Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros




Base de datos
Intervalo de año de publicación
1.
Drug Des Devel Ther ; 18: 2905-2917, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39011542

RESUMEN

Background: Our previous studies in vitro and in vivo have shown anti-severe acute respiratory syndrome coronavirus 2 activity of fingerroot extract (Boesenbergia rotunda) and its phytochemical panduratin A. Aim of Study: Therefore, the objective of this study was to determine the pharmacokinetic profiles of panduratin A, as a pure compound and in fingerroot extract, in rats. Materials and Methods: Male rats were randomly divided into four groups. Rats underwent intravenous administration of 4.5 mg/kg panduratin A, a single oral administration of 45 mg/kg panduratin A, or a multiple oral administration of 45 mg/kg panduratin A-consisted fingerroot extract for 7 consecutive days. The concentrations of panduratin A in plasma, tissues, and excreta were measured by using LCMS with a validated method. Results: The rats showed no change in health status after receiving all test preparations. The absolute oral bioavailability of panduratin A administered as pure panduratin A and fingerroot extract were approximately 9% and 6%, respectively. The peak concentrations for the single oral doses of 45 mg/kg panduratin A and fingerroot extract, were 4833 ± 659 and 3269 ± 819 µg/L, respectively. Panduratin A was mostly distributed in gastrointestinal organs, with the highest tissue-to-plasma ratio in the stomach. Approximately 20-30% of unchanged panduratin A from the administered dose was detected in feces while a negligible amount was found in urine. The major metabolites of administered panduratin A were identified in feces as oxidation and dioxidation products. Conclusion: Panduratin A from fingerroot extract showed low oral bioavailability, good tissue distribution, and partially biotransformed before excretion via feces. These findings will assist in developing fingerroot extract as a phytopharmaceutical product for COVID-19 treatment.


Asunto(s)
Disponibilidad Biológica , Extractos Vegetales , Ratas Sprague-Dawley , Zingiberaceae , Animales , Masculino , Extractos Vegetales/administración & dosificación , Extractos Vegetales/farmacocinética , Administración Oral , Ratas , Zingiberaceae/química , Distribución Tisular , Chalconas
2.
Toxicol Rep ; 12: 397-403, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38590343

RESUMEN

Investigating fine particulate matter (PM2.5) toxicity is crucial for health risk assessment and pollution control. This study explores the developmental toxicity of two PM2.5 sources: standard reference material 2786 (NIST, USA) and PM2.5 from Chakri Naruebodindra Medical Institute (CNMI, Thailand) located in the Bangkok Metropolitan area. Zebrafish embryos exposed to these samples exhibited embryonic mortality, with 50% lethal concentration (LC50) values of 1476 µg/mL for standard PM2.5 and 512 µg/mL for CNMI PM2.5. Morphological analysis revealed malformations, including pericardial and yolk sac edema, and blood clotting in both groups. Gene expression analysis highlighted source-specific effects. Standard PM2.5 downregulated sod1 and cat while upregulating gstp2. Inflammatory genes tnf-α and il-1b were upregulated, and nfkbi-αa was downregulated. Apoptosis-related genes bax, bcl-2, and casp3a were downregulated. CNMI PM2.5 consistently downregulated all examined genes. These findings underscore PM2.5 source variability's significance in biological system impact assessment, providing insights into pollutant-gene expression interactions. The study emphasizes the need for source-specific risk assessment and interventions to address PM2.5 exposure's health impacts effectively.

3.
Environ Toxicol Pharmacol ; 107: 104416, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38492761

RESUMEN

PM2.5-induced airway injury contributes to an increased rate of respiratory morbidity. However, the relationship between PM2.5 toxicants and acute cytotoxic effects remains poorly understood. This study aimed to investigate the mechanisms of PM2.5- and its constituent-induced cytotoxicity in human airway epithelial cells. Exposure to PM2.5 resulted in dose-dependent cytotoxicity within 24 h. Among the PM2.5 constituents examined, Cr(VI) at the dose found in PM2.5 exhibited cytotoxic effects. Both PM2.5 and Cr(VI) cause necrosis while also upregulating the expression of proinflammatory cytokine transcripts. Interestingly, exposure to the conditioned PM, obtained from adsorption in the Cr(VI)-reducing agents, FeSO4 and EDTA, showed a decrease in cytotoxicity. Furthermore, PM2.5 mechanistically enhances programmed pyroptosis through the activation of NLRP3/caspase-1/Gasdermin D pathway and increase of IL-1ß. These pyroptosis markers were reduced when exposure to conditioned PM. These findings provide a deeper understanding of mechanisms underlying PM2.5 and Cr(VI) in acute airway toxicity.


Asunto(s)
Cromo , Inflamasomas , Material Particulado , Humanos , Inflamasomas/metabolismo , Material Particulado/toxicidad , Piroptosis , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Células Epiteliales
4.
Toxicol Rep ; 12: 224-233, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38328737

RESUMEN

Background: Boesenbergia rotunda (fingerroot) rhizome extract contains two major bioactive components, panduratin A and pinostrobin. In our previous study, we found the anti-inflammatory effects of the fingerroot extract against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in golden Syrian hamsters. In the present study, we evaluated the sub-chronic toxicity of a fingerroot extract formulation over 90 consecutive days of oral administration. Methods: We enhanced the water solubility of a fingerroot extract by formulating it with cyclodextrin, containing panduratin A (29% w/w) and pinostrobin (32% w/w). This formulation was administered to male and female Wistar rats at doses of 25, 50, or 100 mg/kg/day for a duration of 90 days. Additionally, two recovery groups, comprising a control group and a high-dose group, were designated for a 14-day observation period to assess the persistence and reversibility of potential adverse effects. Throughout the experiment, we performed clinical and health observations, followed by hematological testing, clinical biochemistry analysis, necropsy examination, and histopathological evaluation at the end of the experiment. Results: The administration of the fingerroot extract formulation at doses of 25, 50, or 100 mg/kg/day did not result in mortality or clinical signs of toxicity. No clinically significant findings were associated with the oral administration of the fingerroot extract formulation. Conclusion: The fingerroot extract formulation showed no serious adverse effects at doses up to 100 mg/kg/day in Wistar rats under the experimental condition. Consequently, the No Observed Adverse Effect Level (NOAEL) was considered to be 100 mg/kg/day. This finding contributes significance for future developments involving fingerroot extract in herbal medicinal products targeting chronic inflammation.

5.
Pharm Biol ; 62(1): 183-194, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38351624

RESUMEN

CONTEXT: The therapeutic potential of andrographolide is hindered by its poor oral bioavailability and unpredictable pharmacokinetics, primarily due to its limited water solubility. OBJECTIVE: This work aimed to enhance the solubility and pharmacokinetics of andrographolide, a bioactive compound in Andrographis paniculata (Burm. f.) Nees (Acanthaceae), using solubilizing agents and a bioenhancer. MATERIALS AND METHODS: Four groups of beagles were compared: (1) A. paniculata powder alone (control), (2) A. paniculata powder with 50% weight/weight (w/w) ß-cyclodextrin solubilizer, (3) A. paniculata powder with 1% w/w sodium dodecyl sulfate (SDS) solubilizer, and (4) A. paniculata powder co-administered with 1% w/w SDS solubilizer and 10% piperine bioenhancer. All groups received a consistent oral dose of 3 mg/kg of andrographolide, administered both as a single dose and multiple doses over seven consecutive days. RESULTS: Thirteen chemical compounds were identified in A. paniculata powder, including 7 diterpenoids, 5 flavonoids, and 1 phenolic compound. A. paniculata co-administration with either 50% w/w ß-cyclodextrin or 1% w/w SDS, alone or in combination with 10% w/w piperine, significantly increased systemic andrographolide exposure by enhancing bioavailability (131.01% to 196.05%) following single and multiple oral co-administration. Glucuronidation is one possible biotransformation pathway for andrographolide, as evidenced by the excretion of glucuronide conjugates in urine and feces. CONCLUSION: The combination of solubilizing agents and a bioenhancer improved the oral bioavailability and pharmacokinetics of andrographolide, indicating potential implications for A. paniculata formulations and clinical therapeutic benefits. Further investigation in clinical studies is warranted.


Asunto(s)
Alcaloides , Andrographis , Benzodioxoles , Diterpenos , Piperidinas , Alcamidas Poliinsaturadas , beta-Ciclodextrinas , Animales , Perros , Andrographis paniculata , Disponibilidad Biológica , Biopotenciadores , Polvos , Andrographis/química , Extractos Vegetales , Excipientes
6.
Food Chem Toxicol ; 185: 114509, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38336016

RESUMEN

BACKGROUND: Centell-S, a water-soluble extract from Centella asiatica, is predominantly composed of madecassoside and asiaticoside, exceeding 80% w/w. Pursuing its development as an herbal medicinal product, Centell-S underwent sub-chronic toxicity assessment adhering to OECD GLP 408 standards. METHODS: In a study involving 100 Wistar rats, varying doses of Centell-S (50, 200, or 800 mg/kg/day) or a vehicle control were administered orally over 90 days. To evaluate Centell-S's safety profile, assessments included clinical observations, health examinations, clinical biochemistry analyses, and detailed anatomical pathology evaluations were conducted. RESULTS: Over the 90 days of treatment, the administration of Centell-S did not lead to any fatalities in the test animals. Clinical observations did not reveal any signs indicative of toxic effects. Notably, an increase in total white blood cell and lymphocyte counts was observed in both sexes, yet these levels returned to normal following a two-week discontinuation period post-treatment. CONCLUSIONS: Under the specific conditions of the OECD GLP 408, Repeated Dose 90-day Oral Toxicity Study in Rodents, the no observed adverse effect level (NOAEL) of Centell-S was 800 mg/kg/day. These findings are promising for the continued development of Centell-S as a phytopharmaceutical for clinical applications.


Asunto(s)
Centella , Triterpenos , Ratas , Masculino , Femenino , Animales , Ratas Wistar , Agua , Extractos Vegetales/toxicidad , Fitoterapia , Triterpenos/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA