Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 791
Filtrar
1.
J Biol Chem ; 300(1): 105564, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38103644

RESUMO

The polysialyltransferases ST8SIA2 and ST8SIA4 and their product, polysialic acid (polySia), are known to be related to cancers and mental disorders. ST8SIA2 and ST8SIA4 have conserved amino acid (AA) sequence motifs essential for the synthesis of the polySia structures on the neural cell adhesion molecule. To search for a new motif in the polysialyltransferases, we adopted the in silico Individual Meta Random Forest program that can predict disease-related AA substitutions. The Individual Meta Random Forest program predicted a new eight-amino-acids sequence motif consisting of highly pathogenic AA residues, thus designated as the pathogenic (P) motif. A series of alanine point mutation experiments in the pathogenic motif (P motif) showed that most P motif mutants lost the polysialylation activity without changing the proper enzyme expression levels or localization in the Golgi. In addition, we evaluated the enzyme stability of the P motif mutants using newly established calculations of mutation energy, demonstrating that the subtle change of the conformational energy regulates the activity. In the AlphaFold2 model, we found that the P motif was a buried ß-strand underneath the known surface motifs unique to ST8SIA2 and ST8SIA4. Taken together, the P motif is a novel buried ß-strand that regulates the full activity of polysialyltransferases from the inside of the molecule.


Assuntos
Mutação , Sialiltransferases , Humanos , Motivos de Aminoácidos/genética , Substituição de Aminoácidos , Simulação por Computador , Complexo de Golgi/enzimologia , Complexo de Golgi/metabolismo , Moléculas de Adesão de Célula Nervosa/química , Moléculas de Adesão de Célula Nervosa/metabolismo , Mutação Puntual , Conformação Proteica em Folha beta , Transporte Proteico , Algoritmo Florestas Aleatórias , Ácidos Siálicos/metabolismo , Sialiltransferases/química , Sialiltransferases/genética , Sialiltransferases/metabolismo
2.
J Biol Chem ; 299(12): 105473, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37979916

RESUMO

Vacuolar H+-ATPases (V-ATPases) are highly conserved multisubunit enzymes that maintain the distinct pH of eukaryotic organelles. The integral membrane a-subunit is encoded by tissue- and organelle-specific isoforms, and its cytosolic N-terminal domain (aNT) modulates organelle-specific regulation and targeting of V-ATPases. Organelle membranes have specific phosphatidylinositol phosphate (PIP) lipid enrichment linked to maintenance of organelle pH. In yeast, the aNT domains of the two a-subunit isoforms bind PIP lipids enriched in the organelle membranes where they reside; these interactions affect activity and regulatory properties of the V-ATPases containing each isoform. Humans have four a-subunit isoforms, and we hypothesize that the aNT domains of these isoforms will also bind to specific PIP lipids. The a1 and a2 isoforms of human V-ATPase a-subunits are localized to endolysosomes and Golgi, respectively. We determined that bacterially expressed Hua1NT and Hua2NT bind specifically to endolysosomal PIP lipids PI(3)P and PI(3,5)P2 and Golgi enriched PI(4)P, respectively. Despite the lack of canonical PIP-binding sites, we identified potential binding sites in the HuaNT domains by sequence comparisons and existing subunit structures and models. We found that mutations at a similar location in the distal loops of both HuaNT isoforms compromise binding to their cognate PIP lipids, suggesting that these loops encode PIP specificity of the a-subunit isoforms. These data suggest a mechanism through which PIP lipid binding could stabilize and activate V-ATPases in distinct organelles.


Assuntos
Fosfatos de Fosfatidilinositol , Subunidades Proteicas , ATPases Vacuolares Próton-Translocadoras , Humanos , Sítios de Ligação , Endossomos/enzimologia , Endossomos/metabolismo , Complexo de Golgi/enzimologia , Complexo de Golgi/metabolismo , Concentração de Íons de Hidrogênio , Lisossomos/enzimologia , Lisossomos/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Saccharomyces cerevisiae/enzimologia , Saccharomyces cerevisiae/metabolismo , Especificidade por Substrato , ATPases Vacuolares Próton-Translocadoras/química , ATPases Vacuolares Próton-Translocadoras/metabolismo , Domínios Proteicos
3.
J Biol Chem ; 299(10): 105192, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37625589

RESUMO

Point mutations in leucine-rich repeat kinase 2 (LRRK2) cause Parkinson's disease and augment LRRK2's kinase activity. However, cellular pathways that endogenously enhance LRRK2 kinase function have not been identified. While overexpressed Rab29 draws LRRK2 to Golgi membranes to increase LRRK2 kinase activity, there is little evidence that endogenous Rab29 performs this function under physiological conditions. Here, we identify Rab38 as a novel physiologic regulator of LRRK2 in melanocytes. In mouse melanocytes, which express high levels of Rab38, Rab32, and Rab29, knockdown (or CRISPR knockout) of Rab38, but not Rab32 or Rab29, decreases phosphorylation of multiple LRRK2 substrates, including Rab10 and Rab12, by both endogenous LRRK2 and exogenous Parkinson's disease-mutant LRRK2. In B16-F10 mouse melanoma cells, Rab38 drives LRRK2 membrane association and overexpressed kinase-active LRRK2 shows striking pericentriolar recruitment, which is dependent on the presence of endogenous Rab38 but not Rab32 or Rab29. Consistently, knockdown or mutation of BLOC-3, the guanine nucleotide exchange factor for Rab38 and Rab32, inhibits Rab38's regulation of LRRK2. Deletion or mutation of LRRK2's Rab38-binding site in the N-terminal armadillo domain decreases LRRK2 membrane association, pericentriolar recruitment, and ability to phosphorylate Rab10. In sum, our data identify Rab38 as a physiologic regulator of LRRK2 function and lend support to a model in which LRRK2 plays a central role in Rab GTPase coordination of vesicular trafficking.


Assuntos
Membranas Intracelulares , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina , Melanócitos , Proteínas rab de Ligação ao GTP , Animais , Camundongos , Complexo de Golgi/enzimologia , Complexo de Golgi/genética , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/genética , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/metabolismo , Melanócitos/metabolismo , Mutação , Doença de Parkinson/metabolismo , Fosforilação , Proteínas rab de Ligação ao GTP/genética , Proteínas rab de Ligação ao GTP/metabolismo , Células HEK293 , Humanos , Expressão Gênica , Domínios Proteicos , Ligação Proteica , Membranas Intracelulares/metabolismo
4.
J Biol Chem ; 298(9): 102281, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35863437

RESUMO

Rab22 and Rab31 belong to the Rab5 subfamily of GTPases that regulates endocytic traffic and endosomal sorting. Rab22 and Rab31 (a.k.a. Rab22b) are closely related and share 87% amino acid sequence similarity, but they show distinct intracellular localization and function in the cell. Rab22 is localized to early endosomes and regulates early endosomal recycling, while Rab31 is mostly localized to the Golgi complex with only a small fraction in the endosomes at steady state. The specific determinants that affect this differential localization, however, are unclear. In this study, we identify a novel membrane targeting domain (MTD) consisting of the C-terminal hypervariable domain (HVD), interswitch loop (ISL), and N-terminal domain as a major determinant of endosomal localization for Rab22 and Rab31, as well as Rab5. Rab22 and Rab31 share the same N-terminal domain, but we find Rab22 chimeras with Rab31 HVD exhibit phenotypic Rab31 localization to the Golgi complex, while Rab31 chimeras with the Rab22 HVD localize to early endosomes, similar to wildtype Rab22. We also find that the Rab22 HVD favors interaction with the early endosomal effector protein Rabenosyn-5, which may stabilize the Rab localization to the endosomes. The importance of effector interaction in endosomal localization is further demonstrated by the disruption of Rab22 endosomal localization in Rabenosyn-5 knockout cells and by the shift of Rab31 to the endosomes in Rabenosyn-5-overexpressing cells. Taken together, we have identified a novel MTD that mediates localization of Rab5 subfamily members to early endosomes via interaction with an effector such as Rabenosyn-5.


Assuntos
Endossomos , Complexo de Golgi , Proteínas rab de Ligação ao GTP , Animais , Cricetinae , Endossomos/enzimologia , Complexo de Golgi/enzimologia , Células HEK293 , Humanos , Células PC12 , Domínios Proteicos , Transporte Proteico , Ratos , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Vesículas Transportadoras/metabolismo , Proteínas rab de Ligação ao GTP/química , Proteínas rab de Ligação ao GTP/genética , Proteínas rab de Ligação ao GTP/metabolismo , Proteínas rab5 de Ligação ao GTP/metabolismo
5.
ACS Chem Biol ; 17(1): 240-251, 2022 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-35000377

RESUMO

Many cellular processes are dependent on correct pH levels, and this is especially important for the secretory pathway. Defects in pH homeostasis in distinct organelles cause a wide range of diseases, including disorders of glycosylation and lysosomal storage diseases. Ratiometric imaging of the pH-sensitive mutant of green fluorescent protein, pHLuorin, has allowed for targeted pH measurements in various organelles, but the required sequential image acquisition is intrinsically slow and therefore the temporal resolution is unsuitable to follow the rapid transit of cargo between organelles. Therefore, we applied fluorescence lifetime imaging microscopy (FLIM) to measure intraorganellar pH with just a single excitation wavelength. We first validated this method by confirming the pH in multiple compartments along the secretory pathway and compared the pH values obtained by the FLIM-based measurements with those obtained by conventional ratiometric imaging. Then, we analyzed the dynamic pH changes within cells treated with Bafilomycin A1, to block the vesicular ATPase, and Brefeldin A, to block endoplasmic reticulum (ER)-Golgi trafficking. Finally, we followed the pH changes of newly synthesized molecules of the inflammatory cytokine tumor necrosis factor-α while they were in transit from the ER via the Golgi to the plasma membrane. The toolbox we present here can be applied to measure intracellular pH with high spatial and temporal resolution and can be used to assess organellar pH in disease models.


Assuntos
Concentração de Íons de Hidrogênio , Imagem Óptica/métodos , Via Secretória , Adenosina Trifosfatases/antagonistas & inibidores , Brefeldina A/farmacologia , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/enzimologia , Retículo Endoplasmático/metabolismo , Complexo de Golgi/efeitos dos fármacos , Complexo de Golgi/enzimologia , Complexo de Golgi/metabolismo , Humanos , Macrolídeos/farmacologia , Microscopia de Fluorescência/métodos , Transporte Proteico
6.
J Biol Chem ; 296: 100315, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33485966

RESUMO

Lipid flipping in the membrane bilayers is a widespread eukaryotic phenomenon that is catalyzed by assorted P4-ATPases. Its occurrence, mechanism, and importance in apicomplexan parasites have remained elusive, however. Here we show that Toxoplasma gondii, an obligate intracellular parasite with high clinical relevance, can salvage phosphatidylserine (PtdSer) and phosphatidylethanolamine (PtdEtn) but not phosphatidylcholine (PtdCho) probes from its milieu. Consistently, the drug analogs of PtdCho are broadly ineffective in the parasite culture. NBD-PtdSer imported to the parasite interior is decarboxylated to NBD-PtdEtn, while the latter is not methylated to yield PtdCho, which confirms the expression of PtdSer decarboxylase but a lack of PtdEtn methyltransferase activity and suggests a role of exogenous lipids in membrane biogenesis of T. gondii. Flow cytometric quantitation of NBD-probes endorsed the selectivity of phospholipid transport and revealed a dependence of the process on energy and protein. Accordingly, our further work identified five P4-ATPases (TgP4-ATPase1-5), all of which harbor the signature residues and motifs required for phospholipid flipping. Of the four proteins expressed during the lytic cycle, TgP4-ATPase1 is present in the apical plasmalemma; TgP4-ATPase3 resides in the Golgi network along with its noncatalytic partner Ligand Effector Module 3 (TgLem3), whereas TgP4-ATPase2 and TgP4-ATPase5 localize in the plasmalemma as well as endo/cytomembranes. Last but not least, auxin-induced degradation of TgP4-ATPase1-3 impaired the parasite growth in human host cells, disclosing their crucial roles during acute infection. In conclusion, we show selective translocation of PtdEtn and PtdSer at the parasite surface and provide the underlying mechanistic and physiological insights in a model eukaryotic pathogen.


Assuntos
Adenosina Trifosfatases/genética , Bicamadas Lipídicas/metabolismo , Toxoplasma/genética , Toxoplasmose/genética , Adenosina Trifosfatases/química , Membrana Celular/genética , Membrana Celular/metabolismo , Citometria de Fluxo , Glicerofosfolipídeos/metabolismo , Complexo de Golgi/química , Complexo de Golgi/enzimologia , Humanos , Bicamadas Lipídicas/química , Lipídeos/química , Lipídeos/genética , Fosfatidilcolinas/genética , Fosfatidilcolinas/metabolismo , Fosfatidiletanolaminas/genética , Fosfatidiletanolaminas/metabolismo , Fosfatidilserinas/metabolismo , Toxoplasma/enzimologia , Toxoplasma/patogenicidade , Toxoplasmose/parasitologia
7.
J Cell Biol ; 219(8)2020 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-32479594

RESUMO

Matrix metalloproteinases (MMPs) degrade several ECM components and are crucial modulators of cell invasion and tissue organization. Although much has been reported about their function in remodeling ECM in health and disease, their trafficking across the Golgi apparatus remains poorly understood. Here we report that the cis-Golgi protein nucleobindin-1 (NUCB1) is critical for MMP2 and MT1-MMP trafficking along the Golgi apparatus. This process is Ca2+-dependent and is required for invasive MDA-MB-231 cell migration as well as for gelatin degradation in primary human macrophages. Our findings emphasize the importance of NUCB1 as an essential component of MMP transport and its overall impact on ECM remodeling.


Assuntos
Neoplasias da Mama/enzimologia , Matriz Extracelular/enzimologia , Complexo de Golgi/enzimologia , Macrófagos/enzimologia , Metaloproteinase 14 da Matriz/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Nucleobindinas/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Cálcio/metabolismo , Sinalização do Cálcio , Movimento Celular , Matriz Extracelular/patologia , Feminino , Gelatina/metabolismo , Células HEK293 , Células HeLa , Humanos , Metaloproteinase 14 da Matriz/genética , Metaloproteinase 2 da Matriz/genética , Nucleobindinas/genética , Transporte Proteico , Proteólise , Fatores de Tempo
8.
mBio ; 11(3)2020 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-32487759

RESUMO

Many species of pathogenic fungi deploy the unfolded protein response (UPR) to expand the folding capacity of the endoplasmic reticulum (ER) in proportion to the demand for virulence-related proteins that traffic through the secretory pathway. Although Ca2+ plays a pivotal role in ER function, the mechanism by which transcriptional upregulation of the protein folding machinery is coordinated with Ca2+ homeostasis is incompletely understood. In this study, we investigated the link between the UPR and genes encoding P-type Ca2+-ATPases in the human-pathogenic mold Aspergillus fumigatus We demonstrate that acute ER stress increases transcription of the srcA gene, encoding a member of the sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) family, as well as that of pmrA, encoding a secretory pathway Ca2+-ATPase (SPCA) in the Golgi membrane. Loss of the UPR transcription factor HacA prevented the induction of srcA and pmrA transcription during ER stress, defining these ER/Golgi Ca2+ pumps as novel downstream targets of this pathway. While deletion of srcA alone caused no major deficiencies, a ΔsrcA/ΔpmrA mutant displayed a severe polarity defect, was hypersensitive to ER stress, and showed attenuated virulence. In addition, cell wall analyses revealed a striking reduction in mannose levels in the absence of both Ca2+ pumps. The ΔhacA mutant was hypersensitive to agents that block calcineurin-dependent signaling, consistent with a functional coupling between the UPR and Ca2+ homeostasis. Together, these findings demonstrate that the UPR integrates the need for increased levels of chaperone and folding enzymes with an influx of Ca2+ into the secretory pathway to support fungal growth, stress adaptation, and pathogenicity.IMPORTANCE The UPR is an intracellular signal transduction pathway that maintains homeostasis of the ER. The pathway is also tightly linked to the expression of virulence-related traits in diverse species of human-pathogenic and plant-pathogenic fungal species, including the predominant mold pathogen infecting humans, Aspergillus fumigatus Despite advances in the understanding of UPR signaling, the linkages and networks that are governed by this pathway are not well defined. In this study, we revealed that the UPR is a major driving force for stimulating Ca2+ influx at the ER and Golgi membranes and that the coupling between the UPR and Ca2+ import is important for virulence, cell wall biosynthesis, and resistance to antifungal compounds that inhibit Ca2+ signaling.


Assuntos
Adenosina Trifosfatases/metabolismo , Aspergillus fumigatus/enzimologia , Aspergillus fumigatus/patogenicidade , Parede Celular/fisiologia , Estresse do Retículo Endoplasmático , Resposta a Proteínas não Dobradas , Células A549 , Células Epiteliais Alveolares/microbiologia , Animais , Aspergillus fumigatus/genética , Cálcio/metabolismo , Retículo Endoplasmático/enzimologia , Feminino , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Complexo de Golgi/enzimologia , Humanos , Masculino , Camundongos , Transdução de Sinais , Virulência
9.
Cell Adh Migr ; 14(1): 96-105, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32419574

RESUMO

CD147 is a widely expressed matrix metalloproteinase inducer involved in the regulation of cell migration. The high glycosylation and ability to undergo oligomerization have been linked to CD147 function, yet there is limited understanding on the molecular mechanisms behind these processes. The current study demonstrates that the expression of Golgi α1,2-mannosidase I is key to maintaining the cell surface organization of CD147 during cell migration. Using an in vitro model of stratified human corneal epithelial wound healing, we show that CD147 is clustered within lateral plasma membranes at the leading edge of adjacent migrating cells. This localization correlates with a surge in matrix metalloproteinase activity and an increase in the expression of α1,2-mannosidase subtype IC (MAN1C1). Global inhibition of α1,2-mannosidase I activity with deoxymannojirimycin markedly attenuates the glycosylation of CD147 and disrupts its surface distribution at the leading edge, concomitantly reducing the expression of matrix metalloproteinase-9. Likewise, treatment with deoxymannojirimycin or siRNA-mediated knockdown of MAN1C1 impairs the ability of the carbohydrate-binding protein galectin-3 to stimulate CD147 clustering in unwounded cells. We conclude that the mannose-trimming activity of α1,2-mannosidase I coordinates the clustering and compartmentalization of CD147 that follows an epithelial injury.


Assuntos
Basigina/metabolismo , Movimento Celular , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Complexo de Golgi/enzimologia , Manosidases/metabolismo , Membrana Celular/metabolismo , Epitélio Corneano/citologia , Galectina 3/metabolismo , Humanos , Polissacarídeos/química , Polissacarídeos/metabolismo
10.
Bioorg Med Chem ; 28(11): 115492, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32291147

RESUMO

Effective chemotherapy for solid cancers is challenging due to a limitation in permeation that prevents anticancer drugs from reaching the center of the tumor, therefore unable to limit cancer cell growth. To circumvent this issue, we planned to apply the drugs directly at the center by first collapsing the outer structure. For this, we focused on cell-cell communication (CCC) between N-glycans and proteins at the tumor cell surface. Mature N-glycans establish CCC; however, CCC is hindered when numerous immature N-glycans are present at the cell surface. Inhibition of Golgi mannosidases (GMs) results in the transport of immature N-glycans to the cell surface. This can be employed to disrupt CCC. Here, we describe the molecular design and synthesis of an improved GM inhibitor with a non-sugar mimic scaffold that was screened from a compound library. The synthesized compounds were tested for enzyme inhibition ability and inhibition of spheroid formation using cell-based methods. Most of the compounds designed and synthesized exhibited GM inhibition at the cellular level. Of those, AR524 had higher inhibitory activity than a known GM inhibitor, kifunensine. Moreover, AR524 inhibited spheroid formation of human malignant cells at low concentration (10 µM), based on the disruption of CCC by GM inhibition.


Assuntos
Desenho de Fármacos , Inibidores Enzimáticos/farmacologia , Complexo de Golgi/enzimologia , Manosidases/antagonistas & inibidores , Esferoides Celulares/efeitos dos fármacos , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Humanos , Manosidases/metabolismo , Estrutura Molecular , Imagem Óptica , Esferoides Celulares/metabolismo , Relação Estrutura-Atividade
11.
Exp Cell Res ; 390(2): 111961, 2020 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-32209306

RESUMO

N-alpha-acetyltransferase 80 (NAA80) was recently demonstrated to acetylate the N-terminus of actin, with NAA80 knockout cells showing actin cytoskeleton-related phenotypes, such as increased formation of membrane protrusions and accelerated migration. Here we report that NAA80 knockout cells additionally display fragmentation of the Golgi apparatus. We further employed rescue assays to demonstrate that this phenotype is connected to the ability of NAA80 to modify actin. Thus, re-expression of NAA80, which leads to re-establishment of actin's N-terminal acetyl group, rescued the Golgi fragmentation, whereas a catalytic dead NAA80 mutant could neither restore actin Nt-acetylation nor Golgi structure. The Golgi phenotype of NAA80 KO cells was shared by both migrating and non-migrating cells and live-cell imaging indicated increased Golgi dynamics in migrating NAA80 KO cells. Finally, we detected a drastic increase in the amount of F-actin in cells lacking NAA80, suggesting a causal relationship between this effect and the observed re-organization of Golgi structure. The findings further underscore the importance of actin Nt-acetylation and provide novel insight into its cellular roles, suggesting a mechanistic link between actin modification state and Golgi organization.


Assuntos
Acetiltransferases/genética , Citoesqueleto de Actina/enzimologia , Actinas/genética , Actinas/metabolismo , Complexo de Golgi/enzimologia , Processamento de Proteína Pós-Traducional , Acetilação , Acetiltransferases/deficiência , Citoesqueleto de Actina/ultraestrutura , Diferenciação Celular , Linhagem Celular Tumoral , Movimento Celular , Fibroblastos/metabolismo , Fibroblastos/ultraestrutura , Complexo de Golgi/ultraestrutura , Humanos , Fenótipo , Imagem com Lapso de Tempo
12.
FEMS Yeast Res ; 20(1)2020 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-31942998

RESUMO

One strategy for overcoming infectious diseases caused by drug-resistant fungi involves combining drugs rendered inactive by resistance with agents targeting the drug resistance mechanism. The antifungal activity of n-dodecanol disappears as incubation time passes. In Saccharomyces cerevisiae, anethole, a principal component of anise oil, prolongs the transient antifungal effect of dodecanol by downregulating genes of multidrug efflux pumps, mainly PDR5. However, the detailed mechanisms of dodecanol's antifungal action and the anethole-induced prolonged antifungal action of dodecanol are unknown. Screening of S. cerevisiae strains lacking genes related to Ca2+ homeostasis and signaling identified a pmr1Δ strain lacking Golgi Ca2+-ATPase as more sensitive to dodecanol than the parental strain. Dodecanol and the dodecanol + anethole combination significantly increased intracellular Ca2+ levels in both strains, but the mutant failed to clear intracellular Ca2+ accumulation. Further, dodecanol and the drug combination reduced PMR1 expression and did not lead to specific localization of Pmr1p in the parental strain after 4-h treatment. By contrast with the parental strain, dodecanol did not stimulate PDR5 expression in pmr1Δ. Based on these observations, we propose that the antifungal activity of dodecanol is related to intracellular Ca2+ accumulation, possibly dependent on PMR1 function, with anethole enabling Ca2+ accumulation by restricting dodecanol efflux.


Assuntos
Anisóis/farmacologia , ATPases Transportadoras de Cálcio/genética , Cálcio/metabolismo , Dodecanol/farmacologia , Deleção de Genes , Chaperonas Moleculares/genética , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/efeitos dos fármacos , Derivados de Alilbenzenos , Anisóis/química , Antifúngicos/química , Antifúngicos/farmacologia , ATPases Transportadoras de Cálcio/efeitos dos fármacos , ATPases Transportadoras de Cálcio/metabolismo , Dodecanol/química , Sinergismo Farmacológico , Citometria de Fluxo , Complexo de Golgi/enzimologia , Chaperonas Moleculares/efeitos dos fármacos , Chaperonas Moleculares/metabolismo , RNA Fúngico/química , RNA Fúngico/isolamento & purificação , Reação em Cadeia da Polimerase em Tempo Real , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/efeitos dos fármacos , Proteínas de Saccharomyces cerevisiae/metabolismo , Transdução de Sinais/genética
13.
Biochem J ; 476(16): 2321-2346, 2019 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-31462439

RESUMO

Phosphatidylinositol 4-phosphate (PI4P) is a membrane glycerophospholipid and a major regulator of the characteristic appearance of the Golgi complex as well as its vesicular trafficking, signalling and metabolic functions. Phosphatidylinositol 4-kinases, and in particular the PI4KIIIß isoform, act in concert with PI4P to recruit macromolecular complexes to initiate the biogenesis of trafficking vesicles for several Golgi exit routes. Dysregulation of Golgi PI4P metabolism and the PI4P protein interactome features in many cancers and is often associated with tumour progression and a poor prognosis. Increased expression of PI4P-binding proteins, such as GOLPH3 or PITPNC1, induces a malignant secretory phenotype and the release of proteins that can remodel the extracellular matrix, promote angiogenesis and enhance cell motility. Aberrant Golgi PI4P metabolism can also result in the impaired post-translational modification of proteins required for focal adhesion formation and cell-matrix interactions, thereby potentiating the development of aggressive metastatic and invasive tumours. Altered expression of the Golgi-targeted PI 4-kinases, PI4KIIIß, PI4KIIα and PI4KIIß, or the PI4P phosphate Sac1, can also modulate oncogenic signalling through effects on TGN-endosomal trafficking. A Golgi trafficking role for a PIP 5-kinase has been recently described, which indicates that PI4P is not the only functionally important phosphoinositide at this subcellular location. This review charts new developments in our understanding of phosphatidylinositol 4-kinase function at the Golgi and how PI4P-dependent trafficking can be deregulated in malignant disease.


Assuntos
1-Fosfatidilinositol 4-Quinase/metabolismo , Complexo de Golgi/enzimologia , Proteínas de Neoplasias/metabolismo , Neoplasias/enzimologia , Fosfatos de Fosfatidilinositol/metabolismo , Vesículas Secretórias/enzimologia , Animais , Complexo de Golgi/patologia , Humanos , Proteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Neoplasias/patologia , Vesículas Secretórias/patologia
14.
Elife ; 82019 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-31433293

RESUMO

Increased adrenergic tone resulting from cardiovascular stress leads to development of heart failure, in part, through chronic stimulation of ß1 adrenergic receptors (ßARs) on cardiac myocytes. Blocking these receptors is part of the basis for ß-blocker therapy for heart failure. Recent data demonstrate that G protein-coupled receptors (GPCRs), including ßARs, are activated intracellularly, although the biological significance is unclear. Here we investigated the functional role of Golgi ßARs in rat cardiac myocytes and found they activate Golgi localized, prohypertrophic, phosphoinositide hydrolysis, that is not accessed by cell surface ßAR stimulation. This pathway is accessed by the physiological neurotransmitter norepinephrine (NE) via an Oct3 organic cation transporter. Blockade of Oct3 or specific blockade of Golgi resident ß1ARs prevents NE dependent cardiac myocyte hypertrophy. This clearly defines a pathway activated by internal GPCRs in a biologically relevant cell type and has implications for development of more efficacious ß-blocker therapies.


Assuntos
Cardiomegalia/fisiopatologia , Complexo de Golgi/metabolismo , Miócitos Cardíacos/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Fosfoinositídeo Fosfolipase C/metabolismo , Receptores Adrenérgicos beta 1/metabolismo , Animais , Complexo de Golgi/enzimologia , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Hidrólise , Miócitos Cardíacos/enzimologia , Ratos
15.
J Biol Chem ; 294(28): 10954-10968, 2019 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-31152060

RESUMO

Neurite outgrowth requires coordinated cytoskeletal rearrangements in the growth cone and directional membrane delivery from the neuronal soma. As an essential Rho guanine nucleotide exchange factor (GEF), TRIO is necessary for cytoskeletal dynamics during neurite outgrowth, but its participation in the membrane delivery is unclear. Using co-localization studies, live-cell imaging, and fluorescence recovery after photobleaching analysis, along with neurite outgrowth assay and various biochemical approaches, we here report that in mouse cerebellar granule neurons, TRIO protein pools at the Golgi and regulates membrane trafficking by controlling the directional maintenance of both RAB8 (member RAS oncogene family 8)- and RAB10-positive membrane vesicles. We found that the spectrin repeats in Golgi-resident TRIO confer RAB8 and RAB10 activation by interacting with and activating the RAB GEF RABIN8. Constitutively active RAB8 or RAB10 could partially restore the neurite outgrowth of TRIO-deficient cerebellar granule neurons, suggesting that TRIO-regulated membrane trafficking has an important functional role in neurite outgrowth. Our results also suggest cross-talk between Rho GEF and Rab GEF in controlling both cytoskeletal dynamics and membrane trafficking during neuronal development. They further highlight how protein pools localized to specific organelles regulate crucial cellular activities and functions. In conclusion, our findings indicate that TRIO regulates membrane trafficking during neurite outgrowth in coordination with its GEF-dependent function in controlling cytoskeletal dynamics via Rho GTPases.


Assuntos
Fatores de Troca do Nucleotídeo Guanina/metabolismo , Neuritos/metabolismo , Crescimento Neuronal/fisiologia , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Movimento Celular , Cerebelo/metabolismo , Complexo de Golgi/enzimologia , Complexo de Golgi/metabolismo , Cones de Crescimento/metabolismo , Fatores de Troca do Nucleotídeo Guanina/fisiologia , Humanos , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Neuritos/fisiologia , Neurônios/metabolismo , Fosfoproteínas/fisiologia , Ligação Proteica , Proteínas Serina-Treonina Quinases/fisiologia , Transporte Proteico , Transdução de Sinais/fisiologia , Proteínas rab de Ligação ao GTP/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo
16.
PLoS One ; 14(5): e0216132, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31067280

RESUMO

Golgi α-mannosidase II (GMII) is a glycoside hydrolase playing a crucial role in the N-glycosylation pathway. In various tumour cell lines, the distribution of N-linked sugars on the cell surface is modified and correlates with the progression of tumour metastasis. GMII therefore is a possible molecular target for anticancer agents. Here, we describe the identification of a non-competitive GMII inhibitor using computer-aided drug design methods including identification of a possible allosteric binding site, pharmacophore search and virtual screening.


Assuntos
Desenho de Fármacos , Complexo de Golgi/enzimologia , alfa-Manosidase/ultraestrutura , Sítio Alostérico , Animais , Sítios de Ligação , Clonagem Molecular , Drosophila melanogaster/enzimologia , Simulação de Acoplamento Molecular , Estrutura Quaternária de Proteína , Proteínas Recombinantes
17.
Hum Mol Genet ; 28(13): 2133-2142, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-30806661

RESUMO

Hereditary multiple osteochondromas (HMO) is a rare autosomal dominant skeletal disorder, caused by heterozygous variants in either EXT1 or EXT2, which encode proteins involved in the biogenesis of heparan sulphate. Pathogenesis and genotype-phenotype correlations remain poorly understood. We studied 114 HMO families (158 affected individuals) with causative EXT1 or EXT2 variants identified by Sanger sequencing, or multiplex ligation-dependent probe amplification and qPCR. Eighty-seven disease-causative variants (55 novel and 32 known) were identified including frameshift (42%), nonsense (32%), missense (11%), splicing (10%) variants and genomic rearrangements (5%). Informative clinical features were available for 42 EXT1 and 27 EXT2 subjects. Osteochondromas were more frequent in EXT1 as compared to EXT2 patients. Anatomical distribution of lesions showed significant differences based on causative gene. Microscopy analysis for selected EXT1 and EXT2 variants verified that EXT1 and EXT2 mutants failed to co-localize each other and loss Golgi localization by surrounding the nucleus and/or assuming a diffuse intracellular distribution. In a cell viability study, cells expressing EXT1 and EXT2 mutants proliferated more slowly than cells expressing wild-type proteins. This confirms the physiological relevance of EXT1 and EXT2 Golgi co-localization and the key role of these proteins in the cell cycle. Taken together, our data expand genotype-phenotype correlations, offer further insights in the pathogenesis of HMO and open the path to future therapies.


Assuntos
Exostose Múltipla Hereditária/genética , N-Acetilglucosaminiltransferases/genética , Proliferação de Células , Sobrevivência Celular , Feminino , Estudos de Associação Genética , Complexo de Golgi/enzimologia , Células HEK293 , Humanos , Masculino , Mutação , N-Acetilglucosaminiltransferases/análise
18.
FEBS J ; 285(24): 4674-4683, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30387551

RESUMO

Egg yolk phosvitins, generated through the fragmentation of vitellogenins (VTGs), are among the most heavily phosphorylated proteins ever described. Despite the early discovery in 1900 that chicken phosvitin is a phosphoprotein and its subsequent employment as an artificial substrate for a number of protein kinases, the identity of the enzyme(s) responsible for its phosphorylation remained a matter of conjecture until present. Here, we provide evidence that phosvitin phosphorylation is catalyzed by a family with sequence similarity 20, member C (Fam20C), an atypical protein kinase recently identified as the genuine casein kinase and responsible for the phosphorylation of many other secreted proteins at residues specified by the S-x-E/pS consensus. Such a conclusion is grounded on the following observations: (a) the levels of Fam20C and phosphorylated VTG rise in parallel upon treatment of zebrafish with oestrogens; (b) zebrafish phosvitin is readily phosphorylated upon coexpression in U2OS cells with Fam20C, but not with its catalytically inactive mutant; (c) a peptide reproducing a stretch of 12 serines, which are phosphorylated in chicken phosvitin despite lacking the C-terminal priming motif S-x-E, is efficiently phosphorylated by both recombinant and native Fam20C. The last finding expands the repertoire of potential targets of Fam20C to include several proteins known to harbor (p-Ser)n clusters not specified by any known kinase consensus.


Assuntos
Caseína Quinase I/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Complexo de Golgi/enzimologia , Peptídeos/metabolismo , Fosfoproteínas/metabolismo , Fosvitina/metabolismo , Sequência de Aminoácidos , Animais , Galinhas , Consenso , Humanos , Fosforilação , Homologia de Sequência , Peixe-Zebra
19.
Am J Physiol Renal Physiol ; 315(5): F1465-F1477, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30089035

RESUMO

Defects in vesicular trafficking underlie a wide variety of human diseases. Genetic disruption of leucine-rich repeat kinase 2 (LRRK2) in rodents results in epithelial vesicular trafficking errors that can also be induced by treatment of animals with LRRK2 kinase inhibitors. Here we demonstrate that defects in human renal cells lacking LRRK2 phenocopy those seen in the kidneys of Lrrk2 knockout mice, characterized by accumulation of intracellular waste vesicles and fragmentation of the Golgi apparatus. This phenotype can be recapitulated by knockdown of N-ethylmaleimide-sensitive factor, which physically associates with LRRK2 in renal cells. Deficiency in either protein leads to a defect in trans-Golgi to lysosome protein trafficking, which compromises the capacity of lysosomes to degrade endocytic and autophagic cargo. In contrast, neither bulk endocytosis nor autophagic flux are impaired when LRRK2 is acutely knocked down in normal immortalized human kidney (HK2) cells. These data collectively suggest that the primary renal defect caused by LRRK2 deficiency is in protein trafficking between the Golgi apparatus and late endosome/lysosome, which leads to progressive impairments in lysosomal function.


Assuntos
Endocitose , Células Epiteliais/enzimologia , Complexo de Golgi/enzimologia , Túbulos Renais Proximais/enzimologia , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/deficiência , Lisossomos/enzimologia , Autofagia , Linhagem Celular , Proliferação de Células , Células Epiteliais/patologia , Técnicas de Silenciamento de Genes , Genótipo , Complexo de Golgi/patologia , Humanos , Túbulos Renais Proximais/patologia , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/genética , Lisossomos/patologia , Proteínas Sensíveis a N-Etilmaleimida/genética , Proteínas Sensíveis a N-Etilmaleimida/metabolismo , Fenótipo , Transporte Proteico , Proteólise
20.
Biochem Soc Trans ; 46(4): 779-788, 2018 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-29903935

RESUMO

Changes in mucin-type O-linked glycosylation are seen in over 90% of breast cancers where increased sialylation is often observed and a change from branched glycans to linear glycans is often seen. There are many mechanisms involved including increased/altered expression of glycosyltransferases and relocalisation to the endoplasmic reticulum of the enzymes responsible for the addition of the first sugar, N-acetyl-d-galactosamine. It is now becoming clear that these changes can contribute to tumour growth and progression by modulating the micro-environment through glycan-sensing lectins expressed on immune cells, by modulating interactions with tumour surface receptors and by binding to selectins. The understanding of how changes in mucin-type O-linked glycosylation influence tumour growth and progression reveals new potential targets for therapeutic intervention in the treatment of breast cancer.


Assuntos
Neoplasias da Mama/metabolismo , Mucina-1/metabolismo , Acetilgalactosamina/metabolismo , Transporte Biológico , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Progressão da Doença , Feminino , Regulação Enzimológica da Expressão Gênica , Glicosídeo Hidrolases/genética , Glicosídeo Hidrolases/metabolismo , Glicosilação , Glicosiltransferases/genética , Glicosiltransferases/metabolismo , Complexo de Golgi/enzimologia , Complexo de Golgi/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Metástase Neoplásica , Microambiente Tumoral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA