Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Clin Invest ; 129(8): 3140-3152, 2019 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-31162142

RESUMO

Elevated blood glucose (hyperglycemia) is a hallmark metabolic abnormality in diabetes. Hyperglycemia is associated with protein kinase A (PKA)-mediated stimulation of L-type Ca2+ channels in arterial myocytes resulting in increased vasoconstriction. However, the mechanisms by which glucose activates PKA remain unclear. Here, we showed that elevating extracellular glucose stimulates cAMP production in arterial myocytes, and that this was specifically dependent on adenylyl cyclase 5 (AC5) activity. Super-resolution imaging suggested nanometer proximity between subpopulations of AC5 and the L-type Ca2+ channel pore-forming subunit CaV1.2. In vitro, in silico, ex vivo and in vivo experiments revealed that this close association is critical for stimulation of L-type Ca2+ channels in arterial myocytes and increased myogenic tone upon acute hyperglycemia. This pathway supported the increase in L-type Ca2+ channel activity and myogenic tone in two animal models of diabetes. Our collective findings demonstrate a unique role for AC5 in PKA-dependent modulation of L-type Ca2+ channel activity and vascular reactivity during acute hyperglycemia and diabetes.


Assuntos
Adenilil Ciclases/metabolismo , Artérias Cerebrais/enzimologia , AMP Cíclico/metabolismo , Diabetes Mellitus Experimental/enzimologia , Hiperglicemia/enzimologia , Músculo Liso Vascular/enzimologia , Miócitos de Músculo Liso/enzimologia , Adenilil Ciclases/genética , Animais , Canais de Cálcio Tipo L/biossíntese , Canais de Cálcio Tipo L/genética , Artérias Cerebrais/patologia , AMP Cíclico/genética , Proteínas Quinases Dependentes de AMP Cíclico/genética , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patologia , Hiperglicemia/genética , Hiperglicemia/patologia , Camundongos , Camundongos Knockout , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia
2.
Elife ; 82019 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-31184584

RESUMO

The cation channel TRPA1 transduces a myriad of noxious chemical stimuli into nociceptor electrical excitation and neuropeptide release, leading to pain and neurogenic inflammation. Despite emergent evidence that TRPA1 is regulated by the membrane environment, it remains unknown whether this channel localizes in membrane microdomains or whether it interacts with cholesterol. Using total internal reflection fluorescence microscopy and density gradient centrifugation we found that mouse TRPA1 localizes preferably into cholesterol-rich domains and functional experiments revealed that cholesterol depletion decreases channel sensitivity to chemical agonists. Moreover, we identified two structural motifs in transmembrane segments 2 and 4 involved in mTRPA1-cholesterol interactions that are necessary for normal agonist sensitivity and plasma membrane localization. We discuss the impact of such interactions on TRPA1 gating mechanisms, regulation by the lipid environment, and role of this channel in sensory membrane microdomains, all of which helps to understand the puzzling pharmacology and pathophysiology of this channel.


Assuntos
Membrana Celular/metabolismo , Colesterol/metabolismo , Canal de Cátion TRPA1/metabolismo , Sequência de Aminoácidos , Animais , Células CHO , Colesterol/química , Cricetinae , Cricetulus , Células HEK293 , Humanos , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Microdomínios da Membrana/metabolismo , Camundongos , Microscopia de Fluorescência/métodos , Modelos Moleculares , Ligação Proteica , Domínios Proteicos , Homologia de Sequência de Aminoácidos , Canal de Cátion TRPA1/química , Canal de Cátion TRPA1/genética , Proteína Vermelha Fluorescente
3.
Elife ; 82019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30821687

RESUMO

Elevated glucose increases vascular reactivity by promoting L-type CaV1.2 channel (LTCC) activity by protein kinase A (PKA). Yet, how glucose activates PKA is unknown. We hypothesized that a Gs-coupled P2Y receptor is an upstream activator of PKA mediating LTCC potentiation during diabetic hyperglycemia. Experiments in apyrase-treated cells suggested involvement of a P2Y receptor underlying the glucose effects on LTTCs. Using human tissue, expression for P2Y11, the only Gs-coupled P2Y receptor, was detected in nanometer proximity to CaV1.2 and PKA. FRET-based experiments revealed that the selective P2Y11 agonist NF546 and elevated glucose stimulate cAMP production resulting in enhanced PKA-dependent LTCC activity. These changes were blocked by the selective P2Y11 inhibitor NF340. Comparable results were observed in mouse tissue, suggesting that a P2Y11-like receptor is mediating the glucose response in these cells. These findings established a key role for P2Y11 in regulating PKA-dependent LTCC function and vascular reactivity during diabetic hyperglycemia.


Assuntos
Vasos Sanguíneos/fisiopatologia , Cálcio/metabolismo , Hiperglicemia , Contração Muscular , Receptores Acoplados a Proteínas G/metabolismo , Receptores Purinérgicos/metabolismo , Animais , Sinalização do Cálcio , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Camundongos Endogâmicos C57BL
4.
J Physiol ; 597(8): 2139-2162, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30714156

RESUMO

KEY POINTS: Prevailing dogma holds that activation of the ß-adrenergic receptor/cAMP/protein kinase A signalling pathway leads to enhanced L-type CaV 1.2 channel activity, resulting in increased Ca2+ influx into ventricular myocytes and a positive inotropic response. However, the full mechanistic and molecular details underlying this phenomenon are incompletely understood. CaV 1.2 channel clusters decorate T-tubule sarcolemmas of ventricular myocytes. Within clusters, nanometer proximity between channels permits Ca2+ -dependent co-operative gating behaviour mediated by physical interactions between adjacent channel C-terminal tails. We report that stimulation of cardiomyocytes with isoproterenol, evokes dynamic, protein kinase A-dependent augmentation of CaV 1.2 channel abundance along cardiomyocyte T-tubules, resulting in the appearance of channel 'super-clusters', and enhanced channel co-operativity that amplifies Ca2+ influx. On the basis of these data, we suggest a new model in which a sub-sarcolemmal pool of pre-synthesized CaV 1.2 channels resides in cardiomyocytes and can be mobilized to the membrane in times of high haemodynamic or metabolic demand, to tune excitation-contraction coupling. ABSTRACT: Voltage-dependent L-type CaV 1.2 channels play an indispensable role in cardiac excitation-contraction coupling. Activation of the ß-adrenergic receptor (ßAR)/cAMP/protein kinase A (PKA) signalling pathway leads to enhanced CaV 1.2 activity, resulting in increased Ca2+ influx into ventricular myocytes and a positive inotropic response. CaV 1.2 channels exhibit a clustered distribution along the T-tubule sarcolemma of ventricular myocytes where nanometer proximity between channels permits Ca2+ -dependent co-operative gating behaviour mediated by dynamic, physical, allosteric interactions between adjacent channel C-terminal tails. This amplifies Ca2+ influx and augments myocyte Ca2+ transient and contraction amplitudes. We investigated whether ßAR signalling could alter CaV 1.2 channel clustering to facilitate co-operative channel interactions and elevate Ca2+ influx in ventricular myocytes. Bimolecular fluorescence complementation experiments reveal that the ßAR agonist, isoproterenol (ISO), promotes enhanced CaV 1.2-CaV 1.2 physical interactions. Super-resolution nanoscopy and dynamic channel tracking indicate that these interactions are expedited by enhanced spatial proximity between channels, resulting in the appearance of CaV 1.2 'super-clusters' along the z-lines of ISO-stimulated cardiomyocytes. The mechanism that leads to super-cluster formation involves rapid, dynamic augmentation of sarcolemmal CaV 1.2 channel abundance after ISO application. Optical and electrophysiological single channel recordings confirm that these newly inserted channels are functional and contribute to overt co-operative gating behaviour of CaV 1.2 channels in ISO stimulated myocytes. The results of the present study reveal a new facet of ßAR-mediated regulation of CaV 1.2 channels in the heart and support the novel concept that a pre-synthesized pool of sub-sarcolemmal CaV 1.2 channel-containing vesicles/endosomes resides in cardiomyocytes and can be mobilized to the sarcolemma to tune excitation-contraction coupling to meet metabolic and/or haemodynamic demands.


Assuntos
Canais de Cálcio Tipo L/fisiologia , Miócitos Cardíacos/fisiologia , Receptores Adrenérgicos beta/fisiologia , Agonistas Adrenérgicos beta/farmacologia , Animais , Linhagem Celular , Feminino , Ventrículos do Coração/citologia , Humanos , Isoproterenol/farmacologia , Masculino , Camundongos Endogâmicos C57BL , Sarcolema/fisiologia
5.
Proc Natl Acad Sci U S A ; 115(49): E11465-E11474, 2018 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-30455320

RESUMO

A-kinase anchoring proteins (AKAPs) shape second-messenger signaling responses by constraining protein kinase A (PKA) at precise intracellular locations. A defining feature of AKAPs is a helical region that binds to regulatory subunits (RII) of PKA. Mining patient-derived databases has identified 42 nonsynonymous SNPs in the PKA-anchoring helices of five AKAPs. Solid-phase RII binding assays confirmed that 21 of these amino acid substitutions disrupt PKA anchoring. The most deleterious side-chain modifications are situated toward C-termini of AKAP helices. More extensive analysis was conducted on a valine-to-methionine variant in the PKA-anchoring helix of AKAP18. Molecular modeling indicates that additional density provided by methionine at position 282 in the AKAP18γ isoform deflects the pitch of the helical anchoring surface outward by 6.6°. Fluorescence polarization measurements show that this subtle topological change reduces RII-binding affinity 8.8-fold and impairs cAMP responsive potentiation of L-type Ca2+ currents in situ. Live-cell imaging of AKAP18γ V282M-GFP adducts led to the unexpected discovery that loss of PKA anchoring promotes nuclear accumulation of this polymorphic variant. Targeting proceeds via a mechanism whereby association with the PKA holoenzyme masks a polybasic nuclear localization signal on the anchoring protein. This led to the discovery of AKAP18ε: an exclusively nuclear isoform that lacks a PKA-anchoring helix. Enzyme-mediated proximity-proteomics reveal that compartment-selective variants of AKAP18 associate with distinct binding partners. Thus, naturally occurring PKA-anchoring-defective AKAP variants not only perturb dissemination of local second-messenger responses, but also may influence the intracellular distribution of certain AKAP18 isoforms.


Assuntos
Proteínas de Ancoragem à Quinase A/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/química , Proteínas Quinases Dependentes de AMP Cíclico/genética , Proteínas de Membrana/metabolismo , Proteínas de Ancoragem à Quinase A/genética , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Regulação Enzimológica da Expressão Gênica , Estudo de Associação Genômica Ampla , Humanos , Proteínas de Membrana/genética , Modelos Moleculares , Polimorfismo de Nucleotídeo Único , Ligação Proteica , Conformação Proteica , Isoformas de Proteínas , Transporte Proteico
6.
Biochim Biophys Acta Mol Cell Res ; 1865(9): 1341-1355, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29959960

RESUMO

L-type CaV1.2 channels are key regulators of gene expression, cell excitability and muscle contraction. CaV1.2 channels organize in clusters throughout the plasma membrane. This channel organization has been suggested to contribute to the concerted activation of adjacent CaV1.2 channels (e.g. cooperative gating). Here, we tested the hypothesis that dynamic intracellular and perimembrane trafficking of CaV1.2 channels is critical for formation and dissolution of functional channel clusters mediating cooperative gating. We found that CaV1.2 moves in vesicular structures of circular and tubular shape with diverse intracellular and submembrane trafficking patterns. Both microtubules and actin filaments are required for dynamic movement of CaV1.2 vesicles. These vesicles undergo constitutive homotypic fusion and fission events that sustain CaV1.2 clustering, channel activity and cooperative gating. Our study suggests that CaV1.2 clusters and activity can be modulated by diverse and unique intracellular and perimembrane vesicular dynamics to fine-tune Ca2+ signals.


Assuntos
Citoesqueleto de Actina/metabolismo , Canais de Cálcio Tipo L/metabolismo , Microtúbulos/metabolismo , Vesículas Transportadoras/metabolismo , Sinalização do Cálcio , Linhagem Celular , Membrana Celular/metabolismo , Citoplasma/metabolismo , Humanos , Ativação do Canal Iônico , Transporte Proteico
7.
Sci Rep ; 7(1): 14058, 2017 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-29070899

RESUMO

Large-conductance Ca2+-activated potassium (BKCa) channels are key determinants of vascular smooth muscle excitability. Impaired BKCa channel function through remodeling of BKCa ß1 expression and function contributes to vascular complications in animal models of diabetes. Yet, whether similar alterations occur in native vascular smooth muscle from humans with type 2 diabetes is unclear. In this study, we evaluated BKCa function in vascular smooth muscle from small resistance adipose arteries of non-diabetic and clinically diagnosed type 2 diabetic patients. We found that BKCa channel activity opposes pressure-induced constriction in human small resistance adipose arteries, and this is compromised in arteries from diabetic patients. Consistent with impairment of BKCa channel function, the amplitude and frequency of spontaneous BKCa currents, but not Ca2+ sparks were lower in cells from diabetic patients. BKCa channels in diabetic cells exhibited reduced Ca2+ sensitivity, single-channel open probability and tamoxifen sensitivity. These effects were associated with decreased functional coupling between BKCa α and ß1 subunits, but no change in total protein abundance. Overall, results suggest impairment in BKCa channel function in vascular smooth muscle from diabetic patients through unique mechanisms, which may contribute to vascular complications in humans with type 2 diabetes.


Assuntos
Artérias/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/fisiopatologia , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/metabolismo , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Potenciais de Ação , Artérias/fisiopatologia , Estudos de Casos e Controles , Células Cultivadas , Humanos , Potenciais da Membrana
8.
Elife ; 52016 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-27449282

RESUMO

Various TRP channels act as polymodal sensors of thermal and chemical stimuli, but the mechanisms whereby chemical ligands impact on TRP channel gating are poorly understood. Here we show that AITC (allyl isothiocyanate; mustard oil) and menthol represent two distinct types of ligands at the mammalian cold sensor TRPM8. Kinetic analysis of channel gating revealed that AITC acts by destabilizing the closed channel, whereas menthol stabilizes the open channel, relative to the transition state. Based on these differences, we classify agonists as either type I (menthol-like) or type II (AITC-like), and provide a kinetic model that faithfully reproduces their differential effects. We further demonstrate that type I and type II agonists have a distinct impact on TRPM8 currents and TRPM8-mediated calcium signals in excitable cells. These findings provide a theoretical framework for understanding the differential actions of TRP channel ligands, with important ramifications for TRP channel structure-function analysis and pharmacology.


Assuntos
Isotiocianatos/metabolismo , Mentol/metabolismo , Canais de Cátion TRPM/agonistas , Canais de Cátion TRPM/metabolismo , Animais , Sinalização do Cálcio , Cinética , Camundongos , Técnicas de Patch-Clamp
10.
Nat Commun ; 7: 10489, 2016 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-26843440

RESUMO

The cation channel TRPM8 plays a central role in the somatosensory system, as a key sensor of innocuously cold temperatures and cooling agents. Although increased functional expression of TRPM8 has been implicated in various forms of pathological cold hypersensitivity, little is known about the cellular and molecular mechanisms that determine TRPM8 abundance at the plasma membrane. Here we demonstrate constitutive transport of TRPM8 towards the plasma membrane in atypical, non-acidic transport vesicles that contain lysosomal-associated membrane protein 1 (LAMP1), and provide evidence that vesicle-associated membrane protein 7 (VAMP7) mediates fusion of these vesicles with the plasma membrane. In line herewith, VAMP7-deficient mice exhibit reduced functional expression of TRPM8 in sensory neurons and concomitant deficits in cold avoidance and icilin-induced cold hypersensitivity. Our results uncover a cellular pathway that controls functional plasma membrane incorporation of a temperature-sensitive TRP channel, and thus regulates thermosensitivity in vivo.


Assuntos
Membrana Celular/metabolismo , Temperatura Baixa , Hiperestesia/genética , Proteínas R-SNARE/genética , Células Receptoras Sensoriais/metabolismo , Canais de Cátion TRPM/metabolismo , Vesículas Transportadoras/metabolismo , Animais , Cálcio/metabolismo , Feminino , Gânglios Espinais/metabolismo , Células HEK293 , Humanos , Hiperestesia/induzido quimicamente , Hiperestesia/metabolismo , Proteínas de Membrana Lisossomal/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia de Fluorescência , Técnicas de Patch-Clamp , Pirimidinonas/toxicidade , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Gânglio Trigeminal/metabolismo
11.
J Gen Physiol ; 146(1): 51-63, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26123194

RESUMO

The transient receptor potential (TRP) channel TRPM3 is a calcium-permeable cation channel activated by heat and by the neurosteroid pregnenolone sulfate (PregS). TRPM3 is highly expressed in sensory neurons, where it plays a key role in heat sensing and inflammatory hyperalgesia, and in pancreatic ß cells, where its activation enhances glucose-induced insulin release. However, despite its functional importance, little is known about the cellular mechanisms that regulate TRPM3 activity. Here, we provide evidence for a dynamic regulation of TRPM3 by membrane phosphatidylinositol phosphates (PIPs). Phosphatidylinositol 4,5-bisphosphate (PI[4,5]P2) and ATP applied to the intracellular side of excised membrane patches promote recovery of TRPM3 from desensitization. The stimulatory effect of cytosolic ATP on TRPM3 reflects activation of phosphatidylinositol kinases (PI-Ks), leading to resynthesis of PIPs in the plasma membrane. Various PIPs directly enhance TRPM3 activity in cell-free inside-out patches, with a potency order PI(3,4,5)P3 > PI(3,5)P2 > PI(4,5)P2 ≈ PI(3,4)P2 >> PI(4)P. Conversely, TRPM3 activity is rapidly and reversibly inhibited by activation of phosphatases that remove the 5-phosphate from PIPs. Finally, we show that recombinant TRPM3, as well as the endogenous TRPM3 in insuloma cells, is rapidly and reversibly inhibited by activation of phospholipase C-coupled muscarinic acetylcholine receptors. Our results reveal basic cellular mechanisms whereby membrane receptors can regulate TRPM3 activity.


Assuntos
Fosfatidilinositóis/metabolismo , Canais de Cátion TRPM/metabolismo , Trifosfato de Adenosina/metabolismo , Linhagem Celular , Membrana Celular/metabolismo , Células HEK293 , Humanos , Hiperalgesia/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Pregnenolona/metabolismo , Células Receptoras Sensoriais/metabolismo
12.
Pflugers Arch ; 467(10): 2107-19, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25559845

RESUMO

Calcium signaling participates in different cellular processes leading to cell migration. TRPV4, a non-selective cation channel that responds to mechano-osmotic stimulation and heat, is also involved in cell migration. However, the mechanistic involvement of TRPV4 in cell migration is currently unknown. We now report that expression of the mutant channel TRPV4-(121)AAWAA (lacking the phosphoinositide-binding site (121)KRWRK(125) and the response to physiological stimuli) altered HEK293 cell migration. Altered migration patterns included periods of fast and persistent motion followed by periods of stalling and turning, and the extension of multiple long cellular protrusions. TRPV4-WT overexpressing cells showed almost complete loss of directionality with frequent turns, no progression, and absence of long protrusions. Traction microscopy revealed higher tractions forces in the tail of TRPV4-(121)AAWAA than in TRPV4-WT expressing cells. These results are consistent with a defective and augmented tail retraction in TRPV4-(121)AAWAA- and TRPV4-WT-expressing cells, respectively. The activity of calpain, a protease implicated in focal adhesion (FA) disassembly, was decreased in TRPV4-(121)AAWAA compared with TRPV4-WT-expressing cells. Consistently, larger focal adhesions were seen in TRPV4-(121)AAWAA compared with TRPV4-WT-expressing HEK293 cells, a result that was also reproduced in T47D and U87 cells. Similarly, overexpression of the pore-dead mutant TRPV4-M680D resumed the TRPV4-(121)AAWAA phenotype presenting larger FA. The migratory phenotype obtained in HEK293 cells overexpressing TRPV4-(121)AAWAA was mimicked by knocking-down TRPC1, a cationic channel that participates in cell migration. Together, our results point to the participation of TRPV4 in the dynamics of trailing adhesions, a function that may require the interplay of TRPV4 with other cation channels or proteins present at the FA sites.


Assuntos
Estruturas da Membrana Celular/metabolismo , Movimento Celular , Canais de Cátion TRPV/metabolismo , Sítios de Ligação , Calpaína/metabolismo , Adesão Celular , Linhagem Celular Tumoral , Células HEK293 , Humanos , Mutação , Fosfatidilinositóis/metabolismo , Ligação Proteica , Canais de Cátion TRPV/química , Canais de Cátion TRPV/genética
13.
Temperature (Austin) ; 2(2): 163-5, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-27227018

RESUMO

TRP ion channels are ubiquitously present in the mammalian body and take part in numerous key physiological functions, including temperature sensing, taste perception, osmo-regulation, cardiac function, renal function, development, and glucose homeostasis. The mechanisms whereby TRP channels are transported to the plasma membrane, where most of them exert their physiological actions, remains a poorly understood aspect of TRP channel biology.

14.
Sci Rep ; 4: 7111, 2014 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-25407951

RESUMO

Transient Receptor Potential (TRP) channels form a broadly expressed and functionally diverse family of cation channels involved in various (patho)physiological processes. Whereas the mechanisms that control opening of TRP channels have been extensively studied, little is known about the transport processes of TRP channels to and within the plasma membrane. Here we used Total Internal Reflection--Fluorescence Recovery after Photobleaching (TIR-FRAP) to selectively visualize and bleach the fluorescently labeled TRP channels TRPV2 and TRPM4 in close proximity of the glass-plasma membrane interface, allowing detailed analysis of their perimembrane dynamics. We show that recovery of TRPM4 occurs via 200-nm diameter transport vesicles, and demonstrate the full fusion of such vesicles with the plasma membrane. In contrast, TRPV2 recovery proceeded mainly via lateral diffusion from non-bleached areas of the plasma membrane. Analysis of the two-dimensional channel diffusion kinetics yielded 2D diffusion coefficients ranging between 0.1 and 0.3 µm(2)/s, suggesting that these TRP channels move relatively unrestricted within the plasma membrane. These data demonstrate distinct modes of TRP channel turnover at the plasma membrane and illustrate the usefulness of TIR-FRAP to monitor these processes with high resolution.


Assuntos
Proteínas de Fluorescência Verde/metabolismo , Canais de Cátion TRPM/metabolismo , Canais de Cátion TRPV/metabolismo , Vesículas Transportadoras/metabolismo , Recuperação de Fluorescência Após Fotodegradação , Expressão Gênica , Proteínas de Fluorescência Verde/genética , Células HEK293 , Humanos , Microscopia de Fluorescência/instrumentação , Microscopia de Fluorescência/métodos , Plasmídeos/química , Plasmídeos/metabolismo , Transporte Proteico , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Canais de Cátion TRPM/genética , Canais de Cátion TRPV/genética , Transfecção , Vesículas Transportadoras/ultraestrutura
15.
EMBO J ; 31(7): 1764-73, 2012 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-22343944

RESUMO

The deregulation of brain cholesterol metabolism is typical in acute neuronal injury (such as stroke, brain trauma and epileptic seizures) and chronic neurodegenerative diseases (Alzheimer's disease). Since both conditions are characterized by excessive stimulation of glutamate receptors, we have here investigated to which extent excitatory neurotransmission plays a role in brain cholesterol homeostasis. We show that a short (30 min) stimulation of glutamatergic neurotransmission induces a small but significant loss of membrane cholesterol, which is paralleled by release to the extracellular milieu of the metabolite 24S-hydroxycholesterol. Consistent with a cause-effect relationship, knockdown of the enzyme cholesterol 24-hydroxylase (CYP46A1) prevented glutamate-mediated cholesterol loss. Functionally, the loss of cholesterol modulates the magnitude of the depolarization-evoked calcium response. Mechanistically, glutamate-induced cholesterol loss requires high levels of intracellular Ca(2+), a functional stromal interaction molecule 2 (STIM2) and mobilization of CYP46A1 towards the plasma membrane. This study underscores the key role of excitatory neurotransmission in the control of membrane lipid composition, and consequently in neuronal membrane organization and function.


Assuntos
Colesterol/metabolismo , Ácido Glutâmico/metabolismo , Hipocampo/metabolismo , Neurônios/metabolismo , Transmissão Sináptica , Animais , Cálcio/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Colesterol 24-Hidroxilase , Técnicas de Silenciamento de Genes , Ácido Glutâmico/farmacologia , Hipocampo/efeitos dos fármacos , Hidroxicolesteróis/metabolismo , Proteínas de Membrana/metabolismo , Neurônios/efeitos dos fármacos , Ratos , Ratos Wistar , Esteroide Hidroxilases/genética , Esteroide Hidroxilases/metabolismo , Molécula 2 de Interação Estromal
16.
Br J Neurosurg ; 25(4): 516-22, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21749185

RESUMO

Spinal cord injury (SCI) occurs in the most productive part of life. Treatment options for treatment of chronic SCI are few and have limited impact on clinical outcome. Central nervous system (CNS) has limited intrinsic regeneration capability. The study included patients with chronic complete SCI. Previously harvested autologous mesenchymal stem cells were administered at the site of injury after a laminectomy. Follow-up was done by a neutral examiner not involved in the surgery every 3 months. One patient had improvement in motor power. Two patients had a patchy improvement in pin prick sensation below the level of injury. Three different, progressively increasing doses did not result in improvement in the clinical outcome. Though the administration of allogenic human mesenchymal stem cells is safe in patients with SCI, it may not be efficacious; especially in patients with chronic SCI.


Assuntos
Transplante de Células-Tronco Mesenquimais/métodos , Traumatismos da Medula Espinal/terapia , Adulto , Doença Crônica , Feminino , Seguimentos , Humanos , Injeções Espinhais , Masculino , Transplante de Células-Tronco Mesenquimais/efeitos adversos , Pessoa de Meia-Idade , Projetos Piloto , Coleta de Tecidos e Órgãos , Transplante Autólogo , Resultado do Tratamento , Adulto Jovem
17.
Proc Natl Acad Sci U S A ; 107(44): 19084-9, 2010 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-20956320

RESUMO

Reduced functional bladder capacity and concomitant increased micturition frequency (pollakisuria) are common lower urinary tract symptoms associated with conditions such as cystitis, prostatic hyperplasia, neurological disease, and overactive bladder syndrome. These symptoms can profoundly affect the quality of life of afflicted individuals, but available pharmacological treatments are often unsatisfactory. Recent work has demonstrated that the cation channel TRPV4 is highly expressed in urothelial cells and plays a role in sensing the normal filling state of the bladder. In this article, we show that the development of cystitis-induced bladder dysfunction is strongly impaired in Trpv4(-/-) mice. Moreover, we describe HC-067047, a previously uncharacterized, potent, and selective TRPV4 antagonist that increases functional bladder capacity and reduces micturition frequency in WT mice and rats with cystitis. HC-067047 did not affect bladder function in Trpv4(-/-) mice, demonstrating that its in vivo effects are on target. These results indicate that TRPV4 antagonists may provide a promising means of treating bladder dysfunction.


Assuntos
Antineoplásicos Alquilantes/efeitos adversos , Ciclofosfamida/efeitos adversos , Cistite , Moduladores de Transporte de Membrana/farmacologia , Morfolinas/farmacologia , Pirróis/farmacologia , Canais de Cátion TRPV/antagonistas & inibidores , Bexiga Urinária/fisiopatologia , Urotélio/fisiopatologia , Animais , Antineoplásicos Alquilantes/farmacologia , Ciclofosfamida/farmacologia , Cistite/induzido quimicamente , Cistite/tratamento farmacológico , Cistite/metabolismo , Cistite/fisiopatologia , Humanos , Camundongos , Camundongos Knockout , Ratos , Ratos Wistar , Canais de Cátion TRPV/genética , Canais de Cátion TRPV/metabolismo , Bexiga Urinária/metabolismo , Micção/efeitos dos fármacos , Urotélio/metabolismo
18.
PLoS One ; 5(4): e9984, 2010 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-20376308

RESUMO

BACKGROUND: Benzo[a]pyrene (B[a]P) belongs to a class of polycyclic aromatic hydrocarbons that serve as micropollutants in the environment. B[a]P has been reported as a probable carcinogen in humans. Exposure to B[a]P can take place by ingestion of contaminated (especially grilled, roasted or smoked) food or water, or inhalation of polluted air. There are reports available that also suggests neurotoxicity as a result of B[a]P exposure, but the exact mechanism of action is unknown. METHODOLOGY/PRINCIPAL FINDINGS: Using neuroblastoma cell line and primary cortical neuron culture, we demonstrated that B[a]P has no direct neurotoxic effect. We utilized both in vivo and in vitro systems to demonstrate that B[a]P causes microglial activation. Using microglial cell line and primary microglial culture, we showed for the first time that B[a]P administration results in elevation of reactive oxygen species within the microglia thereby causing depression of antioxidant protein levels; enhanced expression of inducible nitric oxide synthase, that results in increased production of NO from the cells. Synthesis and secretion of proinflammatory cytokines were also elevated within the microglia, possibly via the p38MAP kinase pathway. All these factors contributed to bystander death of neurons, in vitro. When administered to animals, B[a]P was found to cause microglial activation and astrogliosis in the brain with subsequent increase in proinflammatory cytokine levels. CONCLUSIONS/SIGNIFICANCE: Contrary to earlier published reports we found that B[a]P has no direct neurotoxic activity. However, it kills neurons in a bystander mechanism by activating the immune cells of the brain viz the microglia. For the first time, we have provided conclusive evidence regarding the mechanism by which the micropollutant B[a]P may actually cause damage to the central nervous system. In today's perspective, where rising pollution levels globally are a matter of grave concern, our study throws light on other health hazards that such pollutants may exert.


Assuntos
Benzo(a)pireno/farmacologia , Carcinógenos/farmacologia , Microglia/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Animais , Benzo(a)pireno/toxicidade , Carcinógenos/toxicidade , Morte Celular/efeitos dos fármacos , Linhagem Celular , Células Cultivadas , Citocinas/biossíntese , Citocinas/metabolismo , Poluentes Ambientais/farmacologia , Poluentes Ambientais/toxicidade , Mediadores da Inflamação , Camundongos , Microglia/metabolismo , Neuroblastoma/patologia , Neurônios/citologia , Óxido Nítrico/biossíntese , Espécies Reativas de Oxigênio/metabolismo
19.
PLoS One ; 4(12): e8134, 2009 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-19956550

RESUMO

BACKGROUND: The low immunogenicity of neural stem/progenitor cells (NSPCs) coupled with negligible expression of MHC antigens has popularized their use in transplantation medicine. However, in an inflammatory environment, the NSPCs express costimulatory molecules and MHC antigens, and also exhibit certain immunomodulatory functions. Since NSPCs are the cellular targets in a number of virus infections both during postnatal and adult stages, we wanted to investigate the immunological properties of these stem cells in response to viral pathogen. METHODOLOGY/PRINCIPAL FINDINGS: We utilized both in vivo mouse model and in vitro neurosphere model of Japanese encephalitis virus (JEV) infection for the study. The NSPCs residing in the subventricular zone of the infected brains showed prominent expression of MHC-I and costimulatory molecules CD40, CD80, and CD86. Using Flow cytometry and fluorescence microscopy, we observed increased surface expression of co-stimulatory molecule and MHC class I antigen in NSPCs upon progressive JEV infection in vitro. Moreover, significant production of pro-inflammatory cyto/chemokines was detected in JEV infected NSPCs by Cytokine Bead Array analysis. Interestingly, NSPCs were capable of providing functional costimulation to allogenic T cells and JEV infection resulted in increased proliferation of allogenic T cells, as detected by Mixed Lymphocyte reaction and CFSE experiments. We also report IL-2 production by NSPCs upon JEV infection, which possibly provides mitogenic signals to T cells and trigger their proliferation. CONCLUSION/SIGNIFICANCE: The in vivo and in vitro findings clearly indicate the development of immunogenicity in NSPCs following progressive JEV infection, in our case, JEV infection. Following a neurotropic virus infection, NSPCs possibly behave as immunogenic cells and contribute to both the innate and adaptive immune axes. The newly discovered immunological properties of NSPCs may have implications in assigning a new role of these cells as non-professional antigen presenting cells in the central nervous system.


Assuntos
Vírus da Encefalite Japonesa (Espécie)/imunologia , Imunocompetência/imunologia , Neurônios/citologia , Neurônios/imunologia , Células-Tronco/imunologia , Células-Tronco/virologia , Animais , Proliferação de Células , Ventrículos Cerebrais/imunologia , Ventrículos Cerebrais/patologia , Ventrículos Cerebrais/virologia , Progressão da Doença , Encefalite Japonesa/imunologia , Encefalite Japonesa/patologia , Encefalite Japonesa/virologia , Antígenos de Histocompatibilidade Classe I/imunologia , Interleucina-2/genética , Interleucina-2/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Neurônios/virologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Células-Tronco/citologia , Linfócitos T/citologia , Linfócitos T/imunologia , Linfócitos T/virologia , Regulação para Cima/genética
20.
PLoS Negl Trop Dis ; 3(9): e437, 2009 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-19787040

RESUMO

Japanese encephalitis (JE) is the leading form of viral encephalitis in Asia. It is caused by the JE virus (JEV), which belongs to the family Flaviviridae. JEV is endemic to many parts of Asia, where periodic outbreaks take hundreds of lives. Despite the catastrophes it causes, JE has remained a tropical disease uncommon in the West. With rapid globalization and climatic shift, JEV has started to emerge in areas where the threat was previously unknown. Scientific evidence predicts that JEV will soon become a global pathogen and cause of worldwide pandemics. Although some research documents JEV pathogenesis and drug discovery, worldwide awareness of the need for extensive research to deal with JE is still lacking. This review focuses on the exigency of developing a worldwide effort to acknowledge the prime importance of performing an extensive study of this thus far neglected tropical viral disease. This review also outlines the pathogenesis, the scientific efforts channeled into develop a therapy, and the outlook for a possible future breakthrough addressing this killer disease.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...