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













Base de datos
Intervalo de año de publicación
1.
Hepatology ; 2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38563629

RESUMEN

BACKGROUND AND AIMS: Fibrosis is the common end point for all forms of chronic liver injury, and the progression of fibrosis leads to the development of end-stage liver disease. Activation of HSCs and their transdifferentiation into myofibroblasts results in the accumulation of extracellular matrix proteins that form the fibrotic scar. Long noncoding RNAs regulate the activity of HSCs and provide targets for fibrotic therapies. APPROACH AND RESULTS: We identified long noncoding RNA TILAM located near COL1A1 , expressed in HSCs, and induced with liver fibrosis in humans and mice. Loss-of-function studies in human HSCs and human liver organoids revealed that TILAM regulates the expression of COL1A1 and other extracellular matrix genes. To determine the role of TILAM in vivo, we annotated the mouse ortholog ( Tilam ), generated Tilam- deficient green fluorescent protein-reporter mice, and challenged these mice in 2 different models of liver fibrosis. Single-cell data and analysis of single-data and analysis of Tilam-deficient reporter mice revealed that Tilam is induced in murine HSCs with the development of fibrosis in vivo. Tilam -deficient reporter mice revealed that Tilam is induced in murine HSCs with the development of fibrosis in vivo. Furthermore, loss of Tilam expression attenuated the development of fibrosis in the setting of in vivo liver injury. Finally, we found that TILAM interacts with promyelocytic leukemia nuclear body scaffold protein to regulate a feedback loop by which TGF-ß2 reinforces TILAM expression and nuclear localization of promyelocytic leukemia nuclear body scaffold protein to promote the fibrotic activity of HSCs. CONCLUSIONS: TILAM is activated in HSCs with liver injury and interacts with promyelocytic leukemia nuclear body scaffold protein to drive the development of fibrosis. Depletion of TILAM may serve as a therapeutic approach to combat the development of end-stage liver disease.

2.
Neuron ; 111(21): 3378-3396.e9, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37657442

RESUMEN

A genetically valid animal model could transform our understanding of schizophrenia (SCZ) disease mechanisms. Rare heterozygous loss-of-function (LoF) mutations in GRIN2A, encoding a subunit of the NMDA receptor, greatly increase the risk of SCZ. By transcriptomic, proteomic, and behavioral analyses, we report that heterozygous Grin2a mutant mice show (1) large-scale gene expression changes across multiple brain regions and in neuronal (excitatory and inhibitory) and non-neuronal cells (astrocytes and oligodendrocytes), (2) evidence of hypoactivity in the prefrontal cortex (PFC) and hyperactivity in the hippocampus and striatum, (3) an elevated dopamine signaling in the striatum and hypersensitivity to amphetamine-induced hyperlocomotion (AIH), (4) altered cholesterol biosynthesis in astrocytes, (5) a reduction in glutamatergic receptor signaling proteins in the synapse, and (6) an aberrant locomotor pattern opposite of that induced by antipsychotic drugs. These findings reveal potential pathophysiologic mechanisms, provide support for both the "hypo-glutamate" and "hyper-dopamine" hypotheses of SCZ, and underscore the utility of Grin2a-deficient mice as a genetic model of SCZ.


Asunto(s)
Dopamina , Proteómica , Receptores de N-Metil-D-Aspartato , Animales , Ratones , Encéfalo/metabolismo , Dopamina/metabolismo , Neuroglía/metabolismo , Neuronas/metabolismo , Corteza Prefrontal/metabolismo , Modelos Animales de Enfermedad , Receptores de N-Metil-D-Aspartato/genética
3.
Elife ; 112022 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-35617485

RESUMEN

Chronic liver injury causes fibrosis, characterized by the formation of scar tissue resulting from excessive accumulation of extracellular matrix (ECM) proteins. Hepatic stellate cell (HSC) myofibroblasts are the primary cell type responsible for liver fibrosis, yet there are currently no therapies directed at inhibiting the activity of HSC myofibroblasts. To search for potential anti-fibrotic compounds, we performed a high-throughput compound screen in primary human HSC myofibroblasts and identified 19 small molecules that induce HSC inactivation, including the polyether ionophore nanchangmycin (NCMC). NCMC induces lipid re-accumulation while reducing collagen expression, deposition of collagen in the extracellular matrix, cell proliferation, and migration. We find that NCMC increases cytosolic Ca2+ and reduces the phosphorylated protein levels of FYN, PTK2 (FAK), MAPK1/3 (ERK2/1), HSPB1 (HSP27), and STAT5B. Further, depletion of each of these kinases suppress COL1A1 expression. These studies reveal a signaling network triggered by NCMC to inactivate HSC myofibroblasts and reduce expression of proteins that compose the fibrotic scar. Identification of the antifibrotic effects of NCMC and the elucidation of pathways by which NCMC inhibits fibrosis provide new tools and therapeutic targets that could potentially be utilized to combat the development and progression of liver fibrosis.


Asunto(s)
Cicatriz , Células Estrelladas Hepáticas , Cicatriz/patología , Colágeno/metabolismo , Éteres , Proteínas de la Matriz Extracelular/metabolismo , Fibrosis , Quinasa 1 de Adhesión Focal/metabolismo , Células Estrelladas Hepáticas/metabolismo , Humanos , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Compuestos de Espiro
4.
ACS Pharmacol Transl Sci ; 5(3): 156-168, 2022 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-35311021

RESUMEN

T-type voltage-gated Ca2+ channels have been implicated in many human disorders, and there has been increasing interest in developing highly selective and potent T-type Ca2+ channel modulators for potential clinical use. However, the unique biophysical properties of T-type Ca2+ channels are not conducive for developing high-throughput screening (HTS) assays to identify modulators, particularly potentiators. To illustrate, T-type Ca2+ channels are largely inactivated and unable to open to allow Ca2+ influx at -25 mV, the typical resting membrane potential of the cell lines commonly used in cellular screening assays. To address this issue, we developed cell lines that express Kir2.3 channels to hyperpolarize the membrane potential to -70 mV, thus allowing T-type channels to return to their resting state where they can be subsequently activated by membrane depolarization in the presence of extracellular KCl. Furthermore, to simplify the HTS assay and to reduce reagent cost, we stably expressed a membrane-tethered genetic calcium sensor, GCaMP6s-CAAX, that displays superior signal to the background compared to the untethered GCaMP6s or the synthetic Ca2+ sensor Fluo-4AM. Here, we describe a novel GCaMP6s-CAAX-based calcium assay utilizing a high-throughput fluorometric imaging plate reader (Molecular Devices, Sunnyvale, CA) format that can identify both activators and inhibitors of T-type Ca2+ channels. Lastly, we demonstrate the utility of this novel fluorescence-based assay to evaluate the activities of two distinct G-protein-coupled receptors, thus expanding the use of GCaMP6s-CAAX to a wide range of applications relevant for developing cellular assays in drug discovery.

5.
Aging Brain ; 22022.
Artículo en Inglés | MEDLINE | ID: mdl-36589695

RESUMEN

With the ultimate goal of developing a more representative animal model of Alzheimer's disease (AD), two female amyloid-ß-(Aß) precursor protein-transgenic (APPtg) rhesus monkeys were generated by lentiviral transduction of the APP gene into rhesus oocytes, followed by in vitro fertilization and embryo transfer. The APP-transgene included the AD-associated Swedish K670N/M671L and Indiana V717F mutations (APPSWE/IND) regulated by the human polyubiquitin-C promoter. Overexpression of APP was confirmed in lymphocytes and brain tissue. Upon sacrifice at 10 years of age, one of the monkeys had developed Aß plaques and cerebral Aß-amyloid angiopathy in the occipital, parietal, and caudal temporal neocortices. The induction of Aß deposition more than a decade prior to its usual emergence in the rhesus monkey supports the feasibility of creating a transgenic nonhuman primate model for mechanistic analyses and preclinical testing of treatments for Alzheimer's disease and cerebrovascular amyloidosis.

6.
Nat Cell Biol ; 22(10): 1211-1222, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32895492

RESUMEN

Cooperation between DNA, RNA and protein regulates gene expression and controls differentiation through interactions that connect regions of nucleic acids and protein domains and through the assembly of biomolecular condensates. Here, we report that endoderm differentiation is regulated by the interaction between the long non-coding RNA (lncRNA) DIGIT and the bromodomain and extraterminal domain protein BRD3. BRD3 forms phase-separated condensates of which the formation is promoted by DIGIT, occupies enhancers of endoderm transcription factors and is required for endoderm differentiation. BRD3 binds to histone H3 acetylated at lysine 18 (H3K18ac) in vitro and co-occupies the genome with H3K18ac. DIGIT is also enriched in regions of H3K18ac, and the depletion of DIGIT results in decreased recruitment of BRD3 to these regions. Our findings show that cooperation between DIGIT and BRD3 at regions of H3K18ac regulates the transcription factors that drive endoderm differentiation and suggest that protein-lncRNA phase-separated condensates have a broader role as regulators of transcription.


Asunto(s)
Diferenciación Celular , Endodermo/citología , Histonas/metabolismo , Células Madre Embrionarias Humanas/citología , Transición de Fase , ARN Largo no Codificante/genética , Factores de Transcripción/metabolismo , Acetilación , Endodermo/metabolismo , Genoma Humano , Histonas/genética , Células Madre Embrionarias Humanas/metabolismo , Humanos , Lisina/genética , Lisina/metabolismo , Dominios Proteicos , Procesamiento Proteico-Postraduccional , Factores de Transcripción/genética
7.
Nat Chem Biol ; 16(3): 226-227, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32080631
8.
Sci Signal ; 12(610)2019 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-31796631

RESUMEN

Highly selective, positive allosteric modulators (PAMs) of the M1 subtype of muscarinic acetylcholine receptor have emerged as an exciting new approach to potentially improve cognitive function in patients suffering from Alzheimer's disease and schizophrenia. Discovery programs have produced a structurally diverse range of M1 receptor PAMs with distinct pharmacological properties, including different extents of agonist activity and differences in signal bias. This includes biased M1 receptor PAMs that can potentiate coupling of the receptor to activation of phospholipase C (PLC) but not phospholipase D (PLD). However, little is known about the role of PLD in M1 receptor signaling in native systems, and it is not clear whether biased M1 PAMs display differences in modulating M1-mediated responses in native tissue. Using PLD inhibitors and PLD knockout mice, we showed that PLD was necessary for the induction of M1-dependent long-term depression (LTD) in the prefrontal cortex (PFC). Furthermore, biased M1 PAMs that did not couple to PLD not only failed to potentiate orthosteric agonist-induced LTD but also blocked M1-dependent LTD in the PFC. In contrast, biased and nonbiased M1 PAMs acted similarly in potentiating M1-dependent electrophysiological responses that were PLD independent. These findings demonstrate that PLD plays a critical role in the ability of M1 PAMs to modulate certain central nervous system (CNS) functions and that biased M1 PAMs function differently in brain regions implicated in cognition.


Asunto(s)
Corteza Cerebral/enzimología , Plasticidad Neuronal , Fosfolipasa D/genética , Fosfolipasa D/metabolismo , Receptor Muscarínico M1/genética , Receptor Muscarínico M1/metabolismo , Sitio Alostérico , Animales , Células CHO , Calcio/química , Cognición , Cricetinae , Cricetulus , Electrofisiología , Femenino , Humanos , Depresión Sináptica a Largo Plazo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Corteza Prefrontal/enzimología , Transducción de Señal , Fosfolipasas de Tipo C/metabolismo
9.
Trends Pharmacol Sci ; 40(12): 1006-1020, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31711626

RESUMEN

Muscarinic acetylcholine receptors (mAChR) play important roles in regulating complex behaviors such as cognition, movement, and reward, making them ideally situated as potential drug targets for the treatment of several brain disorders. Recent advances in the discovery of subtype-selective allosteric modulators for mAChRs has provided an unprecedented opportunity for highly specific modulation of signaling by individual mAChR subtypes in the brain. Recently, mAChR allosteric modulators have entered clinical development for Alzheimer's disease (AD) and schizophrenia, and have potential utility for other brain disorders. However, mAChR allosteric modulators can display a diverse array of pharmacological properties, and a more nuanced understanding of the mAChR will be necessary to best translate preclinical findings into successful clinical treatments.


Asunto(s)
Trastornos Mentales/tratamiento farmacológico , Agonistas Muscarínicos/farmacología , Antagonistas Muscarínicos/farmacología , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Receptores Muscarínicos/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Ensayos Clínicos como Asunto , Humanos , Trastornos Mentales/metabolismo , Terapia Molecular Dirigida , Agonistas Muscarínicos/uso terapéutico , Antagonistas Muscarínicos/uso terapéutico , Enfermedades del Sistema Nervioso/metabolismo , Esquizofrenia/tratamiento farmacológico , Esquizofrenia/metabolismo
10.
Biol Psychiatry ; 85(12): 989-1000, 2019 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-31003787

RESUMEN

BACKGROUND: The prefrontal cortex (PFC) integrates information from multiple inputs to exert top-down control allowing for appropriate responses in a given context. In psychiatric disorders such as posttraumatic stress disorder, PFC hyperactivity is associated with inappropriate fear in safe situations. We previously reported a form of muscarinic acetylcholine receptor (mAChR)-dependent long-term depression in the PFC that we hypothesize is involved in appropriate fear responding and could serve to reduce cortical hyperactivity following stress. However, it is unknown whether this long-term depression occurs at fear-related inputs. METHODS: Using optogenetics with extracellular and whole-cell electrophysiology, we assessed the effect of mAChR activation on the synaptic strength of specific PFC inputs. We used selective pharmacological tools to assess the involvement of M1 mAChRs in conditioned fear extinction in control mice and in the stress-enhanced fear-learning model. RESULTS: M1 mAChR activation induced long-term depression at inputs from the ventral hippocampus and basolateral amygdala but not from the mediodorsal nucleus of the thalamus. We found that systemic M1 mAChR antagonism impaired contextual fear extinction. Treatment with an M1 positive allosteric modulator enhanced contextual fear extinction consolidation in stress-enhanced fear learning-conditioned mice. CONCLUSIONS: M1 mAChRs dynamically modulate synaptic transmission at two PFC inputs whose activity is necessary for fear extinction, and M1 mAChR function is required for proper contextual fear extinction. Furthermore, an M1 positive allosteric modulator enhanced the consolidation of fear extinction in the stress-enhanced fear-learning model, suggesting that M1 positive allosteric modulators may provide a novel treatment strategy to facilitate exposure therapy in the clinic for the treatment of posttraumatic stress disorder.


Asunto(s)
Miedo/fisiología , Neuronas/fisiología , Corteza Prefrontal/fisiología , Receptor Muscarínico M1/fisiología , Trastornos por Estrés Postraumático/fisiopatología , Animales , Complejo Nuclear Basolateral/fisiología , Condicionamiento Clásico , Extinción Psicológica/fisiología , Hipocampo/fisiología , Depresión Sináptica a Largo Plazo , Núcleo Talámico Mediodorsal/fisiología , Ratones Endogámicos C57BL , Vías Nerviosas/fisiología , Transmisión Sináptica/fisiología
11.
ACS Chem Neurosci ; 9(9): 2274-2285, 2018 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-29701957

RESUMEN

Selective activation of the M1 subtype of muscarinic acetylcholine receptor, via positive allosteric modulation (PAM), is an exciting strategy to improve cognition in schizophrenia and Alzheimer's disease patients. However, highly potent M1 ago-PAMs, such as MK-7622, PF-06764427, and PF-06827443, can engender excessive activation of M1, leading to agonist actions in the prefrontal cortex (PFC) that impair cognitive function, induce behavioral convulsions, and result in other classic cholinergic adverse events (AEs). Here, we report a fundamentally new and highly selective M1 PAM, VU0486846. VU0486846 possesses only weak agonist activity in M1-expressing cell lines with high receptor reserve and is devoid of agonist actions in the PFC, unlike previously reported ago-PAMs MK-7622, PF-06764427, and PF-06827443. Moreover, VU0486846 shows no interaction with antagonist binding at the orthosteric acetylcholine (ACh) site (e.g., neither bitopic nor displaying negative cooperativity with [3H]-NMS binding at the orthosteric site), no seizure liability at high brain exposures, and no cholinergic AEs. However, as opposed to ago-PAMs, VU0486846 produces robust efficacy in the novel object recognition model of cognitive function. Importantly, we show for the first time that an M1 PAM can reverse the cognitive deficits induced by atypical antipsychotics, such as risperidone. These findings further strengthen the argument that compounds with modest in vitro M1 PAM activity (EC50 > 100 nM) and pure-PAM activity in native tissues display robust procognitive efficacy without AEs mediated by excessive activation of M1. Overall, the combination of compound assessment with recombinant in vitro assays (mindful of receptor reserve), native tissue systems (PFC), and phenotypic screens (behavioral convulsions) is essential to fully understand and evaluate lead compounds and enhance success in clinical development.


Asunto(s)
Cognición/efectos de los fármacos , Condicionamiento Psicológico/efectos de los fármacos , Conducta Exploratoria/efectos de los fármacos , Morfolinas/farmacología , Corteza Prefrontal/efectos de los fármacos , Pirazoles/farmacología , Regulación Alostérica , Animales , Antipsicóticos/toxicidad , Células CHO , Disfunción Cognitiva/inducido químicamente , Disfunción Cognitiva/fisiopatología , Cricetulus , Miedo , Ratones , Morfolinas/toxicidad , Pirazoles/toxicidad , Ratas , Risperidona/toxicidad , Convulsiones/inducido químicamente
12.
ACS Chem Neurosci ; 9(9): 2218-2224, 2018 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-29683646

RESUMEN

Positive allosteric modulators (PAMs) of the M1 subtype of muscarinic acetylcholine receptor have attracted intense interest as an exciting new approach for improving the cognitive deficits in schizophrenia and Alzheimer's disease. Recent evidence suggests that the presence of intrinsic agonist activity of some M1 PAMs may reduce efficacy and contribute to adverse effect liability. However, the M1 PAM PF-06827443 was reported to have only weak agonist activity at human M1 receptors but produced M1-dependent adverse effects. We now report that PF-06827443 is an allosteric agonist in cell lines expressing rat, dog, and human M1 and use of inducible cell lines shows that agonist activity of PF-06827443 is dependent on receptor reserve. Furthermore, PF-06827443 is an agonist in native tissue preparations and induces behavioral convulsions in mice similar to other ago-PAMs. These findings suggest that PF-06827443 is a robust ago-PAM, independent of species, in cell lines and native systems.


Asunto(s)
Isoindoles/farmacología , Oxazoles/farmacología , Corteza Prefrontal/efectos de los fármacos , Receptor Muscarínico M1/agonistas , Convulsiones/inducido químicamente , Convulsiones/metabolismo , Regulación Alostérica , Animales , Células CHO , Calcio/metabolismo , Cricetulus , Perros , Humanos , Ratones , Técnicas de Placa-Clamp , Corteza Prefrontal/metabolismo , Ratas
13.
Neuropsychopharmacology ; 43(8): 1763-1771, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29581537

RESUMEN

Highly selective positive allosteric modulators (PAMs) of the M1 subtype of muscarinic acetylcholine receptor have emerged as an exciting new approach for improving cognitive function in patients suffering from Alzheimer's disease and schizophrenia. However, excessive activation of M1 is known to induce seizure activity and have actions in the prefrontal cortex (PFC) that could impair cognitive function. We now report a series of pharmacological, electrophysiological, and behavioral studies in which we find that recently reported M1 PAMs, PF-06764427 and MK-7622, have robust agonist activity in cell lines and agonist effects in the mouse PFC, and have the potential to overactivate the M1 receptor and disrupt PFC function. In contrast, structurally distinct M1 PAMs (VU0453595 and VU0550164) are devoid of agonist activity in cell lines and maintain activity dependence of M1 activation in the PFC. Consistent with the previously reported effect of PF-06764427, the ago-PAM MK-7622 induces severe behavioral convulsions in mice. In contrast, VU0453595 does not induce behavioral convulsions at doses well above those required for maximal efficacy in enhancing cognitive function. Furthermore, in contrast to the robust efficacy of VU0453595, the ago-PAM MK-7622 failed to improve novel object recognition, a rodent assay of cognitive function. These findings suggest that in vivo cognition-enhancing efficacy of M1 PAMs can be observed with PAMs lacking intrinsic agonist activity and that intrinsic agonist activity of M1 PAMs may contribute to adverse effects and reduced efficacy in improving cognitive function.


Asunto(s)
Colinérgicos/farmacología , Nootrópicos/farmacología , Receptor Muscarínico M1/metabolismo , Regulación Alostérica , Animales , Células CHO , Cricetulus , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Masculino , Ratones Endogámicos C57BL , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/fisiología , Células Piramidales/efectos de los fármacos , Células Piramidales/fisiología , Ratas , Ratas Sprague-Dawley , Receptor Muscarínico M1/agonistas , Receptor Muscarínico M1/genética , Reconocimiento en Psicología/efectos de los fármacos , Técnicas de Cultivo de Tejidos
14.
ACS Chem Neurosci ; 8(10): 2254-2265, 2017 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-28679049

RESUMEN

Selective potentiation of the mGlu5 subtype of metabotropic glutamate (mGlu) receptor using positive allosteric modulators (PAMs) has robust cognition-enhancing effects in rodent models that are relevant for schizophrenia. Until recently, these effects were thought to be due to potentiation of mGlu5-induced modulation of NMDA receptor (NMDAR) currents and NMDAR-dependent synaptic plasticity. However, "biased" mGlu5 PAMs that do not potentiate mGlu5 effects on NMDAR currents show efficacy that is similar to that of prototypical mGlu5 PAMs, suggesting that NMDAR-independent mechanisms must be involved in these actions. We now report that synaptic activation of mGlu5 is required for a form of long-term depression (mLTD) in mouse prefrontal cortex (PFC) that is induced by activation of M1 muscarinic acetylcholine (mAChR) receptors, which was previously thought to be independent of mGlu5 activation. Interestingly, a biased mGlu5 PAM, VU0409551, that does not potentiate mGlu5 modulation of NMDAR currents, potentiated induction of mLTD. Furthermore, coactivation of mGlu5 and M1 receptors increased GABAA-dependent inhibitory tone in the PFC pyramidal neurons, which likely contributes to the observed mLTD. Finally, systemic administration of the biased mGlu5 PAM reversed deficits in mLTD and associated cognitive deficits in a model of cortical disruption caused by repeated phencyclidine exposure that is relevant for schizophrenia and was previously shown to be responsive to selective M1 muscarinic receptor PAMs. These studies provide exciting new insights into a novel mechanism by which mGlu5 PAMs can reverse deficits in PFC function and cognition that is independent of modulation of NMDAR currents.


Asunto(s)
Colinérgicos/farmacología , Corteza Prefrontal/efectos de los fármacos , Receptor del Glutamato Metabotropico 5/metabolismo , Esquizofrenia/tratamiento farmacológico , Transmisión Sináptica/efectos de los fármacos , Animales , Antipsicóticos/farmacología , Cognición/efectos de los fármacos , Ratones Endogámicos C57BL , Plasticidad Neuronal/efectos de los fármacos , Fenciclidina/farmacología , Corteza Prefrontal/metabolismo , Receptores de N-Metil-D-Aspartato/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/metabolismo
15.
Mol Brain ; 10(1): 15, 2017 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-28446243

RESUMEN

Support for the N-methyl-D-aspartate receptor (NMDAR) hypofunction hypothesis of schizophrenia has led to increasing focus on restoring proper glutamatergic signaling as an approach for treatment of this devastating disease. The ability of metabotropic glutamate (mGlu) receptors to modulate glutamatergic neurotransmission has thus attracted considerable attention for the development of novel antipsychotics. Consisting of eight subtypes classified into three groups based on sequence homology, signal transduction, and pharmacology, the mGlu receptors provide a wide range of targets to modulate NMDAR function as well as glutamate release. Recently, allosteric modulators of mGlu receptors have been developed that allow unprecedented selectivity among subtypes, not just groups, facilitating the investigation of the effects of subtype-specific modulation. In preclinical animal models, positive allosteric modulators (PAMs) of the group I mGlu receptor mGlu5 have efficacy across all three symptom domains of schizophrenia (positive, negative, and cognitive). The discovery and development of mGlu5 PAMs that display unique signal bias suggests that efficacy can be retained while avoiding the neurotoxic effects of earlier compounds. Interestingly, mGlu1 negative allosteric modulators (NAMs) appear efficacious in positive symptom models of the disease but are still in early preclinical development. While selective group II mGlu receptor (mGlu2/3) agonists have reached clinical trials but were unsuccessful, specific mGlu2 or mGlu3 receptor targeting still hold great promise. Genetic studies implicated mGlu2 in the antipsychotic effects of group II agonists and mGlu2 PAMs have since entered into clinical trials. Additionally, mGlu3 appears to play an important role in cognition, may confer neuroprotective effects, and thus is a promising target to alleviate cognitive deficits in schizophrenia. Although group III mGlu receptors (mGlu4/6/7/8) have attracted less attention, mGlu4 agonists and PAMs appear to have efficacy across all three symptoms domains in preclinical models. The recent discovery of heterodimers comprising mGlu2 and mGlu4 may explain the efficacy of mGlu4 selective compounds but this remains to be determined. Taken together, compounds targeting mGlu receptors, specifically subtype-selective allosteric modulators, provide a compelling alternative approach to fill the unmet clinical needs for patients with schizophrenia.


Asunto(s)
Terapia Molecular Dirigida , Receptores de Glutamato Metabotrópico/metabolismo , Esquizofrenia/tratamiento farmacológico , Esquizofrenia/metabolismo , Animales , Humanos , Modelos Biológicos
16.
ACS Chem Neurosci ; 7(12): 1706-1716, 2016 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-27617634

RESUMEN

Abnormalities in the signaling of the N-methyl-d-aspartate subtype of the glutamate receptor (NMDAR) within cortical and limbic brain regions are thought to underlie many of the complex cognitive and affective symptoms observed in individuals with schizophrenia. The M1 muscarinic acetylcholine receptor (mAChR) subtype is a closely coupled signaling partner of the NMDAR. Accumulating evidence suggests that development of selective positive allosteric modulators (PAMs) of the M1 receptor represent an important treatment strategy for the potential normalization of disruptions in NMDAR signaling in patients with schizophrenia. In the present studies, we evaluated the effects of the novel and highly potent M1 PAM, VU6004256, in ameliorating selective prefrontal cortical (PFC)-mediated physiologic and cognitive abnormalities in a genetic mouse model of global reduction in the NR1 subunit of the NMDAR (NR1 knockdown [KD]). Using slice-based extracellular field potential recordings, deficits in muscarinic agonist-induced long-term depression (LTD) in layer V of the PFC in the NR1 KD mice were normalized with bath application of VU6004256. Systemic administration of VU6004256 also reduced excessive pyramidal neuron firing in layer V PFC neurons in awake, freely moving NR1 KD mice. Moreover, selective potentiation of M1 by VU6004256 reversed the performance impairments of NR1 KD mice observed in two preclinical models of PFC-mediated learning, specifically the novel object recognition and cue-mediated fear conditioning tasks. VU6004256 also produced a robust, dose-dependent reduction in the hyperlocomotor activity of NR1 KD mice. Taken together, the current findings provide further support for M1 PAMs as a novel therapeutic approach for the PFC-mediated impairments in schizophrenia.


Asunto(s)
Colinérgicos/farmacología , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Proteínas del Tejido Nervioso/deficiencia , Nootrópicos/farmacología , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Receptores de N-Metil-D-Aspartato/deficiencia , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Animales , Colinérgicos/farmacocinética , Trastornos del Conocimiento/tratamiento farmacológico , Trastornos del Conocimiento/metabolismo , Condicionamiento Psicológico/efectos de los fármacos , Condicionamiento Psicológico/fisiología , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Miedo/efectos de los fármacos , Miedo/fisiología , Técnicas de Silenciamiento del Gen , Compuestos Heterocíclicos de 4 o más Anillos/farmacocinética , Depresión Sináptica a Largo Plazo/efectos de los fármacos , Depresión Sináptica a Largo Plazo/fisiología , Masculino , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Proteínas del Tejido Nervioso/genética , Nootrópicos/farmacocinética , Células Piramidales/efectos de los fármacos , Células Piramidales/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Reconocimiento en Psicología/efectos de los fármacos , Reconocimiento en Psicología/fisiología , Técnicas de Cultivo de Tejidos
17.
J Am Assoc Lab Anim Sci ; 55(5): 520-4, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27657705

RESUMEN

Cryopreservation is an important tool routinely used in preserving sperm for assisted reproductive technologies and for genetic preservation of unique animal models. Here we investigated the viability of fresh and frozen sperm from rhesus macaques on the basis of motility, membrane integrity, and acrosome integrity. Sperm motility was determined by visual evaluation; membrane and acrosome integrity were assessed simultaneously through triple staining with Hoechst 33342, propidium iodide, and fluorescein isothiocyanate-peanut agglutinin. We compared thawed semen that had been cryopreserved by using 2 different media with fresh semen from wildtype (WT) macaques; fresh semen from a model of Huntington disease (HD) with fresh WT semen; and fresh HD with cryopreserved-thawed HD semen. Our new freezing media (TEST EQ) preserved the acrosome better, with less net damage, than did traditional TEST (egg yolk extender containing TES and Tris) media. In addition, the percentage of membrane-damaged cells was similar in fresh HD semen (38.6%±2.9%) and WT semen (35.5%±1.9%). Membrane and acrosomal damage were not different between HD and WT sperm after cryopreservation and subsequent thawing. Furthermore, cryopreservation had similar negative effects on the motility of HD and WT sperm. These data illustrate that semen from a rhesus macaque model of HD is similarly cryotoleratant to that from WT animals.


Asunto(s)
Animales Modificados Genéticamente , Criopreservación/veterinaria , Macaca mulatta/genética , Macaca mulatta/fisiología , Preservación de Semen/veterinaria , Animales , Yema de Huevo , Masculino , Motilidad Espermática , Espermatozoides
18.
Brain Behav Immun ; 58: 181-190, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27395434

RESUMEN

Although the most notable clinical symptoms of Huntington's disease (HD) are motor disturbances and brain atrophy, other symptoms include cognitive dysfunction, emotional and hormonal dysregulation. Emotional dysregulation (irritability, anger/aggression, and anxiety) and increased inflammation are early emerging symptoms which can be detected decades before the onset of motor symptoms in HD patients. Despite the advances in understanding the genetic causes of HD there is still no cure or preventative treatment. Thus, to better understand the pathogenesis of HD and develop effective treatments, a holistic understanding of HD is needed, as well as animal models that replicate the full spectrum of HD symptoms. The current study examined the emotional, hormonal, and gene expression responses to an acute stressor of adult male transgenic HD rhesus monkeys (n=2) as compared to wild-type controls (n=2). Results revealed that HD monkeys expressed increased anxiety and irritability/aggression as compared to controls. Reactive cortisol response to the stressor was similar between groups. However, HD monkeys exhibited increased pro-inflammatory cytokines and higher induction of immune pathway genes as compared to controls. Overall, results reveal that HD monkeys exhibit these early emerging symptoms of HD and may be an effective animal model to facilitate the development of new therapeutics for HD patients.


Asunto(s)
Ansiedad , Conducta Animal , Enfermedad de Huntington/inmunología , Enfermedad de Huntington/psicología , Agresión , Animales , Animales Modificados Genéticamente , Proteína C-Reactiva/metabolismo , Modelos Animales de Enfermedad , Expresión Génica , Enfermedad de Huntington/genética , Hidrocortisona/sangre , Interleucina-6/sangre , Leucocitos Mononucleares/metabolismo , Macaca mulatta , Masculino , Estrés Fisiológico , Factor de Necrosis Tumoral alfa/sangre
19.
Stem Cell Reports ; 3(4): 585-93, 2014 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-25358787

RESUMEN

Huntington's disease (HD) is a dominant neurodegenerative disorder caused by the expansion of glutamine residues in the N-terminal region of the huntingtin (HTT) protein. The disease results in progressive neuronal loss, leading to motor, cognitive, and psychiatric impairment. Here, we report the establishment of neural progenitor cell (NPC) lines derived from induced pluripotent stem cells (iPSCs) of transgenic HD monkeys. Upon differentiation to neurons, HD neural cells develop cellular features of HD, including the formation of nuclear inclusions and oligomeric mutant HTT (mHTT) aggregates, as well as increased apoptosis. These phenotypes are rescued by genetic suppression of HTT and pharmacological treatment, demonstrating the ability of our HD cell model to respond to therapeutic treatment. The development and reversal of HD-associated phenotypes in neural cells from HD monkeys provides a unique nonhuman primate (NHP) model for exploring HD pathogenesis and evaluating therapeutics that could be assessed further in HD monkeys.


Asunto(s)
Neuronas GABAérgicas/citología , Enfermedad de Huntington/patología , Células Madre Pluripotentes Inducidas/citología , Células-Madre Neurales/citología , Fenotipo , Animales , Antiparkinsonianos/farmacología , Apoptosis , Células Cultivadas , Neuronas GABAérgicas/efectos de los fármacos , Neuronas GABAérgicas/metabolismo , Haplorrinos , Proteína Huntingtina , Enfermedad de Huntington/genética , Células Madre Pluripotentes Inducidas/metabolismo , Memantina/farmacología , Mutación , Proteínas del Tejido Nervioso/genética , Células-Madre Neurales/metabolismo , Neurogénesis
20.
Theranostics ; 4(10): 972-89, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25161700

RESUMEN

PURPOSE: The ability to longitudinally monitor cell grafts and assess their condition is critical for the clinical translation of stem cell therapy in regenerative medicine. Developing an inducible genetic magnetic resonance imaging (MRI) reporter will enable non-invasive and longitudinal monitoring of stem cell grafts in vivo. METHODS: MagA, a bacterial gene involved in the formation of iron oxide nanocrystals, was genetically modified for in vivo monitoring of cell grafts by MRI. Inducible expression of MagA was regulated by a Tet-On (Tet) switch. A mouse embryonic stem cell-line carrying Tet-MagA (mESC-MagA) was established by lentivirus transduction. The impact of expressing MagA in mESCs was evaluated via proliferation assay, cytotoxicity assay, teratoma formation, MRI, and inductively coupled plasma atomic emission spectroscopy (ICP-OES). Mice were grafted with mESCs with and without MagA (mESC-MagA and mESC-WT). The condition of cell grafts with induced "ON" and non-induced "OFF" expression of MagA was longitudinally monitored in vivo using a 7T MRI scanner. After imaging, whole brain samples were harvested for histological assessment. RESULTS: Expression of MagA in mESCs resulted in significant changes in the transverse relaxation rate (R2 or 1/T2) and susceptibility weighted MRI contrast. The pluripotency of mESCs carrying MagA was not affected in vitro or in vivo. Intracranial mESC-MagA grafts generated sufficient T2 and susceptibility weighted contrast at 7T. The mESC-MagA grafts can be monitored by MRI longitudinally upon induced expression of MagA by administering doxycycline (Dox) via diet. CONCLUSION: Our results demonstrate MagA could be used to monitor cell grafts noninvasively, longitudinally, and repetitively, enabling the assessment of cell graft conditions in vivo.


Asunto(s)
Rastreo Celular/métodos , Células Madre Embrionarias/trasplante , Animales , Proteínas Bacterianas/biosíntesis , Proteínas Bacterianas/genética , Células Madre Embrionarias/fisiología , Expresión Génica , Genes Reporteros , Células HEK293 , Humanos , Imagen por Resonancia Magnética , Ratones de la Cepa 129 , Ratones Desnudos , Ratones SCID , Trasplante de Células Madre/efectos adversos , Teratoma/etiología , Teratoma/metabolismo , Teratoma/patología , Activación Transcripcional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA