Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Int J Mol Sci ; 24(9)2023 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-37175445

RESUMEN

Stem-cell-based therapy is very promising for Alzheimer's disease (AD), yet has not become a reality. A critical challenge is the transplantation microenvironment, which impacts the therapeutic effect of stem cells. In AD brains, amyloid-beta (Aß) peptides and inflammatory cytokines continuously poison the tissue microenvironment, leading to low survival of grafted cells and restricted efficacy. It is necessary to create a growth-supporting microenvironment for transplanted cells. Recent advances in AD studies suggest that the asparaginyl endopeptidase (AEP) is a potential intervention target for modifying pathological changes. We here chose APP/PS1 mice as an AD model and employed pharmacological inhibition of the AEP for one month to improve the brain microenvironment. Thereafter, we transplanted neural stem cells (NSCs) into the hippocampus and maintained therapy for one more month. We found that inhibition of AEPs resulted in a significant decrease of Aß, TNF-α, IL-6 and IL-1ß in their brains. In AD mice receiving NSC transplantation alone, the survival of NSCs was at a low level, while in combination with AEP inhibition pre-treatment the survival rate of engrafted cells was doubled. Within the 2-month treatment period, implantation of NSCs plus pre-inhibition of the AEP significantly enhanced neural plasticity of the hippocampus and rescued cognitive impairment. Neither NSC transplantation alone nor AEP inhibition alone achieved significant efficacy. In conclusion, pharmacological inhibition of the AEP ameliorated brain microenvironment of AD mice, and thus improved the survival and therapeutic efficacy of transplanted stem cells.


Asunto(s)
Enfermedad de Alzheimer , Células-Madre Neurales , Animales , Ratones , Enfermedad de Alzheimer/terapia , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides , Cisteína Endopeptidasas , Modelos Animales de Enfermedad , Ratones Transgénicos , Inhibidores de Cisteína Proteinasa
2.
Int J Mol Sci ; 23(18)2022 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-36142134

RESUMEN

Alzheimer's disease (AD) is incurable dementia closely associated with aging. Most cases of AD are sporadic, and very few are inherited; the pathogenesis of sporadic AD is complex and remains to be elucidated. The asparaginyl endopeptidase (AEP) or legumain is the only recognized cysteine protease that specifically hydrolyzes peptide bonds after asparagine residues in mammals. The expression level of AEPs in healthy brains is far lower than that of peripheral organs. Recently, growing evidence has indicated that aging may upregulate and overactivate brain AEPs. The overactivation of AEPs drives the onset of AD through cleaving tau and amyloid precursor proteins (APP), and SET, an inhibitor of protein phosphatase 2A (PP2A). The AEP-mediated cleavage of these peptides enhances amyloidosis, promotes tau hyperphosphorylation, and ultimately induces neurodegeneration and cognitive impairment. Upregulated AEPs and related deleterious reactions constitute upstream events of amyloid/tau toxicity in the brain, and represent early pathological changes in AD. Thus, upregulated AEPs are an emerging drug target for disease modification and a potential biomarker for predicting preclinical AD. However, the presence of the blood-brain barrier greatly hinders establishing body-fluid-based methods to measure brain AEPs. Research on AEP-activity-based imaging probes and our recent work suggest that the live brain imaging of AEPs could be used to evaluate its predictive efficacy as an AD biomarker. To advance translational research in this area, AEP imaging probes applicable to human brain and AEP inhibitors with good druggability are urgently needed.


Asunto(s)
Enfermedad de Alzheimer , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Asparagina/metabolismo , Biomarcadores/metabolismo , Encéfalo/metabolismo , Cisteína Endopeptidasas/metabolismo , Humanos , Mamíferos/metabolismo , Proteína Fosfatasa 2/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo
3.
Int J Mol Sci ; 23(2)2022 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-35055084

RESUMEN

The role of calcium ion (Ca2+) signaling in tumorigenicity has received increasing attention in melanoma research. Previous Ca2+ signaling studies focused on Ca2+ entry routes, but rarely explored the role of Ca2+ extrusion. Functioning of the Na+/Ca2+ exchanger (NCX) on the plasma membrane is the major way of Ca2+ extrusion, but very few associations between NCX and melanoma have been reported. Here, we explored whether pharmacological modulation of the NCX could suppress melanoma and promise new therapeutic strategies. Methods included cell viability assay, Ca2+ imaging, immunoblotting, and cell death analysis. The NCX inhibitors SN-6 and YM-244769 were used to selectively block reverse operation of the NCX. Bepridil, KB-R7943, and CB-DMB blocked either reverse or forward NCX operation. We found that blocking the reverse NCX with SN-6 or YM-244769 (5-100 µM) did not affect melanoma cells or increase cytosolic Ca2+. Bepridil, KB-R7943, and CB-DMB all significantly suppressed melanoma cells with IC50 values of 3-20 µM. Bepridil and KB-R7943 elevated intracellular Ca2+ level of melanoma. Bepridil-induced melanoma cell death came from cell cycle arrest and enhanced apoptosis, which were all attenuated by the Ca2+ chelator BAPTA-AM. As compared with melanoma, normal melanocytes had lower NCX1 expression and were less sensitive to the cytotoxicity of bepridil. In conclusion, blockade of the forward but not the reverse NCX leads to Ca2+-related cell death in melanoma and the NCX is a potential drug target for cancer therapy.


Asunto(s)
Bloqueadores de los Canales de Calcio/farmacología , Melanoma/metabolismo , Intercambiador de Sodio-Calcio/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Calcio/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Melanoma/etiología , Niacinamida/análogos & derivados , Niacinamida/farmacología , Tiourea/análogos & derivados , Tiourea/farmacología
4.
J Nanobiotechnology ; 19(1): 249, 2021 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-34412639

RESUMEN

BACKGROUND: Discovery of early-stage biomarkers is a long-sought goal of Alzheimer's disease (AD) diagnosis. Age is the greatest risk factor for most AD and accumulating evidence suggests that age-dependent elevation of asparaginyl endopeptidase (AEP) in the brain may represent a new biological marker for predicting AD. However, this speculation remains to be explored with an appropriate assay method because mammalian AEP exists in many organs and the level of AEP in body fluid isn't proportional to its concentration in brain parenchyma. To this end, we here modified gold nanoparticle (AuNPs) into an AEP-responsive imaging probe and choose transgenic APPswe/PS1dE9 (APP/PS1) mice as an animal model of AD. Our aim is to determine whether imaging of brain AEP can be used to predict AD pathology. RESULTS: This AEP-responsive imaging probe AuNPs-Cy5.5-A&C consisted of two particles, AuNPs-Cy5.5-AK and AuNPs-Cy5.5-CABT, which were respectively modified with Ala-Ala-Asn-Cys-Lys (AK) and 2-cyano-6-aminobenzothiazole (CABT). We showed that AuNPs-Cy5.5-A&C could be selectively activated by AEP to aggregate and emit strong fluorescence. Moreover, AuNPs-Cy5.5-A&C displayed a general applicability in various cell lines and its florescence intensity correlated well with AEP activity in these cells. In the brain of APP/PS1 transgenic mice , AEP activity was increased at an early disease stage of AD that precedes formation of senile plaques and cognitive impairment. Pharmacological inhibition of AEP with δ-secretase inhibitor 11 (10 mg kg-1, p.o.) reduced production of ß-amyloid (Aß) and ameliorated memory loss. Therefore, elevation of AEP is an early sign of AD onset. Finally, we showed that live animal imaging with this AEP-responsive probe could monitor the up-regulated AEP in the brain of APP/PS1 mice. CONCLUSIONS: The current work provided a proof of concept that assessment of brain AEP activity by in vivo imaging assay is a potential biomarker for early diagnosis of AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Biomarcadores/metabolismo , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Cisteína Endopeptidasas , Péptidos beta-Amiloides/metabolismo , Animales , Línea Celular Tumoral , Disfunción Cognitiva/metabolismo , Cisteína Endopeptidasas/genética , Modelos Animales de Enfermedad , Diagnóstico Precoz , Femenino , Glioblastoma , Glioma , Oro/metabolismo , Humanos , Masculino , Nanopartículas del Metal/química , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Placa Amiloide/metabolismo
5.
Transl Neurodegener ; 10(1): 12, 2021 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-33789744

RESUMEN

BACKGROUND: Currently, there is no cure for Alzheimer's disease (AD). Therapeutics that can modify the early stage of AD are urgently needed. Recent studies have shown that the pathogenesis of AD is closely regulated by an endo/lysosomal asparaginyl endopeptidase (AEP). Inhibition of AEP has been reported to prevent neural degeneration in transgenic mouse models of AD. However, more than 90% of AD cases are age-related sporadic AD rather than hereditary AD. The therapeutic efficacy of AEP inhibition in ageing-associated sporadic AD remains unknown. METHODS: The senescence-accelerated mouse prone 8 (SAMP8) was chosen as an approximate model of sporadic AD and treated with a selective AEP inhibitor,: δ-secretase inhibitor 11. Activation of AEP was determined by enzymatic activity assay. Concentration of soluble amyloid ß (Aß) in the brain was determined by ELISA. Morris water maze test was performed to assess the learning and memory-related cognitive ability. Pathological changes in the brain were explored by morphological and western blot analyses. RESULTS: The enzymatic activity of AEP in the SAMP8 mouse brain was significantly higher than that in the age-matched SAMR1 mice. The half maximal inhibitory concentration (IC50) for δ-secretase inhibitor 11 to inhibit AEP in vitro is was around 150 nM. Chronic treatment with δ-secretase inhibitor 11 markedly decreased the brain AEP activity, reduced the generation of Aß1-40/42 and ameliorated memory loss. The inhibition of AEP with this reagent not only reduced the AEP-cleaved tau fragments and tau hyperphosphorylation, but also attenuated neuroinflammation in the form of microglial activation. Moreover, treatment with δ-secretase inhibitor 11 prevented the synaptic loss and alleviated dendritic disruption in SAMP8 mouse brain. CONCLUSIONS: Pharmacological inhibition of AEP can intervene and prevent AD-like pathological progress in the model of sporadic AD. The up-regulated AEP in the brain could be a promising target for early treatment of AD. The δ-secretase inhibitor 11 can be used as a lead compound for translational development of AD treatment.


Asunto(s)
Envejecimiento Prematuro/tratamiento farmacológico , Envejecimiento Prematuro/patología , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/patología , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Cisteína Endopeptidasas/metabolismo , Inhibidores de Proteasas/uso terapéutico , Envejecimiento , Péptidos beta-Amiloides/análisis , Animales , Encéfalo/patología , Química Encefálica , Cognición , Cisteína Endopeptidasas/efectos de los fármacos , Humanos , Masculino , Aprendizaje por Laberinto , Memoria , Ratones
6.
Mol Psychiatry ; 26(2): 586-603, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-30382187

RESUMEN

δ-Secretase, an age-dependent asparagine protease, cleaves both amyloid precursor protein (APP) and Tau and is required for amyloid plaque and neurofibrillary tangle pathologies in Alzheimer's disease (AD). However, whether δ-secretase activation is sufficient to trigger AD pathogenesis remains unknown. Here we show that the fragments of δ-secretase-cleavage, APP (586-695) and Tau(1-368), additively drive AD pathogenesis and cognitive dysfunctions. Tau(1-368) strongly augments BACE1 expression and Aß generation in the presence of APP. The Tau(1-368) fragment is more robust than full-length Tau in binding active STAT1, a BACE1 transcription factor, and promotes its nuclear translocation, upregulating BACE1 and Aß production. Notably, Aß-activated SGK1 or JAK2 kinase phosphorylates STAT1 and induces its association with Tau(1-368). Inhibition of these kinases diminishes stimulatory effect of Tau(1-368). Knockout of STAT1 abolishes AD pathologies induced by δ-secretase-generated APP and Tau fragments. Thus, we show that Tau may not only be a downstream effector of Aß in the amyloid hypothesis, but also act as a driving force for Aß, when cleaved by δ-secretase.


Asunto(s)
Enfermedad de Alzheimer , Secretasas de la Proteína Precursora del Amiloide , Enfermedad de Alzheimer/genética , Secretasas de la Proteína Precursora del Amiloide/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Ácido Aspártico Endopeptidasas/genética , Ácido Aspártico Endopeptidasas/metabolismo , Humanos , Ovillos Neurofibrilares , Factor de Transcripción STAT1 , Proteínas tau/metabolismo
7.
World Dev ; 136: 105107, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32834388

RESUMEN

The novel coronavirus disease 2019 (COVID-19) developed into a pandemic on March 11. COVID-19 not only brought life crisis, but also incurred psychological stress: tension, anxiety, fear and despair among affected populations. How to help people overcome traumatic stress reactions and get out of psychological crisis has become a public concern that needs to be resolved in time. This article reported the psychological responses caused by the COVID-19 epidemic in China based on relevant experience and studies. The anti-epidemic measures of self-quarantine and social-distancing were deployed to contain the spread of COVID-19, but inevitably caused a certain extent of side effect: frustration and anxiety in the general public. Especially, the front-line medical rescue staff and COVID-19 patients were more susceptible to developing psychological disorders. Correspondingly, adaptive strategies and public health policies were rapidly implemented in China to deal with outbreak-caused mental stress. The psychological impact of COVID-19 and coping strategies adopted in China provided warning and reference for countries that are and going to be affected by this natural disaster.

8.
J Neurosci ; 39(33): 6571-6594, 2019 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-31263065

RESUMEN

Cell transplantation therapy provides a regenerative strategy for neural repair. We tested the hypothesis that selective excitation of transplanted induced pluripotent stem cell-derived neural progenitor cells (iPS-NPCs) could recapitulate an activity-enriched microenvironment that confers regenerative benefits for the treatment of stroke. Mouse iPS-NPCs were transduced with a novel optochemogenetics fusion protein, luminopsin 3 (LMO3), which consisted of a bioluminescent luciferase, Gaussia luciferase, and an opsin, Volvox Channelrhodopsin 1. These LMO3-iPS-NPCs can be activated by either photostimulation using light or by the luciferase substrate coelenterazine (CTZ). In vitro stimulations of LMO3-iPS-NPCs increased expression of synapsin-1, postsynaptic density 95, brain derived neurotrophic factor (BDNF), and stromal cell-derived factor 1 and promoted neurite outgrowth. After transplantation into the ischemic cortex of mice, LMO3-iPS-NPCs differentiated into mature neurons. Synapse formation between implanted and host neurons was identified using immunogold electron microscopy and patch-clamp recordings. Stimulation of transplanted cells with daily intranasal administration of CTZ enhanced axonal myelination, synaptic transmission, improved thalamocortical connectivity, and functional recovery. Patch-clamp and multielectrode array recordings in brain slices showed that CTZ or light stimulation facilitated synaptic transmission and induced neuroplasticity mimicking the LTP of EPSPs. Stroke mice received the combined LMO3-iPS-NPC/CTZ treatment, but not cell or CTZ alone, showed enhanced neural network connections in the peri-infarct region, promoted optimal functional recoveries after stroke in male and female, young and aged mice. Thus, excitation of transplanted cells via the noninvasive optochemogenetics treatment provides a novel integrative cell therapy with comprehensive regenerative benefits after stroke.SIGNIFICANCE STATEMENT Neural network reconnection is critical for repairing damaged brain. Strategies that promote this repair are expected to improve functional outcomes. This study pioneers the generation and application of an optochemogenetics approach in stem cell transplantation therapy after stroke for optimal neural repair and functional recovery. Using induced pluripotent stem cell-derived neural progenitor cells (iPS-NPCs) expressing the novel optochemogenetic probe luminopsin (LMO3), and intranasally delivered luciferase substrate coelenterazine, we show enhanced regenerative properties of LMO3-iPS-NPCs in vitro and after transplantation into the ischemic brain of different genders and ages. The noninvasive repeated coelenterazine stimulation of transplanted cells is feasible for clinical applications. The synergetic effects of the combinatorial cell therapy may have significant impacts on regenerative approach for treatments of CNS injuries.


Asunto(s)
Células-Madre Neurales/trasplante , Optogenética/métodos , Recuperación de la Función , Trasplante de Células Madre/métodos , Accidente Cerebrovascular , Animales , Diferenciación Celular/fisiología , Femenino , Células Madre Pluripotentes Inducidas/trasplante , Masculino , Ratones , Remielinización/fisiología , Transmisión Sináptica/fisiología
9.
Br J Pharmacol ; 176(15): 2691-2707, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31034096

RESUMEN

BACKGROUND AND PURPOSE: The Na+ /Ca2+ exchanger (NCX) working in either forward or reverse mode participates in maintaining intracellular Ca2+ ([Ca2+ ]i ) homeostasis, which is essential for determining cell fate. Previously, numerous blockers targeting reverse or forward NCX have been developed and studied in ischaemic tissue injury but barely examined in glioblastoma for the purpose of anti-tumour therapy. We assessed the effect of NCX blockers on glioblastoma growth and whether NCX can become a therapeutic target. EXPERIMENTAL APPROACH: Patch-clamp recording, Ca2+ imaging, flow cytometry, and Western blot were used to study the effects of specific and non-specific NCX blockers on cultured glioblastoma cells. In vivo bioluminescent imaging was used to measure effects on grafted glioblastoma. KEY RESULTS: Selectively blocking the reverse NCX with SEA0400, SN-6, and YM-244769 did not affect tumour cell viability. Blocking the forward NCX with bepridil, CB-DMB, or KB-R7943 elevated [Ca2+ ]i and killed glioblastoma cells. Bepridil and CB-DMB caused Ca2+ -dependent cell cycle arrest together with apoptosis, which were all attenuated by a Ca2+ chelator BAPTA-AM. Systemic administration of bepridil inhibited growth of brain-grafted glioblastoma. Bepridil did not appear to have a cytotoxic effect on human astrocytes, which have higher functional expression of NCX than glioblastoma cells. CONCLUSIONS AND IMPLICATIONS: Low expression of the NCX makes glioblastoma cells sensitive to disturbance of [Ca2+ ]i . Interventions designed to block the forward NCX can cause Ca2+ -mediated injury to glioblastoma thus having therapeutic potential. Bepridil could be a lead compound for developing new anti-tumour drugs.


Asunto(s)
Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Bepridil/farmacología , Bepridil/uso terapéutico , Calcio/metabolismo , Glioblastoma/tratamiento farmacológico , Intercambiador de Sodio-Calcio/antagonistas & inhibidores , Amilorida/análogos & derivados , Amilorida/farmacología , Compuestos de Anilina/farmacología , Animales , Astrocitos/efectos de los fármacos , Compuestos de Bencilo/farmacología , Ciclo Celular/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Glioblastoma/metabolismo , Humanos , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Niacinamida/análogos & derivados , Niacinamida/farmacología , Éteres Fenílicos/farmacología , Intercambiador de Sodio-Calcio/fisiología , Tiazolidinas/farmacología , Tiourea/análogos & derivados , Tiourea/farmacología
10.
Phys Chem Chem Phys ; 21(9): 4802-4809, 2019 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-30773579

RESUMEN

We present an atomistic simulation study on the compositional arrangements throughout Cu-Pt icosahedra, with a specific focus on the effects of inherent strain on general segregation trends. The coexistence of radial and site-selective segregation patterns is found in bimetallic nanoparticles for a broad range of sizes and compositions, consistent with prior analytical and atomistic models. Through a thorough comparison between the composition patterns and strain-related patterns, it is suggested that the presence of gradient and site-selective segregation is natural to largely relieve the inherent strain by preferential segregation of big atoms at tensile sites and vice versa, as previously hypothesized in the literature. Analogous to the case of single crystal particles, Cu-rich surface and damped oscillations can also be found in the outer shells of icosahedra, which are dominated by the lowering of both the surface energy and the chemical energy. The thermodynamic stability of segregated icosahedra is similar to segregated cuboctahedra but higher than disordered bulk alloys, validating prior thinking that element segregation driven by strain relief can extend the stability range of multiply-twinned nanoparticles. Our work sheds new light on understanding strain-induced segregation in multiply-twinned nanosystems that have elements with large lattice mismatch and strong alloying ability.

11.
Oncotarget ; 8(57): 97537-97553, 2017 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-29228630

RESUMEN

Ischemic stroke is a leading cause of human death and disability while clinical treatments are limited. The adult brain possesses endogenous regenerative activities that may benefit tissue repair after stroke. Trophic factors such as stromal cell-derived factor 1 alpha (SDF-1α) are upregulated in the ischemic brain, which promote endogenous regeneration. The regenerative response, however, is normally insufficient. Transplantation of exogenous cells has been explored as regenerative therapies. One promising cell type for transplantation is induced pluripotent stem (iPS) cells which are cells genetically reprogrammed from adult somatic cells. We hypothesized that transplanting neural progenitor cells derived from iPS cells (iPS-NPCs) could provide cell replacement and trophic support. The trophic factor SDF-1α was overexpressed in iPS-NPCs by lentiviral transduction to test if SDF-1α could increase regeneration in the ischemic brain. These SDF-1α-iPS-NPCs were differentiated in vitro to express mature neuronal and synaptic markers. Differentiated cells expressed functional Na+ and K+ channels, and fired action potentials. In the oxygen glucose deprivation (OGD) test, SDF-1α-iPS-NPCs survived significantly better compared to control iPS-NPCs. In mice subjected to focal cerebral ischemia in the sensorimotor cortex, iPS-NPCs and SDF-1α-iPS-NPCs were intracranially transplanted into the ischemic cortex 7 days after stroke. Neuronal differentiation of transplanted cells was identified using NeuN 14 days after transplantation. Mice that received SDF-1α-iPS-NPCs had greater numbers of NeuN/BrdU and Glut-1/BrdU co-labeled cells in the peri-infarct area and improved locomotion compared to the control iPS-NPC transplantation. Thus, SDF-1α upregulation in transplanted cells may be a therapeutic strategy to enhance endogenous neurovascular repair after ischemic stroke in adult mice.

12.
J Stroke Cerebrovasc Dis ; 26(12): 2706-2719, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29054733

RESUMEN

BACKGROUND: Stroke is a leading cause of long-term disability. All neuroprotectants targeting excitotoxicity have failed to become stroke medications. In order to explore and identify new therapeutic targets for stroke, we here reviewed present studies of ionic transporters and channels that are involved in ischemic brain damage. METHOD: We surveyed recent literature from animal experiments and clinical reports in the databases of PubMed and Elsevier ScienceDirect to analyze ionic mechanisms underlying ischemic cell damage and suggest promising ideas for stroke therapy. RESULTS: Dysfunction of ionic transporters and disrupted ionic homeostasis are most early changes that underlie ischemic brain injury, thus receiving sustained attention in translational stroke research. The Na+/K+-ATPase, Na+/Ca2+ Exchanger, ionotropic glutamate receptor, acid-sensing ion channels (ASICs), sulfonylurea receptor isoform 1 (SUR1)-regulated NCCa-ATP channels, and transient receptor potential (TRP) channels are critically involved in ischemia-induced cellular degenerating processes such as cytotoxic edema, excitotoxicity, necrosis, apoptosis, and autophagic cell death. Some ionic transporters/channels also act as signalosomes to regulate cell death signaling. For acute stroke treatment, glutamate-mediated excitotoxicity must be interfered within 2 hours after stroke. The SUR1-regulated NCCa-ATP channels, Na+/K+-ATPase, ASICs, and TRP channels have a much longer therapeutic window, providing new therapeutic targets for developing feasible pharmacological treatments toward acute ischemic stroke. CONCLUSION: The next generation of stroke therapy can apply a polypharmacology strategy for which drugs are designed to target multiple ion transporters/channels or their interaction with neurotoxic signaling pathways. But a successful translation of neuroprotectants relies on in-depth analyses of cell death mechanisms and suitable animal models resembling human stroke.


Asunto(s)
Encéfalo/efectos de los fármacos , Transporte Iónico/efectos de los fármacos , Moduladores del Transporte de Membrana/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Accidente Cerebrovascular/tratamiento farmacológico , Equilibrio Hidroelectrolítico/efectos de los fármacos , Canales Iónicos Sensibles al Ácido/efectos de los fármacos , Canales Iónicos Sensibles al Ácido/metabolismo , Adenosina Trifosfatasas/antagonistas & inhibidores , Adenosina Trifosfatasas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Encéfalo/fisiopatología , Humanos , Moduladores del Transporte de Membrana/efectos adversos , Necrosis , Fármacos Neuroprotectores/efectos adversos , Receptores Ionotrópicos de Glutamato/antagonistas & inhibidores , Receptores Ionotrópicos de Glutamato/metabolismo , Transducción de Señal/efectos de los fármacos , ATPasa Intercambiadora de Sodio-Potasio/antagonistas & inhibidores , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/patología , Accidente Cerebrovascular/fisiopatología , Receptores de Sulfonilureas/antagonistas & inhibidores , Receptores de Sulfonilureas/metabolismo , Canales Catiónicos TRPM/antagonistas & inhibidores , Canales Catiónicos TRPM/metabolismo
13.
Nat Commun ; 8: 14740, 2017 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-28345579

RESUMEN

δ-secretase, also known as asparagine endopeptidase (AEP) or legumain, is a lysosomal cysteine protease that cleaves both amyloid precursor protein (APP) and tau, mediating the amyloid-ß and tau pathology in Alzheimer's disease (AD). Here we report the therapeutic effect of an orally bioactive and brain permeable δ-secretase inhibitor in mouse models of AD. We performed a high-throughput screen and identified a non-toxic and selective δ-secretase inhibitor, termed compound 11, that specifically blocks δ-secretase but not other related cysteine proteases. Co-crystal structure analysis revealed a dual active site-directed and allosteric inhibition mode of this compound class. Chronic treatment of tau P301S and 5XFAD transgenic mice with this inhibitor reduces tau and APP cleavage, ameliorates synapse loss and augments long-term potentiation, resulting in protection of memory. Therefore, these findings demonstrate that this δ-secretase inhibitor may be an effective clinical therapeutic agent towards AD.


Asunto(s)
Enfermedad de Alzheimer/enzimología , Enfermedad de Alzheimer/psicología , Cognición/efectos de los fármacos , Cisteína Endopeptidasas/efectos de los fármacos , Modelos Animales de Enfermedad , Inhibidores de Proteasas/farmacología , Regulación Alostérica , Enfermedad de Alzheimer/tratamiento farmacológico , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Ensayos Analíticos de Alto Rendimiento , Humanos , Ratones , Ratones Transgénicos , Inhibidores de Proteasas/química , Inhibidores de Proteasas/uso terapéutico , Proteínas tau/metabolismo
14.
Neurobiol Dis ; 98: 9-24, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27884724

RESUMEN

Neurogenesis in the subventricular zone (SVZ) of the adult brain may contribute to tissue repair after brain injuries. Whether SVZ neurogenesis can be upregulated by specific neuronal activity in vivo and promote functional recovery after stroke is largely unknown. Using the spatial and cell type specific optogenetic technique combined with multiple approaches of in vitro, ex vivo and in vivo examinations, we tested the hypothesis that glutamatergic activation in the striatum could upregulate SVZ neurogenesis in the normal and ischemic brain. In transgenic mice expressing the light-gated channelrhodopsin-2 (ChR2) channel in glutamatergic neurons, optogenetic stimulation of the glutamatergic activity in the striatum triggered glutamate release into SVZ region, evoked membrane currents, Ca2+ influx and increased proliferation of SVZ neuroblasts, mediated by AMPA receptor activation. In ChR2 transgenic mice subjected to focal ischemic stroke, optogenetic stimuli to the striatum started 5days after stroke for 8days not only promoted cell proliferation but also the migration of SVZ neuroblasts into the peri-infarct cortex with increased neuronal differentiation and improved long-term functional recovery. These data provide the first morphological and functional evidence showing a unique striatum-SVZ neuronal regulation via a semi-phasic synaptic mechanism that can boost neurogenic cascades and stroke recovery. The benefits from stimulating endogenous glutamatergic activity suggest a novel regenerative strategy after ischemic stroke and other brain injuries.


Asunto(s)
Cuerpo Estriado/metabolismo , Ácido Glutámico/metabolismo , Neurogénesis/fisiología , Neuronas/metabolismo , Nicho de Células Madre/fisiología , Animales , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Células Cultivadas , Cuerpo Estriado/citología , Cuerpo Estriado/patología , Modelos Animales de Enfermedad , Ventrículos Laterales , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Vías Nerviosas/citología , Vías Nerviosas/metabolismo , Vías Nerviosas/patología , Neuronas/citología , Neuronas/patología , Optogenética , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/patología , Técnicas de Cultivo de Tejidos
15.
Stem Cell Reports ; 5(6): 1239-1249, 2015 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-26651608

RESUMEN

Isolation of ventricular cardiomyocytes (vCMs) has been challenging due to the lack of specific surface markers. Here we show that vCMs can be purified from differentiating mouse embryonic stem cells (mESCs) using molecular beacons (MBs) targeting specific intracellular mRNAs. We designed MBs (IRX4 MBs) to target mRNA encoding Iroquois homeobox protein 4 (Irx4), a transcription factor specific for vCMs. To purify mESC vCMs, IRX4 MBs were delivered into cardiomyogenically differentiating mESCs, and IRX4 MBs-positive cells were FACS-sorted. We found that, of the cells isolated, ~98% displayed vCM-like action potentials by electrophysiological analyses. These MB-purified vCMs continuously maintained their CM characteristics as verified by spontaneous beating, Ca(2+) transient, and expression of vCM-specific proteins. Our study shows the feasibility of isolating pure vCMs via cell sorting without modifying host genes. The homogeneous and functional ventricular CMs generated via the MB-based method can be useful for disease investigation, drug discovery, and cell-based therapies.


Asunto(s)
Separación Celular/métodos , Células Madre Embrionarias/citología , Ventrículos Cardíacos/citología , Proteínas de Homeodominio/genética , Miocitos Cardíacos/citología , Potenciales de Acción , Animales , Secuencia de Bases , Diferenciación Celular , Células Cultivadas , Citometría de Flujo , Ratones , Sondas de Oligonucleótidos/genética , ARN Mensajero/genética
16.
Nat Commun ; 6: 8762, 2015 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-26549211

RESUMEN

The age-dependent deposition of amyloid-ß peptides, derived from amyloid precursor protein (APP), is a neuropathological hallmark of Alzheimer's disease (AD). Despite age being the greatest risk factor for AD, the molecular mechanisms linking ageing to APP processing are unknown. Here we show that asparagine endopeptidase (AEP), a pH-controlled cysteine proteinase, is activated during ageing and mediates APP proteolytic processing. AEP cleaves APP at N373 and N585 residues, selectively influencing the amyloidogenic fragmentation of APP. AEP is activated in normal mice in an age-dependent manner, and is strongly activated in 5XFAD transgenic mouse model and human AD brains. Deletion of AEP from 5XFAD or APP/PS1 mice decreases senile plaque formation, ameliorates synapse loss, elevates long-term potentiation and protects memory. Blockade of APP cleavage by AEP in mice alleviates pathological and behavioural deficits. Thus, AEP acts as a δ-secretase, contributing to the age-dependent pathogenic mechanisms in AD.


Asunto(s)
Envejecimiento , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Encéfalo/metabolismo , Cisteína Endopeptidasas/genética , Hipocampo/metabolismo , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/enzimología , Secretasas de la Proteína Precursora del Amiloide/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Western Blotting , Encéfalo/patología , Cisteína Endopeptidasas/metabolismo , Sinapsis Eléctricas , Ensayo de Inmunoadsorción Enzimática , Femenino , Técnica del Anticuerpo Fluorescente , Células HEK293 , Hipocampo/fisiopatología , Humanos , Inmunohistoquímica , Inmunoprecipitación , Potenciación a Largo Plazo , Masculino , Espectrometría de Masas , Memoria , Ratones , Ratones Noqueados , Ratones Transgénicos , Microscopía Electrónica , Persona de Mediana Edad , Placa Amiloide/metabolismo
17.
Nat Med ; 20(11): 1254-62, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25326800

RESUMEN

Neurofibrillary tangles (NFTs), composed of truncated and hyperphosphorylated tau, are a common feature of numerous aging-related neurodegenerative diseases, including Alzheimer's disease (AD). However, the molecular mechanisms mediating tau truncation and aggregation during aging remain elusive. Here we show that asparagine endopeptidase (AEP), a lysosomal cysteine proteinase, is activated during aging and proteolytically degrades tau, abolishes its microtubule assembly function, induces tau aggregation and triggers neurodegeneration. AEP is upregulated and active during aging and is activated in human AD brain and tau P301S-transgenic mice with synaptic pathology and behavioral impairments, leading to tau truncation in NFTs. Tau P301S-transgenic mice with deletion of the gene encoding AEP show substantially reduced tau hyperphosphorylation, less synapse loss and rescue of impaired hippocampal synaptic function and cognitive deficits. Mice infected with adeno-associated virus encoding an uncleavable tau mutant showed attenuated pathological and behavioral defects compared to mice injected with adeno-associated virus encoding tau P301S. Together, these observations indicate that AEP acts as a crucial mediator of tau-related clinical and neuropathological changes. Inhibition of AEP may be therapeutically useful for treating tau-mediated neurodegenerative diseases.


Asunto(s)
Enfermedad de Alzheimer/enzimología , Enfermedad de Alzheimer/patología , Cisteína Endopeptidasas/metabolismo , Ovillos Neurofibrilares/enzimología , Ovillos Neurofibrilares/patología , Proteínas tau/metabolismo , Anciano , Envejecimiento/patología , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/fisiopatología , Secuencia de Aminoácidos , Animales , Asparagina/metabolismo , Encéfalo/enzimología , Encéfalo/patología , Calpaína/metabolismo , Caspasas/metabolismo , Cognición , Técnicas de Inactivación de Genes , Células HEK293 , Humanos , Trastornos de la Memoria/complicaciones , Trastornos de la Memoria/patología , Trastornos de la Memoria/fisiopatología , Ratones Transgénicos , Datos de Secuencia Molecular , Neurotoxinas/metabolismo , Fosforilación , Estructura Terciaria de Proteína , Solubilidad , Sinapsis/metabolismo , Sinapsis/patología , Regulación hacia Arriba , Proteínas tau/química
18.
BMC Cancer ; 14: 716, 2014 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-25255962

RESUMEN

BACKGROUND: Glioblastoma multiforme (GBM) is very difficult to treat with conventional anti-cancer/anti-apoptotic drugs. We tested the hypothesis that inhibition of Na+/K+-ATPase causes a mixed or hybrid form of concurrent apoptosis and necrosis and therefore should enhance anti-cancer effects of chemotherapy on glioblastoma cells. METHODS: In human LN229 and drug-resistant T98G glioblastoma cell cultures, cell death and signal pathways were measured using immunocytochemistry and Western blotting. Fluorescent dyes were applied to measure intracellular Ca2+, Na+ and K+ changes. RESULTS: The specific Na+/K+-ATPase blocker ouabain (0.1 - 10 µM) induced cell death and disruption of K+ homeostasis in a time- and concentration-dependent manner. Annexin-V translocation and caspase-3 activation indicated an apoptotic component in ouabain cytoxicity, which was accompanied with reduced Bcl-2 expression and mitochondrial membrane potential. Ouabain-induced cell death was partially attenuated by the caspase inhibitor Z-VAD (100 µM). Consistently, the K+ ionophore valinomycin initiated apoptosis in LN229 cells in a K+ efflux-dependent manner. Ouabain caused an initial cell swell, which was followed by a sustained cell volume decrease. Electron microscopy revealed ultrastructural features of both apoptotic and necrotic alterations in the same cells. Finally, human T98G glioblastoma cells that are resistant to the chemotherapy drug temozolomide (TMZ) showed a unique high expression of the Na+/K+-ATPase α2 and α3 subunits compared to the TMZ-sensitive cell line LN229 and normal human astrocytes. At low concentrations, ouabain selectively killed T98G cells. Knocking down the α3 subunit sensitized T98G cells to TMZ and caused more cell death. CONCLUSION: This study suggests that inhibition of Na+/K+-ATPase triggers hybrid cell death and serves as an underlying mechanism for an enhanced chemotherapy effect on glioblastoma cells.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Dacarbazina/análogos & derivados , Ouabaína/farmacología , ATPasa Intercambiadora de Sodio-Potasio/antagonistas & inhibidores , Neoplasias Encefálicas/tratamiento farmacológico , Calcio/metabolismo , Línea Celular Tumoral , Dacarbazina/farmacología , Resistencia a Antineoplásicos , Ensayos de Selección de Medicamentos Antitumorales , Técnicas de Silenciamiento del Gen , Glioblastoma/tratamiento farmacológico , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Sodio/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/genética , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Temozolomida , Valinomicina/farmacología
19.
Stem Cells ; 32(12): 3075-87, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25132189

RESUMEN

Limited treatments are available for perinatal/neonatal stroke. Induced pluripotent stem cells (iPSCs) hold therapeutic promise for stroke treatment, but the benefits of iPSC transplantation in neonates are relatively unknown. We hypothesized that transplanted iPSC-derived neural progenitor cells (iPSC-NPCs) would increase regeneration after stroke. Mouse pluripotent iPSCs were differentiated into neural progenitors using a retinoic acid protocol. Differentiated neural cells were characterized by using multiple criteria and assessments. Ischemic stroke was induced in postnatal day 7 (P7) rats by occluding the right middle cerebral artery and right common carotid artery. iPSC-NPCs (400,000 in 4 µl) were transplanted into the penumbra via intracranial injection 7 days after stroke. Trophic factor expression in the peri-infarct tissue was measured using Western blot analysis. Animals received daily bromodeoxyuridine (BrdU) injections and were sacrificed 21 days after stroke for immunohistochemistry. The vibrissae-elicited forelimb placement test was used to evaluate functional recovery. Differentiated iPSCs expressed mature neuronal markers, functional sodium and potassium channels, and fired action potentials. Several angiogenic and neurogenic trophic factors were identified in iPSC-NPCs. Animals that received iPSC-NPC transplantation had greater expression of stromal cell-derived factor 1-α (SDF-1α) and vascular endothelial growth factor (VEGF) in the peri-infarct region. iPSC-NPCs stained positive for neuronal nuclei (NeuN) or glial fibrillary acidic protein (GFAP) 14 days after transplantation. iPSC-NPC-transplanted animals showed greater numbers of BrdU/NeuN and BrdU/Collagen IV colabeled cells in the peri-infarct area compared with stroke controls and performed better in a sensorimotor functional test after stroke. iPSC-NPC therapy may play multiple therapeutic roles after stroke by providing trophic factors, increasing angiogenesis and neurogenesis, and providing new cells for tissue repair.


Asunto(s)
Isquemia Encefálica/terapia , Células Madre Pluripotentes Inducidas/citología , Células-Madre Neurales/citología , Recuperación de la Función/fisiología , Accidente Cerebrovascular/terapia , Animales , Animales Recién Nacidos , Diferenciación Celular/fisiología , Células Cultivadas , Modelos Animales de Enfermedad , Células Madre Pluripotentes Inducidas/trasplante , Ratones , Neurogénesis/fisiología , Ratas , Factor A de Crecimiento Endotelial Vascular/metabolismo
20.
Neuropsychopharmacology ; 39(3): 638-50, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24022672

RESUMEN

Synaptic loss in the brain correlates well with disease severity in Alzheimer disease (AD). Deficits in brain-derived neurotrophic factor/tropomyosin-receptor-kinase B (TrkB) signaling contribute to the synaptic dysfunction of AD. We have recently identified 7,8-dihydroxyflavone (7,8-DHF) as a potent TrkB agonist that displays therapeutic efficacy toward various neurological diseases. Here we tested the effect of 7,8-DHF on synaptic function in an AD model both in vitro and in vivo. 7,8-DHF protected primary neurons from Aß-induced toxicity and promoted dendrite branching and synaptogenesis. Chronic oral administration of 7,8-DHF activated TrkB signaling and prevented Aß deposition in transgenic mice that coexpress five familial Alzheimer's disease mutations (5XFAD mice). Moreover, 7,8-DHF inhibited the loss of hippocampal synapses, restored synapse number and synaptic plasticity, and prevented memory deficits. These results suggest that 7,8-DHF represents a novel oral bioactive therapeutic agent for treating AD.


Asunto(s)
Enfermedad de Alzheimer , Antipsicóticos/uso terapéutico , Apoptosis/efectos de los fármacos , Flavonas/uso terapéutico , Trastornos de la Memoria/etiología , Trastornos de la Memoria/prevención & control , Sinapsis/patología , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Animales , Células Cultivadas , Corteza Cerebral/citología , Modelos Animales de Enfermedad , Femenino , Hipocampo/patología , Humanos , Técnicas In Vitro , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación/genética , Neuronas/efectos de los fármacos , Neuronas/patología , Neuronas/ultraestructura , Placa Amiloide/patología , Sinapsis/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...