Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 855
Filtrar
1.
Nat Commun ; 15(1): 3987, 2024 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-38734698

RESUMEN

Despite advances in active drug targeting for blood-brain barrier penetration, two key challenges persist: first, attachment of a targeting ligand to the drug or drug carrier does not enhance its brain biodistribution; and second, many brain diseases are intricately linked to microcirculation disorders that significantly impede drug accumulation within brain lesions even after they cross the barrier. Inspired by the neuroprotective properties of vinpocetine, which regulates cerebral blood flow, we propose a molecular library design centered on this class of cyclic tertiary amine compounds and develop a self-enhanced brain-targeted nucleic acid delivery system. Our findings reveal that: (i) vinpocetine-derived ionizable-lipidoid nanoparticles efficiently breach the blood-brain barrier; (ii) they have high gene-loading capacity, facilitating endosomal escape and intracellular transport; (iii) their administration is safe with minimal immunogenicity even with prolonged use; and (iv) they have potent pharmacologic brain-protective activity and may synergize with treatments for brain disorders as demonstrated in male APP/PS1 mice.


Asunto(s)
Barrera Hematoencefálica , Encéfalo , Circulación Cerebrovascular , Nanopartículas , Alcaloides de la Vinca , Animales , Alcaloides de la Vinca/farmacología , Alcaloides de la Vinca/farmacocinética , Alcaloides de la Vinca/administración & dosificación , Alcaloides de la Vinca/química , Nanopartículas/química , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/efectos de los fármacos , Ratones , Circulación Cerebrovascular/efectos de los fármacos , Masculino , Encéfalo/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/irrigación sanguínea , Humanos , Fármacos Neuroprotectores/farmacocinética , Fármacos Neuroprotectores/farmacología , Ratones Endogámicos C57BL , Distribución Tisular , Sistemas de Liberación de Medicamentos , Ratones Transgénicos
2.
Brain Res ; 1834: 148905, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38565372

RESUMEN

Alzheimer's disease (AD) is a primary cause of dementia that affects millions of people worldwide and its prevalence is likely to increase largely in the coming decades. Multiple complex pathways, such as oxidative stress, tau and amyloid-beta (Aß) pathology, and cholinergic dysfunction, are involved in the pathogenesis of Alzheimer's disease. The conventional treatments provide only symptomatic relief and not a complete cure for the disease. On the other hand, recent studies have looked into the possibility of flavonoids as an effective therapeutic strategy for treating AD. Quercetin, a well-known flavonol, has been extensively studied for AD treatment. Therefore, this review mainly focuses on the pharmacokinetics properties of quercetin and its modes of action, such as antioxidant, anti-inflammatory, anti-amyloidogenic, and neuroprotective properties, which are beneficial in treating AD. It also highlights the nano delivery systems of quercetin, including liposomes, nanostructures lipid carriers, solid lipid nanoparticles, nanoemulsions, microemulsions, self-emulsifying drug delivery systems, and nanoparticles reported for AD treatment. The remarkable potential of quercetin nanocarriers has been reflected in enhancing its bioavailability and therapeutic efficacy. Therefore, clinical studies must be conducted to explore it as a therapeutic strategy for Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer , Antioxidantes , Fármacos Neuroprotectores , Quercetina , Quercetina/administración & dosificación , Quercetina/farmacocinética , Quercetina/farmacología , Enfermedad de Alzheimer/tratamiento farmacológico , Humanos , Animales , Antioxidantes/administración & dosificación , Antioxidantes/farmacocinética , Antioxidantes/farmacología , Fármacos Neuroprotectores/farmacocinética , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/farmacología , Sistemas de Liberación de Medicamentos/métodos , Nanopartículas
3.
J Ethnopharmacol ; 330: 118229, 2024 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-38670403

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Thymus quinquecostatus Celak., a member of thymus genus in Lamiaceae family, has been used as a folk medicine for relieving exterior syndrome and alleviating pain in China. The polyphenol-rich fraction (PRF) derived from Thymus quinquecostatus Celak. had been validated that it can protect cerebral ischemia-reperfusion injury (CIRI) by activating Keap1/Nrf2/HO-1 signaling pathway. AIM OF THIS STUDY: To explore effective components and their pharmacokinetic and pharmacodynamic characteristics as well as possible mechanisms of PRF in treating CIRI. MATERIALS AND METHODS: Normal treated group (NTG) and tMCAO model treated group (MTG) rats were administrated PRF intragastrically. The prototype components and metabolites of PRF in plasma and brain were analyzed by the UPLC-Q-Exactive Orbitrap MSn method. Subsequently, the pharmacokinetics properties of indicative components were performed based on HPLC-QQQ-MS/MS. SOD and LDH activities were determined to study the pharmacodynamic (PD) properties of PRF. The PK-PD relationship of PRF was constructed. In addition, the effect of PRF on endogenous metabolites in plasma and brain was investigated using metabolomic method. RESULTS: Salvianic acid A, caffeic acid, rosmarinic acid, scutellarin, and apigenin-7-O-glucuronide were selected as indicative components based on metabolic analysis. The non-compartmental parameters were calculated for indicative components in plasma and brain of NTG and MTG rats. Furthermore, single-component and multi-component PK-PD modeling involved Emax, Imax PD models for effect indexes were fitted as well as ANN models were established, which indicated that these components can work together to regulate SOD and LDH activities in plasma and SOD activity in brain tissue to improve CIRI. Additionally, PRF may ameliorate CIRI by regulating the disorder of endogenous metabolites in lipid metabolism, amino acid metabolism, and purine metabolism pathways in vivo, among which lipid metabolism and purine metabolism are closely related to oxidative stress. CONCLUSION: The PK-PD properties of effect substances and mechanisms of PRF anti-CIRI were further elaborated. The findings provide a convincing foundation for the application of T. quinquecostatus Celak. in the maintenance of human health disorders.


Asunto(s)
Metabolómica , Polifenoles , Ratas Sprague-Dawley , Daño por Reperfusión , Thymus (Planta) , Animales , Masculino , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/metabolismo , Thymus (Planta)/química , Polifenoles/farmacología , Polifenoles/farmacocinética , Ratas , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Extractos Vegetales/farmacología , Extractos Vegetales/farmacocinética , Encéfalo/metabolismo , Encéfalo/efectos de los fármacos , Modelos Animales de Enfermedad , Isquemia Encefálica/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/farmacocinética , Medicamentos Herbarios Chinos/farmacología , Medicamentos Herbarios Chinos/farmacocinética
4.
Expert Opin Drug Deliv ; 21(3): 437-456, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38507231

RESUMEN

INTRODUCTION: The current treatment modalities available for Parkinson's disease (PD) prove inadequate due to the inherent constraints in effectively transporting bioactive compounds across the blood-brain barrier. The utilization of synergistic combinations of multiple drugs in conjunction with advanced nanotechnology, emerges as a promising avenue for the treatment of PD, offering potential breakthroughs in treatment efficacy, targeted therapy, and personalized medicine. AREAS COVERED: This review provides a comprehensive analysis of the efficacy of multifactorial interventions for PD, simultaneously addressing the primary challenges of conventional therapies and highlighting how advanced technologies can help overcome these limitations. Part II focuses on the effectiveness of nanotechnology for improving pharmacokinetics of conventional therapies, through the synergistic use of dual or multiple therapeutic agents into a single nanoformulation. Significant emphasis is laid on the advancements toward innovative integrations, such as CRISPR/Cas9 with neuroprotective agents and stem cells, all effectively synergized with nanocarriers. EXPERT OPINION: By using drug combinations, we can leverage their combined effects to enhance treatment efficacy and mitigate side effects through lower dosages. This article is meant to give nanocarrier-mediated co-delivery of drugs and the strategic incorporation of CRISPR/Cas9, either as an independent intervention or synergized with a neuroprotective agent.


Asunto(s)
Antiparkinsonianos , Portadores de Fármacos , Nanopartículas , Nanotecnología , Fármacos Neuroprotectores , Enfermedad de Parkinson , Humanos , Enfermedad de Parkinson/tratamiento farmacológico , Antiparkinsonianos/administración & dosificación , Antiparkinsonianos/uso terapéutico , Antiparkinsonianos/farmacocinética , Antiparkinsonianos/farmacología , Portadores de Fármacos/química , Animales , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/uso terapéutico , Fármacos Neuroprotectores/farmacocinética , Barrera Hematoencefálica/metabolismo , Sistemas de Liberación de Medicamentos , Medicina de Precisión , Quimioterapia Combinada , Sistemas CRISPR-Cas , Combinación de Medicamentos , Terapia Combinada , Desarrollo de Medicamentos , Diseño de Fármacos
5.
Clin Ther ; 45(12): 1251-1258, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37953075

RESUMEN

PURPOSE: Edaravone is a neuroprotective agent approved as an intravenous treatment for amyotrophic lateral sclerosis (ALS). The intravenous administration of edaravone places a burden on patients and there is a clinical need for oral agents for the treatment of ALS. This report aimed to assess the pharmacokinetics and safety of an edaravone oral suspension in patients with ALS after oral and percutaneous endoscopic gastrostomy (PEG) tube administration. METHODS: Two single-dose, open-label phase 1 clinical studies were conducted. Edaravone oral suspension (105 mg of edaravone in 5 mL aqueous suspension) was administered orally and via PEG tube to 9 and 6 Japanese patients with ALS, respectively. Plasma and urinary pharmacokinetics of unchanged edaravone and its metabolites (sulfate and glucuronide conjugates) were determined. Safety was also evaluated. FINDINGS: After reaching maximum plasma concentration, the mean plasma concentration-time of unchanged edaravone showed a triphasic elimination. Mean plasma concentration-time profiles of the metabolites were higher than those of unchanged edaravone. The mean urinary excretion ratios were higher for the glucuronide conjugate than for either unchanged edaravone or the sulfate conjugate. In patients administered edaravone orally, a single adverse event occurred (blood urine present), which was mild and improved without medical intervention. No adverse drug reactions or serious adverse events were reported. In patients administered edaravone via PEG tube, 5 treatment-emergent adverse events were reported in 3 patients; none were related to the study drug. No adverse drug reactions were reported. IMPLICATIONS: In patients with ALS, a single dose of edaravone oral suspension was well absorbed and mainly eliminated in urine as the glucuronide conjugate. No safety concerns emerged. Pharmacokinetics were similar to those previously reported in healthy participants following oral administration. This indicates that effective drug concentrations were achieved and edaravone can be successfully administered both orally and via a PEG tube in patients with ALS. CLINICAL TRIAL REGISTRATION: ClinicalTrials.gov, NCT04176224 (oral administration) and NCT04254913 (PEG tube administration), www. CLINICALTRIALS: gov.


Asunto(s)
Esclerosis Amiotrófica Lateral , Fármacos Neuroprotectores , Humanos , Esclerosis Amiotrófica Lateral/tratamiento farmacológico , Edaravona/farmacocinética , Glucurónidos/uso terapéutico , Fármacos Neuroprotectores/farmacocinética , Sulfatos/uso terapéutico
6.
J Neurotrauma ; 40(17-18): 1889-1906, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37130044

RESUMEN

To date, no drug therapy has shown significant efficacy in improving functional outcomes in patients with acute spinal cord injury (SCI). Riluzole is an approved benzothiazole sodium channel blocker to attenuate neurodegeneration in amyotrophic lateral sclerosis (ALS) and is of interest for neuroprotection in SCI. In a Phase I clinical trial (ClinicalTrials.gov Identifier: NCT00876889), riluzole was well tolerated with a 2-week treatment at the dose level approved for ALS and exhibited potential efficacy in patients with SCI. The acute and progressive nature of traumatic SCI and the complexity of secondary injury processes alter the pharmacokinetics (PK) of therapeutics. In the PK sub-study of the multi-center, randomized, placebo-controlled, double-blinded Riluzole in Spinal Cord Injury Study (RISCIS) Phase II/III trial (ClinicalTrials.gov Identifier: NCT01597518), a total of 32 SCI patients were enrolled, and most of our patients were middle-age Caucasian males with head and neck injuries. We studied the PK and pharmacodynamics (PD) of riluzole on motor recovery, measured by International Standards for Neurological Classification of SCI (ISNCSCI) Motor Score at injury and at 3-month and 6-month follow-ups, along with levels of the axonal injury biomarker phosphorylated neurofilament heavy chain (pNF-H), during the 2-week treatment. PK modeling, PK/PD correlations were developed to identify the potential effective exposure of riluzole for intended PD outcomes. The longitudinal impacts of SCI on the PK of riluzole are characterized. A time-varying population PK model of riluzole is established, incorporating time-varying clearance and volume of distribution from combined data of Phase I and Phase II/III trials. With the developed model, a rational, optimal dosing scheme can be designed with time-dependent modification to preserve the required therapeutic exposure of riluzole. The PD of riluzole and the relationship between PK and neurological outcomes of the treatment were established. The time course of efficacy in total motor score improvement (ΔTMS) and pNF-H were monitored. A three-dimensional (3D) PK/PD correlation was established for ΔTMS at 6 months with overall riluzole exposure area under the curve for Day 0-Day14 (AUCD0-D14) and baseline TMS for individual patients. Patients with baseline TMS between 1 and 36 benefited from the optimal exposure range of 16-48 mg*h/mL. The PD models of pNF-H revealed the riluzole efficacy, as treated subjects exhibited a diminished increase in progression of pNF-H, indicative of reduced axonal breakdown. The independent parameter of area between effective curves (ABEC) between the time profiles of pNF-H in placebo and treatment groups was statistically identified as a significant predictor for the treatment effect on the biomarker. A mechanistic clinical outcomes (CO)/PD (pNF-H) model was established, and the proposed structure demonstrated the feasibility of PK/PD/CO correlation model. No appreciable hepatic toxicity was observed with the current riluzole treatment regimen. The development of effective treatment for SCI is challenging. However, the future model-informed and PK-guided drug development and regimen modification can be rationally executed with the optimal dosing regimen design based on the developed 3D PK/PD model. The PK/PD/CO model can serve as a rational guide for future drug development, PKPD model refinement, and extension to other studies in SCI settings.


Asunto(s)
Esclerosis Amiotrófica Lateral , Médula Cervical , Traumatismos del Cuello , Fármacos Neuroprotectores , Traumatismos de la Médula Espinal , Masculino , Persona de Mediana Edad , Humanos , Riluzol/efectos adversos , Fármacos Neuroprotectores/efectos adversos , Fármacos Neuroprotectores/farmacocinética , Esclerosis Amiotrófica Lateral/tratamiento farmacológico , Traumatismos de la Médula Espinal/tratamiento farmacológico , Traumatismos del Cuello/tratamiento farmacológico
7.
Inflammopharmacology ; 31(3): 1449-1464, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36856916

RESUMEN

Alzheimer's disease (AD) is one of the most prevalent neurodegenerative illnesses, and yet, no workable treatments have been discovered to prevent or reverse AD. Curcumin (CUR), the major polyphenolic compound of turmeric (Curcuma longa) rhizomes, and Ginkgo biloba extract (GBE) are natural substances derived from conventional Chinese herbs that have long been shown to provide therapeutic advantages for AD. The uptake of curcumin into the brain is severely restricted by its low ability to cross the blood-brain barrier (BBB). Meanwhile, GBE has been shown to improve BBB permeability. The present study evaluated the neuroprotective effects and pharmacokinetic profile of curcumin and GBE combination to find out whether GBE can enhance curcumin's beneficial effects in AD by raising its brain concentration. Results revealed that CUR + GBE achieved significantly higher levels of curcumin in the brain and plasma after 30 min and 1 h of oral administration, compared to curcumin alone, and this was confirmed by reversed phase high-performance liquid chromatography (RP-HPLC). The effect of combined oral treatment, for 28 successive days, on cognitive function and other AD-like alterations was studied in scopolamine-heavy metal mixtures (SCO + HMM) AD model in rats. The combination reversed at least, partially on the learning and memory impairment induced by SCO + HMM. This was associated with a more pronounced inhibitory effect on acetylcholinesterase (AChE), caspase-3, hippocampal amyloid beta (Aß1-42), and phosphorylated tau protein (p-tau) count, and pro-inflammatory cytokines tumor necrosis factor-alpha (TNF-α) and interleukine-1beta (IL-1ß), as compared to the curcumin alone-treated group. Additionally, the combined treatment significantly decreased lipid peroxidation (MDA) and increased levels of reduced glutathione (GSH), when compared with the curcumin alone. These findings support the concept that the combination strategy might be an alternative therapy in the management/prevention of neurological disorders. This study sheds light on a new approach for exploring new phyto-therapies for AD and emphasizes that more research should focus on the synergic effects of herbal drugs in future.


Asunto(s)
Enfermedad de Alzheimer , Curcumina , Fármacos Neuroprotectores , Animales , Ratas , Acetilcolinesterasa/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/metabolismo , Curcumina/farmacocinética , Curcumina/farmacología , Curcumina/uso terapéutico , Fármacos Neuroprotectores/farmacocinética , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Extracto de Ginkgo/farmacocinética , Extracto de Ginkgo/farmacología , Extracto de Ginkgo/uso terapéutico , Humanos , Barrera Hematoencefálica
8.
ACS Chem Neurosci ; 14(4): 554-572, 2023 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-36735764

RESUMEN

Neurodegenerative diseases of the central nervous system (CNS) pose a serious health concern worldwide, with a particular incidence in developed countries as a result of life expectancy increase and the absence of restorative treatments. Presently, treatments for these neurological conditions are focused on managing the symptoms and/or slowing down their progression. As so, the research on novel neuroprotective drugs is of high interest. Glypromate (glycyl-l-prolyl-l-glutamic acid, also known as GPE), an endogenous small peptide widespread in the brain, holds great promise to tackle neurodegenerative diseases such as Parkinson's, Alzheimer's, and Huntington's, s well as other CNS-related disorders like Rett and Down's syndromes. However, the limited pharmacokinetic properties of Glypromate hinder its clinical application. As such, intense research has been devoted to leveraging the pharmacokinetic profile of this neuropeptide. This review aims to offer an updated perspective on Glypromate research by exploring the vast array of chemical derivatizations of more than 100 analogs described in the literature over the past two decades. The collection and discussion of the most relevant structure-activity relationships will hopefully guide the discovery of new Glypromate-based neuroprotective drugs.


Asunto(s)
Enfermedades del Sistema Nervioso Central , Enfermedades Neurodegenerativas , Neuropéptidos , Fármacos Neuroprotectores , Neurociencias , Humanos , Fármacos Neuroprotectores/uso terapéutico , Fármacos Neuroprotectores/farmacocinética , Enfermedades Neurodegenerativas/tratamiento farmacológico
9.
Nanomedicine ; 43: 102547, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35292367

RESUMEN

Baicalin (BA) has a good intervention effect on encephalopathy. In this study, macrophage membrane was modified on the surface of baicalin liposomes (BA-LP) by extrusion method. Macrophage membrane modified BA-LP (MM-BA-LP) was characterized by various analytical techniques, and evaluated for brain targeting. The results presented MM-BA-LP had better brain targeting compared with BA-LP. Pharmacokinetic experiments showed that MM-BA-LP improved pharmacokinetic parameters and increased the residence time of BA. Pharmacodynamic of middle cerebral artery occlusion (MCAO) rat model was studied to verify the therapeutic effect of MM-BA-LP on cerebral ischemia reperfusion injury (CIRI). The results showed that MM-BA-LP could significantly improve the neurological deficit, cerebral infarction volume and brain pathological state of MCAO rats compared with BA-LP. These results suggested that MM-BA-LP could significantly enhance the brain targeting and improve the circulation of BA in blood, and had a significantly better neuroprotective effect on MCAO rats than BA-LP.


Asunto(s)
Isquemia Encefálica , Fármacos Neuroprotectores , Daño por Reperfusión , Animales , Encéfalo , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/patología , Flavonoides , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/patología , Liposomas/farmacocinética , Macrófagos , Fármacos Neuroprotectores/farmacocinética , Fármacos Neuroprotectores/uso terapéutico , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/tratamiento farmacológico
10.
Molecules ; 27(3)2022 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-35164373

RESUMEN

Acanthopanax senticosus (AS) is a medicinal and food homologous plant with many biological activities. In this research, we generated a brain injury model by 60Co -γ ray radiation at 4 Gy, and gavaged adult mice with the extract with AS, Acanthopanax senticocus polysaccharides (ASPS), flavones, syringin and eleutheroside E (EE) to explore the therapeutic effect and metabolic characteristics of AS on the brain injury. Behavioral tests and pathological experiments showed that the AS prevented the irradiated mice from learning and memory ability impairment and protected the neurons of irradiated mice. Meanwhile, the functional components of AS increased the antioxidant activity of irradiated mice. Furthermore, we found the changes of neurotransmitters, especially in the EE and syringin groups. Finally, distribution and pharmacokinetic analysis of AS showed that the functional components, especially EE, could exert their therapeutic effects in brain of irradiated mice. This lays a theoretical foundation for the further research on the treatment of radiation-induced brain injury by AS.


Asunto(s)
Antioxidantes/farmacología , Lesiones Encefálicas/tratamiento farmacológico , Eleutherococcus/química , Fármacos Neuroprotectores/farmacología , Neurotransmisores/metabolismo , Extractos Vegetales/farmacología , Traumatismos por Radiación/tratamiento farmacológico , Animales , Antioxidantes/farmacocinética , Encéfalo/efectos de los fármacos , Lesiones Encefálicas/etiología , Lesiones Encefálicas/patología , Radioisótopos de Cobalto/toxicidad , Masculino , Ratones , Fármacos Neuroprotectores/farmacocinética , Extractos Vegetales/farmacocinética , Traumatismos por Radiación/etiología , Traumatismos por Radiación/patología , Distribución Tisular
11.
Int J Mol Sci ; 23(3)2022 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-35163820

RESUMEN

Globally, stroke is a leading cause of death and long-term disability. Over the past decades, several efforts have attempted to discover new drugs or repurpose existing therapeutics to promote post-stroke neurological recovery. Preclinical stroke studies have reported successes in identifying novel neuroprotective agents; however, none of these compounds have advanced beyond a phase III clinical trial. One reason for these failures is the lack of consideration of blood-brain barrier (BBB) transport mechanisms that can enable these drugs to achieve efficacious concentrations in ischemic brain tissue. Despite the knowledge that drugs with neuroprotective properties (i.e., statins, memantine, metformin) are substrates for endogenous BBB transporters, preclinical stroke research has not extensively studied the role of transporters in central nervous system (CNS) drug delivery. Here, we review current knowledge on specific BBB uptake transporters (i.e., organic anion transporting polypeptides (OATPs in humans; Oatps in rodents); organic cation transporters (OCTs in humans; Octs in rodents) that can be targeted for improved neuroprotective drug delivery. Additionally, we provide state-of-the-art perspectives on how transporter pharmacology can be integrated into preclinical stroke research. Specifically, we discuss the utility of in vivo stroke models to transporter studies and considerations (i.e., species selection, co-morbid conditions) that will optimize the translational success of stroke pharmacotherapeutic experiments.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Accidente Cerebrovascular Isquémico/metabolismo , Transportadores de Anión Orgánico/metabolismo , Proteínas de Transporte de Catión Orgánico/metabolismo , Animales , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Accidente Cerebrovascular Isquémico/tratamiento farmacológico , Terapia Molecular Dirigida , Fármacos Neuroprotectores/farmacocinética , Fármacos Neuroprotectores/uso terapéutico
12.
Nutrients ; 14(2)2022 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-35057429

RESUMEN

For thousands of years, mankind has been using plant extracts or plants themselves as medicinal herbs. Currently, there is a great deal of public interest in naturally occurring medicinal substances that are virtually non-toxic, readily available, and have an impact on well-being and health. It has been noted that dietary curcumin is one of the regulators that may positively influence changes in the brain after ischemia. Curcumin is a natural polyphenolic compound with pleiotropic biological properties. The observed death of pyramidal neurons in the CA1 region of the hippocampus and its atrophy are considered to be typical changes for post-ischemic brain neurodegeneration and for Alzheimer's disease. Additionally, it has been shown that one of the potential mechanisms of severe neuronal death is the accumulation of neurotoxic amyloid and dysfunctional tau protein after cerebral ischemia. Post-ischemic studies of human and animal brains have shown the presence of amyloid plaques and neurofibrillary tangles. The significant therapeutic feature of curcumin is that it can affect the aging-related cellular proteins, i.e., amyloid and tau protein, preventing their aggregation and insolubility after ischemia. Curcumin also decreases the neurotoxicity of amyloid and tau protein by affecting their structure. Studies in animal models of cerebral ischemia have shown that curcumin reduces infarct volume, brain edema, blood-brain barrier permeability, apoptosis, neuroinflammation, glutamate neurotoxicity, inhibits autophagy and oxidative stress, and improves neurological and behavioral deficits. The available data suggest that curcumin may be a new therapeutic substance in both regenerative medicine and the treatment of neurodegenerative disorders such as post-ischemic neurodegeneration.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Isquemia Encefálica/complicaciones , Curcumina/farmacología , Fármacos Neuroprotectores/farmacología , Enfermedad de Alzheimer/etiología , Amiloide/efectos de los fármacos , Amiloide/metabolismo , Animales , Apoptosis/efectos de los fármacos , Atrofia/etiología , Disponibilidad Biológica , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Edema Encefálico/tratamiento farmacológico , Isquemia Encefálica/tratamiento farmacológico , Curcumina/química , Curcumina/farmacocinética , Modelos Animales de Enfermedad , Microbioma Gastrointestinal/fisiología , Gerbillinae , Hipocampo/patología , Humanos , Ratones , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Fármacos Neuroprotectores/química , Fármacos Neuroprotectores/farmacocinética , Estrés Oxidativo/efectos de los fármacos , Ratas , Proteínas tau/efectos de los fármacos , Proteínas tau/metabolismo
13.
Naunyn Schmiedebergs Arch Pharmacol ; 395(2): 167-185, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34988596

RESUMEN

Ischemic stroke presents multifaceted pathological outcomes with overlapping mechanisms of cerebral injury. High mortality and disability with stroke warrant a novel multi-targeted therapeutic approach. The neuroprotection with progesterone (PG) and noscapine (NOS) on cerebral ischemia-reperfusion (I-R) injury was demonstrated individually, but the outcome of combination treatment to alleviate cerebral damage is still unexplored. Randomly divided groups of rats (n = 6) were Sham-operated, I-R, PG (8 mg/kg), NOS (10 mg/kg), and PG + NOS (8 mg/kg + 10 mg/kg). The rats were exposed to bilateral common carotid artery occlusion, except Sham-operated, to investigate the therapeutic outcome of PG and NOS alone and in combination on I-R injury. Besides the alterations in cognitive and motor abilities, we estimated infarct area, oxidative stress, blood-brain barrier (BBB) permeability, and histology after treatment. Pharmacokinetic parameters like Cmax, Tmax, half-life, and AUC0-t were estimated in biological samples to substantiate the therapeutic outcomes of the combination treatment. We report PG and NOS prevent loss of motor ability and improve spatial memory after cerebral I-R injury. Combination treatment significantly reduced inflammation and restricted infarction; it attenuated oxidative stress and BBB damage and improved grip strength. Histopathological analysis demonstrated a significant reduction in leukocyte infiltration with the most profound effect in the combination group. Simultaneous analysis of PG and NOS in plasma revealed enhanced peak drug concentration, improved AUC, and prolonged half-life; the drug levels in the brain have increased significantly for both. We conclude that PG and NOS have beneficial effects against brain damage and the co-administration further reinforced neuroprotection in the cerebral ischemia-reperfusion injury.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Fármacos Neuroprotectores/administración & dosificación , Noscapina/administración & dosificación , Progesterona/administración & dosificación , Animales , Área Bajo la Curva , Barrera Hematoencefálica/metabolismo , Modelos Animales de Enfermedad , Quimioterapia Combinada , Semivida , Accidente Cerebrovascular Isquémico/tratamiento farmacológico , Masculino , Fármacos Neuroprotectores/farmacocinética , Fármacos Neuroprotectores/farmacología , Noscapina/farmacocinética , Noscapina/farmacología , Estrés Oxidativo/efectos de los fármacos , Progesterona/farmacocinética , Progesterona/farmacología , Ratas , Ratas Wistar , Daño por Reperfusión/tratamiento farmacológico
14.
Eur J Med Chem ; 229: 114080, 2022 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-34992038

RESUMEN

Mutations in the Leucine Rich Repeat Protein Kinase 2 gene (LRRK2) are the most common genetic causes of Parkinson's Disease (PD). The G2019S mutation is the most common inherited LRRK2 mutation, occurs in the kinase domain, and results in increased kinase activity. We report the discovery and development of compound 38, an indazole-based, G2019S-selective (>2000-fold vs. WT) LRRK2 inhibitor capable of entering rodent brain (Kp = 0.5) and selectively inhibiting G2019S-LRRK2. The compounds disclosed herein present a starting point for further development of brain penetrant G2019S selective inhibitors that hopefully reduce lung phenotype side-effects and pave the way to providing a precision medicine for people with PD who carry the G2019S mutation.


Asunto(s)
Indazoles/síntesis química , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/antagonistas & inhibidores , Fármacos Neuroprotectores/síntesis química , Enfermedad de Parkinson/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/síntesis química , Animales , Encéfalo , Modelos Animales de Enfermedad , Descubrimiento de Drogas , Humanos , Indazoles/farmacocinética , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Pulmón , Masculino , Ratones , Simulación del Acoplamiento Molecular , Mutación , Fármacos Neuroprotectores/farmacocinética , Fenotipo , Unión Proteica , Conformación Proteica , Inhibidores de Proteínas Quinasas/farmacocinética , Roedores , Relación Estructura-Actividad
15.
Exp Neurol ; 348: 113923, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34780773

RESUMEN

Arginase 1 (A1) is the enzyme that hydrolyzes the amino acid, L-arginine, to ornithine and urea. We have previously shown that A1 deletion worsens retinal ischemic injury, suggesting a protective role of A1. In this translational study, we aimed to study the utility of systemic pegylated A1 (PEG-A1, recombinant human arginase linked to polyethylene glycol) treatment in mouse models of acute retinal and brain injury. Cohorts of WT mice were subjected to retinal ischemia-reperfusion (IR) injury, traumatic optic neuropathy (TON) or brain cerebral ischemia via middle cerebral artery occlusion (MCAO) and treated with intraperitoneal injections of PEG-A1 or vehicle (PEG only). Drug penetration into retina and brain tissues was measured by western blotting and immunolabeling for PEG. Neuroprotection was measured in a blinded fashion by quantitation of NeuN (neuronal marker) immunolabeling of retina flat-mounts and brain infarct area using triphenyl tetrazolium chloride (TTC) staining. Furthermore, ex vivo retina explants and in vitro retina neuron cultures were subjected to oxygen-glucose deprivation (OGD) followed by reoxygenation (R) and treated with PEG-A1. PEG-A1 given systemically did not cross the intact blood-retina/brain barriers in sham controls but reached the retina and brain after injury. PEG-A1 provided neuroprotection after retinal IR injury, TON and cerebral ischemia. PEG-A1 treatment was also neuroprotective in retina explants subjected to OGD/R but did not improve survival in retinal neuronal cultures exposed to OGD/R. In summary, systemic PEG-A1 administration is neuroprotective and provides an excellent route to deliver the drug to the retina and the brain after acute injury.


Asunto(s)
Arginasa/uso terapéutico , Lesiones Encefálicas/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Retina/lesiones , Animales , Arginasa/farmacocinética , Barrera Hematoencefálica , Barrera Hematorretinal , Encéfalo/metabolismo , Isquemia Encefálica/tratamiento farmacológico , Supervivencia Celular/efectos de los fármacos , Humanos , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fármacos Neuroprotectores/farmacocinética , Traumatismos del Nervio Óptico/tratamiento farmacológico , Polietilenglicoles , Proteínas Recombinantes/uso terapéutico , Daño por Reperfusión/prevención & control , Retina/metabolismo
16.
Int J Mol Sci ; 22(22)2021 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-34830148

RESUMEN

Phytosterols constitute a class of natural products that are an important component of diet and have vast applications in foods, cosmetics, and herbal medicines. With many and diverse isolated structures in nature, they exhibit a broad range of biological and pharmacological activities. Among over 200 types of phytosterols, stigmasterol and ß-sitosterol were ubiquitous in many plant species, exhibiting important aspects of activities related to neurodegenerative diseases. Hence, this mini-review presented an overview of the reported studies on selected phytosterols related to neurodegenerative diseases. It covered the major phytosterols based on biosynthetic considerations, including other phytosterols with significant in vitro and in vivo biological activities.


Asunto(s)
Encéfalo/metabolismo , Enfermedades Neurodegenerativas/prevención & control , Fitosteroles/uso terapéutico , Fitoterapia/métodos , Plantas Medicinales/química , Encéfalo/patología , Humanos , Estructura Molecular , Enfermedades Neurodegenerativas/metabolismo , Fármacos Neuroprotectores/química , Fármacos Neuroprotectores/farmacocinética , Fármacos Neuroprotectores/uso terapéutico , Fitosteroles/química , Fitosteroles/farmacocinética , Sitoesteroles/química , Sitoesteroles/farmacocinética , Sitoesteroles/uso terapéutico , Estigmasterol/química , Estigmasterol/farmacocinética , Estigmasterol/uso terapéutico
17.
Cell Mol Biol (Noisy-le-grand) ; 67(1): 101-105, 2021 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-34817361

RESUMEN

Parkinson's disease (PD) is one of the most prevalent neurodegenerative disorders worldwide. It is caused by the degeneration of dopaminergic neurons from the substantia nigra pars compacta. This neuronal loss causes the dopamine deficiency that leads to a series of functional changes within the basal ganglia, producing motor control abnormalities. L-DOPA is considered the gold standard for PD treatment, and it may alleviate its clinical manifestations for some time. However, its prolonged administration produces tolerance and several severe side effects, including dyskinesias and gastrointestinal disorders. Thus, there is an urgent need to find effective medications, and current trends have proposed some natural products as emerging options for this purpose. Concerning this, curcumin represents a promising bioactive compound with high therapeutic potential. Diverse studies in cellular and animal models have suggested that curcumin could be employed for the treatment of PD. Therefore, the objective of this narrative mini-review is to present an overview of the possible therapeutic effects of curcumin and the subjacent molecular mechanisms. Moreover, we describe several possible nanocarrier-based approaches to improve the bioavailability of curcumin and enhance its biological activity.


Asunto(s)
Encéfalo/efectos de los fármacos , Curcumina/administración & dosificación , Nanopartículas/administración & dosificación , Enfermedad de Parkinson/tratamiento farmacológico , Animales , Disponibilidad Biológica , Encéfalo/metabolismo , Curcumina/química , Curcumina/farmacocinética , Liberación de Fármacos , Glutatión Peroxidasa/metabolismo , Humanos , NADPH Oxidasas/antagonistas & inhibidores , NADPH Oxidasas/metabolismo , Nanopartículas/química , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/química , Fármacos Neuroprotectores/farmacocinética , Enfermedad de Parkinson/metabolismo , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/metabolismo , Resultado del Tratamiento , Regulación hacia Arriba/efectos de los fármacos
18.
Drug Deliv ; 28(1): 2469-2479, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34766542

RESUMEN

As a multi-target drug to treat ischemic stroke, N-butylphthalide (NBP) is extremely water-insoluble and exhibits limited oral bioavailability, impeding its wide oral application. Effective treatment of ischemic stroke by NBP requires timely and efficient drug exposure, necessitating the development of new oral formulations. Herein, liposomes containing biosurfactant sodium cholate (CA-liposomes) were systemically investigated as an oral NBP delivery platform because of its high biocompatibility and great potential for clinical applications. The optimized liposomes have a uniform hydrodynamic size of 104.30 ± 1.60 nm and excellent encapsulation efficiency (93.91 ± 1.10%). Intriguingly, NBP-loaded CA-liposomes produced rapid drug release and the cumulative release was up to 88.09 ± 4.04% during 12 h while that for NBP group was only 6.79 ± 0.99%. Caco-2 cell monolayer assay demonstrated the superior cell uptake and transport efficiency of NBP-loaded CA-liposomes than free NBP, which was mediated by passive diffusion via transcellular and paracellular routes. After oral administration to rats, NBP-loaded CA-liposomes exhibited rapid and almost complete drug absorption, with a tmax of 0.70 ± 0.14 h and an absolute bioavailability of 92.65% while NBP suspension demonstrated relatively low bioavailability (21.7%). Meanwhile, NBP-loaded CA-liposomes produced 18.30-fold drug concentration in the brain at 5 min compared with NBP suspension, and the brain bioavailability increased by 2.48-fold. As expected, NBP-loaded CA-liposomes demonstrated significant therapeutic efficacy in a middle cerebral artery occlusion rat model. Our study provides new insights for engineering oral formulations of NBP with fast and sufficient drug exposure against ischemic stroke in the clinic.


Asunto(s)
Benzofuranos/administración & dosificación , Benzofuranos/farmacología , Liposomas/química , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/farmacología , Animales , Área Bajo la Curva , Benzofuranos/farmacocinética , Células CACO-2 , Química Farmacéutica , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Portadores de Fármacos/química , Liberación de Fármacos , Humanos , Accidente Cerebrovascular Isquémico/patología , Masculino , Fármacos Neuroprotectores/farmacocinética , Tamaño de la Partícula , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Colato de Sodio/química , Distribución Tisular
19.
Food Funct ; 12(19): 9211-9228, 2021 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-34606547

RESUMEN

The present study aims to investigate the protective effects of N-(3-methoxybenzyl)-(9Z,12Z,15Z)-octadecatrienamide (M 18:3) on corticosterone-induced neurotoxicity. A neurotoxic model was established by subcutaneous injection of corticosterone (40 mg per kg bw) for 21 days. Depressive behaviors (the percentage of sucrose consumption, the immobility time in the forced swimming test, and the total distance in the open field test) were observed. The levels of the brain-derived neurotrophic factor, the contents of tumor necrosis factor-α and interleukin-6, and the numbers of positive cells of doublecortin and bromodeoxyuridine in the hippocampus were measured. The density of hippocampal neurons was calculated. The morphological changes of hippocampal neurons (the density of dendritic spines, the dendritic length, and the area and volume of dendritic cell bodies) were observed. The expression levels of synaptophysin, synapsin I, and postsynaptic density protein 95 were measured. Behavioral experiments showed that M 18:3 (5 and 25 mg per kg bw) could remarkably improve the depressive behaviors. The enzyme-linked immunosorbent assay showed that M 18:3 could considerably reduce hippocampal neuroinflammation and increase hippocampal neurotrophy. Nissl staining showed that M 18:3 could remarkably improve the corticosterone-induced decrease in the hippocampal neuron density. Immunofluorescence analysis showed that M 18:3 could considerably promote hippocampal neurogenesis. Golgi staining showed that M 18:3 could remarkably improve the corticosterone-induced changes in the hippocampal dendritic structure. Western blotting showed that M 18:3 could considerably increase the expression levels of synaptic-structure-related proteins in the hippocampus. In conclusion, the protective effects of M 18:3 may be attributed to the anti-inflammatory, neurotrophic, and synaptic protection properties.


Asunto(s)
Alquenos/farmacología , Compuestos de Bencilo/farmacología , Hipocampo/efectos de los fármacos , Lepidium , Fármacos Neuroprotectores/farmacología , Alquenos/farmacocinética , Animales , Antiinflamatorios no Esteroideos/farmacología , Antidepresivos/farmacología , Conducta Animal/efectos de los fármacos , Compuestos de Bencilo/farmacocinética , Barrera Hematoencefálica/metabolismo , Recuento de Células , Forma de la Célula , Corticosterona , Depresión/tratamiento farmacológico , Hipocampo/citología , Hipocampo/metabolismo , Hipocampo/fisiología , Masculino , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neurogénesis , Neuronas/citología , Fármacos Neuroprotectores/farmacocinética , Extractos Vegetales/química , Extractos Vegetales/farmacocinética , Extractos Vegetales/farmacología , Ratas , Ratas Wistar , Sinapsis/efectos de los fármacos , Sinapsis/fisiología
20.
Neurosci Lett ; 765: 136249, 2021 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-34536510

RESUMEN

Curcumin as an antioxidant natural herb has shown numerous pharmacological effects. However, the poor bioavailability of curcumin is a significant pharmacological barrier for its antioxidant activities. The present study was conducted to develop curcumin-loaded nanophytosome (CNP) and explore their therapeutic potential in a ketamine (KET)-induced schizophrenia (SCZ) model. The mice in our experiment were treated orally with curcumin and CNP (20 mg/kg) for 30 consecutive days. In addition, the animals received intraperitoneal injection of KET (30 mg/kg/day) from the 16th to the 30th day. SCZ-like behaviors were evaluated employing forced swimming test (FST), open field test (OFT), and novel object recognition test (NORT), and oxidative stress markers in the brain were estimated. Our results revealed that CNP has a greater neuroprotective effect compared to free curcumin. CNP pretreatment significantly ameliorated KET-induced brain injury evidenced by a marked reduction in the depressive and anxiety-like behaviors, memory deficits, and oxidative stress markers in cortical and subcortical tissues. Therefore, CNP, as a suitable drug delivery system, may improve curcumin bioavailability and confer stronger neuroprotective effects against KET-induced behavioral deficits and oxidative damages.


Asunto(s)
Curcumina/administración & dosificación , Sistema de Administración de Fármacos con Nanopartículas , Fármacos Neuroprotectores/administración & dosificación , Esquizofrenia/prevención & control , Administración Oral , Animales , Disponibilidad Biológica , Curcumina/farmacocinética , Modelos Animales de Enfermedad , Humanos , Ketamina/administración & dosificación , Ketamina/toxicidad , Masculino , Ratones , Fármacos Neuroprotectores/farmacocinética , Estrés Oxidativo/efectos de los fármacos , Esquizofrenia/inducido químicamente
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA