Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Cell ; 158(5): 1159-1172, 2014 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-25171414

RESUMEN

In Alzheimer's disease (AD) brain, exposure of axons to Aß causes pathogenic changes that spread retrogradely by unknown mechanisms, affecting the entire neuron. We found that locally applied Aß1-42 initiates axonal synthesis of a defined set of proteins including the transcription factor ATF4. Inhibition of local translation and retrograde transport or knockdown of axonal Atf4 mRNA abolished Aß-induced ATF4 transcriptional activity and cell loss. Aß1-42 injection into the dentate gyrus (DG) of mice caused loss of forebrain neurons whose axons project to the DG. Protein synthesis and Atf4 mRNA were upregulated in these axons, and coinjection of Atf4 siRNA into the DG reduced the effects of Aß1-42 in the forebrain. ATF4 protein and transcripts were found with greater frequency in axons in the brain of AD patients. These results reveal an active role for intra-axonal translation in neurodegeneration and identify ATF4 as a mediator for the spread of AD pathology.


Asunto(s)
Factor de Transcripción Activador 4/análisis , Enfermedad de Alzheimer/patología , Encéfalo/patología , Factor de Transcripción Activador 4/metabolismo , Péptidos beta-Amiloides/genética , Animales , Axones/metabolismo , Encéfalo/citología , Química Encefálica , Factor 2 Eucariótico de Iniciación/metabolismo , Hipocampo , Humanos , Ratones Endogámicos C57BL , Ratas , Factor de Transcripción CHOP/metabolismo
2.
Am J Hum Genet ; 99(5): 1117-1129, 2016 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-27773430

RESUMEN

Lissencephaly is a malformation of cortical development typically caused by deficient neuronal migration resulting in cortical thickening and reduced gyration. Here we describe a "thin" lissencephaly (TLIS) variant characterized by megalencephaly, frontal predominant pachygyria, intellectual disability, and seizures. Trio-based whole-exome sequencing and targeted re-sequencing identified recessive mutations of CRADD in six individuals with TLIS from four unrelated families of diverse ethnic backgrounds. CRADD (also known as RAIDD) is a death-domain-containing adaptor protein that oligomerizes with PIDD and caspase-2 to initiate apoptosis. TLIS variants cluster in the CRADD death domain, a platform for interaction with other death-domain-containing proteins including PIDD. Although caspase-2 is expressed in the developing mammalian brain, little is known about its role in cortical development. CRADD/caspase-2 signaling is implicated in neurotrophic factor withdrawal- and amyloid-ß-induced dendritic spine collapse and neuronal apoptosis, suggesting a role in cortical sculpting and plasticity. TLIS-associated CRADD variants do not disrupt interactions with caspase-2 or PIDD in co-immunoprecipitation assays, but still abolish CRADD's ability to activate caspase-2, resulting in reduced neuronal apoptosis in vitro. Homozygous Cradd knockout mice display megalencephaly and seizures without obvious defects in cortical lamination, supporting a role for CRADD/caspase-2 signaling in mammalian brain development. Megalencephaly and lissencephaly associated with defective programmed cell death from loss of CRADD function in humans implicate reduced apoptosis as an important pathophysiological mechanism of cortical malformation. Our data suggest that CRADD/caspase-2 signaling is critical for normal gyration of the developing human neocortex and for normal cognitive ability.


Asunto(s)
Apoptosis , Proteína Adaptadora de Señalización CRADD/genética , Caspasa 2/metabolismo , Cisteína Endopeptidasas/metabolismo , Lisencefalia/genética , Megalencefalia/genética , Neuronas/metabolismo , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Animales , Caspasa 2/genética , Supervivencia Celular , Clonación Molecular , Cognición , Cisteína Endopeptidasas/genética , Células Dendríticas/metabolismo , Etnicidad/genética , Genes Recesivos , Estudio de Asociación del Genoma Completo , Células HEK293 , Humanos , Inmunoprecipitación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Células PC12 , Ratas , Transducción de Señal
3.
Brain ; 137(Pt 6): 1656-75, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24727569

RESUMEN

We have previously shown that crushing the optic nerve induces death of retinal ganglion cells by apoptosis, but suppression of CASP2, which is predominantly activated in retinal ganglion cells, using a stably modified short interfering RNA CASP2, inhibits retinal ganglion cell apoptosis. Here, we report that combined delivery of short interfering CASP2 and inhibition of CASP6 using a dominant negative CASP6 mutant activates astrocytes and Müller cells, increases CNTF levels in the retina and leads to enhanced retinal ganglion cell axon regeneration. In dissociated adult rat mixed retinal cultures, dominant negative CASP6 mutant + short interfering CASP2 treatment also significantly increases GFAP+ glial activation, increases the expression of CNTF in culture, and subsequently increases the number of retinal ganglion cells with neurites and the mean retinal ganglion cell neurite length. These effects are abrogated by the addition of MAB228 (a monoclonal antibody targeted to the gp130 component of the CNTF receptor) and AG490 (an inhibitor of the JAK/STAT pathway downstream of CNTF signalling). Similarly, in the optic nerve crush injury model, MAB228 and AG490 neutralizes dominant negative CASP6 mutant + short interfering CASP2-mediated retinal ganglion cell axon regeneration, Müller cell activation and CNTF production in the retina without affecting retinal ganglion cell survival. We therefore conclude that axon regeneration promoted by suppression of CASP2 and CASP6 is CNTF-dependent and mediated through the JAK/STAT signalling pathway. This study offers insights for the development of effective therapeutics for promoting retinal ganglion cell survival and axon regeneration.


Asunto(s)
Apoptosis/efectos de los fármacos , Axones/metabolismo , Caspasa 2/metabolismo , Caspasa 6/metabolismo , Factor Neurotrófico Ciliar/metabolismo , Cisteína Endopeptidasas/metabolismo , Células Ganglionares de la Retina/metabolismo , Transducción de Señal , Animales , Apoptosis/genética , Modelos Animales de Enfermedad , Femenino , Regeneración Nerviosa/efectos de los fármacos , Regeneración Nerviosa/genética , Regeneración Nerviosa/fisiología , Nervio Óptico/metabolismo , Traumatismos del Nervio Óptico/metabolismo , Ratas , Ratas Sprague-Dawley , Células Ganglionares de la Retina/citología , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/fisiología
4.
Biochem J ; 455(1): 15-25, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23815625

RESUMEN

Neuronal apoptotic death generally requires de novo transcription, and activation of the transcription factor c-Jun has been shown to be necessary in multiple neuronal death paradigms. Caspase-2 has been implicated in death of neuronal and non-neuronal cells, but its relationship to transcriptional activation has not been clearly elucidated. In the present study, using two different neuronal apoptotic paradigms, ß-amyloid treatment and NGF (nerve growth factor) withdrawal, we examined the hierarchical role of caspase-2 activation in the transcriptional control of neuron death. Both paradigms induce rapid activation of caspase-2 as well as activation of the transcription factor c-Jun and subsequent induction of the pro-apoptotic BH3 (Bcl-homology domain 3)-only protein Bim (Bcl-2-interacting mediator of cell death). Caspase-2 activation is dependent on the adaptor protein RAIDD {RIP (receptor-interacting protein)-associated ICH-1 [ICE (interleukin-1ß-converting enzyme)/CED-3 (cell-death determining 3) homologue 1] protein with a death domain}, and both caspase-2 and RAIDD are required for c-Jun activation and Bim induction. The present study thus shows that rapid caspase-2 activation is essential for c-Jun activation and Bim induction in neurons subjected to apoptotic stimuli. This places caspase-2 at an apical position in the apoptotic cascade and demonstrates for the first time that caspase-2 can regulate transcription.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Proteína Adaptadora de Señalización CRADD/genética , Caspasa 2/genética , Proteínas de la Membrana/genética , Neuronas/metabolismo , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas/genética , Activación Transcripcional/efectos de los fármacos , Péptidos beta-Amiloides/farmacología , Animales , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteína 11 Similar a Bcl2 , Proteína Adaptadora de Señalización CRADD/metabolismo , Caspasa 2/metabolismo , Feto , Proteínas de la Membrana/metabolismo , Factor de Crecimiento Nervioso/deficiencia , Neuronas/citología , Neuronas/efectos de los fármacos , Cultivo Primario de Células , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Transcripción Genética/efectos de los fármacos
5.
Cells ; 13(2)2024 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-38247821

RESUMEN

In the mammalian isocortex, CD44, a cell surface receptor for extracellular matrix molecules, is present in pial-based and fibrous astrocytes of white matter but not in protoplasmic astrocytes. In the hominid isocortex, CD44+ astrocytes comprise the subpial "interlaminar" astrocytes, sending long processes into the cortex. The hippocampus also contains similar astrocytes. We have examined all levels of the human central nervous system and found CD44+ astrocytes in every region. Astrocytes in white matter and astrocytes that interact with large blood vessels but not with capillaries in gray matter are CD44+, the latter extending long processes into the parenchyma. Motor neurons in the brainstem and spinal cord, such as oculomotor, facial, hypoglossal, and in the anterior horn of the spinal cord, are surrounded by CD44+ processes, contrasting with neurons in the cortex, basal ganglia, and thalamus. We found CD44+ processes that intercalate between ependymal cells to reach the ventricle. We also found CD44+ astrocytes in the molecular layer of the cerebellar cortex. Protoplasmic astrocytes, which do not normally contain CD44, acquire it in pathologies like hypoxia and seizures. The pervasive and inducible expression of CD44 in astrocytes is a novel finding that lays the foundations for functional studies into the significance of CD44 in health and disease.


Asunto(s)
Receptores de Hialuranos , Hipoxia , Convulsiones , Animales , Humanos , Astrocitos , Receptores de Hialuranos/metabolismo , Hipoxia/metabolismo , Neocórtex , Convulsiones/metabolismo , Sustancia Blanca
6.
Biochem J ; 444(3): 591-9, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22515271

RESUMEN

Caspase 2 was initially identified as a neuronally expressed developmentally down-regulated gene (HUGO gene nomenclature CASP2) and has been shown to be required for neuronal death induced by several stimuli, including NGF (nerve growth factor) deprivation and Aß (ß-amyloid). In non-neuronal cells the PIDDosome, composed of caspase 2 and two death adaptor proteins, PIDD (p53-inducible protein with a death domain) and RAIDD {RIP (receptor-interacting protein)-associated ICH-1 [ICE (interleukin-1ß-converting enzyme)/CED-3 (cell-death determining 3) homologue 1] protein with a death domain}, has been proposed as the caspase 2 activation complex, although the absolute requirement for the PIDDosome is not clear. To investigate the requirement for the PIDDosome in caspase-2-dependent neuronal death, we have examined the necessity for each component in induction of active caspase 2 and in execution of caspase-2-dependent neuronal death. We find that both NGF deprivation and Aß treatment of neurons induce active caspase 2 and that induction of this activity depends on expression of RAIDD, but is independent of PIDD expression. We show that treatment of wild-type or PIDD-null neurons with Aß or NGF deprivation induces formation of a complex of caspase 2 and RAIDD. We also show that caspase-2-dependent execution of neurons requires RAIDD, not PIDD. Caspase 2 activity can be induced in neurons from PIDD-null mice, and NGF deprivation or Aß use caspase 2 and RAIDD to execute death of these neurons.


Asunto(s)
Proteína Adaptadora de Señalización CRADD/biosíntesis , Caspasa 2/metabolismo , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte/biosíntesis , Neuronas/enzimología , Animales , Animales Recién Nacidos , Células Cultivadas , Activación Enzimática/fisiología , Ratones , Ratones Noqueados , Factor de Crecimiento Nervioso/deficiencia , Factor de Crecimiento Nervioso/farmacología , Células PC12 , Ratas , Ratas Sprague-Dawley
7.
Front Neurosci ; 17: 1209527, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37449272

RESUMEN

Purpose: Retinal vein occlusion (RVO) is a sight-threatening condition typically treated with intravitreal injection of vascular endothelial growth factor (VEGF) antagonists. Treatment response to anti-VEGF therapies is highly variable, with poor visual outcomes and treatment response in patients with significant retinal nonperfusion following RVO. Recently, caspase-9 has been identified as a potent regulator of edema, gliosis, and neuronal dysfunction during acute retinal hypoxia. The purpose of this study was to compare the therapeutic effect of caspase-9 inhibition against VEGF-neutralization in an established mouse model of RVO. Methods: Adult male C57Bl/6 J mice were randomized to induction of RVO and treatment with either vehicle, intravitreal injection of anti-VEGF antibody, topical administration of a selective caspase-9 inhibitor (Pen1-XBir3), or a combination therapy. Animals were followed on days 1, 2, and 8 after RVO with fundus retinal imaging, and with optical coherence tomography (OCT) to capture retinal swelling, capillary nonperfusion (measured by disorganization of retinal inner layers, DRIL), hyperreflective foci (HRF), and retinal atrophy. Focal electroretinography (ERG) measurements were performed on day 7. Histology was performed on retinal sections from day 8. Results: Both VEGF neutralization and caspase-9 inhibition showed significant retinal protection from RVO compared to vehicle treatment arm. Retinal reperfusion of occluded veins was accelerated in eyes receiving caspase-9 inhibitor, but not significantly different from vehicle in the anti-VEGF group. Retinal edema was suppressed in all treatment groups, with approximately 2-fold greater edema reduction with caspase-9 inhibition compared to VEGF neutralization. HRF were reduced similarly across all treatment groups compared to vehicle. Retinal detachment was reduced only in eyes treated with caspase-9 inhibitor monotherapy. Caspase-9 inhibition reduced retinal atrophy and preserved ERG response; VEGF neutralization did not prevent neurodegeneration following RVO. Conclusion: Caspase-9 inhibition confers stronger neuronal and vascular protection compared to VEGF neutralization in the mouse laser-induced model of RVO.

8.
J Neurosci ; 31(24): 8894-904, 2011 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-21677173

RESUMEN

Despite extensive research to develop an effective neuroprotective strategy for the treatment of ischemic stroke, therapeutic options remain limited. Although caspase-dependent death is thought to play a prominent role in neuronal injury, direct evidence of active initiator caspases in stroke and the functional relevance of this activity have not previously been shown. Using an unbiased caspase-trapping technique in vivo, we isolated active caspase-9 from ischemic rat brain within 1 h of reperfusion. Pathogenic relevance of active caspase-9 was shown by intranasal delivery of a novel cell membrane-penetrating highly specific inhibitor for active caspase-9 at 4 h postreperfusion (hpr). Caspase-9 inhibition provided neurofunctional protection and established caspase-6 as its downstream target. The temporal and spatial pattern of expression demonstrates that neuronal caspase-9 activity induces caspase-6 activation, mediating axonal loss by 12 hpr followed by neuronal death within 24 hpr. Collectively, these results support selective inhibition of these specific caspases as an effective therapeutic strategy for stroke.


Asunto(s)
Caspasa 6/fisiología , Inhibidores Enzimáticos/uso terapéutico , Infarto de la Arteria Cerebral Media , Proteínas Inhibidoras de la Apoptosis/uso terapéutico , Enfermedades del Sistema Nervioso , Neuronas/patología , Administración Intranasal , Aldehídos/farmacología , Animales , Infarto Encefálico/tratamiento farmacológico , Infarto Encefálico/etiología , Caspasa 6/deficiencia , Caspasa 9/metabolismo , Inhibidores de Caspasas , Inhibidores de Cisteína Proteinasa/uso terapéutico , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Técnicas In Vitro , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/patología , Proteínas Inhibidoras de la Apoptosis/química , Proteínas Inhibidoras de la Apoptosis/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/metabolismo , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Enfermedades del Sistema Nervioso/etiología , Enfermedades del Sistema Nervioso/patología , Fosfohidrolasa PTEN/química , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/uso terapéutico , Ratas , Ratas Wistar , Factores de Tiempo
9.
Ann Neurol ; 69(5): 819-30, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21472769

RESUMEN

OBJECTIVE: The goal of this study was to investigate the role of endogenous amyloid-ß peptide (Aß) in healthy brain. METHODS: Long-term potentiation (LTP), a type of synaptic plasticity that is thought to be associated with learning and memory, was examined through extracellular field recordings from the CA1 region of hippocampal slices, whereas behavioral techniques were used to assess contextual fear memory and reference memory. Amyloid precursor protein (APP) expression was reduced through small interfering RNA (siRNA) technique. RESULTS: We found that both antirodent Aß antibody and siRNA against murine APP reduced LTP as well as contextual fear memory and reference memory. These effects were rescued by the addition of human Aß42, suggesting that endogenously produced Aß is needed for normal LTP and memory. Furthermore, the effect of endogenous Aß on plasticity and memory was likely due to regulation of transmitter release, activation of α7-containing nicotinic acetylcholine receptors, and Aß42 production. INTERPRETATION: Endogenous Aß42 is a critical player in synaptic plasticity and memory within the normal central nervous system. This needs to be taken into consideration when designing therapies aiming at reducing Aß levels to treat Alzheimer disease.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Hipocampo/fisiología , Potenciación a Largo Plazo/fisiología , Memoria/fisiología , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/inmunología , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Anticuerpos/farmacología , Conducta Animal/efectos de los fármacos , Biofisica/métodos , Estimulación Eléctrica/métodos , Ensayo de Inmunoadsorción Enzimática/métodos , Regulación de la Expresión Génica/fisiología , Hipocampo/efectos de los fármacos , Humanos , Potenciación a Largo Plazo/efectos de los fármacos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Memoria/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Fragmentos de Péptidos/metabolismo , ARN Interferente Pequeño/farmacología
10.
J Vis Exp ; (182)2022 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-35532239

RESUMEN

Advancements in ophthalmic imaging tools offer an unprecedented level of access to researchers working with animal models of neurovascular injury. To properly leverage this greater translatability, there is a need to devise reproducible methods of drawing quantitative data from these images. Optical coherence tomography (OCT) imaging can resolve retinal histology at micrometer resolution and reveal functional differences in vascular blood flow. Here, we delineate noninvasive vascular readouts that we use to characterize pathological damage post vascular insult in an optimized mouse model of retinal vein occlusion (RVO). These readouts include live imaging analysis of retinal morphology, disorganization of retinal inner layers (DRIL) measure of capillary ischemia, and fluorescein angiography measures of retinal edema and vascular density. These techniques correspond directly to those used to examine patients with retinal disease in the clinic. Standardizing these methods enables direct and reproducible comparison of animal models with clinical phenotypes of ophthalmic disease, increasing the translational power of vascular injury models.


Asunto(s)
Lesiones del Sistema Vascular , Animales , Humanos , Ratones , Reproducibilidad de los Resultados , Retina/diagnóstico por imagen , Retina/patología , Vasos Retinianos/diagnóstico por imagen , Estudios Retrospectivos , Tomografía de Coherencia Óptica/métodos , Lesiones del Sistema Vascular/patología , Agudeza Visual
11.
Transl Vis Sci Technol ; 11(8): 5, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35921115

RESUMEN

Purpose: To characterize postnatal ocular pathology in a Ndufs4-/- mouse model of complex I deficiency using noninvasive retinal imaging and visual testing. Methods: Ndufs4-/- mice and wild-type (WT) littermates were analyzed at 3, 5, and 7 weeks postnatal. Retinal morphology was visualized by optical coherence tomography (OCT). OCT images were analyzed for changes in retinal thickness and reflectivity profiles. Visual function was assessed by electroretinogram (ERG) and optomotor reflex (OMR). Results: Ndufs4-/- animals have normal OCT morphology at weaning and develop inner plexiform layer atrophy over weeks 5 to 7. Outer retinal layers show hyporeflectivity of the external limiting membrane (ELM) and photoreceptor ellipsoid zone (EZ). Retinal function is impaired at 3 weeks, with profound deficits in b-wave, a-wave, and oscillatory potential amplitudes. The b-wave and oscillatory potential implicit times are delayed, but the a-wave implicit time is unaffected. Ndufs4-/- animals have normal OMR at 3 weeks and present with increasing acuity and contrast OMR deficits at 5 and 7 weeks. Physiological thinning of inner retinal layers, attenuation of ELM reflectivity, and attenuation of ERG b- and a-wave amplitudes occur in WT C57BL/6 littermates between weeks 3 and 7. Conclusions: Noninvasive ocular imaging captures early-onset retinal degeneration in Ndufs4-/- mice and is a tractable approach for investigating retinal pathology subsequent to complex I deficiency. Translational Relevance: Ophthalmic imaging captures clinically relevant measures of retinal disease in a fast-progressing mouse model of complex I deficiency consistent with human Leigh syndrome.


Asunto(s)
Enfermedades Mitocondriales , Degeneración Retiniana , Animales , Modelos Animales de Enfermedad , Complejo I de Transporte de Electrón/deficiencia , Complejo I de Transporte de Electrón/genética , Electrorretinografía , Humanos , Ratones , Ratones Endogámicos C57BL , Enfermedades Mitocondriales/diagnóstico por imagen , Degeneración Retiniana/diagnóstico por imagen , Degeneración Retiniana/patología
12.
Cell Death Dis ; 13(11): 937, 2022 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-36347836

RESUMEN

Retinal neurovascular injuries are a leading cause of vision loss in young adults presenting unmet therapeutic needs. Neurovascular injuries damage homeostatic communication between endothelial, pericyte, glial, and neuronal cells through signaling pathways that remain to be established. To understand the mechanisms that contribute to neuronal death, we use a mouse model of retinal vein occlusion (RVO). Using this model, we previously discovered that after vascular damage, there was non-apoptotic activation of endothelial caspase-9 (EC Casp9); knock-out of EC Casp9 led to a decrease in retinal edema, capillary ischemia, and neuronal death. In this study, we aimed to explore the role of EC Casp9 in vision loss and inflammation. We found that EC Casp9 is implicated in contrast sensitivity decline, induction of inflammatory cytokines, and glial reactivity. One of the noted glial changes was increased levels of astroglial cl-caspase-6, which we found to be activated cell intrinsically by astroglial caspase-9 (Astro Casp9). Lastly, we discovered that Astro Casp9 contributes to capillary ischemia and contrast sensitivity decline after RVO (P-RVO). These findings reveal specific endothelial and astroglial non-apoptotic caspase-9 roles in inflammation and neurovascular injury respectively; and concomitant relevancy to contrast sensitivity decline.


Asunto(s)
Sensibilidad de Contraste , Oclusión de la Vena Retiniana , Ratones , Animales , Caspasa 9/genética , Caspasa 9/metabolismo , Oclusión de la Vena Retiniana/etiología , Oclusión de la Vena Retiniana/metabolismo , Inflamación/metabolismo , Isquemia/metabolismo , Caspasa 3/metabolismo
13.
J Vis Exp ; (185)2022 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-35938825

RESUMEN

The family of caspases is known to mediate many cellular pathways beyond cell death, including cell differentiation, axonal pathfinding, and proliferation. Since the identification of the family of cell death proteases, there has been a search for tools to identify and expand the function of specific family members in development, health, and disease states. However, many of the currently commercially available caspase tools that are widely used are not specific for the targeted caspase. In this report, we delineate the approach we have used to identify, validate, and target caspase-9 in the nervous system using a novel inhibitor and genetic approaches with immunohistochemical read-outs. Specifically, we used the retinal neuronal tissue as a model to identify and validate the presence and function of caspases. This approach enables the interrogation of cell-type specific apoptotic and non-apoptotic caspase-9 functions and can be applied to other complex tissues and caspases of interest. Understanding the functions of caspases can help to expand current knowledge in cell biology, and can also be advantageous to identify potential therapeutic targets due to their involvement in disease.


Asunto(s)
Caspasas , Retina , Apoptosis , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Caspasas/metabolismo , Diferenciación Celular , Sistema Nervioso , Retina/metabolismo
14.
Cell Death Dis ; 13(11): 959, 2022 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-36379916

RESUMEN

Caspase-2 (Casp2) is a promising therapeutic target in several human diseases, including nonalcoholic steatohepatitis (NASH) and Alzheimer's disease (AD). However, the design of an active-site-directed inhibitor selective to individual caspase family members is challenging because caspases have extremely similar active sites. Here we present new peptidomimetics derived from the VDVAD pentapeptide structure, harboring non-natural modifications at the P2 position and an irreversible warhead. Enzyme kinetics show that these new compounds, such as LJ2 or its specific isomers LJ2a, and LJ3a, strongly and irreversibly inhibit Casp2 with genuine selectivity. In agreement with the established role of Casp2 in cellular stress responses, LJ2 inhibits cell death induced by microtubule destabilization or hydroxamic acid-based deacetylase inhibition. The most potent peptidomimetic, LJ2a, inhibits human Casp2 with a remarkably high inactivation rate (k3/Ki ~5,500,000 M-1 s-1), and the most selective inhibitor, LJ3a, has close to a 1000 times higher inactivation rate on Casp2 as compared to Casp3. Structural analysis of LJ3a shows that the spatial configuration of Cα at the P2 position determines inhibitor efficacy. In transfected human cell lines overexpressing site-1 protease (S1P), sterol regulatory element-binding protein 2 (SREBP2) and Casp2, LJ2a and LJ3a fully inhibit Casp2-mediated S1P cleavage and thus SREBP2 activation, suggesting a potential to prevent NASH development. Furthermore, in primary hippocampal neurons treated with ß-amyloid oligomers, submicromolar concentrations of LJ2a and of LJ3a prevent synapse loss, indicating a potential for further investigations in AD treatment.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Peptidomiméticos , Humanos , Caspasa 2/metabolismo , Caspasa 3/metabolismo , Neuronas/metabolismo , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Peptidomiméticos/farmacología , Peptidomiméticos/metabolismo
15.
Front Pharmacol ; 12: 701301, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34305609

RESUMEN

Caspase-9, a cysteine-aspartic protease known for its role as an initiator of intrinsic apoptosis, regulates physiological cell death and pathological tissue degeneration. Its nonapoptotic functions, including regulation of cellular differentiation/maturation, innate immunity, mitochondrial homeostasis, and autophagy, reveal a multimodal landscape of caspase-9 functions in health and disease. Recent work has demonstrated that caspase-9 can drive neurovascular injury through nonapoptotic endothelial cell dysfunction. CASP9 polymorphisms have been linked with various cancers, neurological disorders, autoimmune pathologies and lumbar disc disease. Clinical reports suggest alterations in caspase-9 expression, activity or function may be associated with acute and chronic neurodegeneration, retinal neuropathy, slow-channel myasthenic syndrome, lumbar disc disease, cardiomyopathies, atherosclerosis and autoimmune disease. Healthy tissues maintain caspase-9 activity at low basal levels, rendering supraphysiological caspase-9 activation a tractable target for therapeutic interventions. Strategies for selective inhibition of caspase-9 include dominant negative caspase-9 mutants and pharmacological inhibitors derived from the XIAP protein, whose Bir3 domain is an endogenous highly selective caspase-9 inhibitor. However, the mechanistic implications of caspase-9 expression and activation remain indeterminate in many pathologies. By assembling clinical reports of caspase-9 genetics, signaling and cellular localization in human tissues, this review identifies gaps between experimental and clinical studies on caspase-9, and presents opportunities for further investigations to examine the consequences of caspase activity in human disease.

16.
J Vis Exp ; (174)2021 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-34424250

RESUMEN

Mouse models of retinal vein occlusion (RVO) are often used in ophthalmology to study hypoxic-ischemic injury in the neural retina. In this report, a detailed method pointing out critical steps is provided with recommendations for optimization to achieve consistently successful occlusion rates across different genetically modified mouse strains. The RVO mouse model consists primarily of the intravenous administration of a photosensitizer dye followed by laser photocoagulation using a retinal imaging microscope attached to an ophthalmic guided laser. Three variables were identified as determinants of occlusion consistency. By adjusting the wait time after rose bengal administration and balancing the baseline and experimental laser output, the variability across experiments can be limited and a higher success rate of occlusions achieved. This method can be used to study retinal diseases that are characterized by retinal edema and hypoxic-ischemic injury. Additionally, as this model induces vascular injury, it can also be applied to study the neurovasculature, neuronal death, and inflammation.


Asunto(s)
Oclusión de la Vena Retiniana , Animales , Modelos Animales de Enfermedad , Hipoxia , Ratones , Retina , Rosa Bengala
17.
J Neurosci ; 29(4): 1011-6, 2009 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-19176810

RESUMEN

Neurodegenerative illnesses such as Parkinson and Alzheimer disease are an increasingly prevalent problem in aging societies, yet no therapies exist that retard or prevent neurodegeneration. Dominant missense mutations in leucine-rich repeat kinase 2 (LRRK2) are the most common genetic cause of Parkinson disease (PD), but the mechanisms by which mutant forms of LRRK2 disrupt neuronal function and cause cell death remain poorly understood. We report that LRRK2 interacts with the death adaptor Fas-associated protein with death domain (FADD), and that in primary neuronal culture LRRK2-mediated neurodegeneration is prevented by the functional inhibition of FADD or depletion of caspase-8, two key elements of the extrinsic cell death pathway. This pathway is activated by disease-triggering mutations, which enhance the LRRK2-FADD association and the consequent recruitment and activation of caspase-8. These results establish a direct molecular link between a mutant PD gene and the activation of programmed cell death signaling, and suggest that FADD/caspase-8 signaling contributes to LRRK2-induced neuronal death.


Asunto(s)
Apoptosis/fisiología , Caspasa 8/metabolismo , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Neuronas/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/genética , Proteínas Portadoras/genética , Línea Celular Transformada , Péptidos de Penetración Celular , Proteína de Dominio de Muerte Asociada a Fas/genética , Proteínas Fluorescentes Verdes/genética , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Mutagénesis Sitio-Dirigida/métodos , Neuronas/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/genética , ARN Interferente Pequeño/farmacología , Transfección/métodos
18.
J Neurosci ; 29(27): 8828-38, 2009 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-19587290

RESUMEN

Oxidative stress contributes to tissue injury in conditions ranging from cardiovascular disease to stroke, spinal cord injury, neurodegeneration, and perhaps even aging. Yet the efficacy of antioxidants in human disease has been mixed at best. We need a better understanding of the mechanisms by which established antioxidants combat oxidative stress. Iron chelators are well established inhibitors of oxidative death in both neural and non-neural tissues, but their precise mechanism of action remains elusive. The prevailing but not completely substantiated view is that iron chelators prevent oxidative injury by suppressing Fenton chemistry and the formation of highly reactive hydroxyl radicals. Here, we show that iron chelation protects, rather unexpectedly, by inhibiting the hypoxia-inducible factor prolyl 4-hydroxylase isoform 1 (PHD1), an iron and 2-oxoglutarate-dependent dioxygenase. PHD1 and its isoforms 2 and 3 are best known for stabilizing transcriptional regulators involved in hypoxic adaptation, such as HIF-1alpha and cAMP response element-binding protein (CREB). Yet we find that global hypoxia-inducible factor (HIF)-PHD inhibition protects neurons even when HIF-1alpha and CREB are directly suppressed. Moreover, two global HIF-PHD inhibitors continued to be neuroprotective even in the presence of diminished HIF-2alpha levels, which itself increases neuronal susceptibility to oxidative stress. Finally, RNA interference to PHD1 but not isoforms PHD2 or PHD3 prevents oxidative death, independent of HIF activation. Together, these studies suggest that iron chelators can prevent normoxic oxidative neuronal death through selective inhibition of PHD1 but independent of HIF-1alpha and CREB; and that HIF-2alpha, not HIF-1alpha, regulates susceptibility to normoxic oxidative neuronal death.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/fisiología , Dioxigenasas/antagonistas & inhibidores , Factor 1 Inducible por Hipoxia/fisiología , Neuronas/fisiología , Proteínas Nucleares/antagonistas & inhibidores , Estrés Oxidativo/fisiología , Transducción de Señal/fisiología , Secuencia de Aminoácidos , Animales , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Línea Celular Transformada , Dioxigenasas/fisiología , Prolina Dioxigenasas del Factor Inducible por Hipoxia , Quelantes del Hierro/farmacología , Ratones , Datos de Secuencia Molecular , Neuronas/efectos de los fármacos , Neuronas/patología , Proteínas Nucleares/fisiología , Estrés Oxidativo/efectos de los fármacos , Procolágeno-Prolina Dioxigenasa , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
19.
ASN Neuro ; 12: 1759091420930865, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32493127

RESUMEN

The p75 neurotrophin receptor (p75NTR) can regulate multiple cellular functions including proliferation, survival, and apoptotic cell death. The p75NTR is widely expressed in the developing brain and is downregulated as the nervous system matures, with only a few neuronal subpopulations retaining expression into adulthood. However, p75NTR expression is induced following damage to the adult brain, including after traumatic brain injury, which is a leading cause of mortality and disability worldwide. A major consequence of traumatic brain injury is the progressive neuronal loss that continues secondary to the initial trauma, which ultimately contributes to cognitive decline. Understanding mechanisms governing this progressive neuronal death is key to developing targeted therapeutic strategies to provide neuroprotection and salvage cognitive function. In this study, we demonstrate that a cortical impact injury to the sensorimotor cortex elicits p75NTR expression in apoptotic neurons in the injury penumbra, confirming previous studies. To establish whether preventing p75NTR induction or blocking the ligands would reduce the extent of secondary neuronal cell death, we used a noninvasive intranasal strategy to deliver either siRNA to block the induction of p75NTR, or function-blocking antibodies to the ligands pro-nerve growth factor and pro-brain-derived neurotrophic factor. We demonstrate that either preventing the induction of p75NTR or blocking the proneurotrophin ligands provides neuroprotection and preserves sensorimotor function.


Asunto(s)
Apoptosis/fisiología , Lesiones Traumáticas del Encéfalo/metabolismo , Neuronas/metabolismo , Receptores de Factor de Crecimiento Nervioso/biosíntesis , Administración Intranasal/métodos , Animales , Lesiones Traumáticas del Encéfalo/patología , Lesiones Traumáticas del Encéfalo/psicología , Muerte Celular/fisiología , Técnicas de Silenciamiento del Gen/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/patología , ARN Interferente Pequeño/administración & dosificación , Receptores de Factor de Crecimiento Nervioso/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA