Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
ACS Omega ; 9(6): 7188-7205, 2024 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-38371771

RESUMO

Background: Inorganic biomaterials are biologically active and are used as implants and drug delivery system. They have therapeutically active elements present in their framework that are released in the physiological milieu. Release of these dopants above the supraphysiological limit may produce adverse effects and physicochemical interactions with the loaded drugs. Therefore, this necessitates evaluating the in vivo release kinetics, biodistribution, and excretion profiles of dopants from barium-doped bioglass (BaBG) that has potential anti-inflammatory, antiulcer, and regenerative properties. Methods: In vitro leaching of Ca, Si, and Ba from BaBG was analyzed in simulated body fluid. Release kinetics post single-dose oral administration (1, 5, and 10 mg/kg) was performed in rats. Blood was collected at different time points, and pharmacokinetic parameters of released elements were calculated. The routes of excretion and biodistribution in major organs were evaluated using ICP-MS. Results: Elements were released after the oral administration of BaBG into the plasma. They showed dose-dependent release kinetics and mean residence time. Cmax was observed at 24 h for all elements, followed by a downhill fall. There was also a dose-dependent increase in the volume of distribution, and the clearance of dopants was mostly through feces. Ba and Si were biodistributed significantly in the liver, spleen, and kidneys. However, by the end of day 7, there was a leveling-off effect observed for all elements. Conclusion: All of the dopants exhibited a dose-dependent increase in release kinetics and biodistribution in vital organs. This study will help in dose optimization and understanding of various physicochemical and pharmacokinetic interactions when BaBG is used for future pharmacological studies.

2.
Toxicology ; 492: 153542, 2023 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-37150287

RESUMO

Indole-3-carbinol (I3C) is reported to have hepatic and neuroprotective properties. However, the I3C role in the protection of the liver and brain in the pathological condition of hepatic encephalopathy has not been investigated. Therefore, in the present study, we have assessed the hepatic and neuroprotective roles of I3C against thioacetamide (TAA)- induced hepatic encephalopathy in Wistar rats. TAA (300 mg/kg) was intraperitoneally administered to Wistar rats to induce hepatic encephalopathy. The elevated levels of ammonia in the blood, liver, and brain were substantially lowered by I3C treatment (25, 50, and 100 mg/kg, oral, 7 days). I3C significantly ameliorated the TAA-induced liver dysfunction by decreasing the alanine transaminase, aspartate transaminase, and alkaline phosphatase enzymes and reduced the elevated cytochrome P4502E1 (CYP2E1) activity in the liver and brain. Further, I3C alleviated mitochondrial dysfunction and oxidative stress in the brain. I3C treatment improved the anti-inflammatory cytokine interleukin (IL)- 10 while reducing inflammatory cytokines such as tumor necrosis factor-1 and IL-6 in hepatic encephalopathy rats. I3C reduced the levels of apoptotic indicators mediated by the mitochondria, including cytochrome c, caspase 9, and caspase 3. Concurrently, I3C mitigated the liver and brain histological abnormalities in hepatic encephalopathy rats. Therefore, the present study concluded that the I3C protected the liver and brain from TAA-induced hepatic encephalopathy injury by inhibiting CYP2E1 enzyme activity and decreasing ammonia, oxidative stress, inflammation, and apoptosis. The present study provides preclinical validation of I3C use as hepatic and neuroprotective for hepatic encephalopathy management.


Assuntos
Encefalopatia Hepática , Ratos , Animais , Encefalopatia Hepática/induzido quimicamente , Encefalopatia Hepática/tratamento farmacológico , Encefalopatia Hepática/prevenção & controle , Tioacetamida/toxicidade , Ratos Wistar , Amônia/efeitos adversos , Amônia/metabolismo , Citocromo P-450 CYP2E1/metabolismo , Fígado/metabolismo , Estresse Oxidativo , Citocinas/metabolismo
3.
Toxicology ; 485: 153409, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36572170

RESUMO

The effects of ELF-PEMF exposure on spontaneous alternation, anxiety, motor coordination, and locomotor activity have been discussed in various pre-clinical and clinical settings. Several epidemiological and experimental studies have demonstrated the potential effects of ELF-PEMF when exposed > âˆ¼1 h/day; however, very few studies have focused on understanding the influence of ELF-PEMF exposure of 1-3 mT with an exposure duration of < 1 h/day on spontaneous alternation, anxiety, motor coordination, and locomotor activity. Hence, we attempted to study the effects of ELF-PEMF exposure of 1-3 mT, 50 Hz with an exposure duration of 20 min each with a 4 h gap (2 times) on the cellular proliferation and morphologies of C6 (Glial) cells and spontaneous alternation, anxiety, motor coordination and locomotor activity of Wistar rats under in vitro and in vivo conditions, respectively. The results showed that ELF-PEMF exposure did not induce any significant levels of cellular fragmentation and changes in the morphology of glial cells. Also, the outcomes revealed no noticeable effects on spontaneous alternation, anxiety, motor coordination, and locomotor activity in PEMF-exposed groups compared with the control. No undesirable side effects were observed at the highest dose (B=3 mT). We also performed histological analysis of the selected brain sections (hippocampus and cortex) following ELF-PEMF exposure. Incidentally, no significant changes were observed in cortical cell counts, tissue structure, and morphology.


Assuntos
Ansiedade , Neuroglia , Ratos , Animais , Ratos Wistar , Proliferação de Células , Ansiedade/induzido quimicamente , Locomoção , Campos Eletromagnéticos
4.
Biomater Adv ; 140: 213080, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35985067

RESUMO

One of the recent challenges in the design/development of prosthetic orthopedic implants is to address the concern of local/systemic toxicity of debris particles, released due to wear or degradation. Such debris particles often lead to inflammation at the implanted site or aseptic loosening of the prosthesis which results in failure of the implant during long run. Several in vitro studies demonstrated the potentiality of piezoelectric sodium potassium niobate [NaxK1-xNbO3 (x = 0.2, 0.5, 0.8), NKN] as an emerging next-generation polarizable orthopedic implant. In this perspective, we performed an in vivo study to examine the local and systemic toxicity of NKN nanoparticulates, as a first report. In the present study, male Wistar rats were intra-articularly injected to the knee joint with 100 µl of NKN nanoparticulates (25 mg/ml in normal saline). After 7 days of exposure, the histopathological analyses demonstrate the absence of any inflammation or dissemination of nanoparticulates in vital organs such as heart, liver, kidney and spleen. The anti-inflammatory cytokines (IL-4 and IL-10) profile analyses suggest the increased anti-inflammatory response in the treated rats as compared to non-injected (control) rats, preferably for the sodium and potassium rich NKN i.e., Na0.8K0.2NbO3 and Na0.2K0.8NbO3. The biochemical analyses revealed no pathological changes in the liver and kidney of particulate treated rats. The present study is the first proof to confirm the non-toxic nature of NKN nanoparticulates which provides a step forward towards the development of prosthetic orthopedic implants using biocompatible piezoelectric NKN ceramics.


Assuntos
Inflamação , Engenharia Tecidual , Animais , Masculino , Nióbio , Óxidos , Potássio , Ratos , Ratos Wistar , Sódio
5.
Life (Basel) ; 12(8)2022 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-36013322

RESUMO

Paclitaxel (PAC) has been approved by FDA for clinical use (Taxol®), yet dose-dependent severe toxicity due to the adjuvant Cremophor EL® in combination with ethanol is a major drawback. The drawbacks of the current therapy can be overcome by (i) finding a suitable vehicle that cannot only bypass the above adjuvant but also be used to deliver drugs orally and (ii) combining the PAC with some other chemotherapeutics to have the enhanced therapeutic efficacy. In the current work, we have used folic acid (FA) functionalized bovine milk-derived exosomes for oral delivery of PAC in combination with 5-fluorouracil (5-FU). Exosomes before and after the drug loading were found to have a particle size in the range of 80-100 nm, polydispersity index (PDI ~0.20), zeta potential (~-25 mV), entrapment efficiency (~82%), practical drug loading (~28%) and sustained drug release for 48 h. Significant decreases in IC50 were observed in the case of exosomes loaded drugs which further improved following the FA functionalization. FA functionalized coumarin-6-loaded exosomes showed remarkably higher cellular uptake in comparison with free coumarin-6. Moreover, FA-functionalized drug-loaded exosomes showed a higher apoptotic index with better control over cell migration. Collectively, data suggested the enhanced efficacy of the combination following its loading to the folic acid functionalized exosomes against breast cancer.

6.
Biomater Sci ; 9(24): 8111-8147, 2021 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-34766608

RESUMO

Tissue engineering (TE), a new paradigm in regenerative medicine, repairs and restores the diseased or damaged tissues and eliminates drawbacks associated with autografts and allografts. In this context, many biomaterials have been developed for regenerating tissues and are considered revolutionary in TE due to their flexibility, biocompatibility, and biodegradability. One such well-documented biomaterial is bioactive glasses (BGs), known for their osteoconductive and osteogenic potential and their abundant orthopedic and dental clinical applications. However, in the last few decades, the soft tissue regenerative potential of BGs has demonstrated great promise. Therefore, this review comprehensively covers the biological application of BGs in the repair and regeneration of tissues outside the skeleton system. BGs promote neovascularization, which is crucial to encourage host tissue integration with the implanted construct, making them suitable biomaterial scaffolds for TE. Moreover, they heal acute and chronic wounds and also have been reported to restore the injured superficial intestinal mucosa, aiding in gastroduodenal regeneration. In addition, BGs promote regeneration of the tissues with minimal renewal capacity like the heart and lungs. Besides, the peripheral nerve and musculoskeletal reparative properties of BGs are also reported. These results show promising soft tissue regenerative potential of BGs under preclinical settings without posing significant adverse effects. Albeit, there is limited bench-to-bedside clinical translation of elucidative research on BGs as they require rigorous pharmacological evaluations using standardized animal models for assessing biomolecular downstream pathways.


Assuntos
Vidro , Engenharia Tecidual , Animais , Materiais Biocompatíveis , Regeneração Óssea , Osteogênese
7.
J Control Release ; 335: 481-497, 2021 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-34087250

RESUMO

Bioactive glasses (BAGs) were invented five decades ago and have been widely used clinically in orthopedic and stomatology. However, in the past two decades, BAGs have been explored immensely by several researchers worldwide as a multifunctional delivery system for a multitude of therapeutics ranging from metal ions to small molecules (e.g., drugs) and macromolecules (e.g., DNA). The impetus for devising a BAG-based delivery system in the 21st century is based upon the facilitative properties it offers for entrapment of a wide range of therapeutic molecules and the tailorable controlled release kinetics to the target tissue site along with the biological activity of the ionic dissolution products in several pathological conditions such as osteoporosis, cancer, infection, and inflammation. This review comprises two parts: the first part discusses the need for a new delivery system and how the journey from melt quench progressed towards template-based sol-gel mesoporous. In the second part, we have comprehended the scientific advancements made so far, emphasizing BAGs as a delivery system ranging from therapeutic ions to phytopharmaceuticals. We have also highlighted a few loopholes that have prevented bench-to-bedside clinical translation of a plethora of elucidative researches done so far.


Assuntos
Vidro , Preparações Farmacêuticas , Materiais Biocompatíveis , Humanos , Íons
8.
Neurochem Int ; 145: 105013, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33689806

RESUMO

Debilitating neuropsychiatric and neurodegenerative conditions are associated with complex multifactorial pathophysiology. Their treatment strategies often only provide symptomatic relief, delaying disease progression without giving a complete cure. Potent and safer treatment alternatives beyond symptomatic relief are sought. Herbal supplements have surely been explored due to their multiple component nature to enhance the effect of western medications. One such well-documented nutraceutical in the ancient Greek, Chinese, and Ayurvedic medicine system known for its various medicinal benefits is Asparagus racemosus. Widely used for its lactogenic properties, A. racemosus is also cited in Ayurveda as a nervine tonic. A. racemosus based nutraceuticals have shown to possess adaptogenic, neuroprotective, antioxidant, anti-inflammatory, and nootropic activity under preclinical and clinical settings without posing significant adverse effects. A. racemosus extracts restore the perturbed neurotransmitters and prevent oxidative neuronal damage. From the available neuropharmacological researches, the physiological actions of A. racemosus can ultimately be directed for either augmentation of cognitive ability or in the management of neurological conditions such as stress, anxiety, depression, epilepsy, Parkinson's, and Alzheimer's disease. The studies focus on the multi-component extract, and the lack of standardization has been a major hurdle in preventing the allotment of reported neuropharmacological activity to one of the phytoconstituent. Herbal standardization of the plant extract based on a specific biomarker can help elucidate the intricate biomolecular pathway and neurocircuitries being involved. This, followed by rigorous standardized clinical trials, fixing dosages, and determining contraindications would facilitate the translation of A. racemosus to a FDA-approved neuromedicine for neurological disorders.


Assuntos
Asparagus , Suplementos Nutricionais , Transtornos Mentais/dietoterapia , Doenças Neurodegenerativas/dietoterapia , Fitoterapia/métodos , Extratos Vegetais/administração & dosagem , Animais , Antioxidantes/administração & dosagem , Antioxidantes/isolamento & purificação , Humanos , Transtornos Mentais/patologia , Doenças Neurodegenerativas/patologia , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Componentes Aéreos da Planta , Extratos Vegetais/isolamento & purificação
9.
Eur J Med Chem ; 215: 113224, 2021 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-33582578

RESUMO

Alzheimer's disease (AD) is multifactorial, progressive neurodegeneration with impaired behavioural and cognitive functions. The multitarget-directed ligand (MTDL) strategies are promising paradigm in drug development, potentially leading to new possible therapy options for complex AD. Herein, a series of novel MTDLs phenylsulfonyl-pyrimidine carboxylate (BS-1 to BS-24) derivatives were designed and synthesized for AD treatment. All the synthesized compounds were validated by 1HNMR, 13CNMR, HRMS, and BS-19 were structurally validated by X-Ray single diffraction analysis. To evaluate the plausible binding affinity of designed compounds, molecular docking study was performed, and the result revealed their significant interaction with active sites of acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE). The synthesized compounds displayed moderate to excellent in vitro enzyme inhibitory activity against AChE and BuChE at nanomolar (nM) concentration. Among 24 compounds (BS-1 to BS-24), the optimal compounds (BS-10 and BS-22) displayed potential inhibition against AChE; IC50 = 47.33 ± 0.02 nM and 51.36 ± 0.04 nM and moderate inhibition against BuChE; IC50 = 159.43 ± 0.72 nM and 153.3 ± 0.74 nM respectively. In the enzyme kinetics study, the compound BS-10 displayed non-competitive inhibition of AChE with Ki = 8 nM. Respective compounds BS-10 and BS-22 inhibited AChE-induced Aß1-42 aggregation in thioflavin T-assay at 10 µM and 20 µM, but BS-10 at 10 µM and 20 µM concentrations are found more potent than BS-22. In addition, the aggregation properties were determined by the dynamic light scattering (DLS) and was found that BS-10 and BS-22 could significantly inhibit self-induced as well as AChE-induced Aß1-42 aggregation. The effect of compounds (BS-10 and BS-22) on the viability of MC65 neuroblastoma cells and their capability to cross the blood-brain barrier (BBB) in PAMPA-BBB were further studied. Further, in silico approach was applied to analyze physicochemical and pharmacokinetics properties of the designed compounds via the SwissADME and PreADMET server. Hence, the novel phenylsulfonyl-pyrimidine carboxylate derivatives can act as promising leads in the development of AChE inhibitors and Aß disaggregator for the treatment of AD.


Assuntos
Fármacos Neuroprotetores/farmacologia , Nootrópicos/farmacologia , Pirimidinas/farmacologia , Sulfonamidas/farmacologia , Acetilcolinesterase/metabolismo , Doença de Alzheimer/tratamento farmacológico , Barreira Hematoencefálica/efeitos dos fármacos , Butirilcolinesterase/metabolismo , Linhagem Celular Tumoral , Inibidores da Colinesterase/síntese química , Inibidores da Colinesterase/metabolismo , Inibidores da Colinesterase/farmacologia , Humanos , Simulação de Acoplamento Molecular , Fármacos Neuroprotetores/síntese química , Fármacos Neuroprotetores/metabolismo , Nootrópicos/síntese química , Nootrópicos/metabolismo , Ligação Proteica , Pirimidinas/síntese química , Pirimidinas/metabolismo , Sulfonamidas/síntese química , Sulfonamidas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA