Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 525
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Plant Cell Physiol ; 61(9): 1669-1682, 2020 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-32645157

RESUMO

Sugars Will Eventually be Exported Transporters (SWEETs) are important in plant biological processes. Expression levels of CsSWEET1a and CsSWEET17 are induced by cold acclimation (CA) and cold stress in Camellia sinensis. Here, we found that CsSWEET17 was alternatively spliced, and its exclusion (Ex) transcript was associated with the CA process. Both plasma membrane-localized CsSWEET1a and CsSWEET17 transport hexoses, but cytoplasm-localized CsSWEET17-Ex does not. These results indicate that alternative splicing may be involved in regulating the function of SWEET transporters in response to low temperature in plants. The extra C-terminal of CsSWEET17, which is not found in the tonoplast fructose transporter AtSWEET17, did not affect its plasma membrane localization but promoted its sugar transport activities. The overexpression (OE) of CsSWEET1a and CsSWEET17 genes resulted in an increased sugar uptake in Arabidopsis, affecting plant germination and growth. The leaf and seed sizes of the CsSWEET17-OE lines were significantly larger than those of the wild type. Moreover, the OE of CsSWEET1a and CsSWEET17 significantly reduced the relative electrolyte leakage levels under freezing stress. Compared with the wild type, the expression of AtCWINV genes was suppressed in both CsSWEET1a-OE and CsSWEET17-OE lines, indicating the alteration in sugar contents in the cell walls of the OE lines. Furthermore, the interaction between CsSWEET1a and CsSWEET17 was confirmed using yeast two-hybrid and bimolecular fluorescence complementation assays. We showed that CsSWEET1a and CsSWEET17 form homo-/heterodimers in the plasma membrane and mediate the partitioning of sugars between the cytoplasm and the apoplast, thereby regulating plant growth and freezing tolerance.


Assuntos
Camellia sinensis/metabolismo , Membrana Celular/metabolismo , Proteínas de Transporte de Monossacarídeos/fisiologia , Proteínas de Plantas/fisiologia , Processamento Alternativo , Arabidopsis , Camellia sinensis/crescimento & desenvolvimento , Camellia sinensis/fisiologia , Resposta ao Choque Frio , Congelamento , Germinação , Glucose/metabolismo , Proteínas de Transporte de Monossacarídeos/metabolismo , Folhas de Planta/metabolismo , Proteínas de Plantas/metabolismo , Plantas Geneticamente Modificadas , beta-Frutofuranosidase/metabolismo
2.
Plant Physiol ; 179(4): 1285-1297, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30305371

RESUMO

Fungal pathogens are a major constraint to global crop production; hence, plant genes encoding pathogen resistance are important tools for combating disease. A few resistance genes identified to date provide partial, durable resistance to multiple pathogens and the wheat (Triticum aestivum) Lr67 hexose transporter variant (Lr67res) fits into this category. Two amino acids differ between the wild-type and resistant alleles - G144R and V387L. Exome sequence data from 267 barley (Hordeum vulgare) landraces and wild accessions was screened and neither of the Lr67res mutations was detected. The barley ortholog of Lr67, HvSTP13, was functionally characterized in yeast as a high affinity hexose transporter. The G144R mutation was introduced into HvSTP13 and abolished Glc uptake, whereas the V387L mutation reduced Glc uptake by ∼ 50%. Glc transport by HvSTP13 heterologously expressed in yeast was reduced when coexpressed with Lr67res Stable transgenic Lr67res barley lines exhibited seedling resistance to the barley-specific pathogens Puccinia hordei and Blumeria graminis f. sp. hordei, which cause leaf rust and powdery mildew, respectively. Barley plants expressing Lr67res exhibited early senescence and higher pathogenesis-related (PR) gene expression. Unlike previous observations implicating flavonoids in the resistance of transgenic sorghum (Sorghum bicolor) expressing Lr34res, another wheat multipathogen resistance gene, barley flavonoids are unlikely to have a role in Lr67res-mediated resistance. Similar to observations made in yeast, Lr67res reduced Glc uptake in planta These results confirm that the pathway by which Lr67res confers resistance to fungal pathogens is conserved between wheat and barley.


Assuntos
Hordeum/imunologia , Proteínas de Transporte de Monossacarídeos/fisiologia , Triticum/genética , Flavonoides/metabolismo , Expressão Gênica , Hordeum/genética , Hordeum/metabolismo , Mutação , Doenças das Plantas/imunologia , Imunidade Vegetal , Proteínas de Plantas/fisiologia , Plantas Geneticamente Modificadas/genética , Plantas Geneticamente Modificadas/imunologia , Plantas Geneticamente Modificadas/metabolismo
3.
Plant Mol Biol ; 100(4-5): 351-365, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31030374

RESUMO

KEY MESSAGE: Present review describes the structure, evolution, transport mechanism and physiological functions of SWEETs. Their application using TALENs and CRISPR/CAS9 based genomic editing approach is discussed. Sugars Will Eventually be Exported Transporters (SWEET) proteins were first identified in plants as the novel family of sugar transporters which mediates the translocation of sugars across cell membranes. The SWEET family of sugar transporters is unique in terms of their structure which contains seven predicted transmembrane domains with two internal triple-helix bundles which possibly originate due to prokaryotic gene duplication. SWEETs perform diverse physiological functions such as pollen nutrition, nectar secretion, seed filling, phloem loading, and pathogen nutrition which we have discussed in the present review. We also discuss how transcriptional activator-like effector nucleases (TALENs) and CRISPR/CAS9 genome editing tools are used to engineer SWEET mutants which modulate pathogen resistance in plants and its applications in the field of agriculture. The expression of SWEETs promises to implement insights into many other cellular transport mechanisms. To conclude, the present review highlights the recent aspects which will further develop better understanding of molecular evolution, structure, and function of SWEET transporters in plants.


Assuntos
Proteínas de Transporte de Monossacarídeos/fisiologia , Proteínas de Plantas/fisiologia , Membrana Celular/metabolismo , Resistência à Doença , Evolução Molecular , Giberelinas/metabolismo , Modelos Moleculares , Proteínas de Transporte de Monossacarídeos/química , Floema/metabolismo , Proteínas de Plantas/química , Plantas/metabolismo , Plantas/microbiologia , Domínios Proteicos , Análise de Sequência de Proteína
4.
Biochim Biophys Acta Mol Cell Res ; 1864(5): 825-838, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28167211

RESUMO

SLC35A4 has been classified in the SLC35A subfamily based on amino acid sequence homology. Most of the proteins belonging to the SLC35 family act as transporters of nucleotide sugars. In this study, the subcellular localization of endogenous SLC35A4 was determined via immunofluorescence staining, and it was demonstrated that SLC35A4 localizes mainly to the Golgi apparatus. In silico topology prediction suggests that SLC35A4 has an uneven number of transmembrane domains and its N-terminus is directed towards the Golgi lumen. However, an experimental assay refuted this prediction: SLC35A4 has an even number of transmembrane regions with both termini facing the cytosol. In vivo interaction analysis using the FLIM-FRET approach revealed that SLC35A4 neither forms homomers nor associates with other members of the SLC35A subfamily except SLC35A5. Additional assays demonstrated that endogenous SLC35A4 is 10 to 40nm proximal to SLC35A2 and SLC35A3. To determine SLC35A4 function SLC35A4 knock-out cells were generated with the CRISPR-Cas9 approach. Although no significant changes in glycosylation were observed, the introduced mutation influenced the subcellular distribution of the SLC35A2/SLC35A3 complexes. Additional FLIM-FRET experiments revealed that overexpression of SLC35A4-BFP together with SLC35A3 and the SLC35A2-Golgi splice variant negatively affects the interaction between the two latter proteins. The results presented here strongly indicate a modulatory role for SLC35A4 in intracellular trafficking of SLC35A2/SLC35A3 complexes.


Assuntos
Proteínas de Transporte de Monossacarídeos/fisiologia , Proteínas de Transporte de Nucleotídeos/fisiologia , Sequência de Aminoácidos , Animais , Transporte Biológico/genética , Células COS , Metabolismo dos Carboidratos/genética , Linhagem Celular Tumoral , Chlorocebus aethiops , Cães , Células HEK293 , Células Hep G2 , Humanos , Células Madin Darby de Rim Canino , Proteínas de Transporte de Monossacarídeos/química , Proteínas de Transporte de Monossacarídeos/genética , Proteínas de Transporte de Nucleotídeos/química , Proteínas de Transporte de Nucleotídeos/genética , Homologia de Sequência de Aminoácidos
5.
Proc Natl Acad Sci U S A ; 112(18): 5809-14, 2015 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-25901322

RESUMO

The innate immune system is the first line of defense encountered by invading pathogens. Delayed and/or inadequate innate immune responses can result in failure to combat pathogens, whereas excessive and/or inappropriate responses cause runaway inflammation. Therefore, immune responses are tightly regulated from initiation to resolution and are repressed during the steady state. It is well known that glycans presented on pathogens play important roles in pathogen recognition and the interactions between host molecules and microbes; however, the function of glycans of host organisms in innate immune responses is less well known. Here, we show that innate immune quiescence and strength of the immune response are controlled by host glycosylation involving a novel UDP-galactose transporter called Senju. In senju mutants, reduced expression of galactose-containing glycans resulted in hyperactivation of the Toll signaling pathway in the absence of immune challenges. Genetic epistasis and biochemical analyses revealed that Senju regulates the Toll signaling pathway at a step that converts Toll ligand Spatzle to its active form. Interestingly, Toll activation in immune-challenged wild type (WT) flies reduced the expression of galactose-containing glycans. Suppression of the degalactosylation by senju overexpression resulted in reduced induction of Toll-dependent expression of an antimicrobial peptide, Drosomycin, and increased susceptibility to infection with Gram-positive bacteria. These data suggest that Senju-mediated galactosylation suppresses undesirable Toll signaling activation during the steady state; however, Toll activation in response to infection leads to degalactosylation, which raises the immune response to an adequate level and contributes to the prompt elimination of pathogens.


Assuntos
Proteínas de Drosophila/fisiologia , Drosophila melanogaster/imunologia , Glicosilação , Imunidade Inata , Proteínas de Transporte de Monossacarídeos/metabolismo , Proteínas de Transporte de Monossacarídeos/fisiologia , Animais , Animais Geneticamente Modificados , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Epistasia Genética , Galactose/química , Sistema Imunitário , Lectinas/química , Proteínas de Transporte de Monossacarídeos/genética , Mutação , Polissacarídeos/química , Recombinação Genética , Receptores Toll-Like/metabolismo
6.
Microb Pathog ; 113: 321-329, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29111323

RESUMO

The LuxS/AI-2 quorum sensing mechanism can regulate the physiological functions of avian pathogenic Escherichia coli (APEC) through internalization of the small molecule autoinducer-2 (AI-2). The ptsI gene encodes enzyme I, which participates in the phosphotransferase system (PTS) that regulates the virulence and AI-2 internalization of bacteria. The aim of the present study was to determine the effect of ptsI on AI-2 internalization and other pathogenesis process in APEC using a ptsI mutant of the APEC strain DE17 (serotype O2), namely DE17ΔptsI. The results showed that deletion of the ptsI gene changed the rdar (red dry and rough) morphotype and decreased motility and biofilm formation in APEC (p < 0.05). Furthermore, scanning electron microscopy showed that the biofilm structure of DE17ΔptsI became sparse and more extracellular, as compared with the wild-type strain DE17. Moreover, AI-2 assay showed that AI-2 was internalized by DE17ΔptsI, while the recombinant PtsI protein had no AI-2 binding activity. Furthermore, deletion of the ptsI gene in APEC significantly increased adherence to DF-1 cells (p < 0.05). The 50% lethal dose of DE17ΔptsI was decreased by 17.8-fold and the bacterial loads of DE17ΔptsI were decreased by 13600-, 68.5-, 131-, and 3600-fold in the blood, liver, spleen, and kidney, respectively, as compared to the DE17. Moreover, histopathological analysis showed that the mutant DE17ΔptsI was associated with reduced pathological changes in the heart, liver, spleen, and kidney of ducklings, respectively, as compared to the wild-type strain DE17. The results of this study will benefit further studies on the functions of the ptsI in APEC.


Assuntos
Doenças das Aves/microbiologia , Infecções por Escherichia coli/microbiologia , Infecções por Escherichia coli/veterinária , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/fisiologia , Escherichia coli/patogenicidade , Homosserina/análogos & derivados , Proteínas de Transporte de Monossacarídeos/genética , Proteínas de Transporte de Monossacarídeos/fisiologia , Sistema Fosfotransferase de Açúcar do Fosfoenolpiruvato/genética , Sistema Fosfotransferase de Açúcar do Fosfoenolpiruvato/fisiologia , Animais , Carga Bacteriana , Proteínas de Bactérias/genética , Proteínas de Bactérias/fisiologia , Biofilmes/crescimento & desenvolvimento , Liases de Carbono-Enxofre , Linhagem Celular , China , Modelos Animais de Doenças , Patos , Escherichia coli/genética , Infecções por Escherichia coli/patologia , Deleção de Genes , Perfilação da Expressão Gênica , Coração/microbiologia , Homosserina/genética , Homosserina/fisiologia , Rim/microbiologia , Rim/patologia , Lactonas , Fígado/microbiologia , Fígado/patologia , Miocárdio/patologia , Fosfotransferases , Percepção de Quorum , Baço/microbiologia , Baço/patologia , Fatores de Virulência/genética
7.
Can J Microbiol ; 63(4): 287-295, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28177798

RESUMO

Bacterial noncoding RNAs (ncRNA) regulate diverse cellular processes, including virulence and environmental fitness. The malS 5' untranslated region (named malS-5'UTR) was identified as a regulatory ncRNA that increases the invasive capacity of Salmonella enterica serovar Typhi. An IntaRNA search suggested base pairing between malS-5'UTR and hisG mRNA, a key gene in the histidine biosynthetic pathway. Overexpression of malS-5'UTR markedly reduced bacterial growth in minimal medium without histidine. Overexpression of malS-5'UTR increased mRNA from his operon genes, independently of the bax gene, and decreased HisG protein in Salmonella Typhi. RNA structure analysis showed base pairing of the malS-5'UTR RNA with the hisG mRNA across the ribosome binding site. Thus, we propose that malS-5'UTR inhibited hisG translation, probably by base pairing to the Shine-Dalgarno sequence.


Assuntos
Regiões 5' não Traduzidas/genética , Proteínas de Bactérias/fisiologia , Histidina/biossíntese , Proteínas de Transporte de Monossacarídeos/fisiologia , Salmonella typhi/metabolismo , Vias Biossintéticas , Salmonella typhi/genética
9.
Plant Cell Environ ; 39(6): 1366-80, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26670204

RESUMO

Sucrose accumulation in leaves in response to various abiotic stresses suggests a specific role of this disaccharide for stress tolerance and adaptation. The high-affinity transporter StSUT1 undergoes substrate-induced endocytosis presenting the question as to whether altered sucrose accumulation in leaves in response to stresses is also related to enhanced endocytosis or altered activity of the sucrose transporter. StSUT1 is known to interact with several stress-inducible proteins; here we investigated whether one of the interacting candidates, StPDI1, affects its subcellular localization in response to stress: StPDI1 expression is induced by ER-stress and salt. Both proteins, StSUT1 and StPDI1, were found in the detergent resistant membrane (DRM) fraction, and this might affect internalization. Knockdown of StPDI1 expression severely affects abiotic stress tolerance of transgenic potato plants. Analysis of these plants does not reveal modified subcellular localization or endocytosis of StSUT1, but rather a disturbed redox homeostasis, reduced detoxification of reactive oxygen species and effects on primary metabolism. Parallel observations with other StSUT1-interacting proteins are discussed. The redox status in leaves seems to be linked to the sugar status in response to various stress stimuli and to play a role in stress tolerance.


Assuntos
Homeostase , Proteínas de Transporte de Monossacarídeos/fisiologia , Oxirredução , Proteínas de Plantas/fisiologia , Isomerases de Dissulfetos de Proteínas/fisiologia , Sacarose/metabolismo , Clonagem Molecular , Técnicas de Silenciamento de Genes , Homeostase/fisiologia , Folhas de Planta/metabolismo , Folhas de Planta/fisiologia , Plantas Geneticamente Modificadas , Reação em Cadeia da Polimerase em Tempo Real , Solanum tuberosum/embriologia , Solanum tuberosum/fisiologia , Estresse Fisiológico/fisiologia
10.
Reproduction ; 152(1): 91-9, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27107036

RESUMO

The Mfsd14a gene, previously called Hiat1, encodes a transmembrane protein of unknown function with homology to the solute carrier protein family. To study the function of the MFSD14A protein, mutant mice (Mus musculus, strain 129S6Sv/Ev) were generated with the Mfsd14a gene disrupted with a LacZ reporter gene. Homozygous mutant mice are viable and healthy, but males are sterile due to a 100-fold reduction in the number of spermatozoa in the vas deferens. Male mice have adequate levels of testosterone and show normal copulatory behaviour. The few spermatozoa that are formed show rounded head defects similar to those found in humans with globozoospermia. Spermatogenesis proceeds normally up to the round spermatid stage, but the subsequent structural changes associated with spermiogenesis are severely disrupted with failure of acrosome formation, sperm head condensation and mitochondrial localization to the mid-piece of the sperm. Staining for ß-galactosidase activity as a surrogate for Mfsd14a expression indicates expression in Sertoli cells, suggesting that MFSD14A may transport a solute from the bloodstream that is required for spermiogenesis.


Assuntos
Infertilidade Masculina/etiologia , Proteínas de Transporte de Monossacarídeos/fisiologia , Células de Sertoli/patologia , Espermatogênese/fisiologia , Teratozoospermia/complicações , Animais , Células Cultivadas , Feminino , Infertilidade Masculina/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Células de Sertoli/metabolismo , Espermatozoides/metabolismo , Teratozoospermia/patologia
11.
Brain ; 138(Pt 6): 1531-47, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25823473

RESUMO

C-type lectins are key players in immune regulation by driving distinct functions of antigen-presenting cells. The C-type lectin CLEC16A gene is located at 16p13, a susceptibility locus for several autoimmune diseases, including multiple sclerosis. However, the function of this gene and its potential contribution to these diseases in humans are poorly understood. In this study, we found a strong upregulation of CLEC16A expression in the white matter of multiple sclerosis patients (n = 14) compared to non-demented controls (n = 11), mainly in perivascular leukocyte infiltrates. Moreover, CLEC16A levels were significantly enhanced in peripheral blood mononuclear cells of multiple sclerosis patients (n = 69) versus healthy controls (n = 46). In peripheral blood mononuclear cells, CLEC16A was most abundant in monocyte-derived dendritic cells, in which it strongly co-localized with human leukocyte antigen class II. Treatment of these professional antigen-presenting cells with vitamin D, a key protective environmental factor in multiple sclerosis, downmodulated CLEC16A in parallel with human leukocyte antigen class II. Knockdown of CLEC16A in distinct types of model and primary antigen-presenting cells resulted in severely impaired cytoplasmic distribution and formation of human leucocyte antigen class II-positive late endosomes, as determined by immunofluorescence and electron microscopy. Mechanistically, CLEC16A participated in the molecular machinery of human leukocyte antigen class II-positive late endosome formation and trafficking to perinuclear regions, involving the dynein motor complex. By performing co-immunoprecipitations, we found that CLEC16A directly binds to two critical members of this complex, RILP and the HOPS complex. CLEC16A silencing in antigen-presenting cells disturbed RILP-mediated recruitment of human leukocyte antigen class II-positive late endosomes to perinuclear regions. Together, we identify CLEC16A as a pivotal gene in multiple sclerosis that serves as a direct regulator of the human leukocyte antigen class II pathway in antigen-presenting cells. These findings are a first step in coupling multiple sclerosis-associated genes to the regulation of the strongest genetic factor in multiple sclerosis, human leukocyte antigen class II.


Assuntos
Endossomos/metabolismo , Predisposição Genética para Doença/genética , Antígenos de Histocompatibilidade Classe II/biossíntese , Lectinas Tipo C/fisiologia , Proteínas de Transporte de Monossacarídeos/fisiologia , Esclerose Múltipla/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Adolescente , Adulto , Idoso , Células Apresentadoras de Antígenos/efeitos dos fármacos , Células Apresentadoras de Antígenos/metabolismo , Estudos de Casos e Controles , Células Cultivadas , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Feminino , Técnicas de Silenciamento de Genes , Humanos , Lectinas Tipo C/genética , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/ultraestrutura , Masculino , Pessoa de Meia-Idade , Proteínas de Transporte de Monossacarídeos/genética , Transporte Proteico/genética , RNA Interferente Pequeno/farmacologia , Regulação para Cima/efeitos dos fármacos , Vitamina D/farmacologia , Substância Branca/metabolismo , Adulto Jovem
12.
Clin Exp Nephrol ; 20(6): 845-852, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26935049

RESUMO

BACKGROUND: Hypouricemia is pathognomonic in syndrome of inappropriate secretion of antidiuretic hormone (SIADH) but the underlying mechanism remains unclear. Based on the previous studies, we hypothesized that V1a receptor may play a principal role in inducing hypouricemia in SIADH and examined uric acid metabolism using a rat model. METHODS: Terlipressin (25 ng/h), a selective V1a agonist, was subcutaneously infused to 7-week-old male Wistar rats (n = 9). Control rats were infused with normal saline (n = 9). The rats were sacrificed to obtain kidney tissues 3 days after treatment. In addition to electrolyte metabolism, changes in expressions of the urate transporters including URAT1 (SLC22A12), GLUT9 (SLC2A9), ABCG2 and NPT1 (SLC17A1) were examined by western blotting and immunohistochemistry. RESULTS: In the terlipressin-treated rats, serum uric acid (UA) significantly decreased and the excretion of urinary UA significantly increased, resulting in marked increase in fractional excretion of UA. Although no change in the expression of URAT1, GLUT9 expression significantly decreased whereas the expressions of ABCG2 and NPT1 significantly increased in the terlipressin group. The results of immunohistochemistry corroborated with those of the western blotting. Aquaporin 2 expression did not change in the medulla, suggesting the independence of V2 receptor stimulation. CONCLUSION: Stimulation of V1a receptor induces the downregulation of GLUT9, reabsorption urate transporter, together with the upregulation of ABCG2 and NPT1, secretion urate transporters, all changes of which clearly lead to increase in renal UA clearance. Hypouricemia seen in SIADH is attributable to V1a receptor stimulation.


Assuntos
Síndrome de Secreção Inadequada de HAD/complicações , Transportadores de Ânions Orgânicos/fisiologia , Receptores de Vasopressinas/fisiologia , Erros Inatos do Transporte Tubular Renal/etiologia , Ácido Úrico/metabolismo , Cálculos Urinários/etiologia , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/fisiologia , Animais , Aquaporina 2/análise , Aquaporina 2/fisiologia , Lipressina/análogos & derivados , Lipressina/farmacologia , Masculino , Taxa de Depuração Metabólica , Proteínas de Transporte de Monossacarídeos/análise , Proteínas de Transporte de Monossacarídeos/fisiologia , Ratos , Ratos Wistar , Proteínas Cotransportadoras de Sódio-Fosfato Tipo III/fisiologia , Terlipressina
13.
New Phytol ; 206(3): 1086-1100, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25678342

RESUMO

The smut Ustilago maydis, a ubiquitous pest of corn, is highly adapted to its host to parasitize on its organic carbon sources. We have identified a hexose transporter, Hxt1, as important for fungal development during both the saprophytic and the pathogenic stage of the fungus. Hxt1 was characterized as a high-affinity transporter for glucose, fructose, and mannose; ∆hxt1 strains show significantly reduced growth on these substrates, setting Hxt1 as the main hexose transporter during saprophytic growth. After plant infection, ∆hxt1 strains show decreased symptom development. However, expression of a Hxt1 protein with a mutation leading to constitutively active signaling in the yeast glucose sensors Snf3p and Rgt2p results in completely apathogenic strains. Fungal development is stalled immediately after plant penetration, implying a dual function of Hxt1 as transporter and sensor. As glucose sensors are only known for yeasts, 'transceptor' as Hxt1 may constitute a general mechanism for sensing of glucose in fungi. In U. maydis, Hxt1 links a nutrient-dependent environmental signal to the developmental program during pathogenic development.


Assuntos
Proteínas Fúngicas/fisiologia , Proteínas de Transporte de Monossacarídeos/fisiologia , Ustilago/patogenicidade , Fatores de Virulência/fisiologia , Zea mays/microbiologia , Substituição de Aminoácidos , Frutose/metabolismo , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Glucose/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Proteínas de Transporte de Monossacarídeos/genética , Proteínas de Transporte de Monossacarídeos/metabolismo , Transdução de Sinais , Ustilago/genética , Ustilago/metabolismo , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
14.
Proc Natl Acad Sci U S A ; 109(18): E1072-81, 2012 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-22493244

RESUMO

Autophagy is a self-degradative process in which cellular material is enclosed within autophagosomes and trafficked to lysosomes for degradation. Autophagosomal biogenesis is well described; however mechanisms controlling the growth and ultimate size of autophagosomes are unclear. Here we demonstrate that the Drosophila membrane protein Ema is required for the growth of autophagosomes. In an ema mutant, autophagosomes form in response to starvation and developmental cues, and these autophagosomes can mature into autolysosomes; however the autophagosomes are very small, and autophagy is impaired. In fat body cells, Ema localizes to the Golgi complex and is recruited to the membrane of autophagosomes in response to starvation. The Drosophila Golgi protein Lva also is recruited to the periphery of autophagosomes in response to starvation, and this recruitment requires ema. Therefore, we propose that Golgi is a membrane source for autophagosomal growth and that Ema facilitates this process. Clec16A, the human ortholog of Ema, is a candidate autoimmune susceptibility locus. Expression of Clec16A can rescue the autophagosome size defect in the ema mutant, suggesting that regulation of autophagosome morphogenesis may be a fundamental function of this gene family.


Assuntos
Proteínas de Drosophila/fisiologia , Lectinas Tipo C/fisiologia , Proteínas de Transporte de Monossacarídeos/fisiologia , Animais , Autofagia/genética , Autofagia/fisiologia , Drosophila/citologia , Drosophila/genética , Drosophila/fisiologia , Proteínas de Drosophila/genética , Endossomos/fisiologia , Corpo Adiposo/citologia , Corpo Adiposo/fisiologia , Genes de Insetos , Complexo de Golgi/fisiologia , Humanos , Lectinas Tipo C/genética , Lisossomos/fisiologia , Fusão de Membrana/fisiologia , Microscopia Eletrônica de Transmissão , Proteínas de Transporte de Monossacarídeos/genética , Mutação , Fagossomos/fisiologia , Fagossomos/ultraestrutura
15.
Nat Med ; 13(11): 1363-7, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17952091

RESUMO

Proteoglycans are a family of extracellular macromolecules comprised of glycosaminoglycan chains of a repeated disaccharide linked to a central core protein. Proteoglycans have critical roles in chondrogenesis and skeletal development. The glycosaminoglycan chains found in cartilage proteoglycans are primarily composed of chondroitin sulfate. The integrity of chondroitin sulfate chains is important to cartilage proteoglycan function; however, chondroitin sulfate metabolism in mammals remains poorly understood. The solute carrier-35 D1 (SLC35D1) gene (SLC35D1) encodes an endoplasmic reticulum nucleotide-sugar transporter (NST) that might transport substrates needed for chondroitin sulfate biosynthesis. Here we created Slc35d1-deficient mice that develop a lethal form of skeletal dysplasia with severe shortening of limbs and facial structures. Epiphyseal cartilage in homozygous mutant mice showed a decreased proliferating zone with round chondrocytes, scarce matrices and reduced proteoglycan aggregates. These mice had short, sparse chondroitin sulfate chains caused by a defect in chondroitin sulfate biosynthesis. We also identified that loss-of-function mutations in human SLC35D1 cause Schneckenbecken dysplasia, a severe skeletal dysplasia. Our findings highlight the crucial role of NSTs in proteoglycan function and cartilage metabolism, thus revealing a new paradigm for skeletal disease and glycobiology.


Assuntos
Osso e Ossos/embriologia , Cartilagem/embriologia , Sulfatos de Condroitina/biossíntese , Proteínas de Transporte de Monossacarídeos/fisiologia , Proteínas de Transporte de Nucleotídeos/fisiologia , Animais , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Cartilagem/metabolismo , Cartilagem/patologia , Células Cultivadas , Condrócitos/metabolismo , Condrócitos/patologia , Epífises/embriologia , Epífises/metabolismo , Epífises/patologia , Ossos Faciais/anormalidades , Ossos Faciais/embriologia , Ossos Faciais/metabolismo , Humanos , Deformidades Congênitas dos Membros/embriologia , Deformidades Congênitas dos Membros/genética , Deformidades Congênitas dos Membros/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteínas de Transporte de Monossacarídeos/deficiência , Proteínas de Transporte de Monossacarídeos/genética , Proteínas de Transporte de Nucleotídeos/genética
16.
Biochem J ; 452(3): 489-97, 2013 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-23495665

RESUMO

Successful colonization and survival in variable environments require a competitive advantage during the initial growth phase after experiencing nutrient changes. Starved yeast cells anticipate exposure to glucose by activating the Hxt5p (hexose transporter 5) glucose transporter, which provides an advantage during early phases after glucose resupply. cAMP and glucose FRET (fluorescence resonance energy transfer) sensors were used to identify three signalling pathways that co-operate in the anticipatory Hxt5p activity in glucose-starved cells: as expected the Snf1 (sucrose nonfermenting 1) AMP kinase pathway, but, surprisingly, the sugar-dependent G-protein-coupled Gpr1 (G-protein-coupled receptor 1)/cAMP/PKA (protein kinase A) pathway and the Pho85 (phosphate metabolism 85)/Plc (phospholipase C) 6/7 pathway. Gpr1/cAMP/PKA are key elements of a G-protein-coupled sugar response pathway that produces a transient cAMP peak to induce growth-related genes. A novel function of the Gpr1/cAMP/PKA pathway was identified in glucose-starved cells: during starvation the Gpr1/cAMP/PKA pathway is required to maintain Hxt5p activity in the absence of glucose-induced cAMP spiking. During starvation, cAMP levels remain low triggering expression of HXT5, whereas cAMP spiking leads to a shift to the high capacity Hxt isoforms.


Assuntos
AMP Cíclico/química , Glucose/metabolismo , Proteínas de Transporte de Monossacarídeos/fisiologia , Proteínas de Saccharomyces cerevisiae/fisiologia , Saccharomyces cerevisiae/fisiologia , Adenilato Quinase/fisiologia , Transporte Biológico Ativo , AMP Cíclico/fisiologia , Proteínas de Transporte de Monossacarídeos/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Transdução de Sinais/fisiologia
17.
Biochem Biophys Res Commun ; 410(3): 471-7, 2011 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-21679696

RESUMO

Myo-inositol is one of the major organic osmolytes in the brain and the kidney. The accumulation of intracellular organic osmolytes allows cells to regulate intracellular osmolality without altering cytoplasmic ionic strength and to adapt to hyperosmotic conditions. Two types of myo-inositol transporters, sodium/myo-inositol transporter and H(+)/myo-inositol transporter (HMIT), have been identified. Sodium/myo-inositol transporters are induced by osmotic stress and might be involved in the intracellular accumulation of myo-inositol in mammals. The role of HMIT, however, remains unknown. In the present study, we characterized three Caenorhabditis elegansHMIT genes, hmit-1.1, hmit-1.2, and hmit-1.3. hmit-1.1 was expressed in the intestine, and hmit-1.2 was expressed in the glia and the excretory canal, which is an osmotic regulatory organ that is functionally analogous to the kidney. hmit-1.3 was expressed in the intestine and the glia. The expression of hmit-1.1 and hmit-1.2 but not hmit-1.3, was markedly induced under hyperosmotic conditions. Animals with mutant hmit-1.1 and hmit-1.2 were hypersensitive to osmotic stress. The defects of hmit-1.1 and hmit-1.2 mutants were rescued by hmit-1.1 and hmit-1.2 transgenes, respectively, and by modified human HMIT. In human cell lines, HMIT expression was induced in hyperosmotic conditions. These findings indicate that the C. elegans HMIT family has a crucial role in the osmoprotective response.


Assuntos
Adaptação Fisiológica/fisiologia , Caenorhabditis elegans/fisiologia , Genes Essenciais/fisiologia , Inositol/metabolismo , Proteínas de Transporte de Monossacarídeos/fisiologia , Estresse Fisiológico/fisiologia , Equilíbrio Hidroeletrolítico/fisiologia , Adaptação Fisiológica/genética , Animais , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Linhagem Celular , Deleção de Genes , Proteínas Facilitadoras de Transporte de Glucose/genética , Proteínas Facilitadoras de Transporte de Glucose/fisiologia , Células HEK293 , Humanos , Proteínas de Transporte de Monossacarídeos/genética , Proteínas de Transporte de Monossacarídeos/metabolismo , Pressão Osmótica , Estresse Fisiológico/genética , Distribuição Tecidual , Equilíbrio Hidroeletrolítico/genética
18.
Curr Opin Cell Biol ; 11(4): 496-502, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10449337

RESUMO

Sugar transporters from one group of the major facilitator superfamily of membrane transporters. A conserved common central pore structure lies at the heart of these transporters and diverse functionality is brought about by alterations to this pore or regions associated with it. Recent mutagenesis studies of sugar transporters within the framework of tenable models for the distantly related lactose permease argue that this model is a good paradigm for other members of the major facilitator superfamily.


Assuntos
Proteínas de Transporte de Monossacarídeos/fisiologia , Sequência de Aminoácidos , Escherichia coli/enzimologia , Transportador de Glucose Tipo 1 , Proteínas de Membrana Transportadoras/metabolismo , Modelos Biológicos , Dados de Sequência Molecular , Proteínas de Transporte de Monossacarídeos/química , Conformação Proteica , Relação Estrutura-Atividade
19.
Sci Rep ; 11(1): 9319, 2021 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-33927318

RESUMO

CLEC16A has been shown to play a role in autophagy/mitophagy processes. Additionally, genetic variants in CLEC16A have been implicated in multiple autoimmune diseases. We generated an inducible whole-body knockout, Clec16aΔUBC mice, to investigate the loss of function of CLEC16A. The mice exhibited a neuronal phenotype including tremors and impaired gait that rapidly progressed to dystonic postures. Nerve conduction studies and pathological analysis revealed loss of sensory axons that are associated with this phenotype. Activated microglia and astrocytes were found in regions of the CNS. Several mitochondrial-related proteins were up- or down-regulated. Upregulation of interferon stimulated gene 15 (IGS15) were observed in neuronal tissues. CLEC16A expression inversely related to IGS15 expression. ISG15 may be the link between CLEC16A and downstream autoimmune, inflammatory processes. Our results demonstrate that a whole-body, inducible knockout of Clec16a in mice results in an inflammatory neurodegenerative phenotype resembling spinocerebellar ataxia.


Assuntos
Lectinas Tipo C/fisiologia , Proteínas de Transporte de Monossacarídeos/fisiologia , Doença Autoimune do Sistema Nervoso Experimental , Ataxias Espinocerebelares , Animais , Citocinas/metabolismo , Feminino , Técnicas de Inativação de Genes , Masculino , Camundongos Knockout , Neurônios/ultraestrutura , Ubiquitinas/metabolismo
20.
Cancer Sci ; 101(10): 2227-33, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20701608

RESUMO

The humanized monoclonal antibody (mAb) against CD317 antigen (anti-HM1.24 antibody; AHM), which is highly expressed on multiple myeloma (MM), induces antibody-dependent cellular cytotoxicity (ADCC). However, the antitumor activity of AHM in the clinical setting has not been clearly demonstrated. In this study, we produced defucosylated AHM and evaluated its potency for clinical application by performing autologous ADCC assays against primary MM cells from patients. Defucosylated AHM that was produced in rat myeloma YB2/0 cells expressing a low level of fucosyltransferase (FUT8) showed significant ADCC activity against three out of six primary MM cells in the presence of autologous PBMC, whereas conventional AHM did not. The results indicate that the potency of AHM to induce ADCC against primary MM cells was insufficient, but was significantly augmented by defucosylation. To generate more homogenous defucosylated monoclonal antibodies (mAb) for fermentation, we disrupted the GFT gene that encodes a GDP-fucose transporter in a CHO/DXB11 cell line by sequential homologous recombination. Analysis of the N-linked oligosaccharide in the defucosylated AHM produced by the established GFT(-/-)CHO cell line showed that a majority (93.4%) of the oligosaccharide was fucose free. The GFT(-/-) cells stably produced defucosylated mAb over passages. These results demonstrate that GTF(-/-)CHO-produced defucosylated AHM (GFTKO-AHM) will be a promising new therapeutic antibody against MM in the clinical setting.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Citotoxicidade Celular Dependente de Anticorpos , Antígenos CD/imunologia , Glicoproteínas de Membrana/imunologia , Mieloma Múltiplo/tratamento farmacológico , Animais , Células CHO , Cricetinae , Cricetulus , Proteínas Ligadas por GPI , Glucosiltransferases/fisiologia , Humanos , Proteínas de Transporte de Monossacarídeos/fisiologia , Mieloma Múltiplo/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA