Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Sci Rep ; 14(1): 4026, 2024 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-38369574

RESUMO

Antibiotics are indispensable to infection management. However, use of antibiotics can cause gut microbiota dysbiosis, which has been linked to cognitive impairment by disrupting communication between the gut microbiota and the brain. We conducted a systematic review and meta-analysis on the effects of long-term antibiotic use on cognitive outcomes. We have searched PubMed, Web of Science, Embase, Cochrane Library and Scopus for English publications before March 2023 following the PRISMA guidelines. Screening, data extraction, and quality assessment were performed in duplicate. 960 articles were screened and 16 studies which evaluated the effect of any antibiotic compared to no antibiotics or placebo were included. Case-reports, in vitro and animal studies were excluded. We found that antibiotic use was associated with worse cognitive outcomes with a pooled effect estimate of - 0.11 (95% CI - 0.15, - 0.07, Z = 5.45; P < 0.00001). Subgroup analyses performed on adult vs pediatric patients showed a similar association of antibiotic on cognition in both subgroups. Antibiotic treatment was not associated with worse cognition on subjects with existing cognitive impairment. On the other hand, antibiotic treatment on subjects with no prior cognitive impairment was associated with worse cognitive performance later in life. This calls for future well-designed and well-powered studies to investigate the impact of antibiotics on cognitive performance.


Assuntos
Disfunção Cognitiva , Microbioma Gastrointestinal , Adulto , Criança , Humanos , Disfunção Cognitiva/tratamento farmacológico , Cognição , Antibacterianos/efeitos adversos , Cabeça
2.
Cell Rep Med ; 4(8): 101156, 2023 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-37586324

RESUMO

We describe a general approach to produce bone and cartilaginous structures utilizing the self-regenerative capacity of the intercostal rib space to treat a deformed metacarpophalangeal joint and microtia. Anatomically precise 3D molds were positioned on the perichondro-periosteal or perichondral flap of the intercostal rib without any other exogenous elements. We find anatomically precise metacarpal head and auricle constructs within the implanted molds after 6 months. The regenerated metacarpal head was used successfully to surgically repair the deformed metacarpophalangeal joint. Auricle reconstructive surgery in five unilateral microtia patients yielded good aesthetic and functional results. Long-term follow-up revealed the auricle constructs were safe and stable. Single-cell RNA sequencing analysis reveal early infiltration of a cell population consistent with mesenchymal stem cells, followed by IL-8-stimulated differentiation into chondrocytes. Our results demonstrate the repair and regeneration of tissues using only endogenous factors and a viable treatment strategy for bone and tissue structural defects.


Assuntos
Microtia Congênita , Células-Tronco Mesenquimais , Humanos , Cartilagem da Orelha/cirurgia , Engenharia Tecidual/métodos , Microtia Congênita/terapia , Condrócitos
3.
Nat Med ; 29(8): 2007-2018, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37524952

RESUMO

Host-pathogen interactions and pathogen evolution are underpinned by protein-protein interactions between viral and host proteins. An understanding of how viral variants affect protein-protein binding is important for predicting viral-host interactions, such as the emergence of new pathogenic SARS-CoV-2 variants. Here we propose an artificial intelligence-based framework called UniBind, in which proteins are represented as a graph at the residue and atom levels. UniBind integrates protein three-dimensional structure and binding affinity and is capable of multi-task learning for heterogeneous biological data integration. In systematic tests on benchmark datasets and further experimental validation, UniBind effectively and scalably predicted the effects of SARS-CoV-2 spike protein variants on their binding affinities to the human ACE2 receptor, as well as to SARS-CoV-2 neutralizing monoclonal antibodies. Furthermore, in a cross-species analysis, UniBind could be applied to predict host susceptibility to SARS-CoV-2 variants and to predict future viral variant evolutionary trends. This in silico approach has the potential to serve as an early warning system for problematic emerging SARS-CoV-2 variants, as well as to facilitate research on protein-protein interactions in general.


Assuntos
COVID-19 , Aprendizado Profundo , Humanos , COVID-19/genética , SARS-CoV-2/genética , Inteligência Artificial , Ligação Proteica
4.
J Clin Med ; 11(18)2022 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-36143101

RESUMO

There is mounting evidence that statin use is beneficial for COVID-19 outcomes. We performed a systematic review and meta-analysis to evaluate the association between statin use and mortality, intensive care unit (ICU) admission and mechanical ventilation in COVID-19 patients, on studies which provided covariate adjusted effect estimates, or performed propensity score matching. We searched PubMed, Embase, Web of Science and Scopus for studies and extracted odds or hazard ratios for specified outcome measures. Data synthesis was performed using a random-effects inverse variance method. Risk of bias, heterogeneity and publication bias were analyzed using standard methods. Our results show that statin use was associated with significant reductions in mortality (OR = 0.72, 95% CI: 0.67-0.77; HR = 0.74, 95% CI: 0.69, 0.79), ICU admission (OR = 0.94, 95% CI: 0.89-0.99; HR = 0.76, 95% CI: 0.60-0.96) and mechanical ventilation (OR = 0.84, 95% CI: 0.78-0.92; HR = 0.67, 95% CI: 0.47-0.97). Nevertheless, current retrospective studies are based on the antecedent use of statins prior to infection and/or continued use of statin after hospital admission. The results may not apply to the de novo commencement of statin treatment after developing COVID-19 infection. Prospective studies are lacking and necessary.

6.
MedComm (2020) ; 3(2): e143, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35592756

RESUMO

The SARS-CoV-2 Omicron BA.1 variant of concern contains more than 30 mutations in the spike protein, with half of these mutations localized in the receptor-binding domain (RBD). Emerging evidence suggests that these large number of mutations impact the neutralizing efficacy of vaccines and monoclonal antibodies. We investigated the relative contributions of spike protein and RBD mutations in Omicron BA.1 variants on infectivity, cell-cell fusion, and their sensitivity to neutralization by monoclonal antibodies or vaccinated sera from individuals who received homologous (CoronaVac, SinoPharm) or heterologous (CoronaVac-BNT162b2, BioNTech) and nonhuman primates that received a recombinant RBD protein vaccine. Our data overall reveal that the mutations in the spike protein reduced infectivity and cell-cell fusion compared to the D614G variant. The impaired infectivity and cell-cell fusion were dependent on non-RBD mutations. We also find reduced sensitivity to neutralization by monoclonal antibodies and vaccinated sera. However, our results also show that nonhuman primates receiving a recombinant RBD protein vaccine show substantial neutralization activity. Our study sheds light on the molecular differences in neutralizing antibody escape by the Omicron BA.1 variant, and highlights the promise of recombinant RBD vaccines in neutralizing the threat posed by the Omicron BA.1 variant.

7.
MedComm (2020) ; 3(1): e126, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35317190

RESUMO

New genetic variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) constantly emerge through unmitigated spread of the virus in the ongoing Coronavirus disease 2019 pandemic. Omicron (B.1.1.529), the latest variant of concern (VOC), has so far shown exceptional spread and infectivity and has established itself as the dominant variant in recent months. The SARS-CoV-2 spike glycoprotein is a key component for the recognition and binding to host cell angiotensin-converting enzyme 2 receptors. The Omicron variant harbors a cluster of substitutions/deletions/insertions, and more than 30 mutations are located in spike. Some noticeable mutations, including K417N, T478K, N501Y, and P681H, are shared with the previous VOCs Alpha, Beta, Gamma, or Delta variants and have been proven to be associated with higher transmissibility, viral infectivity, and immune evasion potential. Studies have revealed that the Omicron variant is partially resistant to the neutralizing activity of therapeutic antibodies and convalescent sera, which poses significant challenges for the clinical effectiveness of the current vaccines and therapeutic antibodies. We provide a comprehensive analysis and summary of the epidemiology and immune escape mechanisms of the Omicron variant. We also suggest some therapeutic strategies against the Omicron variant. This review, therefore, aims to provide information for further research efforts to prevent and contain the impact of new VOCs during the ongoing pandemic.

8.
Adv Physiol Educ ; 45(4): 856-868, 2021 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-34473584

RESUMO

Practical demonstration of cardiomyocyte function requires substantial preparation, a source of freshly isolated animal hearts, and specialized equipment. Even where such resources are available, it is not conducive for demonstration to any more than a few students at a time. These approaches are also not consistent with the 3R principle (replacement, reduction, and refinement) of ethical use of animals. We present an implementation of the LabHEART software, developed by Donald Bers and Jose Puglisi, for medical students. Prior to the activity, students had lectures covering the physiological and pharmacological aspects of cardiac excitation-contraction (EC) coupling. We used this problem-based activity to help students consolidate their knowledge and to allow a hands-on approach to explore the key features of EC coupling. Students simulate and measure action potentials, intracellular calcium changes, and cardiomyocyte contraction. They also apply drugs that target ion channels (e.g., nifedipine or tetrodotoxin) or sympathetic input (using isoproterenol) and explore changes to EC coupling. Furthermore, by modifying the biophysical parameters of key ion channels involved in the electrical activity of the heart, students also explore the effect of channelopathies such as long QT syndromes. We describe approaches to implement this activity in a flipped classroom format, with recorded lecture materials provided ahead of the practical to facilitate active learning. We also describe our experiences implementing this activity online. The content and difficulty of the activity can be altered to suit individual courses and is also amenable to promote peer-driven learning.


Assuntos
Laboratórios , Estudantes de Medicina , Animais , Simulação por Computador , Computadores , Humanos , Aprendizagem Baseada em Problemas
9.
Adv Physiol Educ ; 45(4): 702-708, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34498935

RESUMO

Patch-clamp electrophysiological recordings of neuronal activity require a large amount of space and equipment. The technique is difficult to master and not conducive to demonstration to more than a few medical students. Therefore, neurophysiological education is mostly limited to classroom-based pedagogies such as lectures. However, the demonstration of concepts such as changes in membrane potential and ion channel activity is best achieved with hands-on approaches. This article details an in silico activity suitable for large groups of medical students that demonstrates the key concepts in neurophysiology using the LabAXON simulation software. Learning activities in our practical include 1) measurements of voltage and time parameters of the neuronal action potential and its relationship to the Nernst potentials of Na+ and K+; 2) determination of the stimulus threshold to evoke action potentials; 3) demonstration of the refractory period of an action potential; and 4) voltage-clamp experiments to determine the current-voltage relationship of voltage-gated Na+ and K+ channels and the voltage dependence of, and recovery from, inactivation of voltage-gated Na+ channels. We emphasized the accuracy of quantitative measurements as well as the correct use of units. The level of difficulty of the activity can be altered through different multiple choice questions relating to material introduced in the associated lectures. This practical activity is suitable for different class sizes and is adaptable for delivery with online platforms. Student feedback showed that the students felt the activity helped them consolidate their understanding of the lecture material.


Assuntos
Neurofisiologia , Estudantes de Medicina , Potenciais de Ação , Humanos , Potenciais da Membrana , Sódio
11.
Br J Pharmacol ; 178(8): 1855-1868, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33555037

RESUMO

BACKGROUND AND PURPOSE: ß-Arrestin2 recruitment to µ-receptors may contribute to the development of opioid side effects. This possibility led to the development of TRV130 and PZM21, opioids reportedly biased against ß-arrestin2 recruitment in favour of G-protein signalling. However, low efficacy ß-arrestin2 recruitment by TRV130 and PZM21 may simply reflect partial agonism overlooked due to overexpression of µ-receptors. EXPERIMENTAL APPROACH: Efficacies and apparent potencies of DAMGO, morphine, PZM21 and TRV130 as stimulators of ß-arrestin2 recruitment and inhibitors of cAMP accumulation were assessed in CHO cells stably expressing µ-receptors. Receptor availability was depleted through prior exposure of cells to the irreversible antagonist, ß-FNA. We also examined whether µ-receptor availability influences TRV130 anti-nociception and/or tolerance using the tail withdrawal assay in wild-type C57BL/6 and µ+/- mice. KEY RESULTS: Morphine, PZM21 and TRV130 were partial agonists in the ß-arrestin2 recruitment assay. Only TRV130 exhibited partial agonism in the cAMP assay. Exposure to ß-FNA to reduce µ-receptor availability further limited the efficacy of TRV130 and revealed morphine and PZM21 to be partial agonists. Despite having partial efficacy in vitro, TRV130 caused potent anti-nociception (ED50 : 0.33 mg·kg-1 ) in wild-type mice, without tolerance after daily administration for 10 days. TRV130 caused similar anti-nociception in µ+/- mice, with marked tolerance on day 4 of injections. CONCLUSION AND IMPLICATIONS: Our findings emphasise the importance of receptor reserve when characterising µ-receptor agonists. Reduced receptor availability reveals that TRV130 is a partial agonist capable of tolerance, despite having limited efficacy for ß-arrestin2 recruitment to the µ-receptor.


Assuntos
Morfina , Receptores Opioides mu , Analgésicos Opioides/farmacologia , Animais , Cricetinae , Cricetulus , Tolerância a Medicamentos , Camundongos , Camundongos Endogâmicos C57BL , Morfina/farmacologia , Compostos de Espiro , Tiofenos
12.
Precis Clin Med ; 4(3): 149-154, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35693215

RESUMO

To assess the impact of the key non-synonymous amino acid substitutions in the RBD of the spike protein of SARS-CoV-2 variant B.1.617.1 (dominant variant identified in the current India outbreak) on the infectivity and neutralization activities of the immune sera, L452R and E484Q (L452R-E484Q variant), pseudotyped virus was constructed (with the D614G background). The impact on binding with the neutralizing antibodies was also assessed with an ELISA assay. Pseudotyped virus carrying a L452R-E484Q variant showed a comparable infectivity compared with D614G. However, there was a significant reduction in the neutralization activity of the immune sera from non-human primates vaccinated with a recombinant receptor binding domain (RBD) protein, convalescent patients, and healthy vaccinees vaccinated with an mRNA vaccine. In addition, there was a reduction in binding of L452R-E484Q-D614G protein to the antibodies of the immune sera from vaccinated non-human primates. These results highlight the interplay between infectivity and other biologic factors involved in the natural evolution of SARS-CoV-2. Reduced neutralization activities against the L452R-E484Q variant will have an impact on health authority planning and implications for the vaccination strategy/new vaccine development.

13.
Br J Pharmacol ; 177(15): 3436-3448, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32246840

RESUMO

BACKGROUND AND PURPOSE: A fluorinated derivative (2F-MT-45) of the synthetic µ-opioid receptor agonist MT-45 (1-cyclohexyl-4-(1,2-diphenylethyl)piperazine) was recently identified in a seized illicit tablet. While MT-45 is a Class A drug, banned in a number of countries, nothing is known about the pharmacology of 2F-MT-45. This study compares the pharmacology of MT-45, its fluorinated derivatives and two of its metabolites. EXPERIMENTAL APPROACH: We used a ß-arrestin2 recruitment assay in CHO cells stably expressing µ receptors to quantify the apparent potencies and efficacies of known (MT-45, morphine, fentanyl and DAMGO) and potential agonists. In addition, the GloSensor protein was transiently expressed to quantify changes in cAMP levels. We measured Ca2+ to investigate whether MT-45 and its metabolites have effects on GluN1/N2A NMDA receptors stably expressed in Ltk- cells. KEY RESULTS: The fluorinated MT-45 derivatives have higher apparent potencies (2F-MT-45: 42 nM) than MT-45 (1.3 µM) for inhibition of cAMP accumulation and ß-arrestin2 recruitment (2F-MT-45: 196 nM; MT-45: 23.1 µM). While MT-45 and 2F-MT-45 are poor recruiters of ß-arrestin2, they have similar efficacies for reducing cAMP levels as DAMGO. Two MT-45 metabolites displayed negligible potencies as µ receptor agonists, but one, 1,2-diphenylethylpiperazine, inhibited the NMDA receptor with an IC50 of 29 µM. CONCLUSION AND IMPLICATIONS: Fluorinated derivatives of MT-45 are potent µ receptor agonists and this may pose a danger to illicit opioid users. Inhibition of NMDA receptors by a metabolite of MT-45 may contribute to the reported dissociative effects.


Assuntos
Morfina , Receptores Opioides mu , Analgésicos Opioides/farmacologia , Animais , Cricetinae , Cricetulus , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Piperazina , Piperazinas
14.
J Biol Chem ; 294(7): 2375-2385, 2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30545943

RESUMO

GABAA receptors (GABAARs) are pentameric ligand-gated ion channels that mediate synaptic inhibition throughout the central nervous system. The α1ß2γ2 receptor is the major subtype in the brain; GABA binds at the ß2(+)α1(-) interface. The structure of the homomeric ß3 GABAAR, which is not activated by GABA, has been solved. Recently, four additional heteromeric structures were reported, highlighting key residues required for agonist binding. Here, we used a protein engineering method, taking advantage of knowledge of the key binding residues, to create a ß3(+)α1(-) heteromeric interface in the homomeric human ß3 GABAAR that enables GABA-mediated activation. Substitutions were made in the complementary side of the orthosteric binding site in loop D (Y87F and Q89R), loop E (G152T), and loop G (N66D and A70T). The Q89R and G152T combination enabled low-potency activation by GABA and potentiation by propofol but impaired direct activation by higher propofol concentrations. At higher concentrations, GABA inhibited gating of ß3 GABAAR variants containing Y87F, Q89R, and G152T. Reversion of Phe87 to tyrosine abolished GABA's inhibitory effect and partially recovered direct activation by propofol. This tyrosine is conserved in homomeric GABAARs and in the Erwinia chrysanthemi ligand-gated ion channel and may be essential for the absence of an inhibitory effect of GABA on homomeric channels. This work demonstrated that only two substitutions, Q89R and G152T, in ß3 GABAAR are sufficient to reconstitute GABA-mediated activation and suggests that Tyr87 prevents inhibitory effects of GABA.


Assuntos
Ativação do Canal Iônico , Mutação de Sentido Incorreto , Estrutura Secundária de Proteína , Receptores de GABA-B , Substituição de Aminoácidos , Domínio Catalítico , Dickeya chrysanthemi/química , Dickeya chrysanthemi/genética , Dickeya chrysanthemi/metabolismo , Células HEK293 , Humanos , Propofol/farmacologia , Receptores de GABA-B/química , Receptores de GABA-B/genética , Receptores de GABA-B/metabolismo , Ácido gama-Aminobutírico/química , Ácido gama-Aminobutírico/metabolismo
15.
Anesth Analg ; 127(3): 650-660, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29958221

RESUMO

BACKGROUND: Cardiotoxic effects of local anesthetics (LAs) involve inhibition of NaV1.5 voltage-gated Na channels. Metastatic breast and colon cancer cells also express NaV1.5, predominantly the neonatal splice variant (nNaV1.5) and their inhibition by LAs reduces invasion and migration. It may be advantageous to target cancer cells while sparing cardiac function through selective blockade of nNaV1.5 and/or by preferentially affecting inactivated NaV1.5, which predominate in cancer cells. We tested the hypotheses that lidocaine and levobupivacaine differentially affect (1) adult (aNaV1.5) and nNaV1.5 and (2) the resting and inactivated states of NaV1.5. METHODS: The whole-cell voltage-clamp technique was used to evaluate the actions of lidocaine and levobupivacaine on recombinant NaV1.5 channels expressed in HEK-293 cells. Cells were transiently transfected with cDNAs encoding either aNaV1.5 or nNaV1.5. Voltage protocols were applied to determine depolarizing potentials that either activated or inactivated 50% of maximum conductance (V½ activation and V½ inactivation, respectively). RESULTS: Lidocaine and levobupivacaine potently inhibited aNaV1.5 (IC50 mean [SD]: 20 [22] and 1 [0.6] µM, respectively) and nNaV1.5 (IC50 mean [SD]: 17 [10] and 3 [1.6] µM, respectively) at a holding potential of -80 mV. IC50s differed significantly between lidocaine and levobupivacaine with no influence of splice variant. Levobupivacaine induced a statistically significant depolarizing shift in the V½ activation for aNaV1.5 (mean [SD] from -32 [4.6] mV to -26 [8.1] mV) but had no effect on the voltage dependence of activation of nNaV1.5. Lidocaine had no effect on V½ activation of either variant but caused a significantly greater depression of maximum current mediated by nNaV1.5 compared to aNaV1.5. Similar statistically significant shifts in the V½ inactivation (approximately -10 mV) occurred for both LAs and NaV1.5 variants. Levobupivacaine (1 µM) caused a significantly greater slowing of recovery from inactivation of both variants than did lidocaine (10 µM). Both LAs caused approximately 50% tonic inhibition of aNaV1.5 or nNaV1.5 when holding at -80 mV. Neither LA caused tonic block at a holding potential of either -90 or -120 mV, voltages at which there was little steady-state inactivation. Higher concentrations of either lidocaine (300 µM) or levobupivacaine (100 µM) caused significantly more tonic block at -120 mV. CONCLUSIONS: These data demonstrate that low concentrations of the LAs exhibit inactivation-dependent block of NaV1.5, which may provide a rationale for their use to safely inhibit migration and invasion by metastatic cancer cells without cardiotoxicity.


Assuntos
Anestésicos Locais/farmacologia , Levobupivacaína/farmacologia , Lidocaína/farmacologia , Canal de Sódio Disparado por Voltagem NAV1.5/fisiologia , Bloqueadores dos Canais de Sódio/farmacologia , Adulto , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Células HEK293 , Humanos , Recém-Nascido , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia
16.
Sci Rep ; 7(1): 9969, 2017 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-28855588

RESUMO

The tyrosine kinase, c-Src, participates in mu opioid receptor (MOP) mediated inhibition in sensory neurons in which ß-arrestin2 (ß-arr2) is implicated in its recruitment. Mice lacking ß-arr2 exhibit increased sensitivity to morphine reinforcement; however, whether ß-arr2 and/or c-Src participate in the actions of opioids in neurons within the reward pathway is unknown. It is also unclear whether morphine acts exclusively through MOPs, or involves delta opioid receptors (DOPs). We examined the involvement of MOPs, DOPs, ß-arr2 and c-Src in the inhibition by morphine of GABAergic inhibitory postsynaptic currents (IPSCs) recorded from neurons in the mouse ventral tegmental area. Morphine inhibited spontaneous IPSC frequency, mainly through MOPs, with only a negligible effect remaining in MOP-/- neurons. However, a reduction in the inhibition by morphine for DOP-/- c.f. WT neurons and a DPDPE-induced decrease of IPSC frequency revealed a role for DOPs. The application of the c-Src inhibitor, PP2, to WT neurons also reduced inhibition by morphine, while the inactive PP3, and the MEK inhibitor, SL327, had no effect. Inhibition of IPSC frequency by morphine was also reduced in ß-arr2-/- neurons in which PP2 caused no further reduction. These data suggest that inhibition of IPSCs by morphine involves a ß-arr2/c-Src mediated mechanism.


Assuntos
Analgésicos Opioides/metabolismo , Neurônios GABAérgicos/fisiologia , Morfina/metabolismo , Receptores Opioides mu/agonistas , Área Tegmentar Ventral/efeitos dos fármacos , beta-Arrestina 2/metabolismo , Quinases da Família src/metabolismo , Animais , Proteína Tirosina Quinase CSK , Neurônios GABAérgicos/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Potenciais Sinápticos/efeitos dos fármacos
17.
Anesthesiology ; 127(5): 878-889, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28820778

RESUMO

BACKGROUND: Prolonged opioid administration leads to tolerance characterized by reduced analgesic potency. Pain management is additionally compromised by the hedonic effects of opioids, the cause of their misuse. The multifunctional protein ß-arrestin2 regulates the hedonic effects of morphine and participates in tolerance. These actions might reflect µ opioid receptor up-regulation through reduced endocytosis. ß-Arrestin2 also recruits kinases to µ receptors. We explored the role of Src kinase in morphine analgesic tolerance, locomotor stimulation, and reinforcement in C57BL/6 mice. METHODS: Analgesic (tail withdrawal latency; percentage of maximum possible effect, n = 8 to 16), locomotor (distance traveled, n = 7 to 8), and reinforcing (conditioned place preference, n = 7 to 8) effects of morphine were compared in wild-type, µ, µ, and ß-arrestin2 mice. The influence of c-Src inhibitors dasatinib (n = 8) and PP2 (n = 12) was examined. RESULTS: Analgesia in morphine-treated wild-type mice exhibited tolerance, declining by day 10 to a median of 62% maximum possible effect (interquartile range, 29 to 92%). Tolerance was absent from mice receiving dasatinib. Tolerance was enhanced in µ mice (34% maximum possible effect; interquartile range, 5 to 52% on day 5); dasatinib attenuated tolerance (100% maximum possible effect; interquartile range, 68 to 100%), as did PP2 (91% maximum possible effect; interquartile range, 78 to 100%). By contrast, c-Src inhibition affected neither morphine-evoked locomotor stimulation nor reinforcement. Remarkably, dasatinib not only attenuated tolerance but also reversed established tolerance in µ mice. CONCLUSIONS: The ability of c-Src inhibitors to inhibit tolerance, thereby restoring analgesia, without altering the hedonic effect of morphine, makes c-Src inhibitors promising candidates as adjuncts to opioid analgesics.


Assuntos
Tolerância a Medicamentos/fisiologia , Morfina/farmacologia , Desempenho Psicomotor/fisiologia , Reforço Psicológico , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/metabolismo , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Inibidores de Proteínas Quinases/farmacologia , Desempenho Psicomotor/efeitos dos fármacos
18.
J Physiol ; 595(5): 1725-1741, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-27981574

RESUMO

KEY POINTS: The functional importance of residues in loop G of the GABAA receptor has not been investigated. D43 and T47 in the α1 subunit are of particular significance as their structural modification inhibits activation by GABA. While the T47C substitution had no significant effect, non-conservative substitution of either residue (D43C or T47R) reduced the apparent potency of GABA. Propofol potentiated maximal GABA-evoked currents mediated by α1(D43C)ß2γ2 and α1(T47R)ß2γ2 receptors. Non-stationary variance analysis revealed a reduction in maximal GABA-evoked Popen , suggesting impaired agonist efficacy. Further analysis of α1(T47R)ß2γ2 receptors revealed that the efficacy of the partial agonist THIP (4,5,6,7-tetrahydroisoxazolo[5,4-c]pyridine-3-ol) relative to GABA was impaired. GABA-, THIP- and propofol-evoked currents mediated by α1(T47R)ß2γ2 receptors deactivated faster than those mediated by α1ß2γ2 receptors, indicating that the mutation impairs agonist-evoked gating. Spontaneous gating caused by the ß2(L285R) mutation was also reduced in α1(T47R)ß2(L285R)γ2 compared to α1ß2(L285R)γ2 receptors, confirming that α1(T47R) impairs gating independently of agonist activation. ABSTRACT: The modification of cysteine residues (substituted for D43 and T47) by 2-aminoethyl methanethiosulfonate in the GABAA α1 subunit loop G is known to impair activation of α1ß2γ2 receptors by GABA and propofol. While the T47C substitution had no significant effect, non-conservative substitution of either residue (D43C or T47R) reduced the apparent potency of GABA. Propofol (1 µm), which potentiates sub-maximal but not maximal GABA-evoked currents mediated by α1ß2γ2 receptors, also potentiated maximal currents mediated by α1(D43C)ß2γ2 and α1(T47R)ß2γ2 receptors. Furthermore, the peak open probabilities of α1(D43C)ß2γ2 and α1(T47R)ß2γ2 receptors were reduced. The kinetics of macroscopic currents mediated by α1(D43C)ß2γ2 and α1(T47R)ß2γ2 receptors were characterised by slower desensitisation and faster deactivation. Similar changes in macroscopic current kinetics, together with a slower activation rate, were observed with the loop D α1(F64C) substitution, known to impair both efficacy and agonist binding, and when the partial agonist THIP (4,5,6,7-tetrahydroisoxazolo[5,4-c]pyridine-3-ol) was used to activate WT or α1(T47R)ß2γ2 receptors. Propofol-evoked currents mediated by α1(T47R)ß2γ2 and α1(F64C)ß2γ2 receptors also exhibited faster deactivation than their WT counterparts, revealing that these substitutions impair gating through a mechanism independent of orthosteric binding. Spontaneous gating caused by the introduction of the ß2(L285R) mutation was also reduced in α1(T47R)ß2(L285R)γ2 compared to α1ß2(L285R)γ2 receptors, confirming that α1(T47R) impairs gating independently of activation by any agonist. These findings implicate movement of the GABAA receptor α1 subunit's ß1 strand during agonist-dependent and spontaneous gating. Immobilisation of the ß1 strand may provide a mechanism for the inhibition of gating by inverse agonists such as bicuculline.


Assuntos
Subunidades Proteicas/fisiologia , Receptores de GABA-A/fisiologia , Substituição de Aminoácidos , Animais , GABAérgicos/farmacologia , Células HEK293 , Humanos , Ativação do Canal Iônico/fisiologia , Camundongos , Mutagênese , Propofol/farmacologia , Conformação Proteica , Subunidades Proteicas/química , Subunidades Proteicas/genética , Receptores de GABA-A/química , Receptores de GABA-A/genética , Ácido gama-Aminobutírico/farmacologia
19.
J Physiol ; 594(19): 5555-71, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27195487

RESUMO

KEY POINTS: The role of the ß1 strand in GABAA receptor function is unclear. It lies anti-parallel to the ß2 strand, which is known to participate in receptor activation. Molecular dynamics simulation revealed solvent accessible residues within the ß1 strand of the GABAA ß3 homopentamer that might be amenable to analysis using the substituted Cys accessibility method. Cys substitutions from Asp43 to Thr47 in the GABAA α1 subunit showed that D43C and T47C reduced the apparent potency of GABA. F45C caused a biphasic GABA concentration-response relationship and increased spontaneous gating. Cys43 and Cys47 were accessible to 2-aminoethyl methanethiosulphonate (MTSEA) modification, whereas Cys45 was not. Both GABA and the allosteric agonist propofol reduced MTSEA modification of Cys43 and Cys47. By contrast, modification of Cys64 in the ß2 strand loop D was impeded by GABA but unaffected by propofol. These data reveal movement of ß1 strand loop G residues during agonist activation of the GABAA receptor. ABSTRACT: The GABAA receptor α subunit ß1 strand runs anti-parallel to the ß2 strand, which contains loop D, known to participate in receptor activation and agonist binding. However, a role for the ß1 strand has yet to be established. We used molecular dynamics simulation to quantify the solvent accessible surface area (SASA) of ß1 strand residues in the GABAA ß3 homopentamer structure. Residues in the complementary interface equivalent to those between Asp43 and Thr47 in the α1 subunit have an alternating pattern of high and low SASA consistent with a ß strand structure. We investigated the functional role of these ß1 strand residues in the α1 subunit by individually replacing them with Cys residues. D43C and T47C substitutions reduced the apparent potency of GABA at α1ß2γ2 receptors by 50-fold and eight-fold, respectively, whereas the F45C substitution caused a biphasic GABA concentration-response relationship and increased spontaneous gating. Receptors with D43C or T47C substitutions were sensitive to 2-aminoethyl methanethiosulphonate (MTSEA) modification. However, GABA-evoked currents mediated by α1(F45C)ß2γ2 receptors were unaffected by MTSEA, suggesting that this residue is inaccessible. Both GABA and the allosteric agonist propofol reduced MTSEA modification of α1(D43C)ß2γ2 and α1(T47C)ß2γ2 receptors, indicating movement of the ß1 strand even during allosteric activation. This is in contrast to α1(F64C)ß2γ2 receptors, where only GABA, but not propofol, reduced MTSEA modification. These findings provide the first functional evidence for movement of the ß1 strand during gating of the receptor and identify residues that are critical for maintaining GABAA receptor function.


Assuntos
Receptores de GABA-A/química , Receptores de GABA-A/fisiologia , Metanossulfonato de Etila/análogos & derivados , Metanossulfonato de Etila/farmacologia , Células HEK293 , Humanos , Simulação de Dinâmica Molecular , Propofol/farmacologia , Conformação Proteica em Folha beta , Subunidades Proteicas/química , Subunidades Proteicas/fisiologia , Ácido gama-Aminobutírico/farmacologia
20.
Sci Rep ; 5: 11541, 2015 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-26096612

RESUMO

Functional expression of voltage-gated Na(+) channels (VGSCs) has been demonstrated in multiple cancer cell types where channel activity induces invasive activity. The signaling mechanisms by which VGSCs promote oncogenesis remain poorly understood. We explored the signal transduction process critical to VGSC-mediated invasion on the basis of reports linking channel activity to gene expression changes in excitable cells. Coincidentally, many genes transcriptionally regulated by the SCN5A isoform in colon cancer have an over-representation of cis-acting sites for transcription factors phosphorylated by ERK1/2 MAPK. We hypothesized that VGSC activity promotes MAPK activation to induce transcriptional changes in invasion-related genes. Using pharmacological inhibitors/activators and siRNA-mediated gene knockdowns, we correlated channel activity with Rap1-dependent persistent MAPK activation in the SW620 human colon cancer cell line. We further demonstrated that VGSC activity induces downstream changes in invasion-related gene expression via a PKA/ERK/c-JUN/ELK-1/ETS-1 transcriptional pathway. This is the first study illustrating a molecular mechanism linking functional activity of VGSCs to transcriptional activation of invasion-related genes.


Assuntos
Neoplasias do Colo/genética , Sistema de Sinalização das MAP Quinases/genética , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Invasividade Neoplásica/genética , Transcrição Gênica/genética , Linhagem Celular Tumoral , Neoplasias do Colo/patologia , Ativação Enzimática , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Transporte de Íons/genética , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Interferência de RNA , RNA Interferente Pequeno , Complexo Shelterina , Proteínas de Ligação a Telômeros/metabolismo , Veratridina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA