Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 111
Filtrar
1.
Blood Adv ; 8(7): 1747-1759, 2024 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-38324726

RESUMEN

ABSTRACT: Therapeutic vaccination has long been a promising avenue for cancer immunotherapy but is often limited by tumor heterogeneity. The genetic and molecular diversity between patients often results in variation in the antigens present on cancer cell surfaces. As a result, recent research has focused on personalized cancer vaccines. Although promising, this strategy suffers from time-consuming production, high cost, inaccessibility, and targeting of a limited number of tumor antigens. Instead, we explore an antigen-agnostic polymeric in situ cancer vaccination platform for treating blood malignancies, in our model here with acute myeloid leukemia (AML). Rather than immunizing against specific antigens or targeting adjuvant to specific cell-surface markers, this platform leverages a characteristic metabolic and enzymatic dysregulation in cancer cells that produces an excess of free cysteine thiols on their surfaces. These thiols increase in abundance after treatment with cytotoxic agents such as cytarabine, the current standard of care in AML. The resulting free thiols can undergo efficient disulfide exchange with pyridyl disulfide (PDS) moieties on our construct and allow for in situ covalent attachment to cancer cell surfaces and debris. PDS-functionalized monomers are incorporated into a statistical copolymer with pendant mannose groups and TLR7 agonists to target covalently linked antigen and adjuvant to antigen-presenting cells in the liver and spleen after IV administration. There, the compound initiates an anticancer immune response, including T-cell activation and antibody generation, ultimately prolonging survival in cancer-bearing mice.


Asunto(s)
Cisteína , Leucemia Mieloide Aguda , Humanos , Ratones , Animales , Cisteína/uso terapéutico , Modelos Animales de Enfermedad , Leucemia Mieloide Aguda/tratamiento farmacológico , Adyuvantes Inmunológicos , Antígenos de Neoplasias , Activación de Linfocitos , Disulfuros/uso terapéutico
2.
ACS Appl Mater Interfaces ; 16(6): 6859-6867, 2024 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-38299497

RESUMEN

The rapid development of nanomedicine has considerably advanced precision therapy for cancer treatment. Superior to traditional chemotherapy, emerging theranostic nanoprodrugs can effectively realize inherent self-tracking, targeted drug delivery, stimuli-triggered drug release, and reduced systemic toxicity of chemotherapeutic drugs. However, theranostic nanoprodrugs with real-time drug release monitoring have remained rare so far. In this work, we developed a new glutathione-responsive theranostic nanoprodrug with a high drug-loading content of 59.4 wt % and an average nanoscale size of 46 nm, consisting of the anticancer drug paclitaxel and a fluorescent imaging probe with a high fluorescence quantum yield, which are linked by a disulfide-based glutathione-sensitive self-immolating linker. The strong fluorescence emission of the fluorophore enables efficacious self-tracking and sensitive fluorescence "ON-OFF" glutathione sensing. Upon encountering high-level glutathione in cancer cells, the disulfide bond is cleaved, and the resulting linker halves spontaneously collapse into cyclic small molecules at the same pace, leading to the simultaneous release of the therapeutic drug and the fluorescence-OFF imaging probe. Thereby, the drug release process is efficiently monitored by the fluorescence change in the nanoprodrug. The nanoprodrugs exerted high cytotoxicity toward various cancer cells, especially for A549 and HEK-293 cells, in which the nanoprodrugs generated better therapeutic effects than free paclitaxel. Our work demonstrated a new modality of smart theranostic nanoprodrugs for precise cancer therapy.


Asunto(s)
Nanopartículas , Neoplasias , Humanos , Medicina de Precisión , Línea Celular Tumoral , Liberación de Fármacos , Electrones , Células HEK293 , Paclitaxel/farmacología , Paclitaxel/uso terapéutico , Nanomedicina Teranóstica/métodos , Imagen Óptica/métodos , Glutatión/metabolismo , Disulfuros/uso terapéutico , Nanopartículas/química , Neoplasias/diagnóstico por imagen , Neoplasias/tratamiento farmacológico
3.
ACS Appl Mater Interfaces ; 15(27): 32177-32187, 2023 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-37387421

RESUMEN

The self-association of metabolites into well-ordered assemblies at the nanoscale has significant biological and medical implications. The thiol-containing amino acid cysteine (CYS) can assemble into amyloid-like nanofibrils, and its oxidized form, the disulfide-bonded cystine (CTE), forms hexagonal crystals as those found in cystinuria due to metabolic disorder. Yet, there have been no attempts to connect these two phenomena, especially the fibril-to-crystal transition. Here, we reveal that these are not separated events, and the CYS-forming amyloid fibrils are mechanistically linked to hexagonal CTE crystals. For the first time, we demonstrated that cysteine fibrils are a prerequisite for forming cystine crystals, as observed experimentally. To further understand this mechanism, we studied the effects of thiol-containing cystinuria drugs (tiopronin, TIO; and d-penicillamine, PEN) and the canonical epigallocatechin gallate (EGCG) amyloid inhibitor on fibril formation by CYS. The thiol-containing drugs do not solely interact with monomeric CYS via disulfide bond formation but can disrupt amyloid formation by targeting CYS oligomers. On the other hand, EGCG forms inhibitor-dominant complexes (more than one EGCG molecule per cysteine unit) to prevent CYS fibril formation. Interestingly, while CYS can be oxidized into CTE, the thiol drugs can reduce CTE back to CYS. We thus suggest that the formation of crystals in cystinuria could be halted at the initial stage by targeting CYS fibril formation as an alternative to solubilizing the water-insoluble hexagonal CTE crystals at a later stage. Taken together, we depicted a complex hierarchical organization in a simple amino acid assembly with implications for therapeutic intervention.


Asunto(s)
Cisteína , Cistinuria , Humanos , Cisteína/química , Cistina/química , Cistinuria/tratamiento farmacológico , Aminoácidos/uso terapéutico , Amiloide/química , Disulfuros/uso terapéutico
4.
Int J Mol Sci ; 24(4)2023 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-36835218

RESUMEN

Dairy farming is the most important economic activity in animal husbandry. Mastitis is the most common disease in dairy cattle and has a significant impact on milk quality and yield. The natural extract allicin, which is the main active ingredient of the sulfur-containing organic compounds in garlic, has anti-inflammatory, anticancer, antioxidant, and antibacterial properties; however, the specific mechanism underlying its effect on mastitis in dairy cows needs to be determined. Therefore, in this study, whether allicin can reduce lipopolysaccharide (LPS)-induced inflammation in the mammary epithelium of dairy cows was investigated. A cellular model of mammary inflammation was established by pretreating bovine mammary epithelial cells (MAC-T) with 10 µg/mL LPS, and the cultures were then treated with varying concentrations of allicin (0, 1, 2.5, 5, and 7.5 µM) added to the culture medium. MAC-T cells were examined using RT-qPCR and Western blotting to determine the effect of allicin. Subsequently, the level of phosphorylated nuclear factor kappa-B (NF-κB) was measured to further explore the mechanism underlying the effect of allicin on bovine mammary epithelial cell inflammation. Treatment with 2.5 µM allicin considerably decreased the LPS-induced increase in the levels of the inflammatory cytokines interleukin-1ß (IL-1ß), interleukin-6 (IL-6), interleukin-8 (IL-8), and tumor necrosis factor-α (TNF-α) and inhibited activation of the NOD-like receptor protein 3 (NLRP3) inflammasome in cow mammary epithelial cells. Further research revealed that allicin also inhibited the phosphorylation of inhibitors of nuclear factor kappa-B-α (IκB-α) and NF-κB p65. In mice, LPS-induced mastitis was also ameliorated by allicin. Therefore, we hypothesize that allicin alleviated LPS-induced inflammation in the mammary epithelial cells of cows probably by affecting the TLR4/NF-κB signaling pathway. Allicin will likely become an alternative to antibiotics for the treatment of mastitis in cows.


Asunto(s)
Disulfuros , Mastitis Bovina , FN-kappa B , Ácidos Sulfínicos , Animales , Bovinos , Femenino , Ratones , Disulfuros/uso terapéutico , Células Epiteliales/metabolismo , Inflamación/metabolismo , Interleucina-6/metabolismo , Lipopolisacáridos , Mastitis Bovina/tratamiento farmacológico , FN-kappa B/metabolismo , Transducción de Señal , Ácidos Sulfínicos/uso terapéutico , Receptor Toll-Like 4/metabolismo
5.
Bioconjug Chem ; 34(3): 489-500, 2023 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-36693213

RESUMEN

Dynamic covalent poly(disulfide)-based cross-linked nanoaggregates, termed nanonetworks (NNs), endowed with pH- and redox-responsive degradation features have been fabricated for stable noncovalent encapsulation and triggered cargo release in a controlled fashion. A bioderived lipoic acid-based Gemini surfactant-like amphiphilic molecule was synthesized for the preparation of nanoaggregates. It self-assembles by a entropy-driven self-assembly process in aqueous milieu. To further stabilize the self-assembled nanostructure, the core was cross-linked by ring-opening disulfide exchange polymerization (RODEP) of 1,2-dithiolane rings situated inside the core of the nanoaggregates. The cross-linked nanoaggregates, i.e., nanonetwork, are found to be stable in the presence of blood serum, and also, they maintain the self-assembled structure even below the critical aggregation concentration (CAC) as probed by dynamic light scattering (DLS) experiments. The nanonetwork showed almost 50% reduction in guest leakage compared to that of the nanoaggregates as shown by the release profile in the absence of stimuli, suggesting high encapsulation stability as evidenced by the fluorescence resonance energy transfer (FRET) experiment. The decross-linking of the nanonetwork occurs in response to redox and pH stimuli due to disulfide reduction and ß-thioester hydrolysis, respectively, thus empowering disassembly-mediated controlled cargo release up to ∼87% for 55 h of incubation. The biological evaluation of the doxorubicin (DOX)-loaded nanonetwork revealed environment-specific surface charge modulation-mediated cancer cell-selective cellular uptake and cytotoxicity. The benign nature of the nanonetwork toward normal cells makes the system very promising in targeted drug delivery applications. Thus, the ease of synthesis, nanonetwork fabrication reproducibility, robust stability, triggered drug release in a controlled fashion, and cell-selective cytotoxicity behavior, we believe, will make the system a potential candidate in the development of robust materials for chemotherapeutic applications.


Asunto(s)
Neoplasias , Ácido Tióctico , Ácido Tióctico/química , Antibióticos Antineoplásicos/uso terapéutico , Disulfuros/uso terapéutico , Reproducibilidad de los Resultados , Sistemas de Liberación de Medicamentos , Doxorrubicina/química , Micelas , Concentración de Iones de Hidrógeno , Portadores de Fármacos/química , Neoplasias/tratamiento farmacológico
6.
Saudi J Gastroenterol ; 28(6): 434-440, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35946262

RESUMEN

Background: To investigage the thiol and disulphide levels in Helicobacter pylori-positive patients with non-ulcer dyspepsia and investigate the change in these levels with eradication therapy. Methods: This is a prospective observational study. A total of 320 patients diagnosed with dyspepsia according to Rome IV criteria were included in the study. First, blood samples were drawn from patients to determine their serum thiol and disulphide levels. Endoscopic biopsy was performed on all patients and the biopsy specimens obtained were examined pathologically. Patients positive for H. pylori were administered eradication therapy. Blood samples were drawn from these patients for the second time, and their serum thiol and disulphide levels were measured. The thiol-disulfide levels of the patients who were successful in H. pylori eradication treatment, with those who were not, were compared before and after the treatment. Results: The mean plasma disulphide level decreased significantly from 14.0 ± 6.6 to 10.9 ± 5.9 µmol/L in H. pylori-positive patients that responded to the H. pylori eradication treatment (P = 0.033). On the other hand, there was an insignificant increase in the mean serum thiol level (341.4 ± 30.5 vs. 342.6 ± 29.8 µmol/L; P = 0.273) and an insignificant decrease in the mean serum disulphide level (15.2 ± 2.5 vs. 14.8 ± 2.3 µmol/L; P = 0.163) in H. pylori-positive patients that did not respond to the H. pylori eradication treatment. Conclusion: The inflammation caused by H. pylori shifted the thiol-disulphide equilibrium in the cell redox system towards the direction of disulphide. The study findings suggest that the restoration of the said hemostatic balance with eradication therapy relieved the organism from oxidative stress.


Asunto(s)
Dispepsia , Gastritis , Infecciones por Helicobacter , Helicobacter pylori , Humanos , Dispepsia/tratamiento farmacológico , Infecciones por Helicobacter/complicaciones , Infecciones por Helicobacter/tratamiento farmacológico , Infecciones por Helicobacter/patología , Antibacterianos/uso terapéutico , Gastritis/patología , Estrés Oxidativo , Disulfuros/uso terapéutico , Compuestos de Sulfhidrilo/uso terapéutico , Quimioterapia Combinada
7.
Eur Rev Med Pharmacol Sci ; 26(4): 1283-1292, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-35253185

RESUMEN

OBJECTIVE:   To explore the in vitro and in vivo experimental study of thioredoxin-1(Trx1) inhibitor 1-methylpropyl 2-imidazolyl disulfide (PX-12) promoting multiple myeloma H929 cell apoptosis, investigate the relationship between the inhibitory effect of PX-12 on H929 cells and reactive oxygen species (ROS). MATERIALS AND METHODS: Inhibition of PX-12 on H929 cells in relation to reactive oxygen species (ROS), cell cycle, and apoptosis were assessed by flow cytometry. ELISA kit, IVIS Imaging, Hematoxylin and eosin (H&E) staining and immunohistochemical staining assessment were applied to assess the anti-myeloma effect in the SCID mice model established by H929EL cells. RESULTS: PX-12 inhibited proliferation of H929 cells performed time and dose dependent style. Furthermore, it significantly induced a G2/M phase arrest of the cell cycle in H929 cells. It also increased intracellular ROS and caspase-3 activity in H929 cells indicating that cells have undergone apoptosis. There was an almost 3-5-fold decrease in tumor viability measured by the Living-Imaging system after 21 and 28 days after PX-12 injection compared with the control group. Importantly, PX-12 caused significant decrease in expression of Kappa chain in vivo assessed by immunohistochemical staining. CONCLUSIONS: The results suggest that PX-12 may be a potential strategy for the treatment of MM, and the inhibition of TRX-1 in the treatment of myeloma deserves further research.


Asunto(s)
Mieloma Múltiple , Tiorredoxinas , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular , Disulfuros/farmacología , Disulfuros/uso terapéutico , Imidazoles/farmacología , Imidazoles/uso terapéutico , Ratones , Ratones SCID , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/patología , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo , Tiorredoxinas/metabolismo
8.
Cancer Lett ; 534: 215604, 2022 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-35247515

RESUMEN

Breast cancer mortality remains unacceptably high, indicating a need for safer and more effective therapeutic agents. Disulfide bond Disrupting Agents (DDAs) were previously identified as a novel class of anticancer compounds that selectively kill cancers that overexpress the Epidermal Growth Factor Receptor (EGFR) or its family member HER2. DDAs kill EGFR+ and HER2+ cancer cells via the parallel downregulation of EGFR, HER2, and HER3 and activation/oligomerization of Death Receptors 4 and 5 (DR4/5). However, the mechanisms by which DDAs mediate these effects are unknown. Affinity purification analyses employing biotinylated-DDAs reveal that the Protein Disulfide Isomerase (PDI) family members AGR2, PDIA1, and ERp44 are DDA target proteins. Further analyses demonstrate that shRNA-mediated knockdown of AGR2 and ERp44, or expression of ERp44 mutants, enhance basal DR5 oligomerization. DDA treatment of breast cancer cells disrupts PDIA1 and ERp44 mixed disulfide bonds with their client proteins. Together, the results herein reveal DDAs as the first small molecule, active site inhibitors of AGR2 and ERp44, and demonstrate roles for AGR2 and ERp44 in regulating the activity, stability, and localization of DR4 and DR5, and activation of Caspase 8.


Asunto(s)
Neoplasias de la Mama , Disulfuros , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Muerte Celular , Disulfuros/metabolismo , Disulfuros/uso terapéutico , Receptores ErbB/metabolismo , Femenino , Humanos , Proteínas de la Membrana , Chaperonas Moleculares/metabolismo , Mucoproteínas , Proteínas Oncogénicas/genética , Proteína Disulfuro Isomerasas/genética , Proteína Disulfuro Isomerasas/metabolismo , Proteínas , Receptores de Muerte Celular
9.
Pharmacol Res ; 177: 106118, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35134476

RESUMEN

Phytochemicals have attracted attention in the oncological field because they are biologically friendly and have relevant pharmacological activities. Thanks to the intense and unique spicy aroma, garlic is one of the most used plants for cooking. Its consumption is correlated to health beneficial effects towards several chronic diseases, such as cancer, mainly attributable to allicin, a bioactive sulfur compound stored in different plant parts in a precursor form. The objective of this review is to present and critically discuss the chemistry and biosynthesis of allicin, its pharmacokinetic profile, its anticancer mechanisms and molecular targets, and its selectivity towards tumor cells. The research carried out so far revealed that allicin suppresses the growth of different types of tumors. In particular, it targets many signaling pathways associated with cancer development. Future research directions are also outlined to further characterize this promising natural product.


Asunto(s)
Productos Biológicos , Ajo , Neoplasias , Disulfuros/uso terapéutico , Ajo/química , Humanos , Neoplasias/tratamiento farmacológico , Ácidos Sulfínicos/química , Ácidos Sulfínicos/farmacología , Ácidos Sulfínicos/uso terapéutico
10.
Inflammation ; 45(1): 45-58, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-35061151

RESUMEN

We have previously shown that diallyl disulfide (DADS) protects mice against cerulein-induced acute pancreatitis (AP) and associated lung injury. However, the molecular mechanisms underlying its effect and the components involved have not been studied. We hypothesized that DADS may reduce TNF-α, CSE expression, H2S production, STAT3, and NF-κB activation and induce SOCS3 expression through peroxisome proliferator-activated receptor γ (PPAR-γ) pathway in cerulein-induced mice. Male Swiss mice were treated with hourly intraperitoneal injections of cerulein (50 µg/kg) for 6 h. Diallyl disulfide (200 µg/kg) was administered in the presence or absence of PPAR-γ antagonist GW9662 (0.3 mg/kg) (i.p) 1 h after the induction of AP. Our findings revealed that DADS blocked TNF-α, CSE expression, H2S production, and STAT3, and NF-κB activation was reversed by GW9662. Furthermore, GW9662 abrogated DADS-induced SOCS3 expression. The results show for the first that DADS-induced anti-inflammatory effect in acute pancreatitis is regulated through PPAR-γ.


Asunto(s)
Compuestos Alílicos/farmacología , Antiinflamatorios/farmacología , Disulfuros/farmacología , Lesión Pulmonar/prevención & control , FN-kappa B/metabolismo , PPAR gamma/metabolismo , Pancreatitis/prevención & control , Factor de Transcripción STAT3/metabolismo , Compuestos Alílicos/uso terapéutico , Animales , Antiinflamatorios/uso terapéutico , Ceruletida , Disulfuros/uso terapéutico , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/fisiopatología , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/metabolismo , Lesión Pulmonar/fisiopatología , Masculino , Ratones , Páncreas/efectos de los fármacos , Páncreas/metabolismo , Páncreas/fisiopatología , Pancreatitis/inducido químicamente , Pancreatitis/metabolismo , Pancreatitis/fisiopatología , Distribución Aleatoria , Transducción de Señal/efectos de los fármacos
11.
Mol Biol Rep ; 48(11): 7261-7272, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34626309

RESUMEN

BACKGROUND: The tumor suppressor protein p53 is a most promising target for the development of anticancer drugs. Allicin (diallylthiosulfinate) is one of the most active components of garlic (Alliium sativum L.) and possesses a variety of health-promoting properties with pharmacological applications. However, whether allicin plays an anti-cancer role against breast cancer cells through the induction of p53-mediated apoptosis remains unknown. METHODS AND RESULTS: In this study, we investigate the anti-breast cancer effect of allicin in vitro by using MCF-7 and MD-MBA-231 cells. We found that allicin reduces cell viability, induces apoptosis and cell cycle arrest in both cells. Allicin activated p53 and caspase 3 expressions in both cells but produced different effects on the expression of p53-related biomarkers. In MDA-MB-231 cells, allicin up-regulated the mRNA and protein expression of A1BG and THBS1 while down-regulated the expression of TPM4. Conversely, the mRNA and protein expression of A1BG, THBS1 and TPM4 were all reduced in MCF-7 cells. Hence, allicin induces cell cycle arrest and apoptosis in breast cancer cells through p53 activation but it effects on the expression of p53-related biomarkers were dependent upon the specific type of breast cancer involved. CONCLUSIONS: These findings suggest that allicin induces apoptosis and regulates biomarker expression in breast cancer cell lines through modulating the p53 signaling pathway. Furthermore, our results promote the utility of allicin as compound for further studies as an anticancer drug targeting p53.


Asunto(s)
Apoptosis , Neoplasias de la Mama/tratamiento farmacológico , Puntos de Control del Ciclo Celular , Disulfuros/farmacología , Transducción de Señal , Ácidos Sulfínicos/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Antineoplásicos Fitogénicos/farmacología , Antineoplásicos Fitogénicos/uso terapéutico , Neoplasias de la Mama/fisiopatología , Caspasa 3/genética , Línea Celular Tumoral , Disulfuros/uso terapéutico , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Células MCF-7 , Ácidos Sulfínicos/uso terapéutico , Proteína p53 Supresora de Tumor/genética
13.
ACS Appl Mater Interfaces ; 13(39): 46291-46302, 2021 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-34558902

RESUMEN

The current clinical performance of chemotherapy is far from satisfactory, greatly limited by insufficient delivery efficacy and serious systemic side effects. Dimeric prodrug systems are emerging as valuable strategies for boosting the antitumor outcome. Here, dimeric paclitaxel prodrugs were synthesized with different bridged linkers, and the formed prodrug nanoparticles possessed excellent colloidal stability and ultrahigh drug content. The diselenide bond containing paclitaxel prodrugs could respond to a redox-heterogeneous intracellular microenvironment for on-demand drug release and subsequently show a selective cytotoxicity toward tumor cells against normal cells. Furthermore, the optimal carrier materials were screened out according to their contribution on stability, endocytosis, cytotoxicity, biodistribution, and antitumor efficacy. Compared with DSPE-PEG, human serum albumin, and Fe-tannic acid-based complex, F127 anchored dimeric paclitaxel nanoformulations exhibited preferential tumor accumulation and potent anticancer effect. Our present work provides deep insight into the development of advanced nanoformulations with comprehensive advantages for enhancing cancer therapy.


Asunto(s)
Antineoplásicos Fitogénicos/uso terapéutico , Disulfuros/uso terapéutico , Nanopartículas/uso terapéutico , Neoplasias/tratamiento farmacológico , Paclitaxel/uso terapéutico , Profármacos/uso terapéutico , Animales , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/farmacocinética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Disulfuros/química , Disulfuros/farmacocinética , Liberación de Fármacos , Quimioterapia , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Células 3T3 NIH , Nanopartículas/química , Neoplasias/patología , Oxidación-Reducción , Paclitaxel/análogos & derivados , Paclitaxel/farmacocinética , Profármacos/química , Profármacos/farmacocinética
14.
Molecules ; 26(15)2021 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-34361695

RESUMEN

The active ingredients allicin and curcumin have a wide range of actions against fungi, bacteria, and helminths. Therefore, the study was aimed to evaluate the efficacy of allicin (AL) and curcumin (CU) as antischistosomal drugs and their biochemical effects in normal and Schistosoma mansoni-infected mice. Praziquantel (PZQ) was administrated for two successive days while AL or CU was given for two weeks from the week 7th postinfection (PI). The possible effect of different regimens on Schistosoma worms was evaluated by measuring the percentage of the recovered worms, tissue egg load, and oogram pattern. Serum alanine transaminase activity and levels of triglycerides, cholesterol, and uric acid were measured. Liver tissue malondialdehyde and reduced glutathione levels besides, the activities of glutathione-S-transferase, superoxide dismutase and catalase were assessed for the oxidative/antioxidant condition. DNA electrophoresis of liver tissue was used to indicate the degree of fragmentation. There was a significant reduction in the recovered worms and egg load, with a marked change of oogram pattern in all treated groups with PZQ, AL, and CU in comparison with infected-untreated mice. PZQ, AL, and CU prevented most of the hematological and biochemical disorders, as well as significantly improved the antioxidant capacity and enhanced DNA fragmentation in the liver tissue of schistosomiasis mice compared to the infected-untreated group. These promising results suggest that AL and CU are efficient as antischistosomal drugs, and it would be beneficial to test their combination to understand the mechanism of action and the proper period of treatment leading to the best result.


Asunto(s)
Antioxidantes/uso terapéutico , Curcuma/química , Curcumina/uso terapéutico , Disulfuros/uso terapéutico , Ajo/química , Fitoterapia/métodos , Extractos Vegetales/uso terapéutico , Schistosoma mansoni/efectos de los fármacos , Esquistosomiasis mansoni/tratamiento farmacológico , Esquistosomicidas/uso terapéutico , Ácidos Sulfínicos/uso terapéutico , Animales , Fragmentación del ADN/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Recuento de Huevos de Parásitos , Praziquantel/uso terapéutico , Esquistosomiasis mansoni/parasitología , Resultado del Tratamiento
15.
Angew Chem Int Ed Engl ; 60(43): 23299-23305, 2021 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-34240523

RESUMEN

Development of proteolysis targeting chimeras (PROTACs) is emerging as a promising strategy for targeted protein degradation. However, the drug development using the heterobifunctional PROTAC molecules is generally limited by poor membrane permeability, low in vivo efficacy and indiscriminate distribution. Herein an aptamer-PROTAC conjugation approach was developed as a novel strategy to improve the tumor-specific targeting ability and in vivo antitumor potency of conventional PROTACs. As proof of concept, the first aptamer-PROTAC conjugate (APC) was designed by conjugating a BET-targeting PROTAC to the nucleic acid aptamer AS1411 (AS) via a cleavable linker. Compared with the unmodified BET PROTAC, the designed molecule (APR) showed improved tumor targeting ability in a MCF-7 xenograft model, leading to enhanced in vivo BET degradation and antitumor potency and decreased toxicity. Thus, the APC strategy may pave the way for the design of tumor-specific targeting PROTACs and have broad applications in the development of PROTAC-based drugs.


Asunto(s)
Antineoplásicos/uso terapéutico , Aptámeros de Nucleótidos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Oligodesoxirribonucleótidos/uso terapéutico , Proteolisis/efectos de los fármacos , Animales , Antineoplásicos/síntesis química , Antineoplásicos/toxicidad , Aptámeros de Nucleótidos/síntesis química , Aptámeros de Nucleótidos/toxicidad , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Disulfuros/síntesis química , Disulfuros/uso terapéutico , Disulfuros/toxicidad , Compuestos Heterocíclicos con 3 Anillos/síntesis química , Compuestos Heterocíclicos con 3 Anillos/uso terapéutico , Compuestos Heterocíclicos con 3 Anillos/toxicidad , Humanos , Ratones , Oligodesoxirribonucleótidos/síntesis química , Oligodesoxirribonucleótidos/toxicidad , Prueba de Estudio Conceptual , Pirrolidinas/síntesis química , Pirrolidinas/uso terapéutico , Pirrolidinas/toxicidad , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
16.
J Food Biochem ; 45(6): e13765, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33997994

RESUMEN

Methotrexate (MTX) is a promising chemotherapeutic agent. Its medical use is limited by induced nephropathy. Our study was designed to explore the reno-protective effect of diallyl disulfide (DADS), an organosulfur compound of garlic oil, on MTX-induced nephropathy. Adult rats were randomly divided into 4 groups; normal control, DADS (50 mg kg-1  day-1 , p.o.), MTX (20 mg/kg, i.p.) and DADS+MTX. DADS significantly decreased serum creatinine, urea, uric acid, and albumin levels with an improvement of final body weight. Additionally, DADS markedly attenuated MTX-induced elevations in renal MDA and NO2- contents with an increase in GSH content and SOD activity. Mechanistically, DADS effectively down-regulated mRNA expression level of renal p38 and NF-κB. Additionally, DADS positively regulated the NRF2 gene with a remarkable inhibition of Keap-1 gene. Furthermore, DADS up-regulated BCL2 protein and remarkably suppressed the expression of both BAX and caspase-3 proteins. Overall, DADS has favorable renal protection against MTX-induced nephropathy via modulation of Keap-1/NRF2, p38/NF-κB, and BCL2/BAX/caspase-3 signaling. PRACTICAL APPLICATIONS: Diallyl disulfide is one of the organosulfur compounds of garlic oil. Our study demonstrated that DADS substantially alleviated the decline of kidney function and renal injury induced by MTX. The antioxidative, anti-inflammatory, and anti-apoptotic properties may constitute an important part of its therapeutic applications via regulation of p38/NF-κB, Keap-1/NRF2, and BCL2/BAX/caspase-3 signaling pathways. Therefore, DADS could be a potential therapeutic adjunct in cancer chemotherapy to decrease the associated side effects of MTX. It should be further explored clinically as a protective agent for MTX-treated cancer patients.


Asunto(s)
Metotrexato , Factor 2 Relacionado con NF-E2 , Compuestos Alílicos , Animales , Disulfuros/farmacología , Disulfuros/uso terapéutico , Humanos , Metotrexato/toxicidad , FN-kappa B/genética , Ratas
17.
Curr Pain Headache Rep ; 25(5): 29, 2021 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-33761014

RESUMEN

PURPOSE OF REVIEW: Dual enkephalinase inhibitors (DENKIs) are pain medications that indirectly activate opioid receptors and can be used as an alternative to traditional opioids. Understanding the physiology of enkephalins and their inhibitors and the pharmacology of these drugs will allow for proper clinical application for chronic pain patients in the future. RECENT FINDINGS: DENKIs can be used as an alternative mode of analgesia for patients suffering from chronic pain by preventing the degradation of endogenous opioid ligands. By inhibiting the two major enkephalin-degrading enzymes (neprilysin and aminopeptidase N), DENKIs can provide analgesia with less adverse effects than nonendogenous opioids. The purpose of this paper is to review the current literature investigating DENKIs and explore their contribution to chronic pain management.


Asunto(s)
Antígenos CD13/antagonistas & inhibidores , Dolor Crónico/tratamiento farmacológico , Disulfuros/uso terapéutico , Encefalinas/metabolismo , Inhibidores Enzimáticos/uso terapéutico , Neprilisina/antagonistas & inhibidores , Propionatos/uso terapéutico , Propilaminas/uso terapéutico , Dipéptidos/uso terapéutico , Humanos , Fenilalanina/análogos & derivados , Fenilalanina/uso terapéutico
18.
J Nanobiotechnology ; 19(1): 36, 2021 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-33536031

RESUMEN

Owing to the hypoxia status of the tumor, the reactive oxygen species (ROS) production during photodynamic therapy (PDT) of the tumor is less efficient. Herein, a facile method which involves the synthesis of Mg-Mn-Al layered double hydroxides (LDH) clay with MoS2 doping in the surface and anionic layer space of LDH was presented, to integrate the photo-thermal effect of MoS2 and imaging and catalytic functions of Mg-Mn-Al LDH. The designed LDH-MoS2 (LMM) clay composite was further surface-coated with bovine serum albumin (BSA) to maintain the colloidal stability of LMM in physiological environment. A photosensitizer, chlorin e6 (Ce6), was absorbed at the surface and anionic layer space of LMM@BSA. In the LMM formulation, the magnetic resonance imaging of Mg-Mn-Al LDH was enhanced thanks to the reduced and acid microenvironment of the tumor. Notably, the ROS production and PDT efficiency of Ce6 were significantly improved, because LMM@BSA could catalyze the decomposing of the overexpressed H2O2 in tumors to produce oxygen. The biocompatible LMM@BSA that played the synergism with tumor microenvironment is a promising candidate for the effective treatment of cancer.


Asunto(s)
Catalasa/uso terapéutico , Disulfuros/uso terapéutico , Molibdeno/uso terapéutico , Nanoestructuras/uso terapéutico , Neoplasias/terapia , Fármacos Fotosensibilizantes/uso terapéutico , Porfirinas/uso terapéutico , Animales , Materiales Biomiméticos/síntesis química , Materiales Biomiméticos/uso terapéutico , Clorofilidas , Células HT29 , Humanos , Hidróxidos/uso terapéutico , Imagen por Resonancia Magnética/métodos , Ratones , Neoplasias/diagnóstico por imagen , Neoplasias/metabolismo , Fotoquimioterapia/métodos , Terapia Fototérmica/métodos , Especies Reactivas de Oxígeno/metabolismo , Nanomedicina Teranóstica/métodos
19.
Future Oncol ; 17(8): 877-892, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33251877

RESUMEN

Immune checkpoint inhibitors have demonstrated significant clinical activity across various tumor subtypes; however, their utility in gynecologic malignancies has thus far proven modest. Since the identification of a molecular subclassification system for endometrial cancer (EC), research in immune checkpoint inhibitor therapies has been focusing on certain subgroups predictive for response, particularly microsatellite instability hypermutated/DNA mismatch repair-deficient subtype. Dostarlimab, a PD-1 inhibitor, has demonstrated preliminary evidence of clinical activity and acceptable safety profile in patients with across recurrent EC, particularly microsatellite instability-hypermutated/DNA mismatch repair-deficient EC. This review outlines existing data for the efficacy and safety of dostarlimab in recurrent or advanced-stage EC.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Neoplasias Endometriales/tratamiento farmacológico , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Anticuerpos Monoclonales Humanizados/farmacología , Ensayos Clínicos como Asunto , Reparación de la Incompatibilidad de ADN , Supervivencia sin Enfermedad , Disulfuros/farmacología , Disulfuros/uso terapéutico , Neoplasias Endometriales/diagnóstico , Neoplasias Endometriales/genética , Neoplasias Endometriales/mortalidad , Femenino , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inestabilidad de Microsatélites , Recurrencia Local de Neoplasia/epidemiología , Recurrencia Local de Neoplasia/genética
20.
Cancer Biol Ther ; 21(8): 698-708, 2020 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-32489150

RESUMEN

Non-small cell lung cancer (NSCLC) remains recalcitrant to effective treatment due to tumor relapse and acquired resistance. Cancer stem cells (CSCs) are believed to be one mechanism for relapse and resistance and are consequently considered promising drug targets. We report that chetomin, an active component of Chaetomium globosum, blocks heat shock protein 90/hypoxia-inducible factor 1 alpha (Hsp90/HIF1α) pathway activity. Chetomin also attenuated sphere-forming, a stem cell-like characteristic, of NSCLC CSCs (at ~ nM range) and the proliferation of non-CSCs NSCLC cultures and chemoresistant sublines (at ~ µM range). At these concentrations, chetomin exerted a marginal influence on noncancerous cells originating from several organs. Chetomin markedly decreased in vivo tumor formation in a spontaneous KrasLA1 lung cancer model, flank xenograft models, and a tumor propagation flank implanted model at doses that did not produce an observable toxicity to the animals. Chetomin blocked Hsp90/HIF1α pathway activity via inhibiting the Hsp90-HIF1α binding interaction without affecting Hsp90 or Hsp70 protein levels. This study advocates chetomin as a Hsp90/HIF1α pathway inhibitor and a potent, nontoxic NSCLC CSC-targeting molecule.


Asunto(s)
Disulfuros/uso terapéutico , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Alcaloides Indólicos/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Micotoxinas/uso terapéutico , Células Madre Neoplásicas/efectos de los fármacos , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Disulfuros/farmacología , Humanos , Alcaloides Indólicos/farmacología , Ratones , Micotoxinas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA