Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 248
Filtrar
1.
J Biol Chem ; 300(1): 105576, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38110033

RESUMEN

The sixth family phosphodiesterases (PDE6) are principal effector enzymes of the phototransduction cascade in rods and cones. Maturation of nascent PDE6 protein into a functional enzyme relies on a coordinated action of ubiquitous chaperone HSP90, its specialized cochaperone aryl hydrocarbon receptor-interacting protein-like 1 (AIPL1), and the regulatory Pγ-subunit of PDE6. Deficits in PDE6 maturation and function underlie severe visual disorders and blindness. Here, to elucidate the roles of HSP90, AIPL1, and Pγ in the maturation process, we developed the heterologous expression system of human cone PDE6C in insect cells allowing characterization of the purified enzyme. We demonstrate that in the absence of Pγ, HSP90, and AIPL1 convert the inactive and aggregating PDE6C species into dimeric PDE6C that is predominantly misassembled. Nonetheless, a small fraction of PDE6C is properly assembled and fully functional. From the analysis of mutant mice that lack both rod Pγ and PDE6C, we conclude that, in contrast to the cone enzyme, no maturation of rod PDE6AB occurs in the absence of Pγ. Co-expression of PDE6C with AIPL1 and Pγ in insect cells leads to a fully mature enzyme that is equivalent to retinal PDE6. Lastly, using immature PDE6C and purified chaperone components, we reconstituted the process of the client maturation in vitro. Based on this analysis we propose a scheme for the PDE6 maturation process.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 6 , Células Fotorreceptoras Retinianas Conos , Animales , Humanos , Ratones , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Ceguera/genética , Línea Celular , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 6/química , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 6/deficiencia , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 6/genética , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 6/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Mutación , Multimerización de Proteína , Subunidades de Proteína/química , Subunidades de Proteína/deficiencia , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Células Fotorreceptoras Retinianas Conos/química , Células Fotorreceptoras Retinianas Conos/metabolismo
2.
J Biol Chem ; 299(9): 105132, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37544648

RESUMEN

Voltage-gated sodium (NaV) channels drive the upstroke of the action potential and are comprised of a pore-forming α-subunit and regulatory ß-subunits. The ß-subunits modulate the gating, trafficking, and pharmacology of the α-subunit. These functions are routinely assessed by ectopic expression in heterologous cells. However, currently available expression systems may not capture the full range of these effects since they contain endogenous ß-subunits. To better reveal ß-subunit functions, we engineered a human cell line devoid of endogenous NaV ß-subunits and their immediate phylogenetic relatives. This new cell line, ß-subunit-eliminated eHAP expression (BeHAPe) cells, were derived from haploid eHAP cells by engineering inactivating mutations in the ß-subunits SCN1B, SCN2B, SCN3B, and SCN4B, and other subfamily members MPZ (myelin protein zero(P0)), MPZL1, MPZL2, MPZL3, and JAML. In diploid BeHAPe cells, the cardiac NaV α-subunit, NaV1.5, was highly sensitive to ß-subunit modulation and revealed that each ß-subunit and even MPZ imparted unique gating properties. Furthermore, combining ß1 and ß2 with NaV1.5 generated a sodium channel with hybrid properties, distinct from the effects of the individual subunits. Thus, this approach revealed an expanded ability of ß-subunits to regulate NaV1.5 activity and can be used to improve the characterization of other α/ß NaV complexes.


Asunto(s)
Canal de Sodio Activado por Voltaje NAV1.5 , Subunidades de Proteína , Subunidades beta de Canales de Sodio Activados por Voltaje , Humanos , Potenciales de Acción , Línea Celular , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5/química , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Fosfoproteínas/metabolismo , Subunidades de Proteína/química , Subunidades de Proteína/deficiencia , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Subunidades beta de Canales de Sodio Activados por Voltaje/química , Subunidades beta de Canales de Sodio Activados por Voltaje/deficiencia , Subunidades beta de Canales de Sodio Activados por Voltaje/genética , Subunidades beta de Canales de Sodio Activados por Voltaje/metabolismo , Mutación
3.
Int J Mol Sci ; 22(22)2021 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-34830323

RESUMEN

Sporadic occurrence of inherited eye disorders has been reported in cattle but so far pathogenic variants were found only for rare forms of cataract but not for retinopathies. The aim of this study was to characterize the phenotype and the genetic aetiology of a recessive form of congenital day-blindness observed in several cases of purebred Original Braunvieh cattle. Electroretinography in an affected calf revealed absent cone-mediated function, whereas the rods continue to function normally. Brain areas involved in vision were morphologically normal. When targeting cones by immunofluorescence, a decrease in cone number and an accumulation of beta subunits of cone cyclic-nucleotide gated channel (CNGB3) in the outer plexiform layer of affected animals was obvious. Achromatopsia is a monogenic Mendelian disease characterized by the loss of cone photoreceptor function resulting in day-blindness, total color-blindness, and decreased central visual acuity. After SNP genotyping and subsequent homozygosity mapping with twelve affected cattle, we performed whole-genome sequencing and variant calling of three cases. We identified a single missense variant in the bovine CNGB3 gene situated in a ~2.5 Mb homozygous genome region on chromosome 14 shared between all cases. All affected cattle were homozygous carriers of the p.Asp251Asn mutation that was predicted to be deleterious, affecting an evolutionary conserved residue. In conclusion, we have evidence for the occurrence of a breed-specific novel CNGB3-related form of recessively inherited achromatopsia in Original Braunvieh cattle which we have designated OH1 showing an allele frequency of the deleterious allele of ~8%. The identification of carriers will enable selection against this inherited disorder. The studied cattle might serve as an animal model to further elucidate the function of CNGB3 in mammals.


Asunto(s)
Alelos , Defectos de la Visión Cromática/genética , Canales Catiónicos Regulados por Nucleótidos Cíclicos/genética , Mutación Missense , Subunidades de Proteína/genética , Células Fotorreceptoras Retinianas Conos/metabolismo , Sustitución de Aminoácidos , Animales , Asparagina/metabolismo , Ácido Aspártico/metabolismo , Bovinos , Defectos de la Visión Cromática/diagnóstico por imagen , Defectos de la Visión Cromática/metabolismo , Defectos de la Visión Cromática/patología , Canales Catiónicos Regulados por Nucleótidos Cíclicos/deficiencia , Electrorretinografía , Femenino , Expresión Génica , Frecuencia de los Genes , Homocigoto , Masculino , Fenotipo , Subunidades de Proteína/deficiencia , Células Fotorreceptoras Retinianas Conos/patología , Células Fotorreceptoras Retinianas Bastones/citología , Células Fotorreceptoras Retinianas Bastones/metabolismo , Secuenciación Completa del Genoma
4.
PLoS One ; 16(4): e0249227, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33793620

RESUMEN

Bacteria play an integral role in shaping plant growth and development. However, the genetic factors that facilitate plant-bacteria interactions remain largely unknown. Here, we demonstrated the importance of two bacterial genetic factors that facilitate the interactions between plant-growth-promoting (PGP) bacteria in the genus Caulobacter and the host plant Arabidopsis. Using homologous recombination, we disrupted the cytochrome ubiquinol oxidase (cyo) operon in both C. vibrioides CB13 and C. segnis TK0059 by knocking out the expression of cyoB (critical subunit of the cyo operon) and showed that the mutant strains were unable to enhance the growth of Arabidopsis. In addition, disruption of the cyo operon, metabolomic reconstructions, and pH measurements suggested that both elevated cyoB expression and acid production by strain CB13 contribute to the previously observed inhibition of Arabidopsis seed germination. We also showed that the crescent shape of the PGP bacterial strain C. crescentus CB15 contributes to its ability to enhance plant growth. Thus, we have identified specific genetic factors that explain how select Caulobacter strains interact with Arabidopsis plants.


Asunto(s)
Arabidopsis/crecimiento & desarrollo , Proteínas Bacterianas/genética , Caulobacter/genética , Complejo IV de Transporte de Electrones/genética , Arabidopsis/anatomía & histología , Arabidopsis/microbiología , Proteínas Bacterianas/metabolismo , Caulobacter/clasificación , Complejo IV de Transporte de Electrones/metabolismo , Expresión Génica , Germinación , Recombinación Homóloga , Concentración de Iones de Hidrógeno , Filogenia , Subunidades de Proteína/deficiencia , Subunidades de Proteína/genética , Especies Reactivas de Oxígeno/metabolismo
5.
PLoS Biol ; 18(8): e3000826, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32776935

RESUMEN

Ca2+/calmodulin-dependent kinase II (CaMKII) regulates synaptic plasticity in multiple ways, supposedly including the secretion of neuromodulators like brain-derived neurotrophic factor (BDNF). Here, we show that neuromodulator secretion is indeed reduced in mouse α- and ßCaMKII-deficient (αßCaMKII double-knockout [DKO]) hippocampal neurons. However, this was not due to reduced secretion efficiency or neuromodulator vesicle transport but to 40% reduced neuromodulator levels at synapses and 50% reduced delivery of new neuromodulator vesicles to axons. αßCaMKII depletion drastically reduced neuromodulator expression. Blocking BDNF secretion or BDNF scavenging in wild-type neurons produced a similar reduction. Reduced neuromodulator expression in αßCaMKII DKO neurons was restored by active ßCaMKII but not inactive ßCaMKII or αCaMKII, and by CaMKII downstream effectors that promote cAMP-response element binding protein (CREB) phosphorylation. These data indicate that CaMKII regulates neuromodulation in a feedback loop coupling neuromodulator secretion to ßCaMKII- and CREB-dependent neuromodulator expression and axonal targeting, but CaMKIIs are dispensable for the secretion process itself.


