Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 332
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cell Mol Life Sci ; 81(1): 246, 2024 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-38819479

RESUMO

The glycosylphosphatidylinositol (GPI) biosynthetic pathway in the endoplasmic reticulum (ER) is crucial for generating GPI-anchored proteins (GPI-APs), which are translocated to the cell surface and play a vital role in cell signaling and adhesion. This study focuses on two integral components of the GPI pathway, the PIGL and PIGF proteins, and their significance in trophoblast biology. We show that GPI pathway mutations impact on placental development impairing the differentiation of the syncytiotrophoblast (SynT), and especially the SynT-II layer, which is essential for the establishment of the definitive nutrient exchange area within the placental labyrinth. CRISPR/Cas9 knockout of Pigl and Pigf in mouse trophoblast stem cells (mTSCs) confirms the role of these GPI enzymes in syncytiotrophoblast differentiation. Mechanistically, impaired GPI-AP generation induces an excessive unfolded protein response (UPR) in the ER in mTSCs growing in stem cell conditions, akin to what is observed in human preeclampsia. Upon differentiation, the impairment of the GPI pathway hinders the induction of WNT signaling for early SynT-II development. Remarkably, the transcriptomic profile of Pigl- and Pigf-deficient cells separates human patient placental samples into preeclampsia and control groups, suggesting an involvement of Pigl and Pigf in establishing a preeclamptic gene signature. Our study unveils the pivotal role of GPI biosynthesis in early placentation and uncovers a new preeclampsia gene expression profile associated with mutations in the GPI biosynthesis pathway, providing novel molecular insights into placental development with implications for enhanced patient stratification and timely interventions.


Assuntos
Diferenciação Celular , Glicosilfosfatidilinositóis , Placentação , Trofoblastos , Trofoblastos/metabolismo , Trofoblastos/citologia , Feminino , Gravidez , Animais , Humanos , Camundongos , Placentação/genética , Glicosilfosfatidilinositóis/metabolismo , Glicosilfosfatidilinositóis/biossíntese , Placenta/metabolismo , Placenta/citologia , Via de Sinalização Wnt , Pré-Eclâmpsia/metabolismo , Pré-Eclâmpsia/genética , Pré-Eclâmpsia/patologia , Retículo Endoplasmático/metabolismo , Vias Biossintéticas/genética , Resposta a Proteínas não Dobradas , Sistemas CRISPR-Cas
2.
Am J Hum Genet ; 105(2): 395-402, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31353022

RESUMO

The glycosylphosphatidylinositol (GPI) anchor links over 150 proteins to the cell surface and is present on every cell type. Many of these proteins play crucial roles in neuronal development and function. Mutations in 18 of the 29 genes implicated in the biosynthesis of the GPI anchor have been identified as the cause of GPI biosynthesis deficiencies (GPIBDs) in humans. GPIBDs are associated with intellectual disability and seizures as their cardinal features. An essential component of the GPI transamidase complex is PIGU, along with PIGK, PIGS, PIGT, and GPAA1, all of which link GPI-anchored proteins (GPI-APs) onto the GPI anchor in the endoplasmic reticulum (ER). Here, we report two homozygous missense mutations (c.209T>A [p.Ile70Lys] and c.1149C>A [p.Asn383Lys]) in five individuals from three unrelated families. All individuals presented with global developmental delay, severe-to-profound intellectual disability, muscular hypotonia, seizures, brain anomalies, scoliosis, and mild facial dysmorphism. Using multicolor flow cytometry, we determined a characteristic profile for GPI transamidase deficiency. On granulocytes this profile consisted of reduced cell-surface expression of fluorescein-labeled proaerolysin (FLAER), CD16, and CD24, but not of CD55 and CD59; additionally, B cells showed an increased expression of free GPI anchors determined by T5 antibody. Moreover, computer-assisted facial analysis of different GPIBDs revealed a characteristic facial gestalt shared among individuals with mutations in PIGU and GPAA1. Our findings improve our understanding of the role of the GPI transamidase complex in the development of nervous and skeletal systems and expand the clinical spectrum of disorders belonging to the group of inherited GPI-anchor deficiencies.


Assuntos
Aciltransferases/genética , Encefalopatias/etiologia , Epilepsia/etiologia , Glicosilfosfatidilinositóis/biossíntese , Glicosilfosfatidilinositóis/deficiência , Deficiência Intelectual/etiologia , Mutação , Convulsões/patologia , Adolescente , Adulto , Sequência de Aminoácidos , Encefalopatias/patologia , Criança , Pré-Escolar , Epilepsia/patologia , Feminino , Glicosilfosfatidilinositóis/genética , Humanos , Lactente , Recém-Nascido , Deficiência Intelectual/patologia , Masculino , Linhagem , Convulsões/genética , Homologia de Sequência , Adulto Jovem
3.
Am J Hum Genet ; 105(2): 384-394, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31256876

RESUMO

Proteins anchored to the cell surface via glycosylphosphatidylinositol (GPI) play various key roles in the human body, particularly in development and neurogenesis. As such, many developmental disorders are caused by mutations in genes involved in the GPI biosynthesis and remodeling pathway. We describe ten unrelated families with bi-allelic mutations in PIGB, a gene that encodes phosphatidylinositol glycan class B, which transfers the third mannose to the GPI. Ten different PIGB variants were found in these individuals. Flow cytometric analysis of blood cells and fibroblasts from the affected individuals showed decreased cell surface presence of GPI-anchored proteins. Most of the affected individuals have global developmental and/or intellectual delay, all had seizures, two had polymicrogyria, and four had a peripheral neuropathy. Eight children passed away before four years old. Two of them had a clinical diagnosis of DOORS syndrome (deafness, onychodystrophy, osteodystrophy, mental retardation, and seizures), a condition that includes sensorineural deafness, shortened terminal phalanges with small finger and toenails, intellectual disability, and seizures; this condition overlaps with the severe phenotypes associated with inherited GPI deficiency. Most individuals tested showed elevated alkaline phosphatase, which is a characteristic of the inherited GPI deficiency but not DOORS syndrome. It is notable that two severely affected individuals showed 2-oxoglutaric aciduria, which can be seen in DOORS syndrome, suggesting that severe cases of inherited GPI deficiency and DOORS syndrome might share some molecular pathway disruptions.


Assuntos
Anormalidades Craniofaciais/etiologia , Glicosilfosfatidilinositóis/biossíntese , Glicosilfosfatidilinositóis/deficiência , Deformidades Congênitas da Mão/etiologia , Perda Auditiva Neurossensorial/etiologia , Deficiência Intelectual/etiologia , Manosiltransferases/genética , Doenças Metabólicas/etiologia , Mutação , Unhas Malformadas/etiologia , Doenças do Sistema Nervoso Periférico/etiologia , Convulsões/patologia , Adulto , Criança , Pré-Escolar , Anormalidades Craniofaciais/patologia , Feminino , Glicosilfosfatidilinositóis/genética , Deformidades Congênitas da Mão/patologia , Perda Auditiva Neurossensorial/patologia , Humanos , Lactente , Recém-Nascido , Deficiência Intelectual/patologia , Masculino , Doenças Metabólicas/patologia , Unhas Malformadas/patologia , Linhagem , Doenças do Sistema Nervoso Periférico/patologia , Convulsões/genética , Índice de Gravidade de Doença , Adulto Jovem
4.
Immunity ; 36(4): 572-85, 2012 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-22483804

RESUMO

Histone methyltransferases catalyze site-specific deposition of methyl groups, enabling recruitment of transcriptional regulators. In mammals, trimethylation of lysine 4 in histone H3, a modification localized at the transcription start sites of active genes, is catalyzed by six enzymes (SET1a and SET1b, MLL1-MLL4) whose specific functions are largely unknown. By using a genomic approach, we found that in macrophages, MLL4 (also known as Wbp7) was required for the expression of Pigp, an essential component of the GPI-GlcNAc transferase, the enzyme catalyzing the first step of glycosylphosphatidylinositol (GPI) anchor synthesis. Impaired Pigp expression in Wbp7(-/-) macrophages abolished GPI anchor-dependent loading of proteins on the cell membrane. Consistently, loss of GPI-anchored CD14, the coreceptor for lipopolysaccharide (LPS) and other bacterial molecules, markedly attenuated LPS-triggered intracellular signals and gene expression changes. These data link a histone-modifying enzyme to a biosynthetic pathway and indicate a specialized biological role for Wbp7 in macrophage function and antimicrobial response.


Assuntos
Glicosilfosfatidilinositóis/metabolismo , Macrófagos/metabolismo , Proteínas de Membrana/metabolismo , Proteína de Leucina Linfoide-Mieloide/metabolismo , Animais , Membrana Celular/metabolismo , Células Cultivadas , Glicosilfosfatidilinositóis/biossíntese , Hexosiltransferases/biossíntese , Histona Metiltransferases , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/metabolismo , Receptores de Lipopolissacarídeos/biossíntese , Lipopolissacarídeos/imunologia , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Proteína de Leucina Linfoide-Mieloide/biossíntese , Proteína de Leucina Linfoide-Mieloide/genética , Transdução de Sinais
5.
Neurogenetics ; 21(4): 259-267, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32462292

RESUMO

Deficiency of the endoplasmic reticulum transmembrane protein ARV1 leads to epileptic encephalopathy in humans and in mice. ARV1 is highly conserved, but its function in human cells is unknown. Studies of yeast arv1 null mutants indicate that it is involved in a number of biochemical processes including the synthesis of sphingolipids and glycosylphosphatidylinositol (GPI), a glycolipid anchor that is attached to the C-termini of many membrane bound proteins. GPI anchors are post-translational modifications, enabling proteins to travel from the endoplasmic reticulum (ER) through the Golgi and to attach to plasma membranes. We identified a homozygous pathogenic mutation in ARV1, p.Gly189Arg, in two brothers with infantile encephalopathy, and characterized the biochemical defect caused by this mutation. In addition to reduced expression of ARV1 transcript and protein in patients' fibroblasts, complementation tests in yeast showed that the ARV1 p.Gly189Arg mutation leads to deficient maturation of Gas1, a GPI-anchored protein, but does not affect sphingolipid synthesis. Our results suggest, that similar to mutations in other proteins in the GPI-anchoring pathway, including PIGM, PIGA, and PIGQ, ARV1 p.Gly189Arg causes a GPI anchoring defect and leads to early onset epileptic encephalopathy.


Assuntos
Encefalopatias/genética , Proteínas de Transporte/genética , Glicosilfosfatidilinositóis/biossíntese , Deficiência Intelectual/genética , Proteínas de Membrana/genética , Convulsões/genética , Adolescente , Criança , Retículo Endoplasmático/metabolismo , Fibroblastos/metabolismo , Teste de Complementação Genética , Complexo de Golgi/metabolismo , Homozigoto , Humanos , Lipídeos/química , Masculino , Manosiltransferases/genética , Mutação , Linhagem , Domínios Proteicos , Temperatura
6.
Yeast ; 37(1): 63-72, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31845370

RESUMO

ScGpi12 is a 304 amino residue long endoplasmic reticulum membrane protein, which participates in the de-N-acetylation of N-acetylglucosaminyl phosphatidylinositol to produce glucosaminyl phosphatidylinositol in the second step of GPI anchor biosynthesis pathway in Saccharomyces cerevisiae. ScGpi12 was cloned in a pMAL-c2x vector and expressed heterologously in Rosetta-gami (DE3) strain of E. coli. Affinity purification of the protein yielded low amounts of the MBP-tagged enzyme, which was active. To the best of our knowledge, this is the first successful purification of full-length Gpi12 enzyme, without the accompanying GroEL that was seen in other studies. The presence of the tag did not greatly alter the activity of the enzyme. ScGpi12 was optimally active in the pH range of 6.5-8.5 and at 30 °C. It was not sensitive to treatment with EDTA but was stimulated by multiple divalent cations. The divalent cation did not alter the pH profile of the enzyme, suggesting no role of the divalent metal in creating a nucleophile for catalysis. Divalent cations did, however, enhance the turnover number of the enzyme for its substrate, suggesting that they are probably required for the production of a catalytically competent active site by bringing the active site residues within optimum distance of the substrate for catalysis.


Assuntos
Acetilesterase/metabolismo , Glicosilfosfatidilinositóis/biossíntese , Saccharomyces cerevisiae/enzimologia , Saccharomyces cerevisiae/genética , Acetilesterase/genética , Acetilglucosamina/análogos & derivados , Acetilglucosamina/metabolismo , Vias Biossintéticas , Catálise , Clonagem Molecular , Retículo Endoplasmático/enzimologia , Escherichia coli/genética , Cinética , Fosfatidilinositóis/metabolismo , Especificidade por Substrato
7.
Biochem Soc Trans ; 48(3): 1129-1138, 2020 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-32573677

RESUMO

Glycosylphosphatidylinositol (GPI) is a glycolipid added to the C-terminus of a large variety of proteins in eukaryotes, thereby anchoring these proteins to the cell surface. More than 150 different human proteins are modified with GPI, and GPI-anchored proteins (GPI-APs) play critical roles in embryogenesis, neurogenesis, immunity, and fertilization. GPI-APs are biosynthesized in the endoplasmic reticulum (ER) and transported to the plasma membrane via the Golgi apparatus. During transport, GPI-APs undergo structural remodeling that is important for the efficient folding and sorting of GPI-APs. Asparagine-linked glycan-dependent folding and deacylation by PGAP1 work together to ensure that correctly folded GPI-APs are transported from the ER to the Golgi. Remodeling of the GPI lipid moiety is critical for the association of GPI-APs with lipid rafts. On the cell surface, certain GPI-APs are cleaved by GPI cleavage enzymes and released from the membrane, a key event in processes such as spermatogenesis and neurogenesis. In this review, we discuss the enzymes involved in GPI-AP biosynthesis and the fate of GPI-APs in mammalian cells, with a focus on the assembly, folding, degradation, and cleavage of GPI-APs.


Assuntos
Membrana Celular/metabolismo , Retículo Endoplasmático/enzimologia , Glicosilfosfatidilinositóis/biossíntese , Complexo de Golgi/enzimologia , Animais , Humanos , Masculino , Microdomínios da Membrana/enzimologia , Proteínas de Membrana/metabolismo , Neurogênese , Domínios Proteicos , Dobramento de Proteína , Transporte Proteico , Espermatogênese
8.
Mol Genet Metab ; 130(1): 49-57, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32165008

RESUMO

BACKGROUND: Mutations in the ARV1 Homolog, Fatty Acid Homeostasis Modulator (ARV1), have recently been described in association with early infantile epileptic encephalopathy 38. Affected individuals presented with epilepsy, ataxia, profound intellectual disability, visual impairment, and central hypotonia. In S. cerevisiae, Arv1 is thought to be involved in sphingolipid metabolism and glycophosphatidylinositol (GPI)-anchor synthesis. The function of ARV1 in human cells, however, has not been elucidated. METHODS: Mutations were discovered through whole exome sequencing and alternate splicing was validated on the cDNA level. Expression of the variants was determined by qPCR and Western blot. Expression of GPI-anchored proteins on neutrophils and fibroblasts was analyzed by FACS and immunofluorescence microscopy, respectively. RESULTS: Here we describe seven patients from two unrelated families with biallelic splice mutations in ARV1. The patients presented with early onset epilepsy, global developmental delays, profound hypotonia, delayed speech development, cortical visual impairment, and severe generalized cerebral and cerebellar atrophy. The splice variants resulted in decreased ARV1 expression and significant decreases in GPI-anchored protein on the membranes of neutrophils and fibroblasts, indicating that the loss of ARV1 results in impaired GPI-anchor synthesis. CONCLUSION: Loss of GPI-anchored proteins on our patients' cells confirms that the yeast Arv1 function of GPI-anchor synthesis is conserved in humans. Overlap between the phenotypes in our patients and those reported for other GPI-anchor disorders suggests that ARV1-deficiency is a GPI-anchor synthesis disorder.


Assuntos
Anormalidades Múltiplas/genética , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Deficiências do Desenvolvimento/genética , Epilepsia/genética , Glicosilfosfatidilinositóis/deficiência , Deficiência Intelectual/genética , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Anormalidades Múltiplas/fisiopatologia , Adolescente , Processamento Alternativo/genética , Pré-Escolar , Deficiências do Desenvolvimento/fisiopatologia , Epilepsia/fisiopatologia , Feminino , Fibroblastos/metabolismo , Proteínas Ligadas por GPI/metabolismo , Glicosilfosfatidilinositóis/biossíntese , Homozigoto , Humanos , Lactente , Recém-Nascido , Masculino , Mutação , Neutrófilos/metabolismo , Linhagem , Sequenciamento do Exoma
9.
Am J Med Genet A ; 182(6): 1477-1482, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32198969

RESUMO

Inherited glycosylphosphatidylinositol (GPI) deficiencies are a group of clinically and genetically heterogeneous conditions belonging to the congenital disorders of glycosylation. PIGW is involved in GPI biosynthesis and modification, and biallelic pathogenic variants in this gene cause autosomal recessive GPI biosynthesis defect 11. Only five patients and two fetuses have been reported in the literature thus far. Here we describe a new patient with a novel homozygous missense variant in PIGW, who presented with hypotonia, severe intellectual disability, early-onset epileptic seizures, brain abnormalities, nystagmus, hand stereotypies, recurrent respiratory infections, distinctive facial features, and hyperphosphatasia. Our report expands the phenotype of GPI biosynthesis defect 11 to include stereotypies and recurrent respiratory infections. A detailed and long-term analysis of the electroclinical characteristics and review of the literature suggest that early-onset epileptic seizures are a key manifestation of GPI biosynthesis defect 11. West syndrome and focal-onset epileptic seizures are the most common seizure types, and the fronto-temporal regions may be the most frequently involved areas in these patients.


Assuntos
Aciltransferases/genética , Glicosilfosfatidilinositóis/deficiência , Glicosilfosfatidilinositóis/genética , Deficiência Intelectual/genética , Proteínas de Membrana/genética , Convulsões/genética , Encéfalo/anormalidades , Encéfalo/patologia , Criança , Pré-Escolar , Feminino , Glicosilfosfatidilinositóis/biossíntese , Humanos , Lactente , Deficiência Intelectual/complicações , Deficiência Intelectual/patologia , Masculino , Hipotonia Muscular/complicações , Hipotonia Muscular/genética , Hipotonia Muscular/patologia , Mutação de Sentido Incorreto/genética , Convulsões/complicações , Convulsões/patologia , Convulsões/fisiopatologia
10.
J Biol Chem ; 293(31): 12222-12238, 2018 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-29907567

RESUMO

The ability of Candida albicans to switch between yeast to hyphal form is a property that is primarily associated with the invasion and virulence of this human pathogenic fungus. Several glycosylphosphatidylinositol (GPI)-anchored proteins are expressed only during hyphal morphogenesis. One of the major pathways that controls hyphal morphogenesis is the Ras-signaling pathway. We examine the cross-talk between GPI anchor biosynthesis and Ras signaling in C. albicans. We show that the first step of GPI biosynthesis is activated by Ras in C. albicans This is diametrically opposite to what is reported in Saccharomyces cerevisiae Of the two C. albicans Ras proteins, CaRas1 alone activates GPI-GnT activity; activity is further stimulated by constitutively activated CaRas1. CaRas1 localized to the cytoplasm or endoplasmic reticulum (ER) is sufficient for GPI-GnT activation. Of the six subunits of the GPI-N-acetylglucosaminyltransferase (GPI-GnT) that catalyze the first step of GPI biosynthesis, CaGpi2 is the key player involved in activating Ras signaling and hyphal morphogenesis. Activation of Ras signaling is independent of the catalytic competence of GPI-GnT. This too is unlike what is observed in S. cerevisiae where multiple subunits were identified as inhibiting Ras2. Fluorescence resonance energy transfer (FRET) studies indicate a specific physical interaction between CaRas1 and CaGpi2 in the ER, which would explain the ability of CaRas1 to activate GPI-GnT. CaGpi2, in turn, promotes activation of the Ras-signaling pathway and hyphal morphogenesis. The Cagpi2 mutant is also more susceptible to macrophage-mediated killing, and macrophage cells show better survival when co-cultured with Cagpi2.


Assuntos
Candida albicans/enzimologia , Proteínas Fúngicas/metabolismo , Glicosilfosfatidilinositóis/biossíntese , N-Acetilglucosaminiltransferases/metabolismo , Proteínas ras/metabolismo , Candida albicans/genética , Candida albicans/metabolismo , Retículo Endoplasmático/enzimologia , Retículo Endoplasmático/genética , Retículo Endoplasmático/metabolismo , Proteínas Fúngicas/genética , Hifas/enzimologia , Hifas/genética , Hifas/metabolismo , N-Acetilglucosaminiltransferases/genética , Transporte Proteico , Transdução de Sinais , Proteínas ras/genética
11.
Artigo em Inglês | MEDLINE | ID: mdl-31611349

RESUMO

Manogepix (MGX) targets the conserved fungal Gwt1 enzyme required for acylation of inositol early in the glycosylphosphatidylinositol biosynthesis pathway. The prodrug fosmanogepix is currently in clinical development for the treatment of invasive fungal infections. We determined that the median frequencies of spontaneous mutations conferring reduced susceptibility to MGX in Candida albicans, C. glabrata, and C. parapsilosis ranged from 3 × 10-8 to <1.85 × 10-8 Serial passage on agar identified mutants of C. albicans and C. parapsilosis with reduced susceptibility to MGX; however, this methodology did not result in C. glabrata mutants with reduced susceptibility. Similarly, serial passage in broth resulted in ≤2-fold changes in population MIC values for C. tropicalis, C. auris, and C. glabrata A spontaneous V163A mutation in the Gwt1 protein of C. glabrata and a corresponding C. albicans heterozygous V162A mutant were obtained. A C. glabrata V163A Gwt1 mutant generated using CRISPR, along with V162A and V168A mutants expressed in C. albicans and Saccharomyces cerevisiae Gwt1, respectively, all demonstrated reduced susceptibility to MGX versus control strains, suggesting the importance of this valine residue to MGX binding across different species. Cross-resistance to the three major classes of antifungals was evaluated, but no changes in susceptibility to amphotericin B or caspofungin were observed in any mutant. No change was observed in fluconazole susceptibility, with the exception of a single non-Gwt1 mutant, where a 4-fold increase in the fluconazole MIC was observed. MGX demonstrated a relatively low potential for resistance development, consistent with other approved antifungal agents and those in clinical development.


Assuntos
Aminopiridinas/farmacologia , Antifúngicos/farmacologia , Candida/efeitos dos fármacos , Isoxazóis/farmacologia , Aciltransferases/química , Aciltransferases/genética , Sequência de Aminoácidos , Vias Biossintéticas/efeitos dos fármacos , Vias Biossintéticas/genética , Candida/genética , Candida/metabolismo , Candidíase/tratamento farmacológico , Candidíase/microbiologia , Farmacorresistência Fúngica/genética , Proteínas Fúngicas/antagonistas & inibidores , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Genes Fúngicos , Glicosilfosfatidilinositóis/biossíntese , Glicosilfosfatidilinositóis/química , Glicosilfosfatidilinositóis/genética , Humanos , Proteínas de Membrana/química , Proteínas de Membrana/genética , Testes de Sensibilidade Microbiana , Mutação , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Homologia de Sequência de Aminoácidos , Especificidade da Espécie
12.
Biochem Biophys Res Commun ; 517(4): 755-761, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31402117

RESUMO

Ras proteins are highly conserved small GTPases in eukaryotes. GTP-bound Ras binds to effectors to trigger signaling cascades. In order to understand how extensive is the functional homology between the highly homologous proteins, S. cerevisiae Ras2 and C. albicans Ras1, we examined whether ScRas2 could functionally complement CaRas1 in activating hyphal morphogenesis as well as GPI anchor biosynthesis. We show that ScRas2 functionally complements CaRas1 in rescuing growth as well as activating hyphal growth, a process that involves plasma membrane localized Ras activating cAMP/PKA signaling via Cyr1. However, ScRas2 is unable to activate the GPI-N-acetylglucosaminyl transferase (GPI-GnT) which catalyzes the first step of GPI biosynthesis. That CaRas1 alone activates GPI-GnT and not ScRas2 suggests that this process is cAMP independent. Interestingly, CaRas1 transcriptionally activates CaGPI2, encoding a GPI-GnT subunit that has been shown to interact with CaRas1 physically. In turn, CaGPI2 downregulates CaGPI19, encoding another GPI-GnT subunit. This has direct consequences for expression of CaERG11, encoding the target of azole antifungals. This effect too is specific to CaRas1 and ScRas2 is unable to replicate it.


Assuntos
Candida albicans/metabolismo , Glicosilfosfatidilinositóis/biossíntese , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas ras/metabolismo , Sequência de Aminoácidos , AMP Cíclico/biossíntese , Ergosterol/biossíntese , Proteínas Fúngicas/metabolismo , Teste de Complementação Genética , Hifas/crescimento & desenvolvimento , Hifas/metabolismo , Saccharomyces cerevisiae/crescimento & desenvolvimento , Proteínas de Saccharomyces cerevisiae/química , Homologia de Sequência de Aminoácidos , Proteínas ras/química
13.
Am J Hematol ; 94(1): 93-102, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30370942

RESUMO

Adult B-lymphoblastic leukemia (B-ALL) is a hematological malignancy characterized by genetic heterogeneity. Despite successful remission induction with classical chemotherapeutics and novel targeted agents, enduring remission is often hampered by disease relapse due to outgrowth of a pre-existing subclone resistant against the treatment. In this study, we show that small glycophosphatidylinositol (GPI)-anchor deficient CD52-negative B-cell populations are frequently present already at diagnosis in B-ALL patients, but not in patients suffering from other B-cell malignancies. We demonstrate that the GPI-anchor negative phenotype results from loss of mRNA expression of the PIGH gene, which is involved in the first step of GPI-anchor synthesis. Loss of PIGH mRNA expression within these B-ALL cells follows epigenetic silencing rather than gene mutation or deletion. The coinciding loss of CD52 membrane expression may contribute to the development of resistance to alemtuzumab (ALM) treatment in B-ALL patients resulting in the outgrowth of CD52-negative escape variants. Additional treatment with 5-aza-2'-deoxycytidine may restore expression of CD52 and revert ALM resistance.


Assuntos
Linfócitos B/metabolismo , Antígeno CD52/deficiência , Metilação de DNA/efeitos dos fármacos , Regulação Leucêmica da Expressão Gênica , Inativação Gênica , Glicosilfosfatidilinositóis/deficiência , Proteínas de Membrana/genética , Proteínas de Neoplasias/deficiência , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Alemtuzumab/uso terapêutico , Antimetabólitos Antineoplásicos/farmacologia , Antimetabólitos Antineoplásicos/uso terapêutico , Antineoplásicos Imunológicos/uso terapêutico , Linfócitos B/patologia , Antígeno CD52/biossíntese , Antígeno CD52/genética , Linhagem Celular Tumoral , Decitabina/farmacologia , Decitabina/uso terapêutico , Regulação para Baixo/efeitos dos fármacos , Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Glicosilfosfatidilinositóis/biossíntese , Glicosilfosfatidilinositóis/genética , Humanos , Proteínas de Membrana/biossíntese , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Fenótipo , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Neoplásico/biossíntese , RNA Neoplásico/genética
14.
J Virol ; 91(11)2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28331092

RESUMO

Monkeypox virus (MPXV) is a human pathogen that is a member of the Orthopoxvirus genus, which includes Vaccinia virus and Variola virus (the causative agent of smallpox). Human monkeypox is considered an emerging zoonotic infectious disease. To identify host factors required for MPXV infection, we performed a genome-wide insertional mutagenesis screen in human haploid cells. The screen revealed several candidate genes, including those involved in Golgi trafficking, glycosaminoglycan biosynthesis, and glycosylphosphatidylinositol (GPI)-anchor biosynthesis. We validated the role of a set of vacuolar protein sorting (VPS) genes during infection, VPS51 to VPS54 (VPS51-54), which comprise the Golgi-associated retrograde protein (GARP) complex. The GARP complex is a tethering complex involved in retrograde transport of endosomes to the trans-Golgi apparatus. Our data demonstrate that VPS52 and VPS54 were dispensable for mature virion (MV) production but were required for extracellular virus (EV) formation. For comparison, a known antiviral compound, ST-246, was used in our experiments, demonstrating that EV titers in VPS52 and VPS54 knockout (KO) cells were comparable to levels exhibited by ST-246-treated wild-type cells. Confocal microscopy was used to examine actin tail formation, one of the viral egress mechanisms for cell-to-cell dissemination, and revealed an absence of actin tails in VPS52KO- or VPS54KO-infected cells. Further evaluation of these cells by electron microscopy demonstrated a decrease in levels of wrapped viruses (WVs) compared to those seen with the wild-type control. Collectively, our data demonstrate the role of GARP complex genes in double-membrane wrapping of MVs necessary for EV formation, implicating the host endosomal trafficking pathway in orthopoxvirus infection.IMPORTANCE Human monkeypox is an emerging zoonotic infectious disease caused by Monkeypox virus (MPXV). Of the two MPXV clades, the Congo Basin strain is associated with severe disease, increased mortality, and increased human-to-human transmission relative to the West African strain. Monkeypox is endemic in regions of western and central Africa but was introduced into the United States in 2003 from the importation of infected animals. The threat of MPXV and other orthopoxviruses is increasing due to the absence of routine smallpox vaccination leading to a higher proportion of naive populations. In this study, we have identified and validated candidate genes that are required for MPXV infection, specifically, those associated with the Golgi-associated retrograde protein (GARP) complex. Identifying host targets required for infection that prevents extracellular virus formation such as the GARP complex or the retrograde pathway can provide a potential target for antiviral therapy.


Assuntos
Endossomos/metabolismo , Interações Hospedeiro-Patógeno , Proteínas de Membrana/genética , Monkeypox virus/fisiologia , Proteínas de Transporte Vesicular/metabolismo , Actinas/efeitos dos fármacos , Actinas/metabolismo , Animais , Benzamidas/farmacologia , Transporte Biológico , Linhagem Celular , Genoma Humano , Glicosaminoglicanos/biossíntese , Glicosaminoglicanos/genética , Glicosilfosfatidilinositóis/biossíntese , Complexo de Golgi/genética , Complexo de Golgi/metabolismo , Haploidia , Humanos , Isoindóis/farmacologia , Proteínas de Membrana/metabolismo , Mpox/virologia , Mutagênese Insercional , Proteínas de Transporte Vesicular/genética , Carga Viral , Replicação Viral
15.
IUBMB Life ; 70(5): 355-383, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29679465

RESUMO

Glycosylphosphatidylinositol (GPI)-anchored proteins (GPI-APs) are present ubiquitously at the cell surface in all eukaryotes. They play a crucial role in the interaction of the cell with its external environment, allowing the cell to receive signals, respond to challenges, and mediate adhesion. In yeast and fungi, they also participate in the structural integrity of the cell wall and are often essential for survival. Roughly four decades after the discovery of the first GPI-APs, this review provides an overview of the insights gained from studies of the GPI biosynthetic pathway and the future challenges in the field. In particular, we focus on the biosynthetic pathway in Saccharomyces cerevisiae, which has for long been studied as a model organism. Where available, we also provide information about the GPI biosynthetic steps in other yeast/ fungi. Although the core structure of the GPI anchor is conserved across organisms, several variations are built into the biosynthetic pathway. The present Review specifically highlights these variations and their implications. There is growing evidence to suggest that several phenotypes are common to GPI deficiency and should be expected in GPI biosynthetic mutants. However, it appears that several phenotypes are unique to a specific step in the pathway and may even be species-specific. These could suggest the points at which the GPI biosynthetic pathway intersects with other important cellular pathways and could be points of regulation. They could be of particular significance in the study of pathogenic fungi and in identification of new and specific antifungal drugs/ drug targets. © 2018 IUBMB Life, 70(5):355-383, 2018.


Assuntos
Candida albicans/metabolismo , Proteínas Fúngicas/metabolismo , Glicosilfosfatidilinositóis/biossíntese , Proteínas Ligadas a Lipídeos/metabolismo , Saccharomyces cerevisiae/metabolismo , Antifúngicos/farmacologia , Candida albicans/efeitos dos fármacos , Candida albicans/genética , Sequência de Carboidratos , Proteínas Fúngicas/química , Proteínas Fúngicas/genética , Regulação Fúngica da Expressão Gênica , Glicosilfosfatidilinositóis/química , Glicosilfosfatidilinositóis/deficiência , Glicosilfosfatidilinositóis/genética , Glicosilfosfatidilinositóis/metabolismo , Humanos , Proteínas Ligadas a Lipídeos/química , Proteínas Ligadas a Lipídeos/genética , Doenças Metabólicas/genética , Doenças Metabólicas/metabolismo , Doenças Metabólicas/patologia , Mutação , Fenótipo , Saccharomyces cerevisiae/efeitos dos fármacos , Saccharomyces cerevisiae/genética , Convulsões/genética , Convulsões/metabolismo , Convulsões/patologia , Transdução de Sinais , Especificidade da Espécie
16.
PLoS Genet ; 11(3): e1005082, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25807459

RESUMO

Glycosylphosphatidylinositol (GPI) is a post-translational modification resulting in the attachment of modified proteins to the outer leaflet of the plasma membrane. Tissue culture experiments have shown GPI-anchored proteins (GPI-APs) to be targeted to the apical membrane of epithelial cells. However, the in vivo importance of this targeting has not been investigated since null mutations in GPI biosynthesis enzymes in mice result in very early embryonic lethality. Missense mutations in the human GPI biosynthesis enzyme pigv are associated with a multiple congenital malformation syndrome with a high frequency of Hirschsprung disease and renal anomalies. However, it is currently unknown how these phenotypes are linked to PIGV function. Here, we identify a temperature-sensitive hypomorphic allele of PIGV in Caenorhabditis elegans, pigv-1(qm34), enabling us to study the role of GPI-APs in development. At the restrictive temperature we found a 75% reduction in GPI-APs at the surface of embryonic cells. Consequently, ~80% of pigv-1(qm34) embryos arrested development during the elongation phase of morphogenesis, exhibiting internal cysts and/or surface ruptures. Closer examination of the defects revealed them all to be the result of breaches in epithelial tissues: cysts formed in the intestine and excretory canal, and ruptures occurred through epidermal cells, suggesting weakening of the epithelial membrane or membrane-cortex connection. Knockdown of piga-1, another GPI biosynthesis enzymes resulted in similar phenotypes. Importantly, fortifying the link between the apical membrane and actin cortex by overexpression of the ezrin/radixin/moesin ortholog ERM-1, significantly rescued cyst formation and ruptures in the pigv-1(qm34) mutant. In conclusion, we discovered GPI-APs play a critical role in maintaining the integrity of the epithelial tissues, allowing them to withstand the pressure and stresses of morphogenesis. Our findings may help to explain some of the phenotypes observed in human syndromes associated with pigv mutations.


Assuntos
Proteínas de Caenorhabditis elegans/genética , Proteínas do Citoesqueleto/genética , Desenvolvimento Embrionário/genética , Glicosilfosfatidilinositóis/biossíntese , Doença de Hirschsprung/genética , Manosiltransferases/genética , Animais , Caenorhabditis elegans/genética , Caenorhabditis elegans/crescimento & desenvolvimento , Proteínas de Caenorhabditis elegans/biossíntese , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas do Citoesqueleto/biossíntese , Proteínas do Citoesqueleto/metabolismo , Células Epiteliais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Glicosilfosfatidilinositóis/genética , Doença de Hirschsprung/metabolismo , Humanos , Manosiltransferases/biossíntese , Manosiltransferases/metabolismo , Camundongos , Mutação , Processamento de Proteína Pós-Traducional/genética , Temperatura
17.
J Lipid Res ; 57(1): 6-24, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26563290

RESUMO

Glycosylphosphatidylinositols (GPIs) act as membrane anchors of many eukaryotic cell surface proteins. GPIs in various organisms have a common backbone consisting of ethanolamine phosphate (EtNP), three mannoses (Mans), one non-N-acetylated glucosamine, and inositol phospholipid, whose structure is EtNP-6Manα-2Manα-6Manα-4GlNα-6myoinositol-P-lipid. The lipid part is either phosphatidylinositol of diacyl or 1-alkyl-2-acyl form, or inositol phosphoceramide. GPIs are attached to proteins via an amide bond between the C-terminal carboxyl group and an amino group of EtNP. Fatty chains of inositol phospholipids are inserted into the outer leaflet of the plasma membrane. More than 150 different human proteins are GPI anchored, whose functions include enzymes, adhesion molecules, receptors, protease inhibitors, transcytotic transporters, and complement regulators. GPI modification imparts proteins with unique characteristics, such as association with membrane microdomains or rafts, transient homodimerization, release from the membrane by cleavage in the GPI moiety, and apical sorting in polarized cells. GPI anchoring is essential for mammalian embryogenesis, development, neurogenesis, fertilization, and immune system. Mutations in genes involved in remodeling of the GPI lipid moiety cause human diseases characterized by neurological abnormalities. Yeast Saccharomyces cerevisiae has >60 GPI-anchored proteins (GPI-APs). GPI is essential for growth of yeast. In this review, we discuss biosynthesis of GPI-APs in mammalian cells and yeast with emphasis on the lipid moiety.


Assuntos
Glicosilfosfatidilinositóis/biossíntese , Glicosilfosfatidilinositóis/metabolismo , Lipídeos/biossíntese , Animais , Membrana Celular/metabolismo , Humanos , Microdomínios da Membrana/metabolismo , Proteínas de Membrana/biossíntese , Proteínas de Membrana/metabolismo , Peroxissomos/metabolismo
18.
Yeast ; 33(2): 37-42, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26460143

RESUMO

ARV1 is involved in regulating lipid homeostasis but also in the biosynthesis of glycosylphosphatidylinositol (GPI) in Saccharomyces cerevisiae. Here, we examined whether human ARV1 can complement the role of yeast ARV1 in GPI biosynthesis. Overexpression of human ARV1 could rescue the phenotypes associated with GPI anchor synthesis defect in the yeast arv1Δ mutant. The results suggest that Arv1 function in GPI biosynthesis may be conserved in all eukaryotes, from yeast to humans.


Assuntos
Proteínas de Transporte/metabolismo , Glicosilfosfatidilinositóis/biossíntese , Proteínas de Membrana/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Transporte/genética , Expressão Gênica , Teste de Complementação Genética , Homeostase , Humanos , Metabolismo dos Lipídeos , Proteínas de Membrana/genética , Mutação , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética
19.
Yeast ; 33(8): 365-83, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27337589

RESUMO

CaGpi14 is the catalytic subunit of the first mannosyltransferase that is involved in the glycosylphosphatidylinositol (GPI) biosynthetic pathway in Candida albicans. We show that CaGPI14 is able to rescue a conditionally lethal gpi14 mutant of Saccharomyces cerevisiae, unlike its mammalian homologue. The depletion of this enzyme in C. albicans leads to severe growth defects, besides causing deficiencies in GPI anchor levels. In addition, CaGpi14 depletion results in cell wall defects and upregulation of the cell wall integrity response pathway. This in turn appears to trigger the osmotic-stress dependent activation of the HOG1 pathway and an upregulation of HOG1 as well as its downstream target, SKO1, a known suppressor of expression of hyphae-specific genes. Consistent with this, mutants of CaGPI14 are unable to undergo hyphal transformations in different hyphae-inducing media, under conditions that produce abundant hyphae in the wild-type cells. Hyphal defects in the CaGPI14 mutants could not be attributed either to reduced protein kinase C activation or to defective Ras signalling in these cells but appeared to be driven by perturbations in the HOG1 pathway. Copyright © 2016 John Wiley & Sons, Ltd.


Assuntos
Candida albicans/crescimento & desenvolvimento , Domínio Catalítico , Parede Celular/metabolismo , Glicosilfosfatidilinositóis/biossíntese , Hifas/crescimento & desenvolvimento , Manosiltransferases/metabolismo , Fatores de Transcrição de Zíper de Leucina Básica/genética , Candida albicans/enzimologia , Candida albicans/genética , Genes Letais , Hifas/enzimologia , Hifas/genética , Manosiltransferases/química , Manosiltransferases/genética , Proteínas Quinases Ativadas por Mitógeno/genética , Morfogênese , Mutação , Pressão Osmótica , Proteína Quinase C/metabolismo , Proteínas Repressoras/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Espectrometria de Massas por Ionização por Electrospray
20.
Blood ; 124(18): 2804-11, 2014 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-25237200

RESUMO

Paroxysmal nocturnal hemoglobinuria (PNH) is a rare bone marrow failure disorder that manifests with hemolytic anemia, thrombosis, and peripheral blood cytopenias. The absence of two glycosylphosphatidylinositol (GPI)-anchored proteins, CD55 and CD59, leads to uncontrolled complement activation that accounts for hemolysis and other PNH manifestations. GPI anchor protein deficiency is almost always due to somatic mutations in phosphatidylinositol glycan class A (PIGA), a gene involved in the first step of GPI anchor biosynthesis; however, alternative mutations that cause PNH have recently been discovered. In addition, hypomorphic germ-line PIGA mutations that do not cause PNH have been shown to be responsible for a condition known as multiple congenital anomalies-hypotonia-seizures syndrome 2. Eculizumab, a first-in-class monoclonal antibody that inhibits terminal complement, is the treatment of choice for patients with severe manifestations of PNH. Bone marrow transplantation remains the only cure for PNH but should be reserved for patients with suboptimal response to eculizumab.


Assuntos
Hemoglobinúria Paroxística/genética , Transplante de Medula Óssea , Glicosilfosfatidilinositóis/biossíntese , Hemoglobinúria Paroxística/diagnóstico , Hemoglobinúria Paroxística/fisiopatologia , Hemoglobinúria Paroxística/terapia , Humanos , Monitorização Fisiológica , Mutação/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA