Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 119(23): e2120750119, 2022 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-35648818

RESUMEN

The human voltage-gated proton channel (hHv1) is important for control of intracellular pH. We designed C6, a specific peptide inhibitor of hHv1, to evaluate the roles of the channel in sperm capacitation and in the inflammatory immune response of neutrophils [R. Zhao et al., Proc. Natl. Acad. Sci. U.S.A. 115, E11847­E11856 (2018)]. One C6 binds with nanomolar affinity to each of the two S3­S4 voltage-sensor loops in hHv1 in cooperative fashion so that C6-bound channels require greater depolarization to open and do so more slowly. As depolarization drives hHv1 sensors outwardly, C6 affinity decreases, and inhibition is partial. Here, we identified residues essential to C6­hHv1 binding by scanning mutagenesis, five in the hHv1 S3­S4 loops and seven on C6. A structural model of the C6­hHv1 complex was then generated by molecular dynamics simulations and validated by mutant-cycle analysis. Guided by this model, we created a bivalent C6 peptide (C62) that binds simultaneously to both hHv1 subunits and fully inhibits current with picomolar affinity. The results help delineate the structural basis for C6 state-dependent inhibition, support an anionic lipid-mediated binding mechanism, and offer molecular insight into the effectiveness of engineered C6 as a therapeutic agent or lead.


Asunto(s)
Diseño de Fármacos , Canales Iónicos , Humanos , Canales Iónicos/antagonistas & inhibidores , Canales Iónicos/química , Canales Iónicos/genética , Masculino , Mutagénesis , Péptidos/química , Péptidos/farmacología , Unión Proteica , Protones , Capacitación Espermática
2.
J Biol Chem ; 299(7): 104918, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37315791

RESUMEN

Unlike other members of the voltage-gated ion channel superfamily, voltage-gated proton (Hv) channels are solely composed of voltage sensor domains without separate ion-conducting pores. Due to their unique dependence on both voltage and transmembrane pH gradients, Hv channels normally open to mediate proton efflux. Multiple cellular ligands were also found to regulate the function of Hv channels, including Zn2+, cholesterol, polyunsaturated arachidonic acid, and albumin. Our previous work showed that Zn2+ and cholesterol inhibit the human voltage-gated proton channel (hHv1) by stabilizing its S4 segment at resting state conformations. Released from phospholipids by phospholipase A2 in cells upon infection or injury, arachidonic acid regulates the function of many ion channels, including hHv1. In the present work, we examined the effects of arachidonic acid on purified hHv1 channels using liposome flux assays and revealed underlying structural mechanisms using single-molecule FRET. Our data indicated that arachidonic acid strongly activates hHv1 channels by promoting transitions of the S4 segment toward opening or "preopening" conformations. Moreover, we found that arachidonic acid even activates hHv1 channels inhibited by Zn2+ and cholesterol, providing a biophysical mechanism to activate hHv1 channels in nonexcitable cells upon infection or injury.


Asunto(s)
Ácido Araquidónico , Colesterol , Activación del Canal Iónico , Canales Iónicos , Protones , Zinc , Humanos , Albúminas/farmacología , Ácido Araquidónico/farmacología , Colesterol/farmacología , Transferencia Resonante de Energía de Fluorescencia , Activación del Canal Iónico/efectos de los fármacos , Canales Iónicos/agonistas , Canales Iónicos/antagonistas & inhibidores , Canales Iónicos/química , Canales Iónicos/metabolismo , Liposomas/metabolismo , Fosfolipasas A2/metabolismo , Imagen Individual de Molécula , Zinc/farmacología , Concentración de Iones de Hidrógeno
3.
J Physiol ; 601(18): 4073-4089, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37555355

RESUMEN

ATP is an important molecule implicated in diverse biochemical processes, including the modulation of ion channel and transporter activity. The voltage-gated proton channel (Hv1) controls proton flow through the transmembrane pathway in response to membrane potential, and various molecules regulate its activity. Although it is believed that ATP is not essential for Hv1 activity, a report has indicated that cytosolic ATP may modulate Hv1. However, the detailed molecular mechanism underlying the effect of ATP on Hv1 is unknown, and whether ATP is involved in the physiological regulation of Hv1 activity remains unclear. Here, we report that cytosolic ATP is required to maintain Hv1 activity. To gain insight into the underlying mechanism, we analysed the effects of ATP on the mouse Hv1 channel (mHv1) using electrophysiological and microscale thermophoresis (MST) methods. Intracellular ATP accelerated the activation kinetics of mHv1, thereby increasing the amplitude of the proton current within the physiological concentration range. The increase in proton current was reproduced with a non-hydrolysable ATP analogue, indicating that ATP directly influences Hv1 activity without an enzymatic reaction. The direct molecular interaction between the purified mHv1 protein and ATP was analysed and demonstrated through MST. In addition, ATP facilitation was observed for the endogenous proton current flowing through Hv1 in the physiological concentration range of ATP. These results suggest that ATP influences Hv1 activity via direct molecular interactions and is required for the physiological function of Hv1. KEY POINTS: We found that ATP is required to maintain the activity of voltage-gated proton channels (Hv1) and investigated the underlying molecular mechanism. Application of intracellular ATP increased the amplitude of the proton current flowing through Hv1, accompanied by an acceleration of activation kinetics. The direct interaction between purified Hv1 protein and ATP was quantitatively analysed using microscale thermophoresis. ATP enhanced endogenous proton currents in breast cancer cell lines. These results suggest that ATP influences Hv1 activity via direct molecular interactions and that its functional characteristics are required for the physiological activity of Hv1.


Asunto(s)
Activación del Canal Iónico , Protones , Animales , Ratones , Activación del Canal Iónico/fisiología , Canales Iónicos/metabolismo , Potenciales de la Membrana , Adenosina Trifosfato/farmacología
4.
Glia ; 71(10): 2418-2436, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37395288

RESUMEN

Spinal cord injury (SCI) causes severe functional deficits and neuronal damage, accompanied by intense glial activation. The voltage-gated proton channel Hv1, selectively expressed on microglia, is associated with SCI progression. However, the effect of Hv1 on the phenotypes and functions of reactive astrocytes after SCI remains unclear. Here, we combined Hv1 knockout (Hv1-/- ) mice and T10 spinal cord contusion to investigate the effects of microglial Hv1 on SCI pathophysiology and the phenotypes and functions of reactive astrocytes. After SCI, astrocytes proliferated and activated in the peri-injury area and exhibited an A1-dominant phenotype. Hv1 knockout reduced neurotoxic A1 astrocytes and shifted the dominant phenotype of reactive astrocytes from A1 to A2, enhancing synaptogenesis promotion, phagocytosis, and neurotrophy of astrocytes. Moreover, synaptic and axonal remodeling as well as motor recovery after SCI benefited from the improved astrocytic functions of Hv1 knockout. Furthermore, exogenous and endogenous reactive oxygen species (ROS) in astrocytes after SCI were reduced by Hv1 knockout. Our in vitro results showed that inhibition of ROS reduced the neurotoxic A1 phenotype in primary astrocytes via the STAT3 pathway. Similar to the effect of Hv1 knockout, the application of the ROS scavenger N-acetylcysteine reduced SCI-induced neurotoxic A1 astrocytes in vivo. Based on the in vivo and vitro results, we elucidated that microglial Hv1 knockout promotes synaptic and axonal remodeling in SCI mice by decreasing neurotoxic A1 astrocytes and increasing neuroprotective A2 astrocytes via the ROS/STAT3 pathway. Therefore, the Hv1 proton channel is a promising target for the treatment of SCI.


Asunto(s)
Microglía , Traumatismos de la Médula Espinal , Animales , Ratones , Astrocitos/metabolismo , Canales Iónicos/metabolismo , Ratones Noqueados , Microglía/metabolismo , Protones , Especies Reactivas de Oxígeno/metabolismo , Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/metabolismo
5.
J Neurochem ; 165(1): 29-54, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36625847

RESUMEN

Although the precise mechanisms determining the neurotoxic or neuroprotective activation phenotypes in microglia remain poorly characterized, metabolic changes in these cells appear critical for these processes. As cellular metabolism can be tightly regulated by changes in intracellular pH, we tested whether pharmacological targeting of the microglial voltage-gated proton channel 1 (Hv1), an important regulator of intracellular pH, is critical for activated microglial reprogramming. Using a mouse microglial cell line and mouse primary microglia cultures, either alone, or co-cultured with rat cerebrocortical neurons, we characterized in detail the microglial activation profile in the absence and presence of Hv1 inhibition. We observed that activated microglia neurotoxicity was mainly attributable to the release of tumor necrosis factor alpha, reactive oxygen species, and zinc. Strikingly, pharmacological inhibition of Hv1 largely abrogated inflammatory neurotoxicity not only by reducing the production of cytotoxic mediators but also by promoting neurotrophic molecule production and restraining excessive phagocytic activity. Importantly, the Hv1-sensitive change from a pro-inflammatory to a neuroprotective phenotype was associated with metabolic reprogramming, particularly via a boost in NADH availability and a reduction in lactate. Most critically, Hv1 antagonism not only reduced inflammatory neurotoxicity but also promoted microglia-dependent neuroprotection against a separate excitotoxic injury. Our results strongly suggest that Hv1 blockers may provide an important therapeutic tool against a wide range of inflammatory neurodegenerative disorders.


Asunto(s)
Ácido Glutámico , Microglía , Animales , Ratas , Microglía/metabolismo , Ácido Glutámico/toxicidad , Ácido Glutámico/metabolismo , Canales Iónicos/metabolismo , Neuronas/metabolismo , Especies Reactivas de Oxígeno/metabolismo
6.
J Biol Chem ; 297(4): 101212, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34547291

RESUMEN

Sex as a physiologic factor has a strong association with the features of metabolic syndrome. Our previous study showed that loss of the voltage-gated proton channel Hv1 inhibits insulin secretion and leads to hyperglycemia and glucose intolerance in male mice. However, there are significant differences in blood glucose between male and female Hv1-knockout (KO) mice. Here, we investigated the differences in glucose metabolism and insulin sensitivity between male and female KO mice and how sex steroids contribute to these differences. We found that the fasting blood glucose in female KO mice was visibly lower than that in male KO mice, which was accompanied by hypotestosteronemia. KO mice in both sexes exhibited higher expression of gluconeogenesis-related genes in liver compared with WT mice. Also, the livers from KO males displayed a decrease in glycolysis-related gene expression and an increase in gluconeogenesis-related gene expression compared with KO females. Furthermore, exogenous testosterone supplementation decreased blood glucose levels in male KO mice, as well as enhancing insulin signaling. Taken together, our data demonstrate that knockout of Hv1 results in higher blood glucose levels in male than female mice, despite a decreased insulin secretion in both sexes. This sex-related difference in glucose homeostasis is associated with the glucose metabolism in liver tissue, likely due to the physiological levels of testosterone in KO male mice.


Asunto(s)
Glucemia , Gluconeogénesis , Glucólisis , Canales Iónicos/deficiencia , Hígado/metabolismo , Caracteres Sexuales , Animales , Glucemia/genética , Glucemia/metabolismo , Femenino , Regulación de la Expresión Génica , Canales Iónicos/metabolismo , Masculino , Ratones , Ratones Noqueados , Transducción de Señal
7.
Andrologia ; 54(8): e14471, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35590125

RESUMEN

Prunus japonica var. nakaii is used in traditional Korean medicine to treat various conditions; however, it has not been investigated for treating male infertility. In this study, we investigated the in vitro effects of the ethanolic extract of P. japonica seeds on human sperm motility and identified its mechanism of action. Eleven male volunteers were selected, and the effects of the extract on human spermatozoa were assessed through a computer-assisted semen analysis. The P. japonica seed extract increased the percentage of total and progressive motility of spermatozoa. To understand the mechanism of action, we monitored intracellular alkalization using flow cytometry and obtained electrophysiological recordings of human voltage-gated proton channels hHv1 that were overexpressed in HEK-293 cells. The extract shifted the activation curves in a concentration-dependent manner. Two major constituents of the extract, linoleic acid and oleic acid, exhibited proton channel activity. Our in vitro experiments suggested that P. japonica seed extract could be potentially used to rescue sperm motility in idiopathic infertility patients via pharmacological modulation of the proton channels during capacitation. Therefore, our results indicate the therapeutic potential of P. japonica seed extract for treating male infertility.


Asunto(s)
Infertilidad Masculina , Prunus , Células HEK293 , Humanos , Masculino , Extractos Vegetales/farmacología , Protones , Capacitación Espermática , Motilidad Espermática , Espermatozoides
8.
J Biol Chem ; 295(11): 3601-3613, 2020 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-31949049

RESUMEN

Insulin secretion by pancreatic islet ß-cells is regulated by glucose levels and is accompanied by proton generation. The voltage-gated proton channel Hv1 is present in pancreatic ß-cells and extremely selective for protons. However, whether Hv1 is involved in insulin secretion is unclear. Here we demonstrate that Hv1 promotes insulin secretion of pancreatic ß-cells and glucose homeostasis. Hv1-deficient mice displayed hyperglycemia and glucose intolerance because of reduced insulin secretion but retained normal peripheral insulin sensitivity. Moreover, Hv1 loss contributed much more to severe glucose intolerance as the mice got older. Islets of Hv1-deficient and heterozygous mice were markedly deficient in glucose- and K+-induced insulin secretion. In perifusion assays, Hv1 deletion dramatically reduced the first and second phase of glucose-stimulated insulin secretion. Islet insulin and proinsulin content was reduced, and histological analysis of pancreas slices revealed an accompanying modest reduction of ß-cell mass in Hv1 knockout mice. EM observations also indicated a reduction in insulin granule size, but not granule number or granule docking, in Hv1-deficient mice. Mechanistically, Hv1 loss limited the capacity for glucose-induced membrane depolarization, accompanied by a reduced ability of glucose to raise Ca2+ levels in islets, as evidenced by decreased durations of individual calcium oscillations. Moreover, Hv1 expression was significantly reduced in pancreatic ß-cells from streptozotocin-induced diabetic mice, indicating that Hv1 deficiency is associated with ß-cell dysfunction and diabetes. We conclude that Hv1 regulates insulin secretion and glucose homeostasis through a mechanism that depends on intracellular Ca2+ levels and membrane depolarization.


Asunto(s)
Intolerancia a la Glucosa/complicaciones , Intolerancia a la Glucosa/metabolismo , Hiperglucemia/complicaciones , Hiperglucemia/metabolismo , Secreción de Insulina , Canales Iónicos/metabolismo , Envejecimiento/patología , Animales , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Tamaño de la Célula , Gránulos Citoplasmáticos/metabolismo , Gránulos Citoplasmáticos/ultraestructura , Citosol/metabolismo , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patología , Regulación hacia Abajo/efectos de los fármacos , Eliminación de Gen , Glucosa/farmacología , Concentración de Iones de Hidrógeno , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Células Secretoras de Insulina/ultraestructura , Canales Iónicos/deficiencia , Canales Iónicos/genética , Potenciales de la Membrana , Ratones Endogámicos C57BL , Ratones Noqueados , Acetato de Tetradecanoilforbol/farmacología
9.
J Neurochem ; 157(3): 624-641, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33404063

RESUMEN

The properties of microglia largely differ depending on aging as well as on brain regions. However, there are few studies that investigated the functional importance of such heterogeneous properties of microglia at the molecular level. Voltage-gated proton channel, Hv1/VSOP, could be one of the candidates which confers functional heterogeneity among microglia since it regulates brain oxidative stress in age-dependent manner. In this study, we found that Hv1/VSOP shows brain region-dependent heterogeneity of gene expression with the highest level in the striatum. We studied the importance of Hv1/VSOP in two different brain regions, the cerebral cortex and striatum, and examined their relationship with aging (using mice of different ages). In the cortex, we observed the age-dependent impact of Hv1/VSOP on oxidative stress, microglial morphology, and gene expression profile. On the other hand, we found that the age-dependent significance of Hv1/VSOP was less obvious in the striatum than the cortex. Finally, we performed a battery of behavioral experiments on Hv1/VSOP-deficient mice both at young and aged stages to examine the effect of aging on Hv1/VSOP function. Hv1/VSOP-deficient mice specifically showed a marked difference in behavior in light/dark transition test only at aged stages, indicating that anxiety state is altered in aged Hv1/VSOP mice. This study suggests that a combination of brain region heterogeneity and animal aging underscores the functional importance of Hv1/VSOP in microglia.


Asunto(s)
Envejecimiento/metabolismo , Envejecimiento/fisiología , Química Encefálica/fisiología , Canales Iónicos/metabolismo , Envejecimiento/psicología , Animales , Ansiedad/psicología , Conducta Animal , Corteza Cerebral/metabolismo , Biología Computacional , Regulación de la Expresión Génica , Canales Iónicos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/metabolismo , Neostriado/metabolismo , Carbonilación Proteica , Transcriptoma
10.
Brain Behav Immun ; 91: 267-283, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33039662

RESUMEN

Tissue acidosis is an important secondary injury process in the pathophysiology of traumatic spinal cord injury (SCI). To date, no studies have examined the role of proton extrusion as mechanism of pathological acidosis in SCI. In the present study, we hypothesized that the phagocyte-specific proton channel Hv1 mediates hydrogen proton extrusion after SCI, contributing to increased extracellular acidosis and poor long-term outcomes. Using a contusion model of SCI in adult female mice, we demonstrated that tissue pH levels are markedly lower during the first week after SCI. Acidosis was most evident at the injury site, but also extended into proximal regions of the cervical and lumbar cord. Tissue reactive oxygen species (ROS) levels and expression of Hv1 were significantly increased during the week of injury. Hv1 was exclusively expressed in microglia within the CNS, suggesting that microglia contribute to ROS production and proton extrusion during respiratory burst. Depletion of Hv1 significantly attenuated tissue acidosis, NADPH oxidase 2 (NOX2) expression, and ROS production at 3 d post-injury. Nanostring analysis revealed decreased gene expression of neuroinflammatory and cytokine signaling markers in Hv1 knockout (KO) mice. Furthermore, Hv1 deficiency reduced microglia proliferation, leukocyte infiltration, and phagocytic oxidative burst detected by flow cytometry. Importantly, Hv1 KO mice exhibited significantly improved locomotor function and reduced histopathology. Overall, these data suggest an important role for Hv1 in regulating tissue acidosis, NOX2-mediated ROS production, and functional outcome following SCI. Thus, the Hv1 proton channel represents a potential target that may lead to novel therapeutic strategies for SCI.


Asunto(s)
Acidosis , Contusiones , Traumatismos de la Médula Espinal , Animales , Femenino , Canales Iónicos/genética , Ratones , Protones
11.
Int J Mol Sci ; 22(5)2021 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-33807711

RESUMEN

The voltage-gated proton channel, Hv1, also termed VSOP, was discovered in 2006. It has long been suggested that proton transport through voltage-gated proton channels regulate reactive oxygen species (ROS) production in phagocytes by counteracting the charge imbalance caused by the activation of NADPH oxidase. Discovery of Hv1/VSOP not only confirmed this process in phagocytes, but also led to the elucidation of novel functions in phagocytes. The compensation of charge by Hv1/VSOP sustains ROS production and is also crucial for promoting Ca2+ influx at the plasma membrane. In addition, proton extrusion into neutrophil phagosomes by Hv1/VSOP is necessary to maintain neutral phagosomal pH for the effective killing of bacteria. Contrary to the function of Hv1/VSOP as a positive regulator for ROS generation, it has been revealed that Hv1/VSOP also acts to inhibit ROS production in neutrophils. Hv1/VSOP inhibits hypochlorous acid production by regulating degranulation, leading to reduced inflammation upon fungal infection, and suppresses the activation of extracellular signal-regulated kinase (ERK) signaling by inhibiting ROS production. Thus, Hv1/VSOP is a two-way player regulating ROS production. Here, we review the functions of Hv1/VSOP in neutrophils and discuss future perspectives.


Asunto(s)
Señalización del Calcio , Degranulación de la Célula , Canales Iónicos/metabolismo , Sistema de Señalización de MAP Quinasas , Neutrófilos/metabolismo , Animales , Bacterias/metabolismo , Humanos , Ratones , NADPH Oxidasas/metabolismo , Neutrófilos/microbiología , Especies Reactivas de Oxígeno/metabolismo
12.
Medicina (Kaunas) ; 57(8)2021 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-34441020

RESUMEN

Background and Objectives: Alpha-1 antitrypsin is a serine protease inhibitor that demonstrates an array of immunomodulatory functions. Individuals with the genetic condition of alpha-1 antitrypsin deficiency (AATD) are at increased risk of early onset emphysematous lung disease. This lung disease is partly driven by neutrophil mediated lung destruction in an environment of low AAT. As peripheral neutrophil hyper-responsiveness in AATD leads to excessive degranulation and increased migration to the airways, we examined the expression of the membrane voltage-gated proton channel-1 (HVCN1), which is integrally linked to neutrophil function. The objectives of this study were to evaluate altered HVCN1 in AATD neutrophils, serine protease-dependent degradation of HVCN1, and to investigate the ability of serum AAT to control HVCN1 expression. Materials and Methods: Circulating neutrophils were purified from AATD patients (n = 20), AATD patients receiving AAT augmentation therapy (n = 3) and healthy controls (n = 20). HVCN1 neutrophil expression was assessed by flow cytometry and Western blot analysis. Neutrophil membrane bound elastase was measured by fluorescence resonance energy transfer. Results: In this study we demonstrated that HVCN1 protein is under-expressed in AATD neutrophils (p = 0.02), suggesting a link between reduced HVCN1 expression and AAT deficiency. We have demonstrated that HVCN1 undergoes significant proteolytic degradation in activated neutrophils (p < 0.0001), primarily due to neutrophil elastase activity (p = 0.0004). In addition, the treatment of AATD individuals with AAT augmentation therapy increased neutrophil plasma membrane HVCN1 expression (p = 0.01). Conclusions: Our results demonstrate reduced levels of HVCN1 in peripheral blood neutrophils that may influence the neutrophil-dominated immune response in the AATD airways and highlights the role of antiprotease treatment and specifically AAT augmentation therapy in protecting neutrophil membrane expression of HVCN1.


Asunto(s)
Neutrófilos , Deficiencia de alfa 1-Antitripsina , Humanos , Pulmón , Proteolisis , Protones , Deficiencia de alfa 1-Antitripsina/tratamiento farmacológico , Deficiencia de alfa 1-Antitripsina/genética
13.
Artículo en Inglés | MEDLINE | ID: mdl-32087974

RESUMEN

The pathological process of spinal cord injury (SCI) is complex, particularly during secondary damage that triggers a multiphasic glial reaction consisting of both detrimental and beneficial effects. Deletion of a novel voltage-gated proton channel (Hv1) functionally expressed in microglia has been shown to confer neuroprotection during ischemic stroke. Here, we hypothesized that microglial Hv1 may also participate in the process of SCI through modulating glial responses. To test this hypothesis, we employed an SCI model in Hv1-knockout (Hv1-/-) and wild type (WT) mice and assessed resulting microglial polarization, accumulation of pro-inflammatory cytokines, astrocytic activation, oligodendrocytic apoptosis, lesion sizes, and demyelinated areas. Compared with post-SCI results in WT mice, post-SCI Hv1-/- mice exhibited an M2-dominant microglial polarization, decreased accumulation of microglia, and reduced production of pro-inflammatory factors such as tumor necrosis factor alpha (TNF-α) and interleukin-1 beta (IL-1ß). Additionally, Hv1-/- mice had significantly attenuated reactive astrogliosis and reduced expression of chondroitin sulphate proteoglycans (CSPGs) after SCI. Furthermore, Hv1 deficiency reduced SCI-induced oligodendrocytic apoptosis, demyelinated areas, and cavity formation. Collectively, our results provide the first evidence suggesting that microglial Hv1 may be a multi-mechanism therapeutic target for the treatment of SCI.

14.
J Neuroinflammation ; 17(1): 263, 2020 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-32891159

RESUMEN

BACKGROUND: Spinal cord injury (SCI) causes neurological dysfunction with devastating consequences. SCI pathogenesis is accompanied by inflammasome activation and neuronal damage. But the spatial pattern and the time course of neuronal pyroptosis and apoptosis after SCI should be further elucidated. The microglial voltage-gated proton channel (Hv1) is implicated in reactive oxygen species (ROS)-induced neuronal damage following ischemic stroke. However, there is a lack of quantification on the neuronal pyroptosis and apoptosis associated with microglial Hv1 after SCI. METHODS: We analyzed spatial and temporal characteristics of neuronal pyroptosis and apoptosis following SCI and investigated the effects of Hv1 deficiency on neuronal pyroptosis and the nod-like receptor 3 (NLRP3) inflammasome pathway by using a mouse model of SCI. We tested the effects of Hv1-deficient microglia on ROS production in vivo and examined the relationship between ROS and neuronal pyroptosis in vitro. RESULTS: We observed that apoptosis was detected closer to the injury core than pyroptosis. The incidence of neuronal apoptosis peaked on day 1 after SCI and occurred before pyroptosis. Hv1 deficiency reduced neuronal apoptosis and NLRP3-inflammasome-mediated pyroptosis, improved axonal regeneration, and reduced motor deficits. SCI led to elevated ROS levels, whereas Hv1 deficiency downregulated microglial ROS generation. In vitro, ROS upregulated neuronal pyroptosis and activated the NLRP3 inflammasome pathway, both of which were reversed by addition of a ROS scavenger. Our results suggested that microglial Hv1 regulated neuronal apoptosis and NLRP3-induced neuronal pyroptosis after SCI by mediating ROS production. CONCLUSION: Following SCI, neuronal pyroptosis lasted longer and occurred farther away from the injury core compared with that of neuronal apoptosis. Microglial Hv1 deficiency downregulated microglial ROS generation and reduced apoptosis and NLRP3-induced neuronal pyroptosis. Our findings may provide novel insights into Hv1-associated mechanisms underlying neuronal damage after SCI.


Asunto(s)
Inflamación/metabolismo , Canales Iónicos/metabolismo , Microglía/metabolismo , Neuronas/metabolismo , Piroptosis/fisiología , Traumatismos de la Médula Espinal/metabolismo , Animales , Apoptosis/fisiología , Caspasa 1/metabolismo , Femenino , Inflamación/genética , Inflamación/patología , Canales Iónicos/genética , Ratones , Ratones Noqueados , Microglía/patología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Neuronas/patología , Células PC12 , Ratas , Especies Reactivas de Oxígeno/metabolismo , Traumatismos de la Médula Espinal/genética , Traumatismos de la Médula Espinal/patología
15.
Biochem Biophys Res Commun ; 513(2): 434-438, 2019 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-30967259

RESUMEN

High glucose (HG)-induced oxidative stress contributes to the dysfunction of pancreatic ß cells in diabetes. The voltage-gated proton channel Hv1 has been proposed to support reactive oxygen species (ROS) production during respiratory bursts. However, the effect of Hv1 on glucotoxicity in pancreatic ß cells is not clear yet. In this study, we examined the protective effects of Hv1-deficiency in HG cultured ß cells. Following 48 h of treatment with 30 mM high glucose, Hv1 KO ß cells showed higher cell viability, lower cell apoptosis and a more stable insulin gene expression level compared to WT ß cells. In both control and HG cultured ß cells, deficiency of Hv1 decreased the glucose- and PMA-induced ROS production. Finally, HG incubation led to NOX4 upregulation in WT ß cells, which could be inhibited by HV1 deficiency. In conclusion, Hv1-deficiency prevents the HG treatment-induced NOX4 upregulation and protects ß cells from glucotoxicity.


Asunto(s)
Hiperglucemia/metabolismo , Células Secretoras de Insulina/metabolismo , Canales Iónicos/metabolismo , NADPH Oxidasa 4/metabolismo , Estrés Oxidativo , Animales , Apoptosis , Células Cultivadas , Técnicas de Inactivación de Genes , Glucosa/metabolismo , Hiperglucemia/genética , Hiperglucemia/patología , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/patología , Canales Iónicos/genética , Ratones Endogámicos C57BL , NADPH Oxidasa 4/genética , Especies Reactivas de Oxígeno/metabolismo , Regulación hacia Arriba
16.
Proc Natl Acad Sci U S A ; 113(40): E5962-E5971, 2016 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-27647906

RESUMEN

Voltage-gated proton (Hv1) channels are involved in many physiological processes, such as pH homeostasis and the innate immune response. Zn2+ is an important physiological inhibitor of Hv1. Sperm cells are quiescent in the male reproductive system due to Zn2+ inhibition of Hv1 channels, but become active once introduced into the low-Zn2+-concentration environment of the female reproductive tract. How Zn2+ inhibits Hv1 is not completely understood. In this study, we use the voltage clamp fluorometry technique to identify the molecular mechanism of Zn2+ inhibition of Hv1. We find that Zn2+ binds to both the activated closed and resting closed states of the Hv1 channel, thereby inhibiting both voltage sensor motion and gate opening. Mutations of some Hv1 residues affect only Zn2+ inhibition of the voltage sensor motion, whereas mutations of other residues also affect Zn2+ inhibition of gate opening. These effects are similar in monomeric and dimeric Hv1 channels, suggesting that the Zn2+-binding sites are localized within each subunit of the dimeric Hv1. We propose that Zn2+ binding has two major effects on Hv1: (i) at low concentrations, Zn2+ binds to one site and prevents the opening conformational change of the pore of Hv1, thereby inhibiting proton conduction; and (ii) at high concentrations, Zn2+, in addition, binds to a second site and inhibits the outward movement of the voltage sensor of Hv1. Elucidating the molecular mechanism of how Zn2+ inhibits Hv1 will further our understanding of Hv1 function and might provide valuable information for future drug development for Hv1 channels.


Asunto(s)
Activación del Canal Iónico/genética , Canales Iónicos/genética , Zinc/metabolismo , Animales , Sitios de Unión , Femenino , Fluorometría/métodos , Humanos , Concentración de Iones de Hidrógeno , Inmunidad Innata/genética , Canales Iónicos/metabolismo , Mutación , Técnicas de Placa-Clamp/métodos , Protones , Xenopus laevis/metabolismo , Zinc/química
17.
Biochem Biophys Res Commun ; 498(4): 975-980, 2018 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-29550486

RESUMEN

Reactive oxygen species (ROS) impairs pancreatic ß-cells and plays an important role in development of diabetes. Streptozotocin (STZ) can lead to ß-cell dysfunction via inducing ROS production. The voltage-gated proton channel Hv1 contributes a majority of the charge compensation required for ROS production. Here, we investigated the effects of Hv1 on STZ-induced ß-cell damage. We found that deficiency of Hv1 obviously inhibits STZ-induced glucose intolerance in mice, and prevents the decrease in ß-cell mass and pancreatic insulin content from STZ-treatment. Further studies showed that loss of Hv1 significantly attenuates STZ-induced ß-cell damage and ROS production in pancreatic ß-cells. Our results suggest that Hv1 might contribute to development of diabetes through producing ROS.


Asunto(s)
Diabetes Mellitus/etiología , Células Secretoras de Insulina/patología , Canales Iónicos/deficiencia , Especies Reactivas de Oxígeno/metabolismo , Animales , Intolerancia a la Glucosa/inducido químicamente , Intolerancia a la Glucosa/etiología , Insulina/metabolismo , Canales Iónicos/fisiología , Ratones , Estreptozocina
18.
Biochem Biophys Res Commun ; 498(1): 1-8, 2018 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-28676401

RESUMEN

The contribution of microglial activation to oligodendrocyte precursor cell (OPC) damage in the brain is considered to be a principal pathophysiological feature of periventricular leukomalacia (PVL). Nicotinamide adenine dinucleotide phosphate oxidase (NOX)-dependent reactive oxygen species (ROS) produced in microglia has been shown to be significantly toxic to OPCs. The voltage-gated proton channel Hv1 is selectively expressed in microglia and is essential for NOX-dependent ROS production in the central nervous system. This study aimed to investigate the effects of microglial Hv1 deficiency on the protection of OPCs from oxygen-glucose deprivation (OGD)-induced injury in vitro. In the present study, the levels of OGD-induced ROS and pro-inflammatory cytokine production were dramatically lower in Hv1-deficient microglia (Hv1-/-) than in wild-type (WT) microglia. Following OGD, OPCs co-cultured with WT microglia had increased apoptosis and decreased proliferation and maturation, while those co-cultured with Hv1-/- microglia had attenuated apoptosis and greater proliferation and differentiation. Furthermore, the attenuated damage and enhanced regeneration of OPCs were associated with decreases in extracellular signal-regulated kinase 1/2 and p38 mitogen-activated protein kinase phosphorylation. These results indicate that the protective effects of Hv1 deficiency on OPCs are due to the suppression of ROS and pro-inflammatory cytokine production in microglia. We thus suggest that the microglial proton channel Hv1 may be a potential therapeutic target in PVL.


Asunto(s)
Citocinas/metabolismo , Glucosa/deficiencia , Mediadores de Inflamación/metabolismo , Canales Iónicos/metabolismo , Microglía/metabolismo , Células Precursoras de Oligodendrocitos/patología , Oxígeno/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Apoptosis , Diferenciación Celular , Proliferación Celular , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Canales Iónicos/deficiencia , Sistema de Señalización de MAP Quinasas , Ratones Endogámicos C57BL , Microglía/patología , Células Precursoras de Oligodendrocitos/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
19.
Eur Biophys J ; 47(3): 237-247, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28889176

RESUMEN

The voltage-gated proton channel Hv1 is highly selective for H+ and is activated by membrane depolarization and pH gradient. An increased external and decreased internal pH opens the Hv1 channel. The intracellular C-terminal domain of Hv1 is responsible for channel dimerization, cooperative, and thermosensitive gating. Here, we found that proton pump inhibitors (PPIs) interact with the C-terminal domain of human Hv1. The interaction between PPIs and the C-terminal domain, which is pH-dependent, lowered the thermal and structural stability of the protein at pH 4, but enhanced the thermal and structural stability at pH 8. Furthermore, we investigated in vitro the interaction of PPIs with the C-terminal domain of Hv1 by fluorescence and micro-Raman spectra. Fluorescence quenching measurements revealed that the interaction between the C-terminal domain and PPIs is a mainly hydrophobic interaction. The micro-Raman spectra showed that PPIs did not form stable disulfide bonds with the unique thiol group within this domain (Cys249 residue). The preferential interaction of PPIs with the inactive form of Hv1 stabilizes the high pH inactive state of the C-terminal domain, indicating a mechanism by which PPIs might act explicitly on the stabilization of a closed state of the proton channel.


Asunto(s)
Canales Iónicos/química , Canales Iónicos/metabolismo , Inhibidores de la Bomba de Protones/farmacología , Humanos , Concentración de Iones de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Dominios Proteicos , Multimerización de Proteína , Estabilidad Proteica , Estructura Cuaternaria de Proteína , Temperatura
20.
J Eukaryot Microbiol ; 65(6): 928-933, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29698585

RESUMEN

Bioluminescence in dinoflagellates is controlled by HV 1 proton channels. Database searches of dinoflagellate transcriptomes and genomes yielded hits with sequence features diagnostic of all confirmed HV 1, and show that HV 1 is widely distributed in the dinoflagellate phylogeny including the basal species Oxyrrhis marina. Multiple sequence alignments followed by phylogenetic analysis revealed three major subfamilies of HV 1 that do not correlate with presence of theca, autotrophy, geographic location, or bioluminescence. These data suggest that most dinoflagellates express a HV 1 which has a function separate from bioluminescence. Sequence evidence also suggests that dinoflagellates can contain more than one HV 1 gene.


Asunto(s)
Dinoflagelados/genética , Canales Iónicos/clasificación , Canales Iónicos/genética , Proteínas Luminiscentes/metabolismo , Filogenia , Protones , Análisis por Conglomerados , Dinoflagelados/metabolismo , Genes Protozoarios/genética , Genoma , Canales Iónicos/metabolismo , Alineación de Secuencia , Transcriptoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA