Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Antibiotics (Basel) ; 11(9)2022 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-36139979

RESUMO

The rapid worldwide spread of antimicrobial resistance highlights the significant need for the development of innovative treatments to fight multidrug-resistant bacteria. This study describes the potent antimicrobial activity of the novel peptide OMN6 against a wide array of drug-resistant Acinetobacter baumannii clinical isolates. OMN6 prevented the growth of all tested isolates, regardless of any pre-existing resistance mechanisms. Moreover, in vitro serial-passaging studies demonstrated that no resistance developed against OMN6. Importantly, OMN6 was highly efficacious in treating animal models of lung and blood infections caused by multidrug-resistant A. baumannii. Taken together, these results point to OMN6 as a novel antimicrobial agent with the potential to treat life-threatening infections caused by multidrug-resistant A. baumannii avoiding resistance.

2.
Sci Rep ; 11(1): 6603, 2021 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-33758343

RESUMO

New antimicrobial agents are urgently needed, especially to eliminate multidrug resistant Gram-negative bacteria that stand for most antibiotic-resistant threats. In the following study, we present superior properties of an engineered antimicrobial peptide, OMN6, a 40-amino acid cyclic peptide based on Cecropin A, that presents high efficacy against Gram-negative bacteria with a bactericidal mechanism of action. The target of OMN6 is assumed to be the bacterial membrane in contrast to small molecule-based agents which bind to a specific enzyme or bacterial site. Moreover, OMN6 mechanism of action is effective on Acinetobacter baumannii laboratory strains and clinical isolates, regardless of the bacteria genotype or resistance-phenotype, thus, is by orders-of-magnitude, less likely for mutation-driven development of resistance, recrudescence, or tolerance. OMN6 displays an increase in stability and a significant decrease in proteolytic degradation with full safety margin on erythrocytes and HEK293T cells. Taken together, these results strongly suggest that OMN6 is an efficient, stable, and non-toxic novel antimicrobial agent with the potential to become a therapy for humans.


Assuntos
Acinetobacter baumannii/efeitos dos fármacos , Antibacterianos/farmacologia , Proteínas Citotóxicas Formadoras de Poros/farmacologia , Antibacterianos/química , Peptídeos Catiônicos Antimicrobianos/química , Membrana Externa Bacteriana/efeitos dos fármacos , Farmacorresistência Bacteriana Múltipla , Proteínas Citotóxicas Formadoras de Poros/química , Engenharia de Proteínas , Estabilidade Proteica
3.
Dev Cell ; 28(3): 310-21, 2014 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-24525187

RESUMO

Membrane lipid regulation of cell function is poorly understood. In early development, sterol efflux and the ganglioside GM1 regulate sperm acrosome exocytosis (AE) and fertilization competence through unknown mechanisms. Here, we show that sterol efflux and focal enrichment of GM1 trigger Ca(2+) influx necessary for AE through CaV2.3, whose activity has been highly controversial in sperm. Sperm lacking CaV2.3's pore-forming α1E subunit showed altered Ca(2+) responses, reduced AE, and a strong subfertility phenotype. Surprisingly, AE depended on spatiotemporal information encoded by flux through CaV2.3, not merely the presence/amplitude of Ca(2+) waves. Using studies in both sperm and voltage clamp of Xenopus oocytes, we define a molecular mechanism for GM1/CaV2.3 regulatory interaction, requiring GM1's lipid and sugar components and CaV2.3's α1E and α2δ subunits. Our results provide a mechanistic understanding of membrane lipid regulation of Ca(2+) flux and therefore Ca(2+)-dependent cellular and developmental processes such as exocytosis and fertilization.


Assuntos
Acrossomo/metabolismo , Canais de Cálcio Tipo R/fisiologia , Cálcio/metabolismo , Proteínas de Transporte de Cátions/fisiologia , Exocitose/fisiologia , Fertilização/fisiologia , Gangliosídeo G(M1)/farmacologia , Espermatozoides/metabolismo , Acrossomo/efeitos dos fármacos , Animais , Células Cultivadas , Exocitose/efeitos dos fármacos , Fertilização/efeitos dos fármacos , Masculino , Camundongos , Oócitos/citologia , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Espermatozoides/citologia , Xenopus laevis/crescimento & desenvolvimento , Xenopus laevis/metabolismo
4.
Redox Biol ; 2: 447-56, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24624334

RESUMO

Diabetes is a high risk factor for dementia. High glucose may be a risk factor for dementia even among persons without diabetes, and in transgenic animals it has been shown to cause a potentiation of indices that are pre-symptomatic of Alzheimer's disease. To further elucidate the underlying mechanisms linking inflammatory events elicited in the brain during oxidative stress and diabetes, we monitored the activation of mitogen-activated kinsase (MAPKs), c-jun NH2-terminal kinase (JNK), p38 MAP kinases (p38(MAPK)), and extracellular activating kinsae1/2 (ERK1/2) and the anti-inflammatory effects of the thioredoxin mimetic (TxM) peptides, Ac-Cys-Pro-Cys-amide (CB3) and Ac-Cys-Gly-Pro-Cys-amide (CB4) in the brain of male leptin-receptor-deficient Zucker diabetic fatty (ZDF) rats and human neuroblastoma SH-SY5Y cells. Daily i.p. injection of CB3 to ZDF rats inhibited the phosphorylation of JNK and p38(MAPK), and prevented the expression of thioredoxin-interacting-protein (TXNIP/TBP-2) in ZDF rat brain. Although plasma glucose/insulin remained high, CB3 also increased the phosphorylation of AMP-ribose activating kinase (AMPK) and inhibited p70(S6K) kinase in the brain. Both CB3 and CB4 reversed apoptosis induced by inhibiting thioredoxin reductase as monitored by decreasing caspase 3 cleavage and PARP dissociation in SH-SY5Y cells. The decrease in JNK and p38(MAPK) activity in the absence of a change in plasma glucose implies a decrease in oxidative or neuroinflammatory stress in the ZDF rat brain. CB3 not only attenuated MAPK phosphorylation and activated AMPK in the brain, but it also diminished apoptotic markers, most likely acting via the MAPK-AMPK-mTOR pathway. These results were correlated with CB3 and CB4 inhibiting inflammation progression and protection from oxidative stress induced apoptosis in human neuronal cells. We suggest that by attenuating neuro-inflammatory processes in the brain Trx1 mimetic peptides could become beneficial for preventing neurological disorders associated with diabetes.


Assuntos
Encéfalo/metabolismo , Diabetes Mellitus Tipo 2/tratamento farmacológico , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Obesidade/tratamento farmacológico , Oligopeptídeos/administração & dosagem , Peptidomiméticos/administração & dosagem , Compostos de Sulfidrila/administração & dosagem , Animais , Glicemia/metabolismo , Linhagem Celular Tumoral , Diabetes Mellitus Tipo 2/metabolismo , Modelos Animais de Doenças , Humanos , Insulina/sangue , Masculino , Obesidade/metabolismo , Oligopeptídeos/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Peptidomiméticos/farmacologia , Fosforilação , Ratos , Ratos Zucker , Compostos de Sulfidrila/farmacologia
5.
Sci Rep ; 3: 1620, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23567899

RESUMO

The interaction of syntaxin 1A (Sx1A) with voltage-gated calcium channels (VGCC) is required for depolarization-evoked release. However, it is unclear how the signal is transferred from the channel to the exocytotic machinery and whether assembly of Sx1A and the calcium channel is conformationally linked to triggering synchronous release. Here we demonstrate that depolarization-evoked catecholamine release was decreased in chromaffin cells infected with semliki forest viral vectors encoding Sx1A mutants, Sx1A(C271V), or Sx1A(C272V), or by direct oxidation of these Sx1A transmembrane (TM) cysteine residues. Mutating or oxidizing these highly conserved Sx1A Cys271 and Cys272 equally disrupted the Sx1A interaction with the channel. The results highlight the functional link between the VGCC and the exocytotic machinery, and attribute the redox sensitivity of the release process to the Sx1A TM C271 and C272. This unique intra-membrane signal-transduction pathway enables fast signaling, and triggers synchronous release by conformational-coupling of the channel with Sx1A.


Assuntos
Canais de Cálcio/metabolismo , Membrana Celular/metabolismo , Cisteína/metabolismo , Transdução de Sinais , Sintaxina 1/metabolismo , Sequência de Aminoácidos , Animais , Canais de Cálcio/genética , Canais de Cálcio Tipo L/genética , Canais de Cálcio Tipo L/metabolismo , Bovinos , Células Cromafins/metabolismo , Exocitose , Expressão Gênica , Humanos , Potenciais da Membrana , Modelos Biológicos , Mutação , Oócitos/metabolismo , Oxirredução , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Sintaxina 1/química , Sintaxina 1/genética
6.
Biochem Pharmacol ; 85(7): 977-90, 2013 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-23327993

RESUMO

The thioredoxin reductase/thioredoxin system (TrxR/Trx1) plays a major role in protecting cells from oxidative stress. Disruption of the TrxR-Trx1 system keeps Trx1 in the oxidized state leading to cell death through activation of the ASK1-Trx1 apoptotic pathway. The potential mechanism and ability of tri- and tetra-oligopeptides derived from the canonical -CxxC- motif of the Trx1-active site to mimic and enhance Trx1 cellular activity was examined. The Trx mimetics peptides (TXM) protected insulinoma INS 832/13 cells from oxidative stress induced by selectively inhibiting TrxR with auranofin (AuF). TXM reversed the AuF-effects preventing apoptosis, and increasing cell-viability. The TXM peptides were effective in inhibiting AuF-induced MAPK, JNK and p38(MAPK) phosphorylation, in correlation with preventing caspase-3 cleavage and thereby PARP-1 dissociation. The ability to form a disulfide-bridge-like conformation was estimated from molecular dynamics simulations. The TXM peptides restored insulin secretion and displayed Trx1 denitrosylase activity. Their potency was 10-100-fold higher than redox reagents like NAC, AD4, or ascorbic acid. Unable to reverse ERK1/2 phosphorylation, TXM-CB3 (NAc-Cys-Pro-Cys amide) appeared to function in part, through inhibiting ASK1-Trx dissociation. These highly effective anti-apoptotic effects of Trx1 mimetic peptides exhibited in INS 832/13 cells could become valuable in treating adverse oxidative-stress related disorders such as diabetes.


Assuntos
Apoptose/efeitos dos fármacos , Auranofina/farmacologia , Insulina/metabolismo , Oligopeptídeos/farmacologia , Peptidomiméticos/farmacologia , Tiorredoxinas/metabolismo , Animais , Caspase 3/metabolismo , Domínio Catalítico , Linhagem Celular Tumoral , Radicais Livres/metabolismo , Secreção de Insulina , Insulinoma , Janus Quinases/metabolismo , MAP Quinase Quinase Quinase 5/metabolismo , Simulação de Dinâmica Molecular , Oligopeptídeos/química , Estresse Oxidativo , Peptidomiméticos/química , Fosforilação , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases/metabolismo , Ratos , Relação Estrutura-Atividade , Tiorredoxina Dissulfeto Redutase/antagonistas & inibidores , Tiorredoxinas/química , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
7.
Sci Rep ; 2: 366, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22511998

RESUMO

Ca(2+)-entry in the heart is tightly controlled by Cav1.2 inactivation, which involves Ca(2+)-dependent inactivation (CDI) and voltage-dependent inactivation (VDI) components. Timothy syndrome, a subtype-form of congenital long-QT syndrome, results from a nearly complete elimination of VDI by the G406R mutation in the α(1)1.2 subunit of Cav1.2. Here, we show that a single (A1929P) or a double mutation (H1926A-H1927A) within the CaN-binding site at the human C-terminal tail of α(1)1.2, accelerate the inactivation rate and enhances VDI of both wt and Timothy channels. These results identify the CaN-binding site as the long-sought VDI-regulatory motif of the cardiac channel. The substantial increase in VDI and the accelerated inactivation caused by the selective inhibitors of CaN, cyclosporine A and FK-506, which act at the same CaN-binding site, further support this conclusion. A reversal of enhanced-sympathetic tone by VDI-enhancing CaN inhibitors could be beneficial for improving Timothy syndrome complications such as long-QT and autism.

8.
Int J Endocrinol ; 2011: 746482, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22216029

RESUMO

A PKA consensus phosphorylation site S1928 at the α(1)1.2 subunit of the rabbit cardiac L-type channel, Ca(V)1.2, is involved in the regulation of Ca(V)1.2 kinetics and affects catecholamine secretion. This mutation does not alter basal Ca(V)1.2 current properties or regulation of Ca(V)1.2 current by PKA and the beta-adrenergic receptor, but abolishes Ca(V)1.2 phosphorylation by PKA. Here, we test the contribution of the corresponding PKA phosphorylation site of the human α(1)1.2 subunit S1898, to the regulation of catecholamine secretion in bovine chromaffin cells. Chromaffin cells were infected with a Semliki-Forest viral vector containing either the human wt or a mutated S1898A α(1)1.2 subunit. Both subunits harbor a T1036Y mutation conferring nifedipine insensitivity. Secretion evoked by depolarization in the presence of nifedipine was monitored by amperometry. Depolarization-triggered secretion in cells infected with either the wt α(1)1.2 or α(1)1.2/S1898A mutated subunit was elevated to a similar extent by forskolin. Forskolin, known to directly activate adenylyl-cyclase, increased the rate of secretion in a manner that is largely independent of the presence of S1898. Our results are consistent with the involvement of additional PKA regulatory site(s) at the C-tail of α(1)1.2, the pore forming subunit of Ca(V)1.2.

9.
Channels (Austin) ; 4(4): 266-77, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20495360

RESUMO

It is well-established that syntaxin 1A (Sx1A), SNAP-25 and synaptotagmin (Syt1) either alone or in combination, modify the kinetic properties of voltage-gated Ca(2+) channels (VGCCs). The interaction interface resides mainly at the cytosolic II-III domain of the alpha1 subunit of the channels, while Sx1A interacts with the channel also via two highly conserved cysteine residues at the transmembrane domain. In the present study, we characterized Ca(2+)-independent coupling of the human neuronal P/Q-type calcium channel (Ca(V)2.1) with Sx1A, SNAP-25, Syt1 and synaptobrevin (VAMP) in BAPTA-injected Xenopus oocytes. The co-expression of Ca(V)2.1 with Sx1A, SNAP-25 and Syt1, produced a multiprotein complex with distinctive kinetic properties analogous to the excitosome complexes generated by Ca(V)1.2, Ca(V)2.2 and Ca(V)2.3. The distinct kinetic properties of Ca(V)2.1 acquired by close association with Syt1 and t-SNAREs, suggests that the vesicle is tethered to the neuronal channel and to the exocytotic machinery independently of intracellular Ca(2+). To explore the relevance of these interactions to secretion we exploited a BotC1-and a BotA-sensitive secretion system developed for Xenopus oocytes not buffered by BAPTA, in which depolarization-evoked secretion is monitored by a change in membrane capacitance. The reconstituted release mediated by Ca(V)2.1 is consistent with the model in which the VGCC plays a signaling role in triggering release, acting from within the exocytotic complex. The relevance of these results to secretion posits the role of possible rearrangements within the excitosome subsequent to Ca(2+) entry, setting the stage for the fusion of channel-tethered-vesicles upon the arrival of an action potential.


Assuntos
Canais de Cálcio Tipo N/metabolismo , Canais de Cálcio/metabolismo , Sinalização do Cálcio , Membrana Celular/metabolismo , Exocitose , Ativação do Canal Iônico , Proteínas do Tecido Nervoso/metabolismo , Transmissão Sináptica , Animais , Toxinas Botulínicas/farmacologia , Toxinas Botulínicas Tipo A/farmacologia , Canais de Cálcio/genética , Canais de Cálcio Tipo N/genética , Sinalização do Cálcio/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Quelantes/farmacologia , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Capacitância Elétrica , Potenciais Evocados , Exocitose/efeitos dos fármacos , Feminino , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Cinética , Microinjeções , Proteínas do Tecido Nervoso/genética , Oócitos , Técnicas de Patch-Clamp , Proteínas R-SNARE/metabolismo , Coelhos , Ratos , Transmissão Sináptica/efeitos dos fármacos , Proteína 25 Associada a Sinaptossoma/metabolismo , Sinaptotagmina I/metabolismo , Sintaxina 1/metabolismo , Xenopus laevis
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa