de Gonadotropina/metabolismo , Humanos , Hipotálamo/metabolismo , Hipotálamo/patología , Masculino , Proteínas de la Membrana/genética , Ratones Noqueados , Neuropéptidos/genética , Tamaño de los Órganos , Ovario/patología , Hipófisis/metabolismo , Hipófisis/patología , Espermatogénesis/genética , Testículo/patología
5.
Artículo en Inglés | MEDLINE | ID: mdl-32251724

RESUMEN

Iron is a vital micronutrient for all eukaryotes because it participates as a redox cofactor in multiple metabolic pathways, including lipid biosynthesis. In response to iron deficiency, the Saccharomyces cerevisiae iron-responsive transcription factor Aft1 accumulates in the nucleus and activates a set of genes that promote iron acquisition at the cell surface. In this study, we report that yeast cells lacking the transcription factor Mga2, which promotes the expression of the iron-dependent Δ9-fatty acid desaturase Ole1, display a defect in the activation of the iron regulon during the adaptation to iron limitation. Supplementation with exogenous unsaturated fatty acids (UFAs) or OLE1 expression rescues the iron regulon activation defect of mga2Δ cells. These observations and fatty acid measurements suggest that the mga2Δ defect in iron regulon expression is due to low UFA levels. Subcellular localization studies reveal that low UFAs cause a mislocalization of Aft1 protein to the vacuole upon iron deprivation that prevents its nuclear accumulation. These results indicate that Mga2 and Ole1 are essential to maintain the UFA levels required for Aft1-dependent activation of the iron regulon in response to iron deficiency, and directly connect the biosynthesis of fatty acids to the response to iron depletion.


Asunto(s)
Deficiencias de Hierro , Hierro/metabolismo , Lípidos/química , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/metabolismo , Ácidos Grasos Insaturados/metabolismo , Metabolismo de los Lípidos , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/metabolismo , Saccharomyces cerevisiae/citología , Proteínas de Saccharomyces cerevisiae/metabolismo , Estearoil-CoA Desaturasa/genética , Estearoil-CoA Desaturasa/metabolismo , Factores de Transcripción/deficiencia , Factores de Transcripción/metabolismo
6.
J Ocul Pharmacol Ther ; 36(1): 65-69, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31596637

RESUMEN

Purpose: Age-related macular degeneration (AMD) is a common disease trending towards epidemic proportions and is a leading cause of irreversible vision loss in people over the age of 65. A pathomechanism of AMD is death and/or dysfunction of retinal pigment epithelial (RPE) cells; RPE loss invariably results in photoreceptor atrophy. Treatment options for AMD are very limited, and include vitamin supplements and lifestyle changes. An exciting potential therapy currently being tested in clinical trials is transplantation of stem cell-derived RPE. Methods: We developed a NIH-registered embryonic stem line (CR-4), and in this study set out to determine if CR4-RPE are tolerated in normal mice and in murine models of retinal degeneration by injecting a bolus of CR4-RPE cells in the subretinal space of immunosuppressed wild-type, Mer mutant (Merkd), and Elovl4 deficient mice. Results: Mice with CR-RPE grafts were monitored daily, were examined routinely using OCT, and histology was prepared and examined at terminal end-points. Based on the parameters of the study, none of the animals with CR-RPE grafts (n=36) experienced any obvious adverse reactions. Conclusions: We conclude that transplanted CR-4 hES-derived RPE cells are well tolerated in immunosuppressed healthy and dystrophic murine retinas.


Asunto(s)
Células Madre Embrionarias Humanas/citología , Degeneración Macular/terapia , Epitelio Pigmentado de la Retina/citología , Animales , Modelos Animales de Enfermedad , Proteínas del Ojo/metabolismo , Humanos , Degeneración Macular/metabolismo , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados
7.
ACS Synth Biol ; 8(5): 1037-1046, 2019 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-30990999

RESUMEN

Large hydrophobic molecules, such as carotenoids, cannot be effectively excreted from cells by natural transportation systems. These products accumulate inside the cells and affect normal cellular physiological functions, which hinders further improvement of carotenoid production by microbial cell factories. In this study, we proposed to construct a novel artificial transport system utilizing membrane lipids to carry and transport hydrophobic molecules. Membrane lipids allow the physiological mechanism of membrane dispersion to be reconstructed and amplified to establish a novel artificial membrane vesicle transport system (AMVTS). Specifically, a few proteins in E. coli were reported or proposed to be related to the formation mechanism of outer membrane vesicles, and were individually knocked out or overexpressed to test their physiological functions. The effects on tolR and nlpI were the most significant. Knocking out both tolR and nlpI resulted in a 13.7% increase of secreted ß-carotene with a 35.6% increase of specific production. To supplement the loss of membrane components of the cells due to the increased membrane vesicle dispersion, the synthesis pathway of phosphatidylethanolamine was engineered. While overexpression of AccABCD and PlsBC in TW-013 led to 15% and 17% increases of secreted ß-carotene, respectively, the overexpression of both had a synergistic effect and caused a 53-fold increase of secreted ß-carotene, from 0.2 to 10.7 mg/g dry cell weight (DCW). At the same time, the specific production of ß-carotene increased from 6.9 to 21.9 mg/g DCW, a 3.2-fold increase. The AMVTS was also applied to a ß-carotene hyperproducing strain, CAR025, which led to a 24-fold increase of secreted ß-carotene, from 0.5 to 12.7 mg/g DCW, and a 61% increase of the specific production, from 27.7 to 44.8 mg/g DCW in shake flask fermentation. The AMVTS built in this study establishes a novel artificial transport mechanism different from natural protein-based cellular transport systems, which has great potential to be applied to various cell factories for the excretion of a wide range of hydrophobic compounds.


Asunto(s)
Escherichia coli/metabolismo , Ingeniería Metabólica/métodos , beta Caroteno/metabolismo , Acetil-CoA Carboxilasa/genética , Proteínas Bacterianas/genética , Corynebacterium/metabolismo , Proteínas de Escherichia coli/genética , Ácido Graso Sintasas/genética , Edición Génica , Lipoproteínas/deficiencia , Lipoproteínas/genética , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Membranas Artificiales , Fosfatidiletanolaminas/biosíntesis , Plásmidos/genética , Plásmidos/metabolismo
8.
J Cell Physiol ; 234(10): 18053-18064, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30843215

RESUMEN

The matrix remodeling associated 7 (MXRA7) gene had been ill-studied and its biology remained to be discovered. Inspired by our previous findings and public datasets concerning MXRA7, we hypothesized that the MXRA7 gene might be involved in bone marrow mesenchymal stem cells (BMSCs) functions related to bone formation, which was checked by utilizing in vivo or in vitro methodologies. Micro-computed tomography of MXRA7-deficient mice demonstrated retarded osteogenesis, which was reflected by shorter femurs, lower bone mass in both trabecular and cortical bones compared with wild-type (WT) mice. Histology confirmed the osteopenia-like feature including thinner growth plates in MXRA7-deficient femurs. Immunofluorescence revealed less osteoblasts in MXRA7-deficient femurs. Polymerase chain reaction or western blot analysis showed that when WT BMSCs were induced to differentiate toward osteoblasts or adipocytes in culture, MXRA7 messenger RNA or protein levels were significantly increased alongside osteoblasts induction, but decreased upon adipocytes induction. Cultured MXRA7-deficient BMSCs showed decreased osteogenesis upon osteogenic differentiation induction as reflected by decreased calcium deposition or lower expression of genes responsible for osteogenesis. When recombinant MXRA7 proteins were supplemented in a culture of MXRA7-deficient BMSCs, osteogenesis or gene expression was fully restored. Upon osteoblast induction, the level of active ß-catenin or phospho-extracellular signal-regulated kinase in MXRA7-deficient BMSCs was decreased compared with that in WT BMSCs, and these impairments could be rescued by recombinant MXRA7 proteins. In adipogenesis induction settings, the potency of MXRA7-deficient BMSCs to differentiate into adipocytes was increased over the WT ones. In conclusion, this study demonstrated that MXRA7 influences bone formation via regulating the balance between osteogenesis and adipogenesis in BMSCs.


Asunto(s)
Enfermedades Óseas Metabólicas/metabolismo , Diferenciación Celular , Fémur/metabolismo , Proteínas de la Membrana/metabolismo , Células Madre Mesenquimatosas/metabolismo , Osteoblastos/metabolismo , Osteogénesis , Adipocitos/metabolismo , Adipocitos/patología , Adipogénesis , Animales , Enfermedades Óseas Metabólicas/genética , Enfermedades Óseas Metabólicas/patología , Células Cultivadas , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fémur/patología , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Células Madre Mesenquimatosas/patología , Ratones Noqueados , Osteoblastos/patología , Fenotipo , Transducción de Señal , beta Catenina/metabolismo
9.
J Bacteriol ; 201(7)2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30617243

RESUMEN

CvsSR is a Ca2+-induced two-component system (TCS) in the plant pathogen Pseudomonas syringae pv. tomato DC3000. Here, we discovered that CvsSR is induced by Fe3+, Zn2+, and Cd2+ However, only supplementation of Ca2+ to medium resulted in rugose, opaque colonies in ΔcvsS and ΔcvsR strains. This phenotype corresponded to formation of calcium phosphate precipitation on the surface of ΔcvsS and ΔcvsR colonies. CvsSR regulated swarming motility in P. syringae pv. tomato in a Ca2+-dependent manner, but swarming behavior was not influenced by Fe3+, Zn2+, or Cd2+ We hypothesized that reduced swarming displayed by ΔcvsS and ΔcvsR strains was due to precipitation of calcium phosphate on the surface of ΔcvsS and ΔcvsR cells grown on agar medium supplemented with Ca2+ By reducing the initial pH or adding glucose to the medium, calcium precipitation was inhibited, and swarming was restored to ΔcvsS and ΔcvsR strains, suggesting that calcium precipitation influences swarming ability. Constitutive expression of a CvsSR-regulated carbonic anhydrase and a CvsSR-regulated putative sulfate major facilitator superfamily transporter in ΔcvsS and ΔcvsR strains inhibited formation of calcium precipitates and restored the ability of ΔcvsS and ΔcvsR bacteria to swarm. Lastly, we found that glucose inhibited Ca2+-based induction of CvsSR. Hence, CvsSR is a key regulator that controls calcium precipitation on the surface of bacterial cells.IMPORTANCE Bacteria are capable of precipitating and dissolving minerals. We previously reported the characterization of the two-component system CvsSR in the plant-pathogenic bacterium Pseudomonas syringae CvsSR responds to the presence of calcium and is important for causing disease. Here, we show that CvsSR controls the ability of the bacterium to prevent calcium phosphate precipitation on the surface of cells. We also identified a carbonic anhydrase and transporter that modulate formation of surface-associated calcium precipitates. Furthermore, our results demonstrate that the ability of the bacterium to swarm is controlled by the formation and dissolution of calcium precipitates on the surface of cells. Our study describes new mechanisms for microbially induced mineralization and provides insights into the role of mineral deposits on bacterial physiology. The discoveries may lead to new technological and environmental applications.


Asunto(s)
Fosfatos de Calcio/metabolismo , Proteínas de la Membrana/metabolismo , Pseudomonas syringae/enzimología , Pseudomonas syringae/metabolismo , Factores de Transcripción/metabolismo , Cationes Bivalentes/metabolismo , Medios de Cultivo/química , Eliminación de Gen , Glucosa/metabolismo , Concentración de Iones de Hidrógeno , Locomoción , Proteínas de la Membrana/deficiencia , Proteínas Quinasas/deficiencia , Proteínas Quinasas/metabolismo , Factores de Transcripción/deficiencia
10.
PLoS One ; 13(10): e0205878, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30379953

RESUMEN

Hutchinson-Gilford progeria syndrome (HGPS) is a very rare fatal disease characterized for accelerated aging. Although the causal agent, a point mutation in LMNA gene, was identified more than a decade ago, the molecular mechanisms underlying HGPS are still not fully understood and, currently, there is no cure for the patients, which die at a mean age of thirteen. With the aim of unraveling non-previously altered molecular pathways in the premature aging process, human cell lines from HGPS patients and from healthy parental controls were studied in parallel using Next-Generation Sequencing (RNAseq) and High-Resolution Quantitative Proteomics (iTRAQ) techniques. After selection of significant proteins and transcripts and crosschecking of the results a small set of protein/transcript pairs were chosen for validation. One of those proteins, ribose-phosphate pyrophosphokinase 1 (PRPS1), is essential for nucleotide synthesis. PRPS1 loss-of-function mutants present lower levels of purine. PRPS1 protein and transcript levels are detected as significantly decreased in HGPS cell lines vs. healthy parental controls. This modulation was orthogonally confirmed by targeted techniques in cell lines and also in an animal model of Progeria, the ZMPSTE24 knock-out mouse. In addition, functional experiments through supplementation with S-adenosyl-methionine (SAMe), a metabolite that is an alternative source of purine, were done. Results indicate that SAMe has a positive effect in the proliferative capacity and reduces senescence-associated Beta-galactosidase staining of the HPGS cell lines. Altogether, our data suggests that nucleotide and, specifically, purine-metabolism, are altered in premature aging, opening a new window for the therapeutic treatment of the disease.


Asunto(s)
Lamina Tipo A/genética , Progeria/genética , Purinas/metabolismo , ARN Mensajero/genética , Ribosa-Fosfato Pirofosfoquinasa/genética , Adulto , Animales , Línea Celular , Proliferación Celular , Niño , Biología Computacional/métodos , Modelos Animales de Enfermedad , Femenino , Efecto Fundador , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Lamina Tipo A/deficiencia , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Metaloendopeptidasas/deficiencia , Metaloendopeptidasas/genética , Ratones , Ratones Noqueados , Progeria/tratamiento farmacológico , Progeria/metabolismo , Progeria/patología , ARN Mensajero/metabolismo , Ribosa-Fosfato Pirofosfoquinasa/deficiencia , S-Adenosilmetionina/farmacología , beta-Galactosidasa/genética , beta-Galactosidasa/metabolismo
11.
Dis Model Mech ; 11(9)2018 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-30135069

RESUMEN

Niemann-Pick disease type C1 (NPC1) is a rare autosomal recessive lysosomal storage disease primarily caused by mutations in NPC1 NPC1 is characterized by abnormal accumulation of unesterified cholesterol and glycolipids in late endosomes and lysosomes. Common signs include neonatal jaundice, hepatosplenomegaly, cerebellar ataxia, seizures and cognitive decline. Both mouse and feline models of NPC1 mimic the disease progression in humans and have been used in preclinical studies of 2-hydroxypropyl-ß-cyclodextrin (2HPßCD; VTS-270), a drug that appeared to slow neurological progression in a Phase 1/2 clinical trial. However, there remains a need to identify additional therapeutic agents. High-throughput drug screens have been useful in identifying potential therapeutic compounds; however, current preclinical testing is time and labor intensive. Thus, development of a high-capacity in vivo platform suitable for screening candidate drugs/compounds would be valuable for compound optimization and prioritizing subsequent in vivo testing. Here, we generated and characterize two zebrafish npc1-null mutants using CRISPR/Cas9-mediated gene targeting. The npc1 mutants model both the early liver and later neurological disease phenotypes of NPC1. LysoTracker staining of npc1 mutant larvae was notable for intense staining of lateral line neuromasts, thus providing a robust in vivo screen for lysosomal storage. As a proof of principle, we were able to show that treatment of the npc1 mutant larvae with 2HPßCD significantly reduced neuromast LysoTracker staining. These data demonstrate the potential value of using this zebrafish NPC1 model for efficient and rapid in vivo optimization and screening of potential therapeutic compounds.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Evaluación Preclínica de Medicamentos , Enfermedad de Niemann-Pick Tipo C/tratamiento farmacológico , Pez Cebra/metabolismo , 2-Hidroxipropil-beta-Ciclodextrina/farmacología , Alelos , Animales , Secuencia de Bases , Encéfalo/patología , Colesterol/metabolismo , Modelos Animales de Enfermedad , Larva/metabolismo , Hígado/patología , Hepatopatías/patología , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/metabolismo , Mutación/genética , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Proteína Niemann-Pick C1 , Enfermedad de Niemann-Pick Tipo C/patología , Pez Cebra/crecimiento & desarrollo , Proteínas de Pez Cebra/deficiencia , Proteínas de Pez Cebra/metabolismo
12.
Neuroscience ; 374: 91-103, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29406269

RESUMEN

Wolfram syndrome is caused by mutations in the WFS1 gene. WFS1 protein dysfunction results in a range of neuroendocrine syndromes and is mostly characterized by juvenile-onset diabetes mellitus and optic atrophy. WFS1 has been shown to participate in membrane trafficking, protein processing and Ca2+ homeostasis in the endoplasmic reticulum. Aim of the present study was to find the transcriptomic changes influenced by WFS1 in the hypothalamus and hippocampus using RNA-sequencing. The WFS1-deficient mice were used as a model system to analyze the changes in transcriptional networks. The number of differentially expressed genes between hypothalami of WFS1-deficient (Wfs1KO) and wild-type (WT) mice was 43 and between hippocampi 311 with False Discovery Rate (FDR) <0.05. Avpr1a and Avpr1b were significantly upregulated in the hypothalamus and hippocampus of Wfs1KO mice respectively. Trpm8 was the most upregulated gene in the hippocampus of Wfs1KO mice. The functional analysis revealed significant enrichment of networks and pathways associated with protein synthesis, cell-to-cell signaling and interaction, molecular transport, metabolic disease and nervous system development and function. In conclusion, the transcriptomic profiles of WFS1-deficient hypothalamus and hippocampus do indicate the activation of degenerative molecular pathways causing the clinical occurrences typical to Wolfram syndrome.


Asunto(s)
Hipocampo/metabolismo , Hipotálamo/metabolismo , Proteínas de la Membrana/deficiencia , Síndrome de Wolfram/metabolismo , Animales , Modelos Animales de Enfermedad , Masculino , Proteínas de la Membrana/genética , Ratones de la Cepa 129 , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Vasopresinas/metabolismo , Análisis de Secuencia de ARN , Canales Catiónicos TRPM/metabolismo , Transcriptoma
13.
Am J Physiol Renal Physiol ; 314(5): F702-F714, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28515173

RESUMEN

Ferroptosis is an iron-dependent form of regulated nonapoptotic cell death, which contributes to damage in models of acute kidney injury (AKI). Heme oxygenase-1 (HO-1) is a cytoprotective enzyme induced in response to cellular stress, and is protective against AKI because of its antiapoptotic and anti-inflammatory properties. However, the role of HO-1 in regulating ferroptosis is unclear. The purpose of this study was to elucidate the role of HO-1 in regulating ferroptotic cell death in renal proximal tubule cells (PTCs). Immortalized PTCs obtained from HO-1+/+ and HO-1-/- mice were treated with erastin or RSL3, ferroptosis inducers, in the presence or absence of antioxidants, an iron source, or an iron chelator. Cells were assessed for changes in morphology and metabolic activity as an indicator of cell viability. Treatment of HO-1+/+ PTCs with erastin resulted in a time- and dose-dependent increase in HO-1 gene expression and protein levels compared with vehicle-treated controls. HO-1-/- cells showed increased dose-dependent erastin- or RSL3-induced cell death in comparison to HO-1+/+ PTCs. Iron supplementation with ferric ammonium citrate in erastin-treated cells decreased cell viability further in HO-1-/- PTCs compared with HO-1+/+ cells. Cotreatment with ferrostatin-1 (ferroptosis inhibitor), deferoxamine (iron chelator), or N-acetyl-l-cysteine (glutathione replenisher) significantly increased cell viability and attenuated erastin-induced ferroptosis in both HO-1+/+ and HO-1-/- PTCs. These results demonstrate an important antiferroptotic role of HO-1 in renal epithelial cells.


Asunto(s)
Lesión Renal Aguda/enzimología , Hemo-Oxigenasa 1/metabolismo , Túbulos Renales Proximales/enzimología , Proteínas de la Membrana/metabolismo , Acetilcisteína/farmacología , Lesión Renal Aguda/genética , Lesión Renal Aguda/patología , Lesión Renal Aguda/prevención & control , Animales , Antioxidantes/farmacología , Carbolinas/toxicidad , Muerte Celular , Línea Celular , Ciclohexilaminas/farmacología , Deferoxamina/farmacología , Relación Dosis-Respuesta a Droga , Compuestos Férricos/toxicidad , Glutatión/metabolismo , Hemo-Oxigenasa 1/deficiencia , Hemo-Oxigenasa 1/genética , Quelantes del Hierro/farmacología , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/patología , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones Noqueados , Fenilendiaminas/farmacología , Piperazinas/toxicidad , Compuestos de Amonio Cuaternario/toxicidad , Transducción de Señal , Factores de Tiempo
14.
eNeuro ; 4(5)2017.
Artículo en Inglés | MEDLINE | ID: mdl-28966979

RESUMEN

GABA released from presynaptic sites induces short-lived phasic inhibition mediated by synaptic GABAA receptors (GABAARs) and longer-duration tonic inhibition mediated by extrasynaptic GABAA or GABAB receptors (GABABRs). A number of studies have found that contactin-associated protein 2 (Cntnap2) knockout (KO) mice, a well-established mouse model of autism, exhibit reduced interneuron numbers and aberrant phasic inhibition. However, little is known about whether tonic inhibition is disrupted in Cntnap2 KO mice and when the disruption of inhibition begins to occur during postnatal development. We examined tonic and phasic inhibition in layer 2/3 pyramidal cells of primary visual cortex of Cntnap2 KO at two different developmental stages, three to four and six to eight weeks of age. We found that both phasic inhibition and GABAAR but not GABABR-mediated tonic inhibition was reduced in pyramidal cells from six- to eight-week-old Cntnap2 KO mice, while in three- to four-week-old mice, no significant effects of genotype on tonic or phasic inhibition was observed. We further found that activation of tonic currents mediated by δ-subunit-containing GABAARs reduced neural excitability, an effect that was attenuated by loss of Cntnap2. While the relative contribution of tonic versus phasic inhibition to autism-related symptoms remains unclear, our data suggest that reduced tonic inhibition may play an important role, and δ-subunit-containing GABAARs may be a useful target for therapeutic intervention in autism.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/genética , Proteínas de la Membrana/deficiencia , Proteínas del Tejido Nervioso/deficiencia , Inhibición Neural/genética , Células Piramidales/fisiología , Receptores de GABA-A/metabolismo , Corteza Visual/citología , Animales , Animales Recién Nacidos , Estimulación Eléctrica , Femenino , GABAérgicos/farmacología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Técnicas In Vitro , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Potenciales Postsinápticos Inhibidores/genética , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Corteza Visual/crecimiento & desarrollo , Ácido gamma-Aminobutírico/farmacología
15.
J Clin Endocrinol Metab ; 102(4): 1375-1386, 2017 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-28323990

RESUMEN

CONTEXT: TMEM165 deficiency is a severe multisystem disease that manifests with metabolic, endocrine, and skeletal involvement. It leads to one type of congenital disorders of glycosylation (CDG), a rapidly growing group of inherited diseases in which the glycosylation process is altered. Patients have decreased galactosylation by serum glycan analysis. There are >100 CDGs, but only specific types are treatable. OBJECTIVE: Galactose has been shown to be beneficial in other CDG types with abnormal galactosylation. The aim of this study was to characterize the effects of galactose supplementation on Golgi glycosylation in TMEM165-depleted HEK293 cells, as well as in 2 patients with TMEM165-CDG and in their cultured skin fibroblast cells. DESIGN AND SETTING: Glycosylation was assessed by mass spectrometry, western blot analysis, and transferrin isoelectrofocusing. PATIENTS AND INTERVENTIONS: Both unrelated patients with TMEM165-CDG with the same deep intronic homozygous mutation (c.792+182G>A) were allocated to receive d-galactose in a daily dose of 1 g/kg. RESULTS: We analyzed N-linked glycans and glycolipids in knockout TMEM165 HEK293 cells, revealing severe hypogalactosylation and GalNAc transfer defects. Although these defects were completely corrected by the addition of Mn2+, we demonstrated that the observed N-glycosylation defect could also be overcome by galactose supplementation. We then demonstrated that oral galactose supplementation in patients with TMEM165-deficient CDG improved biochemical and clinical parameters, including a substantial increase in the negatively charged transferrin isoforms, and a decrease in hypogalactosylated total N-glycan structures, endocrine function, and coagulation parameters. CONCLUSION: To our knowledge, this is the first description of abnormal glycosylation of lipids in the TMEM165 defect and the first report of successful dietary treatment in TMEM165 deficiency. We recommend the use of oral d-galactose therapy in TMEM165-CDG.


Asunto(s)
Trastornos Congénitos de Glicosilación/dietoterapia , Trastornos Congénitos de Glicosilación/genética , Galactosa/farmacología , Galactosa/uso terapéutico , Glicosilación/efectos de los fármacos , Proteínas de la Membrana/genética , Adulto , Antiportadores , Proteínas de Transporte de Catión , Niño , Trastornos Congénitos de Glicosilación/patología , Suplementos Dietéticos , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Células HEK293 , Humanos , Masculino , Proteínas de la Membrana/deficiencia , Mutación , Resultado del Tratamiento
16.
J Med Genet ; 54(5): 330-337, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27799409

RESUMEN

BACKGROUND: Acyl-CoA binding domain containing protein 5 (ACBD5) is a peroxisomal membrane protein with a cytosolic acyl-CoA binding domain. Because of its acyl-CoA binding domain, ACBD5 has been assumed to function as an intracellular carrier of acyl-CoA esters. In addition, a role for ACBD5 in pexophagy has been suggested. However, the precise role of ACBD5 in peroxisomal metabolism and/or functioning has not yet been established. Previously, a genetic ACBD5 deficiency was identified in three siblings with retinal dystrophy and white matter disease. We identified a pathogenic mutation in ACBD5 in another patient and studied the consequences of the ACBD5 defect in patient material and in ACBD5-deficient HeLa cells to uncover this role. METHODS: We studied a girl who presented with progressive leukodystrophy, syndromic cleft palate, ataxia and retinal dystrophy. We performed biochemical, cell biological and molecular studies in patient material and in ACBD5-deficient HeLa cells generated by CRISPR-Cas9 genome editing. RESULTS: We identified a homozygous deleterious indel mutation in ACBD5, leading to complete loss of ACBD5 protein in the patient. Our studies showed that ACBD5 deficiency leads to accumulation of very long-chain fatty acids (VLCFAs) due to impaired peroxisomal ß-oxidation. No effect on pexophagy was found. CONCLUSIONS: Our investigations strongly suggest that ACBD5 plays an important role in sequestering C26-CoA in the cytosol and thereby facilitates transport into the peroxisome and subsequent ß-oxidation. Accordingly, ACBD5 deficiency is a novel single peroxisomal enzyme deficiency caused by impaired VLCFA metabolism, leading to retinal dystrophy and white matter disease.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/deficiencia , Ácidos Grasos/metabolismo , Proteínas de la Membrana/deficiencia , Peroxisomas/metabolismo , Acilcoenzima A/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Autofagia , Preescolar , ADN Complementario/genética , Femenino , Fibroblastos/metabolismo , Prueba de Complementación Genética , Células HeLa , Humanos , Lactante , Imagen por Resonancia Magnética , Proteínas de la Membrana/metabolismo , Piel/patología
17.
Hum Mol Genet ; 25(8): 1489-500, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-27008884

RESUMEN

Congenital disorders of glycosylation (CDG) are severe inherited diseases in which aberrant protein glycosylation is a hallmark. From this genetically and clinically heterogenous group, a significant subgroup due to Golgi homeostasis defects is emerging. We previously identified TMEM165 as a Golgi protein involved in CDG. Extremely conserved in the eukaryotic reign, the molecular mechanism by which TMEM165 deficiencies lead to Golgi glycosylation abnormalities is enigmatic. AsGDT1 is the ortholog of TMEM165 in yeast, both gdt1Δ null mutant yeasts and TMEM165 depleted cells were used. We highlighted that the observed Golgi glycosylation defects due to Gdt1p/TMEM165 deficiency result from Golgi manganese homeostasis defect. We discovered that in both yeasts and mammalian Gdt1p/TMEM165-deficient cells, Mn(2+) supplementation could restore a normal glycosylation. We also showed that the GPP130 Mn(2+) sensitivity was altered in TMEM165 depleted cells. This study not only provides novel insights into the molecular causes of glycosylation defects observed in TMEM165-deficient cells but also suggest that TMEM165 is a key determinant for the regulation of Golgi Mn(2+) homeostasis.


Asunto(s)
Proteínas Fúngicas/genética , Aparato de Golgi/fisiología , Manganeso/farmacología , Proteínas de la Membrana/deficiencia , Mutación , Antiportadores , Proteínas de Transporte de Catión , Trastornos Congénitos de Glicosilación/genética , Proteínas Fúngicas/metabolismo , Glicosilación/efectos de los fármacos , Aparato de Golgi/efectos de los fármacos , Aparato de Golgi/metabolismo , Células HEK293 , Células HeLa , Homeostasis , Humanos , Manganeso/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas de Transporte Vesicular/metabolismo
18.
Neuropediatrics ; 47(3): 179-81, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26947510

RESUMEN

Peroxisome biogenesis disorders (PBD) are a heterogeneous group of disorders due to PEX genes mutations, with a broad clinical spectrum comprising severe neonatal disease to mild presentation. Recently, Berendse et al reported an improvement of peroxisomal functions with l-arginine supplementation in fibroblasts with specific mutations of PEX1, PEX6, and PEX12. We report the first treatment by l-arginine in a patient homozygous for the specific PEX12 mutation shown to be l-arginine responsive in fibroblasts. We described the effect of l-arginine on biochemical (decrease of some plasma peroxisomal parameters) and neurophysiological (improvement of deafness) parameters. Some subjective clinical effects have also been observed (no more sialorrhea, behavior improvement). More studies are needed to assess the efficacy of l-arginine in some PBD patients with specific mutations.


Asunto(s)
Arginina/uso terapéutico , Proteínas de la Membrana/genética , Trastorno Peroxisomal/tratamiento farmacológico , Alanina Transaminasa/sangre , Aspartato Aminotransferasas/sangre , Niño , Preescolar , Sordera/etiología , Discapacidades del Desarrollo/etiología , Ácidos Grasos/sangre , Femenino , Humanos , Lactante , Proteínas de la Membrana/deficiencia , Hipotonía Muscular/etiología , Trastorno Peroxisomal/sangre , Trastorno Peroxisomal/complicaciones , Trastorno Peroxisomal/genética , Ácido Fitánico/sangre , Ácidos Pipecólicos/sangre , Sialorrea/etiología
19.
Neurobiol Dis ; 85: 81-92, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26459112

RESUMEN

Absence seizures occur in several types of human epilepsy and result from widespread, synchronous feedback between the cortex and thalamus that produces brief episodes of loss of consciousness. Genetic rodent models have been invaluable for investigating the pathophysiological basis of these seizures. Here, we identify tetratricopeptide-containing Rab8b-interacting protein (TRIP8b) knockout mice as a new model of absence epilepsy, featuring spontaneous spike-wave discharges on electroencephalography (EEG) that are the electrographic hallmark of absence seizures. TRIP8b is an auxiliary subunit of the hyperpolarization-activated cyclic-nucleotide-gated (HCN) channels, which have previously been implicated in the pathogenesis of absence seizures. In contrast to mice lacking the pore-forming HCN channel subunit HCN2, TRIP8b knockout mice exhibited normal cardiac and motor function and a less severe seizure phenotype. Evaluating the circuit that underlies absence seizures, we found that TRIP8b knockout mice had significantly reduced HCN channel expression and function in thalamic-projecting cortical layer 5b neurons and thalamic relay neurons, but preserved function in inhibitory neurons of the reticular thalamic nucleus. Our results expand the known roles of TRIP8b and provide new insight into the region-specific functions of TRIP8b and HCN channels in constraining cortico-thalamo-cortical excitability.


Asunto(s)
Corteza Cerebral/fisiopatología , Epilepsia Tipo Ausencia/fisiopatología , Proteínas de la Membrana/deficiencia , Neuronas/fisiología , Tálamo/fisiopatología , Animales , Western Blotting , Modelos Animales de Enfermedad , Electrocardiografía , Electrocorticografía , Electrodos Implantados , Epilepsia Tipo Ausencia/genética , Inmunohistoquímica , Masculino , Potenciales de la Membrana/fisiología , Proteínas de la Membrana/genética , Ratones Noqueados , Actividad Motora/fisiología , Técnicas de Placa-Clamp , Peroxinas , Prueba de Desempeño de Rotación con Aceleración Constante , Eliminación de Secuencia , Técnicas de Cultivo de Tejidos
20.
Behav Neurosci ; 129(6): 731-43, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26501174

RESUMEN

Genetic epidemiological studies support a role for CNTNAP2 in developmental language disorders such as autism spectrum disorder, specific language impairment, and dyslexia. Atypical language development and function represent a core symptom of autism spectrum disorder (ASD), with evidence suggesting that aberrant auditory processing-including impaired spectrotemporal processing and enhanced pitch perception-may both contribute to an anomalous language phenotype. Investigation of gene-brain-behavior relationships in social and repetitive ASD symptomatology have benefited from experimentation on the Cntnap2 knockout (KO) mouse. However, auditory-processing behavior and effects on neural structures within the central auditory pathway have not been assessed in this model. Thus, this study examined whether auditory-processing abnormalities were associated with mutation of the Cntnap2 gene in mice. Cntnap2 KO mice were assessed on auditory-processing tasks including silent gap detection, embedded tone detection, and pitch discrimination. Cntnap2 knockout mice showed deficits in silent gap detection but a surprising superiority in pitch-related discrimination as compared with controls. Stereological analysis revealed a reduction in the number and density of neurons, as well as a shift in neuronal size distribution toward smaller neurons, in the medial geniculate nucleus of mutant mice. These findings are consistent with a central role for CNTNAP2 in the ontogeny and function of neural systems subserving auditory processing and suggest that developmental disruption of these neural systems could contribute to the atypical language phenotype seen in autism spectrum disorder.


Asunto(s)
Percepción Auditiva/fisiología , Cuerpos Geniculados/patología , Cuerpos Geniculados/fisiopatología , Proteínas de la Membrana/deficiencia , Proteínas del Tejido Nervioso/deficiencia , Estimulación Acústica , Animales , Vías Auditivas/patología , Vías Auditivas/fisiopatología , Trastorno del Espectro Autista , Recuento de Células , Estudios de Cohortes , Pruebas Auditivas , Masculino , Proteínas de la Membrana/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Proteínas del Tejido Nervioso/genética , Neuronas/patología , Reflejo de Sobresalto/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA