Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
1.
J Biol Chem ; 298(12): 102685, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36370845

RESUMO

Brain-specific angiogenesis inhibitor 1 (BAI1; also called ADGRB1 or B1) is an adhesion G protein-coupled receptor known from studies on macrophages to bind to phosphatidylserine (PS) on apoptotic cells via its N-terminal thrombospondin repeats. A separate body of work has shown that B1 regulates postsynaptic function and dendritic spine morphology via signaling pathways involving Rac and Rho. However, it is unknown if PS binding by B1 has any effect on the receptor's signaling activity. To shed light on this subject, we studied G protein-dependent signaling by B1 in the absence and presence of coexpression with the PS flippase ATP11A in human embryonic kidney 293T cells. ATP11A expression reduced the amount of PS exposed extracellularly and also strikingly reduced the signaling activity of coexpressed full-length B1 but not a truncated version of the receptor lacking the thrombospondin repeats. Further experiments with an inactive mutant of ATP11A showed that the PS flippase function of ATP11A was required for modulation of B1 signaling. In coimmunoprecipitation experiments, we made the surprising finding that ATP11A not only modulates B1 signaling but also forms complexes with B1. Parallel studies in which PS in the outer leaflet was reduced by an independent method, deletion of the gene encoding the endogenous lipid scramblase anoctamin 6 (ANO6), revealed that this manipulation also markedly reduced B1 signaling. These findings demonstrate that B1 signaling is modulated by PS exposure and suggest a model in which B1 serves as a PS sensor at synapses and in other cellular contexts.


Assuntos
Fosfatidilserinas , Transdução de Sinais , Humanos , Fosfatidilserinas/genética , Fosfatidilserinas/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/genética , Trombospondinas/metabolismo , Células HEK293
2.
Biochim Biophys Acta Biomembr ; 1864(1): 183810, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34699769

RESUMO

Annexin A2 (AnxA2) is a calcium- and phospholipid-binding protein that plays roles in cellular processes involving membrane and cytoskeleton dynamics and is able to associate to several partner proteins. However, the principal molecular partners of AnxA2 are negatively charged phospholipids such as phosphatidylserine and phosphatidyl-inositol-(4,5)-phosphate. Herein we have studied different aspects of membrane lipid rearrangements induced by AnxA2 membrane binding. X-ray diffraction data revealed that AnxA2 has the property to stabilize lamellar structures and to block the formation of highly curved lipid phases (inverted hexagonal phase, HII). By using pyrene-labelled cholesterol and the environmental probe di-4-ANEPPDHQ, we observed that in model membranes, AnxA2 is able to modify both, cholesterol distribution and lipid compaction. In epithelial cells, we observed that AnxA2 localizes to membranes of different lipid order. The protein binding to membranes resulted in both, increases and/or decreases in membrane order depending on the cellular membrane regions. Overall, AnxA2 showed the capacity to modulate plasma membrane properties by inducing lipid redistribution that may lead to an increase in order or disorder of the membranes.


Assuntos
Anexina A2/genética , Membrana Celular/genética , Lipídeos de Membrana/química , Fosfolipídeos/genética , Anexina A2/química , Fenômenos Biofísicos , Cálcio/química , Proteínas de Transporte/química , Proteínas de Transporte/genética , Comunicação Celular/genética , Membrana Celular/química , Citoesqueleto/química , Citoesqueleto/genética , Humanos , Fosfatidilserinas/química , Fosfatidilserinas/genética , Fosfolipídeos/química
3.
Int J Mol Sci ; 22(22)2021 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-34830477

RESUMO

Singapore grouper iridovirus (SGIV), belonging to genus Ranavirus, family Iridoviridae, causes great economic losses in the aquaculture industry. Previous studies demonstrated the lipid composition of intracellular unenveloped viruses, but the changes in host-cell glyceophospholipids components and the roles of key enzymes during SGIV infection still remain largely unknown. Here, the whole cell lipidomic profiling during SGIV infection was analyzed using UPLC-Q-TOF-MS/MS. The lipidomic data showed that glycerophospholipids (GPs), including phosphatidylcholine (PC), phosphatidylserine (PS), glycerophosphoinositols (PI) and fatty acids (FAs) were significantly elevated in SGIV-infected cells, indicating that SGIV infection disturbed GPs homeostasis, and then affected the metabolism of FAs, especially arachidonic acid (AA). The roles of key enzymes, such as cytosolic phospholipase A2 (cPLA2), 5-Lipoxygenase (5-LOX), and cyclooxygenase (COX) in SGIV infection were further investigated using the corresponding specific inhibitors. The inhibition of cPLA2 by AACOCF3 decreased SGIV replication, suggesting that cPLA2 might play important roles in the process of SGIV infection. Consistent with this result, the ectopic expression of EccPLA2α or knockdown significantly enhanced or suppressed viral replication in vitro, respectively. In addition, the inhibition of both 5-LOX and COX significantly suppressed SGIV replication, indicating that AA metabolism was essential for SGIV infection. Taken together, our results demonstrated for the first time that SGIV infection in vitro disturbed GPs homeostasis and cPLA2 exerted crucial roles in SGIV replication.


Assuntos
Peixes/virologia , Iridovirus/genética , Fosfolipases A2 Citosólicas/genética , Replicação Viral/genética , Animais , Aquicultura , Araquidonato 5-Lipoxigenase/genética , Peixes/genética , Glicerofosfolipídeos/genética , Iridovirus/patogenicidade , Fosfatidilcolinas/genética , Fosfatidilserinas/genética , Singapura
4.
J Biol Chem ; 296: 100232, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33372034

RESUMO

Myosin VI ensembles on endocytic cargo facilitate directed transport through a dense cortical actin network. Myosin VI is recruited to clathrin-coated endosomes via the cargo adaptor Dab2. Canonically, it has been assumed that the interactions between a motor and its cargo adaptor are stable. However, it has been demonstrated that the force generated by multiple stably attached motors disrupts local cytoskeletal architecture, potentially compromising transport. In this study, we demonstrate that dynamic multimerization of myosin VI-Dab2 complexes facilitates cargo processivity without significant reorganization of cortical actin networks. Specifically, we find that Dab2 myosin interacting region (MIR) binds myosin VI with a moderate affinity (184 nM) and single-molecule kinetic measurements demonstrate a high rate of turnover (1 s-1) of the Dab2 MIR-myosin VI interaction. Single-molecule motility shows that saturating Dab2-MIR concentration (2 µM) promotes myosin VI homodimerization and processivity with run lengths comparable with constitutive myosin VI dimers. Cargo-mimetic DNA origami scaffolds patterned with Dab2 MIR-myosin VI complexes are weakly processive, displaying sparse motility on single actin filaments and "stop-and-go" motion on a cellular actin network. On a minimal actin cortex assembled on lipid bilayers, unregulated processive movement by either constitutive myosin V or VI dimers results in actin remodeling and foci formation. In contrast, Dab2 MIR-myosin VI interactions preserve the integrity of a minimal cortical actin network. Taken together, our study demonstrates the importance of dynamic motor-cargo association in enabling cargo transportation without disrupting cytoskeletal organization.


Assuntos
Citoesqueleto de Actina/genética , Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Reguladoras de Apoptose/química , Complexos Multiproteicos/química , Cadeias Pesadas de Miosina/química , Citoesqueleto de Actina/química , Citoesqueleto de Actina/ultraestrutura , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/ultraestrutura , Vesículas Revestidas por Clatrina/química , Vesículas Revestidas por Clatrina/genética , Citoesqueleto/química , Citoesqueleto/genética , Citoesqueleto/ultraestrutura , Endocitose/genética , Endossomos/genética , Humanos , Cinética , Complexos Multiproteicos/genética , Complexos Multiproteicos/ultraestrutura , Cadeias Pesadas de Miosina/genética , Cadeias Pesadas de Miosina/ultraestrutura , Fosfatidilserinas/genética , Ligação Proteica/genética , Multimerização Proteica/genética , Imagem Individual de Molécula
5.
Cell Commun Signal ; 18(1): 190, 2020 12 27.
Artigo em Inglês | MEDLINE | ID: mdl-33357215

RESUMO

The rapid ability of SARS-CoV-2 to spread among humans, along with the clinical complications of coronavirus disease 2019-COVID-19, have represented a significant challenge to the health management systems worldwide. The acute inflammation and coagulation abnormalities appear as the main causes for thousands of deaths worldwide. The intense inflammatory response could be involved with the formation of thrombi. For instance, the presence of uncleaved large multimers of von Willebrand (vWF), due to low ADAMTS13 activity in plasma could be explained by the inhibitory action of pro-inflammatory molecules such as IL-1ß and C reactive protein. In addition, the damage to endothelial cells after viral infection and/or activation of endothelium by pro-inflammatory cytokines, such as IL-1ß, IL-6, IFN-γ, IL-8, and TNF-α induces platelets and monocyte aggregation in the vascular wall and expression of tissue factor (TF). The TF expression may culminate in the formation of thrombi, and activation of cascade by the extrinsic pathway by association with factor VII. In this scenario, the phosphatidylserine-PtdSer exposure on the outer leaflet of the cell membrane as consequence of viral infection emerges as another possible underlying mechanism to acute immune inflammatory response and activation of coagulation cascade. The PtdSer exposure may be an important mechanism related to ADAM17-mediated ACE2, TNF-α, EGFR and IL-6R shedding, and the activation of TF on the surface of infected endothelial cells. In this review, we address the underlying mechanisms involved in the pathophysiology of inflammation and coagulation abnormalities. Moreover, we introduce key biochemical and pathophysiological concepts that support the possible participation of PtdSer exposure on the outer side of the SARS-CoV-2 infected cells membrane, in the pathophysiology of COVID-19. Video Abstract.


Assuntos
COVID-19/genética , Inflamação/genética , Fosfatidilserinas/genética , Trombose/genética , Proteína ADAM17/genética , Proteína ADAMTS13/genética , COVID-19/complicações , COVID-19/patologia , COVID-19/virologia , Células Endoteliais/virologia , Humanos , Inflamação/complicações , Inflamação/virologia , Fosfatidilserinas/metabolismo , Receptores de Interleucina-6/genética , SARS-CoV-2/patogenicidade , Trombose/patologia , Trombose/virologia , Fator de von Willebrand/genética
6.
PLoS Genet ; 16(10): e1009070, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33064773

RESUMO

The major glycerophospholipid phosphatidylethanolamine (PE) in the nervous system is essential for neural development and function. There are two major PE synthesis pathways, the CDP-ethanolamine pathway in the endoplasmic reticulum (ER) and the phosphatidylserine decarboxylase (PSD) pathway in mitochondria. However, the role played by mitochondrial PE synthesis in maintaining cellular PE homeostasis is unknown. Here, we show that Drosophila pect (phosphoethanolamine cytidylyltransferase) mutants lacking the CDP-ethanolamine pathway, exhibited alterations in phospholipid composition, defective phototransduction, and retinal degeneration. Induction of the PSD pathway fully restored levels and composition of cellular PE, thus rescued the retinal degeneration and defective visual responses in pect mutants. Disrupting lipid exchange between mitochondria and ER blocked the ability of PSD to rescue pect mutant phenotypes. These findings provide direct evidence that the synthesis of PE in mitochondria contributes to cellular PE homeostasis, and suggest the induction of mitochondrial PE synthesis as a promising therapeutic approach for disorders associated with PE deficiency.


Assuntos
Carboxiliases/genética , Cistina Difosfato/análogos & derivados , Retículo Endoplasmático/genética , Degeneração Retiniana/genética , Animais , Carboxiliases/metabolismo , Cistina Difosfato/deficiência , Cistina Difosfato/genética , Cistina Difosfato/metabolismo , Modelos Animais de Doenças , Drosophila melanogaster/genética , Retículo Endoplasmático/metabolismo , Etanolaminas/metabolismo , Homeostase/genética , Humanos , Metabolismo dos Lipídeos/genética , Mitocôndrias/genética , Mitocôndrias/metabolismo , Fosfatidilserinas/genética , Fosfatidilserinas/metabolismo , Degeneração Retiniana/metabolismo , Degeneração Retiniana/patologia , Transdução de Sinais/genética
7.
EMBO J ; 39(16): e105380, 2020 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-32657463

RESUMO

Neuronal circuit assembly requires the fine balance between synapse formation and elimination. Microglia, through the elimination of supernumerary synapses, have an established role in this process. While the microglial receptor TREM2 and the soluble complement proteins C1q and C3 are recognized as key players, the neuronal molecular components that specify synapses to be eliminated are still undefined. Here, we show that exposed phosphatidylserine (PS) represents a neuronal "eat-me" signal involved in microglial-mediated pruning. In hippocampal neuron and microglia co-cultures, synapse elimination can be partially prevented by blocking accessibility of exposed PS using Annexin V or through microglial loss of TREM2. In vivo, PS exposure at both hippocampal and retinogeniculate synapses and engulfment of PS-labeled material by microglia occurs during established developmental periods of microglial-mediated synapse elimination. Mice deficient in C1q, which fail to properly refine retinogeniculate connections, have elevated presynaptic PS exposure and reduced PS engulfment by microglia. These data provide mechanistic insight into microglial-mediated synapse pruning and identify a novel role of developmentally regulated neuronal PS exposure that is common among developing brain structures.


Assuntos
Hipocampo/metabolismo , Microglia/metabolismo , Neurônios/metabolismo , Fosfatidilserinas/metabolismo , Sinapses/metabolismo , Animais , Técnicas de Cocultura , Complemento C1q/genética , Complemento C1q/metabolismo , Complemento C3/genética , Complemento C3/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Fosfatidilserinas/genética , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo , Sinapses/genética
8.
EMBO J ; 39(16): e104136, 2020 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-32452062

RESUMO

Developmental synaptic remodeling is important for the formation of precise neural circuitry, and its disruption has been linked to neurodevelopmental disorders such as autism and schizophrenia. Microglia prune synapses, but integration of this synapse pruning with overlapping and concurrent neurodevelopmental processes, remains elusive. Adhesion G protein-coupled receptor ADGRG1/GPR56 controls multiple aspects of brain development in a cell type-specific manner: In neural progenitor cells, GPR56 regulates cortical lamination, whereas in oligodendrocyte progenitor cells, GPR56 controls developmental myelination and myelin repair. Here, we show that microglial GPR56 maintains appropriate synaptic numbers in several brain regions in a time- and circuit-dependent fashion. Phosphatidylserine (PS) on presynaptic elements binds GPR56 in a domain-specific manner, and microglia-specific deletion of Gpr56 leads to increased synapses as a result of reduced microglial engulfment of PS+ presynaptic inputs. Remarkably, a particular alternatively spliced isoform of GPR56 is selectively required for microglia-mediated synaptic pruning. Our present data provide a ligand- and isoform-specific mechanism underlying microglial GPR56-mediated synapse pruning in the context of complex neurodevelopmental processes.


Assuntos
Processamento Alternativo , Microglia/metabolismo , Fosfatidilserinas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Sinapses/metabolismo , Animais , Camundongos , Camundongos Transgênicos , Microglia/citologia , Fosfatidilserinas/genética , Ligação Proteica , Isoformas de Proteínas , Receptores Acoplados a Proteínas G/genética , Sinapses/genética
9.
J Cell Biol ; 219(5)2020 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-32303746

RESUMO

The yeast phosphatidylserine (PtdSer) decarboxylase Psd2 is proposed to engage in a membrane contact site (MCS) for PtdSer decarboxylation to phosphatidylethanolamine (PtdEtn). This proposed MCS harbors Psd2, the Sec14-like phosphatidylinositol transfer protein (PITP) Sfh4, the Stt4 phosphatidylinositol (PtdIns) 4-OH kinase, the Scs2 tether, and an uncharacterized protein. We report that, of these components, only Sfh4 and Stt4 regulate Psd2 activity in vivo. They do so via distinct mechanisms. Sfh4 operates via a mechanism for which its PtdIns-transfer activity is dispensable but requires an Sfh4-Psd2 physical interaction. The other requires Stt4-mediated production of PtdIns-4-phosphate (PtdIns4P), where Stt4 (along with the Sac1 PtdIns4P phosphatase and endoplasmic reticulum-plasma membrane tethers) indirectly modulate Psd2 activity via a PtdIns4P homeostatic mechanism that influences PtdSer accessibility to Psd2. These results identify an example in which the biological function of a Sec14-like PITP is cleanly uncoupled from its canonical in vitro PtdIns-transfer activity and challenge popular functional assumptions regarding lipid-transfer protein involvements in MCS function.


Assuntos
Proteínas de Membrana/genética , Fosfatidilserinas/genética , Proteínas de Transferência de Fosfolipídeos/genética , Proteínas de Saccharomyces cerevisiae/genética , 1-Fosfatidilinositol 4-Quinase/genética , Transporte Biológico/genética , Metabolismo dos Lipídeos/genética , Fosfatidiletanolaminas/genética , Fosfatidiletanolaminas/metabolismo , Fosfatidilserinas/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo
10.
Biochim Biophys Acta Gen Subj ; 1864(1): 129422, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31491457

RESUMO

BACKGROUND: Previous studies suggested that fibrillar human IAPP (hIAPP) is more likely to deposit in ß-cells, resulting in ß-cell injury. However, the changes in the conformation of hIAPP in lipid environment and the mechanism involved in ß-cell damage are unclear. METHODS: Synthetic hIAPP was incubated with five types of free fatty acids and phospholipids 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC) and 1-palmitoyl-2-oleoyl-sn-glycero-3-phospho-l-serine (POPS), which constitute the cell membrane. Thioflavin-T fluorescence assay was conducted to analyze the degree of hIAPP fibrosis, and circular dichroism spectroscopy was performed to detect the ß-fold formation of hIAPP. Furthermore, INS-1 cells were infected with human IAPP delivered by a GV230-EGFP plasmid. The effects of endogenous hIAPP overexpression induced by sodium palmitate on the survival, endoplasmic reticulum (ER) stress, and apoptosis of INS-1 cells were evaluated. RESULTS: The five types of free fatty acids can accelerate the fibrosis of hIAPP. Sodium palmitate also maintained the stability of fibrillar hIAPP. POPS, not POPC, accelerated hIAPP fibrosis. Treatment of INS-1 cells with sodium palmitate increased the expression of hIAPP, activated ER stress and ER stress-dependent apoptosis signaling pathways, and increased the apoptotic rate. CONCLUSION: Free fatty acids and anionic phospholipid can promote ß-fold formation and fibrosis in hIAPP. High lipid induced the overexpression of hIAPP and aggravated ER stress and apoptosis in INS-1 cells, which caused ß-cell death in high lipid environment. GENERAL SIGNIFICANCE: Our study reveals free fatty acids and hIAPP synergistically implicated in endoplasmic reticulum stress and apoptosis of islet ß-cells.


Assuntos
Apoptose/genética , Fibrose/genética , Células Secretoras de Insulina/metabolismo , Polipeptídeo Amiloide das Ilhotas Pancreáticas/genética , Amiloide/genética , Amiloide/metabolismo , Membrana Celular/genética , Membrana Celular/metabolismo , Estresse do Retículo Endoplasmático/genética , Ácidos Graxos não Esterificados/genética , Ácidos Graxos não Esterificados/metabolismo , Fibrose/metabolismo , Fibrose/patologia , Regulação da Expressão Gênica/genética , Humanos , Células Secretoras de Insulina/patologia , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Polipeptídeo Amiloide das Ilhotas Pancreáticas/ultraestrutura , Metabolismo dos Lipídeos/genética , Lipídeos/genética , Ácido Palmítico/metabolismo , Fosfatidilcolinas/genética , Fosfatidilcolinas/metabolismo , Fosfatidilserinas/genética , Fosfatidilserinas/metabolismo , Conformação Proteica em Folha beta , Dobramento de Proteína
11.
Nat Commun ; 10(1): 4456, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31575859

RESUMO

Fertilization is essential for species survival. Although Izumo1 and Juno are critical for initial interaction between gametes, additional molecules necessary for sperm:egg fusion on both the sperm and the oocyte remain to be defined. Here, we show that phosphatidylserine (PtdSer) is exposed on the head region of viable and motile sperm, with PtdSer exposure progressively increasing during sperm transit through the epididymis. Functionally, masking phosphatidylserine on sperm via three different approaches inhibits fertilization. On the oocyte, phosphatidylserine recognition receptors BAI1, CD36, Tim-4, and Mer-TK contribute to fertilization. Further, oocytes lacking the cytoplasmic ELMO1, or functional disruption of RAC1 (both of which signal downstream of BAI1/BAI3), also affect sperm entry into oocytes. Intriguingly, mammalian sperm could fuse with skeletal myoblasts, requiring PtdSer on sperm and BAI1/3, ELMO2, RAC1 in myoblasts. Collectively, these data identify phosphatidylserine on viable sperm and PtdSer recognition receptors on oocytes as key players in sperm:egg fusion.


Assuntos
Oócitos/metabolismo , Fagócitos/metabolismo , Fosfatidilserinas/metabolismo , Interações Espermatozoide-Óvulo/fisiologia , Espermatozoides/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Angiogênicas/metabolismo , Animais , Antígenos CD36/metabolismo , Proteínas do Citoesqueleto/metabolismo , Epididimo , Feminino , Humanos , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Mioblastos Esqueléticos , Proteínas do Tecido Nervoso/metabolismo , Neuropeptídeos/metabolismo , Fosfatidilserinas/genética , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , c-Mer Tirosina Quinase/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo
12.
Int J Mol Sci ; 20(15)2019 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-31382643

RESUMO

Plasma membrane (PM) lipid composition and domain organization are modulated by polarized exocytosis. Conversely, targeting of secretory vesicles at specific domains in the PM is carried out by exocyst complexes, which contain EXO70 subunits that play a significant role in the final recognition of the target membrane. As we have shown previously, a mature Arabidopsis trichome contains a basal domain with a thin cell wall and an apical domain with a thick secondary cell wall, which is developed in an EXO70H4-dependent manner. These domains are separated by a cell wall structure named the Ortmannian ring. Using phospholipid markers, we demonstrate that there are two distinct PM domains corresponding to these cell wall domains. The apical domain is enriched in phosphatidic acid (PA) and phosphatidylserine, with an undetectable amount of phosphatidylinositol 4,5-bisphosphate (PIP2), whereas the basal domain is PIP2-rich. While the apical domain recruits EXO70H4, the basal domain recruits EXO70A1, which corresponds to the lipid-binding capacities of these two paralogs. Loss of EXO70H4 results in a loss of the Ortmannian ring border and decreased apical PA accumulation, which causes the PA and PIP2 domains to merge together. Using transmission electron microscopy, we describe these accumulations as a unique anatomical feature of the apical cell wall-radially distributed rod-shaped membranous pockets, where both EXO70H4 and lipid markers are immobilized.


Assuntos
Proteínas de Arabidopsis/genética , Arabidopsis/genética , Lipídeos de Membrana/genética , Proteínas de Transporte Vesicular/genética , Arabidopsis/química , Proteínas de Arabidopsis/química , Membrana Celular/química , Membrana Celular/genética , Exocitose/genética , Lipídeos de Membrana/metabolismo , Fosfatidilinositol 4,5-Difosfato/química , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfatidilserinas/química , Fosfatidilserinas/genética , Tricomas/química , Tricomas/genética , Proteínas de Transporte Vesicular/química
13.
PLoS One ; 14(4): e0215009, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30958856

RESUMO

Interorganelle phospholipid transfer is critical for eukaryotic membrane biogenesis. In the yeast Saccharomyces cerevisiae, phosphatidylserine (PS) synthesized by PS synthase, Pss1, in the endoplasmic reticulum (ER) is decarboxylated to phosphatidylethanolamine (PE) by PS decarboxylase, Psd1, in the ER and mitochondria or by Psd2 in the endosome, Golgi, and/or vacuole, but the mechanism of interorganelle PS transport remains to be elucidated. Here we report that Sfh1, a member of Sec14 family proteins of S. cerevisiae, possesses the ability to enhance PE production by Psd2. Overexpression of SFH1 in the strain defective in Psd1 restored its growth on non-fermentable carbon sources and increased the intracellular and mitochondrial PE levels. Sfh1 was found to bind various phospholipids, including PS, in vivo. Bacterially expressed and purified Sfh1 was suggested to have the ability to transport fluorescently labeled PS between liposomes by fluorescence dequenching assay in vitro. Biochemical subcellular fractionation suggested that a fraction of Sfh1 localizes to the endosome, Golgi, and/or vacuole. We propose a model that Sfh1 promotes PE production by Psd2 by transferring phospholipids between the ER and endosome.


Assuntos
Carboxiliases/deficiência , Proteínas de Ciclo Celular/biossíntese , Proteínas Cromossômicas não Histona/biossíntese , Mitocôndrias/metabolismo , Modelos Biológicos , Consumo de Oxigênio , Proteínas de Saccharomyces cerevisiae/biossíntese , Saccharomyces cerevisiae/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas Cromossômicas não Histona/genética , Retículo Endoplasmático/genética , Retículo Endoplasmático/metabolismo , Endossomos/genética , Endossomos/metabolismo , Complexo de Golgi/genética , Complexo de Golgi/metabolismo , Mitocôndrias/genética , Fosfatidiletanolaminas/metabolismo , Fosfatidilserinas/genética , Fosfatidilserinas/metabolismo , Proteínas de Transferência de Fosfolipídeos/genética , Proteínas de Transferência de Fosfolipídeos/metabolismo , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Vacúolos/genética , Vacúolos/metabolismo
14.
Biochem J ; 476(5): 783-794, 2019 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-30755463

RESUMO

Type IV P-type ATPases (P4 ATPases) are lipid flippases that catalyze phospholipid transport from the exoplasmic to the cytoplasmic leaflet of cellular membranes, but the mechanism by which they recognize and transport phospholipids through the lipid bilayer remains unknown. In the present study, we succeeded in purifying recombinant aminophospholipid ATPase 2 (ALA2), a member of the P4 ATPase subfamily in Arabidopsis thaliana, in complex with the ALA-interacting subunit 5 (ALIS5). The ATP hydrolytic activity of the ALA2-ALIS5 complex was stimulated in a highly specific manner by phosphatidylserine. Small changes in the stereochemistry or the functional groups of the phosphatidylserine head group affected enzymatic activity, whereas alteration in the length and composition of the acyl chains only had minor effects. Likewise, the enzymatic activity of the ALA2-ALIS5 complex was stimulated by both mono- and di-acyl phosphatidylserines. Taken together, the results identify the lipid head group as the key structural element for substrate recognition by the P4 ATPase.


Assuntos
Adenosina Trifosfatases/química , Proteínas de Arabidopsis/química , Arabidopsis/enzimologia , Fosfatidilserinas/química , Proteínas de Transferência de Fosfolipídeos/química , Adenosina Trifosfatases/genética , Arabidopsis/genética , Proteínas de Arabidopsis/genética , Fosfatidilserinas/genética , Proteínas de Transferência de Fosfolipídeos/genética , Domínios Proteicos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética
15.
J Biol Chem ; 294(7): 2329-2339, 2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30602568

RESUMO

Phospholipids are an integral part of the cellular membrane structure and can be produced by a de novo biosynthetic pathway and, alternatively, by the Kennedy pathway. Studies in several yeast species have shown that the phospholipid phosphatidylserine (PS) is synthesized from CDP-diacylglycerol and serine, a route that is different from its synthesis in mammalian cells, involving a base-exchange reaction from preexisting phospholipids. Fungal-specific PS synthesis has been shown to play an important role in fungal virulence and has been proposed as an attractive drug target. However, PS synthase, which catalyzes this reaction, has not been studied in the human fungal pathogen Cryptococcus neoformans Here, we identified and characterized the PS synthase homolog (Cn Cho1) in this fungus. Heterologous expression of Cn CHO1 in a Saccharomyces cerevisiae cho1Δ mutant rescued the mutant's growth defect in the absence of ethanolamine supplementation. Moreover, an Sc cho1Δ mutant expressing Cn CHO1 had PS synthase activity, confirming that the Cn CHO1 encodes PS synthase. We also found that PS synthase in C. neoformans is localized to the endoplasmic reticulum and that it is essential for mitochondrial function and cell viability. Of note, its deficiency could not be complemented by ethanolamine or choline supplementation for the synthesis of phosphatidylethanolamine (PE) or phosphatidylcholine (PC) via the Kennedy pathway. These findings improve our understanding of phospholipid synthesis in a pathogenic fungus and indicate that PS synthase may be a useful target for antifungal drugs.


Assuntos
Cryptococcus neoformans/metabolismo , Retículo Endoplasmático/metabolismo , Viabilidade Microbiana , Fosfatidilserinas/biossíntese , Animais , CDPdiacilglicerol-Serina O-Fosfatidiltransferase/genética , CDPdiacilglicerol-Serina O-Fosfatidiltransferase/metabolismo , Cryptococcus neoformans/genética , Diglicerídeos de Citidina Difosfato/genética , Diglicerídeos de Citidina Difosfato/metabolismo , Retículo Endoplasmático/genética , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Humanos , Fosfatidilserinas/genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo
16.
Lab Med ; 50(1): 47-53, 2019 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-30016461

RESUMO

OBJECTIVE: To determine the number and intensity of phosphatidylserine (PS) expression of the red blood cells (RBCs), fragmented RBCs, and RBC-derived microparticles (RMPs) in patients with ß-thalassemia/hemoglobin (Hb)E. METHODS: We used flow cytometry to determine the number and levels of PS expression. RESULTS: The number of PS-exposing RBCs was statistically significantly higher (P <.001) than that of PS-exposing fragmented RBCs or RMPs. In contrast, the intensity of PS expression was significantly higher (P <.001) in RMPs than in RBCs or fragmented RBCs. Our study showed a trend of association between RBC distribution width (RDW) and both the number of fragmented RBCs and RMPs and their intensity of PS expression. CONCLUSION: In ß-thalassemia/HbE, PS-exposing RBCs, fragmented RBCs, and RMPs all differed in their numbers and their intensity of PS expression. The effects of these differences among PS-exposing populations on the pathophysiology of the disease require further investigation.


Assuntos
Micropartículas Derivadas de Células/metabolismo , Eritrócitos/metabolismo , Talassemia beta/sangue , Adolescente , Adulto , Estudos de Casos e Controles , Micropartículas Derivadas de Células/patologia , Criança , Pré-Escolar , Eritrócitos/patologia , Feminino , Hemoglobina E/genética , Humanos , Lactente , Masculino , Fosfatidilserinas/genética , Fosfatidilserinas/metabolismo
17.
Sci Rep ; 8(1): 15022, 2018 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-30302009

RESUMO

During the asexual intra-erythrocytic cycle, Plasmodium (P.) falciparum exports parasitic proteins to the surface of infected red blood cells (iRBCs) facilitating its cytoadhesion to various endothelial host receptors. This adhesive behavior is a critical contributor towards disease manifestation. However, little is known about the influence of recurring elevated temperature - a common symptom of the malaria infection - on the adhesive properties of iRBCs to endothelial receptors. To address this, we performed dual-micropipette step-pressure technique between P. falciparum (strain FCR3CSA) iRBCs and Chinese Hamster Ovary cells expressing Chondroitin sulfate A (CHO-CSA) after transient iRBCs incubation at febrile temperatures which revealed increase in adhesion parameters. Furthermore, flow cytometry analysis revealed an increase in phosphatidylserine (PS) expression on the iRBC surface following exposure to febrile temperature. The adhesion between iRBCs and CHO-CSA cells was remarkably reduced in presence of soluble Annexin V, indicating the mediation of PS on the adhesion events. Our results suggest that elevated PS recruitment on iRBC under thermally stressed conditions contributes to the increased adhesive behavior of iRBCs CSA-binding phenotype to CHO-CSA.


Assuntos
Sulfatos de Condroitina/genética , Eritrócitos/metabolismo , Malária Falciparum/genética , Plasmodium falciparum/genética , Animais , Células CHO , Adesão Celular/genética , Sulfatos de Condroitina/biossíntese , Cricetulus , Eritrócitos/parasitologia , Febre/sangue , Febre/genética , Febre/parasitologia , Humanos , Malária Falciparum/sangue , Malária Falciparum/parasitologia , Fosfatidilserinas/biossíntese , Fosfatidilserinas/genética , Plasmodium falciparum/patogenicidade , Propriedades de Superfície , Temperatura
18.
Biomed Res Int ; 2017: 3620510, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28951871

RESUMO

Genetic modifications of the oncolytic vaccinia virus (VV) improve selective tumor cell infection and death, as well as activation of antitumor immunity. We have engineered a double recombinant VV, coding human GM-CSF, and apoptosis-inducing protein apoptin (VV-GMCSF-Apo) for comparing with the earlier constructed double recombinant VV-GMCSF-Lact, coding another apoptosis-inducing protein, lactaptin, which activated different cell death pathways than apoptin. We showed that both these recombinant VVs more considerably activated a set of critical apoptosis markers in infected cells than the recombinant VV coding GM-CSF alone (VV-GMCSF-dGF): these were phosphatidylserine externalization, caspase-3 and caspase-7 activation, DNA fragmentation, and upregulation of proapoptotic protein BAX. However, only VV-GMCSF-Lact efficiently decreased the mitochondrial membrane potential of infected cancer cells. Investigating immunogenic cell death markers in cancer cells infected with recombinant VVs, we demonstrated that all tested recombinant VVs were efficient in calreticulin and HSP70 externalization, decrease of cellular HMGB1, and ATP secretion. The comparison of antitumor activity against advanced MDA-MB-231 tumor revealed that both recombinants VV-GMCSF-Lact and VV-GMCSF-Apo efficiently delay tumor growth. Our results demonstrate that the composition of GM-CSF and apoptosis-inducing proteins in the VV genome is very efficient tool for specific killing of cancer cells and for activation of antitumor immunity.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Apoptose/genética , Transgenes/genética , Vírus Vaccinia/genética , Animais , Antineoplásicos , Biomarcadores Tumorais/genética , Caspase 3/genética , Caspase 7/genética , Morte Celular/genética , Linhagem Celular Tumoral , Chlorocebus aethiops , Fragmentação do DNA , Feminino , Vetores Genéticos/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Proteínas de Choque Térmico HSP70/genética , Humanos , Potencial da Membrana Mitocondrial/genética , Camundongos , Camundongos SCID , Neoplasias/genética , Vírus Oncolíticos/genética , Fosfatidilserinas/genética , Regulação para Cima/genética , Replicação Viral/genética , Proteína X Associada a bcl-2/genética
19.
Mol Cancer Res ; 15(12): 1656-1666, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28923840

RESUMO

Metastases are a major cause of cancer mortality. AXL, a receptor tyrosine kinase aberrantly expressed in many tumors, is a potent oncogenic driver of metastatic cell motility and has been identified as broadly relevant in cancer drug resistance. Despite its frequent association with changes in cancer phenotypes, the precise mechanism leading to AXL activation is incompletely understood. In addition to its ligand growth arrest specific-6 (Gas6), activation of AXL requires the lipid moiety phosphatidylserine (PS). Phosphatidylserine is only available to mediate AXL activation when it is externalized on cell membranes, an event that occurs during certain physiologic processes such as apoptosis. Here, it is reported that exposure of cancer cells to phosphatidylserine-containing vesicles, including synthetic liposomes and apoptotic bodies, contributes to enhanced migration of tumor cells via a PS-Gas6-AXL signaling axis. These findings suggest that anticancer treatments that induce fractional cell killing enhance the motility of surviving cells in AXL-expressing tumors, which may explain the widespread role of AXL in limiting therapeutic efficacy.Implications: This study demonstrates that motility behavior of AXL-expressing tumor cells can be elicited by Gas6-bearing apoptotic bodies generated from tumor treatment with therapeutics that produce killing of a portion of the tumor cells present but not all, hence generating potentially problematic invasive and metastatic behavior of the surviving tumor cells. Mol Cancer Res; 15(12); 1656-66. ©2017 AACR.


Assuntos
Resistencia a Medicamentos Antineoplásicos/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Neoplasias/genética , Proteínas Proto-Oncogênicas/genética , Receptores Proteína Tirosina Quinases/genética , Apoptose/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Vesículas Extracelulares/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Lipossomos/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Neoplasias/patologia , Fosfatidilserinas/genética , Fosfatidilserinas/metabolismo , Transdução de Sinais/genética , Receptor Tirosina Quinase Axl
20.
Biochim Biophys Acta Mol Cell Res ; 1864(11 Pt B): 2082-2087, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28624437

RESUMO

BACKGROUND: ADAM10 and ADAM17 are the best characterized members of the ADAM (A Disintegrin and Metalloproteinase) - family of transmembrane proteases. Both are involved diverse physiological and pathophysiological processes. ADAMs are known to be regulated by posttranslational mechanisms. However, emerging evidence indicates that the plasma membrane with its unique dynamic properties may additionally play an important role in controlling sheddase function. SCOPE OF REVIEW: Membrane events that could contribute to regulation of ADAM-function are summarized. MAJOR CONCLUSIONS: Surface expression of peptidolytic activity should be differentiated from ADAM-sheddase function since the latter additionally requires that the protease finds its substrate in the lipid bilayer. We propose that this is achieved through horizontal and vertical reorganization of membrane nanoarchitecture coordinately occurring at the sites of sheddase activation. Reshuffling of nanodomains thereby guides traffic of enzyme and substrate to each other. For ADAM17 phosphatidylserine exposure is required to then induce its shedding function. GENERAL SIGNIFICANCE: The novel concept that physicochemical properties of the lipid bilayer govern the action of ADAM-proteases may be extendable to other functional proteins that act at the cell surface. This article is part of a Special Issue entitled: Proteolysis as a Regulatory Event in Pathophysiology edited by Stefan Rose-John.


Assuntos
Proteína ADAM10/genética , Proteína ADAM17/genética , Secretases da Proteína Precursora do Amiloide/genética , Proteínas de Membrana/genética , Proteólise , Proteína ADAM10/metabolismo , Proteína ADAM17/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Membrana Celular/genética , Membrana Celular/metabolismo , Humanos , Proteínas de Membrana/metabolismo , Fosfatidilserinas/genética , Fosfatidilserinas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...