Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Thromb Thrombolysis ; 53(2): 291-301, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34757546

RESUMEN

Stroke is the third leading cause of death in the United States and the leading cause of adult disability. Despite enormous research efforts including many clinical trials, tissue plasminogen activator (tPA) remains the only FDA-approved treatment for acute ischemic stroke. Unfortunately, only 1-3% of stroke patients in the US receive this therapy because of the narrow time window and severe side effects for using tPA. The most deadly and damaging side effect is the risk of intracranial bleeding or hemorrhage. For that reason, the dose of tPA and its overall administration are under tight control, which may compromise the effect of thrombolysis. Studies have been focused on improving the effectiveness of tPA for higher rate of reperfusion, and the safety for less adverse bleeding episode. We studied how metal ions (zinc & iron) affect tPA-induced thrombolysis in vitro and in vivo, and proposed a method to improve the rate of thrombolysis. The amount of hemoglobin in the blood clot lysis was measured by a spectrophotometer. The tPA-induced thrombolysis was measured in vivo in femoral artery. Our results showed that Zn2+, Fe3+ and Fe2+ inhibited tPA-induced thrombolysis, with Zn2+ and Fe2+ being the most effective. Metal ion chelating agent EDTA when it was co-applied with tPA significantly enhanced the tPA-induced thrombolysis. The chelation alone did not have noticeable thrombolytic effect. In in vivo study of tPA-induced thrombosis following femoral artery thrombosis, the co-application of tPA and EDTA achieved significant higher rate of reperfusion than that by tPA treatment alone, suggesting that ion chelation facilitates tPA-induced thrombolysis and potentially improves the safety of tPA application by reducing the necessary dose of tPA application. Our results suggest that the co-application of a chelator and tPA improves the efficacy and, potentially, safety of tPA application, by reducing the necessary dose of tPA for thrombolysis.


Asunto(s)
Accidente Cerebrovascular Isquémico , Accidente Cerebrovascular , Adulto , Quelantes/uso terapéutico , Fibrinolíticos/uso terapéutico , Humanos , Iones/uso terapéutico , Accidente Cerebrovascular/inducido químicamente , Accidente Cerebrovascular/tratamiento farmacológico , Terapia Trombolítica/métodos , Activador de Tejido Plasminógeno
2.
Artículo en Inglés | MEDLINE | ID: mdl-34336133

RESUMEN

Zinc (Zn2+) is stored in the nucleus, endoplasmic reticulum (ER), Golgi apparatus, mitochondria, lysosomes, and zinc-binding proteins. The acidity of the microenvironment affects the binding between zinc and proteins in which zinc become free or loosely bound. In this study, when cells were treated with an acidic medium, we started seeing free zinc 'hot spots' or zincosomes where we found bright zinc fluorescence. The rising free zinc quickly across whole cells with both intensity and distribution were pH-dependent. Interestingly, the nucleus was more sensitive to acidic treatment as the increase of nuclear zinc was faster and higher than the increase of cytosolic zinc. In addition, we re-cultured strong acid-challenged cells in a normal medium. Comparing to the control, these cells exhibited multiple zinc 'hot spots' beside the nucleus, suggesting that free zinc became more extensively distributed. To investigate further the function of zinc in cell shaping and morphological changes, we categorized strong acid-challenged cells into different shapes and found that the proportion of each cell shape had changed after the acid challenge. These acid-induced changes of the cell shape percentage were partially reversed by the reduction of zinc, suggesting that zinc participated in directing the cell shapes and morphologies during cell growth. Our findings reveal that acidic pH affects the dynamics of cellular zinc by making zinc more accessible to cellular compartments and zinc-binding proteins, which provided new insights into understanding the cellular behavior and the function of zinc in it.

3.
Artículo en Inglés | MEDLINE | ID: mdl-34093965

RESUMEN

Zinc (Zn2+) is important in cellular processes. In the cell, free zinc is tightly regulated and found in minuscule amounts. However, in an unhealthy cellular environment, such as hypoxia, zinc increases in the cell and zinc overload may occur. Studies have shown that zinc overload causes cellular and mitochondrial stress. Mitochondrial stress affects mitochondrial morphology. In normal cells, mitochondrial morphology resembles a long, tubular shape. In unhealthy cells, mitochondrial morphology resembles fragmented, circular shape. To address whether zinc overload contributes directly to the abnormal changes of mitochondrial morphology, we imaged and analyzed mitochondria that were treated with the application of exogenous zinc. In the first part of the study, exogenous zinc was applied to HeLa cells at 1 µM, 10 µM, 50 µM, 100 µM, or 200 µM zinc chloride along with 10 µM pyrithione. Mitochondrial morphology was analyzed with Mito-Morphology micro in ImageJ. Mitochondrial morphology changed from a healthy tubular shape to an unhealthy circular shape and fragmentation. Mitochondrial morphology changes were observed in a dose-dependent fashion. The second part of the study involved applying the metal ion chelator TPEN after applying 50 µM zinc chloride along with 10 µM pyrithione. TPEN reduced zinc-induced abnormal mitochondrial morphology after zinc treatment. This present study supports that zinc overload may cause morphology changes induced by mitochondrial stress that may lead to cell death.

4.
Mol Cell Biochem ; 476(7): 2813-2821, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33733429

RESUMEN

Environmental changes can stress and alter biology at the molecular and cellular level. For example, metal-protein interaction is a classic physic and biological property of nature, which is fundamentally influenced by acidity. Here, we report a unique cellular reprogramming phenomenon in that a brief strong acid treatment induced the expression of pluripotent stem cell (PSC) markers. We used strong acid to briefly challenge mix-cultured gastric cells, and then subcultured survived cells in a normal cell culture medium. We found that survival acid-treated cells expressed PSC markers detected by commonly used pluripotent antibodies such as SSEA-4 and Oct4. In addition, we observed that the survived cells from the acid challenge grew faster during the second and third weeks of subculture and had a relative short doubling time (DT) than the controls. PSC marker-labeled 'older' cells also presented immature cell-like morphology with some having marker Oct4 in the nucleus. Finally, the expression of the markers appeared to be sensitive to metal ion chelation. Removal of the metals during a brief acid treatment reduced pluripotent marker-positive cells, suggesting the dissociation of metals from metal-binding proteins may be a factor involved in the induction of stem cell markers. Our findings reveal that somatic cells appear to possess a plasticity feature to express pluripotent marker proteins or to select cell subpopulations that express pluripotent marker proteins when cells are transiently exposed to strong acid. It opens new directions for understanding conserved regulatory mechanisms involved in cellular survival under stressful stimulation.


Asunto(s)
Mucosa Gástrica/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Ácido Clorhídrico/farmacología , Factor 3 de Transcripción de Unión a Octámeros/biosíntesis , Antígenos Embrionarios Específico de Estadio/biosíntesis , Animales , Células Cultivadas , Células HeLa , Humanos , Ratones
5.
Eur J Neurosci ; 52(7): 3710-3722, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32302450

RESUMEN

A subset of presynaptic glutamatergic vesicles in the brain co-releases zinc (Zn2+ ) with glutamate into the synapse. However, the role of synaptically released Zn2+ is still under investigation. Here, we studied the effect of Zn2+ on glutamate homeostasis by measuring the evoked extracellular glutamate level (EGL) and the probability of evoked action potential (PEAP ) at the Zn2+ -containing or zincergic mossy fiber-CA3 synapses of the rat hippocampus. We found that the application of Zn2+ (ZnCl2 ) exerted bidirectional effects on both EGL and PEAP : facilitatory at low concentration (~1 µM) while repressive at high concentration (~50 µM). To determine the action of endogenous Zn2+ , we also used extracellular Zn2+ chelator to remove the synaptically released Zn2+ . Zn2+ chelation reduced both EGL and PEAP , suggesting that endogenous Zn2+ has mainly a facilitative role in glutamate secretion on physiological condition. We revealed that calcium/calmodulin-dependent protein kinase II was integral to the mechanism by which Zn2+ facilitated the release of glutamate. Moreover, a glutamate transporter was the molecular entity for the action of Zn2+ on glutamate uptake by which Zn2+ decreases glutamate availability. Taken together, we show a novel action of Zn2+ , which is to biphasically regulate glutamate homeostasis via Zn2+ concentration-dependent synaptic facilitation and depression. Thus, co-released Zn2+ is physiologically important for enhancing weak stimulation, but potentially mitigates excessive stimulation to keep synaptic transmission within optimal physiological range.


Asunto(s)
Transmisión Sináptica , Zinc , Animales , Ácido Glutámico , Hipocampo , Homeostasis , Ratas , Sinapsis
6.
Cell Signal ; 44: 148-157, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29414441

RESUMEN

Protein kinase C delta (PKCδ) is a Ser/Thr-specific kinase involved in many fundamental cellular processes including growth, differentiation and apoptosis. PKCδ is expressed ubiquitously in all known cell types, and can be activated by diacylglycerol, phorbol esters and other kinases. Multiple lines of evidence have indicated that the mode of activation greatly influences the role PKCδ plays in cellular function. Divalent metal ions, such as zinc are released as a response to cellular stress and injury, often resulting in oxidative damage and cell death. In this study, we evaluate the effect increased concentrations of intracellular zinc has on the phosphorylation state and subcellular localization of PKCδ. More specifically, we demonstrate that intracellular zinc inhibits the phosphorylation of PKCδ at Thr505 in a concentration-dependent manner and facilitates the translocation of PKCδ from the cytosol to the Golgi complex. Analysis of a PKCδ structural model revealed a potential His-Cys3 zinc-binding domain adjacent to residue Thr505 and suggests that interaction with a Zn2+ ion may preclude phosphorylation at this site. This study establishes zinc as a potent modulator of PKCδ function and suggests a novel mechanism by which PKCδ is able to "sense" changes in the concentration of intracellular zinc. These findings illuminate a new paradigm of metal ion-protein interaction that may have significant implications on a broad spectrum of cellular processes.


Asunto(s)
Proteína Quinasa C-delta/metabolismo , Zinc/metabolismo , Citosol/metabolismo , Aparato de Golgi/metabolismo , Células HeLa , Humanos , Fosforilación
7.
Artículo en Inglés | MEDLINE | ID: mdl-29209450

RESUMEN

Cardiovascular disorder occurs when a local blood clot obstructs an artery or a vein to its surround organs, causing related tissues to lose function and die. It is one of the leading causes of mortality and a major cause of disability. The effect of thrombolysis induced by injecting intravenous thrombolytic agents is critical for reducing tissue damages. Streptokinase (SK) is a widely used thrombolytic agent in the treatment of thromboembolism in the blood vessels. A high unit of streptokinase is used in thrombolytic therapies for thrombotic disorders and could improve tissue reperfusion. It is a potent plasminogen activator. However, safety concerns for the usage of a high unit of streptokinase have been raised for the hemorrhagic transformation. In the present study, we studied how zinc would affect streptokinase-induced thrombolysis in vitro, and proposed a strategy to improve streptokinase's effectiveness in promoting thrombolysis. The mice whole blood was used to form the blood clot in vitro by incubating with calcium at 37°C for 30 minutes. Streptokinase was used for inducing thrombolysis measured with the spectrophotometer. Zinc and its chelator, Ca-EDTA, were applied with streptokinase, respectively. Results showed that the co-application zinc inhibited the thrombolytic effect of streptokinase in a dose-dependent manner. Zinc chelator, Ca-EDTA, significantly increased the effect of streptokinase-induced thrombolysis. Our results suggest that zinc chelation improved the efficiency of streptokinase in thrombolysis. The results may have a significant clinical implication by potentially reducing the adverse effect of streptokinase application.

8.
Am J Physiol Cell Physiol ; 313(4): C448-C459, 2017 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-28747335

RESUMEN

Both zinc (Zn2+) and reactive oxygen species (ROS) have been shown to accumulate during hypoxic-ischemic stress and play important roles in pathological processes. To understand the cross talk between the two of them, here we studied Zn2+ and ROS accumulation by employing fluorescent probes in HeLa cells to further the understanding of the cause and effect relationship of these two important cellular signaling systems during chemical-ischemia, stimulated by oxygen and glucose deprivation (OGD). We observed two Zn2+ rises that were divided into four phases in the course of 30 min of OGD. The first Zn2+ rise was a transient, which was followed by a latent phase during which Zn2+ levels recovered; however, levels remained above a basal level in most cells. The final phase was the second Zn2+ rise, which reached a sustained plateau called Zn2+ overload. Zn2+ rises were not observed when Zn2+ was removed by TPEN (a Zn2+ chelator) or thapsigargin (depleting Zn2+ from intracellular stores) treatment, indicating that Zn2+ was from intracellular storage. Damaging mitochondria with FCCP significantly reduced the second Zn2+ rise, indicating that the mitochondrial Zn2+ accumulation contributes to Zn2+ overload. We also detected two OGD-induced ROS rises. Two Zn2+ rises preceded two ROS rises. Removal of Zn2+ reduced or delayed OGD- and FCCP-induced ROS generation, indicating that Zn2+ contributes to mitochondrial ROS generation. There was a Zn2+-induced increase in the functional component of NADPH oxidase, p47phox, thus suggesting that NADPH oxidase may mediate Zn2+-induced ROS accumulation. We suggest a new mechanism of cross talk between Zn2+ and mitochondrial ROS through positive feedback processes that eventually causes excessive free Zn2+ and ROS accumulations during the course of ischemic stress.


Asunto(s)
Isquemia Encefálica/metabolismo , Encéfalo/metabolismo , Glucosa/deficiencia , Mitocondrias/metabolismo , Estrés Oxidativo , Oxígeno/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Zinc/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/patología , Isquemia Encefálica/patología , Hipoxia de la Célula , Quelantes/farmacología , Retroalimentación Fisiológica , Células HeLa , Humanos , Técnicas In Vitro , Masculino , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , NADPH Oxidasas/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ionóforos de Protónes/farmacología , Ratas Sprague-Dawley , Transducción de Señal , Factores de Tiempo
9.
Artículo en Inglés | MEDLINE | ID: mdl-27186321

RESUMEN

Zinc (Zn(2+)) is required for numerous cellular functions. As such, the homeostasis and distribution of intracellular zinc can influence cellular metabolism and signaling. However, the exact distribution of free zinc within live cells remains elusive. Previously we showed the release of zinc from thapsigargin/IP3-sensitive endoplasmic reticulum (ER) storage in cortical neurons. In the present study, we investigated if other cellular organelles also contain free chelatable zinc and function as organelle storage for zinc. To identify free zinc within the organelles, live cells were co-stained with Zinpyr-1, a zinc fluorescent dye, and organelle-specific fluorescent dyes (MitoFluor Red 589: mitochondria; ER Tracker Red: endoplasmic reticulum; BODIPY TR ceramide: Golgi apparatus; Syto Red 64: nucleus). We examined organelles that represent potential storing sites for intracellular zinc. We showed that zinc fluorescence staining was co-localized with MitoFluor Red 589, ER Tracker Red, and BODIPY TR ceramide respectively, suggesting the presence of free zinc in mitochondria, endoplasmic reticulum, and the Golgi apparatus. On the other hand, cytosol and nucleus had nearly no detectable zinc fluorescence. It is known that nucleus contains high amount of zinc binding proteins that have high zinc binding affinity. The absence of zinc fluorescence suggests that there is little free zinc in these two regions. It also indicates that the zinc fluorescence detected in mitochondria, ER and Golgi apparatus represents free chelatable zinc. Taken together, our results support that these organelles are potential zinc storing organelles during cellular zinc homeostasis.

10.
Artículo en Inglés | MEDLINE | ID: mdl-27186322

RESUMEN

Mitochondrial reactive oxygen species (ROS) are known to accumulate during chemical hypoxia, causing adverse effects on cell function and survival. Recent studies show important role zinc accumulation plays in dysfunction associated with hypoxia. It is well known that ROS accumulation also plays a major role in cellular damage by hypoxia. In this study, fluorescent imaging and pharmacological methods were used in live HeLa cells to determine role of zinc in initial ROS accumulation in mitochondria during chemical hypoxia (oxygen glucose depravation with 4 mM sodium dithionite). Accumulation of both was observed as a very rapid phenomenon with initial rapid zinc increase (zinc wave) within 60 seconds of hypoxia onset and ROS increase within 4.5 minutes. Zinc chelation with TPEN removed the initial zinc wave which in turn abolished ROS accumulation. Influx of exogenous zinc induced rapid ROS accumulation. Inhibition of NADPH oxidase with apocynin, a NADPH oxidase inhibitor, showed significant and prolonged reduction in zinc induced ROS accumulation. We proposed a novel mechanism of intracellular zinc increase that activates NADPH oxidase which in turn triggers mitochondrial ROS production.

11.
Acta Neurochir Suppl ; 121: 115-9, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26463933

RESUMEN

Thrombotic cerebral ischemia is one of the leading causes of mortality and chronic disability. Animal models provide an essential tool for understanding the complex cellular and molecular pathophysiology of ischemia and for improving treatment and testing novel neuroprotective drugs in the preclinical setting. In this study, we tested zebrafish as a novel model for thrombotic ischemic brain damage. Zebrafish were intraperitoneally injected with Rose Bengal and light exposure was directed onto the optic tectum region of the brain to induce photothrombosis. After full recovery from anesthesia, zebrafish consistently exhibited abnormal swimming patterns, indicating brain injury from the procedure. The staining of 2,3,5-triphenyltetrazolium chloride (TTC) 24 h after the treatment showed lack of staining of the exposed area of the brain, which further confirmed the ischemic injury. Application of Activase®-tPA improved viability of the brain. The tPA treatment also reduced the occurrence of moving disability as well as the mortality rate, demonstrating that the zebrafish model not only showed focal ischemic injury but also responded well to tPA therapy. Our results suggest that the current photothrombotic method induced focal ischemia in zebrafish and produced consistent brain damage that can be measured by behavioral changes and quantified by histological staining.


Asunto(s)
Isquemia Encefálica/patología , Encéfalo/patología , Modelos Animales de Enfermedad , Accidente Cerebrovascular/patología , Trombosis/patología , Pez Cebra , Animales , Encéfalo/efectos de los fármacos , Encéfalo/fisiopatología , Isquemia Encefálica/etiología , Isquemia Encefálica/fisiopatología , Colorantes , Fibrinolíticos/farmacología , Colorantes Fluorescentes/efectos adversos , Luz/efectos adversos , Rosa Bengala/efectos adversos , Accidente Cerebrovascular/etiología , Accidente Cerebrovascular/fisiopatología , Sales de Tetrazolio , Trombosis/etiología , Trombosis/fisiopatología , Activador de Tejido Plasminógeno/farmacología
12.
Acta Neurochir Suppl ; 121: 347-53, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26463973

RESUMEN

Intracellular zinc release and the generation of reactive oxygen species (ROS) have been reported to be common ingredients in numerous toxic signaling mechanisms in neurons. A key source for intracellular zinc release is its liberation from metallothionein-III (MT-III). MT-III binds and regulates intracellular zinc levels under physiological conditions, but the zinc-binding thiols readily react with certain ROS and reactive nitrogen species (RNS) to result in intracellular zinc liberation. Liberated zinc induces ROS and RNS generation by multiple mechanisms, including the induction of mitochondrial ROS production, and also promotes ROS formation outside the mitochondria by interaction with the enzymes NADPH oxidase and 12-lipoxygenase. Of particular relevance to neuronal injury in the context of ischemia and prolonged seizures, the positive feedback cycle between ROS/RNS generation and increasing zinc liberation will be examined.


Asunto(s)
Lesiones Encefálicas/metabolismo , Especies de Nitrógeno Reactivo/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Zinc/metabolismo , Araquidonato 12-Lipooxigenasa/metabolismo , Calcio/metabolismo , Humanos , Mitocondrias/metabolismo , NADPH Oxidasas/metabolismo , Neuronas/metabolismo
13.
Mol Neurobiol ; 53(1): 320-330, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25432887

RESUMEN

Granulocyte colony-stimulating factor (G-CSF) is a hematopoietic growth factor with strong neuroprotective properties. However, it has limited capacity to cross the blood-brain barrier and thus potentially limiting its protective capacity. Recent studies demonstrated that intranasal drug administration is a promising way in delivering neuroprotective agents to the central nervous system. The current study therefore aimed at determining whether intranasal administration of G-CSF increases its delivery to the brain and its neuroprotective effect against ischemic brain injury. Transient focal cerebral ischemia in rat was induced with middle cerebral artery occlusion. Our resulted showed that intranasal administration is 8-12 times more effective than subcutaneous injection in delivering G-CSF to cerebrospinal fluid and brain parenchyma. Intranasal delivery enhanced the protective effects of G-CSF against ischemic injury in rats, indicated by decreased infarct volume and increased recovery of neurological function. The neuroprotective mechanisms of G-CSF involved enhanced upregulation of HO-1 and reduced calcium overload following ischemia. Intranasal G-CSF application also promoted angiogenesis and neurogenesis following brain ischemia. Taken together, G-CSF is a legitimate neuroprotective agent and intranasal administration of G-CSF is more effective in delivery and neuroprotection and could be a practical approach in clinic.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Factor Estimulante de Colonias de Granulocitos/administración & dosificación , Factor Estimulante de Colonias de Granulocitos/uso terapéutico , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/uso terapéutico , Administración Intranasal , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Infarto Encefálico/complicaciones , Infarto Encefálico/tratamiento farmacológico , Infarto Encefálico/fisiopatología , Isquemia Encefálica/complicaciones , Isquemia Encefálica/fisiopatología , Calcio/metabolismo , Citoesqueleto/metabolismo , Factor Estimulante de Colonias de Granulocitos/sangre , Factor Estimulante de Colonias de Granulocitos/líquido cefalorraquídeo , Hemo-Oxigenasa 1/metabolismo , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/fisiopatología , Espacio Intracelular/metabolismo , Masculino , Neovascularización Fisiológica/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Ratas Sprague-Dawley , Tubulina (Proteína)/metabolismo , Regulación hacia Arriba/efectos de los fármacos
14.
Brain Res ; 1615: 129-138, 2015 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-25920370

RESUMEN

Hydrogen-rich water (HRW) has anti-oxidant activities, and it exerts neuroprotective effects during ischemia-reperfusion brain injury. Parvalbumin and hippocalcin are two calcium buffering proteins, which are involved in neuronal differentiation, maturation and apoptosis. The aim of this study was to investigate whether HRW could moderate parvalbumin and hippocalcin expression during ischemic brain injury and glutamate toxicity-induced neuronal cell death. Focal brain ischemia was induced in male Sprague-Dawley rats by middle cerebral artery occlusion (MCAO). Rats were treated with H2O or HRW (6 ml/kg per rat) before and after MCAO, and cerebral cortical tissues were collected 1, 7 and 14 days after MCAO. Based on our results, HRW treatment was able to reduce brain infarct volume and improve neurological function following ischemic brain injury. In addition, HRW prevented the ischemia-induced reduction of parvalbumin and hippocalcin levels in vivo and also reduced the glutamate toxicity-induced death of neurons, including the dose-dependent reduction of glutamate toxicity-associated proteins in vitro. Moreover, HRW attenuated the glutamate toxicity-induced elevate in intracellular Ca(2+) levels. All these results suggest that HRW could protect against ischemic brain injury and that the maintenance of parvalbumin and hippocalcin levels by HRW during ischemic brain injury might contribute to the neuroprotective effects against neuron damage.


Asunto(s)
Isquemia Encefálica/metabolismo , Calcio/metabolismo , Hipocalcina/metabolismo , Hidrógeno/administración & dosificación , Fármacos Neuroprotectores/administración & dosificación , Parvalbúminas/metabolismo , Agua/administración & dosificación , Animales , Isquemia Encefálica/prevención & control , Isquemia Encefálica/psicología , Ácido Glutámico/toxicidad , Infarto de la Arteria Cerebral Media/complicaciones , Masculino , Ratas , Ratas Sprague-Dawley , Agua/química
15.
Endocrine ; 50(1): 110-22, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25771886

RESUMEN

It is well known that zinc (Zn(2+)) is required for the process of insulin biosynthesis and the maturation of insulin secretory granules in pancreatic beta (ß)-cells, and that changes in Zn(2+) levels in the pancreas have been found to be associated with diabetes. Glucose-stimulation causes a rapid co-secretion of Zn(2+) and insulin with similar kinetics. However, we do not know whether Zn(2+) regulates insulin availability and secretion. Here we investigated the effect of Zn(2+) on glucose-stimulated insulin secretion (GSIS) in isolated mouse pancreatic islets. Whereas Zn(2+) alone (control) had no effect on the basal secretion of insulin, it significantly inhibited GSIS. The application of CaEDTA, by removing the secreted Zn(2+) from the extracellular milieu of the islets, resulted in significantly increased GSIS, suggesting an overall inhibitory role of secreted Zn(2+) on GSIS. The inhibitory action of Zn(2+) was mostly mediated through the activities of KATP/Ca(2+) channels. Furthermore, during brief paired-pulse glucose-stimulated Zn(2+) secretion (GSZS), Zn(2+) secretion following the second pulse was significantly attenuated, probably by the secreted endogenous Zn(2+) after the first pulse. Such an inhibition on Zn(2+) secretion following the second pulse was completely reversed by Zn(2+) chelation, suggesting a negative feedback mechanism, in which the initial glucose-stimulated Zn(2+) release inhibits subsequent Zn(2+) secretion, subsequently inhibiting insulin co-secretion as well. Taken together, these data suggest a negative feedback mechanism on GSZS and GSIS by Zn(2+) secreted from ß-cells, and the co-secreted Zn(2+) may act as an autocrine inhibitory modulator.


Asunto(s)
Comunicación Autocrina/fisiología , Glucosa/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Zinc/metabolismo , Animales , Cationes Bivalentes , Células Cultivadas , Femenino , Secreción de Insulina , Ratones , Ratones Endogámicos C57BL
16.
Endocrine ; 45(2): 178-89, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23979673

RESUMEN

Zinc (Zn2+) is an essential element crucial for growth and development, and also plays a role in cell signaling for cellular processes like cell division and apoptosis. In the mammalian pancreas, Zn2+ is essential for the correct processing, storage, secretion, and action of insulin in beta (ß)-cells. Insulin is stored inside secretory vesicles or granules, where two Zn2+ ions coordinate six insulin monomers to form the hexameric-structure on which maturated insulin crystals are based. The total Zn2+ content of the mammalian pancreas is among the highest in the body, and Zn2+ concentration reach millimolar levels in the interior of the dense-core granule. Changes in Zn2+ levels in the pancreas have been found to be associated with diabetes. Hence, the relationship between co-stored Zn2+ and insulin undoubtedly is critical to normal ß-cell function. The advances in the field of Zn2+ biology over the last decade have facilitated our understanding of Zn2+ trafficking, its intracellular distribution and its storage. When exocytosis of insulin occurs, insulin granules fuse with the ß-cell plasma membrane and release their contents, i.e., insulin as well as substantial amount of free Zn2+, into the extracellular space and the local circulation. Studies increasingly indicate that secreted Zn2+ has autocrine or paracrine signaling in ß-cells or the neighboring cells. This review discusses the Zn2+ homeostasis in ß-cells with emphasis on the potential signaling role of Zn2+ to islet biology.


Asunto(s)
Homeostasis/fisiología , Células Secretoras de Insulina/fisiología , Insulina/fisiología , Zinc/fisiología , Animales , Humanos , Células Secretoras de Insulina/citología , Islotes Pancreáticos/citología , Islotes Pancreáticos/fisiología , Modelos Animales , Comunicación Paracrina/fisiología , Transducción de Señal/fisiología
17.
Exp Physiol ; 98(8): 1301-11, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23603373

RESUMEN

Diminished or inappropriate secretion of insulin is associated with type II diabetes. The cellular/molecular mechanism coupled with the regulation of insulin secretion is still under intense investigation. Divalent ion zinc (Zn(2+)) is co-packaged and co-secreted with insulin and is intimately involved in the process of insulin biosynthesis and the maturation of insulin secretory granules. The study reported here investigated glucose-stimulated zinc secretion (GSZS) and the effect of zinc on glucose-stimulated insulin secretion (GSIS) in the HIT-T15 pancreatic ß-cell line. Zinc secretion was measured using a newly developed fluorescent zinc imaging approach, and the insulin secretion was measured using an enzyme-linked immunosorbent assay. There was apparent granular-like zinc staining in ß-cells. The application of glucose induced detectable zinc secretion or GSZS. Like GSIS, GSZS was dependent on the glucose concentration (5-20 mm) and the presence of extracellular calcium. The application of a zinc chelator enhanced GSZS. When brief paired-pulse glucose stimulations, which involve the initial glucose stimulation followed by a second round of glucose stimulation, were applied, zinc secretion or GSZS that followed the first pulse was inhibited. This inhibition was reversed by zinc chelation, suggesting a feedback mechanism on GSZS by zinc secreted from ß-cells. Finally, the application of zinc (50 µm) strongly inhibited GSIS as measured by enzyme-linked immunosorbent assay. The present study suggests that insulin secretion is regulated by co-secreted zinc that may act as an autocrine inhibitory modulator.


Asunto(s)
Glucosa/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/fisiología , Insulina/metabolismo , Zinc/metabolismo , Zinc/farmacología , Animales , Calcio/metabolismo , Línea Celular , Células Secretoras de Insulina/metabolismo , Vesículas Secretoras/efectos de los fármacos , Vesículas Secretoras/metabolismo , Vesículas Secretoras/fisiología
18.
Zebrafish ; 10(1): 30-5, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23461417

RESUMEN

A study was conducted using zebrafish as a model of hypoxic brain injury to investigate the potential neuroprotective effects of zinc (Zn(2+)) chelation. The accumulation of intracellular Zn(2+) is a significant causal factor of the neuronal injury, and has been implicated in cell death followed by ischemic stroke. In this study, the zebrafish was placed in the hypoxia chamber with an extremely low level of dissolved oxygen (less than 0.8 mg/L), which is similar to the conditions in a complete global ischemic stroke. Approximately 50% of zebrafish died after a short period (≈11 min) of hypoxic treatment, suggesting that this is a responsive model system for use in evaluating treatments for hypoxic brain damage. The application of DEDTC reduced intracellular Zn(2+) accumulation and produced a concentration-dependent effect by increasing the survival rate of zebrafish. Zn(2+) chelation also enhanced zebrafish tolerance for hypoxia. When the brain damages were evaluated with TTC staining, the zebrafish that were treated with DEDTC in hypoxic treatment yielded the improvement of TTC staining that was similar to the healthy zebrafish brain. The results support that rising intracellular Zn(2+) plays a critical role in the neuronal damages, and demonstrate the protective effects of Zn(2+) chelation in hypoxic-ischemic brain injury in zebrafish.


Asunto(s)
Quelantes/metabolismo , Modelos Animales de Enfermedad , Ditiocarba/análogos & derivados , Hipoxia-Isquemia Encefálica/prevención & control , Fármacos Neuroprotectores/metabolismo , Pez Cebra , Zinc/metabolismo , Animales , Ditiocarba/metabolismo , Relación Dosis-Respuesta a Droga , Humanos , Hipoxia-Isquemia Encefálica/etiología , Hipoxia-Isquemia Encefálica/metabolismo , Oxígeno/metabolismo , Espectrofotometría , Sales de Tetrazolio/metabolismo
19.
Exp Diabetes Res ; 2012: 190309, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22536213

RESUMEN

Zinc (Zn(2+)) appears to be intimately involved in insulin metabolism since insulin secretion is correlated with zinc secretion in response to glucose stimulation, but little is known about the regulation of zinc homeostasis in pancreatic beta-cells. This study set out to identify the intracellular zinc transient by imaging free cytosolic zinc in HIT-T15 beta-cells with fluorescent zinc indicators. We observed that membrane depolarization by KCl (30-60 mM) was able to induce a rapid increase in cytosolic concentration of zinc. Multiple zinc transients of similar magnitude were elicited during repeated stimulations. The amplitude of zinc responses was not affected by the removal of extracellular calcium or zinc. However, the half-time of the rising slope was significantly slower after removing extracellular zinc with zinc chelator CaEDTA, suggesting that extracellular zinc affect the initial rising phase of zinc response. Glucose (10 mM) induced substantial and progressive increases in intracellular zinc concentration in a similar way as KCl, with variation in the onset and the duration of zinc mobilization. It is known that the depolarization of beta-cell membrane is coupled with the secretion of insulin. Rising intracellular zinc concentration may act as a critical signaling factor in insulin metabolism of pancreatic beta-cells.


Asunto(s)
Glucosa/metabolismo , Homeostasis/fisiología , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Potenciales de la Membrana/fisiología , Zinc/metabolismo , Animales , Línea Celular , Células Cultivadas , Cricetinae , Glucosa/farmacología , Homeostasis/efectos de los fármacos , Secreción de Insulina , Células Secretoras de Insulina/efectos de los fármacos , Potenciales de la Membrana/efectos de los fármacos , Mesocricetus
20.
Artículo en Inglés | MEDLINE | ID: mdl-21760967

RESUMEN

Acute cerebral ischemia is one of the leading causes of mortality and chronic disability. Animal models provide an essential tool for understanding the complex cellular and molecular pathophysiology of hypoxic-ischemia and for testing novel neuroprotective drugs in the pre-clinical setting. In this study we tested zebrafish as a novel model for hypoxic-ischemic brain damage. We built an air-proof chamber where water inside had a low oxygen concentration (0.6-0.8 mg/L) proximate to complete hypoxia. Each zebrafish was placed individually in the hypoxia chamber and was subjected to hypoxia treatment until it became motionless, lying on its side on the bottom of the chamber (time to hypoxia = 679.52 ± 90 seconds, mean ± SD, n =23), followed by transferring into a recovery beaker. Overall, 60.87% of subjects did not recover from hypoxia while 39% survived. The size and distribution of brain injury were determined by triphenyltetrazolium chloride (TTC) staining. Bilateral, moderate to complete TTC decoloration or demarcation of the infarct after 10 minutes of hypoxic treatment was clearly visible in the optic tectum of the optic lobe. The size of the infarct expanded to the deep structure of the optic lobe with longer hypoxic treatments. The zebrafish that survived hypoxia experienced initial twitching followed by unbalanced erratic movements until they regained coordinated, balanced swimming ability. These data indicate that zebrafish are susceptible to hypoxic attack and suggest that the model we present in this study can be used as an alternative model to evaluate hypoxia-induced brain damage.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...