Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 521
Filtrar
1.
J Biol Chem ; 297(5): 101312, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34673028

RESUMEN

Mammalian spermatogenesis is a highly coordinated process that requires cooperation between specific proteins to coordinate diverse biological functions. For example, mouse Parkin coregulated gene (PACRG) recruits meiosis-expressed gene 1 (MEIG1) to the manchette during normal spermiogenesis. Here we mutated Y68 of MEIG1 using the CRISPR/cas9 system and examined the biological and physiological consequences in mice. All homozygous mutant males examined were completely infertile, and sperm count was dramatically reduced. The few developed sperm were immotile and displayed multiple abnormalities. Histological staining showed impaired spermiogenesis in these mutant mice. Immunofluorescent staining further revealed that this mutant MEIG1 was still present in the cell body of spermatocytes, but also that more MEIG1 accumulated in the acrosome region of round spermatids. The mutant MEIG1 and a cargo protein of the MEIG1/PACRG complex, sperm-associated antigen 16L (SPAG16L), were no longer found to be present in the manchette; however, localization of the PACRG component was not changed in the mutants. These findings demonstrate that Y68 of MEIG1 is a key amino acid required for PACRG to recruit MEIG1 to the manchette to transport cargo proteins during sperm flagella formation. Given that MEIG1 and PACRG are conserved in humans, small molecules that block MEIG1/PACRG interaction are likely ideal targets for the development of male contraconception drugs.


Asunto(s)
Acrosoma/metabolismo , Proteínas de Ciclo Celular/metabolismo , Mutación Missense , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Espermatocitos/metabolismo , Sustitución de Aminoácidos , Animales , Transporte Biológico Activo/genética , Proteínas de Ciclo Celular/genética , Masculino , Ratones , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Proteínas Nucleares/genética , Fosfoproteínas/genética
2.
Drug Metab Dispos ; 49(8): 629-637, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34074729

RESUMEN

Exposure to the environmental pollutant cadmium is ubiquitous, as it is present in cigarette smoke and the food supply. Over time, cadmium enters and accumulates in the kidneys, where it causes tubular injury. The breast cancer resistance protein (BCRP, ATP-Binding Cassette G2 ABCG2) is an efflux transporter that mediates the urinary secretion of pharmaceuticals and toxins. The ABCG2 genetic variant Q141K exhibits altered membrane trafficking that results in reduced efflux of BCRP substrates. Here, we sought to 1) evaluate the in vitro and in vivo ability of BCRP to transport cadmium and protect kidney cells from toxicity and 2) determine whether this protection is impaired by the Q141K variant. Cadmium concentrations, cellular stress, and toxicity were quantified in human embryonic kidney 293 cells expressing an empty vector (EV), BCRP wild-type (WT), or variant (Q141K) gene. Treatment with CdCl2 resulted in greater accumulation of cadmium and apoptosis in EV cells relative to WT cells. Exposure to CdCl2 induced expression of stress-related genes and proteins including MT-1A/MT-2A, NAD(P)H quinone dehydrogenase 1, and heme oxygenase-1 to a higher extent in EV cells compared with WT cells. Notably, the Q141K variant protected against CdCl2-induced activation of stress genes and cytotoxicity, but this protection was to a lesser magnitude than observed with WT BCRP. Lastly, concentrations of cadmium in the kidneys of Bcrp knockout mice were 40% higher than in WT mice, confirming that cadmium is an in vivo substrate of BCRP. In conclusion, BCRP prevents the accumulation of cadmium and protects against toxicity, a response that is impaired by the Q141K variant. SIGNIFICANCE STATEMENT: The breast cancer resistance protein transporter lowers cellular accumulation of the toxic heavy metal cadmium. This protective function is partially attenuated by the Q141K genetic variant in the ABCG2 gene.


Asunto(s)
Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Cadmio/farmacocinética , Riñón , Proteínas de Neoplasias , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Animales , Apoptosis/efectos de los fármacos , Transporte Biológico Activo/genética , Cadmio/toxicidad , Células HEK293 , Humanos , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/fisiopatología , Ratones , Ratones Noqueados , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Polimorfismo de Nucleótido Simple , Factores Protectores , Eliminación Renal/fisiología
3.
Crit Rev Biochem Mol Biol ; 56(4): 426-439, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34182846

RESUMEN

Plasma levels of high-density lipoprotein (HDL) inversely correlate with the incidence of cardiovascular diseases (CVD). The causal relationship between plasma HDL-cholesterol levels and CVD has been called into question by Mendelian randomization studies and the majority of clinical trials not showing any benefit of plasma HDL-cholesterol raising drugs on CVD. Nonetheless, recent Mendelian randomization studies including an increased number of CVD cases compared to earlier studies have confirmed that HDL-cholesterol levels and CVD are causally linked. Moreover, several studies in large population cohorts have shown that the cholesterol efflux capacity of HDL inversely correlates with CVD. Cholesterol efflux pathways exert anti-inflammatory and anti-atherogenic effects by suppressing proliferation of hematopoietic stem and progenitor cells, and inflammation and inflammasome activation in macrophages. Cholesterol efflux pathways also suppress the accumulation of cholesteryl esters in macrophages, i.e. macrophage foam cell formation. Recent single-cell RNASeq studies on atherosclerotic plaques have suggested that macrophage foam cells have lower expression of inflammatory genes than non-foam cells, probably reflecting liver X receptor activation, upregulation of ATP Binding Cassette A1 and G1 cholesterol transporters and suppression of inflammation. However, when these pathways are defective lesional foam cells may become pro-inflammatory.


Asunto(s)
Aterosclerosis/metabolismo , HDL-Colesterol/metabolismo , Regulación de la Expresión Génica , RNA-Seq , Análisis de la Célula Individual , Animales , Aterosclerosis/genética , Aterosclerosis/patología , Transporte Biológico Activo/genética , Proliferación Celular , HDL-Colesterol/genética , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Humanos , Inflamasomas , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Receptores X del Hígado/genética , Receptores X del Hígado/metabolismo , Análisis de la Aleatorización Mendeliana
4.
Curr Drug Metab ; 22(9): 709-725, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33992050

RESUMEN

BACKGROUND: Hypoxia has a negative effect on the cardiovascular system, nervous system, and metabolism, which contributes to potential changes in drug absorption, distribution, metabolism, and excretion (ADME). However, hypoxia can also alter the expression of microRNA (miRNA), thereby regulating drug-metabolizing enzymes, transporters, and ADME genes, such as hypoxia-inducible factor, inflammatory cytokine, nuclear receptor, etc. Therefore, it is crucial to study the role of miRNA in the regulation of drug-metabolizing enzymes and transporters under hypoxia. METHODS: A systematic review of published studies was carried out to investigate the role of miRNA in the regulation of drug-metabolizing enzymes and transporters under hypoxia. Data and information on expression changes in miRNA, drug-metabolizing enzymes, and transporters under hypoxia were analyzed and summarized. RESULTS: Hypoxia can up or down-regulate the expression of miRNA. The effect of hypoxia on Cytochrome P450 (CYP450) is still a subject of debate. The widespread belief is that hypoxia decreased the activity and expression of CYP1A1, CYP1A2, CYP2E1, and CYP3A1 and increased those of CYP3A6 and CYP2D1 in rats. Hypoxia increased the expression of a multidrug resistance-associated protein, breast cancer resistance protein, peptide transporter, organic cation transporter, and organic anion transporter. miRNA negatively regulated the expression of drugmetabolizing enzymes and transporters. CONCLUSION: The findings of this review indicated that miRNA plays a key role in the expression changes of drugmetabolizing enzymes and transporters under hypoxia.


Asunto(s)
Biotransformación , Regulación de la Expresión Génica , Hipoxia , MicroARNs/metabolismo , Transporte Biológico Activo/genética , Humanos , Hipoxia/enzimología , Hipoxia/metabolismo , Inactivación Metabólica/genética
5.
Mol Microbiol ; 116(2): 416-426, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33772889

RESUMEN

In the process of natural transformation bacteria import extracellular DNA molecules for integration into their genome. One strand of the incoming DNA molecule is degraded, whereas the remaining strand is transported across the cytoplasmic membrane. The DNA transport channel is provided by the protein ComEC. Many ComEC proteins have an extracellular C-terminal domain (CTD) with homology to the metallo-ß-lactamase fold. Here we show that this CTD binds Mn2+ ions and exhibits Mn2+ -dependent phosphodiesterase and nuclease activities. Inactivation of the enzymatic activity of the CTD severely inhibits natural transformation in Bacillus subtilis. These data suggest that the ComEC CTD is a nuclease responsible for degrading the nontransforming DNA strand during natural transformation and that this process is important for efficient DNA import.


Asunto(s)
Bacillus subtilis/genética , Proteínas Bacterianas/metabolismo , Transporte Biológico Activo/fisiología , Desoxirribonucleasas/metabolismo , Complejos Multienzimáticos/metabolismo , Transformación Bacteriana/genética , Proteínas Bacterianas/genética , Transporte Biológico Activo/genética , Competencia de la Transformación por ADN/genética , Complejos Multienzimáticos/genética , Hidrolasas Diéster Fosfóricas/metabolismo
6.
Int J Mol Sci ; 22(5)2021 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-33669068

RESUMEN

Tunneling nanotubes (TNTs) are recognized long membrane nanotubes connecting distance cells. In the last decade, growing evidence has shown that these subcellular structures mediate the specific transfer of cellular materials, pathogens, and electrical signals between cells. As intercellular bridges, they play a unique role in embryonic development, collective cell migration, injured cell recovery, cancer treatment resistance, and pathogen propagation. Although TNTs have been considered as potential drug targets for treatment, there is still a long way to go to translate the research findings into clinical practice. Herein, we emphasize the heterogeneous nature of TNTs by systemically summarizing the current knowledge on their morphology, structure, and biogenesis in different types of cells. Furthermore, we address the communication efficiency and biological outcomes of TNT-dependent transport related to diseases. Finally, we discuss the opportunities and challenges of TNTs as an exciting therapeutic approach by focusing on the development of efficient and safe drugs targeting TNTs.


Asunto(s)
Comunicación Celular/efectos de los fármacos , Desarrollo de Medicamentos/métodos , Uniones Intercelulares/metabolismo , Neoplasias/metabolismo , Animales , Transporte Biológico Activo/efectos de los fármacos , Transporte Biológico Activo/genética , Transporte Biológico Activo/fisiología , Comunicación Celular/genética , Humanos , Infecciones/tratamiento farmacológico , Infecciones/metabolismo , Uniones Intercelulares/efectos de los fármacos , Uniones Intercelulares/patología , Uniones Intercelulares/ultraestructura , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/metabolismo
7.
J Clin Invest ; 131(2)2021 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-33463537

RESUMEN

T cell-mediated responses are dependent on their secretion of key effector molecules. However, the critical molecular determinants of the secretion of these proteins are largely undefined. Here, we demonstrate that T cell activation increases trafficking via the ER-to-Golgi pathway. To study the functional role of this pathway, we generated mice with a T cell-specific deletion in SEC23B, a core subunit of coat protein complex II (COPII). We found that SEC23B critically regulated the T cell secretome following activation. SEC23B-deficient T cells exhibited a proliferative defect and reduced effector functions in vitro, as well as in experimental models of allogeneic and xenogeneic hematopoietic cell transplantation in vivo. However, T cells derived from 3 patients with congenital dyserythropoietic anemia II (CDAII), which results from Sec23b mutation, did not exhibit a similar phenotype. Mechanistic studies demonstrated that unlike murine KO T cells, T cells from patients with CDAII harbor increased levels of the closely related paralog, SEC23A. In vivo rescue of murine KO by expression of Sec23a from the Sec23b genomic locus restored T cell functions. Together, our data demonstrate a critical role for the COPII pathway, with evidence for functional overlap in vivo between SEC23 paralogs in the regulation of T cell immunity in both mice and humans.


Asunto(s)
Autoinmunidad , Vesículas Cubiertas por Proteínas de Revestimiento/inmunología , Retículo Endoplásmico/inmunología , Aparato de Golgi/inmunología , Linfocitos T/inmunología , Animales , Transporte Biológico Activo/genética , Vesículas Cubiertas por Proteínas de Revestimiento/genética , Retículo Endoplásmico/genética , Aparato de Golgi/genética , Humanos , Ratones , Ratones Noqueados
8.
Cells ; 10(1)2021 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-33477551

RESUMEN

Age-related macular degeneration (AMD) is a blinding disease for which most of the patients remain untreatable. Since the disease affects the macula at the center of the retina, a structure specific to the primate lineage, rodent models to study the pathophysiology of AMD and to develop therapies are very limited. Consequently, our understanding relies mostly on genetic studies highlighting risk alleles at many loci. We are studying the possible implication of a metabolic imbalance associated with risk alleles within the SLC16A8 gene that encodes for a retinal pigment epithelium (RPE)-specific lactate transporter MCT3 and its consequences for vision. As a first approach, we report here the deficit in transepithelial lactate transport of a rare SLC16A8 allele identified during a genome-wide association study. We produced induced pluripotent stem cells (iPSCs) from the unique patient in our cohort that carries two copies of this allele. After in vitro differentiation of the iPSCs into RPE cells and their characterization, we demonstrate that the rare allele results in the retention of intron 2 of the SLC16A8 gene leading to the absence of MCT3 protein. We show using a biochemical assay that these cells have a deficit in transepithelial lactate transport.


Asunto(s)
Empalme Alternativo , Células Epiteliales/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Ácido Láctico/metabolismo , Transportadores de Ácidos Monocarboxílicos/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Transporte Biológico Activo/genética , Células Epiteliales/patología , Humanos , Células Madre Pluripotentes Inducidas/patología , Degeneración Macular/genética , Degeneración Macular/metabolismo , Degeneración Macular/patología , Transportadores de Ácidos Monocarboxílicos/genética , Epitelio Pigmentado de la Retina/patología
9.
Proc Natl Acad Sci U S A ; 118(1)2021 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-33443187

RESUMEN

N-1-naphthylphthalamic acid (NPA) is a key inhibitor of directional (polar) transport of the hormone auxin in plants. For decades, it has been a pivotal tool in elucidating the unique polar auxin transport-based processes underlying plant growth and development. Its exact mode of action has long been sought after and is still being debated, with prevailing mechanistic schemes describing only indirect connections between NPA and the main transporters responsible for directional transport, namely PIN auxin exporters. Here we present data supporting a model in which NPA associates with PINs in a more direct manner than hitherto postulated. We show that NPA inhibits PIN activity in a heterologous oocyte system and that expression of NPA-sensitive PINs in plant, yeast, and oocyte membranes leads to specific saturable NPA binding. We thus propose that PINs are a bona fide NPA target. This offers a straightforward molecular basis for NPA inhibition of PIN-dependent auxin transport and a logical parsimonious explanation for the known physiological effects of NPA on plant growth, as well as an alternative hypothesis to interpret past and future results. We also introduce PIN dimerization and describe an effect of NPA on this, suggesting that NPA binding could be exploited to gain insights into structural aspects of PINs related to their transport mechanism.


Asunto(s)
Transporte Biológico Activo/efectos de los fármacos , Ácidos Indolacéticos/metabolismo , Ftalimidas/metabolismo , Reguladores del Crecimiento de las Plantas/metabolismo , Proteínas de Plantas/metabolismo , Animales , Arabidopsis/efectos de los fármacos , Arabidopsis/metabolismo , Proteínas de Arabidopsis/metabolismo , Transporte Biológico Activo/genética , Dimerización , Espectrometría de Masas , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Oocitos/efectos de los fármacos , Fosforilación , Ftalimidas/farmacología , Reguladores del Crecimiento de las Plantas/antagonistas & inhibidores , Reguladores del Crecimiento de las Plantas/genética , Proteínas de Plantas/genética , Saccharomyces cerevisiae/metabolismo , Nicotiana/efectos de los fármacos , Nicotiana/metabolismo , Xenopus
10.
Int J Mol Sci ; 22(1)2020 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-33375396

RESUMEN

ABCA4 is an ATP-binding cassette (ABC) transporter expressed in photoreceptors, where it transports its substrate, N-retinylidene-phosphatidylethanolamine (N-Ret-PE), across outer segment membranes to facilitate the clearance of retinal from photoreceptors. Mutations in ABCA4 cause Stargardt macular degeneration (STGD1), an autosomal recessive disorder characterized by a loss of central vision and the accumulation of bisretinoid compounds. The purpose of this study was to determine the molecular properties of ABCA4 variants harboring disease-causing missense mutations in the transmembrane domains. Thirty-eight variants expressed in culture cells were analyzed for expression, ATPase activities, and substrate binding. On the basis of these properties, the variants were divided into three classes: Class 1 (severe variants) exhibited significantly reduced ABCA4 expression and basal ATPase activity that was not stimulated by its substrate N-Ret-PE; Class 2 (moderate variants) showed a partial reduction in expression and basal ATPase activity that was modestly stimulated by N-Ret-PE; and Class 3 (mild variants) displayed expression and functional properties comparable to normal ABCA4. The p.R653C variant displayed normal expression and basal ATPase activity, but lacked substrate binding and ATPase activation, suggesting that arginine 653 contributes to N-Ret-PE binding. Our classification provides a basis for better understanding genotype-phenotype correlations and evaluating therapeutic treatments for STGD1.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Transporte Biológico Activo/genética , Enfermedad de Stargardt/genética , Enfermedad de Stargardt/metabolismo , Transportadoras de Casetes de Unión a ATP/química , Animales , Células COS , Chlorocebus aethiops , Técnica del Anticuerpo Fluorescente , Expresión Génica , Estudios de Asociación Genética , Células HEK293 , Humanos , Modelos Moleculares , Mutación Missense , Fosfatidiletanolaminas/metabolismo , Unión Proteica , Dominios Proteicos , Enfermedades de la Retina/congénito , Enfermedades de la Retina/genética , Enfermedades de la Retina/metabolismo , Retinoides/metabolismo , Enfermedad de Stargardt/enzimología
11.
PLoS Genet ; 16(12): e1009232, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33347437

RESUMEN

Motile cilia can beat with distinct patterns, but how motility variations are regulated remain obscure. Here, we have studied the role of the coiled-coil protein CFAP53 in the motility of different cilia-types in the mouse. While node (9+0) cilia of Cfap53 mutants were immotile, tracheal and ependymal (9+2) cilia retained motility, albeit with an altered beat pattern. In node cilia, CFAP53 mainly localized at the base (centriolar satellites), whereas it was also present along the entire axoneme in tracheal cilia. CFAP53 associated tightly with microtubules and interacted with axonemal dyneins and TTC25, a dynein docking complex component. TTC25 and outer dynein arms (ODAs) were lost from node cilia, but were largely maintained in tracheal cilia of Cfap53-/- mice. Thus, CFAP53 at the base of node cilia facilitates axonemal transport of TTC25 and dyneins, while axonemal CFAP53 in 9+2 cilia stabilizes dynein binding to microtubules. Our study establishes how differential localization and function of CFAP53 contributes to the unique motion patterns of two important mammalian cilia-types.


Asunto(s)
Dineínas Axonemales/metabolismo , Axonema/metabolismo , Transporte Biológico Activo/genética , Movimiento Celular/genética , Cilios/metabolismo , Embrión de Mamíferos/metabolismo , Microtúbulos/metabolismo , Animales , Dineínas Axonemales/genética , Axonema/genética , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Cilios/genética , Embrión de Mamíferos/fisiología , Embrión de Mamíferos/ultraestructura , Epéndimo/embriología , Epéndimo/metabolismo , Epéndimo/fisiología , Técnica del Anticuerpo Fluorescente , Genotipo , Inmunoprecipitación , Ratones , Ratones Noqueados , Microscopía Electrónica de Transmisión , Microtúbulos/genética , Mutación , Fenotipo , Tráquea/embriología , Tráquea/metabolismo , Tráquea/fisiología , Tráquea/ultraestructura
12.
Nat Commun ; 11(1): 5565, 2020 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-33149158

RESUMEN

Resistance-nodulation-division efflux pumps play a key role in inherent and evolved multidrug resistance in bacteria. AcrB, a prototypical member of this protein family, extrudes a wide range of antimicrobial agents out of bacteria. Although high-resolution structures exist for AcrB, its conformational fluctuations and their putative role in function are largely unknown. Here, we determine these structural dynamics in the presence of substrates using hydrogen/deuterium exchange mass spectrometry, complemented by molecular dynamics simulations, and bacterial susceptibility studies. We show that an efflux pump inhibitor potentiates antibiotic activity by restraining drug-binding pocket dynamics, rather than preventing antibiotic binding. We also reveal that a drug-binding pocket substitution discovered within a multidrug resistant clinical isolate modifies the plasticity of the transport pathway, which could explain its altered substrate efflux. Our results provide insight into the molecular mechanism of drug export and inhibition of a major multidrug efflux pump and the directive role of its dynamics.


Asunto(s)
Ciprofloxacina/farmacología , Dipéptidos/farmacología , Farmacorresistencia Bacteriana Múltiple/genética , Proteínas de Escherichia coli/química , Escherichia coli/metabolismo , Proteínas de la Membrana/química , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/química , Proteínas Quinasas/química , Antibacterianos/química , Antibacterianos/farmacología , Sitios de Unión/genética , Transporte Biológico Activo/efectos de los fármacos , Transporte Biológico Activo/genética , Ciprofloxacina/química , Dicroismo Circular , Deuterio/química , Dipéptidos/química , Farmacorresistencia Bacteriana Múltiple/efectos de los fármacos , Escherichia coli/genética , Proteínas de Escherichia coli/antagonistas & inhibidores , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Ligandos , Espectrometría de Masas/métodos , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Pruebas de Sensibilidad Microbiana , Simulación de Dinámica Molecular , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Mutación , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo
13.
Biochem Biophys Res Commun ; 533(4): 1083-1087, 2020 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-33019976

RESUMEN

Ferroptosis, a regulated form of cell necrosis was previously reported to be induced upon pharmacological targeting of the cystine transporter SLC7A11 in Head and neck Squamous Cell Carcinoma (HNSCC). Whether tumors arising in a context of chronic infection with Human Papillomavirus (HPV) are sensitive to ferroptosis is unknown. Using RNAseq data (both whole-tumor and single-cell sequencing) we report that HPV positive (HPV+ve) tumors have lower expression levels of SLC7A11 compared to HPV negative (HPV-ve) HNSCC. We examined in vitro the effect of erastin, a specific blocker of SLC7A11, applied on two HNSCC cell lines with stable expression of HPV16 E6 and E7 oncoproteins. We report a decrease in total GSH levels and an increased sensitivity to erastin-induced ferroptosis in E6-E7 cells. Cell sensitivity to ferroptosis was specificaly related to a defect in cystine transport since we found no difference in ferroptosis induced by the direct inhibition of GPX4, and N-Acetyl Cystein abolished the difference between WT and E6-E7-expressing cells. Our findings point to SLC7A11 as an HPV-related biomarker of potential therapeutic relevance in HNSCC. Targeting cystine import to promote ferroptosis might be a promising strategy against HPV+ve HNSCC. (188 words).


Asunto(s)
Sistema de Transporte de Aminoácidos y+/metabolismo , Ferroptosis/genética , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/virología , Papillomavirus Humano 16/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/virología , Acetilcisteína/metabolismo , Sistema de Transporte de Aminoácidos y+/antagonistas & inhibidores , Sistema de Transporte de Aminoácidos y+/genética , Transporte Biológico Activo/efectos de los fármacos , Transporte Biológico Activo/genética , Biomarcadores/metabolismo , Línea Celular Tumoral , Cistina/metabolismo , Ferroptosis/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Glutatión/metabolismo , Neoplasias de Cabeza y Cuello/genética , Humanos , Proteínas Oncogénicas Virales/metabolismo , Proteínas E7 de Papillomavirus/metabolismo , Infecciones por Papillomavirus/virología , Fosfolípido Hidroperóxido Glutatión Peroxidasa/antagonistas & inhibidores , Piperazinas/farmacología , RNA-Seq , Proteínas Represoras/metabolismo , Análisis de la Célula Individual , Carcinoma de Células Escamosas de Cabeza y Cuello/genética
14.
PLoS Genet ; 16(10): e1009016, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33031417

RESUMEN

Several ABC exporters carry a degenerate nucleotide binding site (NBS) that is unable to hydrolyze ATP at a rate sufficient for sustaining transport activity. A hallmark of a degenerate NBS is the lack of the catalytic glutamate in the Walker B motif in the nucleotide binding domain (NBD). The multidrug resistance transporter ABCB1 (P-glycoprotein) has two canonical NBSs, and mutation of the catalytic glutamate E556 in NBS1 renders ABCB1 transport-incompetent. In contrast, the closely related bile salt export pump ABCB11 (BSEP), which shares 49% sequence identity with ABCB1, naturally contains a methionine in place of the catalytic glutamate. The NBD-NBD interfaces of ABCB1 and ABCB11 differ only in four residues, all within NBS1. Mutation of the catalytic glutamate in ABCB1 results in the occlusion of ATP in NBS1, leading to the arrest of the transport cycle. Here we show that despite the catalytic glutamate mutation (E556M), ABCB1 regains its ATP-dependent transport activity, when three additional diverging residues are also replaced. Molecular dynamics simulations revealed that the rescue of ATPase activity is due to the modified geometry of NBS1, resulting in a weaker interaction with ATP, which allows the quadruple mutant to evade the conformationally locked pre-hydrolytic state to proceed to ATP-driven transport. In summary, we show that ABCB1 can be transformed into an active transporter with only one functional catalytic site by preventing the formation of the ATP-locked pre-hydrolytic state in the non-canonical site.


Asunto(s)
Miembro 11 de la Subfamilia B de Transportador de Casetes de Unión al ATP/genética , Transporte Biológico/genética , Proteínas de Ciclo Celular/genética , Proteínas Nucleares/genética , Dominio AAA/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Adenosina Trifosfato/genética , Secuencia de Aminoácidos , Sitios de Unión/genética , Transporte Biológico Activo/genética , Dominio Catalítico/genética , Ácido Glutámico/genética , Humanos , Hidrólisis , Metionina/genética , Simulación de Dinámica Molecular , Mutación/genética , Nucleótidos/genética , Unión Proteica/genética , Dominios Proteicos/genética
15.
EMBO J ; 39(20): e105117, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-32840906

RESUMEN

Heterotetrameric adapter (AP) complexes cooperate with the small GTPase Arf1 or lipids in cargo selection, vesicle formation, and budding at endomembranes in eukaryotic cells. While most AP complexes also require clathrin as the outer vesicle shell, formation of AP-3-coated vesicles involved in Golgi-to-vacuole transport in yeast has been postulated to depend on Vps41, a subunit of the vacuolar HOPS tethering complex. HOPS has also been identified as the tether of AP-3 vesicles on vacuoles. To unravel this conundrum of a dual Vps41 function, we anchored Vps41 stably to the mitochondrial outer membrane. By monitoring AP-3 recruitment, we now show that Vps41 can tether AP-3 vesicles to mitochondria, yet AP-3 vesicles can form in the absence of Vps41 or clathrin. By proximity labeling and mass spectrometry, we identify the Arf1 GTPase-activating protein (GAP) Age2 at the AP-3 coat and show that tethering, but not fusion at the vacuole can occur without complete uncoating. We conclude that AP-3 vesicles retain their coat after budding and that their complete uncoating occurs only after tethering at the vacuole.


Asunto(s)
Factores de Ribosilacion-ADP/metabolismo , Vesículas Citoplasmáticas/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Membranas Mitocondriales/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Vacuolas/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Factores de Ribosilacion-ADP/genética , Transporte Biológico Activo/genética , Quinasa de la Caseína I/genética , Quinasa de la Caseína I/metabolismo , Vesículas Citoplasmáticas/ultraestructura , Proteínas Activadoras de GTPasa/genética , Eliminación de Gen , Aparato de Golgi/metabolismo , Espectrometría de Masas , Fusión de Membrana , Microscopía Electrónica , Membranas Mitocondriales/ultraestructura , Proteínas de Saccharomyces cerevisiae/genética , Vacuolas/ultraestructura , Proteínas de Transporte Vesicular/genética
16.
Mol Syst Biol ; 16(7): e9652, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32697042

RESUMEN

Solute carriers (SLCs) are the largest family of transmembrane transporters in the human genome with more than 400 members. Despite the fact that SLCs mediate critical biological functions and several are important pharmacological targets, a large proportion of them is poorly characterized and present no assigned substrate. A major limitation to systems-level de-orphanization campaigns is the absence of a structured, language-controlled chemical annotation. Here we describe a thorough manual annotation of SLCs based on literature. The annotation of substrates, transport mechanism, coupled ions, and subcellular localization for 446 human SLCs confirmed that ~30% of these were still functionally orphan and lacked known substrates. Application of a substrate-based ontology to transcriptomic datasets identified SLC-specific responses to external perturbations, while a machine-learning approach based on the annotation allowed us to identify potential substrates for several orphan SLCs. The annotation is available at https://opendata.cemm.at/gsflab/slcontology/. Given the increasing availability of large biological datasets and the growing interest in transporters, we expect that the effort presented here will be critical to provide novel insights into the functions of SLCs.


Asunto(s)
Transporte Biológico Activo/genética , Biología Computacional/métodos , Proteínas de Transporte de Membrana/metabolismo , Aminoácidos/metabolismo , Aminoácidos/farmacología , Ontologías Biológicas , Línea Celular , Perfilación de la Expresión Génica , Genoma Humano , Humanos , Aprendizaje Automático
17.
J Cell Biol ; 219(6)2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32357219

RESUMEN

Autophagosome biogenesis involves de novo formation of a membrane that elongates to sequester cytoplasmic cargo and closes to form a double-membrane vesicle (an autophagosome). This process has remained enigmatic since its initial discovery >50 yr ago, but our understanding of the mechanisms involved in autophagosome biogenesis has increased substantially during the last 20 yr. Several key questions do remain open, however, including, What determines the site of autophagosome nucleation? What is the origin and lipid composition of the autophagosome membrane? How is cargo sequestration regulated under nonselective and selective types of autophagy? This review provides key insight into the core molecular mechanisms underlying autophagosome biogenesis, with a specific emphasis on membrane modeling events, and highlights recent conceptual advances in the field.


Asunto(s)
Autofagosomas/metabolismo , Proteínas Relacionadas con la Autofagia/metabolismo , Retículo Endoplásmico/metabolismo , Metabolismo de los Lípidos , Membranas/metabolismo , Autofagosomas/química , Autofagia/genética , Autofagia/fisiología , Proteínas Relacionadas con la Autofagia/genética , Transporte Biológico Activo/genética , Transporte Biológico Activo/fisiología , Humanos , Lípidos/biosíntesis , Lípidos/química , Transducción de Señal/genética , Transducción de Señal/fisiología
18.
Sci Signal ; 13(628)2020 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-32317369

RESUMEN

The tricarboxylic acid (TCA) cycle converts the end products of glycolysis and fatty acid ß-oxidation into the reducing equivalents NADH and FADH2 Although mitochondrial matrix uptake of Ca2+ enhances ATP production, it remains unclear whether deprivation of mitochondrial TCA substrates alters mitochondrial Ca2+ flux. We investigated the effect of TCA cycle substrates on MCU-mediated mitochondrial matrix uptake of Ca2+, mitochondrial bioenergetics, and autophagic flux. Inhibition of glycolysis, mitochondrial pyruvate transport, or mitochondrial fatty acid transport triggered expression of the MCU gatekeeper MICU1 but not the MCU core subunit. Knockdown of mitochondrial pyruvate carrier (MPC) isoforms or expression of the dominant negative mutant MPC1R97W resulted in increased MICU1 protein abundance and inhibition of MCU-mediated mitochondrial matrix uptake of Ca2+ We also found that genetic ablation of MPC1 in hepatocytes and mouse embryonic fibroblasts resulted in reduced resting matrix Ca2+, likely because of increased MICU1 expression, but resulted in changes in mitochondrial morphology. TCA cycle substrate-dependent MICU1 expression was mediated by the transcription factor early growth response 1 (EGR1). Blocking mitochondrial pyruvate or fatty acid flux was linked to increased autophagy marker abundance. These studies reveal a mechanism that controls the MCU-mediated Ca2+ flux machinery and that depends on TCA cycle substrate availability. This mechanism generates a metabolic homeostatic circuit that protects cells from bioenergetic crisis and mitochondrial Ca2+ overload during periods of nutrient stress.


Asunto(s)
Canales de Calcio/metabolismo , Proteínas de Unión al Calcio/metabolismo , Proteínas de Transporte de Catión/metabolismo , Ácidos Grasos/metabolismo , Mitocondrias Hepáticas/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Proteínas Mitocondriales/metabolismo , Ácido Pirúvico/metabolismo , Animales , Transporte Biológico Activo/genética , Canales de Calcio/genética , Proteínas de Unión al Calcio/genética , Proteínas de Transporte de Catión/genética , Células HEK293 , Células HeLa , Células Hep G2 , Humanos , Ratones Noqueados , Mitocondrias Hepáticas/genética , Proteínas de Transporte de Membrana Mitocondrial/genética , Proteínas Mitocondriales/genética
19.
Cells ; 9(4)2020 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-32316189

RESUMEN

The sodium taurocholate cotransporting polypeptide (NTCP) is expressed at the basolateral membrane of hepatocytes, where it mediates the uptake of conjugated bile acids and forms the hepatocyte entry receptor for the hepatitis B and D virus. Here, we aimed to identify novel protein-protein interactions that could play a role in the regulation of NTCP. To this end, NTCP was precipitated from HA-tagged hNTCP-expressing HepG2 cells, and chloride channel CLIC-like 1 (CLCC1) and stomatin were identified as interacting proteins by mass spectrometry. Interaction was confirmed by co-immunoprecipitation. NTCP, CLCC1 and stomatin were found at the plasma membrane in lipid rafts, as demonstrated by a combination of immunofluorescence, cell surface biotinylation and isolation of detergent-resistant membranes. Neither CLCC1 overexpression nor its knockdown had an effect on NTCP function. However, both stomatin overexpression and knockdown increased NTCP-mediated taurocholate uptake while NTCP abundance at the plasma membrane was only increased in stomatin depleted cells. These findings identify stomatin as an interactor of NTCP and show that the interaction modulates bile salt transport.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Transporte Biológico Activo/genética , Hepatocitos/metabolismo , Hígado/metabolismo , Proteínas de la Membrana/metabolismo , Transportadores de Anión Orgánico Sodio-Dependiente/metabolismo , Simportadores/metabolismo , Ácido Taurocólico/metabolismo , Línea Celular Tumoral , Membrana Celular/metabolismo , Canales de Cloruro/genética , Canales de Cloruro/metabolismo , Cromatografía Liquida , Técnicas de Silenciamiento del Gen , Humanos , Microdominios de Membrana/metabolismo , Proteínas de la Membrana/genética , Transportadores de Anión Orgánico Sodio-Dependiente/genética , Unión Proteica , Simportadores/genética , Espectrometría de Masas en Tándem
20.
Mol Microbiol ; 114(1): 151-171, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32198949

RESUMEN

Sinorhizobium (Ensifer) meliloti is a model example of a soil alpha-proteobacterium which induces the formation of nitrogen-fixing symbiotic nodules on the legume roots. In contrast to all other rhizobacterial species, S. meliloti contains multiple homologs of nucleobase transporter genes that belong to NAT/NCS2 family (Nucleobase-Ascorbate Transporter/Nucleobase-Cation Symporter-2). We analyzed functionally all (six) relevant homologs of S. meliloti 1,021 using Escherichia coli K-12 as a host and found that five of them are high-affinity transporters for xanthine (SmLL9), uric acid (SmLL8, SmLL9, SmX28), adenine (SmVC3, SmYE1), guanine (SmVC3), or hypoxanthine (SmVC3). Detailed analysis of substrate profiles showed that two of these transporters display enlarged specificity (SmLL9, SmVC3). SmLL9 is closely related in sequence with the xanthine-specific XanQ of E. coli. We subjected SmLL9 to rationally designed site-directed mutagenesis and found that the role of key binding-site residues of XanQ is conserved in SmLL9, whereas a single amino-acid change (S93N) converts the xanthine/uric-acid transporter SmLL9 to a xanthine-preferring variant, due to disruption of an essential hydrogen bond with the C8 oxygen of uric acid. The results highlight the presence of several different purine nucleobase transporters in S. meliloti and imply that the purine transport might be important in the nodule symbiosis involving S. meliloti.


Asunto(s)
Transporte Biológico Activo/genética , Sinorhizobium meliloti/genética , Sinorhizobium meliloti/metabolismo , Simportadores/genética , Simportadores/metabolismo , Adenina/metabolismo , Escherichia coli K12/genética , Escherichia coli K12/metabolismo , Guanina/metabolismo , Hipoxantina/metabolismo , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Nodulación de la Raíz de la Planta/fisiología , Rizosfera , Nódulos de las Raíces de las Plantas/microbiología , Ácido Úrico/metabolismo , Xantina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA