Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 160
Filtrar
1.
Sci Rep ; 14(1): 19077, 2024 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-39154054

RESUMEN

Parkinson's disease (PD) is a chronic neurodegenerative case. As the disease progresses, the response time to doses of levodopa (L-Dopa) becomes shorter and the effects of the drug are severely limited by some undesirable side effects such as the 'on-off' phenomenon. In several diseases, including Parkinson's, nanoparticles can deliver antioxidant compounds that reduce oxidative stress. This study evaluates and compares the neuroprotective effects of L-Dopa-modified zinc nanoparticles (ZnNPs) in the 6-hydroxydopamine (6-OHDA)-induced PD rat model. For this purpose, the synthesis of NPs was carried out. Scanning electron microscopy, X-ray diffraction and Fourier transform infrared spectrophotometer were used for characterization. The rats were randomized into 9 experimental groups: control, lesion group (6-OHDA), 6-OHDA + 5 mg/kg L-Dopa, 6-OHDA + 10 mg/kg L-Dopa, 6-OHDA + 20 mg/kg L-Dopa, 6-OHDA + 20 mg/kg ZnNPs, 6-OHDA + 40 mg/kg ZnNPs, 6-OHDA + 30 mg/kg ZnNPs + L-Dopa, and 6-OHDA + 60 mg/kg ZnNPs + L-Dopa. Behavioral tests were performed on all groups 14 days after treatment. Phosphatase and tensin homolog, Excitatory amino acid transporter 1/2, and Glutamine synthetase gene analyses were performed on brain samples taken immediately after the tests. In addition, histological and immunohistochemical methods were used to determine the general structure and properties of the tissues. We obtained important findings that L-Dopa-modified ZnNPs increased the activity of glutamate transporters. Our experiment showed that glutamate increases neuronal cell vitality and improves behavioral performance. Therefore, L-Dopa-modified ZnNPs can be used to prevent neurotoxicity. According to what we found, results show that L-Dopa-modified ZnNPs will lend to the effective avoidance and therapy of PD.


Asunto(s)
Modelos Animales de Enfermedad , Levodopa , Fármacos Neuroprotectores , Oxidopamina , Óxido de Zinc , Animales , Levodopa/farmacología , Ratas , Fármacos Neuroprotectores/farmacología , Óxido de Zinc/química , Óxido de Zinc/farmacología , Masculino , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo , Nanopartículas del Metal/química , Nanopartículas/química , Enfermedad de Parkinson Secundaria/inducido químicamente , Enfermedad de Parkinson Secundaria/tratamiento farmacológico , Enfermedad de Parkinson Secundaria/patología , Enfermedad de Parkinson Secundaria/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ratas Wistar
2.
Int J Biol Macromol ; 277(Pt 3): 134398, 2024 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-39097068

RESUMEN

This study presents an innovative bi-layered three-dimensional skin-like nanopad (SLN) engineered for skin tissue regeneration. The SLN integrates a mechanically supportive polycaprolactone nanofibrous layer with a functional chitosan hydrogel film, mimicking natural skin. Our SLN exhibits superior flexibility, with a maximum elongation of 751.71 ± 125 % and exceptional porosity of 95 ± 4.5 %, ensuring effective exudate management due to its high water uptake capacity (4393 ± 72 %). FTIR analysis confirmed a distinctive fiber-hydrogel network within the SLN, which serves as a barrier against Staphylococcus aureus and Pseudomonas aeruginosa infiltration. In vitro cell viability assays with the human fibroblast have consistently demonstrated that 3D bi-layered SLN enhances fibroblast attachment, infiltration, and proliferation by 150 ± 20 %. In vivo studies in a rat model demonstrated significantly faster wound closure, with 60 % on day 7 and 87 % on day 10, compared to the 30 % and 60 % in controls, highlighting the efficacy of SLN. By mimicking the architecture of native skin, this biomimetic bi-layered SLN scaffold provides flexibility and support while accelerating in vivo wound closure by promoting fibroblast proliferation and infiltration. Customizable in size, depth, and shape, the engineered SLN has emerged as a promising platform for advanced wound care and tissue engineering.

