Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Molecules ; 26(18)2021 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-34577156

RESUMEN

DPY19L3 has been identified as a C-mannosyltransferase for thrombospondin type-1 repeat domain-containing proteins. In this study, we focused on the role of DPY19L3 in the myogenic differentiation of C2C12 mouse myoblast cells. We carried out DPY19L3 gene depletion using the CRISPR/Cas9 system. The result showed that these DPY19L3-knockout cells could not be induced for differentiation. Moreover, the phosphorylation levels of MEK/ERK and p70S6K were suppressed in the DPY19L3-knockout cells compared with that of parent cells, suggesting that the protein(s) that is(are) DPY19L3-mediated C-mannosylated and regulate(s) MEK/ERK or p70S6K signaling is(are) required for the differentiation.


Asunto(s)
Diferenciación Celular/genética , Diferenciación Celular/fisiología , Manosiltransferasas/fisiología , Mioblastos/fisiología , Animales , Línea Celular , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Técnicas de Silenciamiento del Gen , Glicosilación , Manosiltransferasas/genética , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Mioblastos/citología , Fosforilación/genética , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Transducción de Señal/genética , Regulación hacia Arriba/genética
2.
Commun Biol ; 4(1): 777, 2021 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-34162996

RESUMEN

Over 100 kinds of proteins are expressed as glycosylphosphatidylinositol (GPI)-anchored proteins (GPI-APs) on the cell surface in mammalian cells. GPI-APs possess unique properties in terms of their intracellular trafficking and association with lipid rafts. Although it is clear that GPI-APs play critical roles in various biological phenomena, it is poorly understood how the GPI moiety contributes to these mechanisms. More than 30 genes are involved in the correct biosynthesis of GPI-APs. We here constructed a cell library in which 32 genes involved in GPI biosynthesis were knocked out in human embryonic kidney 293 cells. Using the cell library, the surface expression and sensitivity to phosphatidylinositol-specific phospholipase C of GPI-APs were analyzed. Furthermore, we identified structural motifs of GPIs that are recognized by a GPI-binding toxin, aerolysin. The cell-based GPI-knockout library could be applied not only to basic researches, but also to applications and methodologies related to GPI-APs.


Asunto(s)
Proteínas Ligadas a GPI/fisiología , Glicosilfosfatidilinositoles/biosíntesis , Toxinas Bacterianas/metabolismo , Técnicas de Inactivación de Genes , Células HEK293 , Humanos , Manosiltransferasas/genética , Manosiltransferasas/fisiología , Proteínas Citotóxicas Formadoras de Poros/metabolismo
3.
Proc Natl Acad Sci U S A ; 118(2)2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33402532

RESUMEN

Pathogenic germline mutations in PIGV lead to glycosylphosphatidylinositol biosynthesis deficiency (GPIBD). Individuals with pathogenic biallelic mutations in genes of the glycosylphosphatidylinositol (GPI)-anchor pathway exhibit cognitive impairments, motor delay, and often epilepsy. Thus far, the pathophysiology underlying the disease remains unclear, and suitable rodent models that mirror all symptoms observed in human patients have not been available. Therefore, we used CRISPR-Cas9 to introduce the most prevalent hypomorphic missense mutation in European patients, Pigv:c.1022C > A (p.A341E), at a site that is conserved in mice. Mirroring the human pathology, mutant Pigv341E mice exhibited deficits in motor coordination, cognitive impairments, and alterations in sociability and sleep patterns, as well as increased seizure susceptibility. Furthermore, immunohistochemistry revealed reduced synaptophysin immunoreactivity in Pigv341E mice, and electrophysiology recordings showed decreased hippocampal synaptic transmission that could underlie impaired memory formation. In single-cell RNA sequencing, Pigv341E-hippocampal cells exhibited changes in gene expression, most prominently in a subtype of microglia and subicular neurons. A significant reduction in Abl1 transcript levels in several cell clusters suggested a link to the signaling pathway of GPI-anchored ephrins. We also observed elevated levels of Hdc transcripts, which might affect histamine metabolism with consequences for circadian rhythm. This mouse model will not only open the doors to further investigation into the pathophysiology of GPIBD, but will also deepen our understanding of the role of GPI-anchor-related pathways in brain development.


Asunto(s)
Glicosilfosfatidilinositoles/genética , Glicosilfosfatidilinositoles/metabolismo , Manosiltransferasas/metabolismo , Anomalías Múltiples/genética , Secuencia de Aminoácidos , Aminoácidos/genética , Animales , Sistemas CRISPR-Cas , Modelos Animales de Enfermedad , Epilepsia/genética , Glicosilfosfatidilinositoles/deficiencia , Hipocampo/metabolismo , Discapacidad Intelectual/genética , Manosiltransferasas/fisiología , Ratones , Ratones Endogámicos C57BL , Mutación , Mutación Missense , Fenotipo , Ingeniería de Proteínas/métodos , Convulsiones/genética , Convulsiones/fisiopatología
4.
Parasitology ; 146(14): 1767-1772, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31559936

RESUMEN

C-mannosylation was recently identified in the thrombospondin-related anonymous protein (TRAP) from Plasmodium falciparum salivary gland sporozoites. A candidate P. falciparum C-mannosyltransferase (PfDPY-19) was demonstrated to modify thrombospondin type 1 repeat (TSR) domains in vitro, exhibiting a different acceptor specificity than their mammalian counterparts. According to the described minimal acceptor of PfDPY19, several TSR domain-containing proteins of P. falciparum could be C-mannosylated in vivo. However, the relevance of this protein modification for the parasite viability remains unknown. In the present study, we used CRISPR/Cas9 technology to generate a PfDPY19 null mutant, demonstrating that this glycosyltransferase is not essential for the asexual blood development of the parasite. PfDPY19 gene disruption was not associated with a growth phenotype, not even under endoplasmic reticulum-stressing conditions that could impair protein folding. The data presented in this work strongly suggest that PfDPY19 is unlikely to play a critical role in the asexual blood stages of the parasite, at least under in vitro conditions.


Asunto(s)
Estadios del Ciclo de Vida , Manosiltransferasas/fisiología , Plasmodium falciparum/enzimología , Plasmodium falciparum/fisiología , Proteínas Protozoarias/fisiología , Sangre/parasitología , Sistemas CRISPR-Cas , Glicosilación , Mutación con Pérdida de Función , Manosiltransferasas/genética , Plasmodium falciparum/genética , Proteínas Protozoarias/genética , Reproducción Asexuada , Glándulas Salivales/parasitología , Trombospondinas/genética , Trombospondinas/fisiología
5.
Mol Biol Rep ; 46(3): 2693-2701, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30805892

RESUMEN

PPFIA family members and ALG3 play important roles in tumorigenesis and tumor progression. However, the exact roles of distinct PPFIA family members and ALG3 in head and neck squamous cell carcinoma (HNSCC) remain unclear. We studied the mRNA expressions of PPFIA family members and ALG3 in a variety of tumor types compared with the normal controls using the Oncomine database along with meta-analyses of their expressions in HNSCC cancer cell line. The mRNA expressions of PPFIA family members and ALG3 in laryngeal squamous cell carcinoma cell line and normal laryngeal cell line were detected by quantitative real-time polymerase chain reaction. Based on the cBioportal database, we further studied mRNA expression alterations and co-occurrence relationships of the PPFIA family members and ALG3 in HNSCC. The relationship between PPFIA1 and ALG3 mRNA expression alterations and prognoses in patients with HNSCC was explored. We found that PPFIA1 and ALG3 were distinctively overexpressed at the mRNA level in HNSCC tissues compared with normal tissues, they had a significant co-occurrence relationship, their mRNA expressions were significantly higher than other PPFIA family members in laryngeal squamous cell carcinoma cell line, and their mRNA expressions were also significantly higher in laryngeal carcinoma cell line than in normal laryngeal cell line. Patients without both PPFIA1 and ALG3 mRNA expression alterations had better overall survival and disease/progression-free survival compared with patients with both PPFIA1 and ALG3 alterations. Based on these findings, PPFIA1 and ALG3 may play roles in oncogene expression in HNSCC. Their combined overexpression is significantly associated with poor survival outcomes. The relationship between them and the mechanism of action in head and neck cancers deserve further investigation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Manosiltransferasas/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/fisiología , Biomarcadores de Tumor/genética , Carcinoma de Células Escamosas/genética , Línea Celular Tumoral , Proliferación Celular/genética , Transformación Celular Neoplásica/genética , Bases de Datos Genéticas , Regulación Neoplásica de la Expresión Génica/genética , Neoplasias de Cabeza y Cuello/genética , Humanos , Neoplasias Laríngeas/genética , Manosiltransferasas/metabolismo , Manosiltransferasas/fisiología , Pronóstico , Reacción en Cadena en Tiempo Real de la Polimerasa , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Transcriptoma/genética
6.
J Neurosci ; 38(7): 1850-1865, 2018 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-29167399

RESUMEN

Genetic defects in protein O-mannosyltransferase 1 (POMT1) and POMT2 underlie severe muscular dystrophies. POMT genes are evolutionarily conserved in metazoan organisms. In Drosophila, both male and female POMT mutants show a clockwise rotation of adult abdominal segments, suggesting a chirality of underlying pathogenic mechanisms. Here we described and analyzed a similar phenotype in POMT mutant embryos that shows left-handed body torsion. Our experiments demonstrated that coordinated muscle contraction waves are associated with asymmetric embryo rolling, unveiling a new chirality marker in Drosophila development. Using genetic and live-imaging approaches, we revealed that the torsion phenotype results from differential rolling and aberrant patterning of peristaltic waves of muscle contractions. Our results demonstrated that peripheral sensory neurons are required for normal contractions that prevent the accumulation of torsion. We found that POMT mutants show abnormal axonal connections of sensory neurons. POMT transgenic expression limited to sensory neurons significantly rescued the torsion phenotype, axonal connectivity defects, and abnormal contractions in POMT mutant embryos. Together, our data suggested that protein O-mannosylation is required for normal sensory feedback to control coordinated muscle contractions and body posture. This mechanism may shed light on analogous functions of POMT genes in mammals and help to elucidate the etiology of neurological defects in muscular dystrophies.SIGNIFICANCE STATEMENT Protein O-mannosyltransferases (POMTs) are evolutionarily conserved in metazoans. Mutations in POMTs cause severe muscular dystrophies associated with pronounced neurological defects. However, neurological functions of POMTs remain poorly understood. We demonstrated that POMT mutations in Drosophila result in abnormal muscle contractions and cause embryo torsion. Our experiments uncovered a chirality of embryo movements and a unique POMT-dependent mechanism that maintains symmetry of a developing system affected by chiral forces. Furthermore, POMTs were found to be required for proper axon connectivity of sensory neurons, suggesting that O-mannosylation regulates the sensory feedback controlling muscle contractions. This novel POMT function in the peripheral nervous system may shed light on analogous functions in mammals and help to elucidate pathomechanisms of neurological abnormalities in muscular dystrophies.


Asunto(s)
Axones/fisiología , Manosiltransferasas/fisiología , Postura/fisiología , Células Receptoras Sensoriales/fisiología , Animales , Animales Modificados Genéticamente , Proteínas de Drosophila/genética , Distroglicanos/genética , Embrión no Mamífero , Retroalimentación Fisiológica , Manosiltransferasas/genética , Contracción Muscular/fisiología , Mutación , Sistema Nervioso Periférico/fisiología , Fenotipo
7.
J Exp Bot ; 67(18): 5473-5484, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27566817

RESUMEN

In Arabidopsis thaliana, the process of abscission, or the shedding of unwanted organs, is mediated by two genes, HAESA (HAE) and HAESA-LIKE 2 (HSL2), encoding receptor-like protein kinases (RLKs). The double loss-of-function mutant hae-3 hsl2-3 is completely deficient in floral abscission, but, interestingly, the hae-3 hsl2-9 mutant displays a less severe defect. This mutant was chosen for an ethyl methanesulfonate (EMS) screen to isolate enhancer and suppressor mutants, and two such suppressors are the focus of this study. Pooled DNA from the F2 generation of a parental backcross was analyzed by genome sequencing to reveal candidate genes, two of which complement the suppressor phenotype. These genes, EMS-MUTAGENIZED BRI1 SUPPRESSOR 3 (EBS3) and EBS4, both encode mannosyltransferases involved in endoplasmic reticulum (ER)-associated degradation (ERAD) of proteins. Further analysis of these suppressor lines revealed that suppressor mutations are acting solely on the partially functional hsl2-9 mutant receptor to modify the abscission phenotype. Expressing a hsl2-9-yellow fluorescent protein (YFP) transgene in ebs3 mutants yields a higher fluorescent signal than in EBS3/ebs3, suggesting that these mutants restore abscission by disrupting ERAD to allow accumulation of the hsl2-9 receptor, which probably escapes degradation to be trafficked to the plasma membrane to regain signaling.


Asunto(s)
Proteínas de Arabidopsis/fisiología , Arabidopsis/genética , Retículo Endoplásmico/metabolismo , Proteínas de Plantas/metabolismo , Proteínas Serina-Treonina Quinasas/fisiología , Arabidopsis/metabolismo , Arabidopsis/fisiología , Proteínas de Arabidopsis/genética , Flores/genética , Flores/fisiología , Manosiltransferasas/genética , Manosiltransferasas/fisiología , Mutación , Proteínas Serina-Treonina Quinasas/genética , Análisis de Secuencia de ADN
8.
Plant J ; 73(1): 105-17, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22966747

RESUMEN

Galactomannans comprise a ß-1,4-mannan backbone substituted with α-1,6-galactosyl residues. Genes encoding the enzymes that are primarily responsible for backbone synthesis and side-chain addition of galactomannans were previously identified and characterized. To identify additional genes involved in galactomannan biosynthesis, we previously performed deep EST profiling of fenugreek (Trigonella foenum-graecum L.) seed endosperm, which accumulates large quantities of galactomannans as a reserve carbohydrate during seed development. One of the candidate genes encodes a protein that is likely to be a glycosyltransferase. Because this protein is involved in mannan biosynthesis, we named it 'mannan synthesis-related' (MSR). Here, we report the characterization of a fenugreek MSR gene (TfMSR) and its two Arabidopsis homologs, AtMSR1 and AtMSR2. TfMSR was highly and specifically expressed in the endosperm. TfMSR, AtMSR1 and AtMSR2 proteins were all determined to be localized to the Golgi by fluorescence confocal microscopy. The level of mannosyl residues in stem glucomannans decreased by approximately 40% for Arabidopsis msr1 single T-DNA insertion mutants and by more than 50% for msr1 msr2 double mutants, but remained unchanged for msr2 single mutants. In addition, in vitro mannan synthase activity from the stems of msr1 single and msr1 msr2 double mutants also decreased. Expression of AtMSR1 or AtMSR2 in the msr1 msr2 double mutant completely or partially restored mannosyl levels. From these results, we conclude that the MSR protein is important for mannan biosynthesis, and offer some ideas about its role.


Asunto(s)
Mananos/biosíntesis , Trigonella/metabolismo , Endospermo/metabolismo , Genes de Plantas/fisiología , Aparato de Golgi/metabolismo , Manosiltransferasas/metabolismo , Manosiltransferasas/fisiología , Microsomas/metabolismo , Proteínas de Plantas/fisiología
9.
PLoS One ; 7(10): e48211, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23118955

RESUMEN

The biosynthesis of mycobacterial mannose-containing lipoglycans, such as lipomannan (LM) and the immunomodulator lipoarabinomanan (LAM), is carried out by the GT-C superfamily of glycosyltransferases that require polyprenylphosphate-based mannose (PPM) as a sugar donor. The essentiality of lipoglycan synthesis for growth makes the glycosyltransferase that synthesizes PPM, a potential drug target in Mycobacterium tuberculosis, the causative agent of tuberculosis. In M. tuberculosis, PPM has been shown to be synthesized by Ppm1 in enzymatic assays. However, genetic evidence for its essentiality and in vivo role in LM/LAM and PPM biosynthesis is lacking. In this study, we demonstrate that MSMEG3859, a Mycobacterium smegmatis gene encoding the homologue of the catalytic domain of M. tuberculosis Ppm1, is essential for survival. Depletion of MSMEG3859 in a conditional mutant of M. smegmatis resulted in the loss of higher order phosphatidyl-myo-inositol mannosides (PIMs) and lipomannan. We were also able to demonstrate that two other M. tuberculosis genes encoding glycosyltransferases that either had been shown to possess PPM synthase activity (Rv3779), or were involved in synthesizing similar polyprenol-linked donors (ppgS), were unable to compensate for the loss of MSMEG3859 in the conditional mutant.


Asunto(s)
Proteínas Bacterianas/genética , Lipopolisacáridos/biosíntesis , Manosiltransferasas/genética , Mycobacterium smegmatis/genética , Proteínas Bacterianas/fisiología , Genes Esenciales , Prueba de Complementación Genética , Manosiltransferasas/fisiología , Viabilidad Microbiana , Mycobacterium smegmatis/enzimología , Mycobacterium smegmatis/crecimiento & desarrollo , Fenotipo , Eliminación de Secuencia
10.
OMICS ; 14(2): 201-10, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20210661

RESUMEN

Chemogenomics, the study of genomic responses to chemical compounds, has the potential to elucidate the basis of cellular resistance to those chemicals. This knowledge can be applied to improve the performance of strains of industrial interest. In this study, a collection of approximately 5,000 haploid single deletion mutants of Saccharomyces cerevisiae in which each nonessential yeast gene was individually deleted, was screened for strains with increased susceptibility toward stress induced by high-glucose concentration (30% w/v), one of the main stresses occurring during industrial alcoholic fermentation processes aiming the production of alcoholic beverages or bio-ethanol. Forty-four determinants of resistance to high-glucose stress were identified. The most significant Gene Ontology (GO) terms enriched in this dataset are vacuolar organization, late endosome to vacuole transport, and regulation of transcription. Clustering the identified resistance determinants by their known physical and genetic interactions further highlighted the importance of nutrient metabolism control in this context. A concentration of 30% (w/v) of glucose was found to perturb vacuolar function, by reducing cell ability to maintain the physiological acidification of the vacuolar lumen. This stress also affects the active rate of proton efflux through the plasma membrane. Based on results of published studies, the present work revealed shared determinants of yeast resistance to high-glucose and ethanol stresses, including genes involved in vacuolar function, cell wall biogenesis (ANP1), and in the transcriptional control of nutrient metabolism (GCN4 and GCR1), with possible impact on the design of more robust strains to be used in industrial alcoholic fermentation processes.


Asunto(s)
Fermentación/fisiología , Glucosa/farmacología , Proteínas de Saccharomyces cerevisiae/fisiología , Saccharomyces cerevisiae/efectos de los fármacos , Saccharomyces cerevisiae/metabolismo , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/fisiología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Etanol/farmacología , Regulación Fúngica de la Expresión Génica/efectos de los fármacos , Regulación Fúngica de la Expresión Génica/genética , Genoma Fúngico/genética , Genómica , Manosiltransferasas/genética , Manosiltransferasas/fisiología , Proteínas de la Membrana/genética , Proteínas de la Membrana/fisiología , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Factores de Transcripción/genética , Factores de Transcripción/fisiología
11.
Plant Cell ; 21(10): 3397-412, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19880800

RESUMEN

In Saccharomyces cerevisiae, the PMT, KRE2/MNT1, and MNN1 mannosyltransferase protein families catalyze the steps of the O-mannosylation pathway, sequentially adding mannoses to target proteins. We have identified members of all three families and analyzed their roles in pathogenesis of the maize smut fungus Ustilago maydis. Furthermore, we have shown that PMT4, one of the three PMT family members in U. maydis, is essential for tumor formation in Zea mays. Significantly, PMT4 seems to be required only for pathogenesis and is dispensable for other aspects of the U. maydis life cycle. We subsequently show that the deletion of pmt4 results in a strong reduction in the frequency of appressorium formation, with the few appressoria that do form lacking the capacity to penetrate the plant cuticle. Our findings suggest that the O-mannosylation pathway plays a key role in the posttranslational modification of proteins involved in the pathogenic development of U. maydis. The fact that PMT homologs are not found in plants may open new avenues for the development of fungal control strategies. Moreover, the discovery of a highly specific requirement for a single O-mannosyltransferase should aid in the identification of the proteins directly involved in fungal plant penetration, thus leading to a better understanding of plant-fungi interactions.


Asunto(s)
Proteínas Fúngicas/fisiología , Manosiltransferasas/fisiología , Ustilago/enzimología , Ustilago/crecimiento & desarrollo , Proteínas Fúngicas/clasificación , Proteínas Fúngicas/genética , Prueba de Complementación Genética , Manosiltransferasas/clasificación , Manosiltransferasas/genética , Microscopía Fluorescente , Datos de Secuencia Molecular , Mutación , Filogenia , Ustilago/genética
12.
J Bacteriol ; 190(10): 3690-9, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18344361

RESUMEN

Lipoarabinomannans (LAMs) and phosphatidylinositol mannosides (PIMs) are abundant glycolipids in the cell walls of all corynebacteria and mycobacteria, including the devastating human pathogen Mycobacterium tuberculosis. We have recently shown that M. smegmatis mutants of the lipoprotein-encoding lpqW gene have a profound defect in LAM biosynthesis. When these mutants are cultured in complex medium, spontaneous bypass mutants consistently evolve in which LAM biosynthesis is restored at the expense of polar PIM synthesis. Here we show that restoration of LAM biosynthesis in the lpqW mutant results from secondary mutations in the pimE gene. PimE is a mannosyltransferase involved in converting AcPIM4, a proposed branch point intermediate in the PIM and LAM biosynthetic pathways, to more polar PIMs. Mutations in pimE arose due to insertion of the mobile genetic element ISMsm1 and independent point mutations that were clustered in predicted extracytoplasmic loops of this polytopic membrane protein. Our findings provide the first strong evidence that LpqW is required to channel intermediates such as AcPIM4 into LAM synthesis and that loss of PimE function results in the accumulation of AcPIM4, bypassing the need for LpqW. These data highlight new mechanisms regulating the biosynthetic pathways of these essential cell wall components.


Asunto(s)
Lipopolisacáridos/biosíntesis , Lipoproteínas/genética , Manosiltransferasas/metabolismo , Manosiltransferasas/fisiología , Mycobacterium smegmatis/metabolismo , Pared Celular/metabolismo , Lipoproteínas/metabolismo , Manosiltransferasas/química , Manosiltransferasas/genética , Mutación , Mycobacterium smegmatis/genética , Mycobacterium smegmatis/crecimiento & desarrollo , Fosfatidilinositoles/biosíntesis , Fosfatidilinositoles/metabolismo
13.
FEMS Yeast Res ; 8(3): 374-85, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18312373

RESUMEN

PAU genes comprise the largest multiple gene family in Saccharomyces cerevisiae with 24 members whose sequence homology ranges from 82% to 100%. Although transcriptional regulation for some of the PAU genes has been reported, none of the Pau proteins has been characterized. We constructed yeast strains encoding a C-terminal tandem affinity purification-tagged Pau5 in the PAU5 locus to study Pau5 production and properties in vivo. Pau5 is highly induced by low temperature, low oxygen and wine fermentation conditions. It is unstable in cells grown under laboratory conditions and is temporarily stabilized by low oxygen, osmotic and ethanol stresses. Pau5 degradation is accompanied by an unknown modification with a gradual increase in molecular mass by 3 kDa. Furthermore, Pau5 is O-mannosylated mainly by Pmt1; mannosylation enhances stability of the protein. The mannosylated Pau5 is soluble whereas the nonmannosylated proform Pau5 is an integral membrane protein. Our findings suggest that the intracellular concentration of Pau5 is regulated by wine making stress both at transcriptional and posttranslational levels; Pau5 might play a role in adaptation of yeast cells during alcoholic fermentations.


Asunto(s)
Proteínas de Saccharomyces cerevisiae/biosíntesis , Saccharomyces cerevisiae/metabolismo , Anaerobiosis , Secuencia de Bases , Frío , Fermentación , Manosa/metabolismo , Manosiltransferasas/fisiología , Proteínas de la Membrana/biosíntesis , Datos de Secuencia Molecular , Saccharomyces cerevisiae/crecimiento & desarrollo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Vino
15.
Eukaryot Cell ; 6(12): 2260-8, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17905922

RESUMEN

Protein O-mannosyltransferases initiate O mannosylation of secretory proteins, which are of fundamental importance in eukaryotes. In this study, the PMT gene family of the human fungal pathogen Aspergillus fumigatus was identified and characterized. Unlike the case in Saccharomyces cerevisiae, where the PMT family is highly redundant, only one member of each PMT subfamily, namely, Afpmt1, Afpmt2, and Afpmt4, is present in A. fumigatus. Mutants with a deletion of Afpmt1 are viable. In vitro and in vivo activity assays confirmed that the protein encoded by Afpmt1 acts as an O-mannosyltransferase (AfPmt1p). Characterization of the DeltaAfpmt1 mutant showed that a lack of AfPmt1p results in sensitivity to elevated temperature and defects in growth and cell wall integrity, thereby affecting cell morphology, conidium formation, and germination. In a mouse model, Afpmt1 was not required for the virulence of A. fumigatus under the experimental conditions used.


Asunto(s)
Aspergillus fumigatus/enzimología , Pared Celular/enzimología , Regulación Fúngica de la Expresión Génica , Manosiltransferasas/metabolismo , Esporas Fúngicas/metabolismo , Animales , Aspergillus fumigatus/genética , Cartilla de ADN/química , Genes Fúngicos , Genoma Fúngico , Masculino , Manosiltransferasas/fisiología , Ratones , Ratones Endogámicos BALB C , Modelos Biológicos , Fenotipo , Temperatura
16.
Eukaryot Cell ; 6(2): 222-34, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17142566

RESUMEN

Protein O mannosylation is initiated in the endoplasmic reticulum by protein O-mannosyltransferases (Pmt proteins) and plays an important role in the secretion, localization, and function of many proteins, as well as in cell wall integrity and morphogenesis in fungi. Three Pmt proteins, each belonging to one of the three respective Pmt subfamilies, are encoded in the genome of the human fungal pathogen Cryptococcus neoformans. Disruption of the C. neoformans PMT4 gene resulted in abnormal growth morphology and defective cell separation. Transmission electron microscopy revealed defective cell wall septum degradation during mother-daughter cell separation in the pmt4 mutant compared to wild-type cells. The pmt4 mutant also demonstrated sensitivity to elevated temperature, sodium dodecyl sulfate, and amphotericin B, suggesting cell wall defects. Further analysis of cell wall protein composition revealed a cell wall proteome defect in the pmt4 mutant, as well as a global decrease in protein mannosylation. Heterologous expression of C. neoformans PMT4 in a Saccharomyces cerevisiae pmt1pmt4 mutant strain functionally complemented the deficient Pmt activity. Furthermore, Pmt4 activity in C. neoformans was required for full virulence in two murine models of disseminated cryptococcal infection. Taken together, these results indicate a central role for Pmt4-mediated protein O mannosylation in growth, cell wall integrity, and virulence of C. neoformans.


Asunto(s)
Cryptococcus neoformans/crecimiento & desarrollo , Cryptococcus neoformans/patogenicidad , Proteínas Fúngicas/fisiología , Manosiltransferasas/fisiología , Morfogénesis , Virulencia , Secuencia de Aminoácidos , Animales , Northern Blotting , Western Blotting , Pared Celular/metabolismo , Cryptococcus neoformans/enzimología , Femenino , Regulación Fúngica de la Expresión Génica , Prueba de Complementación Genética , Ratones , Ratones Endogámicos CBA , Datos de Secuencia Molecular , Mutación , Homología de Secuencia de Aminoácido
17.
Proc Natl Acad Sci U S A ; 103(37): 13664-9, 2006 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-16945913

RESUMEN

Lipoarabinomannan (LAM), one of the few known bacterial glycosylphosphoinositides (GPIs), occurs in various structural forms in Mycobacterium species. It has been implicated in key aspects of the physiology of Mycobacterium tuberculosis and the immunology and pathogenesis of tuberculosis. Yet, little is known of the biosynthesis of LAM. A bioinformatics approach identified putative integral membrane proteins, MSMEG4250 in Mycobacterium smegmatis and Rv2181 in M. tuberculosis, with 10 predicted transmembrane domains and a glycosyltransferase (GT) motif (DID), features that are common to eukaryotic mannosyltransferases (ManTs) of the GT-C superfamily that rely on polyprenyl-linked rather than nucleotide-linked sugar donors. Inactivation of M. smegmatis MSMEG4250 by allelic exchange resulted in altered growth and inability to synthesize lipomannan (LM) but accumulation of a previously uncharacterized, truncated LAM. MALDI-TOF/MS and NMR indicated a structure lower in molecular weight than the native molecule, a preponderance of 6-linked Manp residues, and the absence of 2,6-linked and terminal Manp. Complementation of the mutant with the corresponding ortholog of M. tuberculosis H37Rv restored normal LM/LAM synthesis. The data suggest that MSMEG4250 and Rv2181 are ManTs that are responsible for the addition of alpha(1-->2) branches to the mannan core of LM/LAM and that arrest of this branching in the mutant deters formation of native LAM. The results allow for the presentation of a unique model of LM and LAM biosynthesis. The generation of mutants defective in the synthesis of LM/LAM will help define the role of these GPIs in the immunology and pathogenesis of mycobacterial infections and physiology of the organism.


Asunto(s)
Proteínas Bacterianas/fisiología , Lipopolisacáridos/biosíntesis , Manosiltransferasas/fisiología , Mycobacterium smegmatis/enzimología , Mycobacterium smegmatis/crecimiento & desarrollo , Alelos , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/genética , Biología Computacional , Prueba de Complementación Genética , Manosiltransferasas/antagonistas & inhibidores , Manosiltransferasas/genética , Mycobacterium smegmatis/genética , Mycobacterium tuberculosis/enzimología , Mycobacterium tuberculosis/genética , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
18.
J Biol Chem ; 281(35): 25143-55, 2006 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16803893

RESUMEN

Phosphatidylinositol mannosides (PIMs) are a major class of glycolipids in all mycobacteria. AcPIM2, a dimannosyl PIM, is both an end product and a precursor for polar PIMs, such as hexamannosyl PIM (AcPIM6) and the major cell wall lipoglycan, lipoarabinomannan (LAM). The mannosyltransferases that convert AcPIM2 to AcPIM6 or LAM are dependent on polyprenol-phosphate-mannose (PPM), but have not yet been characterized. Here, we identified a gene, termed pimE that is present in all mycobacteria, and is required for AcPIM6 biosynthesis. PimE was initially identified based on homology with eukaryotic PIG-M mannosyltransferases. PimE-deleted Mycobacterium smegmatis was defective in AcPIM6 synthesis, and accumulated the tetramannosyl PIM, AcPIM4. Loss of PimE had no affect on cell growth or viability, or the biosynthesis of other intracellular and cell wall glycans. However, changes in cell wall hydrophobicity and plasma membrane organization were detected, suggesting a role for AcPIM6 in the structural integrity of the cell wall and plasma membrane. These defects were corrected by ectopic expression of the pimE gene. Metabolic pulse-chase radiolabeling and cell-free PIM biosynthesis assays indicated that PimE catalyzes the alpha1,2-mannosyl transfer for the AcPIM5 synthesis. Mutation of an Asp residue in PimE that is conserved in and required for the activity of human PIG-M resulted in loss of PIM-biosynthetic activity, indicating that PimE is the catalytic component. Finally, PimE was localized to a distinct membrane fraction enriched in AcPIM4-6 biosynthesis. Taken together, PimE represents the first PPM-dependent mannosyl-transferase shown to be involved in PIM biosynthesis, where it mediates the fifth mannose transfer.


Asunto(s)
Manósidos/química , Manosiltransferasas/química , Manosiltransferasas/fisiología , Mycobacterium/metabolismo , Fosfatidilinositoles/química , Secuencia de Aminoácidos , Proliferación Celular , Pared Celular/metabolismo , Sistema Libre de Células , Genoma Bacteriano , Humanos , Manosa/química , Datos de Secuencia Molecular , Mycobacterium smegmatis/metabolismo , Fosfatos , Homología de Secuencia de Aminoácido
19.
J Biol Chem ; 281(28): 19339-45, 2006 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-16698797

RESUMEN

A defect of protein O-mannosylation causes congenital muscular dystrophy with brain malformation and structural eye abnormalities, so-called Walker-Warburg syndrome. Protein O-mannosylation is catalyzed by protein O-mannosyltransferase 1 (POMT1) and its homologue, POMT2. Coexpression of POMT1 and POMT2 is required to show O-mannosylation activity. Here we have shown that POMT1 forms a complex with POMT2 and the complex possesses protein O-mannosyltransferase activity. Results indicate that POMT1 and POMT2 associate physically and functionally in vivo. Recently, three mutations were reported in the POMT1 gene of patients who showed milder phenotypes than typical Walker-Warburg syndrome. We coexpressed these mutant POMT1s with POMT2 and found that none of them had any activity. However, all POMT1 mutants, including previously identified POMT1 mutants, coprecipitated with POMT2. These results indicate that the mutant POMT1s could form heterocomplexes with POMT2 but that such complexes are insufficient for enzymatic activity.


Asunto(s)
Anomalías Múltiples/enzimología , Anomalías del Ojo/patología , Manosiltransferasas/química , Encéfalo/metabolismo , Catálisis , Línea Celular , Detergentes/farmacología , Anomalías del Ojo/metabolismo , Vectores Genéticos , Humanos , Manosiltransferasas/fisiología , Microsomas/metabolismo , Mutación , Unión Proteica , Síndrome
20.
Neurology ; 66(10): 1564-7; discussion 1461, 2006 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-16717220

RESUMEN

Mutations in POMT1 have been identified in Walker-Warburg syndrome and in patients with limb-girdle muscular dystrophy and mental retardation (LGMD2K). The authors report new POMT1 mutations in three unrelated children with severe motor impairment, leg hypertrophy, and mental retardation but without brain and ocular malformations. These patients are similar to LGMD2K but have earlier onset and more severe motor disability. The current findings expand the spectrum of POMT1-associated phenotypes.


Asunto(s)
Discapacidad Intelectual/genética , Manosiltransferasas/deficiencia , Microcefalia/genética , Distrofias Musculares/genética , Adolescente , Edad de Inicio , Preescolar , Codón sin Sentido , Contractura/genética , Progresión de la Enfermedad , Femenino , Glicosilación , Humanos , Hipertrofia , Lactante , Discapacidad Intelectual/patología , Pierna/patología , Imagen por Resonancia Magnética , Masculino , Manosiltransferasas/genética , Manosiltransferasas/fisiología , Microcefalia/patología , Distrofias Musculares/patología , Mutación Missense , Fenotipo , Mutación Puntual , Procesamiento Proteico-Postraduccional , Síndrome
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA