Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cancer Lett ; 593: 216955, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38750720

RESUMEN

Brain cancers, particularly glioblastoma multiforme (GBM), are challenging health issues with frequent unmet aspects. Today, discovering safe and effective therapeutic modalities for brain tumors is among the top research interests. Immunotherapy is an emerging area of investigation in cancer treatment. Since immune checkpoints play fundamental roles in repressing anti-cancer immunity, diverse immune checkpoint inhibitors (ICIs) have been developed, and some monoclonal antibodies have been approved clinically for particular cancers; nevertheless, there are significant concerns regarding their efficacy and safety in brain tumors. Among the various tools to modify the immune checkpoints, phytochemicals show good effectiveness and excellent safety, making them suitable candidates for developing better ICIs. Phytochemicals regulate multiple immunological checkpoint-related signaling pathways in cancer biology; however, their efficacy for clinical cancer immunotherapy remains to be established. Here, we discussed the involvement of immune checkpoints in cancer pathology and summarized recent advancements in applying phytochemicals in modulating immune checkpoints in brain tumors to highlight the state-of-the-art and give constructive prospects for future research.


Asunto(s)
Neoplasias Encefálicas , Inhibidores de Puntos de Control Inmunológico , Fitoquímicos , Humanos , Fitoquímicos/uso terapéutico , Fitoquímicos/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/inmunología , Inmunoterapia/métodos , Glioblastoma/tratamiento farmacológico , Glioblastoma/inmunología , Transducción de Señal/efectos de los fármacos , Animales
2.
Prog Mol Biol Transl Sci ; 204: 133-161, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38458736

RESUMEN

Despite the extensive advancements made in the field of cancer therapy, the outlook of individuals suffering from glioblastoma multiforme remains highly detrimental. The absence of specific treatments for cancerous cells significantly hinders the effectiveness of conventional anticancer techniques. Multiple research studies have demonstrated that the suppression of specific genes or the augmentation of therapeutic proteins through RNA-based therapeutics may represent a valuable approach when combined with chemotherapy or immunotherapy. In recent years, there has been a significant increase in the application of RNA therapeutics in conjunction with chemotherapy and immunotherapy. This emerging field has become a prominent area of research for advancing various types of cancer treatments. The present investigation provides an in-depth overview of the classification and application of RNA therapy, focusing on the mechanisms of RNA antitumor treatment and the current status of clinical studies on RNA drugs.


Asunto(s)
Glioblastoma , Glioma , Humanos , ARN , Glioma/genética , Glioma/terapia , Glioma/patología , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Glioblastoma/patología , Inmunoterapia/métodos
3.
Prog Mol Biol Transl Sci ; 204: 177-218, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38458738

RESUMEN

Liposomal drug delivery systems stand as versatile therapeutic platforms for precisely targeting related elements in cancerous tissues owing to their intrinsic passive and acquired active targeting capabilities and exceptional compatibility with physiologic environments. When the capacity of liposomes as nanocarriers is combined with the revolutionary potential of RNA therapies in affecting undruggable targets, the outcome would be promising drug candidates as game-changers in the cancer treatment arena. However, optimizing liposome composition, physicochemical properties, and surface chemistry is paramount to maximizing their pharmacokinetic and pharmacodynamic attributes. This review highlighted the potential of liposomes as nanovehicles for RNA therapeutics through a literature review and looked at the most recent preclinical and clinical advancements in utilizing liposomal RNA therapeutics for cancer management. Notably, the discovery of novel targets, advancements in liposome engineering, and organizing well-planned clinical trials would help uncover the incredible potential of these nanotherapeutics in cancer patients.


Asunto(s)
Liposomas , Neoplasias , Humanos , Liposomas/uso terapéutico , Sistemas de Liberación de Medicamentos , Neoplasias/tratamiento farmacológico
4.
J Control Release ; 368: 703-727, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38490373

RESUMEN

Drug delivery platforms have gracefully emerged as an indispensable component of novel cancer chemotherapy, bestowing targeted drug distribution, elevating therapeutic effects, and reducing the burden of unwanted side effects. In this context, hybrid delivery systems artfully harnessing the virtues of liposomes and hydrogels bring remarkable benefits, especially for localized cancer therapy, including intensified stability, excellent amenability to hydrophobic and hydrophilic medications, controlled liberation behavior, and appropriate mucoadhesion to mucopenetration shift. Moreover, three-dimensional biocompatible liposome-integrated hydrogel networks have attracted unprecedented interest in tissue regeneration, given their tunable architecture and physicochemical properties, as well as enhanced mechanical support. This review elucidates and presents cutting-edge developments in recruiting liposome-integrated hydrogel systems for cancer treatment and tissue regeneration.


Asunto(s)
Liposomas , Neoplasias , Liposomas/química , Hidrogeles/química , Sistemas de Liberación de Medicamentos/métodos , Neoplasias/tratamiento farmacológico
5.
J Funct Biomater ; 14(8)2023 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-37623653

RESUMEN

Standard cancer chemotherapeutics often produce significant adverse effects and eventually lose their effectiveness due to the emergence of resistance mechanisms. As a result, patients with malignant tumors experience a poor quality of life and a short lifespan. Thus, combination medication regimens provide various advantages, including increased success rate, fewer side effects, and fewer occurrences of resistance. Curcumin (Cur), a potential phytochemical from turmeric, when coupled with traditional chemotherapeutics, has been established to improve the effectiveness of cancer treatment in clinical and preclinical investigations. Cur not only exerts multiple mechanisms resulting in apoptotic cancer cell death but also reduces the resistance to standard chemotherapy drugs, mainly through downregulating the multi-drug resistance (MDR) cargoes. Recent reports showed the beneficial outcomes of Cur combination with many chemotherapeutics in various malignancies. Nevertheless, owing to the limited bioavailability, devising co-delivery strategies for Cur and conventional pharmaceuticals appears to be required for clinical settings. This review summarized various Cur combinations with standard treatments as cancer therapeutics.

6.
Biofactors ; 49(4): 782-819, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37162294

RESUMEN

Regarding the multimechanistic nature of cancers, current chemo- or radiotherapies often fail to eradicate disease pathology, and frequent relapses or resistance to therapies occur. Brain malignancies, particularly glioblastomas, are difficult-to-treat cancers due to their highly malignant and multidimensional biology. Unfortunately, patients suffering from malignant tumors often experience poor prognoses and short survival periods. Thus far, significant efforts have been conducted to discover novel and more effective modalities. To that end, modulation of the ubiquitin-proteasome system (UPS) has attracted tremendous interest since it affects the homeostasis of proteins critically engaged in various cell functions, for example, cell metabolism, survival, proliferation, and differentiation. With their safe and multimodal actions, phytochemicals are among the promising therapeutic tools capable of turning the operation of various UPS elements. The present review, along with an updated outline of the role of UPS dysregulation in multiple cancers, provided a detailed discussion on the impact of phytochemicals on the UPS function in malignancies, especially brain tumors.


Asunto(s)
Neoplasias Encefálicas , Ubiquitina , Humanos , Ubiquitina/genética , Ubiquitina/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/farmacología , Inhibidores de Proteasoma/uso terapéutico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Neoplasias Encefálicas/tratamiento farmacológico , Fitoquímicos/farmacología , Fitoquímicos/uso terapéutico
7.
Biochim Biophys Acta Mol Basis Dis ; 1869(6): 166720, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37062453

RESUMEN

Discovering novel cancer therapies has attracted extreme interest in the last decade. In this regard, multidrug resistance (MDR) to chemotherapies is a key challenge in cancer treatment. Cancerous cells are growingly become resistant to existing chemotherapeutics by employing diverse mechanisms, highlighting the significance of discovering approaches to overcome MDR. One promising strategy is utilizing aptamers as unique tools to target elements or signaling pathways incorporated in resistance mechanisms, or develop actively targeted drug delivery systems or chimeras enabling the precise delivery of novel agents to inhibit the conventionally undruggable resistance elements. Furthermore, due to their advantages over their proteinaceous counterparts, particularly antibodies, including improved targeting action, enhanced thermal stability, easier production, and superior tumor penetration, aptamers are emerging and have frequently been considered for developing cancer therapeutics. Here, we highlighted significant chemoresistance pathways in cancer and discussed the use of aptamers as prospective tools to surmount cancer MDR.


Asunto(s)
Resistencia a Antineoplásicos , Neoplasias , Humanos , Sistemas de Liberación de Medicamentos , Neoplasias/tratamiento farmacológico , Neoplasias/patología
8.
Curr Med Chem ; 30(7): 857-877, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-35796457

RESUMEN

In terms of frequency and aggressiveness, glioblastoma multiforme (GBM) is undoubtedly the most frequent and fatal primary brain tumor. Despite advances in clinical management, the response to current treatments is dismal, with a 2-year survival rate varying between 6 and 12 percent. Metformin, a derivative of biguanide widely used in treating type 2 diabetes, has been shown to extend the lifespan of patients with various malignancies. There is limited evidence available on the long-term survival of GBM patients who have taken metformin. This research examined the literature to assess the connection between metformin's anticancer properties and GBM development. Clinical findings, together with the preclinical data from animal models and cell lines, are included in the present review. This comprehensive review covers not only the association of hyperactivation of the AMPK pathway with the anticancer activity of metformin but also other mechanisms underpinning its role in apoptosis, cell proliferation, metastasis, as well as its chemo-radio-sensitizing behavior against GBM. Current challenges and future directions for developments and applications of metformin-based therapeutics are also discussed.


Asunto(s)
Neoplasias Encefálicas , Diabetes Mellitus Tipo 2 , Glioblastoma , Metformina , Animales , Metformina/farmacología , Metformina/uso terapéutico , Glioblastoma/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Proliferación Celular , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Línea Celular Tumoral
9.
Altern Lab Anim ; 51(1): 30-38, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36550678

RESUMEN

The pathological accumulation of quinolinic acid (QA) is often associated with neuritis and neuronal cell death in several neurodegenerative diseases, through the overproduction of free radicals. Urolithin B and auraptene have been reported to exert potent antioxidant effects - however, little is known about the protective effects of these compounds against QA-induced neurotoxicity. Therefore, this study aimed to explore the in vitro protective effects of urolithin B and auraptene against QA-induced neurotoxicity in the SH-SY5Y neuroblastoma cell line. The MTT assay was used to evaluate cell viability, and flow cytometry was carried out to evaluate effects on the cell cycle and apoptosis. The intracellular levels of reactive oxygen species (ROS) were also determined. Our findings showed that auraptene at non-toxic concentrations had no protective effect on QA-induced toxicity. However, urolithin B at concentrations of 0.6 µM and 2.5 µM enhanced the viability of cells treated with QA. Moreover, while the percentage of apoptotic cells (i.e. in the sub-G1 phase) was shown to significantly increase after QA treatment, pre-treatment with urolithin B reduced the number of these apoptotic cells. Furthermore, urolithin B, as an antioxidant, also significantly reduced QA-induced ROS production. Our findings suggest that urolithin B may possess potent antioxidant and neuroprotective effects against QA-induced neurotoxicity that merit further investigation.


Asunto(s)
Antioxidantes , Neuroblastoma , Humanos , Especies Reactivas de Oxígeno/metabolismo , Especies Reactivas de Oxígeno/farmacología , Antioxidantes/metabolismo , Antioxidantes/farmacología , Ácido Quinolínico/farmacología , Línea Celular Tumoral , Neuroblastoma/metabolismo , Neuroblastoma/patología , Apoptosis , Supervivencia Celular , Estrés Oxidativo/fisiología
10.
Biomed Pharmacother ; 155: 113740, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36166963

RESUMEN

Despite conventional treatment options including chemoradiation, patients with the most aggressive primary brain tumor, glioblastoma multiforme (GBM), experience an average survival time of less than 15 months. Regarding the malignant nature of GBM, extensive research and discovery of novel treatments are urgently required to improve the patients' prognosis. Autophagy, a crucial physiological pathway for the degradation and recycling of cell components, is one of the exciting targets of GBM studies. Interventions aimed at autophagy activation or inhibition have been explored as potential GBM therapeutics. This review, which delves into therapeutic techniques to block or activate autophagy in preclinical and clinical research, aims to expand our understanding of available therapies battling GBM.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Humanos , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/metabolismo , Autofagia
11.
J Control Release ; 348: 287-304, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35644289

RESUMEN

In the biomedical area, the interdisciplinary field of nanotechnology has the potential to bring numerous unique applications, including better tactics for cancer detection, diagnosis, and therapy. Nanoparticles (NPs) have been the topic of many research and material applications throughout the last decade. Unlike small-molecule medications, NPs are defined by distinct physicochemical characteristics, such as a large surface-to-volume ratio, which allows them to permeate live cells with relative ease. The versatility of NPs as both therapeutics and diagnostics makes them ideal for a broad spectrum of illnesses, from infectious diseases to cancer. A significant amount of data has been participated in the current scientific publications, emphasizing the concept that NPs often produce reactive oxygen species (ROS) to a larger degree than micro-sized particles. It is important to note that oxidative stress governs a wide range of cell signaling cascades, many of which are responsible for cancer cell cytotoxicity. Here, we aimed to provide insight into the signaling pathways triggered by oxidative stress in cancer cells in response to several types of nanomaterials, such as metallic and polymeric NPs and quantum dots. We discuss recent advances in developing integrated anticancer medicines based on NPs targeted to destroy malignant cells by increasing their ROS setpoint.


Asunto(s)
Nanopartículas , Neoplasias , Humanos , Nanopartículas/química , Nanotecnología , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo
12.
Semin Cancer Biol ; 86(Pt 2): 860-872, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35115226

RESUMEN

It has become necessary to accept the clinical reality of therapeutic agents targeting the cancer-associated immune system. In recent decades, several investigations have highlighted the role of inflammation in cancer development. It has now been recognized that inflammatory cells secrete mediators, including enzymes, chemokines, and cytokines. These secreted substances produce an inflammatory microenvironment that is critically involved in cancer growth. Inflammation may enhance genomic instability leading to DNA damage, activation of oncogenes, or compromised tumor suppressor activity, all of which may promote various phases of carcinogenesis. Conventional cancer treatment includes surgery, radiation, and chemotherapy. However, treatment failure occurs because current strategies are unable to achieve complete local control due to metastasis. Nanoparticles (NPs) are a broad spectrum of drug carriers typically below the size of 100 nm, targeting tumor sites while reducing off-target consequences. More importantly, NPs can stimulate innate and adaptive immune systems in the tumor microenvironment (TME); hence, they induce a cancer-fighting immune response. Strikingly, targeting cancer cells with NPs helps eliminate drug resistance and tumor recurrence, as well as prevents inflammation. Throughout this review, we provide recent data on the role of inflammation in cancer and explore nano-therapeutic initiatives to target significant mediators, for example, nuclear factor-kappa B (NF-κB), tumor necrosis factor-α (TNF-α), and interleukins (ILs) associated with cancer-related inflammation, to escort the immunomodulators to cancer cells and associated systemic compartments. We also highlight the necessity of better identifying inflammatory pathways in cancer pathophysiology to develop effective treatment plans.


Asunto(s)
Nanopartículas , Neoplasias , Humanos , Sistema de Administración de Fármacos con Nanopartículas , Recurrencia Local de Neoplasia , Neoplasias/patología , FN-kappa B/metabolismo , Inflamación/metabolismo , Microambiente Tumoral
13.
Crit Rev Oncol Hematol ; 172: 103625, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35158070

RESUMEN

The most widespread, malignant, and deadliest type of glial tumor is glioblastoma multiforme (GBM). Despite radiation, chemotherapy, and radical surgery, the median survival of afflicted individuals is about 12 months. Unfortunately, existing therapeutic interventions are abysmal. Dexamethasone (Dex), a synthetic glucocorticoid, has been used for many years to treat brain edema and inflammation caused by GBM. Several investigations have recently shown that Dex also exerts antitumoral effects against GBM. On the other hand, more recent disputed findings have questioned the long-held dogma of Dex treatment for GBM. Unfortunately, steroids are associated with various undesirable side effects, including severe immunosuppression and metabolic changes like hyperglycemia, which may impair the survival of GBM patients. Current ideas and concerns about Dex's effects on GBM cerebral edema, cell proliferation, migration, and its clinical outcomes were investigated in this study.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Neoplasias Encefálicas/patología , Proliferación Celular , Dexametasona/uso terapéutico , Glioblastoma/patología , Humanos
14.
Pharmacol Rep ; 74(2): 323-339, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35050491

RESUMEN

Since the discovery of phosphodiesterase-5 (PDE5) enzyme overexpression in the central nervous system (CNS) malignancies, investigations have explored the potential capacity of current PDE5 inhibitor drugs for repositioning in the treatment of brain tumors, notably glioblastoma multiforme (GBM). It has now been recognized that these drugs increase brain tumors permeability and enhance standard chemotherapeutics effectiveness. More importantly, studies have highlighted the promising antitumor functions of PDE5 inhibitors, e.g., triggering apoptosis, suppressing tumor cell growth and invasion, and reversing tumor microenvironment (TME) immunosuppression in the brain. However, contradictory reports have suggested a pro-oncogenic role for neuronal cyclic guanosine monophosphate (cGMP), indicating the beneficial function of PDE5 in the brain of GBM patients. Unfortunately, due to the inconsistent preclinical findings, only a few clinical trials are evaluating the therapeutic value of PDE5 inhibitors in GBM treatment. Accordingly, additional studies should be conducted to shed light on the precise effect of PDE5 inhibitors in GBM biology regarding the existing molecular heterogeneities among individuals. Here, we highlighted and discussed the previously investigated mechanisms underlying the impacts of PDE5 inhibitors in cancers, focusing on GBM to provide an overview of current knowledge necessary for future studies.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Neoplasias Encefálicas/tratamiento farmacológico , GMP Cíclico , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/uso terapéutico , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Humanos , Inhibidores de Fosfodiesterasa 5/farmacología , Inhibidores de Fosfodiesterasa 5/uso terapéutico , Microambiente Tumoral
15.
Life Sci ; 291: 120299, 2022 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-34999113

RESUMEN

Alzheimer's disease (AD), a type of dementia, is characterized by progressive memory decline and cognition impairment. Despite the considerable body of evidence regarding AD pathophysiology, current therapies merely slow down the disease progression, and a comprehensive therapeutic approach is unavailable. Accordingly, finding an efficient multifunctional remedy is necessary to blunt the increasing rate of AD incidence in the upcoming years. AD shares pathophysiological similarities (e.g., impairment of cognitive functions, insulin sensitivity, and brain glucose metabolism) with noninsulin-dependent diabetes mellitus (NIDDM), which offers the utilization of metformin, a biguanide hypoglycemic agent, as an alternative therapeutic approach in AD therapy. Emerging evidence has revealed the impact of metformin in patients suffering from AD. It has been described that metformin employs multiple mechanisms to improve cognition and memory impairment in pre-clinical AD models, including reduction of hippocampal amyloid-beta (Aß) plaque and neurofibrillary tangles (NFTs) load, suppression of inflammation, amelioration of mitochondrial dysfunction and oxidative stress, restriction of apoptotic neuronal death, and induction of neurogenesis. This review discusses the pre-clinical evidence, which may shed light on the role of metformin in AD and provide a more comprehensive mechanistic insight for future studies in this area of research.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Metformina/uso terapéutico , Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/metabolismo , Animales , Encéfalo/metabolismo , Cognición/efectos de los fármacos , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Humanos , Hipoglucemiantes/farmacología , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/fisiopatología , Metformina/metabolismo , Ovillos Neurofibrilares/metabolismo , Neurogénesis/efectos de los fármacos , Placa Amiloide
16.
J Complement Integr Med ; 19(3): 691-696, 2022 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-33964190

RESUMEN

OBJECTIVES: H-89 (a protein kinase AII [PKA II] inhibitor) impairs the spatial memory in the Morris water maze task in rats. In the present study, we aimed to study the protective effects of nicotine and O-acetyl-L-carnitine against H-89-induced spatial memory deficits. METHODS: Spatial memory impairment was induced by the bilateral intrahippocampal administration of 10 µM H-89 (dissolved in dimethyl sulfoxide, DMSO) to rats. The rats then received bilateral administrations of either nicotine (1 µg/µL, dissolved in saline) or O-acetyl-L-carnitine (100 µM/side, dissolved in deionized water) alone and in combination. Control groups received either saline, deionized water, or DMSO. RESULTS: The H-89-treated animals showed significant increases in the time and distance travelled to find hidden platforms, and there was also a significant decrease in the time spent in the target quadrant compared to DMSO-treated animals. Nicotine and O-acetyl-L-carnitine had no significant effects on H-89-induced spatial learning impairments alone, but the bilateral intrahippocampal co-administration of nicotine and O-acetyl-L-carnitine prevented H-89-induced spatial learning deficits and increased the time spent in the target quadrant in comparison with H-89-treated animals. CONCLUSIONS: Our results indicated the potential synergistic effects of nicotine and O-acetyl-L-carnitine in preventing protein kinase AII inhibitor (H-89)-induced spatial learning impairments.


Asunto(s)
Acetilcarnitina , Nicotina , Acetilcarnitina/metabolismo , Acetilcarnitina/farmacología , Animales , Dimetilsulfóxido/metabolismo , Dimetilsulfóxido/farmacología , Hipocampo/metabolismo , Isoquinolinas , Aprendizaje por Laberinto , Prueba del Laberinto Acuático de Morris , Nicotina/metabolismo , Nicotina/farmacología , Proteínas Quinasas/metabolismo , Proteínas Quinasas/farmacología , Ratas , Ratas Wistar , Aprendizaje Espacial , Sulfonamidas
17.
Neurotoxicology ; 85: 145-159, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34058247

RESUMEN

The amyloid-beta (Aß) fibrillation process seems to execute a principal role in the neuropathology of Alzheimer's disease (AD). Accordingly, novel therapeutic plans have concentrated on the inhibition or degradation of Aß oligomers and fibrils. Biocompatible nanoparticles (NPs), e.g., gold and iron oxide NPs, take a unique capacity in redirecting Aß fibrillation kinetics; nevertheless, their impacts on AD-related memory impairment have not been adequately evaluated in vivo. Here, we examined the effect of commercial PEGylated superparamagnetic iron oxide nanoparticles (SPIONs) on the learning and memory of an AD-animal model. The outcomes demonstrated the dose-dependent effect of SPIONs on Aß fibrillation and learning and memory processes. In vitro and in vivo findings revealed that Low doses of SPIONs inhibited Aß aggregation and ameliorated learning and memory deficit in the AD model, respectively. Enhanced level of hippocampal proteins, including brain-derived neurotrophic factor, BDNF, phosphorylated-cAMP response element-binding protein, p-CREB, and stromal interaction molecules, e.g., STIM1 and STIM2, were also observed. However, at high doses, SPIONs did not improve the detrimental impacts of Aß fibrillation on spatial memory and hippocampal proteins expression. Overall, we revealed the potential capacity of SPIONs on retrieval of behavioral and molecular manifestations of AD in vivo, which needs further investigations to determine the mechanistic effect of SPIONs in the AD conundrum.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Aprendizaje/efectos de los fármacos , Nanopartículas Magnéticas de Óxido de Hierro/administración & dosificación , Trastornos de la Memoria/tratamiento farmacológico , Polietilenglicoles/administración & dosificación , Moléculas de Interacción Estromal , Enfermedad de Alzheimer/inducido químicamente , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/antagonistas & inhibidores , Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/toxicidad , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Reacción de Fuga/efectos de los fármacos , Reacción de Fuga/fisiología , Aprendizaje/fisiología , Masculino , Trastornos de la Memoria/metabolismo , Fragmentos de Péptidos/toxicidad , Ratas , Ratas Wistar , Molécula de Interacción Estromal 1/metabolismo , Molécula de Interacción Estromal 2/metabolismo , Moléculas de Interacción Estromal/metabolismo
18.
Peptides ; 118: 170102, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31199948

RESUMEN

Apelin-13 is known to be one of the predominant neuropeptides with marked protective role in circuits involved in mood disturbances. The most putative hypothesis in pathophysiology of Alzheimer's disease (AD) is Amyloid beta (Aß) aggregation which interrupt proper function of hypothalamic-pituitary-adrenal (HPA) axis and are associated with anxiety. Here, we assessed the potential anxiolytic effect of Apelin-13 in a rodent cognitive impairment model induced by intrahippocampal Aß 25-35 administration. We evaluated the memory impairment and anxiogenic behavior using shuttle box and Elevated plus maze apparatuses. We also measured the glucocorticoid receptor (GR) and FK506 binding protein 51 (FKBP5) expression as important markers showing the proper feedback mechanism within the HPA axis. Our findings showed that Aß 25-35 administration induced memory impairment and anxiety behaviors. Apelin-13 exerted the anxiolytic effects and provided protection against Aß 25-35 -induced passive avoidance memory impairment. Moreover, Apelin-13 caused an increase in GR and a decrease in FKBP5 expression levels in Aß 25-35 treated animals. Taken together, these findings showed the anxiolytic effect of Apelin-13. This effect at least in part, may be mediated through the regulation of GR and FKBP5 expression levels which have a pivotal role in the appropriate negative feedback mechanism within the HPA axis. These data suggest that Apelin-13 might be considered as a potential neuropeptide defense that reduces anxiety along with neuroprotective effect against the Aß 25-35 -induced injury.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Región CA1 Hipocampal/efectos de los fármacos , Región CA1 Hipocampal/metabolismo , Péptidos y Proteínas de Señalización Intercelular/uso terapéutico , Receptores de Glucocorticoides/metabolismo , Proteínas de Unión a Tacrolimus/metabolismo , Péptidos beta-Amiloides/toxicidad , Animales , Ansiedad/inducido químicamente , Ansiedad/tratamiento farmacológico , Ansiedad/metabolismo , Western Blotting , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Sistema Hipotálamo-Hipofisario/metabolismo , Masculino , Trastornos de la Memoria/inducido químicamente , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/metabolismo , Fragmentos de Péptidos/toxicidad , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Sistema Hipófiso-Suprarrenal/metabolismo , Distribución Aleatoria , Ratas , Ratas Wistar
19.
ACS Chem Neurosci ; 10(5): 2299-2309, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30933476

RESUMEN

Alzheimer's disease (AD) is the most common type of neurodegenerative amyloid disorder causing progressive cognitive decline and memory loss. A considerable number of therapies for AD rely on inhibition/delay/dissociation of amyloid beta (Aß) oligomers and fibrils. In this case, nanoparticles (NPs) demonstrated substantial effects on the Aß fibrillation process; however, their effects on progressive cognitive decline and memory have been poorly investigated in vivo. In this study, acquisition and retention of spatial learning and memory are studied in a rat animal model of AD after intrahippocampal (IH) and intraperitoneal (IP) injections of a model NP, i.e., gold NPs (AuNPs). The outcomes revealed that the AuNPs could improve the acquisition and retention of spatial learning and memory in Aß treated rats as indicated by decreased time (Aß: 39.60 ± 3.23 s vs Aß+AuNPs: 25.78 ± 2.80 s) and distance (Aß: 917.98 ± 50.81 cm vs Aß+AuNPs: 589.09 ± 65.96 cm) of finding the hidden platform during training days and by increased time spent in the target quadrant (Aß: 19.40 ± 0.98 s vs Aß+AuNPs: 29.36 ± 1.14 s) in the probe test in Morris water maze (MWM). Expression of brain-derived neurotrophic factor, BDNF, cAMP response element binding protein, CREB, and stromal interaction molecules, e.g., STIM1 and STIM2 was also increased, supporting improved neural survival. Our outcomes may pave a way for mechanistic insights toward the role of NPs on retrieval of the deteriorated behavioral functions in brain tissue after AD outbreak.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/metabolismo , Oro , Nanopartículas del Metal/administración & dosificación , Molécula de Interacción Estromal 1/metabolismo , Molécula de Interacción Estromal 2/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/patología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Memoria/efectos de los fármacos , Nanopartículas del Metal/uso terapéutico , Ratones , Fosforilación/efectos de los fármacos , Ratas , Ratas Wistar
20.
Sensors (Basel) ; 18(11)2018 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-30404144

RESUMEN

Sensors provide aninterface between mechanical systems and the physical world. With the move towardsIndustry 4.0 and cyber-physical systems, demands for cost-effective sensors are rapidly increasing. Conventional sensors used for monitoring manufacturing processes are often bulky and need complex processes. In this study, a novel high-sensitive nanocomposite-based sensor is developed for measuring strain. The developed sensor is comprised of polyvinylidene fluoride (PVDF) as a piezoelectric polymer matrix, and embedded carbon nanotube (CNT) nanoparticles creating a conductive network. Exhibiting both piezoelectric and piezoresistive properties, the developed sensors are capable of strain measurement over a wide frequency band, including static and dynamic measurements. The piezoresistive and piezoelectric properties are fused to improve the overall sensitivity and frequency bandwidth of the sensor. To simulate the sensor, a 3D random walk model and a 2D finite element (FE) model are used to predict the electrical resistivity and the piezoelectric characteristics of the sensor, respectively. The developed models are verified with the experimental results. The developed nanocomposite sensors were employed for strain measurement of a cantilever beam under static load, impulse excitation, free and forced vibrations, collecting both piezoelectric and piezoresistive properties measurements. The obtained signals were fused and compared with those of a reference sensor. The results show that the sensor is capable of strain measurement in the range of 0⁻10 kHz, indicating its effectiveness at measuring both static and high frequency signals which is an important feature of the sensor.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...