3.
Mil Med Res ; 11(1): 35, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38835066

RESUMEN

Neuroendocrine neoplasms (NENs) are highly heterogeneous and potentially malignant tumors arising from secretory cells of the neuroendocrine system. Gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs) are the most common subtype of NENs. Historically, GEP-NENs have been regarded as infrequent and slow-growing malignancies; however, recent data have demonstrated that the worldwide prevalence and incidence of GEP-NENs have increased exponentially over the last three decades. In addition, an increasing number of studies have proven that GEP-NENs result in a limited life expectancy. These findings suggested that the natural biology of GEP-NENs is more aggressive than commonly assumed. Therefore, there is an urgent need for advanced researches focusing on the diagnosis and management of patients with GEP-NENs. In this review, we have summarized the limitations and recent advancements in our comprehension of the epidemiology, clinical presentations, pathology, molecular biology, diagnosis, and treatment of GEP-NETs to identify factors contributing to delays in diagnosis and timely treatment of these patients.


Asunto(s)
Tumores Neuroendocrinos , Neoplasias Pancreáticas , Neoplasias Gástricas , Humanos , Tumores Neuroendocrinos/terapia , Tumores Neuroendocrinos/epidemiología , Tumores Neuroendocrinos/diagnóstico , Neoplasias Pancreáticas/terapia , Neoplasias Pancreáticas/epidemiología , Neoplasias Pancreáticas/diagnóstico , Neoplasias Gástricas/epidemiología , Neoplasias Gástricas/terapia , Neoplasias Gástricas/diagnóstico , Neoplasias Intestinales/terapia , Neoplasias Intestinales/epidemiología , Neoplasias Intestinales/diagnóstico
6.
Toxicol Res (Camb) ; 13(3): tfae075, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38770183

RESUMEN

BACKGROUND: Cisplatin is a potent anticancer agent widely employed in chemotherapy. However, cisplatin leads to toxicity on non-targeted healthy organs, including the liver. We investigated the hepatoprotective mechanism of arbutin (ARB), a glycosylated hydroquinone, against cisplatin-induced hepatotoxicity. METHODS: Rats were orally administered with ARB (ARB1 = 50 mg/kg; ARB2 = 100 mg/kg) for 14 consecutive days against hepatotoxicity induced by a single dose of cisplatin (10 mg/kg) on day 15. Three days after the intraperitoneal cisplatin injection, serum and liver tissue were collected for subsequent analyses. RESULTS: Cisplatin triggered marked increases in serum AST, ALT, and ALP activities, hepatic malondialdehyde (MDA) and reactive oxygen species (ROS) coupled with a considerable diminution in hepatic activities of superoxide dismutase (SOD), catalase (CAT) and the concentration of reduced glutathione (GSH). The gene expressions of interleukin-1ß (IL-1ß), tumor necrosis factor (TNF-α), and IL-6 were notably increased. The pre-administration of ARB1 and ARB2 reduced AST, ALT and ALP in serum and restored SOD, CAT, GSH, ROS, MDA and cytokine levels which was also evidenced by alleviated hepatic lesions. Further, cisplatin-induced prominent alterations in the gene expressions of nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1), iNOS, NF-κB, Bax, Bcl-2, caspase-3 and 8-OHdG in the liver. Interestingly, ARB protected the liver and mitigated the cisplatin-induced alterations in serum AST, ALT, ALP, and reduced hepatic redox markers, 8-OdG, inflammatory markers and gene expressions. CONCLUSION: The findings demonstrate that ARB is a potential protective adjuvant against cisplatin-induced hepatotoxicity via inhibition of hepatic oxidative stress, inflammation, and apoptosis.

7.
Reprod Toxicol ; 127: 108611, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38782144

RESUMEN

The current study aimed to investigate the sensitivity of male testis parenchyma cells to chemotherapy agents and the protective effects and mechanisms of Morinda citrifolia (Noni) administration against structural and functional changes before and after chemotherapy (Paclitaxel (PTX)). For this purpose, rats were randomly assigned into four groups (Control = G1, PTX 5 mg/kg = G2; PTX + Noni 10 mg/kg = G3, PTX + Noni 20 mg/kg = G4). PTX was injected intraperitoneally for 4 consecutive weeks, at a dose of 5 mg/kg to all groups except the control group. Then noni was administrated in 10 (G3) and 20 (G4) mg/kg groups orally (gavage) for 14 days. Biochemical analyses, Real-Time Polymerase Chain Reaction (PCR), and immunohistochemical analyses were performed. According to our results, Total Oxidative Stress (TOS) and Malondialdehyde (MDA) were significantly increased in the PTX group (P < 0.01). Superoxide Dismutase (SOD) enzyme activity and Total Antioxidant Capacity (TAC) levels were decreased (P < 0.01). The changes in the rats treated with PTX + Noni 20 mg/kg were noteworthy. The increased levels of IL1-ß (Interleukin 1 beta) and TNFα (tumor necrosis factor-alpha) with PTX were down-regulated after treatment with PTX + Noni 20 mg/kg (P < 0.01) (9 % and 5 % respectively). In addition, Noni restored the testicular histopathological structure by reducing caspase-3 expression and significantly (61 %) suppressed oxidative DNA damage and apoptosis (by regulating the Bax (bcl-2-like protein 4)/Bcl-2 (B-cell lymphoma gene-2) ratio). In conclusion, Noni reduced cellular apoptosis and drastically changed Caspase 8 and Bax/Bcl-2 levels. Furthermore, it considerably decreases oxidative damage and can be used in testicular degeneration.


Asunto(s)
Antineoplásicos Fitogénicos , Morinda , Estrés Oxidativo , Paclitaxel , Extractos Vegetales , Testículo , Animales , Masculino , Morinda/química , Paclitaxel/toxicidad , Testículo/efectos de los fármacos , Testículo/patología , Testículo/metabolismo , Estrés Oxidativo/efectos de los fármacos , Extractos Vegetales/farmacología , Antineoplásicos Fitogénicos/toxicidad , Antineoplásicos Fitogénicos/farmacología , Superóxido Dismutasa/metabolismo , Malondialdehído/metabolismo , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/genética , Ratas Wistar , Caspasa 3/metabolismo , Interleucina-1beta/metabolismo , Proteína X Asociada a bcl-2/metabolismo , Proteína X Asociada a bcl-2/genética , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/genética , Sustancias Protectoras/farmacología , Ratas
8.
Mol Biol Rep ; 51(1): 532, 2024 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-38637360

RESUMEN

BACKGROUND: Doxorubicin is an effective antineoplastic agent but has limited clinical application because of its cumulative toxicities, including cardiotoxicity. Cardiotoxicity causes lipid peroxidation, genetic impairment, oxidative stress, inhibition of autophagy, and disruption of calcium homeostasis. Doxorubicin-induced cardiotoxicity is frequently tried to be mitigated by phytochemicals, which are derived from plants and possess antioxidant, anti-inflammatory, and anti-apoptotic properties. Arbutin, a natural antioxidant found in the leaves of the bearberry plant, has numerous pharmacological benefits, including antioxidant, anti-bacterial, anti-hyperglycemic, anti-inflammatory, and anti-tumor activity. METHODS AND RESULTS: The study involved male Wistar rats divided into three groups: a control group, a group treated with doxorubicin (20 mg/kg) to induce cardiac toxicity, a group treated with arbutin (100 mg/kg) daily for two weeks before doxorubicin administration. After treatment, plasma and heart tissue samples were collected for analysis. The samples were evaluated for oxidative stress parameters, including superoxide dismutase, malondialdehyde, and catalase, as well as for cardiac biomarkers, including CK, CK-MB, and LDH. The heart tissues were also analyzed using molecular (TNF-α, IL-1ß and Caspase 3), histopathological and immunohistochemical methods (8-OHDG, 4 Hydroxynonenal, and dityrosine). The results showed that arbutin treatment was protective against doxorubicin-induced oxidative damage by increasing SOD and CAT activity and decreasing MDA level. Arbutin treatment was similarly able to reverse the inflammatory response caused by doxorubicin by reducing TNF-α and IL-1ß levels and also reverse the apoptosis by decreasing caspase-3 levels. It was able to prevent doxorubicin-induced cardiac damage by reducing cardiac biomarkers CK, CK-MB and LDH levels. In addition to all these results, histopathological analyzes also show that arbutin may be beneficial against the damage caused by doxorubicin on heart tissue. CONCLUSION: The study suggests that arbutin has the potential to be used to mitigate doxorubicin-induced cardiotoxicity in cancer patients.


Asunto(s)
Antioxidantes , Cardiotoxicidad , Humanos , Ratas , Animales , Antioxidantes/metabolismo , Cardiotoxicidad/tratamiento farmacológico , Cardiotoxicidad/prevención & control , Cardiotoxicidad/etiología , Arbutina/farmacología , Arbutina/metabolismo , Arbutina/uso terapéutico , Miocardio/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Ratas Wistar , Doxorrubicina/efectos adversos , Estrés Oxidativo , Antiinflamatorios/farmacología , Apoptosis , Biomarcadores/metabolismo
9.
Biomedicines ; 12(4)2024 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-38672280

RESUMEN

BACKGROUND: Mitochondrial dysfunction and metabolic abnormalities are acknowledged as significant factors in the onset of neurodegenerative disorders such as Parkinson's disease (PD) and Alzheimer's disease (AD). Our research has demonstrated that the use of combined metabolic activators (CMA) may alleviate metabolic dysfunctions and stimulate mitochondrial metabolism. Therefore, the use of CMA could potentially be an effective therapeutic strategy to slow down or halt the progression of PD and AD. CMAs include substances such as the glutathione precursors (L-serine and N-acetyl cysteine), the NAD+ precursor (nicotinamide riboside), and L-carnitine tartrate. METHODS: Here, we tested the effect of two different formulations, including CMA1 (nicotinamide riboside, L-serine, N-acetyl cysteine, L-carnitine tartrate), and CMA2 (nicotinamide, L-serine, N-acetyl cysteine, L-carnitine tartrate), as well as their individual components, on the animal models of AD and PD. We assessed the brain and liver tissues for pathological changes and immunohistochemical markers. Additionally, in the case of PD, we performed behavioral tests and measured responses to apomorphine-induced rotations. FINDINGS: Histological analysis showed that the administration of both CMA1 and CMA2 formulations led to improvements in hyperemia, degeneration, and necrosis in neurons for both AD and PD models. Moreover, the administration of CMA2 showed a superior effect compared to CMA1. This was further corroborated by immunohistochemical data, which indicated a reduction in immunoreactivity in the neurons. Additionally, notable metabolic enhancements in liver tissues were observed using both formulations. In PD rat models, the administration of both formulations positively influenced the behavioral functions of the animals. INTERPRETATION: Our findings suggest that the administration of both CMA1 and CMA2 markedly enhanced metabolic and behavioral outcomes, aligning with neuro-histological observations. These findings underscore the promise of CMA2 administration as an effective therapeutic strategy for enhancing metabolic parameters and cognitive function in AD and PD patients.

10.
Biotech Histochem ; 99(3): 147-156, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38644776

RESUMEN

The purpose of this study was to evaluate the effects of syringic acid, an anti-oxidant, on indomethacin induced gastric ulcers in rats. Experimental groups were control, ulcer, ulcer treated with 20 mg/kg esomeprazole (a proton pump inhibitor that reduces acid secretion), and ulcer treated with 100 mg/kg syringic acid. Rats were pretreated with esomeprazole or syringic acid two weeks before ulcer induction. Our histopathological observations showed that either syringic acid or esomeprazole attenuated the severity of gastric mucosal damage. Moreover, syringic acid and esomeprazole pretreatments alleviated indomethacin-induced damage by regulating oxidative stress, inflammatory response, the level of transforming growth factor-ß (TGF-ß), expressions of COX and prostaglandin E2, cell proliferation, apoptosis and regulation of the NF-κB signaling pathway. We conclude that either esomeprazole or syringic acid administration protected the gastric mucosa from harmful effects of indomethacin. Syringic acid might, therefore be a potential therapeutic agent for preventing and treating indomethacin-induced gastric damage.


Asunto(s)
Apoptosis , Ácido Gálico , Indometacina , Inflamación , Estrés Oxidativo , Úlcera Gástrica , Animales , Indometacina/farmacología , Indometacina/toxicidad , Úlcera Gástrica/inducido químicamente , Úlcera Gástrica/tratamiento farmacológico , Úlcera Gástrica/patología , Estrés Oxidativo/efectos de los fármacos , Apoptosis/efectos de los fármacos , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Masculino , Ácido Gálico/análogos & derivados , Ácido Gálico/farmacología , Ratas , Ratas Sprague-Dawley , Mucosa Gástrica/efectos de los fármacos , Mucosa Gástrica/patología , Esomeprazol/farmacología
11.
Environ Toxicol Pharmacol ; 107: 104417, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38493879

RESUMEN

The present study was designed to evaluate whether AuNPs (gold nanoparticles) synthesized with the Cynara scolymus (CS) leaf exert protective and/or alleviative effects on arsenic (As)-induced hippocampal neurotoxicity in mice. Neurotoxicity in mice was developed by orally treating 10 mg/kg/day sodium arsenite (NaAsO2) for 21 days. 10 µg/g AuNPs, 1.6 g/kg CS, and 10 µg/g CS-AuNPs were administered orally simultaneously with 10 mg/kg As. CS and CS-AuNPs treatments showed down-regulation of TNF-α and IL-1ß levels. CS and CS-AuNPs also ameliorated apoptosis and reduced the alterations in the expression levels of D1 and D2 dopamine receptors induced by As. Simultaneous treatment with CS and CS-AuNPs improved As-induced learning, memory deficits, and motor coordination in mice assessed by water maze and locomotor tests, respectively. The results of this study provide evidence that CS-AuNPs demonstrated neuroprotective roles with antioxidant, anti-inflammatory, and anti-apoptotic effects, as well as improving D1 and D2 signaling, and eventually reversed neurobehavioral impairments.


Asunto(s)
Arsénico , Cynara scolymus , Nanopartículas del Metal , Extractos Vegetales , Ratones , Animales , Arsénico/metabolismo , Oro , Ratones Endogámicos BALB C , Nanopartículas del Metal/toxicidad , Hipocampo/metabolismo
12.
Cancer Lett ; 587: 216733, 2024 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-38360141

RESUMEN

Despite significant advances in diagnostic techniques and treatment approaches, the prognosis of pancreatic ductal adenocarcinoma (PDAC) is still poor. Previous studies have reported that S-phase kinase-associated protein 2 (SKP2), a subunit of the SCF E3 ubiquitin ligase complex, is engaged in the malignant biological behavior of some tumor entities. However, SKP2 has not been fully investigated in PDAC. In the present study, it was observed that high expression of SKP2 significantly correlates with decreased survival time. Further experiments suggested that SKP2 promotes metastasis by interacting with the putative transcription factor paraspeckle component 1 (PSPC1). According to the results of coimmunoprecipitation and ubiquitination assays, SKP2 depletion resulted in the polyubiquitination of PSPC1, followed by its degradation. Furthermore, the SKP2-mediated ubiquitination of PSPC1 partially depended on the activity of the E3 ligase TRIM21. In addition, inhibition of the SKP2/PSPC1 axis by SMIP004, a traditional inhibitor of SKP2, impaired the migration of PDAC cells. In summary, this study provides novel insight into the mechanisms involved in PDAC malignant progression. Targeting the SKP2/PSPC1 axis is a promising strategy for the treatment of PDAC.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Proteínas Quinasas Asociadas a Fase-S/genética , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Neoplasias Pancreáticas/genética , Ubiquitinación , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Carcinoma Ductal Pancreático/genética , Proteínas de Unión al ARN/metabolismo
13.
J Cell Mol Med ; 28(4): e18118, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38332529

RESUMEN

Opioids can be used for medical and non-medical purposes. Chronic pain such as cancer, as well as the frequent use of such drugs in places such as operating rooms and intensive care units, and in non-medical areas like drug abuse the effects and side effects of these drugs need to be examined in more detail. For this purpose, the effects of fentanyl and remifentanil drugs on neuroinflammation, oxidative stress and cholinesterase metabolism were investigated. Neuron cells (CRL-10742) were used for the evaluation of the toxicity of fentanyl and remifentanil. MTT, PON1 activity and total thiol levels for its effect on oxidative stress, AChE and BChE activities for its effect on the cholinergic system, and TNF, IL-8 and IL-10 gene levels for its neuroinflammation effect were determined. The highest neurotoxic dose of fentanyl and remifentanil was determined as 10 µg/mL. It was observed that the rate of neuron cells in this dose has decreased by up to 61.80% and 56.89%, respectively. The IL-8 gene expression level in both opioids was down-regulated while IL 10 gene level was up-regulated in a dose-dependent manner compared to the control. In our results, the TNF gene expression level differs between the two opioids. In the fentanyl group, it was seen to be up-regulated in a dose-dependent manner compared to the control. Fentanyl and remifentanil showed an inhibitory effect against PON1, while remifentanil showed an increase in total thiol levels. PON1, BChE and total thiol activities showed similarity with MTT.


Asunto(s)
Dolor Crónico , Fentanilo , Humanos , Fentanilo/toxicidad , Remifentanilo/farmacología , Piperidinas/toxicidad , Interleucina-8 , Enfermedades Neuroinflamatorias , Analgésicos Opioides/toxicidad , Estrés Oxidativo , Neuronas , Dolor Crónico/inducido químicamente , Compuestos de Sulfhidrilo , Arildialquilfosfatasa
14.
Chem Biol Interact ; 387: 110793, 2024 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-37949423

RESUMEN

The chemotherapeutic agent paclitaxel (PTX) causes testicular toxicity due to oxidative stress. Parthenolide (PTL), the active ingredient of the Tanacetum parthenium plant, is used to treat inflammation, dizziness, and spasms. In the present study, we evaluated the therapeutic effect of PTL on PTX-induced testicular toxicity in rats and its role in reproductive function. To this end, 6 groups were formed: control, PTX, sham, T1, T2, and T3. After testicular toxicity was induced in rats with 8 mg/kg PTX, the rats were treated with 1 mg/kg, 2 mg/kg, and 4 mg/kg PTL for 14 days. GSH and MDA levels were measured in rat testicular tissue after the last dose of PTL was administered. To determine the damage caused by PTX to testicular tissue by detecting 8-OHdG and iNOS, sections were prepared and examined histopathologically and immunohistochemically. Furthermore, the gene expressions and enzymatic activities of SOD, CAT, GPx, GST, and GR were investigated in all groups. After PTL treatment, MDA, 8-OHdG, and iNOS levels decreased while GSH levels increased in testicular tissue. Increased levels of antioxidant genes and enzymes also reduced oxidative stress. Additionally, the expression levels of the Dazl, Ddx4, and Amh genes, which are involved in gametogenesis and sperm production, decreased in case of toxicity and increased with PTL treatment. The data from this study show that PTL may have a therapeutic effect in the treatment of testicular damage by eliminating the oxidative stress-induced damage caused by PTX in testicular tissue, providing an effective approach to alleviating testicular toxicity, and playing an important role in reproduction/sperm production, especially at a dose of 4 mg/kg.


Asunto(s)
Paclitaxel , Semen , Ratas , Masculino , Animales , Paclitaxel/farmacología , Semen/metabolismo , Estrés Oxidativo , Testículo , Antioxidantes/metabolismo
15.
Tissue Cell ; 86: 102275, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37979397

RESUMEN

The prevalence of tendinopathy in patients with diabetes is well documented. Despite efforts to improve diabetes management, there is a lack of research on therapeutic agents targeting the core features of tendinopathy, namely, tenocyte apoptosis and extracellular matrix (ECM) damage. In this study, we investigated the potential of ginsenoside compound K (CK), known for its antidiabetic properties, to mitigate tenocyte apoptosis, inflammation, oxidative stress, and the metalloproteinase (MMP) system under hyperglycemic conditions. Our research also aimed to unravel the molecular mechanism underlying the effects of CK. The assessment of apoptosis involved observing intracellular chromatin condensation and measuring caspase 3 activity. To gauge oxidative stress, we examined cellular ROS levels and hydrogen peroxide and malondialdehyde concentrations. Western blotting was employed to determine the expression of various proteins. Our findings indicate that CK treatment effectively countered high glucose-induced apoptosis, inflammation, and oxidative stress in cultured tenocytes. Furthermore, CK normalized the expression of MMP-9, MMP-13, and TIMP-1. Notably, CK treatment boosted the expression of PPARγ and antioxidant enzymes. We conducted small interfering (si) RNA experiments targeting PPARγ, revealing its role in mediating CK's effects on tendinopathy features in hyperglycemic tenocytes. In conclusion, these in vitro results offer valuable insights into the potential therapeutic role of CK in managing tendinopathy among individuals with diabetes. By addressing crucial aspects of tendinopathy, CK presents itself as a promising avenue for future research and treatment development in this domain.


Asunto(s)
Diabetes Mellitus , Ginsenósidos , Tendinopatía , Humanos , Tenocitos/metabolismo , PPAR gamma/metabolismo , PPAR gamma/farmacología , PPAR gamma/uso terapéutico , Diabetes Mellitus/tratamiento farmacológico , Diabetes Mellitus/metabolismo , Matriz Extracelular/metabolismo , Apoptosis , Tendinopatía/tratamiento farmacológico , Tendinopatía/metabolismo , Inflamación/metabolismo
16.
Vet Med Sci ; 10(1): e1336, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-38109225

RESUMEN

OBJECTIVES: Bee venom is used for medicinal purposes, including the treatment of neurological and liver diseases, but its use as a primary health care approach for preventive purposes requires further exploration. The aim of this study was to provide the first investigation into the possible protective effects of bee venom against hepatic encephalopathy, a serious neurodegenerative disease. MATERIALS AND METHODS: An experimental animal study was conducted in which healthy albino Sprague-Dawley rats were randomized into three groups: healthy, control and bee venom groups. All rats were tested for locomotor activity at the beginning and end of the study. No intervention was made in the healthy group, whereas hepatic encephalopathy was induced in the control and bee venom groups by the administration of thioacetamide (TAA) (200 mg/kg/day). The bee venom group also received bee venom (5 mg/kg/day) subcutaneously every day for 14 days prior to the TAA administration. RESULTS: The results for the final locomotor activity tests were statistically better in the bee venom group than in the control group, supporting a beneficial effect of prophylactic bee venom application. Blood ammonia levels and liver weights, determined as indicators of inflammation, were lower in the bee venom group than in the control group and were close to levels in the healthy group, but not statistically significant. CONCLUSIONS: Bee venom administration has protective effects against the development of hepatic encephalopathy and offers a promising therapeutic opportunity in preventive medicine.


Asunto(s)
Venenos de Abeja , Encefalopatía Hepática , Enfermedades Neurodegenerativas , Animales , Ratas , Venenos de Abeja/uso terapéutico , Encefalopatía Hepática/prevención & control , Encefalopatía Hepática/veterinaria , Encefalopatía Hepática/tratamiento farmacológico , Enfermedades Neurodegenerativas/veterinaria , Ratas Sprague-Dawley
17.
Forensic Toxicol ; 42(2): 125-141, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38102417

RESUMEN

PURPOSE: There is very little information about the toxicological and pathological effects of synthetic cannabinoids, which have cannabis-like properties. This study was carried out to histopathologically, hematologically, and biochemically determine the toxic effects of acute and subacute exposure to a novel synthetic cannabinoid 1-(4-cyanobutyl)-N-(2-phenylpropan-2-yl)indazole-3-carboxamide in internal organs of adult male rats. METHODS: The cannabinoid was injected intraperitoneally at three doses (0.5, 1.0, and 2.0 mg/kg, body weight). The cannabinoid was administered to acute groups for 2 days and to subacute groups for 14 days. Observations were made for 14 days and various changes such as mortality, injury, and illness were recorded daily. Hematological and biochemical changes were evaluated and histopathological analyses in lung, liver, and kidney tissues were also performed. RESULTS: No mortality was observed. It was observed that there were fluctuations in hematological and serum biochemical parameters. Among the oxidative stress parameters, significant decreases in superoxide dismutase, catalase levels and significant increases in lipid peroxidation levels were determined. Serious pathological changes such as necrosis, vacuolation, congestion, and fibrosis were observed in the internal organs in a dose-dependent and time-dependent manner. It was also found that the synthetic cannabinoid triggered apoptosis in the organs. The results demonstrated that the most affected organ by the cannabinoid was the kidney. CONCLUSION: This study showed for the first time that CUMYL-4CN-BINACA adversely affects healthy male albino rats. It can be estimated that the abuse of the cannabinoid may harm human health in the same way.


Asunto(s)
Cannabinoides , Riñón , Peroxidación de Lípido , Hígado , Pulmón , Estrés Oxidativo , Animales , Masculino , Riñón/efectos de los fármacos , Riñón/patología , Riñón/metabolismo , Cannabinoides/toxicidad , Ratas , Hígado/efectos de los fármacos , Hígado/patología , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Superóxido Dismutasa/metabolismo , Indazoles/toxicidad , Catalasa/metabolismo , Relación Dosis-Respuesta a Droga , Pruebas de Toxicidad Subaguda , Ratas Wistar , Pruebas de Toxicidad Aguda
18.
Environ Sci Pollut Res Int ; 30(55): 117609-117623, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37872332

RESUMEN

Agents that will accelerate wound healing maintain their clinical importance in all aspects. The aim of this study is to determine the antimicrobial activity of zinc oxide nanoparticles (ZnO NPs) ZnO nanoparticles obtained by green synthesis from Capparis spinosa L. extract and their effect on in vitro wound healing. ZnO NPs were synthesized and characterized using Capparis spinosa L. extract. ZnO NPs were tested against nine ATCC-coded pathogen strains to determine antimicrobial activity. The effects of different doses (0.0390625-20 µg/mL) of NPs on cell viability were determined by MTT assay. The effect of ZnO NPs doses (0.0390625 µg/mL, 0.078125 µg/mL, 0.15625 µg/mL, 0.3125 µg/mL, 0.625 µg/mL, 1.25 µg/mL) that increase proliferation and migration on wound healing was investigated in an in vitro wound experiment. Cell culture medium obtained from the in vitro wound assay was used for biochemical analysis, and plate alcohol-fixed cells were used for immunohistochemical staining. It was determined that NPs formed an inhibition zone against the tested Gram-positive bacteria. The ZnO NPs doses determined in the MTT test provided faster wound closure in in-vitro conditions compared to the DMSO group. Biochemical analyses showed that inflammation and oxidative status decreased, while antioxidant levels increased in ZnO NPs groups. Immunohistochemical analyses showed increased expression levels of Bek/FGFR2, IGF, and TGF-ß associated with wound healing. The findings reveal the antimicrobial effect of ZnO nanoparticles obtained using Capparis spinosa L. extract in vitro and their potential applications in wound healing.


Asunto(s)
Antiinfecciosos , Capparis , Nanopartículas del Metal , Nanopartículas , Óxido de Zinc , Óxido de Zinc/química , Capparis/metabolismo , Nanopartículas/química , Cicatrización de Heridas , Antiinfecciosos/farmacología , Antiinfecciosos/química , Extractos Vegetales/farmacología , Extractos Vegetales/química , Nanopartículas del Metal/química , Antibacterianos/farmacología , Pruebas de Sensibilidad Microbiana
19.
Mol Biol Rep ; 50(11): 9143-9151, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37768465

RESUMEN

BACKGROUND: Quantum dots are usually particles smaller than 100 nm and have a low toxic effect. This study aimed to bioconjugate the anticancer effective melatonin agonist to quantum dots and demonstrate its effects in two cancer lines. This is the first study that aims to examine the anticancer activity of ramelteon bioconjugation to quantum dots, providing a new perspective on the use of Melatonin and its derivatives in cancer. METHODS AND RESULTS: For this purpose, first of all, cobalt sulfide (CoS) quantum dots were synthesized, bioconjugated and characterized with Punica granatum extract by green synthesis method. The effects of synthesized nanomaterials on neuroblastoma and prostate cancer cells were investigated. It was noted that nanomaterials reduced cell viability by 50% in neuroblastoma and prostate cancer lines at a dose of 50 µg/mL. Ramelteon bioconjugated nanomaterials reduced cancer cell viability by 1.4 times more than free melatonin. CoS quantum dots were determined to double the oxidative stress in the neuroblastoma cell line compared to the control, while no change was observed in prostate cancer. In the gene expression findings, it was observed that CoS nanoparticles caused an increase in the expression levels of apoptosis-related genes in the neuroblastoma cell line and induced key protein expression levels of pathways such as ROR-alpha in the prostate cancer cell line. CONCLUSION: As a result, it was observed that the viability of the neuroblastoma cell line decreased with apoptosis induced by oxidative stress, while this effect was observed in the DU-145 cell line via the ROR-alpha pathway.


Asunto(s)
Melatonina , Neuroblastoma , Neoplasias de la Próstata , Puntos Cuánticos , Humanos , Masculino , Melatonina/farmacología , Neoplasias de la Próstata/tratamiento farmacológico , Línea Celular Tumoral
20.
Drug Resist Updat ; 71: 101005, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37647746

RESUMEN

AIMS: Multidrug resistance in pancreatic cancer poses a significant challenge in clinical treatment. Bufalin (BA), a compound found in secretions from the glands of toads, may help overcome this problem. However, severe cardiotoxicity thus far has hindered its clinical application. Hence, the present study aimed to develop a cell membrane-camouflaged and BA-loaded polylactic-co-glycolic acid nanoparticle (CBAP) and assess its potential to counter chemoresistance in pancreatic cancer. METHODS: The toxicity of CBAP was evaluated by electrocardiogram, body weight, distress score, and nesting behavior of mice. In addition, the anticarcinoma activity and underlying mechanism were investigated both in vitro and in vivo. RESULTS: CBAP significantly mitigated BA-mediated acute cardiotoxicity and enhanced the sensitivity of pancreatic cancer to several clinical drugs, such as gemcitabine, 5-fluorouracil, and FOLFIRINOX. Mechanistically, CBAP directly bound to nucleotide-binding and oligomerization domain containing protein 2 (NOD2) and inhibited the expression of nuclear factor kappa-light-chain-enhancer of activated B cells. This inhibits the expression of ATP-binding cassette transporters, which are responsible for chemoresistance in cancer cells. CONCLUSIONS: Our findings indicate that CBAP directly inhibits NOD2. Combining CBAP with standard-of-care chemotherapeutics represents a safe and efficient strategy for the treatment of pancreatic cancer.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica , Neoplasias Pancreáticas , Animales , Ratones , Neoplasias Pancreáticas/tratamiento farmacológico , Cardiotoxicidad , Membrana Celular , Resistencia a Múltiples Medicamentos , Neoplasias Pancreáticas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...