Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
1.
J Neurosci ; 42(4): 552-566, 2022 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-34872928

RESUMO

Fluorescence imaging is an indispensable method for analysis of diverse cellular and molecular processes, enabling, for example, detection of ions, second messengers, or metabolites. Intensity-based approaches, however, are prone to artifacts introduced by changes in fluorophore concentrations. This drawback can be overcome by fluorescence lifetime imaging (FLIM) based on time-correlated single-photon counting. FLIM often necessitates long photon collection times, resulting in strong temporal binning of dynamic processes. Recently, rapidFLIM was introduced, exploiting ultra-low dead-time photodetectors together with rapid electronics. Here, we demonstrate the applicability of rapidFLIM, combined with new and improved correction schemes, for spatiotemporal fluorescence lifetime imaging of low-emission fluorophores in a biological system. Using tissue slices of hippocampi of mice of either sex, loaded with the Na+ indicator ING2, we show that improved rapidFLIM enables quantitative, dynamic imaging of neuronal Na+ signals at a full-frame temporal resolution of 0.5 Hz. Induction of transient chemical ischemia resulted in unexpectedly large Na+ influx, accompanied by considerable cell swelling. Both Na+ loading and cell swelling were dampened on inhibition of TRPV4 channels. Together, rapidFLIM enabled the spatiotemporal visualization and quantification of neuronal Na+ transients at unprecedented speed and independent from changes in cell volume. Moreover, our experiments identified TRPV4 channels as hitherto unappreciated contributors to neuronal Na+ loading on metabolic failure, suggesting this pathway as a possible target to ameliorate excitotoxic damage. Finally, rapidFLIM will allow faster and more sensitive detection of a wide range of dynamic signals with other FLIM probes, most notably those with intrinsic low-photon emission.SIGNIFICANCE STATEMENT FLIM is an indispensable method for analysis of cellular processes. FLIM often necessitates long photon collection periods, requiring the sacrifice of temporal resolution at the expense of spatial information. Here, we demonstrate the applicability of the recently introduced rapidFLIM for quantitative, dynamic imaging with low-emission fluorophores in brain slices. RapidFLIM, combined with improved correction schemes, enabled intensity-independent recording of neuronal Na+ transients at unprecedented full-frame rates of 0.5 Hz. It also allowed quantitative imaging independent from changes in cell volume, revealing a surprisingly strong and hitherto uncovered contribution of TRPV4 channels to Na+ loading on energy failure. Collectively, our study thus provides a novel, unexpected insight into the mechanisms that are responsible for Na+ changes on energy depletion.


Assuntos
Isquemia Encefálica/metabolismo , Neurônios/metabolismo , Imagem Óptica/métodos , Sódio/metabolismo , Canais de Cátion TRPV/metabolismo , Animais , Isquemia Encefálica/patologia , Feminino , Hipocampo/metabolismo , Hipocampo/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Neurônios/química , Técnicas de Cultura de Órgãos , Canais de Cátion TRPV/análise
2.
Biol Pharm Bull ; 45(11): 1684-1691, 2022 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-35989293

RESUMO

Pulmonary arterial hypertension (PAH) is characterized by vascular remodeling of the pulmonary artery, which is mainly attributed to the excessive proliferation of pulmonary arterial smooth muscle cells (PASMCs) comprising the medial layer of pulmonary arteries. The activity of ion channels associated with cytosolic Ca2+ signaling regulates the pathogenesis of PAH. Limited information is currently available on the role of Cl- channels in PASMCs. Therefore, the functional expression of ClC3 channels/transporters was herein investigated in the PASMCs of normal subjects and patients with idiopathic pulmonary arterial hypertension (IPAH). Expression analyses revealed the upregulated expression of ClC3 channels/transporters at the mRNA and protein levels in IPAH-PASMCs. Hypoosmotic perfusion (230 mOsm) evoked swelling-activated Cl- currents (ICl-swell) in normal-PASMCs, whereas 100 µM 4,4'-diisothiocyanatostilbene-2,2'-disulfonic acid (DIDS) exerted the opposite effects. The small interfering RNA (siRNA) knockdown of ClC3 did not affect ICl-swell. On the other hand, ICl-swell was larger in IPAH-PASMCs and inhibited by DIDS and the siRNA knockdown of ClC3. IPAH-PASMCs grew more than normal-PASMCs. The growth of IPAH-PASMCs was suppressed by niflumic acid and DIDS, but not by 9-anthracenecarboxylic acid or T16Ainh-A01. The siRNA knockdown of ClC3 also inhibited the proliferation of IPAH-PASMCs. Collectively, the present results indicate that upregulated ClC3 channels/transporters are involved in ICl-swell and the excessive proliferation of IPAH-PASMCs, thereby contributing to the pathogenesis of PAH. Therefore, ClC3 channels/transporters have potential as a target of therapeutic drugs for the treatment of PAH.


Assuntos
Miócitos de Músculo Liso , Humanos , Hipertensão Pulmonar Primária Familiar/tratamento farmacológico , Hipertensão Pulmonar Primária Familiar/genética , Hipertensão Pulmonar Primária Familiar/patologia , Ácido 4,4'-Di-Isotiocianoestilbeno-2,2'-Dissulfônico/farmacologia , RNA Interferente Pequeno/farmacologia , Miócitos de Músculo Liso/metabolismo , Proliferação de Células , Células Cultivadas
3.
Mikrochim Acta ; 189(11): 423, 2022 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-36255523

RESUMO

A novel ratiometric electrochemiluminescence (ECL) system based on gold nanostars (AuNSs) support was constructed for the determination of hypotonicity-induced ATP release from HepG2 cells. AuNS@Lu nanoprobe was used as anodic luminophore and K2S2O8 as cathodic luminophore as well as anodic co-reactant. AuNS with the large specific surface was adopted to adsorb plentiful luminol to form solid-state probe and as affinity support to immobilize ATP aptamer (Apt). The obtained nanocomposite (Apt-AuNS@Lu) generated a strong ECL signal at + 0.4 V (vs. Ag/AgCl) with co-reactant K2S2O8, because of excellent conductivity and catalytic activity of AuNS. Furthermore, graphene oxide was reduced onto indium tin oxide (ITO) electrodes to facilitate the electron transfer. Following, polydopamine (PDA) film was formed via self-polymerization, improving stability and adhesion of the electrode surface. To immobilize ATP capture aptamer (AptC), abounding AuNSs were attached to RGO/PDA surface. When the sensor was incubated in the mixture solution of Apt-AuNS@Lu and target ATP, the ECL signal of Apt-AuNS@Lu increased with the increase of ATP concentration, meanwhile, the signal of K2S2O8 declined. The ratio of the two luminophores was used for the quantitative determination of ATP. The linear range was 5 to 250 nM, and the limit of detection was 1.4 nM at (3σ)/S. The method was successfully applied to analyze ATP release from HepG2 cells stimulated by 0.45% NaCl hypotonic solution. The results showed that the release kinetics profile of ATP had a sigmoidal shape with rapid release within 10 min and then slowed. Compared to the isotonic groups, the intracellular ATP concentration was 3.7 ± 0.3 µM (n = 3) decreasing by 40.3% and the extracellular was 23.4 ± 1.2 nM (n = 3) increasing by 9.2 times in the hypotonicity for 10 min, which showed ATP release from cells and good agreement with commercial ELISA test. The proposed strategy would be beneficial to broadening application of ECL technology in studying cell biological functions.


Assuntos
Luminol , Nanopartículas Metálicas , Medições Luminescentes , Cloreto de Sódio , Soluções Hipotônicas , Ouro , Trifosfato de Adenosina/análise
4.
Glia ; 69(1): 28-41, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32506554

RESUMO

The mammalian brain consists of 80% water, which is continuously shifted between different compartments and cellular structures by mechanisms that are, to a large extent, unresolved. Aquaporin 4 (AQP4) is abundantly expressed in glia and ependymal cells of the mammalian brain and has been proposed to act as a gatekeeper for brain water dynamics, predominantly based on studies utilizing AQP4-deficient mice. However, these mice have a range of secondary effects due to the gene deletion. An efficient and selective AQP4 inhibitor has thus been sorely needed to validate the results obtained in the AQP4-/- mice to quantify the contribution of AQP4 to brain fluid dynamics. In AQP4-expressing Xenopus laevis oocytes monitored by a high-resolution volume recording system, we here demonstrate that the compound TGN-020 is such a selective AQP4 inhibitor. TGN-020 targets the tested species of AQP4 with an IC50 of ~3.5 µM, but displays no inhibitory effect on the other AQPs (AQP1-AQP9). With this tool, we employed rat hippocampal slices and ion-sensitive microelectrodes to determine the role of AQP4 in glia cell swelling following neuronal activity. TGN-020-mediated inhibition of AQP4 did not prevent stimulus-induced extracellular space shrinkage, nor did it slow clearance of the activity-evoked K+ transient. These data, obtained with a verified isoform-selective AQP4 inhibitor, indicate that AQP4 is not required for the astrocytic contribution to the K+ clearance or the associated extracellular space shrinkage.


Assuntos
Neuroglia , Animais , Aquaporina 4/genética , Aquaporinas , Astrócitos/metabolismo , Edema , Camundongos , Neuroglia/metabolismo , Isoformas de Proteínas , Ratos , Água/metabolismo
5.
Biol Chem ; 402(9): 1033-1045, 2021 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-33915604

RESUMO

Liver cell hydration (cell volume) is dynamic and can change within minutes under the influence of hormones, nutrients, and oxidative stress. Such volume changes were identified as a novel and important modulator of cell function. It provides an early example for the interaction between a physical parameter (cell volume) on the one hand and metabolism, transport, and gene expression on the other. Such events involve mechanotransduction (osmosensing) which triggers signaling cascades towards liver function (osmosignaling). This article reviews our own work on this topic with emphasis on the role of ß1 integrins as (osmo-)mechanosensors in the liver, but also on their role in bile acid signaling.


Assuntos
Mecanotransdução Celular , Ácidos e Sais Biliares , Tamanho Celular , Hepatócitos , Integrinas , Transdução de Sinais
6.
Arch Biochem Biophys ; 704: 108891, 2021 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-33901485

RESUMO

A few Bacillus thuringiensis Cry proteins, known as parasporins, have demonstrated cell proliferation inhibition of human cancer cells in vitro after protease activation. In this work, eight peptides derived from the Cry11Bb protoxin produced by B. thuringiensis subsp. medellin were selected and evaluated to investigate their membrane permeabilization and cytolytic activities, using red blood cells and cancer cell lines A549, MCF-7 and Caco-2, respectively. The most active peptides permeabilized red blood cells in a membrane potential-dependent manner. Half maximal inhibitory concentration in cancer cells was in the range 0.78-7.63 µM. At the same time, at peptides concentration of 25 µM, the hemolysis percentage varied in the range of 4.6-32.4%. The peptides BTM-P1 and BTM-P4 in D form had the lowest IC50 values on the MCF-7 cell line and they are considered as the most promising peptides among the evaluated. Fluorescence microscopy using AnnexinV-FLUOS staining indicates that the possible cause of MCF-7 cell death by peptide BTM-P1, is apoptosis. Real time PCR analysis showed an increased transcription of p53 in MCF-7 cells, thus confirming the probable pro-apoptotic effect of the peptide BTM-P1. In general, this study suggests that the cytolytic activity of the polycationic peptides derived from the Cry11Bb protoxin could be mediated by a pro-apoptotic mechanism that might include potential-dependent membrane permeabilization. Further studies might be accomplished to establish whether the peptides are cytolytic to other cancer cell lines and to solid tumors.


Assuntos
Bacillus thuringiensis/química , Proteínas de Bactérias/química , Toxinas Bacterianas/química , Citotoxinas , Membrana Eritrocítica/metabolismo , Hemólise/efeitos dos fármacos , Peptídeos , Transcrição Gênica/efeitos dos fármacos , Proteína Supressora de Tumor p53/biossíntese , Células A549 , Células CACO-2 , Citotoxinas/química , Citotoxinas/farmacologia , Humanos , Células MCF-7 , Peptídeos/química , Peptídeos/farmacologia
7.
Int J Mol Sci ; 22(11)2021 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-34073593

RESUMO

Ischemic stroke is a leading cause of mortality and chronic disability. Either recovery or progression towards irreversible failure of neurons and astrocytes occurs within minutes to days, depending on remaining perfusion levels. Initial damage arises from energy depletion resulting in a failure to maintain homeostasis and ion gradients between extra- and intracellular spaces. Astrocytes play a key role in these processes and are thus central players in the dynamics towards recovery or progression of stroke-induced brain damage. Here, we present a synopsis of the pivotal functions of astrocytes at the tripartite synapse, which form the basis of physiological brain functioning. We summarize the evidence of astrocytic failure and its consequences under ischemic conditions. Special emphasis is put on the homeostasis and stroke-induced dysregulation of the major monovalent ions, namely Na+, K+, H+, and Cl-, and their involvement in maintenance of cellular volume and generation of cerebral edema.


Assuntos
Astrócitos/metabolismo , Edema Encefálico/metabolismo , Lesões Encefálicas/metabolismo , Homeostase , Acidente Vascular Cerebral/metabolismo , Astrócitos/patologia , Edema Encefálico/patologia , Lesões Encefálicas/patologia , Humanos , Transporte de Íons , Acidente Vascular Cerebral/patologia
8.
J Biol Chem ; 294(48): 18421-18434, 2019 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-31619514

RESUMO

Many retinal diseases are associated with pathological cell swelling, but the underlying etiology remains to be established. A key component of the volume-sensitive machinery, the transient receptor potential vanilloid 4 (TRPV4) ion channel, may represent a sensor and transducer of cell swelling, but the molecular link between the swelling and TRPV4 activation is unresolved. Here, our results from experiments using electrophysiology, cell volumetric measurements, and fluorescence imaging conducted in murine retinal cells and Xenopus oocytes indicated that cell swelling in the physiological range activated TRPV4 in Müller glia and Xenopus oocytes, but required phospholipase A2 (PLA2) activity exclusively in Müller cells. Volume-dependent TRPV4 gating was independent of cytoskeletal rearrangements and phosphorylation. Our findings also revealed that TRPV4-mediated transduction of volume changes is dependent by its N terminus, more specifically by its distal-most part. We conclude that the volume sensitivity and function of TRPV4 in situ depend critically on its functional and cell type-specific interactions.


Assuntos
Células Ependimogliais/metabolismo , Ativação do Canal Iônico/fisiologia , Neuroglia/metabolismo , Oócitos/metabolismo , Canais de Cátion TRPV/metabolismo , Animais , Tamanho Celular , Células Ependimogliais/citologia , Feminino , Ativação do Canal Iônico/genética , Camundongos , Neuroglia/citologia , Neurônios/citologia , Neurônios/metabolismo , Oócitos/citologia , Técnicas de Patch-Clamp , Fosfolipases A2/metabolismo , Fosforilação , Ratos , Canais de Cátion TRPV/genética , Xenopus laevis
9.
Glia ; 68(11): 2192-2211, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32181522

RESUMO

Neuronal signaling in the central nervous system (CNS) associates with release of K+ into the extracellular space resulting in transient increases in [K+ ]o . This elevated K+ is swiftly removed, in part, via uptake by neighboring glia cells. This process occurs in parallel to the [K+ ]o elevation and glia cells thus act as K+ sinks during the neuronal activity, while releasing it at the termination of the pulse. The molecular transport mechanisms governing this glial K+ absorption remain a point of debate. Passive distribution of K+ via Kir4.1-mediated spatial buffering of K+ has become a favorite within the glial field, although evidence for a quantitatively significant contribution from this ion channel to K+ clearance from the extracellular space is sparse. The Na+ /K+ -ATPase, but not the Na+ /K+ /Cl- cotransporter, NKCC1, shapes the activity-evoked K+ transient. The different isoform combinations of the Na+ /K+ -ATPase expressed in glia cells and neurons display different kinetic characteristics and are thereby distinctly geared toward their temporal and quantitative contribution to K+ clearance. The glia cell swelling occurring with the K+ transient was long assumed to be directly associated with K+ uptake and/or AQP4, although accumulating evidence suggests that they are not. Rather, activation of bicarbonate- and lactate transporters appear to lead to glial cell swelling via the activity-evoked alkaline transient, K+ -mediated glial depolarization, and metabolic demand. This review covers evidence, or lack thereof, accumulated over the last half century on the molecular mechanisms supporting activity-evoked K+ and extracellular space dynamics.


Assuntos
Espaço Extracelular , Espaço Extracelular/metabolismo , Neuroglia/metabolismo , Potássio/metabolismo , Sódio/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo
10.
Am J Physiol Cell Physiol ; 316(3): C434-C443, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30649920

RESUMO

ß-Cell swelling induces membrane depolarization, which has been suggested to be caused at least partly by the activation of cation channels. Here, we show the identification of the cation channels. In isolated mouse pancreatic ß-cells, the exposure to 30% hypotonic solution elicited an increase in cytosolic Ca2+ concentration ([Ca2+]c). The [Ca2+]c elevation was partially inhibited by ruthenium red, a blocker of several Ca2+-permeable channels including transient receptor potential vanilloid receptors [transient receptor potential cation channel subfamily V (TRPV)], and by nicardipine, but not by the depletion of intracellular Ca2+ stores with thapsigargin and caffeine. The hypotonic stimulation also increased insulin secretion from isolated mouse islets, which was significantly suppressed by ruthenium red. Expression of TRPV2 and TRPV4 was confirmed in mouse pancreatic islets and the MIN6 ß-cell line by RT-PCR, Western blot, and immunohistochemical analyses. However, neither 4α-phorbol 12,13-didecanoate nor GSK1016790A, TRPV4 activators, showed any apparent effect on [Ca2+]c in isolated mouse ß-cells or in MIN6 cells. In contrast, probenecid, a TRPV2 activator, induced an increase in [Ca2+]c in MIN6 cells, which was attenuated by ruthenium red. Moreover, the [Ca2+]c elevation induced by 30% hypotonic stimulation was significantly reduced by knockdown of TRPV2 with siRNA and by tranilast, a TRPV2 inhibitor. The knockdown of TRPV2 also decreased insulin secretion induced by the hypotonic stimulation. In addition, glucose-stimulated insulin secretion was also significantly reduced in the TRPV2-knockdown MIN6 cells. These results suggest that osmotic cell swelling activates TRPV2 in mouse ß-cells, thereby causing membrane depolarization and subsequent activation of voltage-dependent Ca2+ channels and insulin secretion.


Assuntos
Canais de Cálcio/metabolismo , Secreção de Insulina/fisiologia , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Canais de Cátion TRPV/metabolismo , Animais , Cálcio/metabolismo , Sinalização do Cálcio/fisiologia , Membrana Celular/metabolismo , Células Cultivadas , Citosol/metabolismo , Glucose/metabolismo , Masculino , Camundongos , RNA Interferente Pequeno/metabolismo
11.
J Neurophysiol ; 121(5): 1718-1734, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30786219

RESUMO

Two macroscopic parameters describe the interstitial diffusion of substances in the extracellular space (ECS) of the brain, the ECS volume fraction α and the diffusion tortuosity λ. Past methods based on sampling the extracellular concentration of a membrane-impermeable ion tracer, such as tetramethylammonium (TMA+), can characterize either the dynamic α(t) alone or the constant α and λ in resting state but never the dynamic α(t) and λ(t) simultaneously in short-lived brain events. In this work, we propose to use a sinusoidal method of TMA+ to provide time-resolved quantification of α(t) and λ(t) in acute brain events. This method iontophoretically injects TMA+ in the brain ECS by a sinusoidal time pattern, samples the resulting TMA+ diffusion waveform at a distance, and analyzes the transient modulations of the amplitude and phase lag of the sampled TMA+ waveform to infer α(t) and λ(t). Applicability of the sinusoidal method was verified through computer simulations of the sinusoidal TMA+ diffusion waveform in cortical spreading depression. Parameter sensitivity analysis identified the sinusoidal frequency and the interelectrode distance as two key operating parameters. Compared with other TMA+-based methods, the sinusoidal method can more accurately capture the dynamic α(t) and λ(t) in acute brain events and is equally applicable to other pathological episodes such as epilepsy, transient ischemic attack, and brain injury. Future improvement of the method should focus on high-fidelity extraction of the waveform amplitude and phase angle. NEW & NOTEWORTHY An iontophoretic sinusoidal method of tetramethylammonium is described to capture the dynamic brain extracellular space volume fraction α and diffusion tortuosity λ. The sinusoidal frequency and interelectrode distance are two key operating parameters affecting the method's accuracy in capturing α(t) and λ(t). High-fidelity extraction of the waveform amplitude and phase lag is critical to successful sinusoidal analyses.


Assuntos
Encéfalo/citologia , Potenciais Evocados , Espaço Extracelular/fisiologia , Modelos Neurológicos , Técnicas de Rastreamento Neuroanatômico/métodos , Algoritmos , Animais , Encéfalo/fisiologia , Difusão , Espaço Extracelular/metabolismo , Marcadores do Trato Nervoso/farmacocinética , Compostos de Amônio Quaternário/farmacocinética , Ratos , Tempo de Reação
12.
Amino Acids ; 51(9): 1337-1351, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31428912

RESUMO

The application of high concentrations of taurine induces long-lasting potentiation of synaptic responses and axon excitability. This phenomenon seems to require the contribution of a transport system with a low affinity for taurine. The prototypic taurine transporter TauT (SLC6A6) was discarded by experimental evidence obtained in TauT-KO mice. The purpose of the present study was to determine whether the proton-coupled amino acid transporter 1 (PAT1; SLC36A1) which is a transport system with low affinity and high capacity for a great variety of amino acids including taurine, contributes to the taurine-induced synaptic potentiation. In rat hippocampal slices, the application of several amino acids (L- and D-alanine, L-glutamine, ß-guanidinopropionic acid, glycine, L-histidine, L- and D-serine, sarcosine, L- and D-threonine) imitated the synaptic potentiation induced by taurine. The magnitude of the potentiation caused by some of these amino acids was even greater than that induced by taurine. By contrast, the application of other amino acids (L-arginine, betaine, L-leucine, L-methionine, L- and D-proline, and L-valine) did not induce potentiation. The behaviour of these different amino acids on synaptic potentiation is not compatible with a role of PAT1 in synaptic potentiation. There was a positive correlation between the accumulation of the different amino acids in the slice and the magnitude of synaptic potentiation induced by them. Some of the amino acids inducing synaptic potentiation, like taurine and L-threonine, also increased electrical resistance of the slice, whereas L-leucine did not modify this parameter. Modifications induced by either taurine or L-threonine in synaptic potentiation, slice resistance and amino acid accumulation were dependent on extracellular chloride concentration. These findings support the idea that the accumulation of amino acids throughout the action of transporters causes cell swelling enhancing the electrical resistance of the slice, which by itself could be sufficient to increase field synaptic potentials.


Assuntos
Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Aminoácidos/metabolismo , Hipocampo/fisiologia , Simportadores/metabolismo , Potenciais Sinápticos , Aminoácidos/química , Aminoácidos/farmacologia , Animais , Impedância Elétrica , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley , Taurina/metabolismo , Taurina/farmacologia , Treonina/metabolismo , Treonina/farmacologia
13.
Neurochem Res ; 43(2): 397-406, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29150743

RESUMO

Brain edema and the associated increase in intracranial pressure are major consequences of traumatic brain injury (TBI) that accounts for most early deaths after TBI. We recently showed that acute severe trauma to cultured astrocytes results in cell swelling. We further examined whether trauma induces cell swelling in neurons and microglia. We found that severe trauma also caused cell swelling in cultured neurons, whereas no swelling was observed in microglia. While severe trauma caused cell swelling in both astrocytes and neurons, mild trauma to astrocytes, neurons, and microglia failed to cell swelling. Since extracellular levels of glutamate are increased in brain post-TBI and microglia are known to release cytokine, and direct exposure of astrocytes to these molecules are known to stimulate cell swelling, we examined whether glutamate or cytokines have any additive effect on trauma-induced cell swelling. Exposure of cultured astrocytes to trauma caused cell swelling, and such swelling was potentiated by the exposure of traumatized astrocytes to glutamate and cytokines. Conditioned medium (CM) from traumatized astrocytes had no effect on neuronal swelling post-trauma, while CM from traumatized neurons and microglia potentiated the effect of trauma on astrocyte swelling. Further, trauma significantly increased the Na-K-Cl co-transporter (NKCC) activity in neurons, and that inhibition of NKCC activity diminished the trauma-induced neuronal swelling. Our results indicate that a differential sensitivity to trauma-induced cell swelling exists in neural cells and that neurons and microglia are likely to be involved in the potentiation of the astrocyte swelling post-trauma.


Assuntos
Astrócitos/metabolismo , Lesões Encefálicas/complicações , Microglia/metabolismo , Neurônios/metabolismo , Animais , Animais Recém-Nascidos , Edema Encefálico/etiologia , Edema Encefálico/metabolismo , Lesões Encefálicas/metabolismo , Células Cultivadas , Citocinas/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Ratos
14.
Plant Cell Rep ; 37(4): 665-676, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29354881

RESUMO

KEY MESSAGE: It is the first time that formononetin produced by cell culture and its accumulation was shown to be triggered by specific stress signalling linked jasmonate pathway. Callerya speciosa, an endangered traditional Chinese medicine plant, is intensively used in traditional folk medicine. To develop sustainable alternatives for the overexploitation of natural resources, a suspension cell line was created from C. speciosa. Ingredients of C. speciosa, for instance the isoflavone formononetin, are formed during a peculiar swelling response of the root, which is considered as a quality trait for commercial application. A cell strain with elongated cells was obtained by using synthetic cytokinin 6-benzylaminopurine (6-BA) and synthetic auxin picloram. Both, picloram and 6-BA, promote cell division, whereas picloram was shown to be crucial for the maintenance of axial cell expansion. We addressed the question, whether the loss of axiality observed in the maturating root is necessary and sufficient for the accumulation of formononetin. While we were able to mimic a loss of axiality for cell expansion, either by specific combinations of 6-BA and picloram, or by treatment with the anti-microtubular compound oryzalin, formononetin was not detectable. However, formononetin could be induced by the stress hormone methyl jasmonate (MeJA), as well as by the bacterial elicitor flagellin peptide (flg22), but not by a necrosis inducing protein. Combined the fact that none of these treatments induced the loss of axiality, we conclude that formononetin accumulates in response to basal defence and unrelated with cell swelling.


Assuntos
Forma Celular/fisiologia , Fabaceae/metabolismo , Isoflavonas/metabolismo , Raízes de Plantas/metabolismo , Acetatos/farmacologia , Linhagem Celular , Forma Celular/efeitos dos fármacos , Ciclopentanos/farmacologia , Fabaceae/citologia , Fabaceae/efeitos dos fármacos , Oxilipinas/farmacologia , Reguladores de Crescimento de Plantas/farmacologia , Raízes de Plantas/citologia , Raízes de Plantas/efeitos dos fármacos
15.
Curr Top Membr ; 81: 97-123, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30243442

RESUMO

The effects of osmotic swelling on key cellular biomechanical properties are explored in this chapter. We present the governing equations and theoretical backgrounds of the models employed to estimate cell membrane tension and elastic moduli from experimental methods, and provide a summary of the prevailing experimental approaches used to obtain these biomechanical parameters. A detailed analysis of the current evidence of the effects of osmotic swelling on membrane tension and elastic moduli is provided. Briefly, due to the buffering effect of unfolding membrane reservoirs, mild hypotonic swelling does not change membrane tension or the adhesion of the membrane to the underlying cytoskeleton. Conversely, osmotic swelling causes the cell membrane envelope to stiffen, measured as an increase in the membrane elastic modulus.


Assuntos
Membrana Celular/metabolismo , Módulo de Elasticidade/fisiologia , Animais , Tamanho Celular , Citoesqueleto/metabolismo , Humanos , Osmose/fisiologia
16.
Curr Top Membr ; 81: 3-52, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30243436

RESUMO

From early unicellular organisms that formed in salty water environments to complex organisms that live on land away from water, cells have had to protect a homeostatic internal environment favorable to the biochemical reactions necessary for life. In this chapter, we will outline what steps were necessary to conserve the water within our cells and how mechanisms have evolved to maintain and regulate our cellular and organismal volume. We will first examine whole body water homeostasis and the relationship between kidney function, regulation of blood pressure, and blood filtration in the process of producing urine. We will then discuss how the composition of the lipid-rich bilayer affects its permeability to water and salts, and how the cell uses this differential to drive physiological and biochemical cellular functions. The capacity to maintain cell volume is vital to epithelial transport, neurotransmission, cell cycle, apoptosis, and cell migration. Finally, we will wrap up the chapter by discussing in some detail specific channels, cotransporters, and exchangers that have evolved to facilitate the movement of cations and anions otherwise unable to cross the lipid-rich bilayer and that are involved in maintaining or regulating cell volume.


Assuntos
Tamanho Celular , Animais , Homeostase/fisiologia , Humanos , Transdução de Sinais/fisiologia
17.
Curr Top Membr ; 81: 385-455, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30243438

RESUMO

Regulation of cellular volume is a critical homeostatic process that is intimately linked to ionic and osmotic balance in the brain tissue. Because the brain is encased in the rigid skull and has a very complex cellular architecture, even minute changes in the volume of extracellular and intracellular compartments have a very strong impact on tissue excitability and function. The failure of cell volume control is a major feature of several neuropathologies, such as hyponatremia, stroke, epilepsy, hyperammonemia, and others. There is strong evidence that such dysregulation, especially uncontrolled cell swelling, plays a major role in adverse pathological outcomes. To protect themselves, brain cells utilize a variety of mechanisms to maintain their optimal volume, primarily by releasing or taking in ions and small organic molecules through diverse volume-sensitive ion channels and transporters. In principle, the mechanisms of cell volume regulation are not unique to the brain and share many commonalities with other tissues. However, because ions and some organic osmolytes (e.g., major amino acid neurotransmitters) have a strong impact on neuronal excitability, cell volume regulation in the brain is a surprisingly treacherous process, which may cause more harm than good. This topical review covers the established and emerging information in this rapidly developing area of physiology.


Assuntos
Encéfalo/citologia , Encéfalo/metabolismo , Tamanho Celular , Neuropatologia , Animais , Encéfalo/patologia , Humanos , Canais Iônicos/metabolismo
18.
Curr Top Membr ; 81: 177-203, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30243432

RESUMO

The Volume-Regulated Anion Channel (VRAC) is activated by cell swelling and plays a key role in cell volume regulation. VRAC is ubiquitously expressed in vertebrate cells and also implicated in many other physiological and cellular processes including fluid secretion, glutamate release, membrane potential regulation, cell proliferation, migration, and apoptosis. Although its biophysical properties have been well characterized, the molecular identity of VRAC remained a mystery for almost three decades. The field was transformed by recent discoveries showing that the leucine-rich repeat-containing protein 8A (LRRC8A, also named SWELL1) and its four other homologs form heteromeric VRAC channels. The composition of LRRC8 subunits determines channel properties and substrate selectivity of a large variety of different VRACs. Incorporating purified SWELL1-containing protein complexes into lipid bilayers is sufficient to reconstitute channel activities, a finding that supports the decrease in intracellular ionic strength as the mechanism of VRAC activation during cell swelling. Characterization of Swell1 knockout mice uncovers the important role of VRAC in T cell development, pancreatic ß-cell glucose-stimulated insulin secretion, and adipocyte metabolic function. The ability to permeate organic osmolytes and metabolites is a major feature of VRAC. The list of VRAC substrates is expected to grow, now also including some cancer drugs and antibiotics even under non-cell swelling conditions. Therefore, a critical role of VRAC in drug resistance and cell-cell communication is emerging. This review summarizes the exciting recent progress on the structure-function relationship and physiology of VRAC and discusses key future questions to be solved.


Assuntos
Ânions/metabolismo , Tamanho Celular , Canais Iônicos/metabolismo , Animais , Transporte Biológico , Humanos , Transdução de Sinais
19.
Curr Top Membr ; 81: 457-496, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30243439

RESUMO

When they become simultaneously leaky to both Na+ and Cl-, excitable cells are vulnerable to potentially lethal cytotoxic swelling. Swelling ensues in spite of an isosmotic milieu because the entering ions add osmolytes to the cytoplasm's high concentration of impermeant anionic osmolytes. An influx of osmotically-obliged water is unavoidable. A cell that cannot stanch at least one the leaks will succumb to death by Donnan effect. "Sick excitable cells" are those injured through ischemia, trauma, inflammation, hyperactivity, genetically-impaired membrane skeletons and other insults, all of which foster bleb-damage to regions of the plasma membrane. Nav channels resident in damaged membrane exhibit left-shifted kinetics; the corresponding Nav window conductance constitutes a Na+-leak. In cortical neurons, sustained depolarization to ∼-20mV elicits a sustained lethal gCl. Underlying Vrest in skeletal muscle is a constitutively active gCl; not surprisingly therefore, dystrophic muscle fibers, which are prone to bleb damage and which exhibit Nav-leak and Na+-overload, are prone to cytotoxic swelling. To restore viability in cytotoxically swelling neurons and muscle, the imperative of fully functional ion homeostasis is well-recognized. However, as emphasized here, in a healthy excitable cell, fully functional membrane tension homeostasis is also imperative. ATPase-pumps keep plasma membrane batteries charged, and ATPase-motor proteins maintain membrane tone. In sick excitable cells, neither condition prevails.


Assuntos
Tamanho Celular , Neurônios/citologia , Neurônios/metabolismo , Animais , Membrana Celular/metabolismo , Homeostase/fisiologia , Humanos , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo
20.
Curr Top Membr ; 81: 207-235, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30243433

RESUMO

Ion Transport across the cell membrane is required to maintain cell volume homeostasis. In response to changes in extracellular osmolarity, most cells activate specific metabolic or membrane-transport pathways to respond to cell swelling or shrinkage and return their volume to its normal resting state. This process involves the rapid adjustment of the activities of channels and transporters that mediate flux of K+, Na+, Cl-, and small organic osmolytes. Cation chloride cotransporters (CCCs) NKCCs and KCCs are a family of membrane proteins modulated by changes in cell volume and/or in the intracellular chloride concentration ([Cl-]i). Cell swelling triggers regulatory volume decrease (RVD), promoting solute and water efflux to restore normal cell volume. Swelling-activated KCCs mediate RVD in most cell types. In contrast, cell shrinkage triggers regulatory volume increase (RVI), which involves the activation of the NKCC1 cotransporter of the CCC family. Regulation of the CCCs during RVI and RVD by protein phosphorylation is a well-characterized mechanism, where WNK kinases and their downstream kinase substrates, SPAK and OSR1 constitute the essential phospho-regulators. WNKs-SPAK/OSR1-CCCs complex is required to regulate cell shrinkage-induced RVI or cell swelling-induced RVD via activating or inhibitory phosphorylation of NKCCs or KCCs, respectively. WNK1 and WNK4 kinases have been established as [Cl-]i sensors/regulators, while a role for WNK3 kinase as a cell volume-sensing kinase has emerged and is proposed in this chapter.


Assuntos
Tamanho Celular , Animais , Cloretos/metabolismo , Humanos , Transporte de Íons/fisiologia , Fosforilação , Sódio/metabolismo , Simportadores de Cloreto de Sódio-Potássio/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa