Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Pflugers Arch ; 2024 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-38538988

RESUMO

Human-induced pluripotent stem cells (iPS cells) are efficiently differentiated into sensory neurons. These cells express the voltage-gated sodium channel NaV1.7, which is a validated pain target. NaV1.7 deficiency leads to pain insensitivity, whereas NaV1.7 gain-of-function mutants are associated with chronic pain. During differentiation, the sensory neurons start spontaneous action potential firing around day 22, with increasing firing rate until day 40. Here, we used CRISPR/Cas9 genome editing to generate a HA-tag NaV1.7 to follow its expression during differentiation. We used two protocols to generate sensory neurons: the classical small molecule approach and a directed differentiation methodology and assessed surface NaV1.7 expression by Airyscan high-resolution microscopy. Our results show that maturation of at least 49 days is necessary to observe robust NaV1.7 surface expression in both protocols. Electric activity of the sensory neurons precedes NaV1.7 surface expression. A clinically effective NaV1.7 blocker is still missing, and we expect this iPS cell model system to be useful for drug discovery and disease modeling.

3.
Front Mol Neurosci ; 14: 709228, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34385907

RESUMO

Apart from the most prominent symptoms in Autism spectrum disorder (ASD), namely deficits in social interaction, communication and repetitive behavior, patients often show abnormal sensory reactivity to environmental stimuli. Especially potentially painful stimuli are reported to be experienced in a different way compared to healthy persons. In our present study, we identified an ASD patient carrying compound heterozygous mutations in the voltage-gated sodium channel (VGSC) Na v 1.8, which is preferentially expressed in sensory neurons. We expressed both mutations, p.I1511M and p.R512∗, in a heterologous expression system and investigated their biophysical properties using patch-clamp recordings. The results of these experiments reveal that the p.R512∗ mutation renders the channel non-functional, while the p.I1511M mutation showed only minor effects on the channel's function. Behavioral experiments in a Na v 1.8 loss-of-function mouse model additionally revealed that Na v 1.8 may play a role in autism-like symptomatology. Our results present Na v 1.8 as a protein potentially involved in ASD pathophysiology and may therefore offer new insights into the genetic basis of this disease.

4.
Sci Rep ; 11(1): 6934, 2021 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-33767215

RESUMO

Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder caused by an expanded polyglutamine repeat in the huntingtin gene. The neuropathology of HD is characterized by the decline of a specific neuronal population within the brain, the striatal medium spiny neurons (MSNs). The origins of this extreme vulnerability remain unknown. Human induced pluripotent stem cell (hiPS cell)-derived MSNs represent a powerful tool to study this genetic disease. However, the differentiation protocols published so far show a high heterogeneity of neuronal populations in vitro. Here, we compared two previously published protocols to obtain hiPS cell-derived striatal neurons from both healthy donors and HD patients. Patch-clamp experiments, immunostaining and RT-qPCR were performed to characterize the neurons in culture. While the neurons were mature enough to fire action potentials, a majority failed to express markers typical for MSNs. Voltage-clamp experiments on voltage-gated sodium (Nav) channels revealed a large variability between the two differentiation protocols. Action potential analysis did not reveal changes induced by the HD mutation. This study attempts to demonstrate the current challenges in reproducing data of previously published differentiation protocols and in generating hiPS cell-derived striatal MSNs to model a genetic neurodegenerative disorder in vitro.


Assuntos
Técnicas de Cultura de Células , Diferenciação Celular , Doença de Huntington , Neurônios/fisiologia , Potenciais de Ação , Animais , Cálcio/metabolismo , Estudos de Casos e Controles , Linhagem Celular , Humanos , Células-Tronco Pluripotentes Induzidas , Camundongos Endogâmicos C57BL , Subunidade beta-4 do Canal de Sódio Disparado por Voltagem/metabolismo , Ácido gama-Aminobutírico/metabolismo
5.
Channels (Austin) ; 15(1): 208-228, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-33487118

RESUMO

Mutations in the voltage-gated sodium channel Nav1.7 are linked to human pain. The Nav1.7/N1245S variant was described before in several patients suffering from primary erythromelalgia and/or olfactory hypersensitivity. We have identified this variant in a pain patient and a patient suffering from severe and life-threatening orthostatic hypotension. In addition, we report a female patient suffering from muscle pain and carrying the Nav1.7/E1139K variant. We tested both Nav1.7 variants by whole-cell voltage-clamp recordings in HEK293 cells, revealing a slightly enhanced current density for the N1245S variant when co-expressed with the ß1 subunit. This effect was counteracted by an enhanced slow inactivation. Both variants showed similar voltage dependence of activation and steady-state fast inactivation, as well as kinetics of fast inactivation, deactivation, and use-dependency compared to WT Nav1.7. Finally, homology modeling revealed that the N1245S substitution results in different intramolecular interaction partners. Taken together, these experiments do not point to a clear pathogenic effect of either the N1245S or E1139K variant and suggest they may not be solely responsible for the patients' pain symptoms. As discussed previously for other variants, investigations in heterologous expression systems may not sufficiently mimic the pathophysiological situation in pain patients, and single nucleotide variants in other genes or modulatory proteins are necessary for these specific variants to show their effect. Our findings stress that biophysical investigations of ion channel mutations need to be evaluated with care and should preferably be supplemented with studies investigating the mutations in their context, ideally in human sensory neurons.


Assuntos
Canal de Sódio Disparado por Voltagem NAV1.7 , Eritromelalgia , Células HEK293 , Humanos , Potenciais da Membrana , Técnicas de Patch-Clamp
6.
J Biol Chem ; 296: 100227, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33361158

RESUMO

Mutations in voltage-gated sodium channels (Navs) can cause alterations in pain sensation, such as chronic pain diseases like inherited erythromelalgia. The mutation causing inherited erythromelalgia, Nav1.7 p.I848T, is known to induce a hyperpolarized shift in the voltage dependence of activation in Nav1.7. So far, however, the mechanism to explain this increase in voltage sensitivity remains unknown. In the present study, we show that phosphorylation of the newly introduced Thr residue explains the functional change. We expressed wildtype human Nav1.7, the I848T mutant, or other mutations in HEK293T cells and performed whole-cell patch-clamp electrophysiology. As the insertion of a Thr residue potentially creates a novel phosphorylation site for Ser/Thr kinases and because Nav1.7 had been shown in Xenopus oocytes to be affected by protein kinases C and A, we used different nonselective and selective kinase inhibitors and activators to test the effect of phosphorylation on Nav1.7 in a human system. We identify protein kinase C, but not protein kinase A, to be responsible for the phosphorylation of T848 and thereby for the shift in voltage sensitivity. Introducing a negatively charged amino acid instead of the putative phosphorylation site mimics the effect on voltage gating to a lesser extent. 3D modeling using the published cryo-EM structure of human Nav1.7 showed that introduction of this negatively charged site seems to alter the interaction of this residue with the surrounding amino acids and thus to influence channel function. These results could provide new opportunities for the development of novel treatment options for patients with chronic pain.


Assuntos
Potenciais da Membrana/fisiologia , Canal de Sódio Disparado por Voltagem NAV1.7/química , Proteína Quinase C/metabolismo , Processamento de Proteína Pós-Traducional , Treonina/química , Substituição de Aminoácidos , Sítios de Ligação , Dor Crônica/genética , Dor Crônica/metabolismo , Dor Crônica/fisiopatologia , Eritromelalgia/genética , Eritromelalgia/metabolismo , Eritromelalgia/fisiopatologia , Expressão Gênica , Células HEK293 , Humanos , Ativação do Canal Iônico/fisiologia , Isoleucina/química , Isoleucina/metabolismo , Modelos Moleculares , Mutação , Canal de Sódio Disparado por Voltagem NAV1.7/genética , Canal de Sódio Disparado por Voltagem NAV1.7/metabolismo , Técnicas de Patch-Clamp , Fosforilação/efeitos dos fármacos , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/genética , Inibidores de Proteínas Quinases/farmacologia , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Estaurosporina/farmacologia , Treonina/metabolismo
7.
Br J Pharmacol ; 177(19): 4481-4496, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32663327

RESUMO

BACKGROUND AND PURPOSE: The voltage-gated sodium channel Nav 1.7 is essential for adequate perception of painful stimuli. Mutations in the encoding gene, SCN9A, cause various pain syndromes in humans. The hNav 1.7/A1632E channel mutant causes symptoms of erythromelalgia and paroxysmal extreme pain disorder (PEPD), and its main gating change is a strongly enhanced persistent current. On the basis of recently published 3D structures of voltage-gated sodium channels, we investigated how the inactivation particle binds to the channel, how this mechanism is altered by the hNav 1.7/A1632E mutation, and how dimerization modifies function of the pain-linked mutation. EXPERIMENTAL APPROACH: We applied atomistic molecular simulations to demonstrate the effect of the mutation on channel fast inactivation. Native PAGE was used to demonstrate channel dimerization, and electrophysiological measurements in HEK cells and Xenopus laevis oocytes were used to analyze the links between functional channel dimerization and impairment of fast inactivation by the hNav 1.7/A1632E mutation. KEY RESULTS: Enhanced persistent current through hNav 1.7/A1632E channels was caused by impaired binding of the inactivation particle, which inhibits proper functioning of the recently proposed allosteric fast inactivation mechanism. hNav 1.7 channels form dimers and the disease-associated persistent current through hNav 1.7/A1632E channels depends on their functional dimerization status: Expression of the synthetic peptide difopein, a 14-3-3 inhibitor known to functionally uncouple dimers, decreased hNav 1.7/A1632E channel-induced persistent currents. CONCLUSION AND IMPLICATIONS: Functional uncoupling of mutant hNav 1.7/A1632E channel dimers restored their defective allosteric fast inactivation mechanism. Our findings support the concept of sodium channel dimerization and reveal its potential relevance for human pain syndromes.


Assuntos
Eritromelalgia , Canal de Sódio Disparado por Voltagem NAV1.7 , Humanos , Mutação , Canal de Sódio Disparado por Voltagem NAV1.7/genética , Dor , Fenótipo
8.
Pain ; 160(6): 1327-1341, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30720580

RESUMO

The chronic pain syndrome inherited erythromelalgia (IEM) is attributed to mutations in the voltage-gated sodium channel (NaV) 1.7. Still, recent studies targeting NaV1.7 in clinical trials have provided conflicting results. Here, we differentiated induced pluripotent stem cells from IEM patients with the NaV1.7/I848T mutation into sensory nociceptors. Action potentials in these IEM nociceptors displayed a decreased firing threshold, an enhanced upstroke, and afterhyperpolarization, all of which may explain the increased pain experienced by patients. Subsequently, we investigated the voltage dependence of the tetrodotoxin-sensitive NaV activation in these human sensory neurons using a specific prepulse voltage protocol. The IEM mutation induced a hyperpolarizing shift of NaV activation, which leads to activation of NaV1.7 at more negative potentials. Our results indicate that NaV1.7 is not active during subthreshold depolarizations, but that its activity defines the action potential threshold and contributes significantly to the action potential upstroke. Thus, our model system with induced pluripotent stem cell-derived sensory neurons provides a new rationale for NaV1.7 function and promises to be valuable as a translational tool to profile and develop more efficacious clinical analgesics.


Assuntos
Eritromelalgia/fisiopatologia , Células-Tronco Pluripotentes Induzidas/citologia , Canal de Sódio Disparado por Voltagem NAV1.7/metabolismo , Células Receptoras Sensoriais/metabolismo , Potenciais de Ação/efeitos dos fármacos , Estimulação Elétrica/métodos , Eritromelalgia/genética , Gânglios Espinais/citologia , Humanos , Potenciais da Membrana/efeitos dos fármacos , Canal de Sódio Disparado por Voltagem NAV1.7/genética , Nociceptores/fisiologia , Dor/diagnóstico , Dor/genética , Técnicas de Patch-Clamp/métodos , Tetrodotoxina/farmacologia
9.
Pflugers Arch ; 470(12): 1787-1801, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30099632

RESUMO

Mutations in voltage-gated sodium channels are associated with altered pain perception in humans. Most of these mutations studied to date present with a direct and intuitive link between the altered electrophysiological function of the channel and the phenotype of the patient. In this study, we characterize a variant of Nav1.8, D1639N, which has been previously identified in a patient suffering from the chronic pain syndrome "small fiber neuropathy". Using a heterologous expression system and patch-clamp analysis, we show that Nav1.8/D1639N reduces current density without altering biophysical gating properties of Nav1.8. Therefore, the D1639N variant causes a loss-of-function of the Nav1.8 sodium channel in a patient suffering from chronic pain. Using immunocytochemistry and biochemical approaches, we show that Nav1.8/D1639N impairs trafficking of the channel to the cell membrane. Neither co-expression of ß1 or ß3 subunit, nor overnight incubation at 27 °C rescued current density of the D1639N variant. On the other hand, overnight incubation with lidocaine fully restored current density of Nav1.8/D1639N most likely by overcoming the trafficking defect, whereas phenytoin failed to do so. Since lidocaine rescues the loss-of-function of Nav1.8/D1639N, it may offer a future therapeutic option for the patient carrying this variant. These results demonstrate that the D1639N variant, identified in a patient suffering from chronic pain, causes loss-of-function of the channel due to impaired cell surface trafficking and that this trafficking defect can be rescued by lidocaine.


Assuntos
Anestésicos Locais/farmacologia , Dor Crônica/genética , Lidocaína/farmacologia , Mutação com Perda de Função , Canal de Sódio Disparado por Voltagem NAV1.8/genética , Potenciais de Ação , Animais , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Membrana Celular/fisiologia , Humanos , Canal de Sódio Disparado por Voltagem NAV1.8/metabolismo , Transporte Proteico/efeitos dos fármacos , Xenopus
10.
Am J Pathol ; 185(8): 2132-42, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26216283

RESUMO

We have identified platelet-derived growth factor (PDGF)-CC as a potent profibrotic mediator in kidney fibrosis and pro-angiogenic mediator in glomeruli. Because renal fibrosis is associated with progressive capillary rarefaction, we asked whether PDGF-CC neutralization in fibrosis might have detrimental anti-angiogenic effects leading to aggravated peritubular capillary loss. We analyzed capillary rarefaction in mice with and without PDGF-CC neutralization (using genetically deficient mice and neutralizing antibodies), in three different models of renal interstitial fibrosis, unilateral ureteral obstruction, unilateral ischemia-reperfusion, Col4a3-deficient (Alport) mice, and healthy animals. Independent of the effect of PDGF-CC neutralization on renal fibrosis, we found no difference in capillary rarefaction between PDGF-CC-neutralized mice and mice with intact PDGF-CC. We also found no differences in microvascular leakage (determined by extravasation of Evans Blue Dye) and in renal relative blood volume quantified using in vivo microcomputed tomography. PDGF-CC neutralization had no effects on renal microvasculature in healthy animals. Capillary endothelium did not express PDGF receptor-α, suggesting that potential PDGF-CC effects would have to be indirect. PDGF-CC neutralization or deficiency was not associated with preservation or accelerated loss of peritubular capillaries, suggesting no significant pro-angiogenic effects of PDGF-CC during renal fibrosis. From a clinical perspective, the profibrotic effects of PDGF-CC outweigh the pro-angiogenic effects and, thus, do not limit a potential therapeutic use of PDGF-CC inhibition in renal fibrosis.


Assuntos
Capilares/metabolismo , Nefropatias/metabolismo , Rim/metabolismo , Linfocinas/metabolismo , Fator de Crescimento Derivado de Plaquetas/metabolismo , Animais , Capilares/patologia , Modelos Animais de Doenças , Fibrose/metabolismo , Fibrose/patologia , Rim/patologia , Nefropatias/patologia , Glomérulos Renais/metabolismo , Glomérulos Renais/patologia , Linfocinas/genética , Camundongos , Camundongos Knockout , Fator de Crescimento Derivado de Plaquetas/genética , Obstrução Ureteral/metabolismo , Obstrução Ureteral/patologia
11.
Nano Lett ; 10(1): 52-8, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19968235

RESUMO

Effective and specific targeting of nanoparticles is of paramount importance in the fields of targeted therapeutics and diagnostics. In the current study, we investigated the targeting efficacy of nanoparticles that were functionalized with two angiogenesis-specific targeting ligands, an alpha(v)beta(3) integrin-specific and a galectin-1-specific peptide. We show in vitro, using optical techniques and MRI, that the dual-targeting approach produces synergistic targeting effects, causing a dramatically elevated uptake of nanoparticles as compared to single ligand targeting.


Assuntos
Galectina 1/química , Integrina alfaVbeta3/química , Lipossomos , Imageamento por Ressonância Magnética/métodos , Nanotecnologia/métodos , Neovascularização Patológica , Inibidores da Angiogênese/farmacologia , Células Cultivadas , Humanos , Ligantes , Magnetismo , Microscopia Confocal/métodos , Microscopia de Fluorescência/métodos , Nanopartículas/química , Peptídeos/química
12.
FASEB J ; 21(2): 378-83, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17202248

RESUMO

Noninvasive diagnostic imaging methods to establish the efficacy of angiostatic therapies are becoming increasingly important with the first Food and Drug Administration approvals of such agents. Magnetic resonance molecular imaging is an imaging technique that allows the visualization of pathological processes in vivo with a better spatial resolution as compared with nuclear methods, such as photon emission tomography and single photon emission computed tomography. In this study, we used alpha(v)beta3 targeted bimodal liposomes to quantitate angiogenesis in a tumor mouse model with magnetic resonance imaging (MRI) and to evaluate the therapeutic efficacy of the angiogenesis inhibitors anginex and endostatin. The MRI findings were validated with fluorescence microscopy and showed a very good correlation with the microvessel density. In conclusion, this study provides evidence that molecular MRI can be used to noninvasively measure the efficacy of angiogenesis inhibitors during the course of therapy.


Assuntos
Inibidores da Angiogênese/farmacologia , Endostatinas/farmacologia , Imageamento por Ressonância Magnética/métodos , Melanoma/tratamento farmacológico , Proteínas/farmacologia , Inibidores da Angiogênese/administração & dosagem , Inibidores da Angiogênese/farmacocinética , Animais , Linhagem Celular Tumoral , Endostatinas/administração & dosagem , Endostatinas/farmacocinética , Lipossomos , Melanoma/irrigação sanguínea , Melanoma/patologia , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Neovascularização Patológica/patologia , Neovascularização Patológica/prevenção & controle , Peptídeos , Proteínas/administração & dosagem , Proteínas/farmacocinética , Fatores de Tempo , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...