Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.140
Filtrar
1.
Front Immunol ; 13: 820131, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35251001

RESUMO

Coronavirus disease 2019 (COVID-19) is currently a worldwide emergency caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). In observational clinical studies, statins have been identified as beneficial to hospitalized patients with COVID-19. However, experimental evidence of underlying statins protection against SARS-CoV-2 remains elusive. Here we reported for the first-time experimental evidence of the protective effects of simvastatin treatment both in vitro and in vivo. We found that treatment with simvastatin significantly reduced the viral replication and lung damage in vivo, delaying SARS-CoV-2-associated physiopathology and mortality in the K18-hACE2-transgenic mice model. Moreover, simvastatin also downregulated the inflammation triggered by SARS-CoV-2 infection in pulmonary tissue and in human neutrophils, peripheral blood monocytes, and lung epithelial Calu-3 cells in vitro, showing its potential to modulate the inflammatory response both at the site of infection and systemically. Additionally, we also observed that simvastatin affected the course of SARS-CoV-2 infection through displacing ACE2 on cell membrane lipid rafts. In conclusion, our results show that simvastatin exhibits early protective effects on SARS-CoV-2 infection by inhibiting virus cell entry and inflammatory cytokine production, through mechanisms at least in part dependent on lipid rafts disruption.


Assuntos
Tratamento Farmacológico da COVID-19 , Regulação para Baixo/efeitos dos fármacos , Inflamação/tratamento farmacológico , Microdomínios da Membrana/efeitos dos fármacos , SARS-CoV-2/patogenicidade , Sinvastatina/farmacologia , Animais , COVID-19/virologia , Modelos Animais de Doenças , Humanos , Inflamação/virologia , Pulmão/virologia , Camundongos , Camundongos Transgênicos , Replicação Viral/efeitos dos fármacos
2.
Brain Res ; 1774: 147709, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34758347

RESUMO

The increase in Aß1-42 is a neurotoxic effect induced by aluminum which can lead to impairment of learning and memory, but its mechanism has yet to be fully elucidated. Studies have shown that APP palmitoylation is appears to be involved in the production process of Aß1-42. Here, we investigated whether APP palmitoylation is related to the increase in Aß caused by aluminum and its specific mechanism of action. In this study, APP palmitoylation was studied in the setting of aluminum-induced increases in Aß1-42 from two perspectives: whole animal experiments and in vitro cell experiments. First, the learning and memory of rats were impaired and the number of rat cortical neurons was decreased after staining with aluminum. Second, the expression of palmitoyl APP, APP in lipid rafts and palmitoyl acyltransferase zDHHC7 both in rat cerebral cortex and PC12 cells increased with the production of Aß1-42 induced by aluminum in a dose-dependent manner. Finally, the intervention with the palmitoylation inhibitors 2-BP and siRNA zDHHC7 in PC12 cells reduced levels of palmitoyl APP, the expression of APP in lipid rafts and the content of Aß1-42 induced by aluminum to a certain extent. Our results indicate that increased APP palmitoylation levels may be related to the increase in Aß1-42 caused by aluminum, and the mechanism may involve APP palmitoylation promoting the accumulation of APP protein on lipid rafts and the cleavage of APP by BACE1 in amyloidogenic pathway. The increase in expression of zDHHC7 may be one of the reasons for the increase in levels of APP palmitoylation caused by aluminum.


Assuntos
Alumínio/farmacologia , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Neurônios/efeitos dos fármacos , Fragmentos de Peptídeos/metabolismo , Acetiltransferases/metabolismo , Animais , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Aprendizagem/efeitos dos fármacos , Lipoilação/efeitos dos fármacos , Microdomínios da Membrana/efeitos dos fármacos , Microdomínios da Membrana/metabolismo , Memória/efeitos dos fármacos , Neurônios/metabolismo , Células PC12 , Ratos
3.
Cell Rep ; 37(13): 110160, 2021 12 28.
Artigo em Inglês | MEDLINE | ID: mdl-34965434

RESUMO

The lipid raft-resident protein, MAL2, has been implicated as contributing to the pathogenesis of several malignancies, including breast cancer, but the underlying mechanism for its effects on tumorigenesis is unknown. Here, we show that MAL2-mediated lipid raft formation leads to HER2 plasma membrane retention and enhanced HER2 signaling in breast cancer cells. We demonstrate physical interactions between HER2 and MAL2 in lipid rafts using proximity ligation assays. Super-resolution structured illumination microscopy imaging displays the structural organization of the HER2/Ezrin/NHERF1/PMCA2 protein complex. Formation of this protein complex maintains low intracellular calcium concentrations in the vicinity of the plasma membrane. HER2/MAL2 protein interactions in lipid rafts are enhanced in trastuzumab-resistant breast cancer cells. Our findings suggest that MAL2 is crucial for lipid raft formation, HER2 signaling, and HER2 membrane stability in breast cancer cells, suggesting MAL2 as a potential therapeutic target.


Assuntos
Neoplasias da Mama/patologia , Proteínas do Citoesqueleto/metabolismo , Resistencia a Medicamentos Antineoplásicos , Microdomínios da Membrana/metabolismo , Proteínas Proteolipídicas Associadas a Linfócitos e Mielina/metabolismo , Fosfoproteínas/metabolismo , ATPases Transportadoras de Cálcio da Membrana Plasmática/metabolismo , Receptor ErbB-2/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Antineoplásicos Imunológicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Proliferação de Células , Proteínas do Citoesqueleto/genética , Endocitose , Feminino , Humanos , Microdomínios da Membrana/efeitos dos fármacos , Proteínas Proteolipídicas Associadas a Linfócitos e Mielina/genética , Fosfoproteínas/genética , ATPases Transportadoras de Cálcio da Membrana Plasmática/genética , Receptor ErbB-2/genética , Trocadores de Sódio-Hidrogênio/genética , Trastuzumab/farmacologia , Células Tumorais Cultivadas
4.
Eur Rev Med Pharmacol Sci ; 25(1 Suppl): 90-100, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34890039

RESUMO

OBJECTIVE: The aim of the study was to show the effect that two naturally occurring compounds, a cyclodextrin and hydroxytyrosol, can have on the entry of SARS-CoV-2 into human cells. MATERIALS AND METHODS: The PubMed database was searched to retrieve studies published from 2000 to 2020, satisfying the inclusion criteria. The search keywords were: SARS-CoV, SARS-CoV-2, coronavirus, lipid raft, endocytosis, hydroxytyrosol, cyclodextrin. Modeling of alpha-cyclodextrin and hydroxytyrosol were done using UCSF Chimera 1.14. RESULTS: The search results indicated that cyclodextrins can reduce the efficiency of viral endocytosis and that hydroxytyrosol has antiviral properties. Bioinformatic docking studies showed that alpha-cyclodextrin and hydroxytyrosol, alone or in combination, interact with the viral spike protein and its host cell receptor ACE2, thereby potentially influencing the endocytosis process. CONCLUSIONS: Hydroxytyrosol and alpha-cyclodextrin can be useful against the spread of SARS-CoV-2.


Assuntos
Álcool Feniletílico/análogos & derivados , SARS-CoV-2/fisiologia , Internalização do Vírus/efeitos dos fármacos , alfa-Ciclodextrinas/farmacologia , Enzima de Conversão de Angiotensina 2/química , Enzima de Conversão de Angiotensina 2/metabolismo , Sítios de Ligação , COVID-19/patologia , COVID-19/prevenção & controle , COVID-19/virologia , Biologia Computacional/métodos , Humanos , Microdomínios da Membrana/efeitos dos fármacos , Microdomínios da Membrana/metabolismo , Microdomínios da Membrana/virologia , Simulação de Acoplamento Molecular , Álcool Feniletílico/química , Álcool Feniletílico/metabolismo , Álcool Feniletílico/farmacologia , Álcool Feniletílico/uso terapêutico , Ligação Proteica , SARS-CoV-2/isolamento & purificação , Glicoproteína da Espícula de Coronavírus/química , Glicoproteína da Espícula de Coronavírus/metabolismo , alfa-Ciclodextrinas/química , alfa-Ciclodextrinas/metabolismo , alfa-Ciclodextrinas/uso terapêutico
5.
Clin Transl Med ; 11(11): e552, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34841679

RESUMO

BACKGROUND: Lipid rafts (LRs), cholesterol-enriched microdomains on cell membranes, are increasingly viewed as signalling platforms governing critical facets of cancer progression. The phenotype of cancer stem-like cells (CSCs) presents significant hurdles for successful cancer treatment, and the expression of several CSC markers is associated with LR integrity. However, LR implications in CSCs remain unclear. METHODS: This study evaluated the biological and molecular functions of LRs in colorectal cancer (CRC) by using an LR-disrupting alkylphospholipid (APL) drug, miltefosine. The mechanistic role of miltefosine in CSC inhibition was examined through normal or tumour intestinal mouse organoid, human CRC cell, CRC xenograft and miltefosine treatment gene expression profile analyses. RESULTS: Miltefosine suppresses CSC populations and their self-renewal activities in CRC cells, a CSC-targeting effect leading to irreversible disruption of tumour-initiating potential in vivo. Mechanistically, miltefosine reduced the expression of a set of genes, leading to stem cell death. Among them, miltefosine transcriptionally inhibited checkpoint kinase 1 (CHEK1), indicating that LR integrity is essential for CHEK1 expression regulation. In isolated CD44high CSCs, we found that CSCs exhibited stronger therapy resistance than non-CSC counterparts by preventing cell death through CHEK1-mediated cell cycle checkpoints. However, inhibition of the LR/CHEK1 axis by miltefosine released cell cycle checkpoints, forcing CSCs to enter inappropriate mitosis with accumulated DNA damage and resulting in catastrophic cell death. CONCLUSION: Our findings underscore the therapeutic potential of LR-targeting APLs for CRC treatment that overcomes the therapy-resistant phenotype of CSCs, highlighting the importance of the LR/CHEK1 axis as a novel mechanism of APLs.


Assuntos
Neoplasias Colorretais/tratamento farmacológico , Microdomínios da Membrana/efeitos dos fármacos , Fosforilcolina/análogos & derivados , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Proliferação de Células/efeitos dos fármacos , Neoplasias Colorretais/fisiopatologia , Modelos Animais de Doenças , Camundongos , Fosforilcolina/farmacologia , Fosforilcolina/uso terapêutico
6.
Mol Pharmacol ; 100(5): 502-512, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34475108

RESUMO

The activity of local anesthetics (LAs) has been attributed to the inhibition of ion channels, causing anesthesia. However, there is a growing body of research showing that LAs act on a wide range of receptors and channel proteins far beyond simple analgesia. The current concept of ligand recognition may no longer explain the multitude of protein targets influenced by LAs. We hypothesize that LAs can cause anesthesia without directly binding to the receptor proteins just by changing the physical properties of the lipid bilayer surrounding these proteins and ion channels based on LAs' amphiphilicity. It is possible that LAs act in one of the following ways: They 1) dissolve raft-like membrane microdomains, 2) impede nerve impulse propagation by lowering the lipid phase transition temperature, or 3) modulate the lateral pressure profile of the lipid bilayer. This could also explain the numerous additional effects of LAs besides anesthesia. Furthermore, the concepts of membrane-mediated activity and binding to ion channels do not have to exclude each other. If we were to consider LA as the middle part of a continuum between unspecific membrane-mediated activity on one end and highly specific ligand binding on the other end, we could describe LA as the link between the unspecific action of general anesthetics and toxins with their highly specific receptor binding. This comprehensive membrane-mediated model offers a fresh perspective to clinical and pharmaceutical research and therapeutic applications of local anesthetics. SIGNIFICANCE STATEMENT: Local anesthetics, according to the World Health Organization, belong to the most important drugs available to mankind. Their rediscovery as therapeutics and not only anesthetics marks a milestone in global pain therapy. The membrane-mediated mechanism of action proposed in this review can explain their puzzling variety of target proteins and their thus far inexplicable therapeutic effects. The new concept presented here places LAs on a continuum of structures and molecular mechanisms in between small general anesthetics and the more complex molecular toxins.


Assuntos
Potenciais de Ação/fisiologia , Anestésicos Locais/metabolismo , Fenômenos Fisiológicos Celulares/fisiologia , Microdomínios da Membrana/metabolismo , Potenciais de Ação/efeitos dos fármacos , Anestésicos Locais/administração & dosagem , Anestésicos Locais/química , Animais , Sítios de Ligação/efeitos dos fármacos , Sítios de Ligação/fisiologia , Fenômenos Fisiológicos Celulares/efeitos dos fármacos , Humanos , Canais Iônicos/antagonistas & inibidores , Canais Iônicos/metabolismo , Bicamadas Lipídicas/metabolismo , Microdomínios da Membrana/efeitos dos fármacos , Estrutura Secundária de Proteína
7.
Toxins (Basel) ; 13(9)2021 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-34564674

RESUMO

Equinatoxin II (EqtII) and Fragaceatoxin C (FraC) are pore-forming toxins (PFTs) from the actinoporin family that have enhanced membrane affinity in the presence of sphingomyelin (SM) and phase coexistence in the membrane. However, little is known about the effect of these proteins on the nanoscopic properties of membrane domains. Here, we used combined confocal microscopy and force mapping by atomic force microscopy to study the effect of EqtII and FraC on the organization of phase-separated phosphatidylcholine/SM/cholesterol membranes. To this aim, we developed a fast, high-throughput processing tool to correlate structural and nano-mechanical information from force mapping. We found that both proteins changed the lipid domain shape. Strikingly, they induced a reduction in the domain area and circularity, suggesting a decrease in the line tension due to a lipid phase height mismatch, which correlated with proteins binding to the domain interfaces. Moreover, force mapping suggested that the proteins affected the mechanical properties at the edge, but not in the bulk, of the domains. This effect could not be revealed by ensemble force spectroscopy measurements supporting the suitability of force mapping to study local membrane topographical and mechanical alterations by membranotropic proteins.


Assuntos
Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Venenos de Cnidários/metabolismo , Venenos de Cnidários/toxicidade , Microdomínios da Membrana/metabolismo , Anêmonas-do-Mar/química , Anêmonas-do-Mar/metabolismo , Esfingomielinas/metabolismo , Animais , Microdomínios da Membrana/efeitos dos fármacos , Microscopia de Força Atômica , Microscopia Confocal
8.
Cell Rep Med ; 2(7): 100345, 2021 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-34337561

RESUMO

Hereditary sensory neuropathy type 1 (HSN1) is caused by mutations in the SPTLC1 or SPTLC2 sub-units of the enzyme serine palmitoyltransferase, resulting in the production of toxic 1-deoxysphingolipid bases (DSBs). We used induced pluripotent stem cells (iPSCs) from patients with HSN1 to determine whether endogenous DSBs are neurotoxic, patho-mechanisms of toxicity and response to therapy. HSN1 iPSC-derived sensory neurons (iPSCdSNs) endogenously produce neurotoxic DSBs. Complex gangliosides, which are essential for membrane micro-domains and signaling, are reduced, and neurotrophin signaling is impaired, resulting in reduced neurite outgrowth. In HSN1 myelinating cocultures, we find a major disruption of nodal complex proteins after 8 weeks, which leads to complete myelin breakdown after 6 months. HSN1 iPSC models have, therefore, revealed that SPTLC1 mutation alters lipid metabolism, impairs the formation of complex gangliosides, and reduces axon and myelin stability. Many of these changes are prevented by l-serine supplementation, supporting its use as a rational therapy.


Assuntos
Axônios/metabolismo , Gangliosídeos/metabolismo , Neuropatias Hereditárias Sensoriais e Autônomas/patologia , Células-Tronco Pluripotentes Induzidas/patologia , Modelos Biológicos , Neuroglia/metabolismo , Serina/farmacologia , Envelhecimento/patologia , Axônios/efeitos dos fármacos , Axônios/ultraestrutura , Sequência de Bases , Caspase 3/metabolismo , Linhagem Celular , Regulação da Expressão Gênica/efeitos dos fármacos , Neuropatias Hereditárias Sensoriais e Autônomas/genética , Humanos , Células-Tronco Pluripotentes Induzidas/ultraestrutura , Microdomínios da Membrana/efeitos dos fármacos , Microdomínios da Membrana/metabolismo , Microdomínios da Membrana/ultraestrutura , Bainha de Mielina/metabolismo , Fatores de Crescimento Neural/metabolismo , Neuroglia/efeitos dos fármacos , Crescimento Neuronal/efeitos dos fármacos , Proteína Nodal/metabolismo , Células Receptoras Sensoriais/efeitos dos fármacos , Células Receptoras Sensoriais/metabolismo , Células Receptoras Sensoriais/patologia , Células Receptoras Sensoriais/ultraestrutura , Transdução de Sinais/efeitos dos fármacos , Esfingolipídeos/metabolismo , Transcriptoma/genética
9.
Molecules ; 26(15)2021 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-34361779

RESUMO

Delivering nucleic acids into the endothelium has great potential in treating vascular diseases. However, endothelial cells, which line the vasculature, are considered as sensitive in nature and hard to transfect. Low transfection efficacies in endothelial cells limit their potential therapeutic applications. Towards improving the transfection efficiency, we made an effort to understand the internalization of lipoplexes into the cells, which is the first and most critical step in nucleic acid transfections. In this study, we demonstrated that the transient modulation of caveolae/lipid rafts mediated endocytosis with the cholesterol-sequestrating agents, nystatin, filipin III, and siRNA against Cav-1, which significantly increased the transfection properties of cationic lipid-(2-hydroxy-N-methyl-N,N-bis(2-tetradecanamidoethyl)ethanaminium chloride), namely, amide liposomes in combination with 1,2-Dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE) (AD Liposomes) in liver sinusoidal endothelial cells (SK-Hep1). In particular, nystatin was found to be highly effective with 2-3-fold enhanced transfection efficacy when compared with amide liposomes in combination with Cholesterol (AC), by switching lipoplex internalization predominantly through clathrin-mediated endocytosis and macropinocytosis.


Assuntos
Cavéolas/efeitos dos fármacos , Colesterol/química , Células Endoteliais/efeitos dos fármacos , Lipossomos/química , Microdomínios da Membrana/efeitos dos fármacos , Transfecção/métodos , Animais , Cavéolas/química , Cavéolas/metabolismo , Caveolina 1/antagonistas & inibidores , Caveolina 1/genética , Caveolina 1/metabolismo , Linhagem Celular Transformada , Colesterol/metabolismo , Clatrina/metabolismo , DNA/química , DNA/metabolismo , Endocitose/efeitos dos fármacos , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Filipina/química , Filipina/farmacologia , Expressão Gênica , Lipossomos/metabolismo , Microdomínios da Membrana/química , Microdomínios da Membrana/metabolismo , Nistatina/química , Nistatina/farmacologia , Fosfatidiletanolaminas/química , Fosfatidiletanolaminas/farmacologia , Pinocitose/efeitos dos fármacos , Plasmídeos/química , Plasmídeos/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos
10.
Mol Pharmacol ; 100(2): 66-81, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34011569

RESUMO

Termination of antidepressant therapy often has negative consequences. Although symptoms of antidepressant withdrawal are widely recognized, the molecular processes that underlie them are not well characterized. We show that certain aspects of Gα s signaling remain suppressed after antidepressant withdrawal, even after others have reverted to baseline. Antidepressant treatment causes translocation of Gα s protein from lipid rafts to nonraft membrane regions. This results in augmented Gα s signaling, including facilitated activation of adenylyl cyclase and increased cAMP accumulation. Using CC6 or SK-N-SH cells and a lipid raft-localized cAMP sensor, we show that Gα s signaling is reduced in lipid rafts, even while signaling is enhanced elsewhere in the cell. These signaling changes mirror the changes in Gα s localization observed after antidepressant treatment. Furthermore, we show that suppression of Gα s signaling in lipid rafts persists at least 24 hours after cessation of antidepressant treatment. Gα s localization was quantified after membrane isolation and sequential detergent extraction. We show that suppression of lipid raft Gα s signaling persists for an extended time period after antidepressant withdrawal, whereas increased nonraft membrane Gα s signaling reverts partially or fully upon cessation of antidepressant treatment. Translocation of Gα s out of lipid rafts is also persistent. These events may reflect cellular adaptations to antidepressant treatment that contribute to antidepressant discontinuation syndromes and may aid in the discovery of new treatments and strategies to mitigate the symptoms of depression and antidepressant withdrawal. SIGNIFICANCE STATEMENT: This work explores, for the first time, the effects of antidepressants on Gα s signaling after drug withdrawal. This provides novel insight into the cellular and molecular processes affected by antidepressant drugs and their persistence after discontinuation of treatment.


Assuntos
Antidepressivos/farmacologia , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Microdomínios da Membrana/metabolismo , Animais , Linhagem Celular , Regulação da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Humanos , Microdomínios da Membrana/efeitos dos fármacos , Ratos , Transdução de Sinais/efeitos dos fármacos
11.
Int J Mol Sci ; 22(6)2021 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-33799511

RESUMO

In uremic patients, high-density lipoprotein (HDL) loses its anti-inflammatory features and can even become pro-inflammatory due to an altered protein composition. In chronic kidney disease (CKD), impaired functions of polymorphonuclear leukocytes (PMNLs) contribute to inflammation and an increased risk of cardiovascular disease. This study investigated the effect of HDL from CKD and hemodialysis (HD) patients on the CD14 expression on PMNLs. HDL was isolated using a one-step density gradient centrifugation. Isolation of PMNLs was carried out by discontinuous Ficoll-Hypaque density gradient centrifugation. CD14 surface expression was quantified by flow cytometry. The activity of the small GTPase Rac1 was determined by means of an activation pull-down assay. HDL increased the CD14 surface expression on PMNLs. This effect was more pronounced for HDL isolated from uremic patients. The acute phase protein serum amyloid A (SAA) caused higher CD14 expression, while SAA as part of an HDL particle did not. Lipid raft disruption with methyl-ß-cyclodextrin led to a reduced CD14 expression in the absence and presence of HDL. HDL from healthy subjects but not from HD patients decreased the activity of Rac1. Considering the known anti-inflammatory effects of HDL, the finding that even HDL from healthy subjects increased the CD14 expression was unexpected. The pathophysiological relevance of this result needs further investigation.


Assuntos
Receptores de Lipopolissacarídeos/genética , Lipoproteínas HDL/farmacologia , Neutrófilos/efeitos dos fármacos , Insuficiência Renal Crônica/genética , Uremia/genética , Idoso , Estudos de Casos e Controles , Feminino , Humanos , Receptores de Lipopolissacarídeos/metabolismo , Lipoproteínas HDL/isolamento & purificação , Masculino , Microdomínios da Membrana/química , Microdomínios da Membrana/efeitos dos fármacos , Microdomínios da Membrana/metabolismo , Pessoa de Meia-Idade , Neutrófilos/metabolismo , Neutrófilos/patologia , Cultura Primária de Células , Diálise Renal , Insuficiência Renal Crônica/metabolismo , Insuficiência Renal Crônica/fisiopatologia , Insuficiência Renal Crônica/terapia , Proteína Amiloide A Sérica/genética , Proteína Amiloide A Sérica/metabolismo , Uremia/metabolismo , Uremia/fisiopatologia , Uremia/terapia , beta-Ciclodextrinas/farmacologia , Proteínas rac1 de Ligação ao GTP/genética , Proteínas rac1 de Ligação ao GTP/metabolismo
12.
Thromb Haemost ; 121(12): 1628-1636, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-33851387

RESUMO

BACKGROUND: Platelet cytosolic cyclic adenosine monophosphate (cAMP) levels are balanced by synthesis, degradation, and efflux. Efflux can occur via multidrug resistant protein-4 (MRP4; ABCC4) present on dense granule and/or plasma membranes. As lipid rafts have been shown to interfere on cAMP homeostasis, we evaluated the relationships between the distribution and activity of MRP4 in lipid rafts and cAMP efflux. METHODS: Platelet activation and cAMP homeostasis were analyzed in human and wild-type or MRP4-deleted mouse platelets in the presence of methyl-ß-cyclodextrin (MßCD) to disrupt lipid rafts, and of activators of the cAMP signalling pathways. Human platelet MRP4 and effector proteins of the cAMP pathway were analyzed by immunoblots in lipid rafts isolated by differential centrifugation. RESULTS: MßCD dose dependently inhibited human and mouse platelet aggregation without affecting per se cAMP levels. An additive inhibitory effect existed between the adenylate cyclase (AC) activator forskolin and MßCD that was accompanied by an overincrease of cAMP, and which was significantly enhanced upon MRP4 deletion. Finally, an efflux of cAMP out of resting platelets incubated with prostaglandin E1 (PGE1) was observed that was partly dependent on MRP4. Lipid rafts contained a small fraction (≈15%) of MRP4 and most of the inhibitory G-protein Gi, whereas Gs protein, AC3, and phosphodiesterases PDE2 and PDE3A were all present as only trace amounts. CONCLUSION: Our results are in favour of part of MRP4 present at the platelet surface, including in lipid rafts. Lipid raft integrity is necessary for cAMP signalling regulation, although MRP4 and most players of cAMP homeostasis are essentially located outside rafts.


Assuntos
Plaquetas/metabolismo , AMP Cíclico/sangue , Microdomínios da Membrana/metabolismo , Proteínas Associadas à Resistência a Múltiplos Medicamentos/sangue , Agregação Plaquetária , Sistemas do Segundo Mensageiro , Alprostadil/farmacologia , Animais , Plaquetas/efeitos dos fármacos , Microdomínios da Membrana/efeitos dos fármacos , Microdomínios da Membrana/genética , Camundongos Knockout , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Agregação Plaquetária/efeitos dos fármacos , beta-Ciclodextrinas/farmacologia
13.
Toxins (Basel) ; 13(2)2021 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-33673393

RESUMO

Human kidney epithelial cells are supposed to be directly involved in the pathogenesis of the hemolytic-uremic syndrome (HUS) caused by Shiga toxin (Stx)-producing enterohemorrhagic Escherichia coli (EHEC). The characterization of the major and minor Stx-binding glycosphingolipids (GSLs) globotriaosylceramide (Gb3Cer) and globotetraosylceramide (Gb4Cer), respectively, of primary human renal cortical epithelial cells (pHRCEpiCs) revealed GSLs with Cer (d18:1, C16:0), Cer (d18:1, C22:0), and Cer (d18:1, C24:1/C24:0) as the dominant lipoforms. Using detergent-resistant membranes (DRMs) and non-DRMs, Gb3Cer and Gb4Cer prevailed in the DRM fractions, suggesting their association with microdomains in the liquid-ordered membrane phase. A preference of Gb3Cer and Gb4Cer endowed with C24:0 fatty acid accompanied by minor monounsaturated C24:1-harboring counterparts was observed in DRMs, whereas the C24:1 fatty acid increased in relation to the saturated equivalents in non-DRMs. A shift of the dominant phospholipid phosphatidylcholine with saturated fatty acids in the DRM to unsaturated species in the non-DRM fractions correlated with the GSL distribution. Cytotoxicity assays gave a moderate susceptibility of pHRCEpiCs to the Stx1a and Stx2a subtypes when compared to highly sensitive Vero-B4 cells. The results indicate that presence of Stx-binding GSLs per se and preferred occurrence in microdomains do not necessarily lead to a high cellular susceptibility towards Stx.


Assuntos
Células Epiteliais/metabolismo , Globosídeos/metabolismo , Córtex Renal/metabolismo , Toxina Shiga I/toxicidade , Toxina Shiga II/toxicidade , Triexosilceramidas/metabolismo , Animais , Sobrevivência Celular/efeitos dos fármacos , Chlorocebus aethiops , Células Epiteliais/patologia , Infecções por Escherichia coli/microbiologia , Síndrome Hemolítico-Urêmica/microbiologia , Humanos , Córtex Renal/patologia , Microdomínios da Membrana/efeitos dos fármacos , Microdomínios da Membrana/metabolismo , Microdomínios da Membrana/patologia , Cultura Primária de Células , Ligação Proteica , Toxina Shiga I/metabolismo , Toxina Shiga II/metabolismo , Escherichia coli Shiga Toxigênica/metabolismo , Escherichia coli Shiga Toxigênica/patogenicidade , Células Vero
14.
Life Sci ; 273: 119300, 2021 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-33662433

RESUMO

AIMS: Plasma hyperlipidemia is a protective factor in amyotrophic lateral sclerosis (ALS) while cholesterol-lowering drugs aggravate the pathology. We hypothesize that this phenomenon can be linked with membrane lipid alterations in the neuromuscular junctions (NMJs) occurring before motor neuron loss. METHODS: Neurotransmitter release in parallel with lipid membrane properties in diaphragm NMJs of SOD1G93A (mSOD) mice at nine weeks of age (pre-onset stage) were assessed. KEY FINDINGS: Despite on slight changes in spontaneous and evoked quantum release of acetylcholine, extracellular levels of choline at resting conditions, an indicator of non-quantum release, were significantly increased in mSOD mice. The use of lipid-sensitive fluorescent probes points to lipid raft disruption in the NMJs of mSOD mice. However, content of cholesterol, a key raft component was unchanged implying another pathway responsible for the loss of raft integrity. In the mSOD mice we found marked increase in levels of raft-destabilizing lipid ceramide. This was accompanied by enhanced ability to uptake of exogenous ceramide in NMJs. Acute and chronic administration of 25-hydroxycholesterol, whose levels increase due to hypercholesterolemia, recovered early alterations in membrane properties. Furthermore, chronic treatment with 25-hydroxycholesterol prevented increase in ceramide and extracellular choline levels as well as suppressed lipid peroxidation of NMJ membranes and fragmentation of end plates. SIGNIFICANCE: Thus, lipid raft disruption likely due to ceramide accumulation could be early event in ALS which may trigger neuromuscular abnormalities. Cholesterol derivative 25-hydroxycholesterol may serve as a molecule restoring the membrane and functional properties of NMJs at the early stage.


Assuntos
Esclerose Amiotrófica Lateral/tratamento farmacológico , Modelos Animais de Doenças , Hidroxicolesteróis/farmacologia , Microdomínios da Membrana/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Superóxido Dismutase-1/fisiologia , Acetilcolina/metabolismo , Esclerose Amiotrófica Lateral/metabolismo , Esclerose Amiotrófica Lateral/patologia , Animais , Ceramidas/metabolismo , Colesterol/metabolismo , Feminino , Masculino , Microdomínios da Membrana/metabolismo , Microdomínios da Membrana/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Junção Neuromuscular , Transmissão Sináptica
15.
Immunopharmacol Immunotoxicol ; 43(2): 138-144, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33509007

RESUMO

OBJECTIVE: Polydatin has been used in the treatment of various inflammatory diseases. However, its role in the regulation of neuroinflammation has not been reported. In this study, we designed to investigate the anti-inflammatory effects of polydatin in LPS-stimulated BV2 microglia cells. METHODS: Inflammatory mediators TNF-α, IL-1ß, NO, and PGE2 production were measured by ELISA. The protein of signaling pathways were detected by western blot analysis. RESULTS: The results showed that polydatin significantly ameliorated the production of TNF-α, IL-1ß, NO, and PGE2 up-regulated by LPS. Polydatin also blocked LPS-induced NF-κB activation. In addition, PI3K and AKT, the up-stream molecules of NF-κB signaling pathway, were inhibited by the treatment of polydatin. Also, we found the formation of lipid rafts was inhibited by polydatin through attenuating the cholesterol content. Finally, polydatin was found to increase the expression of ABCA1 and ABCG1. CONCLUSION: In conclusion, the results of the present study suggested that polydatin exhibited its anti-inflammatory effects in BV2 cells through disrupting lipid rafts, which subsequently inhibiting PI3K/AKT signaling pathway.


Assuntos
Anti-Inflamatórios/farmacologia , Glucosídeos/farmacologia , Mediadores da Inflamação/antagonistas & inibidores , Lipopolissacarídeos/toxicidade , Microdomínios da Membrana/efeitos dos fármacos , Microglia/efeitos dos fármacos , Estilbenos/farmacologia , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Relação Dose-Resposta a Droga , Medicamentos de Ervas Chinesas/farmacologia , Mediadores da Inflamação/metabolismo , Microdomínios da Membrana/metabolismo , Camundongos , Microglia/metabolismo
16.
Int J Mol Sci ; 22(2)2021 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-33466887

RESUMO

The binding of vascular endothelial growth factor A (VEGF) to VEGF receptor-2 (VEGFR-2) stimulates angiogenic signaling. Lipid rafts are cholesterol-dense regions of the plasma membrane that serve as an organizational platform for biomolecules. Although VEGFR2 has been shown to colocalize with lipid rafts to regulate its activation, the effect of lipid rafts on non-activated VEGFR2 has not been explored. Here, we characterized the involvement of lipid rafts in modulating the stability of non-activated VEGFR2 in endothelial cells using raft disrupting agents: methyl-ß-cyclodextrin, sphingomyelinase and simvastatin. Disrupting lipid rafts selectively decreased the levels of non-activated VEGFR2 as a result of increased lysosomal degradation. The decreased expression of VEGFR2 translated to reduced VEGF-activation of the extracellular signal-regulated protein kinases (ERK). Overall, our results indicate that lipid rafts stabilize VEGFR2 and its associated signal transduction activities required for angiogenesis. Thus, modulation of lipid rafts may provide a means to regulate the sensitivity of endothelial cells to VEGF stimulation. Indeed, the ability of simvastatin to down regulate VEGFR2 and inhibit VEGF activity suggest a potential mechanism underlying the observation that this drug improves outcomes in the treatment of certain cancers.


Assuntos
Células Endoteliais/metabolismo , Microdomínios da Membrana/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Anticolesterolemiantes/farmacologia , Vasos Sanguíneos/efeitos dos fármacos , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/fisiologia , Bovinos , Células Cultivadas , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Microdomínios da Membrana/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Estabilidade Proteica/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Sinvastatina/farmacologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
17.
Int J Biol Macromol ; 171: 343-357, 2021 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-33421469

RESUMO

Lactoferrin (Lf) is a bioactive milk-derived protein with remarkable wide-spectrum antifungal activity. To deepen our understanding of the molecular mechanisms underlying Lf cytotoxicity, the role of plasma membrane ergosterol- and sphingolipid-rich lipid rafts and their association with the proton pump Pma1p was explored. Pma1p was previously identified as a Lf-binding protein. Results showed that bovine Lf (bLf) perturbs ergosterol-rich lipid rafts organization by inducing intracellular accumulation of ergosterol. Using yeast mutant strains lacking lipid rafts-associated proteins or enzymes involved in the synthesis of ergosterol and sphingolipids, we found that perturbations in the composition of these membrane domains increase resistance to bLf-induced yeast cell death. Also, when Pma1p-lipid rafts association is compromised in the Pma1-10 mutant and in the absence of the Pma1p-binding protein Ast1p, the bLf killing activity is impaired. Altogether, results showed that the perturbation of lipid rafts and the inhibition of both Pma1p and V-ATPase activities mediate the antifungal activity of bLf. Since it is suggested that the combination of conventional antifungals with lipid rafts-disrupting compounds is a powerful antifungal approach, our data will help to pave the way for the use of bLf alone or in combination for the treatment/eradication of clinically and agronomically relevant yeast pathogens/fungi.


Assuntos
Antifúngicos/farmacologia , Lactoferrina/farmacologia , Microdomínios da Membrana/efeitos dos fármacos , ATPases Translocadoras de Prótons/fisiologia , Proteínas de Saccharomyces cerevisiae/fisiologia , Saccharomyces cerevisiae/efeitos dos fármacos , Trifosfato de Adenosina/metabolismo , Farmacorresistência Fúngica , Ergosterol/metabolismo , Filipina , Proteínas de Fluorescência Verde/análise , Microdomínios da Membrana/química , Mutação Puntual , ATPases Translocadoras de Prótons/biossíntese , ATPases Translocadoras de Prótons/genética , Proteínas Recombinantes de Fusão/análise , Proteínas Recombinantes de Fusão/biossíntese , Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/ultraestrutura , Proteínas de Saccharomyces cerevisiae/biossíntese , Proteínas de Saccharomyces cerevisiae/genética , Vacúolos/efeitos dos fármacos , Vacúolos/enzimologia , beta-Ciclodextrinas/farmacologia
18.
Int J Mol Sci ; 22(2)2021 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-33450869

RESUMO

Lipid rafts are critical cell membrane lipid platforms enriched in sphingolipid and cholesterol content involved in diverse cellular processes. They have been proposed to influence membrane properties and to accommodate receptors within themselves by facilitating their interaction with ligands. Over the past decade, technical advances have improved our understanding of lipid rafts as bioactive structures. In this review, we will cover the more recent findings about cholesterol, sphingolipids and lipid rafts located in cellular and nuclear membranes in cancer. Collectively, the data provide insights on the role of lipid rafts as biomolecular targets in cancer with good perspectives for the development of innovative therapeutic strategies.


Assuntos
Antineoplásicos/farmacologia , Colesterol/metabolismo , Microdomínios da Membrana/efeitos dos fármacos , Microdomínios da Membrana/metabolismo , Neoplasias/metabolismo , Esfingolipídeos/metabolismo , Animais , Antineoplásicos/uso terapêutico , Biomarcadores Tumorais , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Humanos , Terapia de Alvo Molecular , Neoplasias/tratamento farmacológico , Neoplasias/etiologia , Membrana Nuclear/metabolismo
19.
Neurobiol Dis ; 150: 105259, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33434618

RESUMO

Neuronal regeneration in the injured central nervous system is hampered by multiple extracellular proteins. These proteins exert their inhibitory action through interactions with receptors that are located in cholesterol rich compartments of the membrane termed lipid rafts. Here we show that cholesterol-synthesis inhibition prevents the association of the Neogenin receptor with lipid rafts. Furthermore, we show that cholesterol-synthesis inhibition enhances axonal growth both on inhibitory -myelin and -RGMa substrates. Following optic nerve injury, lowering cholesterol synthesis with both drugs and siRNA-strategies allows for robust axonal regeneration and promotes neuronal survival. Cholesterol inhibition also enhanced photoreceptor survival in a model of Retinitis Pigmentosa. Our data reveal that Lovastatin leads to several opposing effects on regenerating axons: cholesterol synthesis inhibition promotes regeneration whereas altered prenylation impairs regeneration. We also show that the lactone prodrug form of lovastatin has differing effects on regeneration when compared to the ring-open hydroxy-acid form. Thus the association of cell surface receptors with lipid rafts contributes to axonal regeneration inhibition, and blocking cholesterol synthesis provides a potential therapeutic approach to promote neuronal regeneration and survival in the diseased Central Nervous System. SIGNIFICANCE STATEMENT: Statins have been intensively used to treat high levels of cholesterol in humans. However, the effect of cholesterol inhibition in both the healthy and the diseased brain remains controversial. In particular, it is unclear whether cholesterol inhibition with statins can promote regeneration and survival following injuries. Here we show that late stage cholesterol inhibition promotes robust axonal regeneration following optic nerve injury. We identified distinct mechanisms of action for activated vs non-activated Lovastatin that may account for discrepancies found in the literature. We show that late stage cholesterol synthesis inhibition alters Neogenin association with lipid rafts, thereby i) neutralizing the inhibitory function of its ligand and ii) offering a novel opportunity to promote CNS regeneration and survival following injuries.


Assuntos
Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Lovastatina/farmacologia , Regeneração Nervosa/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Nervo Óptico/efeitos dos fármacos , Animais , Anticolesterolemiantes/farmacologia , Axônios/efeitos dos fármacos , Axônios/patologia , Sobrevivência Celular , Embrião de Galinha , Microdomínios da Membrana/efeitos dos fármacos , Microdomínios da Membrana/metabolismo , Proteínas de Membrana/efeitos dos fármacos , Proteínas de Membrana/metabolismo , Bainha de Mielina , Neurônios/metabolismo , Nervo Óptico/metabolismo , Nervo Óptico/patologia , Traumatismos do Nervo Óptico/metabolismo , Traumatismos do Nervo Óptico/patologia , Células Fotorreceptoras , Prenilação , Pró-Fármacos , Ratos , Retina , Retinite Pigmentosa , Dicloridrato de trans-1,4-Bis(2-clorobenzaminometil)ciclo-hexano/farmacologia
20.
Front Immunol ; 12: 815020, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35126371

RESUMO

Lipid rafts, also known as microdomains, are important components of cell membranes and are enriched in cholesterol, glycophospholipids and receptors. They are involved in various essential cellular processes, including endocytosis, exocytosis and cellular signaling. Receptors are concentrated at lipid rafts, through which cellular signaling can be transmitted. Pathogens exploit these signaling mechanisms to enter cells, proliferate and egress. However, lipid rafts also play an important role in initiating antimicrobial responses by sensing pathogens via clustered pathogen-sensing receptors and triggering downstream signaling events such as programmed cell death or cytokine production for pathogen clearance. In this review, we discuss how both host and pathogens use lipid rafts and associated proteins in an arms race to survive. Special attention is given to the involvement of the major vault protein, the main constituent of a ribonucleoprotein complex, which is enriched in lipid rafts upon infection with vaccinia virus.


Assuntos
Suscetibilidade a Doenças , Interações Hospedeiro-Patógeno , Microdomínios da Membrana/metabolismo , Animais , Apoptose , Citocinas/biossíntese , Endocitose , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunomodulação , Microdomínios da Membrana/efeitos dos fármacos , Oxirredução , Ligação Proteica , Receptores de Reconhecimento de Padrão/metabolismo , Transdução de Sinais , Partículas de Ribonucleoproteínas em Forma de Abóbada/genética , Partículas de Ribonucleoproteínas em Forma de Abóbada/metabolismo , Internalização do Vírus
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...