Asunto(s)
Astrocitos/metabolismo , Factor Neurotrófico Derivado del Encéfalo/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Calcio/metabolismo , Neuronas/metabolismo , Subunidades de Proteína/genética , Animales , Astrocitos/citología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/deficiencia , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Retroalimentación Fisiológica , Regulación de la Expresión Génica , Hipocampo/citología , Hipocampo/metabolismo , Ratones , Ratones Transgénicos , Neuronas/citología , Fosforilación , Cultivo Primario de Células , Subunidades de Proteína/deficiencia , Sinapsis/fisiología , Transmisión Sináptica , Imagen de Lapso de Tiempo
6.
Cell Rep ; 29(13): 4285-4294.e5, 2019 12 24.
Artículo en Inglés | MEDLINE | ID: mdl-31875540

RESUMEN

NMDA receptors (NMDARs) are critical for physiological synaptic plasticity, learning, and memory and for pathological plasticity and neuronal death. The GluN1 subunit is encoded by a single gene, GRIN1, with 8 splice variants, but whether the diversity generated by this splicing has physiological consequences remains enigmatic. Here, we generate mice lacking from the GluN1 exon 5-encoded N1 cassette (GluN1a mice) or compulsorily expressing this exon (GluN1b mice). Despite no differences in basal synaptic transmission, long-term potentiation in the hippocampus is significantly enhanced in GluN1a mice compared with that in GluN1b mice. Furthermore, GluN1a mice learn more quickly and have significantly better spatial memory performance than do GluN1b mice. In addition, in human iPSC-derived neurons in autism spectrum disorder NMDARs show characteristics of N1-lacking GluN1. Our findings indicate that alternative splicing of GluN1 is a mechanism for controlling physiological long-lasting synaptic potentiation, learning, and memory.


Asunto(s)
Empalme Alternativo , Trastorno del Espectro Autista/genética , Potenciación a Largo Plazo/genética , Proteínas del Tejido Nervioso/genética , Neuronas/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Animales , Trastorno del Espectro Autista/metabolismo , Trastorno del Espectro Autista/patología , Muerte Celular/genética , Diferenciación Celular , Potenciales Postsinápticos Excitadores/fisiología , Exones , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/deficiencia , Neuronas/patología , Cultivo Primario de Células , Subunidades de Proteína/deficiencia , Subunidades de Proteína/genética , Receptores de N-Metil-D-Aspartato/deficiencia , Memoria Espacial/fisiología , Sinapsis/metabolismo , Transmisión Sináptica
7.
Biomolecules ; 9(6)2019 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-31159305

RESUMEN

The 26S proteasome is a key player in the degradation of ubiquitinated proteins, comprising a 20S core particle (CP) and a 19S regulatory particle (RP). The RP is further divided into base and lid subcomplexes, which are assembled independently from each other. We have previously demonstrated the assembly pathway of the CP and the base by observing assembly intermediates resulting from knockdowns of each proteasome subunit and the assembly chaperones. In this study, we examine the assembly pathway of the mammalian lid, which remains to be elucidated. We show that the lid assembly pathway is conserved between humans and yeast. The final step is the incorporation of Rpn12 into the assembly intermediate consisting of two modular complexes, Rpn3-7-15 and Rpn5-6-8-9-11, in both humans and yeast. Furthermore, we dissect the assembly pathways of the two modular complexes by the knockdown of each lid subunit.


Asunto(s)
Complejo de la Endopetidasa Proteasomal/química , Complejo de la Endopetidasa Proteasomal/metabolismo , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Subunidades de Proteína/deficiencia , Subunidades de Proteína/genética , Interferencia de ARN , ARN Interferente Pequeño/genética
8.
Cell Microbiol ; 21(8): e13041, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31087807

RESUMEN

AP-2 complex is widely distributed in eukaryotes in the form of heterotetramer that functions in the uptake of membrane proteins during mammalian/plant clathrin-mediated endocytosis. However, its biological function remains mysterious in pathogenic fungi. In this study, the wheat scab fungus, Fusarium graminearum, was used to characterise the biological function of the AP-2 complex. Our study shows that FgAP-2 complex plays a critical role in the maintenance of hyphal polarity. Lack of any subunit (FgAP2α , FgAP2ß , FgAP2σ , and FgAP2mu ) of the FgAP-2 complex significantly affects the fungal vegetative growth, conidial morphology, and germination. Remarkably, FgAP-2 complex is important for the fungal pathogenicity, especially during colonisation and extension after infecting the host. The FgAP-2 complex is expressed ubiquitously at all developmental stages but having more concentrated protein distribution at the subapical collar and septa in young growing hyphae. Although FgAP-2 complex displays similar dynamic behaviour to the actin patch components and accumulates at endocytic sites, it is dispensable for general endocytosis. We further demonstrated that FgAP-2 complex is required for polar localisation of the lipid flippases FgDnfA and FgDnfB, which led to the proposal that FgAP-2 functions as a cargo-specific adaptor that promotes polar growth and colonising ability of F. graminearum.


Asunto(s)
Complejo 2 de Proteína Adaptadora/genética , Proteínas Fúngicas/genética , Fusarium/genética , Fusarium/patogenicidad , Regulación Fúngica de la Expresión Génica , Proteínas de Transferencia de Fosfolípidos/genética , Actinas/genética , Actinas/metabolismo , Complejo 2 de Proteína Adaptadora/metabolismo , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Endocitosis/genética , Proteínas Fúngicas/metabolismo , Fusarium/crecimiento & desarrollo , Fusarium/metabolismo , Eliminación de Gen , Hifa/genética , Hifa/crecimiento & desarrollo , Hifa/metabolismo , Hifa/patogenicidad , Isoenzimas/genética , Isoenzimas/metabolismo , Lípidos de la Membrana/metabolismo , Proteínas de Transferencia de Fosfolípidos/metabolismo , Enfermedades de las Plantas/microbiología , Subunidades de Proteína/deficiencia , Subunidades de Proteína/genética , Esporas Fúngicas/genética , Esporas Fúngicas/crecimiento & desarrollo , Esporas Fúngicas/metabolismo , Esporas Fúngicas/patogenicidad , Triticum/microbiología , Técnicas del Sistema de Dos Híbridos , Virulencia
9.
Sci Rep ; 9(1): 7159, 2019 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-31073221

RESUMEN

The nucleosome, composed of DNA and a histone core, is the basic structural unit of chromatin. The fission yeast Schizosaccharomyces pombe has two genes of histone H2A, hta1+ and hta2+; these genes encode two protein species of histone H2A (H2Aα and H2Aß, respectively), which differ in three amino acid residues, and only hta2+ is upregulated during meiosis. However, it is unknown whether S. pombe H2Aα and H2Aß have functional differences. Therefore, in this study, we examined the possible functional differences between H2Aα and H2Aß during meiosis in S. pombe. We found that deletion of hta2+, but not hta1+, causes defects in chromosome segregation and spore formation during meiosis. Meiotic defects in hta2+ deletion cells were rescued by expressing additional copies of hta1+ or by expressing hta1+ from the hta2 promoter. This indicated that the defects were caused by insufficient amounts of histone H2A, and not by the amino acid residue differences between H2Aα and H2Aß. Microscopic observation attributed the chromosome segregation defects to anaphase bridge formation in a chromosomal region at the repeats of ribosomal RNA genes (rDNA repeats). These results suggest that histone H2A insufficiency affects the chromatin structures of rDNA repeats, leading to chromosome missegregation in S. pombe.


Asunto(s)
Segregación Cromosómica/fisiología , Cromosomas Fúngicos/metabolismo , ADN Ribosómico/genética , Histonas/genética , Proteínas de Schizosaccharomyces pombe/genética , Schizosaccharomyces/metabolismo , Anafase , Cromatina/metabolismo , Histonas/deficiencia , Histonas/metabolismo , Regiones Promotoras Genéticas , Subunidades de Proteína/deficiencia , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Proteínas de Schizosaccharomyces pombe/metabolismo , Esporas Fúngicas/metabolismo , Regulación hacia Arriba
10.
EBioMedicine ; 41: 333-344, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30827928

RESUMEN

BACKGROUND: Innate lymphoid cells (ILCs) are a newly discovered family of immune cells that have similar cytokine-secreting profiles as T helper cell subsets. Although ILCs are critical for host defense against infections and tissue homeostasis, their roles in tumor development are not well established. METHODS: We studied the function of ILC3 cells in the liver for the development of hepatocellular carcinoma (HCC) in murine HCC models using flow cytometry, adoptive transfer, and in vitro functional assays. FINDINGS: We found that ILC3 lacking the natural cytotoxicity-triggering receptor (NCR-ILC3) promoted the development of HCC in response to interleukin 23 (IL-23). IL-23 serum level is elevated in HCC patients and its high expression is associated with poor clinical outcomes. We found that IL-23 could promote tumor development in murine HCC tumor models. IL-23 promoted the expansion of NCR-ILC3 and its differentiation from group 1 ILCs (ILC1s). Furthermore, NCR-ILC3 initiated IL-17 production upon IL-23 stimulation and directly inhibited CD8+ T cell immunity by promoting lymphocyte apoptosis and limiting their proliferation. INTERPRETATION: Together, our findings suggest that NCR-ILC3 initiates the IL-17-rich immunosuppressive tumor microenvironment and promotes the development of HCC, thus may serve as a promising target for future cancer immunotherapy. FUND: This work was supported by grants from National Natural Science Foundation of China (81471586, 81571556), the Priority Academic Program Development of Jiangsu Higher Education Institutions, the collaborative Innovation Center of Hematology, start-up grant from National University of Singapore, the Cancer Prevention and Research Institute of Texas CPRIT (RR180017), and the National Cancer Institute's Cancer Center Support (Core) Grant CA016672 (to The University of Texas MD Anderson Cancer Center).


