Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Heliyon ; 10(10): e31276, 2024 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-38803956

RESUMO

Uncoordinated mutant number-45 myosin chaperone A (UNC-45A), a protein highly conserved throughout evolution, is ubiquitously expressed in somatic cells. It is correlated with tumorigenesis, proliferation, metastasis, and invasion of multiple malignant tumors. The current understanding of the role of UNC-45A in tumor progression is mainly related to the regulation of non-muscle myosin II (NM-II). However, many studies have suggested that the mechanisms by which UNC-45A is involved in tumor progression are far greater than those of NM-II regulation. UNC-45A can also promote tumor cell proliferation by regulating checkpoint kinase 1 (ChK1) phosphorylation or the transcriptional activity of nuclear receptors, and induces chemoresistance to paclitaxel in tumor cells by destabilizing microtubule activity. In this review, we discuss the recent advances illuminating the role of UNC-45A in tumor progression. We also put forward therapeutic strategies targeting UNC-45A, in the hope of paving the way the development of UNC-45A-targeted therapies for patients with malignant tumors.

2.
J Chem Inf Model ; 64(9): 3874-3883, 2024 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-38652138

RESUMO

The lipid raft subdomains in cancer cell membranes play a key role in signal transduction, biomolecule recruitment, and drug transmembrane transport. Augmented membrane rigidity due to the formation of a lipid raft is unfavorable for the entry of drugs, a limiting factor in clinical oncology. The short-chain ceramide (CER) has been reported to promote drug entry into membranes and disrupt lipid raft formation, but the underlying mechanism is not well understood. We recently explored the carrier-membrane fusion dynamics of PEG-DPPE micelles in delivering doxorubicin (DOX). Based on the phase-segregated membrane model composed of DPPC/DIPC/CHOL/GM1/PIP2, we aim to explore the dynamic mechanism of the PEG-DPPE micelle-encapsulating DOXs in association with the raft-included cell membrane modulated by C8 acyl tail CERs. The results show that the lipid raft remains integrated and DOX-resistant subjected to free DOXs and the micelle-encapsulating ones. Addition of CERs disorganizes the lipid raft by pushing CHOL aside from DPPC. It subsequently allows for a good permeability for PEG-DPPE micelle-encapsulated DOXs, which penetrate deeper as CER concentration increases. GM1 is significant in guiding drugs' redistributing between bilayer phases, and the anionic PIP2 further helps DOXs attain the inner bilayer surface. These results elaborate on the perturbing effect of CERs on lipid raft stability, which provides a new comprehensive approach for further design of drug delivery systems.


Assuntos
Ceramidas , Doxorrubicina , Microdomínios da Membrana , Micelas , Simulação de Dinâmica Molecular , Polietilenoglicóis , Polietilenoglicóis/química , Doxorrubicina/química , Doxorrubicina/farmacologia , Doxorrubicina/metabolismo , Ceramidas/química , Microdomínios da Membrana/metabolismo , Microdomínios da Membrana/química , Fosfatidiletanolaminas/química , Humanos
3.
Phys Chem Chem Phys ; 25(23): 16114-16125, 2023 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-37278335

RESUMO

Exploiting micelles of polyethylene glycol-dipalmitoylglycerophosphoethanolamine (PEG-DPPE) as a drug delivery approach is of great promise for improving therapeutic targeting and the half-lives of drugs. To optimize the micelle carriers, pending issues concerning the kinetics underlying the carrier-membrane interplay and the specific contributions of the micelle hydrophobic/hydrophilic components remain to be addressed. Relying on MARTINI coarse-grain (CG) molecular dynamics simulations, we explored the carrier-membrane fusion dynamics of PEG-DPPE micelles with different PEG repetitions in delivering doxorubicin (DOX). A bilayer model composed of 20% phosphatidylglycerol (POPG) and 80% phosphatidylcholine (POPC) was constructed to mimic anionic cancer cell membranes. The CG model of DOX was pioneeringly constructed herein, and it was found to distribute at the hydrophilic/hydrophobic interface of the PEGylated micelles, in agreement with experimental results. The free DOXs cause insignificant disorder of the membrane organization, whereas the PEG-DPPE micelles encapsulating DOX lead to a remarkable membrane invasion supported by the order parameter of the lipid acyl carbon tails and the membrane permeation free energy of DOX. The carrier-bilayer interaction shows a stepwise form attributed to the rearrangement of the zwitterionic/anionic lipids upon the absorption of the DOX-micelle complex on a membrane locality, which initiates the rapid release of DOX to the bilayer interior. Benefiting from the enhanced micelle-membrane interplay, the PEG1250-DPPE micelles result in severe bilayer breakage and deeper membrane insertion of DOX compared to the PEG2000-DPPE micelles. This study provides new theoretical insights into the mechanism of PEG-DPPE micelles in delivering drugs through membranes, which is of benefit for further optimization of PEGylated delivery systems.


Assuntos
Micelas , Polietilenoglicóis , Polietilenoglicóis/química , Linhagem Celular Tumoral , Doxorrubicina/química , Portadores de Fármacos/química
4.
J Chem Inf Model ; 63(14): 4423-4432, 2023 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-37382878

RESUMO

Transmissibility of SARS-CoV-2 initially relies on its trimeric Spike-RBDs to tether the ACE-2 on host cells, and enhanced self-association of ACE-2 engaged with Spike facilitates the viral infection. Two primary packing modes of Spike-ACE2 heteroproteins exist potentially due to discrepant amounts of RBDs loading on ACE-2, but the resultant self-association difference is inherently unclear. We used extensive coarse-grained dynamic simulations to characterize the self-association efficiency, the conformation relevance, and the molecular mechanism of ACE-2 with different RBD amounts. It was revealed that the ACE-2 hanging two/full RBDs (Mode-A) rapidly dimerized into the heteroprotein complex in a compact "linear" conformation, while the bare ACE-2 showed weakened self-association and a protein complex. The RBD-tethered ectodomains of ACE-2 presented a more upright conformation relative to the membrane, and the intermolecular ectodomains were predominantly packed by the neck domains, which was obligated to the rapid protein self-association in a compact pattern. Noted is the fact that the ACE-2 tethered by a single RBD (Mode-B) retained considerable self-association efficiency and clustering capability, which unravels the interrelation of ACE-2 colocalization and protein cross-linkage. The molecular perspectives in this study expound the self-association potency of ACE-2 with different RBD amounts and the viral activity implications, which can greatly enhance our comprehension of SARS-CoV-2 infection details.


Assuntos
COVID-19 , Humanos , Análise por Conglomerados , Dimerização , Simulação de Dinâmica Molecular , Ligação Proteica , SARS-CoV-2
5.
Phys Chem Chem Phys ; 25(27): 18495, 2023 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-37377001

RESUMO

Correction for 'Delivery mechanism of doxorubicin by PEG-DPPE micelles on membrane invasion by dynamic simulations' by Lina Zhao et al., Phys. Chem. Chem. Phys., 2023, 25, 16114-16125, https://doi.org/10.1039/D2CP05946K.

6.
Biophys J ; 122(13): 2675-2685, 2023 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-37218130

RESUMO

Association of the cellular adhesive protein CD44 and the N-terminal (FERM) domain of cytoskeleton adaptors is critical for cell proliferation, migration, and signaling. Phosphorylation of the cytoplasmic domain (CTD) of CD44 acts as an important regulator of the protein association, but the structural transformation and dynamics mechanism remain enigmatic. In this study, extensive coarse-grained simulations were employed to explore the molecular details in the formation of CD44-FERM complex under S291 and S325 phosphorylation, a modification path known to exert reciprocal effects on the protein association. We find that phosphorylation of S291 inhibits complexation by causing the CTD of CD44 to adopt a more closed structure. In contrast, S325 phosphorylation liberates the CD44-CTD from the membrane surface and promotes the linkage with FERM. The phosphorylation-driven transformation is found to occur in a PIP2-dependent manner, with PIP2 effecting the relative stability of the closed and open conformation, and a replacement of PIP2 by POPS greatly abrogates this effect. The revealed interdependent regulation mechanism by phosphorylation and PIP2 in the association of CD44 and FERM further strengthens our understanding of the molecular basis of cellular signaling and migration.


Assuntos
Citoesqueleto , Proteínas , Transdução de Sinais , Conformação Molecular , Ligação Proteica
7.
Zool Res ; 43(5): 886-896, 2022 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-36052553

RESUMO

Various peptide toxins in animal venom inhibit voltage-gated sodium ion channel Nav1.7, including Nav-targeting spider toxin (NaSpTx) Family I. Toxins in NaSpTx Family I share a similar structure, i.e., N-terminal, loops 1-4, and C-terminal. Here, we used Mu-theraphotoxin-Ca2a (Ca2a), a peptide isolated from Cyriopagopus albostriatus, as a template to investigate the general properties of toxins in NaSpTx Family I. The toxins interacted with the cell membrane prior to binding to Nav1.7 via similar hydrophobic residues. Residues in loop 1, loop 4, and the C-terminal primarily interacted with the S3-S4 linker of domain II, especially basic amino acids binding to E818. We also identified the critical role of loop 2 in Ca2a regarding its affinity to Nav1.7. Our results provide further evidence that NaSpTx Family I toxins share similar structures and mechanisms of binding to Nav1.7.


Assuntos
Venenos de Aranha , Animais , Peptídeos/química , Canais de Sódio , Venenos de Aranha/química , Venenos de Aranha/genética , Venenos de Aranha/farmacologia , Bloqueadores do Canal de Sódio Disparado por Voltagem/química , Bloqueadores do Canal de Sódio Disparado por Voltagem/farmacologia , Bloqueadores do Canal de Sódio Disparado por Voltagem/uso terapêutico
8.
ACS Nano ; 16(9): 13783-13799, 2022 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-36099446

RESUMO

Anticancer peptides are promising drug candidates for cancer treatment, but the short circulation time and low delivery efficiency limit their clinical applications. Herein, we designed several lasso-like self-assembling anticancer peptides (LASAPs) integrated with multiple functions by a computer-aided approach. Among these LASAPs, LASAP1 (CRGDKGPDCGKAFRRFLGALFKALSHLL, 1-9 disulfide bond) was determined to be superior to the others because it can self-assemble into homogeneous nanoparticles and exhibits improved stability in serum. Thus, LASAP1 was chosen for proving the design idea. LASAP1 can self-assemble into nanoparticles displaying iRGD on the surface because of its amphiphilic structure and accumulate to the tumor site after injection because of the EPR effect and iRGD targeting to αVß3 integrin. The nanoparticles could disassemble in the acidic microenvironment of the solid tumor, and cleaved by the overexpressed hK2, which was secreted by prostate tumor cells, to release the effector peptide PTP-7b (FLGALFKALSHLL), which was further activated by the acidic pH. Therefore, LASAP1 could target the orthotopic prostate tumor in the model mice after intraperitoneal injection and specifically inhibit tumor growth, with low systematic toxicity. Combining the multiple targeting functions, LASAP1 represents a promising design of self-delivery of peptide drugs for targeted cancer treatments.


Assuntos
Antineoplásicos , Nanopartículas , Neoplasias da Próstata , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Desenho Assistido por Computador , Dissulfetos , Sistemas de Liberação de Medicamentos , Humanos , Integrinas , Masculino , Camundongos , Nanopartículas/química , Peptídeos/química , Neoplasias da Próstata/tratamento farmacológico , Microambiente Tumoral
9.
iScience ; 25(6): 104423, 2022 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-35663038

RESUMO

The exploitation of biocompatible ice-control materials especially the small molecules for non-vitreous cryopreservation remains challenging. Here, we report a small molecule of fulvic acid (FA) with strong hydration ability, which enables non-vitreous cellular cryopreservation by reducing ice growth during freezing and reducing ice recrystallization/promoting ice melting during thawing. Without adding any other cryoprotectants, FA can enhance the recovery of sheep red blood cells (RBCs) by three times as compared with a commercial cryoprotectant (hydroxyethyl starch) under a stringent test condition. Investigation of water mobility reveals that the ice-control properties of FA can be ascribed to its strong bondage to water molecules. Furthermore, we found that FA can be absorbed by RBCs and mainly locates on membranes, suggesting the possible contribution of FA to cell protection through stabilizing membranes. This work bespeaks a bright future for small-molecule cryoprotectants in non-vitreous cryopreservation application.

10.
Biophys J ; 121(14): 2671-2683, 2022 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-35733341

RESUMO

The homodimerization of CD44 plays a key role in an intercellular-to-extracellular signal transduction and tumor progression. Acylated modification and specific membrane environments have been reported to mediate translocation and oligomerization of CD44; however, the underlying molecular mechanism remains elusive. In this study, extensive molecular dynamics simulations are performed to characterize the dimerization of palmitoylated CD44 variants in different bilayer environments. CD44 forms homodimer depending on the cysteines on the juxta-membrane domains, and the dimerization efficiency and packing configurations are defected by their palmitoylated modifications. In the phase-segregated (raft included) membrane, homodimerization of the palmitoylated CD44 is hardly observed, whereas PIP2 addition compensates to realize dimerization. However, the structure of CD44 homodimer formed in the phase-segregated bilayer turns susceptive and PIP2 addition allows for an extensive conformation of the cytoplasmic domain, a proposal prerequisite to access the cytoskeleton linker proteins. The results unravel a delicate competitive relationship between PIP2, lipid raft, and palmitoylation in mediating protein homodimerization, which helps to clarify the dynamic dimer conformations and involved cellular signaling of the CD44 likewise proteins.


Assuntos
Lipoilação , Microdomínios da Membrana , Membrana Celular/metabolismo , Dimerização , Microdomínios da Membrana/metabolismo , Simulação de Dinâmica Molecular , Proteínas/metabolismo
11.
J Biol Chem ; 298(3): 101731, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35176281

RESUMO

Lung cancer has the highest mortality among cancers worldwide due to its high incidence and lack of the effective cures. We have previously demonstrated that the membrane ion channel TMEM16A is a potential drug target for the treatment of lung adenocarcinoma and have identified a pocket of inhibitor binding that provides the basis for screening promising new inhibitors. However, conventional drug discovery strategies are lengthy and costly, and the unpredictable side effects lead to a high failure rate in drug development. Therefore, finding new therapeutic directions for already marketed drugs may be a feasible strategy to obtain safe and effective therapeutic drugs. Here, we screened a library of over 1400 Food and Drug Administration-approved drugs through virtual screening and activity testing. We identified a drug candidate, Zafirlukast (ZAF), clinically approved for the treatment of asthma, that could inhibit the TMEM16A channel in a concentration-dependent manner. Molecular dynamics simulations and site-directed mutagenesis experiments showed that ZAF can bind to S387/N533/R535 in the nonselective inhibitor binding pocket, thereby blocking the channel pore. Furthermore, we demonstrate ZAF can target TMEM16A channel to inhibit the proliferation and migration of lung adenocarcinoma LA795 cells. In vivo experiments showed that ZAF can significantly inhibit lung adenocarcinoma tumor growth in mice. Taken together, we identified ZAF as a novel TMEM16A channel inhibitor with excellent anticancer activity, and as such, it represents a promising candidate for future preclinical and clinical studies.


Assuntos
Adenocarcinoma de Pulmão , Anoctamina-1 , Indóis , Neoplasias Pulmonares , Fenilcarbamatos , Sulfonamidas , Adenocarcinoma de Pulmão/tratamento farmacológico , Adenocarcinoma de Pulmão/genética , Adenocarcinoma de Pulmão/metabolismo , Animais , Anoctamina-1/antagonistas & inibidores , Anoctamina-1/metabolismo , Canais de Cloreto , Indóis/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Camundongos , Fenilcarbamatos/farmacologia , Sulfonamidas/farmacologia
12.
Biochim Biophys Acta Biomembr ; 1864(1): 183777, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34537214

RESUMO

TMEM16A, the calcium-activated chloride channel, is broadly expressed and plays pivotal roles in diverse physiological processes. To understand the structural and functional relationships of TMEM16A, it is necessary to fully clarify the structural basis of the gating of the TMEM16A channel. Herein, we performed the protein electrostatic analysis and molecular dynamics simulation on the TMEM16A in the presence and absence of Ca2+. Data showed that the separation of TM4 and TM6 causes pore expansion, and Q646 may be a key residue for the formation of π-helix in the middle segment of TM6. Moreover, E705 was found to form a group of H-bond interactions with D554/K588/K645 below the hydrophobic gate to stabilize the closed conformation of the pore in the Ca2+-free state. Interestingly, in the Ca2+ bound state, the E705 side chain swings 100o to serve as Ca2+-binding coordination and released K645. K645 is closer to the hydrophobic gate in the calcium-bound state, which facilitates the provision of electrostatic forces for chloride ions as the ions pass through the hydrophobic gate. Our findings provide the structural-based insights to understanding the mechanisms of gating of TMEM16A.


Assuntos
Anoctamina-1/ultraestrutura , Comunicação Celular/genética , Conformação Proteica em alfa-Hélice/genética , Conformação Proteica , Anoctamina-1/química , Anoctamina-1/genética , Cálcio/química , Humanos , Interações Hidrofóbicas e Hidrofílicas , Simulação de Dinâmica Molecular , Eletricidade Estática , Relação Estrutura-Atividade
13.
J Biol Chem ; 297(3): 101016, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34329684

RESUMO

As a calcium-activated chloride channel regulated by the intracellular Ca2+ concentration and membrane potential, TMEM16A has attracted considerable attention and has been proposed as a novel anticancer drug target. We have previously reported that the pocket above the ion conductance pore could be a nonselective inhibitor-binding pocket. However, whether this pocket is druggable remains unexplored. In this study, we performed virtual screening to target the presumed inhibitor-binding pocket and identified a highly effective TMEM16A inhibitor, theaflavin (TF: a tea polyphenol in black tea). Molecular dynamics simulations revealed that theaflavin adopts a "wedge insertion mode" to block the ion conduction pore and induces pore closure. Moreover, the binding mode showed that the TF pedestal plays an important role in pore blockade, and R515, R535, T539, K603, E623, and E633 were determined to be most likely to interact directly with the pedestal. Mutagenesis experiment results corroborated the mechanism through which TF binds to this pocket. Combined with the quantitative calculation results, our data indicated that the three hydroxyl groups on the pedestal may be the most crucial pharmacophores for TMEM16A inhibition by TF. Finally, antitumor experiments revealed that TF could target TMEM16A to inhibit the proliferation and migration of LA795 cells, indicating the potential therapeutic effect of TF on the growth of lung adenocarcinoma with high TMEM16A expression. The successful application of drug screening strategies based on this binding pocket highlights new directions for discovering superior modulators and contributes to the development of novel therapeutics for lung adenocarcinoma.


Assuntos
Adenocarcinoma de Pulmão/patologia , Anoctamina-1/metabolismo , Biflavonoides/metabolismo , Catequina/metabolismo , Neoplasias Pulmonares/patologia , Proteínas de Neoplasias/metabolismo , Adenocarcinoma de Pulmão/tratamento farmacológico , Adenocarcinoma de Pulmão/metabolismo , Antineoplásicos/farmacologia , Biflavonoides/farmacologia , Sítios de Ligação , Catequina/farmacologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Simulação de Dinâmica Molecular
14.
Arch Biochem Biophys ; 695: 108650, 2020 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-33132191

RESUMO

TMEM16A is a calcium-activated chloride channel that is associate with several diseases, including pulmonary diseases, hypertension, diarrhea and cancer. The CaCCinh-A01 (A01) is widely recognized as an efficient blocker of TMEM16A and has been used as a tool drug to inhibit TMEM16A currents in the laboratory. A01 also has excellent pharmacokinetic properties and can be developed as a drug to target TMEM16A. However, the molecular mechanism how A01 inhibits TMEM16A is still elusive, which slows down its drug development process. Here, calculations identified that the binding pocket of A01 was located above the pore, and it was also discovered that the binding of A01 to TMEM16A not only blocked the pore but also led to its collapse. The interaction model analysis predicted that R515/K603/E623 were crucial residues for the binding between TMEM16A and A01, and the site-directed mutagenesis studies confirmed the above results. The binding mode and quantum chemical calculations showed that the carboxyl and the amide oxygen atom of A01 were the key interaction sites between TMEM16A and A01. Therefore, our study proposed the inhibitory mechanism of TMEM16A current by A01 and revealed how A01 inhibits TMEM16A at the molecular level. These findings will shed light on both the development of A01 as a potential drug for TMEM16A dysfunction-related disorders and drug screening targeting the pocket.


Assuntos
Anoctamina-1 , Simulação de Acoplamento Molecular , Proteínas de Neoplasias , Tiofenos/química , Substituição de Aminoácidos , Anoctamina-1/antagonistas & inibidores , Anoctamina-1/química , Anoctamina-1/genética , Anoctamina-1/metabolismo , Sítios de Ligação , Células HEK293 , Humanos , Mutagênese Sítio-Dirigida , Mutação de Sentido Incorreto , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo
15.
PLoS Comput Biol ; 16(4): e1007777, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32271757

RESUMO

The co-localization of Cluster-of-Differentiation-44 protein (CD44) and cytoplasmic adaptors in specific membrane environments is crucial for cell adhesion and migration. The process is controlled by two different pathways: On the one hand palmitoylation keeps CD44 in lipid raft domains and disables the linking to the cytoplasmic adaptor, whereas on the other hand, the presence of phosphatidylinositol-4,5-biphosphate (PIP2) lipids accelerates the formation of the CD44-adaptor complex. The molecular mechanism explaining how CD44 is migrating into and out of the lipid raft domains and its dependence on both palmitoylations and the presence of PIP2 remains, however, elusive. In this study, we performed extensive molecular dynamics simulations to study the raft affinity and translocation of CD44 in phase separated model membranes as well as more realistic plasma membrane environments. We observe a delicate balance between the influence of the palmitoylations and the presence of PIP2 lipids: whereas the palmitoylations of CD44 increases the affinity for raft domains, PIP2 lipids have the opposite effect. Additionally, we studied the association between CD44 and the membrane adaptor FERM in dependence of these factors. We find that the presence of PIP2 lipids allows CD44 and FERM to associate in an experimentally observed binding mode whereas the highly palmitoylated species shows no binding affinity. Together, our results shed light on the sophisticated mechanism on how membrane translocation and peripheral protein association can be controlled by both protein modifications and membrane composition.


Assuntos
Receptores de Hialuronatos , Lipoilação/fisiologia , Microdomínios da Membrana , Simulação de Dinâmica Molecular , Fosfatidilinositol 4,5-Difosfato , Membrana Celular/química , Membrana Celular/metabolismo , Biologia Computacional , Citoplasma/química , Citoplasma/metabolismo , Receptores de Hialuronatos/química , Receptores de Hialuronatos/metabolismo , Microdomínios da Membrana/química , Microdomínios da Membrana/metabolismo , Fosfatidilinositol 4,5-Difosfato/química , Fosfatidilinositol 4,5-Difosfato/metabolismo
16.
Medicine (Baltimore) ; 99(9): e19323, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32118762

RESUMO

BACKGROUND: Low intra-abdominal pressure (IAP) and deep neuromuscular blockade (NMB) are frequently used in laparoscopic abdominal surgery to improve surgical space conditions and decrease postoperative pain. The evidence supporting operations using low IAP and deep NMB is open to debate. METHODS: The feasibility of the routine use of low IAP +deep NMB during laparoscopic surgery was examined. A meta-analysis is conducted with randomized controlled trials (RCTs) to compare the influence of low IAP + deep NMB vs. low IAP + moderate NMB, standard IAP +deep NMB, and standard IAP + moderate NMB during laparoscopic procedures on surgical space conditions, the duration of surgery and postoperative pain. RCTs were identified using the Cochrane, Embase, PubMed, and Web of Science databases from initiation to June 2019. Our search identified 9 eligible studies on the use of low IAP + deep NMB and surgical space conditions. RESULTS: Low IAP + deep NMB during laparoscopic surgery did not improve the surgical space conditions when compared with the use of moderate NMB, with a mean difference (MD) of -0.09 (95% confidence interval (CI): -0.55-0.37). Subgroup analyses showed improved surgical space conditions with the use of low IAP + deep NMB compared with low IAP + moderate NMB, (MD = 0.63 [95% CI:0.06-1.19]), and slightly worse conditions compared with the use of standard IAP + deep NMB and standard IAP + moderate NMB, with MDs of -1.13(95% CI:-1.47 to 0.79) and -0.87(95% CI:-1.30 to 0.43), respectively. The duration of surgery did not improve with low IAP + deep NMB, (MD = 1.72 [95% CI: -1.69 to 5.14]), and no significant reduction in early postoperative pain was found in the deep-NMB group (MD = -0.14 [95% CI: -0.51 to 0.23]). CONCLUSION: Low IAP +deep NMB is not significantly more effective than other IAP +NMB combinations for optimizing surgical space conditions, duration of surgery, or postoperative pain in this meta-analysis. Whether the use of low IAP + deep NMB results in fewer intraoperative complications, enhanced quality of recovery or both after laparoscopic surgery should be studied in the future.


Assuntos
Cavidade Abdominal/fisiopatologia , Bloqueio Neuromuscular/efeitos adversos , Pressão/efeitos adversos , Cavidade Abdominal/irrigação sanguínea , Humanos , Complicações Intraoperatórias , Laparoscopia/métodos , Bloqueio Neuromuscular/métodos
17.
J Membr Biol ; 253(2): 167-181, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32170353

RESUMO

KCNQ2 channel is one of the important members of potassium voltage-gated channel. KCNQ2 is closely related to neuronal excitatory diseases including epilepsy and neuropathic pain, and also acts as a drug target of the anti-epileptic drug, retigabine (RTG). In the past few decades, RTG has shown strong efficacy in the treatment of refractory epilepsy but has been withdrawn from clinical use due to its multiple adverse effects in clinical phase III trials. To overcome the drawbacks of RTG, several RTG analogues have been developed with different activation potency to KCNQ2. However, the detailed molecular mechanism by which these RTG analogues regulate KCNQ2 channel remains obscure. In this study, we used molecular simulations to analyse the interaction mode between the RTG analogues and KCNQ2, and to determine their molecular mechanism of action. Our data show that the van der Waals interactions, hydrophobic interactions, hydrogen bond, halogen bond, and π-π stacking work together to maintain the binding stability of the drugs in the binding pocket. On an atomic scale, the amide group in the carbamate and the amino group in the 2-aminophenyl moiety of RTG and RL648_81 are identified as key interaction sites. Our finding provides insight into the molecular mechanism by which KCNQ2 channels are regulated by RTG analogues. It also provides direct theoretical support for optimizing design of the KCNQ2 channel openers in the future, which will help treat refractory epilepsy caused by nerve excitability.


Assuntos
Carbamatos/farmacologia , Ativação do Canal Iônico/efeitos dos fármacos , Canal de Potássio KCNQ2/química , Canal de Potássio KCNQ2/fisiologia , Moduladores de Transporte de Membrana/farmacologia , Fenilenodiaminas/farmacologia , Sequência de Aminoácidos , Sítios de Ligação , Ligação de Hidrogênio , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Ligação Proteica , Conformação Proteica , Relação Estrutura-Atividade
18.
J Mol Biol ; 432(2): 467-483, 2020 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-31805282

RESUMO

The RNA-binding protein fused in sarcoma (FUS) forms physiological granules and pathological fibrils, which facilitate RNA functions and cause neurodegenerative diseases, respectively. Phosphorylation at Ser/Thr residues may regulate the functional assembly of FUS and prevent pathological aggregation in cells. However, the low-complexity nature of the FUS sequence makes it challenging to characterize how phosphorylation of specific sites within the core amyloid-forming segment affects aggregation. Taking advantage of the recently solved molecular structures of the fibrillar core of the FUS low-complexity (FUS-LC) domain, we systematically investigated the aggregation of repeated segments within the core. We identified a segment with a strong amyloid-forming tendency that induced the aggregation of FUS-LC domain in phase-separated liquid droplets and further seeded the aggregation of full-length FUS. The aggregation propensity and seeding ability of this amyloid-forming segment were modulated by site-specific phosphorylation. Solid-state nuclear magnetic resonance (NMR) spectroscopy and computational modeling implied that site-specific phosphorylation at Ser61 plays key roles in FUS assembly by disrupting both intra- and intermolecular interactions that maintain the amyloid core structure.


Assuntos
Amiloide/genética , Amiloidose/genética , Agregação Patológica de Proteínas/genética , Proteína FUS de Ligação a RNA/genética , Proteínas de Ligação a RNA/genética , Amiloide/ultraestrutura , Proteínas Amiloidogênicas/genética , Proteínas Amiloidogênicas/ultraestrutura , Amiloidose/patologia , Humanos , Estrutura Molecular , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/patologia , Ressonância Magnética Nuclear Biomolecular , Fosforilação/genética , Agregação Patológica de Proteínas/patologia , Conformação Proteica , Domínios Proteicos/genética , Proteína FUS de Ligação a RNA/ultraestrutura , Proteínas de Ligação a RNA/ultraestrutura
19.
Biophys J ; 114(8): 1858-1868, 2018 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-29694864

RESUMO

Phosphatidylinositol 4,5-bisphosphate (PIP2) acts as a signaling lipid, mediating membrane trafficking and recruitment of proteins to membranes. A key example is the PIP2-dependent regulation of the adhesion of L-selectin to the cytoskeleton adaptors of the N-terminal subdomain of ezrin-radixin-moesin (FERM). The molecular details of the mediating behavior of multivalent anionic PIP2 lipids in this process, however, remain unclear. Here, we use coarse-grained molecular dynamics simulation to explore the mechanistic details of PIP2 in the transformation, translocation, and association of the FERM/L-selectin complex. We compare membranes of different compositions and find that anionic phospholipids are necessary for both FERM and the cytoplasmic domain of L-selectin to absorb on the membrane surface. The subsequent formation of the FERM/L-selectin complex is strongly favored by the presence of PIP2, which clusters around both proteins and triggers a conformational transition in the cytoplasmic domain of L-selectin. We are able to quantify the effect of PIP2 on the association free energy of the complex by means of a potential of mean force. We conclude that PIP2 behaves as an adhesive agent to enhance the stability of the FERM/L-selectin complex and identify key residues involved. The molecular information revealed in this study highlights the specific role of membrane lipids such as PIP2 in protein translocation and potential signaling.


Assuntos
Domínios FERM , Selectina L/metabolismo , Simulação de Dinâmica Molecular , Fosfatidilinositol 4,5-Difosfato/metabolismo , Membrana Celular/metabolismo , Selectina L/química , Ligação Proteica , Transporte Proteico
20.
Proteins ; 86(8): 844-852, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29569285

RESUMO

P-selectin glycoprotein ligand-1 (PSGL-1) is a homodimeric mucin ligand that is important to mediate the earliest adhesive event during an inflammatory response by rapidly forming and dissociating the selectin-ligand adhesive bonds. Recent research indicates that the noncovalent associations between the PSGL-1 transmembrane domains (TMDs) can substitute for the C320-dependent covalent bond to mediate the dimerization of PSGL-1. In this article, we combined TOXCAT assays and molecular dynamics (MD) simulations to probe the mechanism of PSGL-1 dimerization. The results of TOXCAT assays and Martini coarse-grained molecular dynamics (CG MD) simulations demonstrated that PSGL-1 TMDs strongly dimerized in a natural membrane and a leucine zipper motif was responsible for the noncovalent dimerization of PSGL-1 TMD since mutations of the residues that occupied a or d positions in an (abcdefg)n leucine heptad repeat motif significantly reduced the dimer activity. Furthermore, we studied the effects of the disulfide bond on the PSGL-1 dimer using MD simulations. The disulfide bond was critical to form the leucine zipper structure, by which the disulfide bond further improved the stability of the PSGL-1 dimer. These findings provide insights to understand the transmembrane association of PSGL-1 that is an important structural basis for PSGL-1 preferentially binding to P-selectin to achieve its biochemical and biophysical functions.


Assuntos
Zíper de Leucina , Glicoproteínas de Membrana/química , Multimerização Proteica , Sequência de Aminoácidos , Animais , Escherichia coli/genética , Humanos , Simulação de Dinâmica Molecular , Ligação Proteica , Domínios Proteicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA