Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 995
Filtrar
1.
J Nanobiotechnology ; 22(1): 251, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38750597

RESUMEN

BACKGROUND: Hypothermia is a promising therapy for traumatic brain injury (TBI) in the clinic. However, the neuroprotective outcomes of hypothermia-treated TBI patients in clinical studies are inconsistent due to several severe side effects. Here, an injectable refrigerated hydrogel was designed to deliver 3-iodothyronamine (T1AM) to achieve a longer period of local hypothermia for TBI treatment. Hydrogel has four advantages: (1) It can be injected into injured sites after TBI, where it forms a hydrogel and avoids the side effects of whole-body cooling. (2) Hydrogels can biodegrade and be used for controlled drug release. (3) Released T1AM can induce hypothermia. (4) This hydrogel has increased medical value given its simple operation and ability to achieve timely treatment. METHODS: Pol/T hydrogels were prepared by a low-temperature mixing method and characterized. The effect of the Pol/T hydrogel on traumatic brain injury in mice was studied. The degradation of the hydrogel at the body level was observed with a small animal imager. Brain temperature and body temperature were measured by brain thermometer and body thermometer, respectively. The apoptosis of peripheral nerve cells was detected by immunohistochemical staining. The protective effect of the hydrogels on the blood-brain barrier (BBB) after TBI was evaluated by the Evans blue penetration test. The protective effect of hydrogel on brain edema after injury in mice was detected by Magnetic resonance (MR) in small animals. The enzyme linked immunosorbent assay (ELISA) method was used to measure the levels of inflammatory factors. The effects of behavioral tests on the learning ability and exercise ability of mice after injury were evaluated. RESULTS: This hydrogel was able to cool the brain to hypothermia for 12 h while maintaining body temperature within the normal range after TBI in mice. More importantly, hypothermia induced by this hydrogel leads to the maintenance of BBB integrity, the prevention of cell death, the reduction of the inflammatory response and brain edema, and the promotion of functional recovery after TBI in mice. This cooling method could be developed as a new approach for hypothermia treatment in TBI patients. CONCLUSION: Our study showed that injectable and biodegradable frozen Pol/T hydrogels to induce local hypothermia in TBI mice can be used for the treatment of traumatic brain injury.


Asunto(s)
Barrera Hematoencefálica , Lesiones Traumáticas del Encéfalo , Hidrogeles , Hipotermia Inducida , Animales , Lesiones Traumáticas del Encéfalo/terapia , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Ratones , Hidrogeles/química , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/efectos de los fármacos , Masculino , Hipotermia Inducida/métodos , Neuroprotección/efectos de los fármacos , Encéfalo/patología , Modelos Animales de Enfermedad , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/química , Temperatura Corporal , Ratones Endogámicos C57BL
3.
Neurosurg Rev ; 47(1): 193, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38662220

RESUMEN

This critique examines a 12-year retrospective study on serum magnesium concentration-guided administration of magnesium sulfate in 548 patients with aneurysmal subarachnoid hemorrhage (aSAH). The study reported that maintaining serum magnesium levels between 2 and 2.5 mmol/L reduced rates of delayed cerebral infarction and improved clinical outcomes. However, limitations due to its retrospective nature, single-center design, and unequal treatment group sizes may affect generalizability. Future multicentric randomized controlled trials are recommended to validate these findings and refine magnesium dosing strategies for aSAH treatment.


Asunto(s)
Sulfato de Magnesio , Fármacos Neuroprotectores , Hemorragia Subaracnoidea , Humanos , Hemorragia Subaracnoidea/tratamiento farmacológico , Hemorragia Subaracnoidea/complicaciones , Sulfato de Magnesio/administración & dosificación , Estudios Retrospectivos , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/uso terapéutico , Resultado del Tratamiento , Femenino , Administración Intravenosa , Persona de Mediana Edad , Masculino , Neuroprotección/efectos de los fármacos , Infarto Cerebral/prevención & control , Infarto Cerebral/tratamiento farmacológico , Adulto
4.
Acta Neuropathol Commun ; 12(1): 65, 2024 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-38649962

RESUMEN

The progressive and irreversible degeneration of retinal ganglion cells (RGCs) and their axons is the major characteristic of glaucoma, a leading cause of irreversible blindness worldwide. Nicotinamide adenine dinucleotide (NAD) is a cofactor and metabolite of redox reaction critical for neuronal survival. Supplementation with nicotinamide (NAM), a precursor of NAD, can confer neuroprotective effects against glaucomatous damage caused by an age-related decline of NAD or mitochondrial dysfunction, reflecting the high metabolic activity of RGCs. However, oral supplementation of drug is relatively less efficient in terms of transmissibility to RGCs compared to direct delivery methods such as intraocular injection or delivery using subconjunctival depots. Neither method is ideal, given the risks of infection and subconjunctival scarring without novel techniques. By contrast, extracellular vesicles (EVs) have advantages as a drug delivery system with low immunogeneity and tissue interactions. We have evaluated the EV delivery of NAM as an RGC protective agent using a quantitative assessment of dendritic integrity using DiOlistics, which is confirmed to be a more sensitive measure of neuronal health in our mouse glaucoma model than the evaluation of somatic loss via the immunostaining method. NAM or NAM-loaded EVs showed a significant neuroprotective effect in the mouse retinal explant model. Furthermore, NAM-loaded EVs can penetrate the sclera once deployed in the subconjunctival space. These results confirm the feasibility of using subconjunctival injection of EVs to deliver NAM to intraocular targets.


Asunto(s)
Vesículas Extracelulares , Glaucoma , Ratones Endogámicos C57BL , Fármacos Neuroprotectores , Niacinamida , Células Ganglionares de la Retina , Animales , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/efectos de los fármacos , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/metabolismo , Niacinamida/administración & dosificación , Niacinamida/farmacología , Ratones , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/farmacología , Glaucoma/metabolismo , Glaucoma/tratamiento farmacológico , Neuroprotección/efectos de los fármacos , Esclerótica/metabolismo , Esclerótica/efectos de los fármacos , Sistemas de Liberación de Medicamentos/métodos , Masculino
5.
Brain Res ; 1834: 148906, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38570152

RESUMEN

Alzherimer's disease (AD) is an age-dependent ubiquitous ailment worldwide with limited therapies that only alleviate the symptoms of AD but do not cure them entirely because of the restricted blood-brain barrier passage of the drug. Hence with new advanced technology, nanoparticles can offer an opportunity as the active candidate to overcome the above limitations. Aurothioglucose, a synthetic glucose derivative of the gold compound, has been clinically proven to be an effective anti-inflammatory drug for rheumatic arthritis. Recently, several scientific groups have developed gold nanoparticle preparations and tested them for the treatment of dementia. This study was planned to prepare the PLGA nanoparticles of aurothioglucose (ATG) and check the neuroprotective potential against STZ-induced AD in rats. The nanoparticles were prepared using the double emulsion solvent evaporation method and characterized for various parameters such as drug-excipient interaction, particle size, zeta potential, and morphology. Then, rats were injected STZ (3 mg/kg/i.c.v., days 1 and 3) and ATG (5 and 10 mg/kg/s.c.), ATG NPs (2.5 and 5 mg/kg/s.c.) and donepezil (2 mg/kg/p.o) from 15th to 29th day. Behavior parameters were performed using an actophotometer, MWM, and ORT. On the 30th day, all the animals were sacrificed, and the brains were isolated for estimating biochemical, neurochemical, and proinflammatory markers. It was observed that ATG NPs significantly restored all behavior and neurotransmitter alterations caused by STZ. Also, it increased antioxidant levels and decreased inflammatory cytokines significantly, then ATG alone. Thus, the study suggests that ATG loaded PLGA NPs could be used as a novel therapeutic strategy to slow the process of AD.


Asunto(s)
Enfermedad de Alzheimer , Nanopartículas , Fármacos Neuroprotectores , Estreptozocina , Animales , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Ratas , Fármacos Neuroprotectores/farmacología , Estreptozocina/farmacología , Masculino , Nanopartículas/administración & dosificación , Ratas Wistar , Neuroprotección/efectos de los fármacos , Modelos Animales de Enfermedad , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo
6.
Basic Clin Pharmacol Toxicol ; 134(6): 770-777, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38566316

RESUMEN

Neurodegenerative diseases, such as Alzheimer's disease and Parkinson's disease, are characterised by the progressive loss of specific neuronal cell populations due to multifactorial factors, including neurochemical and immunological disturbances. Consequently, patients can develop cognitive, motor and behavioural dysfunctions, which lead to impairments in their quality of life. Over the years, studies have reported on the neuroprotective properties inherent in phenolic compounds. Therefore, this review highlights the most recent scientific findings regarding phenolic compounds as promising neuroprotective molecules against neurodegenerative diseases.


Asunto(s)
Fármacos Neuroprotectores , Fenoles , Fármacos Neuroprotectores/farmacología , Animales , Humanos , Fenoles/farmacología , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedad de Alzheimer/tratamiento farmacológico , Modelos Animales de Enfermedad , Enfermedad de Parkinson/tratamiento farmacológico , Neuroprotección/efectos de los fármacos
7.
Cell Rep ; 43(4): 113980, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38520693

RESUMEN

In the brain, the role of matrilin-3, an extracellular matrix component in cartilage, is unknown. Here, we identify that matrilin-3 decreased in reactive astrocytes but was unchanged in neurons after ischemic stroke in animals. Importantly, it declined in serum of patients with acute ischemic stroke. Genetic or pharmacological inhibition or supplementation of matrilin-3 aggravates or reduces brain injury, astrocytic cell death, and glial scar, respectively, but has no direct effect on neuronal cell death. RNA sequencing demonstrates that Matn3-/- mice display an increased inflammatory response profile in the ischemic brain, including the nuclear factor κB (NF-κB) signaling pathway. Both endogenous and exogenous matrilin-3 reduce inflammatory mediators. Mechanistically, extracellular matrilin-3 enters astrocytes via caveolin-1-mediated endocytosis. Cytoplasmic matrilin-3 translocates into the nucleus by binding to NF-κB p65, suppressing inflammatory cytokine transcription. Extracellular matrilin-3 binds to BMP-2, blocking the BMP-2/Smads pathway. Thus, matrilin-3 is required for astrocytes to exert neuroprotection, at least partially, by suppressing astrocyte-mediated neuroinflammation.


Asunto(s)
Astrocitos , Accidente Cerebrovascular Isquémico , Proteínas Matrilinas , Ratones Endogámicos C57BL , Enfermedades Neuroinflamatorias , Neuroprotección , Animales , Humanos , Masculino , Ratones , Astrocitos/metabolismo , Astrocitos/efectos de los fármacos , Accidente Cerebrovascular Isquémico/metabolismo , Accidente Cerebrovascular Isquémico/patología , Proteínas Matrilinas/metabolismo , Ratones Noqueados , Enfermedades Neuroinflamatorias/metabolismo , Enfermedades Neuroinflamatorias/patología , Neuroprotección/efectos de los fármacos , FN-kappa B/metabolismo , Transducción de Señal
8.
FEBS Lett ; 598(9): 995-1007, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38413095

RESUMEN

Resveratrol prevents various neurodegenerative diseases in animal models despite reaching only low nanomolar concentrations in the brain after oral administration. In this study, based on the quenching of intrinsic tryptophan fluorescence and molecular docking, we found that trans-resveratrol, its conjugates (glucuronide and sulfate), and dihydro-resveratrol (intestinal microbial metabolite) bind with high affinities (Kd, 0.2-2 nm) to the peptide G palindromic sequence (near glycosaminoglycan-binding motif) of the 67-kDa laminin receptor (67LR). Preconditioning with low concentrations (0.01-10 nm) of these polyphenols, especially resveratrol-glucuronide, protected neuronal cells from death induced by serum withdrawal via activation of cAMP-mediated signaling pathways. This protection was prevented by a 67LR-blocking antibody, suggesting a role for this cell-surface receptor in neuroprotection by resveratrol metabolites.


Asunto(s)
Fármacos Neuroprotectores , Receptores de Laminina , Resveratrol , Resveratrol/farmacología , Resveratrol/metabolismo , Resveratrol/química , Receptores de Laminina/metabolismo , Receptores de Laminina/genética , Humanos , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/metabolismo , Simulación del Acoplamiento Molecular , Animales , Unión Proteica , Neuronas/metabolismo , Neuronas/efectos de los fármacos , Estilbenos/farmacología , Estilbenos/metabolismo , Estilbenos/química , Neuroprotección/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Sitios de Unión , Glucurónidos/metabolismo , Glucurónidos/química , Proteínas Ribosómicas
10.
Phytomedicine ; 119: 155015, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37597362

RESUMEN

BACKGROUND: Depression is a severe mental illness that endangers human health. Depressed individuals are prone to sleep less and to the loss of appetite for food; their thinking and cognition processes, as well as mood, may even be affected. Danzhi Xiaoyao San (DXS), documented in the Internal Medicine Summary, has been used for hundreds of years in China and is widely applied traditionally to treat liver qi stagnation, liver and spleen blood deficiency, menstrual disorders, and spontaneous and night sweating. DXS can also clear heat and drain the liver. Presently, it is used frequently in the treatment of depression based on its ability to clear the liver and alleviate depression. PURPOSE: To summarize clinical and preclinical studies on the antidepressant-like effects of DXS, understand the material basis and mechanisms of these effects, and offer new suggestions and methods for the clinical treatment of depression. METHODS: "Danzhi Xiaoyao", "Danzhixiaoyao", "Xiaoyao", "depression" and active ingredients were entered as keywords in PubMed, Google Scholar, CNKI and WANFANG DATA databases in the search for material on DXS and its active ingredients. The PRISMA guidelines were followed in this review process. RESULTS: Per clinical reports, DXS has a therapeutic effect on patients with depression but few side effects. DXS and its active ingredients allegedly produce their neuroprotective antidepressant-like effects by modulating monoamine neurotransmitter levels, inhibiting the hypothalamic-pituitary-adrenal (HPA) axis hyperfunction, reducing neuroinflammation and increasing neurotrophic factors. CONCLUSION: Overall, DXS influences multiple potential mechanisms to exert its antidepressant-like effects thanks to its multicomponent character. Because depression is not caused by a single mechanism, probing the antidepressant-like effects of DXS could further help understand the pathogenesis of depression and discover new antidepressant drugs.


Asunto(s)
Antidepresivos , Medicina Tradicional China , Antidepresivos/química , Antidepresivos/farmacología , Humanos , Animales , Neurotransmisores/química , Neurotransmisores/farmacología , Neuroprotección/efectos de los fármacos , Metabolómica
11.
Adv Neurobiol ; 32: 271-313, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37480464

RESUMEN

Environmental temperature adversely affects the outcome of concussive head injury (CHI)-induced brain pathology. Studies from our laboratory showed that animals reared at either cold environment or at hot environment exacerbate brain pathology following CHI. Our previous experiments showed that nanowired delivery of oxiracetam significantly attenuated CHI-induced brain pathology and associated neurovascular changes. Military personnel are the most susceptible to CHI caused by explosion, blasts, missile or blunt head trauma leading to lifetime functional and cognitive impairments affecting the quality of life. Severe CHI leads to instant death and/or lifetime paralysis. Military personnel engaged in combat operations are often subjected to extreme high or low environmental temperature zones across the globe. Thus, further exploration of novel therapeutic agents at cold or hot ambient temperatures following CHI are the need of the hour. CHI is also a major risk factor for developing Alzheimer's disease by enhancing amyloid beta peptide deposits in the brain. In this review, effect of hot environment on CHI-induced brain pathology is discussed. In addition, whether nanodelivery of oxiracetam together with neprilysin and monoclonal antibodies (mAb) to amyloid beta peptide and p-tau could lead to superior neuroprotection in CHI is explored. Our results show that co-administration of oxiracetam with neprilysin and mAb to AßP and p-tau significantly induced superior neuroprotection following CHI in hot environment, not reported earlier.


Asunto(s)
Anticuerpos Monoclonales , Lesiones Traumáticas del Encéfalo , Neprilisina , Pirrolidinas , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/patología , Animales , Calor , Pirrolidinas/administración & dosificación , Humanos , Nanocables/química , Encéfalo/patología , Neprilisina/administración & dosificación , Anticuerpos Monoclonales/administración & dosificación , Péptidos beta-Amiloides/metabolismo , Proteínas tau/metabolismo , Neuroprotección/efectos de los fármacos
12.
Int J Biol Macromol ; 234: 123669, 2023 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-36796555

RESUMEN

Ulvans are water-soluble sulfated polysaccharides predominantly found in the cell wall of green algae. They hold unique characteristics that are attributed to their 3D conformation, functional groups along with the presence of saccharides and sulfate ions. Traditionally, ulvans are widely used as food supplements and probiotics owing to the high content of carbohydrates. Despite their widespread usage in food industry, an in-depth understanding is required for extrapolating their potential application as a nutraceutical and medicinal agent which could be beneficial in promoting human health and well-being. This review emphasizes novel therapeutic avenues where ulvan polysaccharides can be used beyond their nutritional applications. A collection of literature points towards multifarious applications of ulvan in various biomedical fields. Structural aspects along with extraction and purification methods have been discussed. The underlying molecular mechanisms associated with its biomedical potential in different therapeutic fields like oncology, infectious diseases, inflammation, neuroprotection and tissue engineering, etc. have been unravelled. Challenges associated with clinical translation and future perspectives have been deliberated.


Asunto(s)
Productos Biológicos , Polisacáridos , Animales , Humanos , Productos Biológicos/química , Productos Biológicos/farmacología , Productos Biológicos/uso terapéutico , Chlorophyta/química , Suplementos Dietéticos , Polisacáridos/farmacología , Polisacáridos/uso terapéutico , Polisacáridos/química , Neoplasias/tratamiento farmacológico , Cicatrización de Heridas/efectos de los fármacos , Infecciones/tratamiento farmacológico , Neuroprotección/efectos de los fármacos , Antiinflamatorios/farmacología , Antioxidantes/farmacología , Ciencia Traslacional Biomédica , Anticoagulantes/farmacología , Ingeniería de Tejidos , Regeneración/efectos de los fármacos
13.
Int J Mol Sci ; 24(4)2023 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-36835155

RESUMEN

Neurodegenerative diseases (NDs) affect the West due to the increase in life expectancy. Nervous cells accumulate oxidative damage, which is one of the factors that triggers and accelerates neurodegeneration. However, cells have mechanisms that scavenge reactive oxygen species (ROS) and alleviate oxidative stress (OS). Many of these endogenous antioxidant systems are regulated at the gene expression level by the transcription factor Nrf2 (nuclear factor erythroid 2-related factor 2). In the presence of prooxidant conditions, Nrf2 translocates to the nucleus and induces the transcription of genes containing ARE (antioxidant response element). In recent years, there has been an increase in the study of the Nrf2 pathway and the natural products that positively regulate it to reduce oxidative damage to the nervous system, both in in vitro models with neurons and microglia subjected to stress factors and in vivo models using mainly murine models. Quercetin, curcumin, anthocyanins, tea polyphenols, and other less studied phenolic compounds such as kaempferol, hesperetin, and icariin can also modulate Nrf2 by regulating several Nrf2 upstream activators. Another group of phytochemical compounds that upregulate this pathway are terpenoids, including monoterpenes (aucubin, catapol), diterpenes (ginkgolides), triterpenes (ginsenosides), and carotenoids (astaxanthin, lycopene). This review aims to update the knowledge on the influence of secondary metabolites of health interest on the activation of the Nrf2 pathway and their potential as treatments for NDs.


Asunto(s)
Productos Biológicos , Factor 2 Relacionado con NF-E2 , Neuroprotección , Animales , Ratones , Antocianinas/metabolismo , Antioxidantes/farmacología , Productos Biológicos/farmacología , Neuroprotección/efectos de los fármacos , Factor 2 Relacionado con NF-E2/efectos de los fármacos , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos
14.
Neurol India ; 70(4): 1601-1609, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36076665

RESUMEN

Background: Neuroprotection in traumatic brain injury (TBI) is an unmet medical need. Objective: We evaluated two agents, aglepristone (progesterone receptor antagonist) and N-salicyloyltryptamine (STP) (activator of Maxi-K channel in GH3 cells), for neuroprotection in Feeney's weight drop model of TBI. Material and Methods: Forty-eight male Wistar rats were divided into six groups (n = 8 per group). A battery of six neurobehavioral tests was evaluated at the end of the first week (EO1W), second week (EO2W), and third week (EO3W). In addition, histopathological and immunohistochemistry (BAX, Bcl-2, and M30 Cytodeath) tests were performed at EO3W. Results: Aglepristone at 10 mg/kg showed significant neuroprotection compared to control as assessed by Rota-rod test at EO1W, VEFP right paw and 28-point neurobehavioral test at EO2W, MWM test at EO3W, and positive histopathological and IHC findings. Aglepristone at 20 mg/kg showed negative results as assessed by BAX expression, downregulation of Bcl-2, and positive M30 Cytodeath, thereby suggesting toxicity at higher doses. STP 100 mg/kg showed modest neuroprotective activity but failed to show a dose-response relationship at a dose of 50 mg/kg. Conclusion: The study shows that progesterone receptor antagonists have neuroprotection at lower doses and toxicity at higher doses.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Lesiones Encefálicas , Canales de Potasio de Gran Conductancia Activados por el Calcio , Fármacos Neuroprotectores , Receptores de Progesterona , Animales , Lesiones Encefálicas/patología , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Proteínas Portadoras , Modelos Animales de Enfermedad , Canales de Potasio de Gran Conductancia Activados por el Calcio/agonistas , Masculino , Neuroprotección/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Receptores de Progesterona/antagonistas & inhibidores , Proteína X Asociada a bcl-2/metabolismo
15.
Cells ; 11(15)2022 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-35892581

RESUMEN

Alzheimer's disease (AD) is a neurodegenerative disorder, and no effective treatments are available to treat this disorder. Therefore, researchers have been investigating Hericium erinaceus, or the monkey head mushroom, an edible medicinal mushroom, as a possible treatment for AD. In this narrative review, we evaluated six preclinical and three clinical studies of the therapeutic effects of Hericium erinaceus on AD. Preclinical trials have successfully demonstrated that extracts and bioactive compounds of Hericium erinaceus have potential beneficial effects in ameliorating cognitive functioning and behavioral deficits in animal models of AD. A limited number of clinical studies have been conducted and several clinical trials are ongoing, which have thus far shown analogous outcomes to the preclinical studies. Nonetheless, future research on Hericium erinaceus needs to focus on elucidating the specific neuroprotective mechanisms and the target sites in AD. Additionally, standardized treatment parameters and universal regulatory systems need to be established to further ensure treatment safety and efficacy. In conclusion, Hericium erinaceus has therapeutic potential and may facilitate memory enhancement in patients with AD.


Asunto(s)
Enfermedad de Alzheimer , Hericium , Memoria , Enfermedad de Alzheimer/tratamiento farmacológico , Animales , Extractos Celulares/farmacología , Extractos Celulares/uso terapéutico , Modelos Animales de Enfermedad , Hericium/química , Humanos , Memoria/efectos de los fármacos , Neuroprotección/efectos de los fármacos
16.
Acta Pharmacol Sin ; 43(10): 2527-2541, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35347247

RESUMEN

Oxidative stress is extensively involved in neurodegeneration. Clinical evidence shows that keeping the mind active through mentally-stimulating physical activities can effectively slow down the progression of neurodegeneration. With increased physical activities, more neurotransmitters would be released in the brain. In the present study, we investigated whether some of the released neurotransmitters might have a beneficial effect against oxidative neurodegeneration in vitro. Glutamate-induced, glutathione depletion-associated oxidative cytotoxicity in HT22 mouse hippocampal neuronal cells was used as an experimental model. We showed that norepinephrine (NE, 50 µM) or dopamine (DA, 50 µM) exerted potent protective effect against glutamate-induced cytotoxicity, but this effect was not observed when other neurotransmitters such as histamine, γ-aminobutyric acid, serotonin, glycine and acetylcholine were tested. In glutamate-treated HT22 cells, both NE and DA significantly suppressed glutathione depletion-associated mitochondrial dysfunction including mitochondrial superoxide accumulation, ATP depletion and mitochondrial AIF release. Moreover, both NE and DA inhibited glutathione depletion-associated MAPKs activation, p53 phosphorylation and GADD45α activation. Molecular docking analysis revealed that NE and DA could bind to protein disulfide isomerase (PDI). In biochemical enzymatic assay in vitro, NE and DA dose-dependently inhibited the reductive activity of PDI. We further revealed that the protective effect of NE and DA against glutamate-induced oxidative cytotoxicity was mediated through inhibition of PDI-catalyzed dimerization of the neuronal nitric oxide synthase. Collectively, the results of this study suggest that NE and DA may have a protective effect against oxidative neurodegeneration through inhibition of protein disulfide isomerase and the subsequent activation of the MAPKs‒p53‒GADD45α oxidative cascade.


Asunto(s)
Muerte Celular , Dopamina , Neuroprotección , Norepinefrina , Proteína Disulfuro Isomerasas , Acetilcolina/farmacología , Adenosina Trifosfato/metabolismo , Animales , Muerte Celular/efectos de los fármacos , Dopamina/farmacología , Ácido Glutámico/metabolismo , Glutatión/metabolismo , Glicina/farmacología , Histamina/metabolismo , Ratones , Simulación del Acoplamiento Molecular , Neuroprotección/efectos de los fármacos , Neurotransmisores , Óxido Nítrico Sintasa de Tipo I/metabolismo , Norepinefrina/farmacología , Estrés Oxidativo , Proteína Disulfuro Isomerasas/efectos de los fármacos , Proteína Disulfuro Isomerasas/metabolismo , Serotonina/metabolismo , Serotonina/farmacología , Superóxidos/metabolismo , Superóxidos/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Ácido gamma-Aminobutírico/metabolismo
17.
Mar Drugs ; 20(3)2022 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-35323490

RESUMEN

Oxidative stress has been implicated in the etiology of Parkinson's disease (PD). Molecules non-covalently binding to the Keap1-Nrf2 complex could be a promising therapeutic approach for PD. Herein, two novel prenylated indole alkaloids asperpenazine (1), and asperpendoline (2) with a scarce skeleton of pyrimido[1,6-a]indole were discovered from the co-cultivated fungi of Aspergillus ochraceus MCCC 3A00521 and Penicillium sp. HUBU 0120. Compound 2 exhibited potential neuroprotective activity on SH-SY5Y cells against oxidative stress. Molecular mechanism research demonstrated that 2 inhibited Keap1 expression, resulting in the translocation of Nrf2 from the cytoplasm to the nucleus, activating the downstream genes expression of HO-1 and NQO1, leading to the reduction in reactive oxygen species (ROS) and the augment of glutathione. Molecular docking and dynamic simulation analyses manifested that 2 interacted with Keap1 (PDB ID: 1X2R) via forming typical hydrogen and hydrophobic bonds with residues and presented less fluctuation of RMSD and RMSF during a natural physiological condition.


Asunto(s)
Alcaloides Indólicos/farmacología , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Fármacos Neuroprotectores/farmacología , Aspergillus ochraceus/química , Aspergillus ochraceus/metabolismo , Línea Celular Tumoral , Glutatión/metabolismo , Humanos , Alcaloides Indólicos/química , Alcaloides Indólicos/metabolismo , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Estructura Molecular , Neuroprotección/efectos de los fármacos , Fármacos Neuroprotectores/química , Fármacos Neuroprotectores/metabolismo , Estrés Oxidativo/efectos de los fármacos , Penicillium/química , Penicillium/metabolismo , Prenilación
18.
Int J Mol Sci ; 23(4)2022 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-35216504

RESUMEN

Spinal cord injury (SCI) causes significant mortality and morbidity. Currently, no FDA-approved pharmacotherapy is available for treating SCI. Previously, low doses of estrogen (17ß-estradiol, E2) were shown to improve the post-injury outcome in a rat SCI model. However, the range of associated side effects makes advocating its therapeutic use difficult. Therefore, this study aimed at investigating the therapeutic efficacy of Premarin (PRM) in SCI. PRM is an FDA-approved E2 (10%) formulation, which is used for hormone replacement therapy with minimal risk of serious side effects. The effects of PRM on SCI were examined by magnetic resonance imaging, immunofluorescent staining, and western blot analysis in a rat model. SCI animals treated with vehicle alone, PRM, E2 receptor antagonist (ICI), or PRM + ICI were graded in a blinded way for locomotor function by using the Basso-Beattie-Bresnahan (BBB) locomotor scale. PRM treatment for 7 days decreased post-SCI lesion volume and attenuated neuronal cell death, inflammation, and axonal damage. PRM also altered the balance of pro- and anti-apoptotic proteins in favor of cell survival and improved angiogenesis and microvascular growth. Increased expression of estrogen receptors (ERs) ERα and ERß following PRM treatment and their inhibition by ER inhibitor indicated that the neuroprotection associated with PRM treatment might be E2-receptor mediated. The attenuation of glial activation with decreased inflammation and cell death, and increased angiogenesis by PRM led to improved functional outcome as determined by the BBB locomotor scale. These results suggest that PRM treatment has significant therapeutic implications for the improvement of post-SCI outcome.


Asunto(s)
Estrógenos Conjugados (USP)/farmacología , Enfermedades Neurodegenerativas/tratamiento farmacológico , Traumatismos de la Médula Espinal/tratamiento farmacológico , Animales , Axones/efectos de los fármacos , Axones/metabolismo , Modelos Animales de Enfermedad , Estradiol/metabolismo , Estrógenos/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Locomoción/efectos de los fármacos , Masculino , Actividad Motora/efectos de los fármacos , Enfermedades Neurodegenerativas/metabolismo , Neuroprotección/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Recuperación de la Función/efectos de los fármacos , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/metabolismo
19.
Mol Neurobiol ; 59(1): 574-589, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34735672

RESUMEN

Phosphodiesterase 10A (PDE10A) hydrolyzes adenosine 3',5'-cyclic monophosphate (cAMP) and guanosine 3',5'-cyclic monophosphate (cGMP). It is highly expressed in the striatum. Recent evidence implied that PDE10A may be involved in the inflammatory processes following injury, such as ischemic stroke. Its role in ischemic injury was unknown. Herein, we exposed mice to 90 or 30-min middle cerebral artery occlusion, followed by the delivery of the highly selective PDE10A inhibitor TAK-063 (0.3 mg/kg or 3 mg/kg) immediately after reperfusion. Animals were sacrificed after 24 or 72 h, respectively. Both TAK-063 doses enhanced neurological function, reduced infarct volume, increased neuronal survival, reduced brain edema, and increased blood-brain barrier integrity, alongside cerebral microcirculation improvements. Post-ischemic neuroprotection was associated with increased phosphorylation (i.e., activation) of pro-survival Akt, Erk-1/2, GSK-3α/ß and anti-apoptotic Bcl-xL abundance, decreased phosphorylation of pro-survival mTOR, and HIF-1α, MMP-9 and pro-apoptotic Bax abundance. Interestingly, PDE10A inhibition reduced inflammatory cytokines/chemokines, including IFN-γ and TNF-α, analyzed by planar surface immunoassay. In addition, liquid chromatography-tandem mass spectrometry revealed 40 proteins were significantly altered by TAK-063. Our study established PDE10A as a target for ischemic stroke therapy.


Asunto(s)
Edema Encefálico/tratamiento farmacológico , Supervivencia Celular/efectos de los fármacos , Accidente Cerebrovascular Isquémico/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Inhibidores de Fosfodiesterasa/uso terapéutico , Hidrolasas Diéster Fosfóricas/metabolismo , Animales , Edema Encefálico/metabolismo , Modelos Animales de Enfermedad , Accidente Cerebrovascular Isquémico/metabolismo , Ratones , Microcirculación/efectos de los fármacos , Neuroprotección/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Inhibidores de Fosfodiesterasa/farmacología , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirazoles/farmacología , Pirazoles/uso terapéutico , Piridazinas/farmacología , Piridazinas/uso terapéutico , Transducción de Señal/efectos de los fármacos
20.
Ann N Y Acad Sci ; 1507(1): 49-59, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34060087

RESUMEN

Cardiac arrest has many implications for morbidity and mortality. Few interventions have been shown to improve return of spontaneous circulation (ROSC) and long-term outcomes after cardiac arrest. Ischemic-reperfusion injury upon achieving ROSC creates an imbalance between oxygen supply and demand. Multiple events occur in the postcardiac arrest period, including excitotoxicity, mitochondrial dysfunction, and oxidative stress and inflammation, all of which contribute to ongoing brain injury and cellular death. Given that complex pathophysiology underlies global brain hypoxic ischemia, neuroprotective strategies targeting multiple stages of the neuropathologic cascade should be considered as a means of mitigating secondary neuronal injury and improving neurologic outcomes and survival in cardiac arrest victims. In this review article, we discuss a number of different pharmacologic agents that may have a potential role in targeting these injurious pathways following cardiac arrest. Pharmacologic therapies most relevant for discussion currently include memantine, perampanel, magnesium, propofol, thiamine, methylene blue, vitamin C, vitamin E, coenzyme Q10 , minocycline, steroids, and aspirin.


Asunto(s)
Lesiones Encefálicas/prevención & control , Isquemia Encefálica/prevención & control , Paro Cardíaco/tratamiento farmacológico , Neuroprotección/efectos de los fármacos , Fármacos Neuroprotectores/administración & dosificación , Animales , Antioxidantes/administración & dosificación , Lesiones Encefálicas/etiología , Lesiones Encefálicas/metabolismo , Isquemia Encefálica/etiología , Isquemia Encefálica/metabolismo , Paro Cardíaco/complicaciones , Paro Cardíaco/metabolismo , Humanos , Memantina/administración & dosificación , Neuroprotección/fisiología , Nitrilos/administración & dosificación , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Piridonas/administración & dosificación , Tiamina/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...