Asunto(s)
Carcinoma Hepatocelular/patología , Interleucina-17/metabolismo , Interleucina-23/metabolismo , Neoplasias Hepáticas/patología , Animales , Apoptosis , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/metabolismo , Diferenciación Celular , Línea Celular Tumoral , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Inmunidad Innata , Interleucina-12/metabolismo , Interleucina-17/análisis , Interleucina-23/análisis , Interleucina-23/genética , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/metabolismo , Linfocitos/citología , Linfocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Subunidades de Proteína/deficiencia , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Trasplante Homólogo
11.
DNA Repair (Amst) ; 73: 64-70, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30470508

RESUMEN

Human DNA polymerase δ is normally present in unstressed, non-dividing cells as a heterotetramer (Pol δ4). Its smallest subunit, p12, is transiently degraded in response to UV damage, as well as during the entry into S-phase, resulting in the conversion of Pol δ4 to a trimer (Pol δ3). In order to further understand the specific cellular roles of these two forms of Pol δ, the gene (POLD4) encoding p12 was disrupted by CRISPR/Cas9 to produce p12 knockout (p12KO) cells. Thus, Pol δ4 is absent in p12KO cells, leaving Pol δ3 as the sole source of Pol δ activity. GFP reporter assays revealed that the p12KO cells exhibited a defect in homologous recombination (HR) repair, indicating that Pol δ4, but not Pol δ3, is required for HR. Expression of Flag-tagged p12 in p12KO cells to restore Pol δ4 alleviated the HR defect. These results establish a specific requirement for Pol δ4 in HR repair. This leads to the prediction that p12KO cells should be more sensitive to chemotherapeutic agents, and should exhibit synthetic lethal killing by PARP inhibitors. These predictions were confirmed by clonogenic cell survival assays of p12KO cells treated with cisplatin and mitomycin C, and with the PARP inhibitors Olaparib, Talazoparib, Rucaparib, and Niraparib. The sensitivity to PARP inhibitors in H1299-p12KO cells was alleviated by expression of Flag-p12. These findings have clinical significance, as the expression levels of p12 could be a predictive biomarker of tumor response to PARP inhibitors. In addition, small cell lung cancers (SCLC) are known to exhibit a defect in p12 expression. Analysis of several SCLC cell lines showed that they exhibit hypersensitivity to PARP inhibitors, providing evidence that loss of p12 expression could represent a novel molecular basis for HR deficiency.


Asunto(s)
ADN Polimerasa III/antagonistas & inhibidores , ADN Polimerasa III/genética , Técnicas de Inactivación de Genes , Recombinación Homóloga/genética , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Subunidades de Proteína/deficiencia , Subunidades de Proteína/genética , Línea Celular Tumoral , Cisplatino/farmacología , Roturas del ADN de Doble Cadena/efectos de los fármacos , ADN Polimerasa III/deficiencia , Células HeLa , Recombinación Homóloga/efectos de los fármacos , Humanos , Mitomicina/farmacología
12.
Am J Physiol Cell Physiol ; 316(2): C154-C161, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30427720

RESUMEN

Voltage-gated Kv1.1 potassium channel α-subunits, encoded by the Kcna1 gene, have traditionally been regarded as neural-specific with no expression or function in the heart. However, recent data revealed that Kv1.1 subunits are expressed in atria where they may have an overlooked role in controlling repolarization and arrhythmia susceptibility independent of the nervous system. To explore this concept in more detail and to identify functional and molecular effects of Kv1.1 channel impairment in the heart, atrial cardiomyocyte patch-clamp electrophysiology and gene expression analyses were performed using Kcna1 knockout ( Kcna1-/-) mice. Specifically, we hypothesized that Kv1.1 subunits contribute to outward repolarizing K+ currents in mouse atria and that their absence prolongs cardiac action potentials. In voltage-clamp experiments, dendrotoxin-K (DTX-K), a Kv1.1-specific inhibitor, significantly reduced peak outward K+ currents in wild-type (WT) atrial cells but not Kcna1-/- cells, demonstrating an important contribution by Kv1.1-containing channels to mouse atrial repolarizing currents. In current-clamp recordings, Kcna1-/- atrial myocytes exhibited significant action potential prolongation which was exacerbated in right atria, effects that were partially recapitulated in WT cells by application of DTX-K. Quantitative RT-PCR measurements showed mRNA expression remodeling in Kcna1-/- atria for several ion channel genes that contribute to the atrial action potential including the Kcna5, Kcnh2, and Kcnj2 potassium channel genes and the Scn5a sodium channel gene. This study demonstrates a previously undescribed heart-intrinsic role for Kv1.1 subunits in mediating atrial repolarization, thereby adding a new member to the already diverse collection of known K+ channels in the heart.


Asunto(s)
Potenciales de Acción/fisiología , Atrios Cardíacos/metabolismo , Canal de Potasio Kv.1.1/antagonistas & inhibidores , Canal de Potasio Kv.1.1/genética , Miocitos Cardíacos/metabolismo , Bloqueadores de los Canales de Potasio/farmacología , Potenciales de Acción/efectos de los fármacos , Animales , Femenino , Atrios Cardíacos/citología , Atrios Cardíacos/efectos de los fármacos , Canal de Potasio Kv.1.1/deficiencia , Masculino , Ratones , Miocitos Cardíacos/efectos de los fármacos , Subunidades de Proteína/deficiencia , Subunidades de Proteína/genética
13.
Hum Mol Genet ; 28(6): 912-927, 2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30445451

RESUMEN

Polyglutamine (polyQ) expansion in Ataxin-7 (ATXN7) results in spinocerebellar ataxia type 7 (SCA7) and causes visual impairment. SCA7 photoreceptors progressively lose their outer segments (OSs), a structure essential for their visual function. ATXN7 is a subunit of the transcriptional coactivator Spt-Ada-Gcn5 Acetyltransferase complex, implicated in the development of the visual system in flies. To determine the function of ATXN7 in the vertebrate eye, we have inactivated ATXN7 in zebrafish. While ATXN7 depletion in flies led to gross retinal degeneration, in zebrafish, it primarily results in ocular coloboma, a structural malformation responsible for pediatric visual impairment in humans. ATXN7 inactivation leads to elevated Hedgehog signaling in the forebrain, causing an alteration of proximo-distal patterning of the optic vesicle during early eye development and coloboma. At later developmental stages, malformations of photoreceptors due to incomplete formation of their OSs are observed and correlate with altered expression of crx, a key transcription factor involved in the formation of photoreceptor OS. Therefore, we propose that a primary toxic effect of polyQ expansion is the alteration of ATXN7 function in the daily renewal of OS in SCA7. Together, our data indicate that ATXN7 plays an essential role in vertebrate eye morphogenesis and photoreceptor differentiation, and its loss of function may contribute to the development of human coloboma.


Asunto(s)
Ataxina-7/deficiencia , Coloboma/etiología , Coloboma/metabolismo , Predisposición Genética a la Enfermedad , Células Fotorreceptoras/metabolismo , Subunidades de Proteína/deficiencia , Transactivadores/genética , Animales , Animales Modificados Genéticamente , Biomarcadores , Tipificación del Cuerpo/genética , Diferenciación Celular , Coloboma/patología , Modelos Animales de Enfermedad , Edición Génica , Regulación de la Expresión Génica , Histonas/metabolismo , Inmunohistoquímica , Modelos Biológicos , Nervio Óptico/embriología , Nervio Óptico/metabolismo , Organogénesis/genética , Fenotipo , Células Fotorreceptoras/patología , Procesamiento Proteico-Postraduccional , Transactivadores/química , Transactivadores/metabolismo , Pez Cebra
14.
J Proteome Res ; 17(11): 3959-3975, 2018 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-30285449

RESUMEN

The α7-nicotinic acetylcholine receptor (α7-nAChR) is a ligand-gated ion channel that is expressed widely in vertebrates and is the principal high-affinity α-bungarotoxin (α-bgtx) binding protein in the mammalian CNS. α7-nAChRs associate with proteins that can modulate its properties. The α7-nAChR interactome is the summation of proteins interacting or associating with α7-nAChRs in a protein complex. To identify an α7-nAChR interactome in neural tissue, we isolated α-bgtx-affinity protein complexes from wild-type and α7-nAChR knockout (α7 KO) mouse whole brain tissue homogenates using α-bgtx-affinity beads. Affinity precipitated proteins were trypsinized and analyzed with an Orbitrap Fusion mass spectrometer. Proteins isolated with the α7-nAChR specific ligand, α-bgtx, were determined to be α7-nAChR associated proteins. The α7-nAChR subunit and 120 additional proteins were identified. Additionally, 369 proteins were identified as binding to α-bgtx in the absence of α7-nAChR expression, thereby identifying nonspecific proteins for α7-nAChR investigations using α-bgtx enrichment. These results expand on our previous investigations of α7-nAChR interacting proteins using α-bgtx-affinity bead isolation by controlling for differences between α7-nAChR and α-bgtx-specific proteins, developing an improved protein isolation methodology, and incorporating the latest technology in mass spectrometry. The α7-nAChR interactome identified in this study includes proteins associated with the expression, localization, function, or modulation of α7-nAChRs, and it provides a foundation for future studies to elucidate how these interactions contribute to human disease.


Asunto(s)
Encéfalo/metabolismo , Bungarotoxinas/química , Mapeo de Interacción de Proteínas/métodos , Subunidades de Proteína/química , Proteoma/metabolismo , Receptor Nicotínico de Acetilcolina alfa 7/química , Animales , Encéfalo/citología , Bungarotoxinas/metabolismo , Cromatografía de Afinidad , Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/citología , Neuronas/metabolismo , Péptidos/análisis , Unión Proteica , Subunidades de Proteína/deficiencia , Subunidades de Proteína/genética , Proteolisis , Proteoma/genética , Proteómica/métodos , Receptor Nicotínico de Acetilcolina alfa 7/deficiencia , Receptor Nicotínico de Acetilcolina alfa 7/genética
15.
Cell ; 174(6): 1436-1449.e20, 2018 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-30146163

RESUMEN

Synaptic vesicle and active zone proteins are required for synaptogenesis. The molecular mechanisms for coordinated synthesis of these proteins are not understood. Using forward genetic screens, we identified the conserved THO nuclear export complex (THOC) as an important regulator of presynapse development in C. elegans dopaminergic neurons. In THOC mutants, synaptic messenger RNAs are retained in the nucleus, resulting in dramatic decrease of synaptic protein expression, near complete loss of synapses, and compromised dopamine function. CRE binding protein (CREB) interacts with THOC to mark synaptic transcripts for efficient nuclear export. Deletion of Thoc5, a THOC subunit, in mouse dopaminergic neurons causes severe defects in synapse maintenance and subsequent neuronal death in the substantia nigra compacta. These cellular defects lead to abrogated dopamine release, ataxia, and animal death. Together, our results argue that nuclear export mechanisms can select specific mRNAs and be a rate-limiting step for neuronal differentiation and survival.


Asunto(s)
Proteínas de Caenorhabditis elegans/genética , Neuronas Dopaminérgicas/metabolismo , Proteínas Nucleares/genética , Sinapsis/metabolismo , Transporte Activo de Núcleo Celular , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Animales , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/metabolismo , Señalización del Calcio , Núcleo Celular/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutagénesis , Mutación Missense , Proteínas Nucleares/deficiencia , Proteínas Nucleares/metabolismo , Subunidades de Proteína/deficiencia , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo
16.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1863(9): 1121-1131, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29902570

RESUMEN

Phosphoinositides are bioactive lipids essential in the regulation of cell signaling as well as cytoskeleton and membrane dynamics. Their metabolism is highly active in blood platelets where they play a critical role during activation, at least through two well identified pathways involving phospholipase C and phosphoinositide 3-kinases (PI3K). Here, using a sensitive high-performance liquid chromatography-mass spectrometry method recently developed, we monitored for the first time the profiling of phosphatidylinositol (PI), PIP, PIP2 and PIP3 molecular species (fatty-acyl profiles) in human and mouse platelets during the course of stimulation by thrombin and collagen-related peptide. Furthermore, using class IA PI3K p110α or p110ß deficient mouse platelets and a pharmacological inhibitor, we show the crucial role of p110ß and the more subtle role of p110α in the production of PIP3 molecular species following stimulation. This comprehensive platelet phosphoinositides profiling provides important resources for future studies and reveals new information on phosphoinositides biology, similarities and differences in mouse and human platelets and unexpected dramatic increase in low-abundance molecular species of PIP2 during stimulation, opening new perspectives in phosphoinositide signaling in platelets.


Asunto(s)
Plaquetas/efectos de los fármacos , Fosfatidilinositol 3-Quinasa Clase I/genética , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Animales , Plaquetas/citología , Plaquetas/metabolismo , Proteínas Portadoras/farmacología , Fosfatidilinositol 3-Quinasa Clase I/antagonistas & inhibidores , Fosfatidilinositol 3-Quinasa Clase I/deficiencia , Inhibidores Enzimáticos/farmacología , Expresión Génica , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Péptidos/farmacología , Activación Plaquetaria/efectos de los fármacos , Cultivo Primario de Células , Subunidades de Proteína/antagonistas & inhibidores , Subunidades de Proteína/deficiencia , Subunidades de Proteína/genética , Pirimidinonas/farmacología , Trombina/farmacología , ortoaminobenzoatos/farmacología
17.
Mitochondrion ; 38: 48-57, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28801230

RESUMEN

Proteomic analyses were carried out on isolated mitochondrial samples of C. albicans from gene-deleted mutants (nuo1Δ, nuo2Δ and goa1Δ) as well as the parental strain in order to better understand the contribution of these three fungal-specific mitochondrial ETC complex I (CI) subunits to cellular activities. Herein, we identify 2333 putative proteins from four strains, in which a total of 663 proteins (28.5%) are putatively located in mitochondria. Comparison of protein abundances between mutants and the parental strain reveal 146 differentially-expressed proteins, of which 78 are decreased and 68 are increased in at least one mutant. The common changes across the three mutants include the down-regulation of nuclear-encoded CI subunit proteins as well as phospholipid, ergosterol and cell wall mannan synthesis, and up-regulated proteins in CIV and the alternative oxidase (AOX2). As for gene-specific functions, we find that NUO1 participates in nucleotide synthesis and ribosomal biogenesis; NUO2 is involved in vesicle trafficking; and GOA1 appears to regulate membrane transporter proteins, ROS removal, and substrates trafficking between peroxisomes and mitochondria. The proteomic view of general as well as mutant-specific proteins further extends our understanding of the functional roles of non-mammalian CI-specific subunit proteins in cell processes. Particularly intriguing is the confirmation of a regulatory role for GOA1 on ETC function, a protein found almost exclusively in Candida species. SIGNIFICANCE: Fungal mitochondria are critical for fungal pathogenesis. The absence of any of the three fungal specific CI subunits in mitochondria causes an avirulence phenotype of C. albicans in a murine model of invasive disease. As model yeast (Saccharomyces cerevisiae) lacks a CI and is rarely a pathogen of humans, C. albicans is a better choice for establishing a link between mitochondrial CI and pathogenesis. Apart from the general effects of CI mutants on respiration, previous phenotyping of these mutants were quite similar to each other or to CI conservative subunit. By comparison to transcriptional data, the proteomic data obtained in this study indicate that biosynthetic events in each mutant such as cell wall and cell membrane phospholipids and ergosterol are generally decreased in both transcriptomal and translational levels. However, in the case of mitochondrial function, glycolysis/gluconeogenesis, and ROS scavengers, often gene changes are opposite that of proteomic data in mutants. We hypothesize that the loss of energy production in mutants is compensated by increases in protein levels of glycolysis, gluconeogenesis, and anti-ROS scavengers that at least extend mutant survival.


Asunto(s)
Candida albicans/enzimología , Complejo I de Transporte de Electrón/deficiencia , Proteínas Fúngicas/análisis , Mitocondrias/enzimología , Proteoma/análisis , Eliminación de Gen , Subunidades de Proteína/deficiencia , Saccharomyces cerevisiae
18.
Sci Rep ; 7(1): 17055, 2017 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-29213114

RESUMEN

Chromatin remodelers have emerged as prominent regulators of epigenetic processes and potential drivers of various human pathologies. The multi-subunit chromatin-remodeling SWI/SNF complex determines gene expression programs and, consequently, contributes to the differentiation, maturation and plasticity of neurons. Here, we investigate the elusive biological function of Bcl7a and Bcl7b, two newly identified subunits of the SWI/SNF complex that are highly expressed throughout the brain. We generated ubiquitous and neuron-specific Bcl7a and Bcl7b single and double knockout mice. We provide evidence that Bcl7b is dispensable for animal survival as well as behavioral plasticity. Conversely, ubiquitous Bcl7a knockout results in perinatal lethality, while genetic deletion of Bcl7a in postmitotic neurons elicits motor abnormalities and affects dendritic branching of Purkinje cells, with no obvious synergistic relationship with Bcl7b. Collectively, our findings reveal novel insights into the cellular processes linked to BCL7-containing SWI/SNF complexes and their unrecognized roles in the brain.


Asunto(s)
Conducta Animal/fisiología , Proteínas de Microfilamentos/metabolismo , Animales , Peso Corporal , Encéfalo/metabolismo , Células Cultivadas , Femenino , Locomoción/fisiología , Masculino , Ratones , Ratones Noqueados , Proteínas de Microfilamentos/deficiencia , Proteínas de Microfilamentos/genética , Subunidades de Proteína/deficiencia , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Células de Purkinje/citología , Células de Purkinje/metabolismo
19.
Pathog Dis ; 75(6)2017 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-28859308

RESUMEN

Salmonella enterica Enteritidis forms biofilms and survives in agricultural environments, infecting poultry and eggs. Bacteria in biofilms are difficult to eradicate compared to planktonic cells, causing serious problems in industry and public health. In this study, we evaluated the role of ihfA and ihfB in biofilm formation by S. enterica Enteritidis by employing different microbiology techniques. Our data indicate that ihf mutant strains are impaired in biofilm formation, showing a reduction in matrix formation and a decrease in viability and metabolic activity. Phenotypic analysis also showed that deletion of ihf causes a deficiency in curli fimbriae expression, cellulose production and pellicle formation. These results show that integration host factor has an important regulatory role in biofilm formation by S. enterica Enteritidis.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Fimbrias Bacterianas/genética , Regulación Bacteriana de la Expresión Génica , Factores de Integración del Huésped/genética , Plancton/genética , Salmonella enteritidis/genética , Celulosa/biosíntesis , Fimbrias Bacterianas/metabolismo , Eliminación de Gen , Aptitud Genética , Factores de Integración del Huésped/deficiencia , Plancton/crecimiento & desarrollo , Plancton/metabolismo , Polisacáridos Bacterianos/biosíntesis , Polisacáridos Bacterianos/deficiencia , Subunidades de Proteína/deficiencia , Subunidades de Proteína/genética , Salmonella enteritidis/crecimiento & desarrollo , Salmonella enteritidis/metabolismo , Salmonella enteritidis/patogenicidad
20.
Sci Rep ; 7(1): 7424, 2017 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-28785046

RESUMEN

The GluA1 subunit of the AMPA receptor has been implicated in schizophrenia. While GluA1 is important for cognition, it is not clear what the role of GluA1 is in hedonic responses that are relevant to the negative symptoms of disorders such as schizophrenia. Here, we tested mice that lack GluA1 (Gria1 -/- mice) on consumption of sucrose solutions using a licking microstructure analysis. GluA1 deletion drastically reduced palatability (as measured by the mean lick cluster size) across a range of sucrose concentrations. Although initial lick rates were reduced, measures of consumption across long periods of access to sucrose solutions were not affected by GluA1 deletion and Gria1 -/- mice showed normal satiety responses to high sucrose concentrations. GluA1 deletion also failed to impair flavour conditioning, in which increased intake of a flavour occurred as a consequence of prior pairing with a high sucrose concentration. These results demonstrate that GluA1 plays a role in responding on the basis of palatability rather than other properties, such as the automatic and learnt post-ingestive, nutritional consequences of sucrose. Therefore, Gria1 -/- mice provide a potential model of anhedonia, adding converging evidence to the role of glutamatergic dysfunction in various symptoms of schizophrenia and related disorders.


Asunto(s)
Conducta Alimentaria , Subunidades de Proteína/deficiencia , Receptores AMPA/deficiencia , Respuesta de Saciedad , Sacarosa/metabolismo , Animales , Eliminación de Gen , Ratones , Ratones Noqueados
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA