Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Proc Natl Acad Sci U S A ; 118(12)2021 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-33737395

RESUMEN

Acute kidney injury is highly prevalent and associated with high morbidity and mortality, and there are no approved drugs for its prevention and treatment. Vagus nerve stimulation (VNS) alleviates inflammatory diseases including kidney disease; however, neural circuits involved in VNS-induced tissue protection remain poorly understood. The vagus nerve, a heterogeneous group of neural fibers, innervates numerous organs. VNS broadly stimulates these fibers without specificity. We used optogenetics to selectively stimulate vagus efferent or afferent fibers. Anterograde efferent fiber stimulation or anterograde (centripetal) sensory afferent fiber stimulation both conferred kidney protection from ischemia-reperfusion injury. We identified the C1 neurons-sympathetic nervous system-splenic nerve-spleen-kidney axis as the downstream pathway of vagus afferent fiber stimulation. Our study provides a map of the neural circuits important for kidney protection induced by VNS, which is critical for the safe and effective clinical application of VNS for protection from acute kidney injury.


Asunto(s)
Lesión Renal Aguda/etiología , Susceptibilidad a Enfermedades , Neuroinmunomodulación , Bazo/inmunología , Bazo/inervación , Estimulación del Nervio Vago , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Animales , Ratones , Neuronas , Sistema Nervioso Simpático/fisiología
2.
Kidney Int ; 100(3): 613-620, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34224760

RESUMEN

Microcirculatory changes and oxidative stress have long been associated with acute kidney injury. Despite substantial progress made by two-photon microscopy of microvascular responses to acute kidney injury in rodent models, little is known about the underlying changes in blood oxygen delivery and tissue oxygen metabolism. To fill this gap, we developed a label-free kidney imaging technique based on photoacoustic microscopy, which enables simultaneous quantification of hemoglobin concentration, oxygen saturation of hemoglobin, and blood flow in peritubular capillaries in vivo. Based on these microvascular parameters, microregional oxygen metabolism was quantified. We demonstrated the utility of this technique by studying kidney hemodynamic and oxygen-metabolic responses to acute kidney injury in mice subject to lipopolysaccharide-induced sepsis. Dynamic photoacoustic microscopy of the peritubular capillary function and tissue oxygen metabolism revealed that sepsis induced an acute and significant reduction in peritubular capillary oxygen saturation of hemoglobin, concomitant with a marked reduction in kidney ATP levels and contrasted with nominal changes in peritubular capillary flow and plasma creatinine. Thus, our technique opens new opportunities to study microvascular and metabolic dysfunction in acute and chronic kidney diseases.


Asunto(s)
Capilares , Microscopía , Animales , Riñón , Ratones , Microcirculación , Oxígeno
3.
Kidney Int ; 95(3): 563-576, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30670317

RESUMEN

The cholinergic anti-inflammatory pathway (CAP) links the nervous and immune systems and modulates innate and adaptive immunity. Activation of the CAP by vagus nerve stimulation exerts protective effects in a wide variety of clinical disorders including rheumatoid arthritis and Crohn's disease, and in murine models of acute kidney injury including ischemia/reperfusion injury (IRI). The canonical CAP pathway involves activation of splenic alpha7-nicotinic acetylcholine receptor (α7nAChR)-positive macrophages by splenic ß2-adrenergic receptor-positive CD4+ T cells. Here we demonstrate that ultrasound or vagus nerve stimulation also activated α7nAChR-positive peritoneal macrophages, and that adoptive transfer of these activated peritoneal macrophages reduced IRI in recipient mice. The protective effect required α7nAChR, and did not occur in splenectomized mice or in mice lacking T and B cells, suggesting a bidirectional interaction between α7nAChR-positive peritoneal macrophages and other immune cells including ß2-adrenergic receptor-positive CD4+ T cells. We also found that expression of hairy and enhancer of split-1 (Hes1), a basic helix-loop-helix DNA-binding protein, is induced in peritoneal macrophages by ultrasound or vagus nerve stimulation. Adoptive transfer of Hes1-overexpressing peritoneal macrophages reduced kidney IRI. Our data suggest that Hes1 is downstream of α7nAChR and is important to fully activate the CAP. Taken together, these results suggest that peritoneal macrophages play a previously unrecognized role in mediating the protective effect of CAP activation in kidney injury, and that Hes1 is a new candidate pharmacological target to activate the CAP.


Asunto(s)
Lesión Renal Aguda/inmunología , Macrófagos Peritoneales/inmunología , Daño por Reperfusión/inmunología , Factor de Transcripción HES-1/metabolismo , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo , Lesión Renal Aguda/patología , Lesión Renal Aguda/terapia , Traslado Adoptivo , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/efectos de la radiación , Linfocitos T CD4-Positivos/trasplante , Modelos Animales de Enfermedad , Técnicas de Silenciamiento del Gen , Humanos , Activación de Macrófagos , Macrófagos Peritoneales/metabolismo , Macrófagos Peritoneales/trasplante , Masculino , Ratones , Neuroinmunomodulación/efectos de la radiación , Células RAW 264.7 , Daño por Reperfusión/patología , Daño por Reperfusión/terapia , Factor de Transcripción HES-1/genética , Factor de Transcripción HES-1/inmunología , Terapia por Ultrasonido , Regulación hacia Arriba/efectos de la radiación , Estimulación del Nervio Vago , Receptor Nicotínico de Acetilcolina alfa 7/inmunología
4.
Proc Natl Acad Sci U S A ; 111(47): 16889-94, 2014 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-25385587

RESUMEN

Although dominant gain-of-function triplet repeat expansions in the Huntingtin (HTT) gene are the underlying cause of Huntington disease (HD), understanding the normal functions of nonmutant HTT protein has remained a challenge. We report here findings that suggest that HTT plays a significant role in selective autophagy. Loss of HTT function in Drosophila disrupts starvation-induced autophagy in larvae and conditional knockout of HTT in the mouse CNS causes characteristic cellular hallmarks of disrupted autophagy, including an accumulation of striatal p62/SQSTM1 over time. We observe that specific domains of HTT have structural similarities to yeast Atg proteins that function in selective autophagy, and in particular that the C-terminal domain of HTT shares structural similarity to yeast Atg11, an autophagic scaffold protein. To explore possible functional similarity between HTT and Atg11, we investigated whether the C-terminal domain of HTT interacts with mammalian counterparts of yeast Atg11-interacting proteins. Strikingly, this domain of HTT coimmunoprecipitates with several key Atg11 interactors, including the Atg1/Unc-51-like autophagy activating kinase 1 kinase complex, autophagic receptor proteins, and mammalian Atg8 homologs. Mutation of a phylogenetically conserved WXXL domain in a C-terminal HTT fragment reduces coprecipitation with mammalian Atg8 homolog GABARAPL1, suggesting a direct interaction. Collectively, these data support a possible central role for HTT as an Atg11-like scaffold protein. These findings have relevance to both mechanisms of disease pathogenesis and to therapeutic intervention strategies that reduce levels of both mutant and normal HTT.


Asunto(s)
Autofagia , Proteínas Asociadas a Microtúbulos/fisiología , Animales , Animales Modificados Genéticamente , Drosophila , Proteínas de Drosophila , Proteína Huntingtina , Ratones , Proteínas Asociadas a Microtúbulos/genética
5.
Nature ; 467(7313): 333-7, 2010 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-20844538

RESUMEN

Apoptosis and the subsequent clearance of dying cells occurs throughout development and adult life in many tissues. Failure to promptly clear apoptotic cells has been linked to many diseases. ELMO1 is an evolutionarily conserved cytoplasmic engulfment protein that functions downstream of the phosphatidylserine receptor BAI1, and, along with DOCK1 and the GTPase RAC1, promotes internalization of the dying cells. Here we report the generation of ELMO1-deficient mice, which we found to be unexpectedly viable and grossly normal. However, they had a striking testicular pathology, with disrupted seminiferous epithelium, multinucleated giant cells, uncleared apoptotic germ cells and decreased sperm output. Subsequent in vitro and in vivo analyses revealed a crucial role for ELMO1 in the phagocytic clearance of apoptotic germ cells by Sertoli cells lining the seminiferous epithelium. The engulfment receptor BAI1 and RAC1 (upstream and downstream of ELMO1, respectively) were also important for Sertoli-cell-mediated engulfment. Collectively, these findings uncover a selective requirement for ELMO1 in Sertoli-cell-mediated removal of apoptotic germ cells and make a compelling case for a relationship between engulfment and tissue homeostasis in vivo.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Apoptosis , Fagocitosis/fisiología , Células de Sertoli/citología , Células de Sertoli/metabolismo , Espermatozoides/citología , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Angiogénicas/metabolismo , Animales , Línea Celular , Homeostasis , Masculino , Ratones , Ratones Endogámicos C57BL , Neuropéptidos/metabolismo , Fosfatidilserinas/metabolismo , Epitelio Seminífero/citología , Epitelio Seminífero/patología , Células de Sertoli/patología , Transducción de Señal , Espermatozoides/patología , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rac1
6.
J Biol Chem ; 287(26): 21599-614, 2012 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-22556411

RESUMEN

Huntington disease is a neurodegenerative disorder caused by a CAG repeat amplification in the gene huntingtin (HTT) that is reflected by a polyglutamine expansion in the Htt protein. Nearly 20 years of research have uncovered roles for Htt in a wide range of cellular processes, and many of these discoveries stemmed from the identification of Htt-interacting proteins. However, no study has employed an impartial and comprehensive strategy to identify proteins that differentially associate with full-length wild-type and mutant Htt in brain tissue, the most relevant sample source to the disease condition. We analyzed Htt affinity-purified complexes from wild-type and HTT mutant juvenile mouse brain from two different biochemical fractions by tandem mass spectrometry. We compared variations in protein spectral counts relative to Htt to identify those proteins that are the most significantly contrasted between wild-type and mutant Htt purifications. Previously unreported Htt interactions with Myo5a, Prkra (PACT), Gnb2l1 (RACK1), Rps6, and Syt2 were confirmed by Western blot analysis. Gene Ontology analysis of these and other Htt-associated proteins revealed a statistically significant enrichment for proteins involved in translation among other categories. Furthermore, Htt co-sedimentation with polysomes in cytoplasmic mouse brain extracts is dependent upon the presence of intact ribosomes. Finally, wild-type or mutant Htt overexpression inhibits cap-dependent translation of a reporter mRNA in an in vitro system. Cumulatively, these data support a new role for Htt in translation and provide impetus for further study into the link between protein synthesis and Huntington disease pathogenesis.


Asunto(s)
Encéfalo/metabolismo , Enfermedad de Huntington/metabolismo , Mutación , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteómica/métodos , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Animales , Modelos Animales de Enfermedad , Silenciador del Gen , Células HeLa , Humanos , Proteína Huntingtina , Ratones , Modelos Estadísticos , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , Biosíntesis de Proteínas , Proteoma , ARN/metabolismo , Espectrometría de Masas en Tándem/métodos
7.
PLoS Genet ; 6(2): e1000838, 2010 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-20140187

RESUMEN

Expansion of a stretch of polyglutamine in huntingtin (htt), the protein product of the IT15 gene, causes Huntington's disease (HD). Previous investigations into the role of the polyglutamine stretch (polyQ) in htt function have suggested that its length may modulate a normal htt function involved in regulating energy homeostasis. Here we show that expression of full-length htt lacking its polyglutamine stretch (DeltaQ-htt) in a knockin mouse model for HD (Hdh(140Q/DeltaQ)), reduces significantly neuropil mutant htt aggregates, ameliorates motor/behavioral deficits, and extends lifespan in comparison to the HD model mice (Hdh(140Q/+)). The rescue of HD model phenotypes is accompanied by the normalization of lipofuscin levels in the brain and an increase in the steady-state levels of the mammalian autophagy marker microtubule-associate protein 1 light chain 3-II (LC3-II). We also find that DeltaQ-htt expression in vitro increases autophagosome synthesis and stimulates the Atg5-dependent clearance of truncated N-terminal htt aggregates. DeltaQ-htt's effect on autophagy most likely represents a gain-of-function, as overexpression of full-length wild-type htt in vitro does not increase autophagosome synthesis. Moreover, Hdh(DeltaQ/DeltaQ) mice live significantly longer than wild-type mice, suggesting that autophagy upregulation may be beneficial both in diseases caused by toxic intracellular aggregate-prone proteins and also as a lifespan extender in normal mammals.


Asunto(s)
Autofagia , Longevidad , Proteínas del Tejido Nervioso/genética , Neuronas/patología , Proteínas Nucleares/genética , Péptidos/genética , Eliminación de Secuencia/genética , Animales , Proteína 5 Relacionada con la Autofagia , Conducta Animal , Línea Celular , Modelos Animales de Enfermedad , Humanos , Proteína Huntingtina , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lipofuscina/metabolismo , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Actividad Motora , Neostriado/metabolismo , Neostriado/patología , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Neurópilo/metabolismo , Neurópilo/patología , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Fagosomas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Estructura Cuaternaria de Proteína , Transducción de Señal , Serina-Treonina Quinasas TOR
8.
J Exp Med ; 220(12)2023 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-37733279

RESUMEN

A key process in central sensory circuit development involves activity-dependent pruning of exuberant terminals. Here, we studied gustatory terminal field maturation in the postnatal mouse nucleus of the solitary tract (NST) during normal development and in mice where their mothers were fed a low NaCl diet for a limited period soon after conception. Pruning of terminal fields of gustatory nerves in controls involved the complement system and is likely driven by NaCl-elicited taste activity. In contrast, offspring of mothers with an early dietary manipulation failed to prune gustatory terminal fields even though peripheral taste activity developed normally. The ability to prune in these mice was rescued by activating myeloid cells postnatally, and conversely, pruning was arrested in controls with the loss of myeloid cell function. The altered pruning and myeloid cell function appear to be programmed before the peripheral gustatory system is assembled and corresponds to the embryonic period when microglia progenitors derived from the yolk sac migrate to and colonize the brain.


Asunto(s)
Microglía , Cloruro de Sodio , Animales , Ratones , Embarazo , Femenino , Gusto , Dieta , Encéfalo
9.
Front Med (Lausanne) ; 9: 931293, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35966871

RESUMEN

Adenosine triphosphate (ATP) released from injured or dying cells is a potent pro-inflammatory "danger" signal. Alkaline phosphatase (AP), an endogenous enzyme that de-phosphorylates extracellular ATP, likely plays an anti-inflammatory role in immune responses. We hypothesized that ilofotase alfa, a human recombinant AP, protects kidneys from ischemia-reperfusion injury (IRI), a model of acute kidney injury (AKI), by metabolizing extracellular ATP to adenosine, which is known to activate adenosine receptors. Ilofotase alfa (iv) with or without ZM241,385 (sc), a selective adenosine A2A receptor (A2AR) antagonist, was administered 1 h before bilateral IRI in WT, A2AR KO (Adora2a-/- ) or CD73-/- mice. In additional studies recombinant alkaline phosphatase was given after IRI. In an AKI-on-chronic kidney disease (CKD) ischemic rat model, ilofotase alfa was given after the three instances of IRI and rats were followed for 56 days. Ilofotase alfa in a dose dependent manner decreased IRI in WT mice, an effect prevented by ZM241,385 and partially prevented in Adora2a-/- mice. Enzymatically inactive ilofotase alfa was not protective. Ilofotase alfa rescued CD73-/- mice, which lack a 5'-ectonucleotidase that dephosphorylates AMP to adenosine; ZM241,385 inhibited that protection. In both rats and mice ilofotase alfa ameliorated IRI when administered after injury, thus providing relevance for therapeutic dosing of ilofotase alfa following established AKI. In an AKI-on-CKD ischemic rat model, ilofotase alfa given after the third instance of IRI reduced injury. These results suggest that ilofotase alfa promotes production of adenosine from liberated ATP in injured kidney tissue, thereby amplifying endogenous mechanisms that can reverse tissue injury, in part through A2AR-and non-A2AR-dependent signaling pathways.

10.
Sci Transl Med ; 14(658): eabj2681, 2022 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-35976996

RESUMEN

Chronic kidney disease (CKD), characterized by sustained inflammation and progressive fibrosis, is highly prevalent and can eventually progress to end-stage kidney disease. However, current treatments to slow CKD progression are limited. Sphingosine 1-phosphate (S1P), a product of sphingolipid catabolism, is a pleiotropic mediator involved in many cellular functions, and drugs targeting S1P signaling have previously been studied particularly for autoimmune diseases. The primary mechanism of most of these drugs is functional antagonism of S1P receptor-1 (S1P1) expressed on lymphocytes and the resultant immunosuppressive effect. Here, we documented the role of local S1P signaling in perivascular cells in the progression of kidney fibrosis using primary kidney perivascular cells and several conditional mouse models. S1P was predominantly produced by sphingosine kinase 2 in kidney perivascular cells and exported via spinster homolog 2 (Spns2). It bound to S1P1 expressed in perivascular cells to enhance production of proinflammatory cytokines/chemokines upon injury, leading to immune cell infiltration and subsequent fibrosis. A small-molecule Spns2 inhibitor blocked S1P transport, resulting in suppression of inflammatory signaling in human and mouse kidney perivascular cells in vitro and amelioration of kidney fibrosis in mice. Our study provides insight into the regulation of inflammation and fibrosis by S1P and demonstrates the potential of Spns2 inhibition as a treatment for CKD and potentially other inflammatory and fibrotic diseases that avoids the adverse events associated with systemic modulation of S1P receptors.


Asunto(s)
Inflamación , Insuficiencia Renal Crónica , Animales , Fibrosis , Humanos , Inflamación/metabolismo , Riñón/metabolismo , Lisofosfolípidos , Ratones , Esfingosina/análogos & derivados
11.
Nephron ; 144(12): 621-625, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33147592

RESUMEN

Understanding and measuring parameters responsible for the pathogenesis of sepsis-induced AKI (SI-AKI) is critical in developing therapies. Blood flow to the kidney is heterogeneous, partly due to the existence of dynamic networks of capillaries in various regions, responding differentially to oxygen demand in cortex versus medulla. High energy demand regions, especially the outer medulla, are susceptible to hypoxia and subject to damage during SI-AKI. Proximal tubule epithelial cells in the cortex and the outer medulla can also undergo metabolic reprogramming during SI-AKI to maintain basal physiological status and to avoid potential damage. Current data on the assessment of renal hemodynamics and oxygen metabolism during sepsis is limited. Preclinical and clinical studies show changes in renal hemodynamics associated with SI-AKI, and in clinical settings, interventions to manage renal hemodynamics seem to help improve disease outcomes in some cases. Lack of proper tools to assess temporospatial changes in peritubular blood flow and tissue oxygen metabolism is a barrier to our ability to understand microcirculatory dynamics and oxygen consumption and their role in the pathogenesis of SI-AKI. Current tools to assess renal oxygenation are limited in their usability as these cannot perform continuous simultaneous measurement of renal hemodynamics and oxygen metabolism. Multi-parametric photo-acoustic microscopy (PAM) is a new tool that can measure real-time changes in microhemodynamics and oxygen metabolism. Use of multi-parametric PAM in combination with advanced intravital imaging techniques has the potential to understand the contribution of microhemodynamic and tissue oxygenation alterations to SI-AKI.


Asunto(s)
Lesión Renal Aguda/metabolismo , Capilares/metabolismo , Consumo de Oxígeno , Sepsis/metabolismo , Lesión Renal Aguda/etiología , Animales , Modelos Animales de Enfermedad , Humanos , Hierro/metabolismo , Sepsis/complicaciones
12.
Brain Res ; 1054(2): 143-51, 2005 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-16081051

RESUMEN

A brain slice model was used to test the hypothesis that preconditioning with isoflurane, a commonly used volatile anesthetic in clinical practice, reduces neuronal injury caused by overstimulation of glutamate receptors. Glutamate receptors were stimulated by various concentrations of glutamate for 20 min, N-methyl-d-aspartate (NMDA) for 15 min or alpha-amino-3-hydroxy-5-methyl-4-isoxazol propionic acid (AMPA) for 15 min. Morphology of Purkinje neurons in the cerebellar slices of adult male Sprague-Dawley rats was evaluated 5 h after the agonist stimulation. Glutamate, NMDA and AMPA induced a dose-dependent decrease in the percentage of morphologically normal Purkinje neurons. The concentration to induce the maximal neurotoxic effect was 300 microM for glutamate, 300 microM for NMDA and 30 microM for AMPA. Isoflurane preconditioning (2% isoflurane for 30 min and then a 15-min rest period before the agonist stimulation) significantly reduced the neurotoxicity induced by 300 microM glutamate, 300 microM NMDA or 30 microM AMPA. Isoflurane preconditioning-induced protection against glutamate neurotoxicity was abolished by two protein kinase C (PKC) inhibitors, calphostin C (0.5 microM) and chelerythrine (5 microM), or a nitric oxide synthase (NOS) inhibitor, l-nitro(G)-arginine methyl ester (l-NAME, 1.5 mM), but was not affected by an adenosine A1 receptor inhibitor, 8-cyclopentyl-1,3-dipropylxanthine (DPCPX, 300 nM), or a Gi protein inhibitor, pertussis toxin (PTX, 200 ng/ml). Isoflurane preconditioning-induced protection against NMDA neurotoxicity was also abolished by calphostin C, chelerythrine or l-NAME. Thus, isoflurane preconditioning reduced glutamate receptor overstimulation-induced neuronal injury/death. This neuroprotection may be PKC- and NOS-dependent.


Asunto(s)
Cerebelo/citología , Isoflurano/farmacología , Células de Purkinje/efectos de los fármacos , Receptores de Glutamato/metabolismo , 6-Ciano 7-nitroquinoxalina 2,3-diona/farmacología , Anestésicos por Inhalación/farmacología , Animales , Supervivencia Celular/efectos de los fármacos , Maleato de Dizocilpina/farmacología , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Inhibidores Enzimáticos/farmacología , Agonistas de Aminoácidos Excitadores/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Ácido Glutámico/toxicidad , Técnicas In Vitro , Masculino , N-Metilaspartato/farmacología , Toxina del Pertussis/farmacología , Células de Purkinje/fisiología , Ratas , Ratas Sprague-Dawley , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/farmacología
13.
Mol Brain ; 5: 28, 2012 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-22892315

RESUMEN

BACKGROUND: Huntington's disease (HD) is an autosomal dominant neurodegenerative disease that is caused by the expansion of a polyglutamine (polyQ) stretch within Huntingtin (htt), the protein product of the HD gene. Although studies in vitro have suggested that the mutant htt can act in a potentially dominant negative fashion by sequestering wild-type htt into insoluble protein aggregates, the role of the length of the normal htt polyQ stretch, and the adjacent proline-rich region (PRR) in modulating HD mouse model pathogenesis is currently unknown. RESULTS: We describe the generation and characterization of a series of knock-in HD mouse models that express versions of the mouse HD gene (Hdh) encoding N-terminal hemaglutinin (HA) or 3xFlag epitope tagged full-length htt with different polyQ lengths (HA7Q-, 3xFlag7Q-, 3xFlag20Q-, and 3xFlag140Q-htt) and substitution of the adjacent mouse PRR with the human PRR (3xFlag20Q- and 3xFlag140Q-htt). Using co-immunoprecipitation and immunohistochemistry analyses, we detect no significant interaction between soluble full-length normal 7Q- htt and mutant (140Q) htt, but we do observe N-terminal fragments of epitope-tagged normal htt in mutant htt aggregates. When the sequences encoding normal mouse htt's polyQ stretch and PRR are replaced with non-pathogenic human sequence in mice also expressing 140Q-htt, aggregation foci within the striatum, and the mean size of htt inclusions are increased, along with an increase in striatal lipofuscin and gliosis. CONCLUSION: In mice, soluble full-length normal and mutant htt are predominantly monomeric. In heterozygous knock-in HD mouse models, substituting the normal mouse polyQ and PRR with normal human sequence can exacerbate some neuropathological phenotypes.


Asunto(s)
Alelos , Epítopos/química , Enfermedad de Huntington/metabolismo , Proteínas Mutantes/metabolismo , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Péptidos/química , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Encéfalo/metabolismo , Encéfalo/patología , Distribución de Chi-Cuadrado , Cruzamientos Genéticos , Modelos Animales de Enfermedad , Exones/genética , Femenino , Gliosis/metabolismo , Gliosis/patología , Hemicigoto , Heterocigoto , Humanos , Proteína Huntingtina , Enfermedad de Huntington/patología , Lipofuscina/metabolismo , Masculino , Ratones , Datos de Secuencia Molecular , Proteínas Mutantes/química , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Unión Proteica , Expansión de Repetición de Trinucleótido/genética
14.
Cell Tissue Res ; 328(2): 411-9, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17265069

RESUMEN

Ischemia-reperfusion (IR) of the testis results in germ-cell-specific apoptosis (GCA) and a reduction in daily sperm production. This has been correlated with and is dependent upon neutrophil recruitment to the testis. In a rat model of testicular IR, this has also been correlated with an increase in reactive oxygen species (ROS). We have investigated ROS in the mouse testis after IR and determined whether the observed GCA is mediated via a mitochondrial caspase-9-dependent pathway involving the upstream mediators caspase 2 and BAX. Mice were subjected to a 2-h period of testicular ischemia followed by reperfusion. An accumulation of 8-isoprostane, a marker of oxidative stress, occurred 4 h after reperfusion. Activation of a mitochondrial dependent pathway to GCA after testicular IR was determined based on the observations that both BAX and caspase 2 translocated to the mitochondria, and that an increase occurred in cytoplasmic cytochrome c. Moreover, microinfusion of a specific caspase 9 inhibitor significantly reduced active caspase 3 after testicular IR and the number of apoptotic germ cells. These results suggest that oxidative stress products accumulate in the testis following IR and demonstrate that the observed GCA is stimulated through a mitochondrial caspase-9-dependent pathway. The identification of the germ-cell apoptotic pathway induced after testicular IR, including the key players in the pathway subsequent to ROS (BAX, caspase 9, and caspase 2), aids our understanding of IR injury in the testis and provides a wider background for the development of therapeutic interventions to rescue testis function.


Asunto(s)
Apoptosis , Caspasa 2/metabolismo , Caspasa 9/metabolismo , Estrés Oxidativo , Espermatozoides/citología , Espermatozoides/enzimología , Proteína X Asociada a bcl-2/metabolismo , Animales , Caspasa 3/metabolismo , Citocromos c/metabolismo , Dinoprost/análogos & derivados , Dinoprost/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Daño por Reperfusión , Torsión del Cordón Espermático/inducido químicamente , Testículo/irrigación sanguínea , Testículo/citología
15.
J Biol Chem ; 281(30): 21250-21255, 2006 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-16723357

RESUMEN

Glutamate transporters (excitatory amino acid transporters, EAAT) play an important role in maintaining extracellular glutamate homeostasis and regulating glutamate neurotransmission. However, very few studies have investigated the regulation of EAAT expression. A binding sequence for the regulatory factor X1 (RFX1) exists in the promoter region of the gene encoding for EAAT3, a neuronal EAAT, but not in the promoter regions of the genes encoding for EAAT1 and EAAT2, two glial EAATs. RFX proteins are transcription factors binding to X-boxes of DNA sequences. Although RFX proteins are necessary for the normal function of sensory neurons in Caenorhabditis elegans, their roles in the mammalian brain are not known. We showed that RFX1 increased EAAT3 expression and activity in C6 glioma cells. RFX1 binding complexes were found in the nuclear extracts of C6 cells. The activity of EAAT3 promoter as measured by luciferase reporter activity was increased by RFX1 in C6 cells and the neuron-like SH-SY5Y cells. However, RFX1 did not change the expression of EAAT2 proteins in the NRK52E cells. RFX1 proteins were expressed in the neurons of rat brain. A high expression level of RFX1 proteins was found in the neurons of cerebral cortex and Purkinje cells. Knockdown of the RFX1 expression by RFX1 antisense oligonucleotides decreased EAAT3 expression in rat cortical neurons in culture. These results suggest that RFX1 enhances the activity of EAAT3 promoter to increase the expression of EAAT3 proteins. This study provides initial evidence for the regulation of gene expression in the nervous cells by RFX1.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Transportador 3 de Aminoácidos Excitadores/biosíntesis , Regulación del Desarrollo de la Expresión Génica , Factores de Transcripción/fisiología , Animales , Línea Celular Tumoral , Corteza Cerebral/embriología , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/química , Proteínas de Transporte de Glutamato en la Membrana Plasmática/metabolismo , Neuronas/metabolismo , Plásmidos/metabolismo , Regiones Promotoras Genéticas , Ratas , Ratas Sprague-Dawley , Factores de Transcripción del Factor Regulador X , Factor Regulador X1 , Factores de Transcripción/biosíntesis , Factores de Transcripción/química , Transfección
16.
Biol Reprod ; 74(6): 1026-33, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16481596

RESUMEN

Early in postnatal life the first phase of spermatogenesis is accompanied by an initial wave of germ cell apoptosis. This wave of germ cell death is thought to reflect an adjustment of germ cell numbers that can be adequately maintained by Sertoli cells. Caspase 2 is an initiator caspase whose activation has been found to stimulate apoptosis through the mitochondria. The present study investigates if germ cell apoptosis during the first phase of spermatogenesis involves activation of caspase 2. Germ cell apoptosis was found to peak at Postnatal Days (pnds) 15 and 16 in male C57BL/6 mice. Western blot analysis revealed that caspase 2 also increased in the testes at pnd 16. Immunolocalization of total caspase 2 showed staining of germ cells in the periphery of the seminiferous tubules as well as germ cells more centrally located in an area where apoptotic germ cells were observed. Cytoplasmic as well as nuclear staining was observed. Western blot analysis of cytoplasmic and nuclear proteins from pnd 16 testis revealed pro-caspase 2 in both fractions. Further Western blot analysis for caspase 2 detected an increase in the activation of caspase 2 at pnd 16 in proteins isolated from the cytoplasm but not from the nucleus. Proteins isolated from mitochondria from pnd 16 testes revealed an increase in pro-caspase 2 as well as activated caspase 2 corresponding with an increase in cytochrome c in cytoplasmic fractions. Injection of the caspase 2-specific inhibitor z-VDVAD-fmk directly into the testis significantly reduced the observed germ cell apoptosis at pnds 15 and 16. These results suggest that caspase 2 is present in germ cells in the murine testis in early postnatal life and increases in expression in correspondence to the initial wave of germ cell apoptosis. Caspase 2 also localizes to mitochondria, where it is correlated with a release of cytochrome c and germ cell apoptosis. Blockade of caspase 2 activation reduced the number of apoptotic germ cells in the initial wave of germ cell apoptosis, indicating that caspase 2 plays an important role upstream of the mitochondria in germ cell apoptosis during the first phase of spermatogenesis.


Asunto(s)
Apoptosis/fisiología , Caspasas/metabolismo , Espermatogénesis/fisiología , Espermatozoides/citología , Espermatozoides/fisiología , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Caspasa 2 , Citocromos c/análisis , Citocromos c/fisiología , Activación Enzimática/fisiología , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/química , Mitocondrias/enzimología , Oligopéptidos/farmacología , Epitelio Seminífero/química , Epitelio Seminífero/citología , Epitelio Seminífero/enzimología , Espermatozoides/química , Espermatozoides/enzimología , Testículo/química , Testículo/citología , Testículo/enzimología
17.
Mol Pharmacol ; 65(5): 1172-80, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15102945

RESUMEN

A brief exposure to the volatile anesthetic isoflurane (preconditioning) induces ischemic tolerance in rat brain. However, whether isoflurane preconditioning improves long-term neurological outcome after brain ischemia and the mechanisms for this neuroprotection are not known. Here, we report that isoflurane preconditioning (2% isoflurane for 30 min at 24 h before brain ischemia) reduced brain infarct sizes and improved neurological deficit scores assessed 6, 24, and 72 h after permanent right middle cerebral arterial occlusion (MCAO) in adult male rats. More morphologically intact neurons and fewer dying cells existed in the ipsilateral frontal cortex area 1 and rostral subventricular zone of caudate putamen of isoflurane-preconditioned rats than rats undergoing MCAO alone at 14 days after the MCAO. This neuroprotection was abolished by an inhibitor of p38 mitogen-activated protein kinases (MAPK), 4-(4-fluorophenyl)-2-(4-methylsulfinylphenyl)-5-(4-pyridyl)1H-imidazole (SB203580) (the percentages of infarct volumes in the ipsilateral hemisphere volumes were 34 +/- 7% for MCAO, 24 +/- 6% for isoflurane preconditioning plus MCAO, and 30 +/- 6% for SB203580 plus isoflurane preconditioning plus MCAO, n = 8, P < 0.05 for isoflurane preconditioning plus MCAO to compare with MCAO alone or with SB203580 plus isoflurane preconditioning plus MCAO) and mimicked by an activator of these kinases, anisomycin. Isoflurane induced a rapid and prolonged increase of the phosphorylated p38 MAPK in cerebral neocortex. These active kinases distributed mainly in perikaryal regions of neurons. These results suggest that isoflurane preconditioning may improve long-term neurological outcome after focal brain ischemia and that the effects may be mediated by activating p38 MAPK.


Asunto(s)
Isquemia Encefálica/prevención & control , Isoflurano/uso terapéutico , Proteínas Quinasas Activadas por Mitógenos/fisiología , Fármacos Neuroprotectores/uso terapéutico , Animales , Antiinflamatorios no Esteroideos/farmacología , Isquemia Encefálica/complicaciones , Isquemia Encefálica/etiología , Modelos Animales de Enfermedad , Imidazoles/farmacología , Precondicionamiento Isquémico , Masculino , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosforilación , Piridinas/farmacología , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Proteínas Quinasas p38 Activadas por Mitógenos
18.
Anesthesiology ; 100(2): 331-7, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14739808

RESUMEN

BACKGROUND: A period of hypothermia before ischemia (hypothermic preconditioning) induces a delayed phase of ischemic tolerance in rat brain. However, whether hypothermic preconditioning induces an acute phase (within a few hours after the hypothermia) of ischemic tolerance remains unknown. This study was designed to determine the time window of the hypothermic preconditioning-induced acute phase of neuroprotection, which is useful information for situations during surgery with anticipated ischemic episodes, and its involved mechanisms. METHODS: Survival of Purkinje cells in rat cerebellar slices was evaluated after a 20-min oxygen-glucose deprivation (OGD, in vitro simulated ischemia) followed by a 4-h recovery. Mild hypothermia (33 degrees C) for 20 min was applied at various times before the OGD. RESULTS: The hypothermia applied immediately to 3 h before the OGD equally effectively reduced OGD-induced Purkinje cell death/injury. Glibenclamide, a selective KATP channel blocker; 8-cyclopentyl-1,3-dipropylxanthine, a selective adenosine A1 receptor antagonist; and farnesyl protein transferase inhibitor III, a selective inhibitor to reduce Ras farnesylation, abolished hypothermic preconditioning-induced neuroprotection when applied during the hypothermia. OGD increased the expression of high-mobility group I(Y) proteins, which are nuclear transcription factors to enhance the expression of putatively damaging proteins such as cyclooxygenase-2, in cerebellar slices. This increase was attenuated by hypothermic preconditioning. CONCLUSIONS: Hypothermic preconditioning induces an acute phase of neuroprotection. This neuroprotection depends on activation of the signaling molecules, adenosine A1 receptors, KATP channels, and Ras. Inhibition of putatively damaging proteins via the effects of hypothermic preconditioning on high-mobility group I(Y) expression may also be involved in hypothermic preconditioning-induced neuroprotection.


Asunto(s)
Isquemia Encefálica/patología , Encéfalo/irrigación sanguínea , Supervivencia Celular , Hipotermia Inducida , Precondicionamiento Isquémico/métodos , Células de Purkinje/metabolismo , Animales , Gliburida/farmacología , Masculino , Prenilación de Proteína/efectos de los fármacos , Células de Purkinje/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
19.
Anesthesiology ; 100(3): 562-8, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15108969

RESUMEN

BACKGROUND: Morphine pretreatment via activation of delta1-opioid receptors induces cardioprotection. In this study, the authors determined whether morphine preconditioning induces ischemic tolerance in neurons. METHODS: Cerebellar brain slices from adult Sprague-Dawley rats were incubated with morphine at 0.1-10 microM in the presence or absence of various antagonists for 30 min. They were then kept in morphine- and antagonist-free buffer for 30 min before they were subjected to simulated ischemia (oxygen-glucose deprivation) for 20 min. After being recovered in oxygenated artificial cerebrospinal fluid for 5 h, they were fixed for morphologic examination to determine the percentage of undamaged Purkinje cells. RESULTS: The survival rate of Purkinje cells was significantly higher in slices preconditioned with morphine (> or = 0.3 microM) before the oxygen-glucose deprivation (57 +/- 4% at 0.3 microM morphine) than that of the oxygen-glucose deprivation alone (39 +/- 3%, P < 0.05). This morphine preconditioning-induced neuroprotection was abolished by naloxone, a non-type-selective opioid receptor antagonist, by naltrindole, a selective delta-opioid receptor antagonist, or by 7-benzylidenenaltrexone, a selective delta1-opioid receptor antagonist. However, the effects were not blocked by the mu-, kappa-, or delta2-opioid receptor antagonists, beta-funaltrexamine, nor-binaltorphimine, or naltriben, respectively. Morphine preconditioning-induced neuroprotection was partially blocked by the selective mitochondrial adenosine triphosphate-sensitive potassium channel antagonist, 5-hydroxydecanoate, or the mitochondrial electron transport inhibitor, myxothiazol. None of the inhibitors used in this study alone affected the simulated ischemia-induced neuronal death. CONCLUSIONS: These data suggest that morphine preconditioning is neuroprotective. This neuroprotection may be delta1-opioid receptor dependent and may involve mitochondrial adenosine triphosphate-sensitive potassium channel activation and free radical production. Because morphine is a commonly used analgesic, morphine preconditioning may be explored further for potential clinical use to reduce ischemic brain injury.


Asunto(s)
Precondicionamiento Isquémico , Morfina/farmacología , Narcóticos/farmacología , Fármacos Neuroprotectores , Células de Purkinje/efectos de los fármacos , Daño por Reperfusión/patología , Animales , Muerte Celular/efectos de los fármacos , Cerebelo/patología , Relación Dosis-Respuesta a Droga , Transporte de Electrón/efectos de los fármacos , Glucosa/fisiología , Hipoxia/patología , Técnicas In Vitro , Masculino , Proteínas de la Membrana/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Mitocondrias/metabolismo , Antagonistas de Narcóticos/farmacología , Canales de Potasio , Ratas , Ratas Sprague-Dawley , Receptores Opioides delta/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA