Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Curr Top Membr ; 92: 199-231, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38007268

RESUMO

Cancer and neurodegenerative disease, albeit fundamental differences, share some common pathogenic mechanisms. Accordingly, both conditions are associated with aberrant cell proliferation and migration. Here, we review the causative role played by potassium (K+) channels, a fundamental class of proteins, in cancer and neurodegenerative disease. The concept that emerges from the review of the literature is that K+ channels can promote the development and progression of cancerous and neurodegenerative pathologies by dysregulating cell proliferation and migration. K+ channels appear to control these cellular functions in ways that not necessarily depend on their conducting properties and that involve the ability to directly or indirectly engage growth and survival signaling pathways. As cancer and neurodegenerative disease represent global health concerns, identifying commonalities may help understand the molecular basis for those devastating conditions and may facilitate the design of new drugs or the repurposing of existing drugs.


Assuntos
Neoplasias , Doenças Neurodegenerativas , Humanos , Canais de Potássio/metabolismo , Neoplasias/tratamento farmacológico , Transdução de Sinais , Proliferação de Células/fisiologia
2.
J Vis Exp ; (198)2023 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-37639634

RESUMO

The hypothalamus regulates fundamental metabolic processes by controlling functions as varied as food intake, body temperature, and hormone release. As the functions of the hypothalamus are controlled by specific subsets of neuronal populations, the ability to isolate them provides a major tool for studying metabolic mechanisms. In this regard, the neuronal complexity of the hypothalamus poses exceptional challenges. For these reasons, new techniques, such as Magnetic-Activated Cell Sorting (MACS), have been explored. This paper describes a new application of magnetic-activated cell sorting (MACS) using microbead technology to isolate a targeted neuronal population from prenatal mice brains. The technique is simple and guarantees a highly pure and viable primary hypothalamic neuron culture with high reproducibility. The hypothalamus is gently dissociated, neurons are selectively isolated and separated from glial cells, and finally, using a specific antibody for a cell surface marker, the population of interest is selected. Once isolated, targeted neurons can be used to investigate their morphological, electrical, and endocrine characteristics and their responses in normal or pathological conditions. Furthermore, given the variegated roles of the hypothalamus in regulating feeding, metabolism, stress, sleep, and motivation, a closer look at targeted and region-specific neurons may provide insight into their tasks in this complex environment.


Assuntos
Hipotálamo , Neurônios , Animais , Camundongos , Feminino , Gravidez , Reprodutibilidade dos Testes , Neuroglia , Anticorpos
3.
Neural Regen Res ; 18(11): 2365-2369, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37282454

RESUMO

Ion channels modulate cellular excitability by regulating ionic fluxes across biological membranes. Pathogenic mutations in ion channel genes give rise to epileptic disorders that are among the most frequent neurological diseases affecting millions of individuals worldwide. Epilepsies are triggered by an imbalance between excitatory and inhibitory conductances. However, pathogenic mutations in the same allele can give rise to loss-of-function and/or gain-of-function variants, all able to trigger epilepsy. Furthermore, certain alleles are associated with brain malformations even in the absence of a clear electrical phenotype. This body of evidence argues that the underlying epileptogenic mechanisms of ion channels are more diverse than originally thought. Studies focusing on ion channels in prenatal cortical development have shed light on this apparent paradox. The picture that emerges is that ion channels play crucial roles in landmark neurodevelopmental processes, including neuronal migration, neurite outgrowth, and synapse formation. Thus, pathogenic channel mutants can not only cause epileptic disorders by altering excitability, but further, by inducing morphological and synaptic abnormalities that are initiated during neocortex formation and may persist into the adult brain.

4.
Cell Death Differ ; 30(3): 687-701, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36207442

RESUMO

Potassium (K+) channels are robustly expressed during prenatal brain development, including in progenitor cells and migrating neurons, but their function is poorly understood. Here, we investigate the role of voltage-gated K+ channel KCNB1 (Kv2.1) in neocortical development. Neuronal migration of glutamatergic neurons was impaired in the neocortices of KCNB1 null mice. Migratory defects persisted into the adult brains, along with disrupted morphology and synaptic connectivity. Mice developed seizure phenotype, anxiety, and compulsive behavior. To determine whether defective KCNB1 can give rise to developmental channelopathy, we constructed Knock In (KI) mice, harboring the gene variant Kcnb1R312H (R312H mice) found in children with developmental and epileptic encephalopathies (DEEs). The R312H mice exhibited a similar phenotype to the null mice. Wild type (WT) and R312H KCNB1 channels made complexes with integrins α5ß5 (Integrin_K+ channel_Complexes, IKCs), whose biochemical signaling was impaired in R312H brains. Treatment with Angiotensin II in vitro, an agonist of Focal Adhesion kinase, a key component of IKC signaling machinery, corrected the neuronal abnormalities. Thus, a genetic mutation in a K+ channel induces severe neuromorphological abnormalities through non-conducting mechanisms, that can be rescued by pharmacological intervention. This underscores a previously unknown role of IKCs as key players in neuronal development, and implicate developmental channelopathies in the etiology of DEEs.


Assuntos
Epilepsia , Neocórtex , Animais , Camundongos , Epilepsia/genética , Integrinas/genética , Camundongos Knockout , Mutação , Canais de Potássio/genética
6.
Channels (Austin) ; 16(1): 185-197, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35942524

RESUMO

Started as an academic curiosity more than two decades ago, the idea that ion channels can regulate cellular processes in ways that do not depend on their conducting properties (non-ionic functions) gained traction and is now a flourishing area of research. Channels can regulate physiological processes including actin cytoskeletal remodeling, cell motility, excitation-contraction coupling, non-associative learning and embryogenesis, just to mention some, through non-ionic functions. When defective, non-ionic functions can give rise to channelopathies involved in cancer, neurodegenerative disease and brain trauma. Ion channels exert their non-ionic functions through a variety of mechanisms that range from physical coupling with other proteins, to possessing enzymatic activity, to assembling with signaling molecules. In this article, we take stock of the field and review recent findings. The concept that emerges, is that one of the most common ways through which channels acquire non-ionic attributes, is by assembling with integrins. These integrin-channel complexes exhibit broad genotypic and phenotypic heterogeneity and reveal a pleiotropic nature, as they appear to be capable of influencing both physiological and pathological processes.


Assuntos
Integrinas , Doenças Neurodegenerativas , Membrana Celular/metabolismo , Humanos , Integrinas/metabolismo , Canais Iônicos/metabolismo , Doenças Neurodegenerativas/metabolismo , Transdução de Sinais
7.
FASEB J ; 36(5): e22292, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35357039

RESUMO

Complexes formed with α5-integrins and the voltage-gated potassium (K+ ) channel KCNB1 (Kv2.1), known as IKCs, transduce the electrical activity at the plasma membrane into biochemical events that impinge on cytoskeletal remodeling, cell differentiation, and migration. However, when cells are subject to stress of oxidative nature IKCs turn toxic and cause inflammation and death. Here, biochemical, pharmacological, and cell viability evidence demonstrates that in response to oxidative insults, IKCs activate an apoptotic Mitogen-activated protein kinase/extracellular signal-regulated kinase (Ras-MAPK) signaling pathway. Simultaneously, wild-type (WT) KCNB1 channels sequester protein kinase B (Akt) causing dephosphorylation of BCL2-associated agonist of cell death (BAD), a major sentinel of apoptosis progression. In contrast, IKCs formed with C73A KCNB1 variant that does not induce apoptosis (IKCC73A ), do not sequester Akt and thus are able to engage cell survival mechanisms. Taken together, these data suggest that apoptotic and survival forces co-exist in IKCs. Integrins send death signals through Ras-MAPK and KCNB1 channels simultaneously sabotage survival mechanisms. Thus, the combined action of integrins and KCNB1 channels advances life or death.


Assuntos
Integrinas , Proteínas Proto-Oncogênicas c-akt , Apoptose/fisiologia , Sobrevivência Celular/fisiologia , Integrinas/fisiologia , Transdução de Sinais/fisiologia
8.
PLoS One ; 15(10): e0240255, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33035268

RESUMO

Biological organisms respond to environmental stressors by recruiting multiple cellular cascades that act to mitigate damage and ultimately enhance survival. This implies that compounds that interact with any of those pathways might improve organism's survival. Here, we report on an initial attempt to develop a drug screening assay based on the heat shock (HS) response of Caenorhabditis elegans nematodes. The protocol works by subjecting the worms to two HS conditions in the absence/presence of the test compounds. Post-heat shock survival is quantified manually or in semi-automatic manner by analyzing z-stack pictures. We blindly screened a cassette of 72 compounds in different developmental stages provided by Eli Lilly through their Open Innovation Drug Discovery program. The analysis indicated that, on average, therapeutically useful drugs increase survival to HS compared to compounds used in non-clinical settings. We developed a formalism that estimates the probability of a compound to enhance survival based on a comparison with a set of parameters calculated from a pool of 35 FDA-approved drugs. The method correctly identified the developmental stages of the Lilly compounds based on their relative abilities to enhance survival to the HS. Taken together these data provide proof of principle that an assay that measures the HS response of C. elegans can offer physiological and pharmacological insight in a cost- and time-efficient manner.


Assuntos
Bioensaio/métodos , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/fisiologia , Resposta ao Choque Térmico/fisiologia , Animais , Proteínas de Caenorhabditis elegans/genética , Avaliação Pré-Clínica de Medicamentos/métodos , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Resposta ao Choque Térmico/genética
9.
FASEB J ; 33(12): 14680-14689, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31682765

RESUMO

Voltage-gated potassium (K+) channel subfamily B member 1 (KCNB1, Kv2.1) and integrin-α5 form macromolecular complexes-named integrin-α5-KCNB1 complexes (IKCs)-in the human brain, but their function was poorly understood. Here we report that membrane depolarization triggered IKC intracellular signals mediated by small GTPases of the Ras subfamily and protein kinase B (Akt) to advance the development of filopodia and lamellipodia in Chinese hamster ovary cells, stimulate their motility, and enhance neurite outgrowth in mouse neuroblastoma Neuro2a cells. Five KCNB1 mutants (L211P, R312H G379R, G381R, and F416L) linked to severe infancy or early-onset epileptic encephalopathy exhibited markedly defective conduction. However, although L211P, G379R, and G381R normally engaged Ras/Akt and stimulated cell migration, R312H and F416L failed to activate Ras/Akt signaling and did not enhance cell migration. Taken together, these data suggest that IKCs modulate cellular plasticity via Ras and Akt signaling. As such, defective IKCs may cause epilepsy through mechanisms other than dysregulated excitability such as, for example, abnormal neuronal development and resulting synaptic connectivity.-Yu, W., Shin, M. R., Sesti, F. Complexes formed with integrin-α5 and KCNB1 potassium channel wild type or epilepsy-susceptibility variants modulate cellular plasticity via Ras and Akt signaling.


Assuntos
Epilepsia/genética , Integrina alfa5/metabolismo , Mutação , Plasticidade Neuronal , Canais de Potássio Shab/metabolismo , Animais , Células CHO , Linhagem Celular Tumoral , Movimento Celular , Cricetinae , Cricetulus , Camundongos , Neurônios/metabolismo , Neurônios/fisiologia , Ligação Proteica , Proteínas Proto-Oncogênicas c-akt/metabolismo , Canais de Potássio Shab/genética , Transdução de Sinais , Proteínas ras/metabolismo
10.
Cell Death Dis ; 10(10): 756, 2019 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-31591393

RESUMO

Since the publication of this article the authors have noted that there were two typos that could have caused confusion to the readers:1) References to "integrin alpha chain V" should have been "integrin alpha 5". These appear in the Abstract and the first paragraph of the Results section.2) In the Results section under the header "Integrins activate Fyn tyrosine kinases" and the legend of Figure 6a, "tyr530" should be replaced with "tyr416".3) In the the legend of Figure 6b, "pSrc" with should be replaced with "pFyn".

11.
Biochem Biophys Res Commun ; 512(4): 665-669, 2019 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-30922570

RESUMO

Voltage-gated potassium (K+) channel sub-family B member 1 (KCNB1, Kv2.1) is known to undergo oxidation-induced oligomerization during aging but whether this process affects brain's physiology was not known. Here, we used 10, 16 and 22 month-old transgenic mice overexpressing a KCNB1 variant that does not oligomerize (Tg-C73A) and as control, mice overexpressing the wild type (Tg-WT) channel and non-transgenic (non-Tg) mice to elucidate the effects of channel's oxidation on cognitive function. Aging mice in which KCNB1 oligomerization is negligible (Tg-C73A), performed significantly better in the Morris Water Maze (MWM) test of working memory compared to non-Tg or Tg-WT mice. KCNB1 and synapsin-1 co-immunoprecipitated and the cognitive impairment in the MWM was associated with moderate loss of synapsin-1 in pre-synaptic structures of the hippocampus, whereas neurodegeneration and neuronal loss were not significantly different in the various genotypes. We conclude that moderate oxidation of the KCNB1 channel during aging can influence neuronal networks by affecting synaptic function.


Assuntos
Envelhecimento , Disfunção Cognitiva/metabolismo , Estresse Oxidativo , Canais de Potássio Shab/metabolismo , Animais , Disfunção Cognitiva/genética , Disfunção Cognitiva/fisiopatologia , Expressão Gênica , Variação Genética , Humanos , Memória de Curto Prazo , Camundongos , Camundongos Transgênicos , Oxirredução , Multimerização Proteica , Canais de Potássio Shab/química , Canais de Potássio Shab/genética
12.
Cell Death Dis ; 9(8): 820, 2018 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-30050035

RESUMO

Oxidative modification of the voltage-gated K+ channel subfamily B member 1 (KCNB1, Kv2.1) is emerging as a mechanism of neuronal vulnerability potentially capable of affecting multiple conditions associated with oxidative stress, from normal aging to neurodegenerative disease. In this study we report that oxidation of KCNB1 channels is exacerbated in the post mortem brains of Alzheimer's disease (AD) donors compared to age-matched controls. In addition, phosphorylation of Focal Adhesion kinases (FAK) and Src tyrosine kinases, two key signaling steps that follow KCNB1 oxidation, is also strengthened in AD vs. control brains. Quadruple transgenic mice expressing a non-oxidizable form of KCNB1 in the 3xTg-AD background (APPSWE, PS1M146V, and tauP301L), exhibit improved working memory along with reduced brain inflammation, protein carbonylation and intraneuronal ß-amyloid (Aß) compared to 3xTg-AD mice or mice expressing the wild type (WT) KCNB1 channel. We conclude that oxidation of KCNB1 channels is a mechanism of neuronal vulnerability that is pervasive in the vertebrate brain.


Assuntos
Doença de Alzheimer/patologia , Encéfalo/metabolismo , Canais de Potássio Shab/metabolismo , Idoso de 80 Anos ou mais , Doença de Alzheimer/metabolismo , Doença de Alzheimer/prevenção & controle , Peptídeos beta-Amiloides/metabolismo , Animais , Dasatinibe/uso terapêutico , Modelos Animais de Doenças , Feminino , Quinase 1 de Adesão Focal/metabolismo , Gliose , Humanos , Masculino , Memória de Curto Prazo , Camundongos , Camundongos Transgênicos , Estresse Oxidativo , Carbonilação Proteica , Canais de Potássio Shab/química , Canais de Potássio Shab/genética , Quinases da Família src/metabolismo
13.
Biochem Biophys Res Commun ; 492(3): 338-342, 2017 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-28859988

RESUMO

Biological systems are highly sensitive to changes in their environment. Indeed, the molecular basis of the environmental stress response suggests that the specialized stress responses share more commonalities than previously believed. Here, we used the nematode C. elegans to gain insight into the role of Rho signaling during two common environmental challenges, oxidative and thermal stress. In response to heat shock (HS), wild type (N2) worms demonstrated reduced viability which was rescued by genetic suppression of CDC42 and RHO-1. Visualization of F-actin by phalloidin-rhodamine underscored a strict correlation between the levels of F-actin following GTPase suppression and survival. Additionally, genetic ablation of OSG-1, a Guanine Nucleotide Exchange Factor (GEF) previously implicated in oxidative stress, was associated with constitutively lower levels of F-actin and increased mortality. However, upon an oxidative insult F-actin stability decreased in N2 worms, a rescue of this affect was observed in OSG-1 null worms, consistent with the resistance exhibited by these worms to oxidative stress (OS). Together these data suggest that during conditions of thermal or oxidative stress Rho signaling promotes vulnerability by altering actin dynamics. Thus, the stability of the actin cytoskeleton, in part through a conserved mechanism mediated by Rho signaling, is a crucial factor for the cell's survival to environmental challenges.


Assuntos
Caenorhabditis elegans/citologia , Caenorhabditis elegans/metabolismo , Citoesqueleto/metabolismo , Estresse Oxidativo , Temperatura , Proteínas rho de Ligação ao GTP/metabolismo , Animais , Caenorhabditis elegans/enzimologia
14.
Cell Death Dis ; 8(4): e2737, 2017 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-28383553

RESUMO

Oxidative modification of the voltage-gated potassium (K+) channel KCNB1 promotes apoptosis in the neurons of cortex and hippocampus through a signaling pathway mediated by Src tyrosine kinases. How oxidation of the channel is transduced into Src recruitment and activation, however, was not known. Here we show that the apoptotic signal originates from integrins, which form macromolecular complexes with KCNB1 channels. The initial stimulus is transduced to Fyn and possibly other Src family members by focal adhesion kinase (FAK). Thus KCNB1 and integrin alpha chain V (integrin-α5) coimmunoprecipitated in the mouse brain and these interactions were retained upon channel's oxidation. Pharmacological inhibition of integrin signaling or FAK suppressed apoptosis induced by oxidation of KCNB1, as well as FAK and Src/Fyn activation. Most importantly, the activation of the integrin-FAK-Src/Fyn cascade was negligible in the presence of non-oxidizable C73A KCNB1 mutant channels, even though they normally interacted with integrin-α5. This leads us to conclude that the transition between the non-oxidized and oxidized state of KCNB1 activates integrin signaling. KCNB1 oxidation may favor integrin clustering, thereby facilitating the recruitment and activation of FAK and Src/Fyn kinases.


Assuntos
Apoptose/fisiologia , Encéfalo/metabolismo , Integrina alfa5/metabolismo , Canais de Potássio Shab/metabolismo , Transdução de Sinais/fisiologia , Substituição de Aminoácidos , Animais , Células CHO , Cricetinae , Cricetulus , Quinase 1 de Adesão Focal/genética , Quinase 1 de Adesão Focal/metabolismo , Integrina alfa5/genética , Camundongos , Mutação de Sentido Incorreto , Proteínas Proto-Oncogênicas c-fyn/genética , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Canais de Potássio Shab/genética , Quinases da Família src/genética , Quinases da Família src/metabolismo
15.
J Neurosci ; 36(43): 11084-11096, 2016 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-27798188

RESUMO

The delayed rectifier potassium (K+) channel KCNB1 (Kv2.1), which conducts a major somatodendritic current in cortex and hippocampus, is known to undergo oxidation in the brain, but whether this can cause neurodegeneration and cognitive impairment is not known. Here, we used transgenic mice harboring human KCNB1 wild-type (Tg-WT) or a nonoxidable C73A mutant (Tg-C73A) in cortex and hippocampus to determine whether oxidized KCNB1 channels affect brain function. Animals were subjected to moderate traumatic brain injury (TBI), a condition characterized by extensive oxidative stress. Dasatinib, a Food and Drug Administration-approved inhibitor of Src tyrosine kinases, was used to impinge on the proapoptotic signaling pathway activated by oxidized KCNB1 channels. Thus, typical lesions of brain injury, namely, inflammation (astrocytosis), neurodegeneration, and cell death, were markedly reduced in Tg-C73A and dasatinib-treated non-Tg animals. Accordingly, Tg-C73A mice and non-Tg mice treated with dasatinib exhibited improved behavioral outcomes in motor (rotarod) and cognitive (Morris water maze) assays compared to controls. Moreover, the activity of Src kinases, along with oxidative stress, were significantly diminished in Tg-C73A brains. Together, these data demonstrate that oxidation of KCNB1 channels is a contributing mechanism to cellular and behavioral deficits in vertebrates and suggest a new therapeutic approach to TBI. SIGNIFICANCE STATEMENT: This study provides the first experimental evidence that oxidation of a K+ channel constitutes a mechanism of neuronal and cognitive impairment in vertebrates. Specifically, the interaction of KCNB1 channels with reactive oxygen species plays a major role in the etiology of mouse model of traumatic brain injury (TBI), a condition associated with extensive oxidative stress. In addition, a Food and Drug Administration-approved drug ameliorates the outcome of TBI in mouse, by directly impinging on the toxic pathway activated in response to oxidation of the KCNB1 channel. These findings elucidate a basic mechanism of neurotoxicity in vertebrates and might lead to a new therapeutic approach to TBI in humans, which, despite significant efforts, is a condition that remains without effective pharmacological treatments.


Assuntos
Lesões Encefálicas Traumáticas/fisiopatologia , Transtornos Cognitivos/fisiopatologia , Hipocampo/fisiopatologia , Neurônios/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Canais de Potássio Shab/metabolismo , Animais , Apoptose , Lesões Encefálicas Traumáticas/patologia , Transtornos Cognitivos/patologia , Dasatinibe/administração & dosagem , Hipocampo/patologia , Masculino , Camundongos , Camundongos Transgênicos , Doenças Neurodegenerativas , Neurônios/patologia , Oxirredução/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Inibidores de Proteínas Quinases/administração & dosagem
16.
Aging Dis ; 7(2): 130-5, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27114846

RESUMO

Reversible regulation of proteins by reactive oxygen species (ROS) is an important mechanism of neuronal plasticity. In particular, ROS have been shown to act as modulatory molecules of ion channels-which are key to neuronal excitability-in several physiological processes. However ROS are also fundamental contributors to aging vulnerability. When the level of excess ROS increases in the cell during aging, DNA is damaged, proteins are oxidized, lipids are degraded and more ROS are produced, all culminating in significant cell injury. From this arose the idea that oxidation of ion channels by ROS is one of the culprits for neuronal aging. Aging-dependent oxidative modification of voltage-gated potassium (K(+)) channels was initially demonstrated in the nematode Caenorhabditis elegans and more recently in the mammalian brain. Specifically, oxidation of the delayed rectifier KCNB1 (Kv2.1) and of Ca(2+)- and voltage sensitive K(+) channels have been established suggesting that their redox sensitivity contributes to altered excitability, progression of healthy aging and of neurodegenerative disease. Here I discuss the implications that oxidation of K(+) channels by ROS may have for normal aging, as well as for neurodegenerative disease.

17.
Brain Res ; 1639: 174-85, 2016 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-26947620

RESUMO

Ion channels are integral membrane proteins that allow passive diffusion of ions across membranes. In neurons and in other excitable cells, the harmonious coordination between the numerous types of ion channels shape and propagate electrical signals. Increased accumulation of reactive oxidative species (ROS), and subsequent oxidation of proteins, including ion channels, is a hallmark feature of aging and may contribute to cell failure as a result. In this review we discuss the effects of ROS on three major types of ion channels of the central nervous system, namely the potassium (K(+)), calcium (Ca(2+)) and sodium (Na(+)) channels. We examine two general mechanisms through which ROS affect ion channels: via direct oxidation of specific residues and via indirect interference of pathways that regulate the channels. The overall status of the present studies indicates that the interaction of ion channels with ROS is multimodal and pervasive in the central nervous system and likely constitutes a general mechanism of aging susceptibility.


Assuntos
Envelhecimento/metabolismo , Sistema Nervoso Central/metabolismo , Canais Iônicos/metabolismo , Animais , Humanos , Oxirredução , Estresse Oxidativo/fisiologia
18.
Genetics ; 199(2): 487-96, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25527286

RESUMO

Rho signaling regulates a variety of biological processes, but whether it is implicated in aging remains an open question. Here we show that a guanine nucleotide exchange factor of the Dbl family, OSG-1, confers functional aging by dysregulating Rho GTPases activities in C. elegans. Thus, gene reporter analysis revealed widespread OSG-1 expression in muscle and neurons. Loss of OSG-1 gene function was not associated with developmental defects. In contrast, suppression of OSG-1 lessened loss of function (chemotaxis) in ASE sensory neurons subjected to conditions of oxidative stress generated during natural aging, by oxidative challenges, or by genetic mutations. RNAi analysis showed that OSG-1 was specific toward activation of RHO-1 GTPase signaling. RNAi further implicated actin-binding proteins ARX-3 and ARX-5, thus the actin cytoskeleton, as one of the targets of OSG-1/RHO-1 signaling. Taken together these data suggest that OSG-1 is recruited under conditions of oxidative stress, a hallmark of aging, and contributes to promote loss of neuronal function by affecting the actin cytoskeleton via altered RHO-1 activity.


Assuntos
Envelhecimento/genética , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Neurônios/metabolismo , Transdução de Sinais , Proteínas rho de Ligação ao GTP/metabolismo , Peptídeos beta-Amiloides/genética , Animais , Animais Geneticamente Modificados , Mapeamento Cromossômico , Dendritos/metabolismo , Expressão Gênica , Genes Reporter , Loci Gênicos , Humanos , Longevidade/genética , Músculos/metabolismo , Estresse Oxidativo , Fenótipo , Ligação Proteica
19.
World J Biol Chem ; 5(2): 85-92, 2014 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-24921000

RESUMO

KCNB1, a voltage-gated potassium (K(+)) channel that conducts a major delayed rectifier current in the brain, pancreas and cardiovascular system is a key player in apoptotic programs associated with oxidative stress. As a result, this protein represents a bona fide drug target for limiting the toxic effects of oxygen radicals. Until recently the consensus view was that reactive oxygen species trigger a pro-apoptotic surge in KCNB1 current via phosphorylation and SNARE-dependent incorporation of KCNB1 channels into the plasma membrane. However, new evidence shows that KCNB1 can be modified by oxidants and that oxidized KCNB1 channels can directly activate pro-apoptotic signaling pathways. Hence, a more articulated picture of the pro-apoptotic role of KCNB1 is emerging in which the protein induces cell's death through distinct molecular mechanisms and activation of multiple pathways. In this review article we discuss the diverse functional, toxic and protective roles that KCNB1 channels play in the major organs where they are expressed.

20.
PLoS One ; 8(11): e79790, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24223195

RESUMO

BACKGROUND: Aging is a biological process strongly determined by genetics. However, only a few single nucleotide polymorphisms (SNPs) have been reported to be consistently associated with aging. While investigating whether copy number variations (CNVs) could fill this gap, we focused on CNVs that have not been studied in previous SNP-based searches via tagging SNPs. METHODS: TaqMan qPCR assays were developed to quantify 20 common CNVs in 222 senior American Caucasians in order to reveal possible association with longevity. The replication study was comprised of 1283 community-dwelling senior European Caucasians. Replicated CNVs were further investigated for association with healthy aging and aging-related diseases, while association with longevity was additionally tested in Caenorhabditis elegans. RESULTS: In the discovery study of ≥80 vs.<80 years old seniors, a homozygous intronic CNV deletion in the CNTNAP4 gene was inversely associated with survival to the age of 80 (OR=0.51, 95%CI 0.29-0.87, p=0.015 before correction for multiple testing). After stratification by sex, association remained significant in females (OR=0.41, 95%CI 0.21-0.77, p=0.007), but not in males (OR=0.97, 95%CI 0.33-2.79, p=1). The finding was validated in a replication study (OR=0.66, 95%CI 0.48-0.90, p=0.011 for females). CNTNAP4 association with longevity was supported by a marked 25% lifespan change in C. elegans after knocking down the ortholog gene. An inverse association of the CNV del/del variant with female healthy aging was observed (OR=0.39, 95%CI 0.19-0.76, p=0.006). A corresponding positive association with aging-related diseases was revealed for cognitive impairment (OR=2.17, 95%CI 1.11-4.22, p=0.024) and, in independent studies, for Alzheimer's (OR=4.07, 95%CI 1.17-14.14, p=0.036) and Parkinson's (OR=1.59, 95%CI 1.03-2.42, p=0.041) diseases. CONCLUSION: This is the first demonstration for association of the CNTNAP4 gene and one of its intronic CNV polymorphisms with aging. Association with particular aging-related diseases awaits replication and independent validation.


Assuntos
Envelhecimento/genética , Variações do Número de Cópias de DNA/genética , Proteínas do Tecido Nervoso/genética , Idoso de 80 Anos ou mais , Envelhecimento/fisiologia , Feminino , Estudo de Associação Genômica Ampla , Humanos , Longevidade/genética , Masculino , Modelos Teóricos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA