Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biochim Biophys Acta Mol Basis Dis ; 1863(4): 929-935, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28131914

RESUMEN

Events responsible for cerebrovascular disease in diabetes are not fully understood. Pericyte loss is an early event that leads to endothelial cell death, microaneurysms, and cognitive impairment. A biochemical mechanism underlying pericyte loss is rapid respiration (oxidative metabolism of glucose). This escalation in respiration results from free influx of glucose into insulin-insensitive tissues in the face of high glucose levels in the blood. Rapid respiration generates superoxide, the precursor to all reactive oxygen species (ROS), and results in pericyte death. Respiration is regulated by carbonic anhydrases (CAs) VA and VB, the two isozymes expressed in mitochondria, and their pharmacologic inhibition with topiramate reduces respiration, ROS, and pericyte death. Topiramate inhibits both isozymes; therefore, in the earlier studies, their individual roles were not discerned. In a recent genetic study, we showed that mitochondrial CA VA plays a significant role in regulation of reactive oxygen species and pericyte death. The role of CA VB was not addressed. In this report, genetic knockdown and overexpression studies confirm that mitochondrial CA VA regulates respiration in pericytes, whereas mitochondrial CA VB does not contribute significantly. Identification of mitochondrial CA VA as a sole regulator of respiration provides a specific target to develop new drugs with fewer side effects that may be better tolerated and can protect the brain from diabetic injury. Since similar events occur in the capillary beds of other insulin-insensitive tissues such as the eye and kidney, these drugs may also slow the onset and progression of diabetic disease in these tissues.


Asunto(s)
Apoptosis , Encéfalo/enzimología , Anhidrasa Carbónica V/metabolismo , Trastornos Cerebrovasculares/enzimología , Angiopatías Diabéticas/prevención & control , Mitocondrias/enzimología , Proteínas Mitocondriales/metabolismo , Pericitos/enzimología , Animales , Encéfalo/patología , Anhidrasa Carbónica V/genética , Línea Celular Transformada , Trastornos Cerebrovasculares/genética , Trastornos Cerebrovasculares/patología , Angiopatías Diabéticas/enzimología , Angiopatías Diabéticas/genética , Angiopatías Diabéticas/patología , Ratones , Mitocondrias/patología , Proteínas Mitocondriales/genética , Pericitos/patología
2.
J Pharmacol Exp Ther ; 359(3): 452-459, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27729477

RESUMEN

All forms of diabetes mellitus are characterized by chronic hyperglycemia, resulting in the development of a number of microvascular and macrovascular pathologies. Diabetes is also associated with changes in brain microvasculature, leading to dysfunction and ultimately disruption of the blood-brain barrier (BBB). These changes are correlated with a decline in cognitive function. In diabetes, BBB damage is associated with increased oxidative stress and reactive oxygen species. This occurs because of the increased oxidative metabolism of glucose caused by hyperglycemia. Decreasing the production of bicarbonate with the use of a mitochondrial carbonic anhydrase inhibitor (mCAi) limits oxidative metabolism and the production of reactive oxygen species. In this study, we have demonstrated that 1) streptozotocin-induced diabetes resulted in BBB disruption, 2) ultrastructural studies showed a breakdown of the BBB and changes to the neurovascular unit (NVU), including a loss of brain pericytes and retraction of astrocytes, the two cell types that maintain the BBB, and 3) treatment with topiramate, a mCAi, attenuated the effects of diabetes on BBB disruption and ultrastructural changes in the neurovascular unit.


Asunto(s)
Vasos Sanguíneos/fisiopatología , Barrera Hematoencefálica/efectos de los fármacos , Inhibidores de Anhidrasa Carbónica/farmacología , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Fructosa/análogos & derivados , Mitocondrias/enzimología , Animales , Glucemia/metabolismo , Vasos Sanguíneos/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Diabetes Mellitus Experimental/fisiopatología , Fructosa/farmacología , Masculino , Ratones , Permeabilidad/efectos de los fármacos , Topiramato
3.
Artículo en Inglés | MEDLINE | ID: mdl-26167540

RESUMEN

Hyperglycemia in diabetes mellitus causes oxidative stress and pericyte depletion from the microvasculature of the brain thus leading to the Blood-Brain Barrier (BBB) disruption. The compromised BBB exposes the brain to circulating substances, resulting in neurotoxicity and neuronal cell death. The decline in pericyte numbers in diabetic mouse brain and pericyte apoptosis in high glucose cultures are caused by excess superoxide produced during enhanced respiration (mitochondrial oxidative metabolism of glucose). Superoxide is precursor to all Reactive Oxygen Species (ROS) which, in turn, cause oxidative stress. The rate of respiration and thus the ROS production is regulated by mitochondrial carbonic anhydrases (mCA) VA and VB, the two isoforms expressed in the mitochondria. Inhibition of both mCA: decreases the oxidative stress and restores the pericyte numbers in diabetic brain; and reduces high glucose-induced respiration, ROS, oxidative stress, and apoptosis in cultured brain pericytes. However, the individual role of the two isoforms has not been established. To investigate the contribution of mCA VA in ROS production and apoptosis, a mCA VA overexpressing brain pericyte cell line was engineered. These cells were exposed to high glucose and analyzed for the changes in ROS and apoptosis. Overexpression of mCA VA significantly increased pericyte ROS and apoptosis. Inhibition of mCA VA with topiramate prevented increases both in glucose-induced ROS and pericyte death. These results demonstrate, for the first time, that mCA VA regulates the rate of pericyte respiration. These findings identify mCA VA as a novel and specific therapeutic target to protect the cerebromicrovascular bed in diabetes.

4.
Int Libr Diabetes Metab ; 1(1): 6-12, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26120599

RESUMEN

Diabetes mellitus-associated damage to the microvasculature of the brain is caused by hyperglycemia-induced oxidative stress, which results in pericyte loss, blood-brain barrier disruption, and impaired cognitive function. Oxidative stress, in diabetes, is caused by reactive oxygen species produced during accelerated respiration (mitochondrial oxidative metabolism of glucose). The rate of respiration is regulated by mitochondrial carbonic anhydrases (CAs). Inhibition of these enzymes protects the brain from diabetic damage. Previously, we reported that topiramate, a mitochondrial CA inhibitor, at a dose of 50 mg/kg/day protects the brain in diabetes by reducing oxidative stress and restoring pericyte numbers. Topiramate has high affinity for both mitochondrial CAs; therefore, it is conceivable that a much lower dose may inhibit these enzymes and thus protect the brain from hyperglycemia-induced oxidative damage. Therefore, in an effort to reduce the toxicity associated with higher doses of topiramate, the current study was designed to investigate the effect of 1.0 mg/kg topiramate on reducing oxidative stress, restoring pericyte numbers in the brain, and improving the impaired learning behavior in diabetic mouse. Diabetes was induced by a one-time injection of streptozotocin and topiramate was administered daily for 12 weeks. Levels of oxidative stress, reduced glutathione (GSH) and 4-hydroxy-2-trans-nonenal (HNE) were measured in the brain and pericyte/endothelial cell ratios in isolated brain microvessels. Learning behavior was assessed by T-maze foot shock avoidance test. A significant decrease in GSH (control, 12.2 ± 0.4 vs. diabetic, 10.8 ± 0.4 vs. diabetic + topiramate, 12.6 ± 0.6, p<0.05) and an increase in HNE (control, 100 ± 4.2, vs. diabetic, 127.3 ± 8.8 vs. diabetic + topiramate, 93.9 ± 8.4 p<0.05) in diabetic mice were corrected by topiramate treatment. Topiramate treatment also resulted in restoration of pericyte numbers in diabetic mice (control, 25.89 ± 0.85 vs. diabetic, 18.14 ± 0.66 vs. diabetic + topiramate, 24.35 ± 0.53, p<0.001) and improvement in learning behavior. In conclusion, these data clearly demonstrate that topiramate at 1.0 mg/kg protects the mouse brain from diabetic damage. A 1.0 mg/kg topiramate in the mouse translates to a 5.0 mg daily dose in a 60 kg human, which may help slow the onset and progression of diabetic complications in the human brain.

5.
J Alzheimers Dis ; 46(2): 535-48, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25854933

RESUMEN

The senescence-accelerated mouse (SAMP8) strain exhibits an age-related decrease in memory accompanied by an increase in hippocampal amyloid-ß protein precursor (AßPP) and amyloid-ß peptide (Aß). We have shown that administration of an antisense oligonucleotide against the Aß region of AßPP (AßPP antisense) reverses the memory deficits. The purpose of this study was to determine the effect of peripheral (IV) administration of AßPP antisense on hippocampal gene expression. The AßPP antisense reversed the memory deficits and altered expression of 944 hippocampal genes. Pathway analysis showed significant gene expression changes in nine pathways. These include the MAPK signaling pathway (p = 0.0078) and the phosphatidylinositol signaling pathway (p = 0.043), which we have previously shown to be altered in SAMP8 mice. The changes in these pathways contributed to significant changes in the neurotropin (p = 0.0083) and insulin signaling (p = 0.015) pathways, which are known to be important in learning and memory. Changes in these pathways were accompanied by phosphorylation changes in the downstream target proteins p70S6K, GSK3ß, ERK, and CREB. These changes in hippocampal gene expression and protein phosphorylation may suggest specific new targets for antisense therapy aimed at improving memory.


Asunto(s)
Péptidos beta-Amiloides/química , Hipocampo/efectos de los fármacos , Aprendizaje por Laberinto/efectos de los fármacos , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/genética , Memoria/efectos de los fármacos , Oligonucleótidos Antisentido/administración & dosificación , Animales , Modelos Animales de Enfermedad , Expresión Génica , Ratones , Fosforilación , Transducción de Señal
6.
PLoS One ; 9(9): e108034, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25251220

RESUMEN

Traumatic brain injury (TBI) in its various forms has emerged as a major problem for modern society. Acute TBI can transform into a chronic condition and be a risk factor for neurodegenerative diseases such as Alzheimer's and Parkinson's diseases, probably through induction of oxidative stress and neuroinflammation. Here, we examined the ability of the antioxidant molecular hydrogen given in drinking water (molecular hydrogen water; mHW) to alter the acute changes induced by controlled cortical impact (CCI), a commonly used experimental model of TBI. We found that mHW reversed CCI-induced edema by about half, completely blocked pathological tau expression, accentuated an early increase seen in several cytokines but attenuated that increase by day 7, reversed changes seen in the protein levels of aquaporin-4, HIF-1, MMP-2, and MMP-9, but not for amyloid beta peptide 1-40 or 1-42. Treatment with mHW also reversed the increase seen 4 h after CCI in gene expression related to oxidation/carbohydrate metabolism, cytokine release, leukocyte or cell migration, cytokine transport, ATP and nucleotide binding. Finally, we found that mHW preserved or increased ATP levels and propose a new mechanism for mHW, that of ATP production through the Jagendorf reaction. These results show that molecular hydrogen given in drinking water reverses many of the sequelae of CCI and suggests that it could be an easily administered, highly effective treatment for TBI.


Asunto(s)
Antioxidantes/uso terapéutico , Edema Encefálico/tratamiento farmacológico , Lesiones Encefálicas/tratamiento farmacológico , Agua Potable , Hidrógeno/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Animales , Antioxidantes/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/patología , Edema Encefálico/sangre , Edema Encefálico/etiología , Edema Encefálico/patología , Lesiones Encefálicas/sangre , Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/patología , Citocinas/análisis , Citocinas/sangre , Agua Potable/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Hidrógeno/metabolismo , Masculino , Ratones Endogámicos C57BL , Fármacos Neuroprotectores/metabolismo
7.
Cell Mol Neurobiol ; 34(4): 473-8, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24643366

RESUMEN

In order to define the molecular anatomy of the blood-brain barrier (BBB) that may be relevant to either barrier or transport function, proteins that are overexpressed in the cerebral microvessels should be identified. We used differential display to identify novel proteins that are overexpressed or unique to the BBB. DNA sequence analysis is one of the differentially expressed transcripts showed that it is highly homologous with the ATPase class I, type 8B, and member 1 (ATP8B1) protein and contains an ATPase domain and a phospholipid-binding domain. ATP8B1 is expressed in the BBB microvessels but not brain tissue lacking microvessels. Likewise, ATP8B1 was enriched in BBB microvessels similar to glucose transporter 1. Immunohistochemistry using an ATP8B1-specific antibody demonstrated preferential staining of the microvessels within the cerebral tissue. These results suggest that ATP8B1, a P-type aminophospholipid translocase, is enriched in cerebral microvessels and may have a role in plasma membrane lipid transport.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Barrera Hematoencefálica/metabolismo , Membrana Celular/metabolismo , Endotelio Vascular/metabolismo , Microvasos/metabolismo , Proteínas de Transferencia de Fosfolípidos/metabolismo , Animales , Perfilación de la Expresión Génica , Proteínas de Transporte de Membrana/metabolismo , Ratas Endogámicas F344
8.
Neurobiol Aging ; 35(1): 159-68, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23969180

RESUMEN

The senescence-accelerated mouse (SAMP8) strain exhibits decreased learning and memory and increased amyloid beta (Aß) peptide accumulation at 12 months. To detect differences in gene expression in SAMP8 mice, we used a control mouse that was a 50% cross between SAMP8 and CD-1 mice and which showed no memory deficits (50% SAMs). We then compared gene expression in the hippocampus of 4- and 12-month-old SAMP8 and control mice using Affymetrix gene arrays. At 12 months, but not at 4 months, pathway analysis revealed significant differences in the long term potentiation (6 genes), phosphatidylinositol signaling (6 genes), and endocytosis (10 genes) pathways. The changes in long term potentiation included mitogen-activated protein kinase (MAPK) signaling (N-ras, cAMP responsive element binding protein [CREB], protein phosphatase inhibitor 1) and Ca-dependent signaling (inositol triphosphate [ITP] receptors 1 and 2 and phospholipase C). Changes in phosphatidylinositol signaling genes suggested altered signaling through phosphatidylinositol-3-kinase, and Western blotting revealed phosphorylation changes in serine/threonine protein kinase AKT and 70S6K. Changes in the endocytosis pathway involved genes related to clathrin-mediated endocytosis (dynamin and clathrin). Endocytosis is required for receptor recycling, is involved in Aß metabolism, and is regulated by phosphatidylinositol signaling. In summary, these studies demonstrate altered gene expression in 3 SAMP8 hippocampal pathways associated with memory formation and consolidation. These pathways might provide new therapeutic targets in addition to targeting Aß metabolism itself.


Asunto(s)
Envejecimiento/genética , Endocitosis/genética , Endocitosis/fisiología , Regulación del Desarrollo de la Expresión Génica/genética , Expresión Génica , Hipocampo , Potenciación a Largo Plazo/genética , Trastornos de la Memoria/genética , Fosfatidilinositoles/genética , Transducción de Señal/genética , Envejecimiento/fisiología , Péptidos beta-Amiloides/metabolismo , Animales , Regulación del Desarrollo de la Expresión Génica/fisiología , Hipocampo/metabolismo , Hipocampo/fisiología , Aprendizaje/fisiología , Potenciación a Largo Plazo/fisiología , Memoria/fisiología , Ratones , Ratones Endogámicos , Fosfatidilinositoles/fisiología , Transducción de Señal/fisiología
9.
Biochem Biophys Res Commun ; 440(2): 354-8, 2013 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-24076121

RESUMEN

Hyperglycemia-induced oxidative stress leads to diabetes-associated damage to the microvasculature of the brain. Pericytes in close proximity to endothelial cells in the brain microvessels are vital to the integrity of the blood-brain barrier and are especially susceptible to oxidative stress. According to our recently published results, streptozotocin-diabetic mouse brain exhibits oxidative stress and loose pericytes by twelve weeks of diabetes, and cerebral pericytes cultured in high glucose media suffer intracellular oxidative stress and apoptosis. Oxidative stress in diabetes is hypothesized to be caused by reactive oxygen species (ROS) produced during hyperglycemia-induced enhanced oxidative metabolism of glucose (respiration). To test this hypothesis, we investigated the effect of high glucose on respiration rate and ROS production in mouse cerebral pericytes. Previously, we showed that pharmacological inhibition of mitochondrial carbonic anhydrases protects the brain from oxidative stress and pericyte loss. The high glucose-induced intracellular oxidative stress and apoptosis of pericytes in culture were also reversed by inhibition of mitochondrial carbonic anhydrases. Therefore, we extended our current study to determine the effect of these inhibitors on high glucose-induced increases in pericyte respiration and ROS. We now report that both the respiration and ROS are significantly increased in pericytes challenged with high glucose. Furthermore, inhibition of mitochondrial carbonic anhydrases significantly slowed down both the rate of respiration and ROS production. These data provide new evidence that pharmacological inhibitors of mitochondrial carbonic anhydrases, already in clinical use, may prove beneficial in protecting the brain from oxidative stress caused by ROS produced as a consequence of hyperglycemia-induced enhanced respiration.


Asunto(s)
Inhibidores de Anhidrasa Carbónica/farmacología , Glucosa/administración & dosificación , Mitocondrias/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Pericitos/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Animales , Barrera Hematoencefálica/efectos de los fármacos , Respiración de la Célula/efectos de los fármacos , Células Cultivadas , Diabetes Mellitus Experimental/metabolismo , Etoxzolamida/farmacología , Fructosa/análogos & derivados , Fructosa/farmacología , Hiperglucemia/fisiopatología , Ratones , Mitocondrias/metabolismo , Pericitos/metabolismo , Topiramato
10.
Proc Natl Acad Sci U S A ; 110(18): 7423-8, 2013 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-23589845

RESUMEN

Prior studies with carbonic anhydrase (CA) inhibitors implicated mitochondrial CA in ureagenesis and gluconeogenesis. Subsequent studies identified two mitochondrial CAs. To distinguish the contribution of each enzyme, we studied the effects of targeted disruption of the murine CA genes, called Car5A and Car5B. The Car5A mutation had several deleterious consequences. Car5A null mice were smaller than wild-type littermates and bred poorly. However, on sodium-potassium citrate-supplemented water, they produced offspring in expected numbers. Their blood ammonia concentrations were markedly elevated, but their fasting blood sugars were normal. By contrast, Car5B null mice showed normal growth and normal blood ammonia levels. They too had normal fasting blood sugars. Car5A/B double-knockout (DKO) mice showed additional abnormalities. Impaired growth was more severe than for Car5A null mice. Hyperammonemia was even greater as well. Although fertile, DKO animals were produced in less-than-predicted numbers even when supplemented with sodium-potassium citrate in their drinking water. Survival after weaning was also reduced, especially for males. In addition, fasting blood glucose levels for DKO mice were significantly lower than for controls (153 ± 33 vs. 230 ± 24 mg/dL). The enhanced hyperammonemia and lower fasting blood sugar, which are both seen in the DKO mice, indicate that both Car5A and Car5B contribute to both ammonia detoxification (ureagenesis) and regulation of fasting blood sugar (gluconeogenesis). Car5A, which is expressed mainly in liver, clearly has the predominant role in ammonia detoxification. The contribution of Car5B to ureagenesis and gluconeogenesis was evident only on a Car5A null background.


Asunto(s)
Amoníaco/metabolismo , Anhidrasa Carbónica V/genética , Marcación de Gen , Glucosa/metabolismo , Mitocondrias/enzimología , Mutagénesis/genética , Amoníaco/sangre , Animales , Glucemia/metabolismo , Anhidrasa Carbónica V/metabolismo , Femenino , Genotipo , Inactivación Metabólica , Masculino , Ratones , Ratones Noqueados , ARN Mensajero/genética , ARN Mensajero/metabolismo , Análisis de Supervivencia , Aumento de Peso
11.
J Pharmacol Exp Ther ; 344(3): 637-45, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23249625

RESUMEN

Diabetes-associated complications in the microvasculature of the brain are caused by oxidative stress, generated by overproduction of reactive oxygen species from hyperglycemia-induced accelerated oxidative metabolism of glucose. Pericytes, essential for the viability of the microvasculature, are especially susceptible to oxidative stress. Mitochondrial carbonic anhydrases, regulators of the oxidative metabolism of glucose, determine the rate of reactive oxygen species production and inhibition of mitochondrial carbonic anhydrases rescues glucose-induced pericyte loss in the diabetic mouse brain. We hypothesized that high glucose induces intracellular oxidative stress and pericyte apoptosis and that inhibition of mitochondrial carbonic anhydrases protects pericytes from oxidative stress-induced apoptosis. To validate our hypothesis, conditionally immortalized cerebral pericyte (IPC) cultures were established from Immortomice to investigate the effect of high glucose on oxidative stress and pericyte apoptosis. The IPCs expressed pericyte markers and induced high transendothelial electrical resistance and low permeability in brain endothelial cell monolayers comparable with pericytes in primary cultures. The IPCs also secreted cytokines constitutively and in response to lipopolysaccharide similar to pericytes. High glucose caused oxidative stress and apoptosis of these cells, with both oxidative stress and apoptosis significantly reduced after mitochondrial carbonic anhydrase inhibition. These results provide the first evidence that pharmacological inhibition of mitochondrial carbonic anhydrases attenuates pericyte apoptosis caused by high glucose-induced oxidative stress. Carbonic anhydrase inhibitors have a long history of safe clinical use and can be immediately evaluated for this new indication in translational research. Thus, mitochondrial carbonic anhydrases may provide a new therapeutic target for oxidative stress-related illnesses of the brain.


Asunto(s)
Apoptosis/efectos de los fármacos , Inhibidores de Anhidrasa Carbónica/farmacología , Glucosa/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/enzimología , Estrés Oxidativo/efectos de los fármacos , Pericitos/efectos de los fármacos , Animales , Anhidrasas Carbónicas/metabolismo , Células Cultivadas , Cerebro/efectos de los fármacos , Cerebro/enzimología , Cerebro/metabolismo , Quimiocinas/metabolismo , Citocinas/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/enzimología , Células Endoteliales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Neuroglía/efectos de los fármacos , Neuroglía/enzimología , Pericitos/enzimología , Pericitos/metabolismo , Especies Reactivas de Oxígeno/metabolismo
12.
J Neurosci ; 32(47): 16754-62, 2012 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-23175829

RESUMEN

In the hippocampus, extracellular carbonic anhydrase (Car) speeds the buffering of an activity-generated rise in extracellular pH that impacts H(+)-sensitive NMDA receptors (NMDARs). We studied the role of Car14 in this brain structure, in which it is expressed solely on neurons. Current-clamp responses were recorded from CA1 pyramidal neurons in wild-type (WT) versus Car14 knock-out (KO) mice 2 s before (control) and after (test) a 10 pulse, 100 Hz afferent train. In both WT and KO, the half-width (HW) of the test response, and its number of spikes, were augmented relative to the control. An increase in presynaptic release was not involved, because AMPAR-mediated EPSCs were depressed after a train. The increases in HW and spike number were both greater in the Car14 KO. In 0 Mg(2+) saline with picrotoxin (using a 20 Hz train), the HW measures were still greater in the KO. The Car inhibitor benzolamide (BZ) enhanced the test response HW in the WT but had no effect on the already-prolonged HW in the KO. With intracellular MK-801 [(+)-5-methyl-10,11-dihydro-5H-dibenzo [a,d]-cyclohepten-5,10-imine maleate], the curtailed WT and KO responses were indistinguishable, and BZ caused no change. In contrast, the extracellular alkaline changes evoked by the train were not different between WT and KO, and BZ amplified these alkalinizations similarly. These data suggest that Car14 regulates pH transients in the perisynaptic microenvironment and govern their impact on NMDARs but plays little role in buffering pH shifts in the broader, macroscopic, extracellular space.


Asunto(s)
Anhidrasas Carbónicas/fisiología , Potenciales Postsinápticos Excitadores/fisiología , Receptores de N-Metil-D-Aspartato/fisiología , Animales , Benzodiazepinas/farmacología , Región CA1 Hipocampal/citología , Región CA1 Hipocampal/fisiología , Anhidrasas Carbónicas/efectos de los fármacos , Anhidrasas Carbónicas/genética , Interpretación Estadística de Datos , Fenómenos Electrofisiológicos/efectos de los fármacos , Fenómenos Electrofisiológicos/fisiología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Antagonistas del GABA/farmacología , Moduladores del GABA/farmacología , Hipocampo/fisiología , Concentración de Iones de Hidrógeno , Magnesio/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microelectrodos , Plasticidad Neuronal/efectos de los fármacos , Plasticidad Neuronal/fisiología , Técnicas de Placa-Clamp , Picrotoxina/farmacología , Células Piramidales/efectos de los fármacos , Células Piramidales/fisiología , Receptores de N-Metil-D-Aspartato/efectos de los fármacos , Sinapsis/fisiología
13.
Endocrinology ; 153(1): 362-72, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22109883

RESUMEN

Diabetes mellitus causes cerebral microvasculature deterioration and cognitive decline. The specialized endothelial cells of cerebral microvasculature comprise the blood-brain barrier, and the pericytes (PC) that are in immediate contact with these endothelial cells are vital for blood-brain barrier integrity. In diabetes, increased mitochondrial oxidative stress is implicated as a mechanism for hyperglycemia-induced PC loss as a prerequisite leading to blood-brain barrier disruption. Mitochondrial carbonic anhydrases (CA) regulate the oxidative metabolism of glucose and thus play an important role in the generation of reactive oxygen species and oxidative stress. We hypothesize that the inhibition of mitochondrial CA would reduce mitochondrial oxidative stress, rescue cerebral PC loss caused by diabetes-induced oxidative stress, and preserve blood-brain barrier integrity. We studied the effects of pharmacological inhibition of mitochondrial CA activity on streptozotocin-diabetes-induced oxidative stress and PC loss in the mouse brain. At 3 wk of diabetes, there was significant oxidative stress; the levels of reduced glutathione were lower and those of 3-nitrotyrosine, 4-hydroxy-2-trans-nonenal, and superoxide dismutase were higher. Treatment of diabetic mice with topiramate, a potent mitochondrial CA inhibitor, prevented the oxidative stress caused by 3 wk of diabetes. A significant decline in cerebral PC numbers, at 12 wk of diabetes, was also rescued by topiramate treatment. These results provide the first evidence that inhibition of mitochondrial CA activity reduces diabetes-induced oxidative stress in the mouse brain and rescues cerebral PC dropout. Thus, mitochondrial CA may provide a new therapeutic target for oxidative stress related illnesses of the central nervous system.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Inhibidores de Anhidrasa Carbónica/farmacología , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Fructosa/análogos & derivados , Hiperglucemia/tratamiento farmacológico , Hiperglucemia/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Anhidrasa Carbónica V/antagonistas & inhibidores , Anhidrasa Carbónica V/deficiencia , Anhidrasa Carbónica V/genética , Células Cultivadas , Diabetes Mellitus Experimental/patología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Fructosa/farmacología , Hiperglucemia/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Estrés Oxidativo/efectos de los fármacos , Pericitos/efectos de los fármacos , Pericitos/patología , Topiramato
14.
Proc Natl Acad Sci U S A ; 107(14): 6448-52, 2010 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-20308551

RESUMEN

Mutations in the human carbonic anhydrase IV (hCAIV) have been associated with retinal degeneration in an autosomal-dominant form of retinitis pigmentosa (RP17). Prior in vitro cell culture studies confirmed that all of the RP17-associated hCAIV mutations cause protein misfolding, leading to endoplasmic reticulum (ER) stress-induced apoptosis in cells expressing the mutant proteins. To evaluate the physiological impacts of these folding mutants in other carbonic anhydrase IV-producing tissues, we generated two transgenic mouse lines expressing R219S or R14W hCAIV under control of the endogenous hCAIV promoter. Expression of either of these mutant proteins in kidneys caused progressive renal injury in male transgenic mice as evidenced by an age-dependent increase in the tubule cell apoptosis starting at approximately 20 weeks of age and vacuolization throughout the renal cortex in older mice. Up-regulation of the ER chaperone, BiP, was observed in the cells of the renal cortex of the male transgenic mice, suggesting ER stress as a causal factor for the renal injury. The renal injury inflicted by expression of the folding mutants was markedly enhanced by haploinsufficiency of the ER cochaperone p58(IPK). The transgenic mice expressing the hCAIV folding mutants on a p58(IPK) heterozygous background showed extensive renal tubular apoptosis by approximately 10 weeks of age in both male and female mice. These data indicate that expression of the RP17-associated folding mutants of hCAIV can adversely affect tissues beyond the retina and their in vivo proteotoxicity is sensitive to modulation of the protein folding environment of the ER.


Asunto(s)
Anhidrasa Carbónica IV/metabolismo , Progresión de la Enfermedad , Proteínas del Choque Térmico HSP40/metabolismo , Riñón/enzimología , Riñón/patología , Mutación , Pliegue de Proteína , Animales , Apoptosis , Secuencia de Bases , Anhidrasa Carbónica IV/genética , Retículo Endoplásmico/metabolismo , Femenino , Proteínas del Choque Térmico HSP40/deficiencia , Humanos , Riñón/lesiones , Masculino , Ratones , Ratones Transgénicos , Estrés Fisiológico , Regulación hacia Arriba
15.
Am J Physiol Cell Physiol ; 297(5): C1091-102, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19692653

RESUMEN

Bicarbonate transport and metabolism are key elements of normal cellular function. Two alternate transcripts of anion exchanger 3 (AE3), full-length (AE3fl) and cardiac (AE3c), are expressed in central nervous system (CNS), where AE3 catalyzes electroneutral Cl(-)/HCO(3)(-) exchange across the plasma membrane of neuronal and glial cells of CNS. Anion exchanger isoforms, AE3fl and AE3c, associate with the carbonic anhydrases (CA) CAII and CAIV, forming a HCO(3)(-) transport metabolon, to maximize HCO(3)(-) flux across the plasma membrane. CAXIV, with catalytic domain anchored to the extracellular surface, is also expressed in CNS. Here physical association of AE3 and CAXIV was examined by coimmunoprecipitation experiments, using mouse brain and retinal lysates. CAXIV immunoprecipitated with anti-AE3 antibody, and both AE3 isoforms were immunoprecipitated using anti-CAXIV antibody, indicating CAXIV and AE3 interaction in the CNS. Confocal images revealed colocalization of CAXIV and AE3 in Müller and horizontal cells, in the mouse retina. Cl(-)/HCO(3)(-) exchange activity of AE3fl was investigated in transiently transfected human embryonic kidney 293 cells, using intracellular fluorescence measurements of BCECF, to monitor intracellular pH. CAXIV increased the rate of AE3fl-mediated HCO(3)(-) transport by up to 120%, which was suppressed by the CA inhibitor acetazolamide. Association of AE3 and CAXIV may represent a mechanism to enhance disposal of waste CO(2) and to balance pH in excitable tissues.


Asunto(s)
Antiportadores/metabolismo , Bicarbonatos/metabolismo , Anhidrasas Carbónicas/metabolismo , Homeostasis/fisiología , Neuronas/metabolismo , Animales , Encéfalo/metabolismo , Dióxido de Carbono/metabolismo , Línea Celular , Humanos , Concentración de Iones de Hidrógeno , Inmunohistoquímica , Inmunoprecipitación , Ratones , Ratones Noqueados , Microscopía Confocal
16.
Brain Behav Immun ; 23(4): 507-17, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19486646

RESUMEN

Alzheimer's disease (AD) brains are characterized by accumulation of amyloid beta protein (Abeta) and neuroinflammation. Increased blood-to-brain influx and decreased brain-to-blood efflux across the blood-brain barrier (BBB) have been proposed as mechanisms for Abeta accumulation. Epidemiological studies suggest that the nonsteroidal anti-inflammatory drug (NSAID) indomethacin slows the progression of AD. We hypothesized that inflammation alters BBB handling of Abeta. Mice treated with lipopolysaccharide (LPS) had increased brain influx and decreased brain efflux of Abeta, recapitulating the findings in AD. Neither influx nor efflux was mediated by LPS acting directly on BBB cells. Increased influx was mediated by a blood-borne factor, indomethacin-independent, blocked by the triglyceride triolein, and not related to expression of the blood-to-brain transporter of Abeta, RAGE. Serum levels of IL-6, IL-10, IL-13, and MCP-1 mirrored changes in Abeta influx. Decreased efflux was blocked by indomethacin and accompanied by decreased protein expression of the brain-to-blood transporter of Abeta, LRP-1. LPS paradoxically increased expression of neuronal LRP-1, a major source of Abeta. Thus, inflammation potentially increases brain levels of Abeta by three mechanisms: increased influx, decreased efflux, and increased neuronal production.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Barrera Hematoencefálica/efectos de los fármacos , Inflamación/metabolismo , Transporte de Proteínas/efectos de los fármacos , Enfermedad de Alzheimer/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Western Blotting , Encéfalo/metabolismo , Inhibidores de la Ciclooxigenasa/farmacología , Citocinas/metabolismo , Progresión de la Enfermedad , Relación Dosis-Respuesta a Droga , Indometacina/farmacología , Lipopolisacáridos/administración & dosificación , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad , Masculino , Ratones , Estrés Oxidativo/efectos de los fármacos , Transporte de Proteínas/fisiología , Receptor para Productos Finales de Glicación Avanzada , Receptores Inmunológicos/metabolismo , Receptores de LDL/metabolismo , Trioleína/farmacología , Proteínas Supresoras de Tumor/metabolismo
17.
J Gerontol A Biol Sci Med Sci ; 64(10): 1025-30, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19531769

RESUMEN

Amyloid beta protein (Abeta) in Alzheimer's disease induces oxidative stress through several mechanisms, including stimulation of nitric oxide synthase (NOS) activity. We examined NOS activity and expression in the senescence-accelerated mouse P8 (SAMP8) line. The SAMP8 strain develops with aging cognitive impairments, increases in Abeta, and oxidative stress, all reversed by amyloid precursor protein antisense or Abeta antibody treatment. We found here that hippocampal NOS activity in 12-month-old SAMP8 mice was nearly double that of 2-month-old SAMP8 or CD-1 mice, but with no change in NOS isoenzyme mRNA and protein levels. Antisense or antibody treatment further increased NOS activity in aged SAMP8 mice. Antisense treatment increased inducible NOS (iNOS) mRNA levels, decreased neuronal NOS mRNA and protein levels, but did not affect endothelial NOS (eNOS) or iNOS protein or eNOS mRNA levels. These results suggest a complex relation between Abeta and NOS in the SAMP8 that is largely mediated through posttranslational mechanisms.


Asunto(s)
Enfermedad de Alzheimer/enzimología , Péptidos beta-Amiloides/inmunología , Anticuerpos/uso terapéutico , Elementos sin Sentido (Genética)/uso terapéutico , Factores Inmunológicos/uso terapéutico , Óxido Nítrico Sintasa/metabolismo , Factores de Edad , Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/terapia , Animales , Modelos Animales de Enfermedad , Hipocampo/enzimología , Isoenzimas/genética , Isoenzimas/metabolismo , Masculino , Ratones , Óxido Nítrico Sintasa/genética , ARN Mensajero/metabolismo
18.
Proc Natl Acad Sci U S A ; 106(9): 3437-42, 2009 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-19211803

RESUMEN

Missense mutations in the carbonic anhydrase IV (CA IV) gene have been identified in patients with an autosomal dominant form of retinitis pigmentosa (RP17). We used two transient expression systems to investigate the molecular mechanism by which the newly identified CA IV mutations, R69H and R219S, contribute to retinal pathogenesis. Although the R219S mutation drastically reduced the activity of the enzyme, the R69H mutation had a minimal effect, suggesting that loss of CA activity is not the molecular basis for their pathogenesis. Defective processing was apparent for both mutant proteins. Cell surface-labeling techniques showed that the R69H and R219S mutations both impaired the trafficking of CA IV to the cell surface, resulting in their abnormal intracellular retention. Expression of both CA IV mutants induced elevated levels of the endoplasmic reticulum (ER) stress markers, BiP and CHOP, and led to cell death by apoptosis. They also had a dominant-negative effect on the secretory function of the ER. These properties are similar to those of R14W CA IV, the signal sequence variant found in the original patients with RP17. These findings suggest that toxic gain of function involving ER stress-induced apoptosis is the common mechanism for pathogenesis of this autosomal-dominant disease. Apoptosis induced by the CA IV mutants could be prevented, at least partially, by treating the cells with dorzolamide, a CA inhibitor. Thus, the use of a CA inhibitor as a chemical chaperone to reduce ER stress may delay or prevent the onset of blindness in RP17.


Asunto(s)
Apoptosis , Anhidrasa Carbónica IV/metabolismo , Retinitis Pigmentosa/enzimología , Retinitis Pigmentosa/patología , Animales , Apoptosis/efectos de los fármacos , Anhidrasa Carbónica IV/antagonistas & inhibidores , Anhidrasa Carbónica IV/genética , Línea Celular , Membrana Celular/enzimología , Chlorocebus aethiops , Retículo Endoplásmico/metabolismo , Inhibidores Enzimáticos/farmacología , Regulación Enzimológica de la Expresión Génica , Humanos , Mutación/genética , Transporte de Proteínas , Retinitis Pigmentosa/genética
19.
Proc Natl Acad Sci U S A ; 105(24): 8375-80, 2008 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-18544647

RESUMEN

Glycosaminoglycan storage begins in prenatal life in patients with mucopolysaccharidosis (MPS). In fact, prenatal hydrops is a common manifestation of MPS VII because of beta-glucuronidase (GUS) deficiency. One way to address prenatal storage might be to deliver the missing enzyme across the placenta into the fetal circulation. Maternal IgG is transported across the placenta by the neonatal Fc receptor (FcRn), which recognizes the Fc domain of IgG and mediates transcytosis from maternal to fetal circulation. We hypothesized that we could exploit this process to deliver corrective enzyme to the fetus. To test this hypothesis, the C-terminal fusion protein, GUS-Fc, was compared with native, untagged, recombinant GUS for clearance from the maternal circulation, delivery to the fetus, and reduction of lysosomal storage in offspring of MPS VII mice. We observed that GUS-Fc, infused into pregnant mothers on embryonic days 17 and 18, was transported across the placenta. Similarly infused untagged GUS was not delivered to the fetus. GUS-Fc plasma enzyme activity in newborn MPS VII mice was 1,000 times that seen after administration of untagged GUS and approximately 100 times that of untreated WT newborns. Reduced lysosomal storage in heart valves, liver, and spleen provided evidence that in utero enzyme replacement therapy with GUS-Fc targeted sites of storage in the MPS VII fetus. We hypothesize that this noninvasive approach could deliver the missing lysosomal enzyme to a fetus with any lysosomal storage disease. It might also provide a method for inducing immune tolerance to the missing enzyme or another foreign protein.


Asunto(s)
Glucuronidasa/uso terapéutico , Mucopolisacaridosis VII/prevención & control , Placenta/metabolismo , Receptores Fc/uso terapéutico , Proteínas Recombinantes de Fusión/uso terapéutico , Útero , Animales , Femenino , Glucuronidasa/administración & dosificación , Glucuronidasa/farmacocinética , Glicosaminoglicanos/antagonistas & inhibidores , Infusiones Parenterales , Lisosomas/metabolismo , Ratones , Embarazo , Receptores Fc/administración & dosificación , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/farmacocinética , Distribución Tisular
20.
Am J Physiol Cell Physiol ; 294(2): C402-12, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18003750

RESUMEN

The subcellular localization of carbonic anhydrase (CA) IV and CA IX in mouse skeletal muscle fibers has been studied immunohistochemically by confocal laser scanning microscopy. CA IV has been found to be located on the plasma membrane as well as on the sarcoplasmic reticulum (SR) membrane. CA IX is not localized in the plasma membrane but in the region of the t-tubular (TT)/terminal SR membrane. CA IV contributes 20% and CA IX 60% to the total CA activity of SR membrane vesicles isolated from mouse skeletal muscles. Our aim was to examine whether SR CA IV and TT/SR CA IX affect muscle contraction. Isolated fiber bundles of fast-twitch extensor digitorum longus and slow-twitch soleus muscle from mouse were investigated for isometric twitch and tetanic contractions and by a fatigue test. The muscle functions of CA IV knockout (KO) fibers and of CA IX KO fibers do not differ from the function of wild-type (WT) fibers. Muscle function of CA IV/XIV double KO mice unexpectedly shows a decrease in rise and relaxation time and in force of single twitches. In contrast, the CA inhibitor dorzolamide, whether applied to WT or to double KO muscle fibers, leads to a significant increase in rise time and force of twitches. It is concluded that the function of mouse skeletal muscle fibers expressing three membrane-associated CAs, IV, IX, and XIV, is not affected by the lack of one isoform but is possibly affected by the lack of all three CAs, as indicated by the inhibition studies.


Asunto(s)
Anhidrasa Carbónica IV/metabolismo , Anhidrasas Carbónicas/metabolismo , Músculo Esquelético/enzimología , Animales , Anhidrasa Carbónica IV/genética , Anhidrasa Carbónica IX , Anhidrasas Carbónicas/genética , Inhibidores Enzimáticos/farmacología , Regulación Enzimológica de la Expresión Génica/genética , Inmunohistoquímica , Ratones , Ratones Noqueados , Microscopía Confocal , Contracción Muscular/genética , Fibras Musculares de Contracción Rápida/enzimología , Fibras Musculares de Contracción Rápida/ultraestructura , Fibras Musculares de Contracción Lenta/enzimología , Fibras Musculares de Contracción Lenta/ultraestructura , Músculo Esquelético/ultraestructura , Sarcolema/enzimología , Sarcolema/ultraestructura , Retículo Sarcoplasmático/enzimología , Retículo Sarcoplasmático/ultraestructura , Sulfonamidas/farmacología , Tiofenos/farmacología , Factores de Tiempo , Vesículas Transportadoras/enzimología , Vesículas Transportadoras/ultraestructura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...