Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Channels (Austin) ; 18(1): 2341077, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38601983

RESUMO

Voltage-gated calcium channels (VGCCs) are the major conduits for calcium ions (Ca2+) within excitable cells. Recent studies have highlighted the non-ionotropic functionality of VGCCs, revealing their capacity to activate intracellular pathways independently of ion flow. This non-ionotropic signaling mode plays a pivotal role in excitation-coupling processes, including gene transcription through excitation-transcription (ET), synaptic transmission via excitation-secretion (ES), and cardiac contraction through excitation-contraction (EC). However, it is noteworthy that these excitation-coupling processes require extracellular calcium (Ca2+) and Ca2+ occupancy of the channel ion pore. Analogous to the "non-canonical" characterization of the non-ionotropic signaling exhibited by the N-methyl-D-aspartate receptor (NMDA), which requires extracellular Ca2+ without the influx of ions, VGCC activation requires depolarization-triggered conformational change(s) concomitant with Ca2+ binding to the open channel. Here, we discuss the contributions of VGCCs to ES, ET, and EC coupling as Ca2+ binding macromolecules that transduces external stimuli to intracellular input prior to elevating intracellular Ca2+. We emphasize the recognition of calcium ion occupancy within the open ion-pore and its contribution to the excitation coupling processes that precede the influx of calcium. The non-ionotropic activation of VGCCs, triggered by the upstroke of an action potential, provides a conceptual framework to elucidate the mechanistic aspects underlying the microseconds nature of synaptic transmission, cardiac contractility, and the rapid induction of first-wave genes.


Assuntos
Canais de Cálcio , Cálcio , Cálcio/metabolismo , Canais de Cálcio/metabolismo , Transdução de Sinais , Acoplamento Excitação-Contração , Íons/metabolismo , Sinalização do Cálcio/fisiologia , Canais de Cálcio Tipo L/metabolismo
2.
Antioxidants (Basel) ; 13(3)2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38539884

RESUMO

Albumin (HSA) is the most abundant circulating protein and plays a pivotal role in maintaining the redox state of the plasma. Three HSA proteoforms have been identified based on the redox state of cysteine 34. These proteoforms comprise of the reduced state (HSA-SH) referred to as mercaptoalbumin, non-mercaptoalbumin-1, containing a disulfide with small thiols such as cysteine (HSA-Cys), and non-mercaptoalbumin-2, representing the higher oxidized proteoform. Several clinical studies have shown a relationship between an individual's serum HSA redox status and the severity of diseases such as heart failure, diabetes mellitus, and liver disease. Furthermore, when HSA undergoes oxidation, it can worsen certain health conditions and contribute to their advancement. This study aimed to evaluate the ability of the redox compounds AD4/NACA and the thioredoxin mimetic (TXM) peptides TXM-CB3, TXM-CB13, and TXM-CB30 to regenerate HSA-SH and to enhance its redox activity. The HSA proteoforms were quantified by LC-MS, and the antioxidant activity was determined using dichlorofluorescin. Each of the compounds exhibited a significant increase in HSA-SH and a reduction in HSA-Cys levels. The increase in HSA-SH was associated with a recovery of its antioxidant activity. In this work, we unveil a novel mechanistic facet of the antioxidant activity of AD4/NACA and TXM peptides. These results suggest an additional therapeutic approach for addressing oxidative stress-related conditions.

3.
Proc Natl Acad Sci U S A ; 121(8): e2317343121, 2024 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-38359293

RESUMO

Glucose and amino acid metabolism are critical for glioblastoma (GBM) growth, but little is known about the specific metabolic alterations in GBM that are targetable with FDA-approved compounds. To investigate tumor metabolism signatures unique to GBM, we interrogated The Cancer Genome Atlas for alterations in glucose and amino acid signatures in GBM relative to other human cancers and found that GBM exhibits the highest levels of cysteine and methionine pathway gene expression of 32 human cancers. Treatment of patient-derived GBM cells with the FDA-approved single cysteine compound N-acetylcysteine (NAC) reduced GBM cell growth and mitochondrial oxygen consumption, which was worsened by glucose starvation. Normal brain cells and other cancer cells showed no response to NAC. Mechanistic experiments revealed that cysteine compounds induce rapid mitochondrial H2O2 production and reductive stress in GBM cells, an effect blocked by oxidized glutathione, thioredoxin, and redox enzyme overexpression. From analysis of the clinical proteomic tumor analysis consortium (CPTAC) database, we found that GBM cells exhibit lower expression of mitochondrial redox enzymes than four other cancers whose proteomic data are available in CPTAC. Knockdown of mitochondrial thioredoxin-2 in lung cancer cells induced NAC susceptibility, indicating the importance of mitochondrial redox enzyme expression in mitigating reductive stress. Intraperitoneal treatment of mice bearing orthotopic GBM xenografts with a two-cysteine peptide induced H2O2 in brain tumors in vivo. These findings indicate that GBM is uniquely susceptible to NAC-driven reductive stress and could synergize with glucose-lowering treatments for GBM.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Humanos , Camundongos , Animais , Peróxido de Hidrogênio , Peróxidos , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Glioblastoma/metabolismo , Proteômica , Acetilcisteína/farmacologia , Glucose , Linhagem Celular Tumoral , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética
4.
Antiviral Res ; 222: 105806, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-38211737

RESUMO

After three years of the SARS-CoV-2 pandemic, the search and availability of relatively low-cost benchtop therapeutics for people not at high risk for a severe disease are still ongoing. Although vaccines and new SARS-CoV-2 variants reduce the death toll, the long COVID-19 along with neurologic symptoms can develop and persist even after a mild initial infection. Reinfections, which further increase the risk of sequelae in multiple organ systems as well as the risk of death, continue to require caution. The spike protein of SARS-CoV-2 is an important target for both vaccines and therapeutics. The presence of disulfide bonds in the receptor binding domain (RBD) of the spike protein is essential for its binding to the human ACE2 receptor and cell entry. Here, we demonstrate that thiol-reducing peptides based on the active site of oxidoreductase thioredoxin 1, called thioredoxin mimetic (TXM) peptides, can prevent syncytia formation, SARS-CoV-2 entry into cells, and infection in a mouse model. We also show that TXM peptides inhibit the redox-sensitive HIV pseudotyped viral cell entry. These results support disulfide targeting as a common therapeutic strategy for treating infections caused by viruses using redox-sensitive fusion. Furthermore, TXM peptides exert anti-inflammatory properties by lowering the activation of NF-κB and IRF signaling pathways, mitogen-activated protein kinases (MAPKs) and lipopolysaccharide (LPS)-induced cytokines in mice. The antioxidant and anti-inflammatory effects of the TXM peptides, which also cross the blood-brain barrier, in combination with prevention of viral infections, may provide a beneficial clinical strategy to lower viral infections and mitigate severe consequences of COVID-19.


Assuntos
COVID-19 , Vacinas , Animais , Humanos , Camundongos , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus , Síndrome de COVID-19 Pós-Aguda , Peptídeos/farmacologia , Vacinas/farmacologia , Tiorredoxinas/química , Tiorredoxinas/metabolismo , Tiorredoxinas/farmacologia , Anti-Inflamatórios/farmacologia , Dissulfetos/farmacologia , Células Gigantes , Ligação Proteica
5.
Antioxidants (Basel) ; 12(7)2023 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-37507934

RESUMO

In the present study, we tested the effect of small-molecular-weight redox molecules on collagen-induced platelet aggregation. We used N-acetylcysteine amide (AD4/NACA), the amide form of N-acetylcysteine (NAC), a thiol antioxidant with improved lipophilicity and bioavailability compared to NAC, and the thioredoxin-mimetic (TXM) peptides, TXM-CB3, TXM-CB13, and TXM-CB30. All compounds significantly inhibited platelet aggregation induced by collagen, with TXM-peptides and AD4 being more effective than NAC. The levels of TxB2 and 12-HETE, the main metabolites derived from the cyclooxygenase and lipoxygenase pathways following platelet activation, were significantly reduced in the presence of AD4, TXM peptides, or NAC, when tested at the highest concentration (0.6 mM). The effects of AD4, TXM-peptides, and NAC were also tested on the clotting time (CT) of whole blood. TXM-CB3 and TXM-CB30 showed the greatest increase in CT. Furthermore, two representative compounds, TXM-CB3 and NAC, showed an increase in the anti-oxidant free sulfhydryl groups of plasma detected via Ellman's method, suggesting a contribution of plasma factors to the antiaggregating effects. Our results suggest that these small-molecular-weight redox peptides might become useful for the prevention and/or treatment of oxidative stress conditions associated with platelet activation.

6.
Cell Calcium ; 108: 102672, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36427431

RESUMO

Membrane depolarization triggers gene expression through voltage-gated calcium channels (VGCC) in a process called Excitation-transcription (ET) coupling. Mutations in the channel subunits α11.2, or ß2d, are associated with neurodevelopmental disorders such as ASD. Here, we found that two mutations S143F and G113S within the rat Cavß2a corresponding to autistic related mutations Cavß2dS197F and Cavß2dG167S in the human Cavß2d, activate ET-coupling via the RAS/ERK/CREB pathway. Membrane depolarization of HEK293 cells co-expressing α11.2 and α2δ with Cavß2aS143F or Cavß2aG113S triggers constitutive transcriptional activation, which is correlated with facilitated channel activity. Similar to the Timothy-associated autistic mutation α11.2G406R, constitutive gene activation is attributed to a hyperpolarizing shift in the activation kinetics of Cav1.2. Pulldown of RasGRF2 and RhoGEF by wt and the Cavß2a autistic mutants is consistent with Cavß2/Ras activation in ET coupling and implicates Rho signaling as yet another molecular pathway activated by Cavα11.2/Cavß2 . Facilitated spontaneous channel activity preceding enhanced gene activation via the Ras/ERK/CREB pathway, appears a general molecular mechanism for Ca2+ channel mediated ASD and other neurodevelopmental disorders.


Assuntos
Transtorno Autístico , Canais de Cálcio Tipo L , Animais , Humanos , Ratos , Transtorno Autístico/genética , Expressão Gênica , Células HEK293 , Mutação , Canais de Cálcio Tipo L/genética
7.
Prog Neurobiol ; 216: 102312, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35760141

RESUMO

Measurements of the time elapsed during synaptic transmission has shown that synaptic vesicle (SV) fusion lags behind Ca2+-influx by approximately 60 microseconds (µsec). The conventional model cannot explain this extreme rapidity of the release event. Synaptic transmission occurs at the active zone (AZ), which comprises of two pools of SV, non-releasable "tethered" vesicles, and a readily-releasable pool of channel-associated Ca2+-primed vesicles, "RRP". A recent TIRF study at cerebellar-mossy fiber-terminal, showed that subsequent to an action potential, newly "tethered" vesicles, became fusion-competent in a Ca2+-dependent manner, 300-400 ms after tethering, but were not fused. This time resolution may correspond to priming of tethered vesicles through Ca2+-binding to Syt1/Munc13-1/complexin. It confirms that Ca2+-priming and Ca2+-influx-independent fusion, are two distinct events. Notably, we have established that Ca2+ channel signals evoked-release in an ion flux-independent manner, demonstrated by Ca2+-impermeable channel, or by substitution of Ca2+ with channel -impermeable La3+. Thus, conformational changes in a channel coupled to RRP appear to directly activate the release machinery and account for a µsec Ca2+-influx-independent vesicle fusion. Rapid vesicle fusion driven by non-ionotropic channel signaling strengthens a conformational-coupling mechanism of synaptic transmission, and contributes to better understanding of neuronal communication vital for brain function.


Assuntos
Transmissão Sináptica , Vesículas Sinápticas , Cálcio , Exocitose/fisiologia , Humanos , Neurônios , Transmissão Sináptica/fisiologia , Vesículas Sinápticas/fisiologia
8.
Cell Death Dis ; 13(3): 227, 2022 03 11.
Artigo em Inglês | MEDLINE | ID: mdl-35277478

RESUMO

Parkinson's disease (PD) is characterized by a gradual degeneration of the dopaminergic neurons in the substantia nigra pars compacta (SNpC). Levodopa, the standard PD treatment, provides the missing dopamine in SNpC, but ultimately after a honeymoon with levodopa treatment the neurodegenerative process and the progression of the disease continue. Aimed at prolonging the life of dopaminergic cells, we prepared the levodopa precursors SuperDopa (SD) and SueprDopamide (SDA), in which levodopa is merged with the antioxidant N-acetylcysteine (NAC) into a single molecule. Rotenone is a mitochondrial complex inhibitor often used as experimental model of PD. In vivo, SD and SDA treatment show a significant relief of motor disabilities in rotenone-injected rats. SD and SDA also lower rotenone-induced-α-synuclein (α-syn) expression in human SH-SY5Y cells, and α-syn oligomerization in α-syn-overexpressing-HEK293 cells. In the neuronal SH-SY5Y cells, SD and SDA reverse oxidative stress-induced phosphorylation of cJun-N-terminal kinase (JNK) and p38-mitogen-activated kinase (p38MAPK). Attenuation of the MAPK-inflammatory/apoptotic pathway in SH-SY5Y cells concurrent with protection of rotenone-triggered motor impairment in rats, is a manifestation of the combined antioxidant/anti-inflammatory activity of SD and SDA together with levodopa release. The concept of joined therapies into a single molecule, where levodopa precursors confer antioxidant activity by enabling NAC delivery across the BBB, provides a potential disease-modifying treatment for slowing PD progression.


Assuntos
Fármacos Neuroprotetores , Doença de Parkinson , Animais , Antioxidantes/metabolismo , Neurônios Dopaminérgicos/metabolismo , Células HEK293 , Humanos , Levodopa/metabolismo , Levodopa/farmacologia , Fármacos Neuroprotetores/metabolismo , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/metabolismo , Ratos , Rotenona/farmacologia
9.
Free Radic Biol Med ; 176: 120-141, 2021 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-34481041

RESUMO

Understanding neurodegenerative diseases have challenged scientists for decades. It has become apparent that a decrease in life span is often correlated with the development of neurodegenerative disorders. Oxidative stress and the subsequent inflammatory damages appear to contribute to the different molecular and biochemical mechanisms associated with neurodegeneration. In this review, I examine the protective properties of novel amino acid based compounds, comprising the AD series (AD1-AD7) in particular N-acetylcysteine amide, AD4, also called NACA, and the series of thioredoxin mimetic (TXM) peptides, TXM-CB3-TXM-CB16. Designed to cross the blood-brain-barrier (BBB) and permeate the cell membrane, these antioxidant/anti-inflammatory compounds may enable effective treatment of neurodegenerative related disorders. The review addresses the molecular mechanism of cellular protection exhibited by these new reagents, focusing on the reversal of oxidative stress, mitochondrial stress, inflammatory damages, and prevention of premature cell death. In addition, it will cover the outlook of the clinical prospects of AD4/NACA and the thioredoxin-mimetic peptides, which are currently in development.


Assuntos
Doenças Neurodegenerativas , Compostos de Sulfidrila , Acetilcisteína/análogos & derivados , Amidas , Humanos , Doenças Neurodegenerativas/tratamento farmacológico , Peptídeos , Tiorredoxinas
10.
Prog Neurobiol ; 191: 101820, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32437834

RESUMO

Timothy syndrome (TS) is a neurodevelopmental disorder caused by mutations in the pore-forming subunit α11.2 of the L-type voltage-gated Ca2+-channel Cav1.2, at positions G406R or G402S. Although both mutations cause cardiac arrhythmias, only Cav1.2G406R is associated with the autism-spectrum-disorder (ASD). We show that transcriptional activation by Cav1.2G406R and Cav1.2G402S is driven by membrane depolarization through the Ras/ERK/CREB pathway in a process called excitation-transcription (ET) coupling, as previously shown for wt Cav1.2. This process requires the presence of the intracellular ß-subunit of the channel. We found that only the autism-associated mutant Cav1.2G406R, as opposed to the non-autistic mutated channel Cav1.2G402S, exhibits a depolarization-independent CREB phosphorylation, and spontaneous transcription of cFos and MeCP2. A leftward voltage-shift typical of Cav1.2G406R activation, increases channel opening at subthreshold potentials, resulting in an enhanced channel activity, as opposed to a rightward shift in Cav1.2G402S. We suggest that the enhanced spontaneous Cav1.2G406R activity accounts for the increase in basal transcriptional activation. This uncontroled transcriptional activation may result in the manifestation of long-term dysregulations such as autism. Thus, gating changes provide a mechanistic framework for understanding the molecular events underlying the autistic phenomena caused by the G406R Timothy mutation. They might clarify whether a constitutive transcriptional activation accompanies other VGCC that exhibit a leftward voltage-shift of activation and are also associated with long-term cognitive disorders.


Assuntos
Transtorno do Espectro Autista , Canais de Cálcio Tipo L/fisiologia , Transdução de Sinais/fisiologia , Ativação Transcricional/fisiologia , Transtorno do Espectro Autista/genética , Transtorno do Espectro Autista/metabolismo , Transtorno do Espectro Autista/fisiopatologia , Transtorno Autístico/genética , Transtorno Autístico/metabolismo , Transtorno Autístico/fisiopatologia , Canais de Cálcio Tipo L/genética , Canais de Cálcio Tipo L/metabolismo , Células HEK293 , Humanos , Síndrome do QT Longo/genética , Síndrome do QT Longo/metabolismo , Síndrome do QT Longo/fisiopatologia , Mutação , Transdução de Sinais/genética , Sindactilia/genética , Sindactilia/metabolismo , Sindactilia/fisiopatologia , Ativação Transcricional/genética
11.
Cell Calcium ; 84: 102102, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31733625

RESUMO

During membrane depolarization the voltage-gated calcium channel (VGCC) activates gene expression in excitable cells by means of a signal-transduction pathway termed excitation transcription (ET) coupling. The L-type calcium channel Cav1.2 can drive nuclear activity by either the ERK-CREB pathway, the Ca2+/calmodulin-dependent protein kinase II (CaMKII) cascade, or via the Ca2+-dependent protein phosphatase calcineurin. The ERK-CREB pathway mediates nuclear activity via a direct interaction of the intracellular ß subunit of VGCC with the Ras/GRF1 complex. Here we show that ET coupling in HEK293 cells transfected with wt Cav1.2 or the Timothy mutant Cav1.2G406R is mediated by substituting Ca2+ with the impermeable lanthanum (La3+). In the absence of extracellular Ca2+ or La3+, ET coupling was not triggered. This implies that cation occupancy of the selectivity filter, as opposed to calcium influx, plays an essential role in depolarization triggered signaling to the nucleus. ET coupling triggered by membrane depolarization in Cav1.2 transfected HEK293 cells and neuroendocrine PC12 cells was also supported by substituting Ba2+ for Ca2+ as the charge carrier. Since Ba2+ ions do not bind to calmodulin this implies activation of ET coupling via a Ca2+/calmodulin-independent pathway. Together, these results suggest a model whereby nuclear signaling through the ERK-CREB pathway is driven by voltage-dependent conformational change that requires channel pore occupancy and is Ca2+ influx-independent. This model is also consistent with the previous observation that ET coupling can be driven by the Ca2+-impermeable Cav1.2L745P mutant. Thus, the conversion of synaptic stimuli to transcriptional activation is mediated by the metabotropic function (Ca2+-inflow independent) of Cav1.2, similar to the ion-influx independent depolarization-triggered transmitter release and transcription activation mediated by the NMDA receptors.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Cálcio/química , Íons/química , Lantânio/química , Calcineurina/metabolismo , Cálcio/metabolismo , Canais de Cálcio Tipo L/química , Canais de Cálcio Tipo L/genética , Sinalização do Cálcio , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Calmodulina , Acoplamento Excitação-Contração , Células HEK293 , Humanos , Sistema de Sinalização das MAP Quinases , Potenciais da Membrana , Relação Estrutura-Atividade , Ativação Transcricional
12.
Proc Natl Acad Sci U S A ; 115(37): E8624-E8633, 2018 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-30150369

RESUMO

Depolarization-induced signaling to the nucleus by the L-type voltage-gated calcium channel Cav1.2 is widely assumed to proceed by elevating intracellular calcium. The apparent lack of quantitative correlation between Ca2+ influx and gene activation suggests an alternative activation pathway. Here, we demonstrate that membrane depolarization of HEK293 cells transfected with α11.2/ß2b/α2δ subunits (Cav1.2) triggers c-Fos and MeCP2 activation via the Ras/ERK/CREB pathway. Nuclear signaling is lost either by absence of the intracellular ß2 subunit or by transfecting the cells with the channel mutant α11.2W440A/ß2b/α2δ, a mutation that disrupts the interaction between α11.2 and ß2 subunits. Pulldown assays in neuronal SH-SY5Y cells and in vitro binding of recombinant H-Ras and ß2 confirmed the importance of the intracellular ß2 subunit for depolarization-induced gene activation. Using a Ca2+-impermeable mutant channel α11.2L745P/ß2b/α2δ or disrupting Ca2+/calmodulin binding to the channel using the channel mutant α11.2I1624A/ß2b/α2δ, we demonstrate that depolarization-induced c-Fos and MeCP2 activation does not depend on Ca2+ transport by the channel. Thus, in contrast to the paradigm that elevated intracellular Ca2+ drives nuclear signaling, we show that Cav1.2-triggered c-Fos or MeCP2 is dependent on extracellular Ca2+ and Ca2+ occupancy of the open channel pore, but is Ca2+-influx independent. An indispensable ß-subunit interaction with H-Ras, which is triggered by conformational changes at α11.2 independently of Ca2+ flux, brings to light a master regulatory role of ß2 in transcriptional activation via the ERK/CREB pathway. This mode of H-Ras activation could have broad implications for understanding the coupling of membrane depolarization to the rapid induction of gene transcription.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Transdução de Sinais , Cálcio/metabolismo , Canais de Cálcio Tipo L/genética , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Células HEK293 , Humanos , Proteína 2 de Ligação a Metil-CpG/genética , Proteína 2 de Ligação a Metil-CpG/metabolismo , Mutação , Neurônios/metabolismo , Ligação Proteica , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo
13.
Sci Rep ; 7(1): 11350, 2017 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-28900128

RESUMO

The secretory signal elicited by membrane depolarization traverses from the Ca2+-bound α11.2 pore-forming subunit of the L-type Ca2+-channel (Cav1.2) to syntaxin 1 A (Sx1A) via an intra-membrane signaling mechanism. Here, we report the use of two-color Photo-Activated-Localization-Microscopy (PALM) to determine the relation between Cav1.2 and Sx1A in single-molecule detail. We observed nanoscale co-clusters of PAmCherry-tagged Sx1A and Dronpa-tagged α11.2 at a ~1:1 ratio. PAmCherry-tagged Sx1AC145A, or PAmCherry-tagged Sx2, an inactive Cav1.2 modulator, in which Cys145 is a Ser residue, showed no co-clustering. These results are  consistent with the crucial role of the single cytosolic Sx1ACys145 in clustering with Cav1.2. Cav1.2 and the functionally inactive transmembrane-domain double mutant Sx1AC271V/C272V engendered clusters with a ~2:1 ratio. A higher extent of co-clustering, which coincides with compromised depolarization-evoked transmitter-release, was observed also by oxidation of Sx1ACys271 and Cys272. Our super-resolution-imaging results set the stage for studying co-clustering of the channel with other exocytotic proteins at a single-molecule level.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Membrana Celular/metabolismo , Sintaxina 1/metabolismo , Canais de Cálcio Tipo L/genética , Linhagem Celular , Exocitose , Humanos , Microscopia de Fluorescência , Imagem Molecular/métodos , Oxirredução , Ligação Proteica , Transporte Proteico , Transdução de Sinais , Sintaxina 1/genética
14.
Free Radic Biol Med ; 99: 557-571, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27658743

RESUMO

Impaired insulin signaling and the associated insulin-resistance in liver, adipose tissue, and skeletal muscle, represents a hallmark of the pathogenesis of type 2-diabetes-mellitus. Here we show that in the liver of db/db mice, a murine model of obesity, type 2 diabetes, and dyslipidemia, the elevated activities of mitogen-activated protein kinases (MAPK; ERK1/2 and p38MAPK), and Akt/PKB are abolished by rosiglitazone-treatment, which normalizes blood glucose in db/db mice. This is unequivocal evidence of a functional link between the activation of the MAPK specific inflammatory-pathway and high-blood sugar. A similar reduction in ERK1/2, p38MAPK, and Akt activities but without affecting blood-glucose was observed in the liver of db/db mice treated with a molecule that mimics the action of thioredoxin, called thioredoxin-mimetic peptide (TXM). N-Acetyl-Cys-Pro-Cys-amide (TXM-CB3) is a free radical scavenger, a reducing and denitrosylating reagent that protects the cells from early death induced by inflammatory pathways. TXM-CB3 also lowered MAPK signaling activated by the disruption of the thioredoxin-reductase-thioredoxin (Trx-TrxR) redox-system and restored Akt activity in rat hepatoma FAO cells. Similarly, two other TXM-peptides, N-Acetyl-Cys-Met-Lys-Cys-amide (TXM-CB13; DY70), and N-Acetyl-Cys-γGlu-Cys-Cys-amide (TXM-CB16; DY71), lowered insulin- and oxidative stress-induced ERK1/2 activation, and rescued HepG2 cells from cell death. The potential impact of TXM-peptides on inhibiting inflammatory pathways associated with high-glucose could be effective in reversing low-grade inflammation. TXM-peptides might also have the potential to improve insulin resistance by protecting from posttranslational modifications like nitrosylation.


Assuntos
Glicemia/metabolismo , Diabetes Mellitus Tipo 2/tratamento farmacológico , Hipoglicemiantes/farmacologia , Fígado/efeitos dos fármacos , Oligopeptídeos/farmacologia , Peptídeos/farmacologia , Animais , Linhagem Celular Tumoral , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Modelos Animais de Doenças , Regulação da Expressão Gênica , Células Hep G2 , Humanos , Fígado/metabolismo , Fígado/patologia , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Mimetismo Molecular , Estresse Oxidativo/efeitos dos fármacos , Peptídeos/síntese química , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Rosiglitazona , Transdução de Sinais , Tiazolidinedionas/farmacologia , Tiorredoxinas/química , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
15.
Psychopharmacology (Berl) ; 233(18): 3437-48, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27469021

RESUMO

RATIONALE: Chronic exposure to drugs of abuse changes glutamatergic transmission in human addicts and animal models. N-acetylcysteine (NAC) is a cysteine prodrug that indirectly activates cysteine-glutamate antiporters. In the extrasynaptic space, NAC restores basal glutamate levels during drug abstinence and normalizes increased glutamatergic tone in rats during reinstatement to drugs of abuse. In initial clinical trials, repeated NAC administration seems to be promising for reduced craving in cocaine addicts. OBJECTIVE: In this study, NAC-amide, called AD4 or NACA, was examined in intravenous cocaine self-administration and extinction/reinstatement procedures in rats. We investigated the behavioral effects of AD4 in the olfactory bulbectomized (OBX) rats, considered an animal model of depression. Finally, we tested rats injected with AD4 or NAC during 10-daily extinction training sessions to examine subsequent cocaine seeking. RESULTS: AD4 (25-75 mg kg(-1)) given acutely did not alter the rewarding effects of cocaine in OBX rats and sham-operated controls. However, at 6.25-50 mg kg(-1), AD4 decreased dose-dependently cocaine seeking and relapse triggered by cocaine priming or drug-associated conditioned cues in both phenotypes. Furthermore, repeated treatment with AD4 (25 mg kg(-1)) or NAC (100 mg kg(-1)) during daily extinction trials reduced reinstatement of drug-seeking behavior in sham-operated controls. In the OBX rats only, AD4 effectively blocked cocaine-seeking behavior. CONCLUSIONS: Our results demonstrate that AD4 is effective at blocking cocaine-seeking behavior, highlighting its potential clinical use toward cocaine use disorder.


Assuntos
Acetilcisteína/análogos & derivados , Comportamento Animal/efeitos dos fármacos , Cocaína/administração & dosagem , Inibidores da Captação de Dopamina/administração & dosagem , Comportamento de Procura de Droga/efeitos dos fármacos , Extinção Psicológica/efeitos dos fármacos , Acetilcisteína/farmacologia , Animais , Transtornos Relacionados ao Uso de Cocaína , Sinais (Psicologia) , Depressão , Masculino , Bulbo Olfatório/cirurgia , Ratos , Ratos Wistar , Recompensa , Autoadministração
16.
PLoS One ; 11(6): e0157064, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27285176

RESUMO

Mild traumatic brain injury (mTBI) is recognized as a common injury among children, sportsmen, and elderly population. mTBI lacks visible objective structural brain damage but patients frequently suffer from long-lasting cognitive, behavioral and emotional difficulties associated with biochemical and cellular changes. Currently there is no effective treatment for patients with mTBI. The thioredoxin reductase/thioredoxin pathway (TrxR/Trx1) has both anti-inflammatory and anti-oxidative properties. If the system is compromised, Trx1 remains oxidized and triggers cell death via an ASK1-Trx1 signal transduction mechanism. We previously showed tri and tetra peptides which were derived from the canonical -CxxC- motif of the Trx1-active site, called thioredoxin mimetic (TXM) peptides, reversed inflammatory and oxidative stress damage mimicking Trx1 activity. Here, TXM-peptides were examined for protecting cognitive function following weight drop closed-head injury in a mouse model of mTBI. TXM-CB3 (AcCys-Pro-CysNH2), TXM-CB13 (DY-70; AcCys-Met-Lys-CysNH2) or AD4 (ACysNH2) were administered at 50 mg/kg, 60 min after injury and cognitive performance was monitored by the novel-object-recognition and Y-maze tests. Behavioral deficits subsequent to mTBI injury were reversed by a single dose of TXM-CB3, TXM-CB13 and, to a lesser extent, by AD4. TXM-CB13 similar to TXM-CB3 and AD4 reversed oxidative stress-induced phosphorylation of mitogen-activated kinases, p38MAPK and c-Jun N-terminal kinase, (JNK) in human neuronal SH-SY5Y cells. We conclude that significantly improved cognitive behavior post mTBI by the TXM-peptides could result from anti-apoptotic, and/or anti-inflammatory activities. Future preclinical studies are required to establish the TXM-peptides as potential therapeutic drugs for brain injuries.


Assuntos
Concussão Encefálica/tratamento farmacológico , Cognição/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Peptídeos/farmacologia , Tiorredoxinas/química , Animais , Comportamento Animal/efeitos dos fármacos , Biomimética , Concussão Encefálica/patologia , Concussão Encefálica/fisiopatologia , Concussão Encefálica/psicologia , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Endogâmicos ICR , Fármacos Neuroprotetores/química , Peptídeos/química , Tiorredoxinas/farmacologia
17.
CNS Neurosci Ther ; 22(6): 461-7, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26861609

RESUMO

BACKGROUND: Long-term l-dihydroxyphenylalanine (l-DOPA) treatment of Parkinson's disease (PD) is associated with motor complications known as l-DOPA-induced dyskinesias (LID) and on/off fluctuations, which are linked to unsteady pulsatile dopaminergic stimulation. AIM: The objective of this study was to improve l-DOPA treatment by slowing and stabilizing dopamine (DA) production in the brain and increasing water solubility to provide a rescue therapy for PD. RESULTS: We synthesized l-DOPA-amide, a novel l-DOPA precursor called DopAmide. DopAmide is water soluble and, as a prodrug, requires hydrolysis prior to decarboxylation by the aromatic l-amino acid decarboxylase (EC 4.1.1.28; AAAD). In the 6-OH-dopamine (6-OHDA)-lesioned rats, DopAmide maintained steady rotations for up to 4 h compared with 2 h by l-DOPA, suggesting that this rate-limiting step generated a sustained level of DA at dopaminergic neurons. Pharmacokinetic studies showed elimination half-life of l-DOPA in the plasma after DopAmide treatment of t1/2 = 4.1 h, significantly longer than t1/2 = 2.9 h after treatment with l-DOPA, consistent with the 6-OHDA results. CONCLUSIONS: The slow conversion of DopAmide to l-DOPA provides a sustained level of DA in the dopaminergic cells, shown by the long 6-OHDA steady rotations. The water solubility and improved bioavailability may help reduce medication frequency associated with l-DOPA treatment of PD. Sustained levels of DA might lower the super-sensitization of DA signaling and potentially attenuate l-DOPA adverse effects.


Assuntos
Antiparkinsonianos/farmacologia , Dopamina/metabolismo , Neurônios Dopaminérgicos/efeitos dos fármacos , Levodopa/farmacologia , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/patologia , Anfetamina/farmacologia , Análise de Variância , Animais , Antiparkinsonianos/uso terapêutico , Inibidores das Descarboxilases de Aminoácidos Aromáticos/farmacologia , Carbidopa/farmacologia , Estimulantes do Sistema Nervoso Central/farmacologia , Modelos Animais de Doenças , Dopamina/análogos & derivados , Relação Dose-Resposta a Droga , Levodopa/uso terapêutico , Masculino , Oxidopamina/toxicidade , Doença de Parkinson/etiologia , Ratos , Ratos Wistar , Rotação , Simpatolíticos/toxicidade , Fatores de Tempo
18.
Free Radic Biol Med ; 79: 138-46, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25483557

RESUMO

S-nitrosylation, the coupling of a nitric oxide moiety to a reactive cysteine residue to form an S-nitrosothiol (SNO), is an important posttranslational mechanism for regulating protein activity. Growing evidence indicates that hyper-S-nitrosylation may contribute to cellular dysfunction associated with various human diseases. It is also increasingly appreciated that thioredoxin and thioredoxin reductase play significant roles in the cellular catabolism of SNO and protection from nitrosative stress. Here, we investigated the SNO reductase activity and protective effects of thioredoxin-mimetic peptides (TXMs), Ac-Cys-Pro-Cys-amide (CB3) and Ac-Cys-Gly-Pro-Cys-amide (CB4), both under cell-free conditions and in nitrosatively stressed cultured cells. In vitro biochemical analyses revealed that the TXM peptides reduced small-molecule SNO compounds, such as S-nitrosoglutathione (GSNO), and acted as general and efficient protein-denitrosylating agents. In particular, CB3 was found to be a highly potent SNO-metabolizing agent. Notably, CB3 mimicked the activity of thioredoxin by coupling with thioredoxin reductase to enhance GSNO reduction. Moreover, in a cell-free lysate system, both CB3 and CB4 synergized with an NADPH-dependent activity to denitrosylate proteins. Further investigation revealed that the TXM peptides protect the peroxiredoxin-thioredoxin system from SNO-dependent inhibition. Indeed, SNO-inhibited Prx1 was efficiently denitrosylated and reactivated by CB3 or CB4. In addition, CB3 protected thioredoxin reductase from SNO-mediated inactivation both in vitro and in intact cells. Finally, CB3 and CB4 partially rescued human neuroblastoma SH-SY5Y cells and rat insulinoma INS-1 832/13 cells from GSNO-induced growth inhibition. Collectively, the present findings indicate the efficient denitrosylation activity and protective effects of TXM peptides and suggest their potential therapeutic value in treating pathological conditions related to nitrosative stress.


Assuntos
Mimetismo Molecular , Nitrosação , Tiorredoxinas/metabolismo , Catálise , Linhagem Celular , Humanos , Peso Molecular , Tiorredoxinas/química
19.
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
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA