Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 207
Filtrar
1.
Biomed Pharmacother ; 179: 117405, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39236478

RESUMEN

Traumatic brain injury (TBI) is a significant contributor to global mortality and disability, and there is still no specific drug available to treat cognitive deficits in survivors. Vanillic acid (VA), a bioactive phenolic compound, has shown protective effects in various models of neurodegeneration; however, its impact on TBI outcomes remains elusive. Therefore, this study aimed to elucidate the possible role of VA in ameliorating TBI-induced cognitive decline and to reveal the mechanisms involved. TBI was induced using the Marmarou impact acceleration model to deliver an impact force of 300 g, and treatment with VA (50 mg/kg; P.O.) was initiated 30 minutes post-TBI. The cognitive performance, hippocampal long-term potentiation (LTP), oxidative stress markers, neurological function, cerebral edema, and morphological changes were assessed at scheduled points in time. TBI resulted in cognitive decline in the passive avoidance task, impaired LTP in the perforant path-dentate gyrus (PP-DG) pathway, increased hippocampal oxidative stress, cerebral edema, neurological deficits, and neuronal loss in the rat hippocampus. In contrast, acute VA administration mitigated all the aforementioned TBI outcomes. The data suggest that reducing synaptic plasticity impairment, regulating oxidative and antioxidant defense, alleviating cerebral edema, and preventing neuronal loss by VA can be at least partially attributed to its protection against TBI-induced cognitive decline.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Disfunción Cognitiva , Hipocampo , Potenciación a Largo Plazo , Estrés Oxidativo , Ácido Vanílico , Animales , Ácido Vanílico/farmacología , Masculino , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/etiología , Disfunción Cognitiva/prevención & control , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/psicología , Estrés Oxidativo/efectos de los fármacos , Ratas , Potenciación a Largo Plazo/efectos de los fármacos , Hipocampo/efectos de los fármacos , Hipocampo/patología , Hipocampo/metabolismo , Fármacos Neuroprotectores/farmacología , Edema Encefálico/tratamiento farmacológico , Ratas Wistar , Modelos Animales de Enfermedad , Antioxidantes/farmacología , Cognición/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos
2.
Neuron ; 2024 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-39241780

RESUMEN

Tau interacts with α-Synuclein (α-Syn) and co-localizes with it in the Lewy bodies, influencing α-Syn pathology in Parkinson's disease (PD). However, whether these biochemical events regulate α-Syn pathology spreading from the gut into the brain remains incompletely understood. Here, we show that α-Syn and Tau co-pathology is spread into the brain in gut-inducible SYN103+/- and/or TAU368+/- transgenic mouse models, eliciting behavioral defects. Gut pathology was initially observed, and α-Syn or Tau pathology was subsequently propagated into the DMV or NTS and then to other brain regions. Remarkably, more extensive spreading and widespread neuronal loss were found in double transgenic mice (Both) than in single transgenic mice. Truncal vagotomy and α-Syn deficiency significantly inhibited synucleinopathy or tauopathy spreading. The α-Syn PET tracer [18F]-F0502B detected α-Syn aggregates in the gut and brain. Thus, α-Syn and Tau co-pathology can propagate from the gut to the brain, triggering behavioral disorders.

3.
Brain Res ; 1844: 149196, 2024 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-39181223

RESUMEN

This work was done to investigate the ameliorating impact of 4-methylumbilliferon (4-MU) on spatial learning and memory dysfunction and restraint stress (STR)-induced anxiety-like behaviors in male Wistar rats and the underlying mechanisms. Thirty-two animals were assigned into 4 cohorts: control, 4-MU, STR, and STR+4-MU. Animals were exposed to STR for 4 h per day for 14 consecutive days or kept in normal conditions (healthy animals without exposure to stress). 4-MU (25 mg/kg) was intraperitoneally administered once daily to STR rats before restraint stress for 14 consecutive days. The behavioral tests were performed through Morris water maze tests and elevated-plus maze to examine learning/memory function, and anxiety levels, respectively. The levels of the antioxidant defense biomarkers (GPX, SOD) and MDA as an oxidant molecule in the brain tissues were measured using commercial ELISA kits. Neuronal loss or density of neurons was evaluated using Nissl staining. STR exposure could cause significant alterations in the levels of the antioxidant defense biomarkers (MDA, GPX, and SOD) in the prefrontal cortex and hippocampus, induce anxiety, and impair spatial learning and memory function. Treatment with 4-MU markedly reduced anxiety levels and improved spatial learning and memory dysfunction via restoring the antioxidant defense biomarkers to normal values and reducing MDA levels. Moreover, more intact cells with normal morphologies were detected in STR-induced animals treated with 4-MU. 4-MU could attenuate the STR-induced anxiety-like behaviors and spatial learning and memory dysfunction by reducing oxidative damage and neuronal loss in the prefrontal cortex and hippocampus region. Taken together, our findings provide new insights regarding the potential therapeutic effects of 4-MU against neurobehavioral disorders induced by STR.


Asunto(s)
Ansiedad , Muerte Celular , Trastornos de la Memoria , Neuronas , Estrés Oxidativo , Ratas Wistar , Animales , Estrés Oxidativo/efectos de los fármacos , Masculino , Ansiedad/tratamiento farmacológico , Ansiedad/metabolismo , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Ratas , Muerte Celular/efectos de los fármacos , Estrés Psicológico/tratamiento farmacológico , Estrés Psicológico/metabolismo , Estrés Psicológico/complicaciones , Aprendizaje por Laberinto/efectos de los fármacos , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Antioxidantes/farmacología , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo
4.
Brain Commun ; 6(4): fcae255, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39130514

RESUMEN

Cerebral toxoplasmosis, the most common opportunistic infection in immunocompromised individuals, is increasingly reported in immunocompetent individuals due to mutant strains of Toxoplasma gondii, which, furthermore, are reported to be resistant to available treatments. We assessed the therapeutic potential of Garcinia kola, a medicinal plant reported to have antiplasmodial and neuroprotective properties, against experimental toxoplasmosis in rats. Severe toxoplasmosis was induced in male Wistar rats (156.7 ± 4.1 g) by injecting them with 10 million tachyzoites in suspension in 500 µl of saline (intraperitoneal), and exclusive feeding with a low-protein diet [7% protein (weight by weight)]. Then, animals were treated with hexane, dichloromethane, and ethyl acetate fractions of Garcinia kola. Footprints were analysed and open-field and elevated plus maze ethological tests were performed when symptoms of severe disease were observed in the infected controls. After sacrifice, blood samples were processed for Giemsa staining, organs were processed for haematoxylin and eosin staining, and brains were processed for Nissl staining and cell counting. Compared with non-infected animals, the infected control animals had significantly lower body weights (30.27%↓, P = 0.001), higher body temperatures (P = 0.033) during the sacrifice, together with signs of cognitive impairment and neurologic deficits such as lower open-field arena centre entries (P < 0.001), elevated plus maze open-arm time (P = 0.029) and decreased stride lengths and step widths (P < 0.001), as well as neuronal loss in various brain areas. The ethyl acetate fraction of Garcinia kola prevented or mitigated most of these signs. Our data suggest that the ethyl acetate fraction of Garcinia kola has therapeutic potential against cerebral toxoplasmosis.

5.
Aging Cell ; : e14263, 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38961613

RESUMEN

Frailty is a geriatric, multi-dimensional syndrome that reflects multisystem physiological change and is a transversal measure of reduced resilience to negative events. It is characterized by weakness, frequent falls, cognitive decline, increased hospitalization and dead and represents a risk factor for the development of Alzheimer's disease (AD). The fact that frailty is recognized as a reversible condition encourages the identification of earlier biomarkers to timely predict and prevent its occurrence. SAMP8 (Senescence-Accelerated Mouse Prone-8) mice represent the most appropriate preclinical model to this aim and were used in this study to carry transcriptional and metabolic analyses in the brain and plasma, respectively, upon a characterization at cognitive, motor, structural, and neuropathological level at 2.5, 6, and 9 months of age. At 2.5 months, SAMP8 mice started displaying memory deficits, muscle weakness, and motor impairment. Functional alterations were associated with a neurodevelopmental deficiency associated with reduced neuronal density and glial cell loss. Through transcriptomics, we identified specific genetic signatures well distinguishing SAMP8 mice at 6 months, whereas plasma metabolomics allowed to segregate SAMP8 mice from SAMR1 already at 2.5 months of age by detecting constitutively lower levels of acylcarnitines and lipids in SAMP8 at all ages investigated correlating with functional deficits and neuropathological signs. Our findings suggest that specific genetic alterations at central level, as well as metabolomic changes in plasma, might allow to early assess a frail condition leading to dementia development, which paves the foundation for future investigation in a clinical setting.

6.
Int J Mol Sci ; 25(14)2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-39062781

RESUMEN

Due to their inhibition of acetylcholinesterase, organophosphates are among the most toxic of chemicals. Pralidoxime (a.k.a 2-PAM) is the only acetylcholinesterase reactivator approved in the U.S., but 2-PAM only poorly traverses the blood-brain barrier. Previously, we have demonstrated that scL-2PAM, a nanoformulation designed to enter the brain via receptor-mediated transcytosis, is superior to unencapsulated 2-PAM for reactivating brain acetylcholinesterase, ameliorating cholinergic crisis, and improving survival rates for paraoxon-exposed mice. Here, we employ histology and transcriptome analyses to assess the ability of scL-2PAM to prevent neurological sequelae including microglial activation, expression of inflammatory cytokines, and ultimately loss of neurons in mice surviving paraoxon exposures. Levels of the mRNA encoding chemokine ligand 2 (CCL2) were significantly upregulated after paraoxon exposures, with CCL2 mRNA levels in the brain correlating well with the intensity and duration of cholinergic symptoms. Our nanoformulation of 2-PAM was found to be superior to unencapsulated 2-PAM in reducing the levels of the CCL2 transcript. Moreover, brain histology revealed that scL-2PAM was more effective than unencapsulated 2-PAM in preventing microglial activation and the subsequent loss of neurons. Thus, scL-2PAM appears to be a new and improved countermeasure for reducing neuroinflammation and mitigating brain damage in survivors of organophosphate exposures.


Asunto(s)
Inhibidores de la Colinesterasa , Enfermedades Neuroinflamatorias , Neuronas , Paraoxon , Animales , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Inhibidores de la Colinesterasa/farmacología , Paraoxon/toxicidad , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Enfermedades Neuroinflamatorias/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Quimiocina CCL2/metabolismo , Quimiocina CCL2/genética , Microglía/efectos de los fármacos , Microglía/metabolismo , Masculino , Organofosfatos/farmacología , Acetilcolinesterasa/metabolismo , Ratones Endogámicos C57BL
7.
CNS Neurosci Ther ; 30(7): e14886, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39072940

RESUMEN

BACKGROUND: Oxidative stress is a well-known pathological factor driving neuronal loss and age-related neurodegenerative diseases. Melatonin, coenzyme Q10 and lecithin are three common nutrients with an antioxidative capacity. Here, we examined the effectiveness of them administrated individually and in combination in protecting against oxidative stress-induced neuronal death in vitro, and neurodegenerative conditions such as Alzheimer's disease and associated deficits in vivo. METHODS: Mouse neuroblastoma Neuro-2a (N2a) cells were exposed with H2O2 for 6 h, and subsequently treated with melatonin, coenzyme Q10, and lecithin alone or in combination for further 24 h. Cell viability was assessed using the CCK-8 assay. Eight-week-old male mice were intraperitoneally injected with D-(+)-galactose for 10 weeks and administrated with melatonin, coenzyme Q10, lecithin, or in combination for 5 weeks starting from the sixth week, followed by behavioral tests to assess the effectiveness in mitigating neurological deficits, and biochemical assays to explore the underlying mechanisms. RESULTS: Exposure to H2O2 significantly reduced the viability of N2a cells and increased oxidative stress and tau phosphorylation, all of which were alleviated by treatment with melatonin, coenzyme Q10, lecithin alone, and, most noticeably, by combined treatment. Administration of mice with D-(+)-galactose-induced oxidative stress and tau phosphorylation, brain aging, impairments in learning and memory, anxiety- and depression-like behaviors, and such detrimental effects were mitigated by melatonin, coenzyme Q10, lecithin alone, and, most consistently, by combined treatment. CONCLUSIONS: These results suggest that targeting oxidative stress via supplementation of antioxidant nutrients, particularly in combination, is a better strategy to alleviate oxidative stress-mediated neuronal loss and brain dysfunction due to age-related neurodegenerative conditions.


Asunto(s)
Antioxidantes , Peróxido de Hidrógeno , Neuronas , Estrés Oxidativo , Ubiquinona , Animales , Estrés Oxidativo/efectos de los fármacos , Ratones , Ubiquinona/análogos & derivados , Ubiquinona/farmacología , Ubiquinona/administración & dosificación , Masculino , Antioxidantes/farmacología , Peróxido de Hidrógeno/toxicidad , Neuronas/efectos de los fármacos , Neuronas/patología , Línea Celular Tumoral , Melatonina/farmacología , Melatonina/uso terapéutico , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Proteínas tau/metabolismo , Fármacos Neuroprotectores/farmacología , Galactosa/toxicidad , Quimioterapia Combinada
8.
Brain Res Bull ; 214: 110994, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38830486

RESUMEN

Synaptotagmin-7 (SYT7) has been proposed as an innovative therapeutic strategy for treating cognitive impairment, while its contribution to Alzheimer's disease (AD) alleviation remains unclear. In this study, we investigated the role and potential mechanisms of SYT7 in AD. APP/PS1 mice were induced as an AD mouse model, and RNA-sequencing was conducted to analyze the transcriptomic differences between the brain tissues of AD mice and controls. SYT7, which was the most significantly differentially expressed gene in the RNA-sequencing, was found to be reduced in AD-like mice, and overexpression of SYT7 alleviated cognitive dysfunction and attenuated neuroinflammation and neuronal loss in the hippocampal tissues of mice with AD. Transcription factor double-strand-break repair protein rad21 homolog (RAD21) bound to the promoter of SYT7 to activate SYT7 transcription. SYT7 and RAD21 were expressed in microglia. SYT7 and RAD21 both promoted M2 polarization of microglia, while silencing of SYT7 repressed the M2 polarization of microglia in the presence of RAD21 overexpression. Overall, our results indicate that RAD21 mediated transcriptional activation of SYT7 to promote M2 polarization of microglia, thereby alleviating AD-like symptoms in mice, which might provide prospective cues for developing therapeutic strategies to improve cognitive impairment and AD course.


Asunto(s)
Enfermedad de Alzheimer , Microglía , Sinaptotagminas , Animales , Masculino , Ratones , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/genética , Disfunción Cognitiva/metabolismo , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Ratones Transgénicos , Microglía/metabolismo , Sinaptotagminas/metabolismo , Sinaptotagminas/genética
9.
Pharmacol Rep ; 76(4): 644-664, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38904713

RESUMEN

Neurodegenerative diseases such as Parkinson's disease, Alzheimer's disease, Huntington's disease, and multiple sclerosis affect millions of people around the world. In addition to age, which is a key factor contributing to the development of all neurodegenerative diseases, genetic and environmental components are also important risk factors. Current methods of treating neurodegenerative diseases are mostly symptomatic and do not eliminate the cause of the disease. Many studies focus on searching for natural substances with neuroprotective properties that could be used as an adjuvant therapy in the inhibition of the neurodegeneration process. These compounds include flavonoids, such as luteolin, showing significant anti-inflammatory, antioxidant, and neuroprotective activity. Increasing evidence suggests that luteolin may confer protection against neurodegeneration. In this review, we summarize the scientific reports from preclinical in vitro and in vivo studies regarding the beneficial effects of luteolin in neurodegenerative diseases. Luteolin was studied most extensively in various models of Alzheimer's disease but there are also several reports showing its neuroprotective effects in models of Parkinson's disease. Though very limited, studies on possible protective effects of luteolin against Huntington's disease and multiple sclerosis are also discussed here. Overall, although preclinical studies show the potential benefits of luteolin in neurodegenerative disorders, clinical evidence on its therapeutic efficacy is still deficient.


Asunto(s)
Luteolina , Enfermedades Neurodegenerativas , Fármacos Neuroprotectores , Luteolina/farmacología , Luteolina/uso terapéutico , Humanos , Animales , Enfermedades Neurodegenerativas/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Fármacos Neuroprotectores/farmacología , Antioxidantes/farmacología , Antioxidantes/uso terapéutico
10.
Brain ; 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38743596

RESUMEN

Protein Kinase A (PKA) neuronal function is controlled by the interaction of a regulatory (R) subunit dimer to two catalytic (C) subunits. Recently, the L50R variant in the gene encoding the RIß subunit was identified in individuals with a novel neurodegenerative disease. However, the mechanisms driving the disease phenotype remained unknown. In this study, we generated a mouse model carrying the RIß-L50R mutation to replicate the human disease phenotype and study its progression with age. We examined postmortem brains of affected individuals as well as live cell cultures. Employing biochemical assays, immunohistochemistry, and behavioral assessments, we investigated the impact of the mutation on PKA complex assembly, protein aggregation and neuronal degeneration. We reveal that RIß is an aggregation-prone protein that progressively accumulates in wildtype and Alzheimer's mouse models with age, while aggregation is accelerated in the RIß-L50R mouse model. We define RIß-L50R as a causal mutation driving an age-dependent behavioral and disease phenotype in human and mouse models. Mechanistically, this mutation disrupts RIß dimerization, leading to aggregation of its monomers. Intriguingly, interaction with the C-subunit protects the RIß-L50R from self-aggregating, in a dose-dependent manner. Furthermore, cAMP signaling induces RIß-L50R aggregation. The pathophysiological mechanism elucidated here for a newly recognized neurodegenerative disease, in which protein aggregation is the result of disrupted homodimerization, sheds light on a remarkably under-appreciated but potentially common mechanism across several neurodegenerative diseases.

11.
Res Sq ; 2024 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-38562908

RESUMEN

Background: The G2019S mutation of LRRK2, which enhances kinase activity of the protein, confers a substantial risk of developing Parkinson's disease (PD). However, the mutation demonstrates incomplete penetrance, suggesting the involvement of other genetic or environmental modulating factors. Here, we investigated whether LRRK2 G2019S knock-in (KI) mice treated with the inflammogen lipopolysaccharide (LPS) could model LRRK2 PD. Results: We found that short-term (2 weeks) treatment with LPS did not result in the loss of dopaminergic neurons in either LRRK2 G2019S KI or wild-type (WT) mice. Compared with WT mice, LRRK2 G2019S-KI mice showed incomplete recovery from LPS-induced weight loss. In LRRK2 G2019S KI mice, LPS treatment led to upregulated phosphorylation of LRRK2 at the autophosphorylation site Serine 1292, which is known as a direct readout of LRRK2 kinase activity. LPS treatment caused a greater increase in the activated astrocyte marker glial fibrillary acidic protein (GFAP) in the striatum and substantia nigra of LRRK2 G2019S mice than in those of WT mice. The administration of caffeine, which was recently identified as a biomarker of resistance to developing PD in individuals with LRRK2 mutations, attenuated LPS-induced astrocyte activation specifically in LRRK2 G2019S KI mice. Conclusions: Our findings suggest that 2 weeks of exposure to LPS is not sufficient to cause dopaminergic neuronal loss in LRRK2 G2019S KI mice but rather results in increased astrocyte activation, which can be ameliorated by caffeine.

12.
Physiol Behav ; 278: 114508, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38460779

RESUMEN

BACKGROUND: Neurodegenerative disorders are associated with chronic neuroinflammation, which contributes to their pathogenesis and progression. Resveratrol (RSV) is a polyphenolic compound with strong antioxidant and anti-inflammatory properties. In the present study, we investigated whether RSV could protect against cognitive impairment and inflammatory response in a mouse model of chronic neuroinflammation induced by lipopolysaccharide (LPS). METHOD: Mice received oral RSV (30 mg/kg) or vehicle for two weeks, and injected with LPS (0.75 mg/kg) or saline daily for the last seven days. After two weeks, mice were subjected to behavioral assessments using the Morris water maze and Y-maze. Moreover, mRNA expression of several inflammatory markers, neuronal loss, and glial density were evaluated in the hippocampus of treated mice. RESULTS: Our findings showed that RSV treatment effectively improved spatial and working memory impairments induced by LPS. In addition, RSV significantly reduced hippocampal glial densities and neuronal loss in LPS-injected mice. Moreover, RSV treatment suppressed LPS-induced upregulation of NF-κB, IL-6, IL-1ß, and GFAP in the hippocampus of treated mice. CONCLUSION: Taken together, our results highlight the detrimental effect of systemic inflammation on the hippocampus and the potential of natural products with anti-inflammatory effects to counteract this impact.


Asunto(s)
Disfunción Cognitiva , Lipopolisacáridos , Ratones , Animales , Resveratrol/uso terapéutico , Lipopolisacáridos/toxicidad , Enfermedades Neuroinflamatorias , Microglía/metabolismo , Disfunción Cognitiva/inducido químicamente , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/metabolismo , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Modelos Animales de Enfermedad , FN-kappa B/metabolismo , Hipocampo/metabolismo , Aprendizaje por Laberinto
13.
Expert Opin Drug Deliv ; 21(3): 423-435, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38481172

RESUMEN

INTRODUCTION: Parkinson's disease (PD) is a neurological condition defined by a substantial reduction in dopamine-containing cells in the substantia nigra. Levodopa (L-Dopa) is considered the gold standard in treatment. Recent research has clearly shown that resistance to existing therapies can develop. Moreover, the involvement of multiple pathways in the nigrostriatal dopaminergic neuronal loss suggests that modifying the treatment strategy could effectively reduce this degeneration. AREAS COVERED: This review summarizes the key concerns with treating PD patients and the combinations, aimed at effectively managing PD. Part I focuses on the clinical diagnosis at every stage of the disease as well as the pharmacological treatment strategies that are applied throughout its course. It methodically elucidates the potency of multifactorial interventions in attenuating the disease trajectory, substantiating the rationale for co-administration of dual or multiple therapeutic agents. Significant emphasis is laid on evidence-based pharmacological combinations for PD management. EXPERT OPINION: By utilizing multiple drugs in a combination fashion, this approach can leverage the additive or synergistic effects of these agents, amplify the spectrum of treatment, and curtail the risk of side effects by reducing the dose of each drug, demonstrating significantly greater efficacy.


Asunto(s)
Antiparkinsonianos , Quimioterapia Combinada , Levodopa , Enfermedad de Parkinson , Enfermedad de Parkinson/tratamiento farmacológico , Humanos , Antiparkinsonianos/administración & dosificación , Antiparkinsonianos/uso terapéutico , Antiparkinsonianos/farmacología , Levodopa/administración & dosificación , Levodopa/uso terapéutico , Animales , Portadores de Fármacos/química , Nanopartículas , Sinergismo Farmacológico
14.
Pharmacol Biochem Behav ; 237: 173721, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38307465

RESUMEN

Autism spectrum disorder (ASD) is a neurodevelopment disorder that mainly arises due to abnormalities in different brain regions, resulting in behavioral deficits. Besides its diverse phenotypical features, ASD is associated with complex and varied etiology, presenting challenges in understanding its precise neuro-pathophysiology. Pioglitazone was reported to have a fundamental role in neuroprotection in various other neurological disorders. The present study aimed to investigate the therapeutic potential of pioglitazone in the prenatal valproic acid (VPA)-model of ASD in Wistar rats. Pregnant female Wistar rats received VPA on Embryonic day (E.D12.5) to induce autistic-like-behavioral and neurobiological alterations in their offspring. VPA-exposed rats presented core behavioral symptoms of ASD such as deficits in social interaction, poor spatial and learning behavior, increased anxiety, locomotory and repetitive activity, and decreased exploratory activity. Apart from these, VPA exposure also stimulated neurochemical and histopathological neurodegeneration in various brain regions. We administered three different doses of pioglitazone i.e., 2.5, 5, and 10 mg/kg in rats to assess various parameters. Of all the doses, our study highlighted that 10 mg/kg pioglitazone efficiently attenuated the autistic symptoms along with other neurochemical alterations such as oxidative stress, neuroinflammation, and apoptosis. Moreover, pioglitazone significantly attenuated the neurodegeneration by restoring the neuronal loss in the hippocampus and cerebellum. Taken together, our study suggests that pioglitazone exhibits therapeutic potential in alleviating behavioral abnormalities induced by prenatal VPA exposure in rats. However, further research is needed to fully understand and establish pioglitazone's effectiveness in treating ASD.


Asunto(s)
Trastorno del Espectro Autista , Trastorno Autístico , Efectos Tardíos de la Exposición Prenatal , Embarazo , Ratas , Femenino , Animales , Humanos , Ácido Valproico/farmacología , Trastorno del Espectro Autista/inducido químicamente , Trastorno del Espectro Autista/tratamiento farmacológico , Ratas Wistar , Pioglitazona/farmacología , Trastorno Autístico/inducido químicamente , Conducta Social , Conducta Animal , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/tratamiento farmacológico , Modelos Animales de Enfermedad
15.
Epigenomics ; 16(5): 331-358, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38321930

RESUMEN

Recent genomics and epigenetic advances have empowered the exploration of DNA/RNA methylation and histone modifications crucial for gene expression in response to stress, aging and disease. Interest in understanding neuronal plasticity's epigenetic mechanisms, influencing brain rewiring amid development, aging and neurodegenerative disorders, continues to grow. Histone acetylation dysregulation, a commonality in diverse brain disorders, has become a therapeutic focus. Histone acetyltransferases and histone deacetylases have emerged as promising targets for neurodegenerative disorder treatment. This review delves into histone acetylation regulation, potential therapies and future perspectives for disorders like Alzheimer's, Parkinson's and Huntington's. Exploring genetic-environmental interplay through models and studies reveals molecular changes, behavioral insights and early intervention possibilities targeting the epigenome in at-risk individuals.


Scientists have made progress in understanding how our genes and their chemical modifications play a role in how our brains respond to stress, age and diseases. They are particularly interested in how these processes affect the flexibility of our brain circuits, which is important during growth and aging and in conditions like Alzheimer's and Parkinson's. One key area of focus is controlling a specific chemical change called histone acetylation, which tends to go awry in various brain disorders. Researchers are looking at potential treatments that target specific proteins related to this process. This review explores how these chemical changes might be regulated, potential treatments and the future for disorders like Alzheimer's, Parkinson's and Huntington's. By studying the interaction between our genes and the environment, scientists are uncovering changes at the molecular level, gaining insights into behavior and exploring ways to intervene early for people who are at risk.


Asunto(s)
Histonas , Enfermedades Neurodegenerativas , Humanos , Histonas/metabolismo , Acetilación , Enfermedades Neurodegenerativas/genética , Metilación de ADN , Epigénesis Genética
16.
Toxicol Appl Pharmacol ; 483: 116830, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38246289

RESUMEN

BACKGROUND: There is strong evidence that prenatal infection during a specific period of brain development increases the risk of neurodevelopmental disorders, partly through immune-inflammatory pathways. This suggests that anti-inflammatory agents could prevent these disorders by targeting the maternal inflammatory response. In the present study, we used a rat model of maternal immune activation (MIA) to examine whether maternal quercetin (QE) supplementation can alleviate behavioral deficits and inflammatory mediators in the prefrontal cortex (PFC) and hippocampus of adult male offspring. METHODS: Pregnant rats were supplemented with QE (50 mg/kg) or vehicle throughout pregnancy and injected with either lipopolysaccharide (0.5 mg/kg) or saline on gestational days 15/16. At postnatal day 60, we evaluated the offspring's behavior, hippocampal and prefrontal cortex glial density, pro-inflammatory gene expression, and neuronal survival. RESULTS: Our data showed that maternal QE supplementation can prevent working and recognition memory impairments in adult MIA offspring. This behavioral improvement correlates with the decrease in MIA-induced expression of pro-inflammatory genes, microglia, and astrocyte densities, without affecting neuronal survival, in both PFC and CA1 hippocampus areas. CONCLUSION: Therefore, our study supports the potential preventive effect of QE on MIA-induced behavioral dysfunctions, at least in part, by suppressing the glial-mediated inflammatory response.


Asunto(s)
Lipopolisacáridos , Efectos Tardíos de la Exposición Prenatal , Embarazo , Femenino , Humanos , Ratas , Animales , Masculino , Lipopolisacáridos/toxicidad , Quercetina/farmacología , Quercetina/uso terapéutico , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Cognición , Suplementos Dietéticos , Conducta Animal , Modelos Animales de Enfermedad
17.
J Neuroimmunol ; 387: 578292, 2024 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-38278081

RESUMEN

Systemic inflammation can cause chronic neuroinflammation, which is a significant risk factor for neurodegenerative disorders. Therefore, anti-inflammatory agents that reduce peripheral inflammation are potential targets for the prevention or treatment of these debilitating diseases. In the present study, we investigated whether gamma-oryzanol (ORY) could protect against chronic neuroinflammation induced by lipopolysaccharide (LPS) in adult male mice. Mice were injected with LPS (0.75 mg/kg/day) or saline for 7 consecutive days and orally received ORY (100 mg/kg) or vehicle for 14 days (7 days before LPS injections and 7 days co-treated with LPS). After two weeks, mice were subjected to behavioral assessments using the Morris water maze and Y-maze. Moreover, the expression level of several inflammatory mediators was measured in the hippocampus of treated animals. Also, neuronal loss, microglia, and astrocyte densities were evaluated in the CA1 and CA3 hippocampus. We found that ORY treatment significantly improved spatial and working memory in LPS-treated mice. This behavioral improvement was accompanied by a significant reduction in the number of microglia and astrocytes in the CA1 and CA3 hippocampus. Moreover, ORY treatment effectively prevented LPS-induced increases in the expression of inflammatory mediators and enhanced neuronal survival in the CA1 hippocampus. Our findings suggest that ORY treatment can be a therapeutic option to improve cognitive impairments and neuroinflammation induced by endotoxins.


Asunto(s)
Disfunción Cognitiva , Lipopolisacáridos , Fenilpropionatos , Ratones , Animales , Masculino , Lipopolisacáridos/toxicidad , Lipopolisacáridos/metabolismo , Enfermedades Neuroinflamatorias , Disfunción Cognitiva/inducido químicamente , Disfunción Cognitiva/tratamiento farmacológico , Hipocampo , Microglía/metabolismo , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Mediadores de Inflamación/metabolismo , Ratones Endogámicos C57BL
18.
Neurol Sci ; 45(3): 1163-1171, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37837508

RESUMEN

OBJECTIVE: The relationship between the cell body layer and the dendritic network layer of the retina and cognitive performance (CP) in MS patients has not been examined separately. The objective of this study is to predict cognitive impairment (CI) in RRMS patients and to examine the relationship between CP and ganglion cell layer (GCL), inner plexiform layer (IPL), and GCL divided by IPL (GCL/IPL). METHODS: Ophthalmological evaluation, retinal segmentation, and Symbol Digit Modalities Test (SDMT) were performed on 102 RRMS patients and 54 healthy subjects. The relationships of GCL, IPL, and GCL/IPL with CP in eyes without a history of optic neuritis were investigated using Spearman's correlation. Models were created by accepting 1 standard deviation less of the SDMT mean of the control group as the limit for CI. The cutoff value of the GCL/IPL variable that could predict CI was calculated by ROC analysis, and the ability to accurately predict CI was tested with binary logistic regression. RESULTS: No correlation was found between OCT parameters and CP in healthy subjects. Correlation was found between GCL thickness and GCL/IPL variable and CP in RRMS patients (r=0.235, r=0.667 respectively). A GCL/IPL value of 1.255 was able to identify CI with 81.8% sensitivity and 75.9% specificity (AUC=0.844, LR=3.38) and predicted CI with 74.5% accuracy (Nagelkerke R2=0.439). CONCLUSION: In RRMS patients, the IPL thickness is unrelated to CP. Therewithal, the GCL/IPL-CP relationship is stronger than the GCL-CP relationship and GCL/IPL can predict CI.


Asunto(s)
Esclerosis Múltiple , Neuritis Óptica , Humanos , Células Ganglionares de la Retina , Esclerosis Múltiple/complicaciones , Esclerosis Múltiple/diagnóstico por imagen , Tomografía de Coherencia Óptica , Retina/diagnóstico por imagen
19.
J Cereb Blood Flow Metab ; 44(4): 477-490, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37988123

RESUMEN

Neuronal loss in the ipsilateral thalamus after focal cortical infarction participates in post-stroke cognitive deficits, and enhanced angiogenesis in the thalamus is expected to reduce neuronal damage. We hypothesize that novel translocator protein (TSPO) ligand, 2-Cl-MGV-1, can promote angiogenesis, attenuate neuronal loss in the thalamus, and ameliorate post-stroke cognitive deficits. Cortical infarction was induced by distal middle cerebral artery occlusion (dMCAO) in stroke-prone renovascular hypertensive rats. 2-Cl-MGV-1 or dimethyl sulfoxide was administered 24 h after dMCAO and then for 6 or 13 days. Spatial learning and memory were assessed using the Morris water maze. Neuronal loss, TSPO expression, angiogenesis, and intrinsic pathway were determined by immunofluorescence and immunoblotting 7 and 14 days after dMCAO. Cortical infarction caused post-stroke cognitive deficits and secondary neuronal loss with gliosis in the ipsilateral thalamus within 14 days of dMCAO. Increased angiogenesis and elevated expression of vascular TSPO were detected in the ipsilateral thalamus, and treatment with 2-Cl-MGV-1 enhanced angiogenesis by stimulating the PI3K-AKT-mTOR pathway. The effects of 2-Cl-MGV-1 on angiogenesis coincided with reduced neuronal loss in the thalamus and contributed to improvements in post-stroke cognitive deficits. Our findings suggest that 2-Cl-MGV-1 stimulates angiogenesis, ameliorates neuronal loss in the thalamus, and improves post-stroke cognitive deficits.


Asunto(s)
Angiogénesis , Carbamatos , Quinazolinas , Accidente Cerebrovascular , Ratas , Animales , Ratas Sprague-Dawley , Ligandos , Fosfatidilinositol 3-Quinasas/metabolismo , Infarto de la Arteria Cerebral Media/complicaciones , Accidente Cerebrovascular/metabolismo , Tálamo/metabolismo , Cognición
20.
J Ethnopharmacol ; 323: 117679, 2024 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-38160863

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: SuanZaoRen Decoction (SZRD), a famous herbal prescription, and has been widely proven to have positive therapeutic effects on insomnia, depression and Alzheimer's disease (AD). However, the anti-AD molecular mechanism of SZRD remains to be further investigated. AIM OF THE STUDY: To elucidate the molecular mechanism of SZRD's improvement in AD's neuronal loss, synaptic damage and ferroptosis by regulating DJ-1/Nrf2 signaling pathway. MATERIALS AND METHODS: LC-MS/MS was used to detect the active ingredients from SZRD. APP/PS1 mice was treated with SZRD and a ferroptosis inhibitor (Liproxstatin-1), respectively. Upon the completion of behavioral tests, Nissl staining, FJB staining, Golgi staining, immunofluorescence, immunohistochemistry, and transmission electron microscopy were preformed to evaluate the effects of SZRD on neuronal loss, synaptic damage, Aß deposition. Iron staining, transmission electron microscopy, and iron assay kit was performed to estimate the effects of SZRD on ferroptosis. SOD kit, MDA kit, GSH kit, and GSH/GSSG kit were utilized to measure the oxidative stress levels in the hippocampus. The protein expression of TfR1, FTH1, FTL, FPN1, DJ-1, Nrf2, GPX4, SLC7A11, and ACSL4 were detected by Western blot. RESULTS: A total of 16 active ingredients were identified from SZRD extract. SZRD SZRD significantly alleviated learning and memory impairment in APP/PS1 mice. SZRD improved the hippocampal neuronal loss and degenerated neurons in APP/PS1 mice via inhibiting the Aß deposit. SZRD mitigated the hippocampal synaptic damage in APP/PS1 mice. SZRD inhibited iron accumulation, and alleviated the oxidative stress level in the hippocampus of APP/PS1 mice. Meanwhile, SZRD could up-regulate the protein expression level of FPN1, DJ-1, Nrf2, GPX4 and SLC7A11 in the hippocampus, and inhibit TfR1, FTH1, FTL, and ACSL4 protein expression. CONCLUSION: SZRD alleviated neuronal loss, synaptic damage and ferroptosis in AD via activating DJ-1/Nrf2 signaling pathway.


Asunto(s)
Enfermedad de Alzheimer , Medicamentos Herbarios Chinos , Ferroptosis , Animales , Ratones , Cromatografía Liquida , Factor 2 Relacionado con NF-E2 , Espectrometría de Masas en Tándem , Transducción de Señal , Hierro
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA