Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
ACS Biomater Sci Eng ; 2024 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-39285678

RESUMEN

Multi drug resistance (MDR) in breast carcinoma still poses a significant impairment to successful chemotherapy. As the arsenal of anticancer agents increases with improved preclinical methods, the growth of therapeutic drug combinations is now unprecedented. The malignancies addressed by mono drugs often fail to limit cancer progression, resulting in resistant cancer, thereby offering combinatorial therapies a terrific edge over monodrug regimes. However, the selection of drug combinations required enough preliminary evidence for their synergistic effect. The fundamental mechanisms of MDR to chemotherapeutics are associated with the overexpression of membrane efflux pumps, alternations in drug targets, and increased drug metabolism. Unfortunately, it is very difficult for drugs to overcome resistance produced on their own or by another different drug action. In this context, herein, we report a simple delivery system for coencapsulation and intracellular codelivery of dual-drug thymoquinone (TQ) and doxorubicin (DOX) to resensitize DOX-resistant MDA MB231 cell line (231 R). The 231 R cell line developed in our lab showed an enhanced expression of the ATP-binding cassette (ABC) transporters P-gp1/MDR-1 and a declined miR-298 expression. The present delivery system is based on amine-functionalized mesoporous silica nanoparticles (MSNs), in which the side chain amine functional group was used to react with the carbonyl group of TQ, which acts as a pro-drug system (TQ-MSN) to release TQ and DOX simultaneously. DOX was encapsulated later into the above TQ-MSN by a simple diffusion method. The drugs containing MSNs were further coated with a hyaluronic acid-conjugated PEG-PLGA polymer (HA@TQ-DOX-MSN). This simple nanostrategy interferes with the MDR-1/miR-298 cross-talk, thereby allowing a significant reduction in drug efflux from the cell and highlighting a promising nanotechnology-based combinatorial delivery approach in managing breast cancer chemoresistance.

2.
Biofactors ; 2024 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-39096306

RESUMEN

Groundwater arsenic is a notorious toxicant and exposure to environmentally relevant concentrations persists as a healthcare burden across the world. Arsenic has been reported to jeopardize the normal functioning of the immune system, but there are still gaps in the understanding of thymic T cell biology. Immunotoxic influence of arsenic in thymic integrity demands a potent restorative molecule. The objectives of this study were to examine key signaling cross-talks associated with arsenic-induced immune alterations in the thymus and propose melatonin as a potential candidate against immunological complications arising from arsenic exposure. Swiss albino mice were exposed to sodium arsenite (0.05 mg/L; in drinking water) and melatonin (IP:10 mg/kg BW) for 28 days. Melatonin successfully protected thymus from arsenic-mediated tissue degeneration and maintained immune homeostasis including T cell maturation and proliferation by mitigating oxidative stress through Nrf2 upregulation. Additionally, melatonin exerted ameliorative effect against arsenic-induced apoptosis and inflammation by inhibiting p53-mediated mitochondrial cell death pathway and NF-κB-p65/STAT3-mediated proinflammatory pathway, respectively. For the first time, we showed that arsenic-induced profibrotic changes were inhibited by melatonin through targeting of inflammation-associated EMT. Our findings clearly demonstrate that melatonin can be a viable and promising candidate in combating arsenic-induced immune toxicity with no collateral damage, making it an important research target.

3.
Biochim Biophys Acta Mol Basis Dis ; 1870(7): 167313, 2024 10.
Artículo en Inglés | MEDLINE | ID: mdl-38901652

RESUMEN

BACKGROUND: Chronic pancreatic dysfunction is frequently observed as a consequence of prolonged high-fat diet consumption and is a serious public health concern. This pro-diabetic insult aggravates inflammation-influenced fibrotic lesions and is associated with deregulated autophagy. Metformin, a conventional anti-hyperglycemic drug, might be beneficial for pancreatic health, but the complex molecular regulations are not clarified. Considering the worldwide prevalence of chronic pancreatic dysfunction in obese individuals, we aimed to unwind the molecular intricacies explaining the involvement of oxidative stress, inflammation and fibrosis and to approbate metformin as a plausible intervention in this crossroad. MAIN METHODS: Age-matched Swiss Albino mice were exposed to high-fat diet (60 kcal%) against control diet (10 kcal%) to establish diet-induced stress model. Metformin treatment was introduced after 4 weeks to metformin-control and HFD-exposed metformin groups. After 8 weeks, metabolic and molecular outcomes were assessed to establish the impact of metformin on chronic consequences of HFD-mediated injury. KEY FINDINGS: High-fat diet administration to healthy mice primes oxidative stress-mediated chronic inflammation through Nrf2/Keap1/NF-κB interplay. Besides, pro-inflammatory cytokine bias leading to fibrotic (increased TGF-ß, α-SMA, and MMP9) and pro-EMT (Twist1, Slug, Vimentin, E-cadherin) repercussions in pancreatic lobules were evident. Metformin distinctly rescues high-fat diet-induced remodeling of pancreatic pro-diabetic alterations and cellular survival/death switch. Further, metformin abrogates the p62-Twist1 crosstalk in an autophagy-dependent manner (elevated beclin1, LC3-II/I, Lamp2) to restore pancreatic homeostasis. CONCLUSION: Our research validates the therapeutic potential of metformin in the inflammation-fibrosis nexus to ameliorate high-fat diet-induced pancreatic dysfunction and related metabolic alterations.


Asunto(s)
Autofagia , Dieta Alta en Grasa , Fibrosis , Metformina , Estrés Oxidativo , Animales , Metformina/farmacología , Dieta Alta en Grasa/efectos adversos , Autofagia/efectos de los fármacos , Ratones , Estrés Oxidativo/efectos de los fármacos , Masculino , Transición Epitelial-Mesenquimal/efectos de los fármacos , Inflamación/patología , Inflamación/metabolismo , Inflamación/tratamiento farmacológico , Páncreas/patología , Páncreas/efectos de los fármacos , Páncreas/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo
4.
Heliyon ; 10(1): e23870, 2024 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-38226217

RESUMEN

Epidemiological as well as experimental studies have established that the pineal hormone melatonin has inhibitory effects on different types of cancers. Several mechanisms have been proposed for the anticancer activities of melatonin, but the fundamental molecular pathways still require clarity. We developed a mouse model of breast cancer using Ehrlich's ascites carcinoma (injected in the 4th mammary fat pad of female Swiss albino mice) and investigated the possibility of targeting the autophagy-inflammation-EMT colloquy to restrict breast tumor progression using melatonin as intervention. Contrary to its conventional antioxidant role, melatonin was shown to augment intracellular ROS and initiate ROS-dependent apoptosis in our system, by modulating the p53/JNK & NF-κB/pJNK expressions/interactions. Melatonin-induced ROS promoted SIRT1 activity. Interplay between SIRT1 and NF-κB/p65 is known to play a pivotal role in regulating the crosstalk between autophagy and inflammation. Persistent inflammation in the tumor microenvironment and subsequent activation of the IL-6/STAT3/NF-κB feedback loop promoted EMT and suppression of autophagy through activation of PI3K/Akt/mTOR signaling pathway. Melatonin disrupted NF-κB/SIRT1 interactions blocking IL-6/STAT3/NF-κB pathway. This led to reversal of pro-inflammatory bias in the breast tumor microenvironment and augmented autophagic responses. The interactions between p62/Twist1, NF-κB/Beclin1 and NF-κB/Slug were altered by melatonin to strike a balance between autophagy, inflammation and EMT, leading to tumor regression. This study provides critical insights into how melatonin could be utilized in treating breast cancer via inhibition of the PI3K/Akt/mTOR signaling and differential modulation of SIRT1 and NF-κB proteins, leading to the establishment of apoptotic and autophagic fates in breast cancer cells.

5.
Apoptosis ; 29(1-2): 191-209, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37945815

RESUMEN

During cancer cell invasion, integrin undergoes constant endo/exocytic trafficking. It has been found that the recycling ability of integrin ß1 through Rab11-controlled long loop pathways is directly associated with cancer invasion. Previous studies showed that gain-of-function mutant p53 regulates the Rab-coupling protein [RCP]-mediated integrin ß1 recycling by inactivating tumor suppressor TAp63. So, we were interested to investigate the involvement of miR-205 in this process. In the current study first, we evaluated that the lower expression of miR-205 in MDA-MB-231 cell line is associated with high motility and invasiveness. Further investigation corroborated that miR-205 directly targets RCP resulting in attenuated RCP-mediated integrin ß1 recycling. Overexpression of TAp63 validates our in vitro findings. To appraise the anti-metastatic role of miR-205, we developed two in vivo experimental models- xenograft-chick embryo and xenograft-immunosuppressed BALB/c mice. Our in vivo results support the negative effect of miR-205 on metastasis. Therefore, these findings advocate the tumor suppressor activity of miR-205 in breast cancer cells and suggest that in the future development of miR-205-targeting RNAi therapeutics could be a smart alternative approach to prevent the metastatic fate of the disease.


Asunto(s)
Neoplasias de la Mama , MicroARNs , Animales , Embrión de Pollo , Femenino , Humanos , Ratones , Apoptosis , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular/genética , Integrina beta1/genética , Integrina beta1/metabolismo , MicroARNs/genética , Invasividad Neoplásica , Metástasis de la Neoplasia
6.
Fitoterapia ; 169: 105601, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37406886

RESUMEN

Cancer continues to threat mortal alongside scientific community with burgeoning grasp. Most efforts directed to tame Cancer such as radiotherapy or chemotherapy, all came at a cost of severe side effects. The plant derived bioactive compounds on the other hand carries an inevitable advantage of being safer, bioavailable & less toxic compared to contemporary chemotherapeutics. Our strategic approach employed solvent extraction of Black Seed Oil (BSO) to highlight the orchestrated use of its oil soluble phytochemicals - Thymoquinone, Carvacrol & Trans-Anethole when used in cohort. These anti-cancer agents in unbelievably modest amounts present in BSO shows better potential to delineate migratory properties in breast cancer cells as compared to when treated individually. BSO was also observed to have apoptotic calibre when investigated in MDA-MB-231 and MCF-7 cell lines. We performed chemical characterization of the individual phytochemical as well as the oil in-whole to demonstrate the bioactive oil-soluble entities present in whole extract. BSO was observed to have significant anti-cancerous properties in cumulative proportion that is reportedly higher than the individual three components. Besides, this study also reports micro-RNA regulation on BSO administration, thereby playing a pivotal role in breast cancer alleviation. Thus, synergistic action of the integrants serves better combat force against breast cancer in the form of whole extract, hence aiming at a more lucrative paradigm while significantly regulating microRNAs associated with breast cancer migration and apoptosis.


Asunto(s)
Neoplasias de la Mama , MicroARNs , Nigella sativa , Humanos , Femenino , Neoplasias de la Mama/tratamiento farmacológico , Nigella sativa/química , Estructura Molecular , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico
7.
Int Immunopharmacol ; 120: 110301, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37224648

RESUMEN

AIMS: Circumstantial anxiety as well as chronic stress may stimulate the release of stress hormones including catecholamines. Adrenaline toxicity has been implicated in many cardiovascular conditions. Considering previous literature that suggests the oxidative potential of the adrenaline-copper entity, we have investigated its potential nocuous role in isolated adult rat cardiomyocytes, the underlying molecular mechanism, and its possible protection by melatonin. MAIN METHODS: Given the mechanistic congruity of adrenaline-copper (AC) with the well-established H2O2-copper-ascorbate (HCA) system of free radical generation, we have used the latter as a representative model to study the cytotoxic nature of AC. We further investigated the cardioprotective efficacy of melatonin in both the stress models through scanning electron microscopy, immunofluorescence, flow cytometry, and western blot analysis. KEY FINDINGS: Results show that melatonin significantly protects AC-treated cardiomyocytes from ROS-mediated membrane damage, disruption of mitochondrial membrane potential, antioxidant imbalance, and distortion of cellular morphology. Melatonin protects cardiomyocytes from inflammation by downregulating pro-inflammatory mediators viz., COX-2, NF-κB, TNF-α, and upregulating anti-inflammatory IL-10. Melatonin significantly ameliorated cardiomyocyte apoptosis in AC and HCA-treated cells as evidenced by decreased BAX/BCL-2 ratio and subsequent suppression of caspase-9 and caspase-3 levels. The isothermal calorimetric study revealed that melatonin inhibits the binding of adrenaline bitartrate with copper in solution, which fairly explains the rescue potential of melatonin against AC-mediated toxicity in cardiomyocytes. SIGNIFICANCE: Findings suggest that the multipronged strategy of melatonin that includes its antioxidant, anti-inflammatory, anti-apoptotic, and overall cardioprotective ability may substantiate its potential therapeutic efficacy against adrenaline-copper-induced damage and death of adult rat cardiomyocytes.


Asunto(s)
Melatonina , Ratas , Animales , Melatonina/farmacología , Melatonina/metabolismo , Antioxidantes/farmacología , Antioxidantes/metabolismo , Cobre/toxicidad , Cobre/metabolismo , Miocitos Cardíacos/metabolismo , Peróxido de Hidrógeno/metabolismo , Estrés Oxidativo , Apoptosis , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Epinefrina/metabolismo
8.
J Nutr Biochem ; 115: 109283, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36791995

RESUMEN

One of the key biochemical features that distinguish a cancer cell from normal cells is its persistent pro-oxidative state that leads to intrinsic oxidative stress. Malignant cells have evolved sophisticated adaptation systems that involve high dependency on antioxidant functions and upregulation of pro-survival molecules to counteract the deleterious effects of reactive species and to maintain dynamic redox balance. This situation renders them vulnerable to further oxidative challenges by exogenous agents. In the present study, we advocated that pomegranate polyphenols act as pro-oxidants and trigger ROS-mediated apoptosis in cancer cells. With the help of both in vitro and in vivo models, we have established that pomegranate fruit extract (PFE) can cause a significant reduction in tumor proliferation while leaving normal tissues and cells unharmed. Administration of PFE (0.2% v/v) in Erhlich's ascites carcinoma-bearing mice for 3 weeks, inhibited the nuclear factor (erythroid-derived 2)-like 2-antioxidant response element signaling cascade, increased intracellular reactive oxygen species content, altered glutathione cycle thereby activating reactive oxygen species-induced apoptotic pathway in Erhlich's ascites carcinoma cells. Moreover, PFE mitigated epithelial to mesenchymal transition and migration in triple negative breast cancer cells (MDA-MB 231 cells) by down-regulating nuclear factor kappa light-chain-enhancer of activated B cells. Pre-treatment of tumor cells with N-acetyl cysteine protected these cells from undergoing PFE-induced apoptosis while siRNA-mediated silencing of Nuclear factor (erythroid-derived 2)-like 2 and nuclear factor kappa light-chain-enhancer of activated B cells in tumor cells increased the cytotoxic potential and pro-oxidative activity of PFE, indicating a clear role of these transcription factors in orchestrating the anticancer/pro-oxidative properties of PFE. The seminal findings provided may be exploited to develop potential therapeutic targets for selective killing of malignant cells.


Asunto(s)
Carcinoma , Granada (Fruta) , Animales , Ratones , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Antioxidantes/química , Especies Reactivas de Oxígeno/metabolismo , Frutas/química , Ascitis , Polifenoles/farmacología , Polifenoles/análisis , Transición Epitelial-Mesenquimal , Estrés Oxidativo , Factor 2 Relacionado con NF-E2/metabolismo , FN-kappa B/metabolismo , Apoptosis
9.
Cell Signal ; 101: 110521, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36375715

RESUMEN

The unending lifestyle stressors along with genetic predisposition, environmental factors and infections have pushed the immune system into a state of constant activity, leading to unresolved inflammation and increased vulnerability to chronic diseases. Liver fibrosis, an early-stage liver condition that increases the risk of developing liver diseases like cirrhosis and hepatocellular carcinoma, is among the various diseases linked to inflammation that dominate worldwide morbidity and mortality. We developed a mouse model with low-grade lipopolysaccharide (LPS) exposure that shows hepatic damage and a pro-inflammatory condition in the liver. We show that inflammation and oxidative changes increase autophagy in liver cells, a degradation process critical in maintaining cellular homeostasis. Our findings from in vivo and in vitro studies also show that induction of both inflammation and autophagy trigger epithelial-mesenchymal transition (EMT) and pro-fibrotic changes in hepatocytes. Inhibiting the inflammatory pathways with a naturally occurring NF-κB inhibitor and antioxidant, melatonin, could assuage the changes in autophagy and activation of EMT/fibrotic pathways in hepatocytes. Taken together, this study shows a pathway linking inflammation and autophagy which could be targeted for future drug development to delay the progression of liver fibrosis.


Asunto(s)
Neoplasias Hepáticas , Melatonina , Ratones , Animales , Transición Epitelial-Mesenquimal/genética , Melatonina/farmacología , Melatonina/metabolismo , Hepatocitos/metabolismo , Cirrosis Hepática/metabolismo , Hígado/metabolismo , Autofagia , Inflamación/metabolismo , Neoplasias Hepáticas/patología
10.
Life Sci ; 314: 121290, 2023 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-36549349

RESUMEN

AIM: Arsenic contamination in drinking water is a world-wide public health concern. Sustained arsenic ingestion leads to immune alterations and subsequent development of inflammatory and autoimmune diseases; however, the underlying cellular and molecular intricacies of immunotoxicity remains uncharacterized. We aim to understand how exposure to arsenic at different concentrations affects the immune system differentially and whether arsenic-induced differential inflammation dictates altered T-regulatory cell bias and emphasize the role of autophagy in the pathway. MAIN METHODS: Swiss albino mice were exposed to environmentally relevant concentrations of arsenic in drinking water for 28 days. Examination of thymic cyto-architecture was done to evaluate thymic damage. ELISA was performed for key cytokines. Flow cytometry, western blotting, and immunostaining were performed for cell surface and intracellular proteins. Co-immunoprecipitation and transfection with siRNA were performed to examine the direct physical interactions between proteins. KEY FINDINGS: Our study distinctly demonstrates that arsenic-induced oxidative stress instigates NF-κB activation, which not only provokes pro-inflammatory responses, but also exhibits immune-suppressive activity depending on the dose of arsenic. Co-immunoprecipitation of NF-κBp65 and pSTAT-3 reveals that arsenic alters their physical interaction, thereby suppressing IL-6/STAT-3/IL-17A feedback loop. Flow cytometry and silencing studies demonstrate that NF-κB-driven Treg cell differentiation induces immune-suppression through FoxP3 up-regulation at the highest dose of arsenic and such immune-suppression is actively supported by NF-κB-driven autophagy activation. SIGNIFICANCE: Collectively, our findings reveal that exposure to arsenic differentially impacts the immune system and understanding the molecular cascade might provide direction for prevention/treatment of arsenic-induced inflammatory and autoimmune diseases.


Asunto(s)
Arsénico , Enfermedades Autoinmunes , Agua Potable , Animales , Ratones , FN-kappa B/metabolismo , Arsénico/toxicidad , Linfocitos T Reguladores/metabolismo , Autofagia
11.
Biomater Sci ; 9(24): 8285-8312, 2021 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-34766965

RESUMEN

Epidermal growth factor receptor (EGFR) normally over-expresses in non-small cell lung cancer (NSCLC) cells. Its mutations act as oncogenic drivers in the cellular signal transduction pathway, and induce the downstream activation of numerous key cellular events involved in cellular proliferation and survival. EGFR tyrosine kinase inhibitors (EGFR-TK inhibitors), such as gefitinib and erlotinib, have been used for a long time in the treatment of NSCLC. However, they fail to overcome the EGFR-TK mutation due to the acquisition of drug resistance. It is strongly believed that the epithelial-to-mesenchymal transition (EMT) is a key player for acquired resistance and consequent limitation of the clinical efficiency of EGFR-TKIs. Therefore, a new strategy needs to be developed to overcome the resistance in NSCLC. In this current study, we have disclosed for the first time the efficiency of transferrin-modified PLGA-thymoquinone-nanoparticles in combination with gefitinib (NP-dual-1, NP-dual-2 and NP-dual-3) towards gefitinib-resistant A549 cells. The gefitinib-resistant A549 cells (A549/GR) showed 12.3-fold more resistance to gefitinib in comparison to non-resistant A549 cells. The phenotypic alteration resembling spindle-cell shape and increased pseudopodia integuments featured the EMT phenomena in A549/GR cells. EMT in A549/GR was later coupled with the loss of Ecad and expansion of Ncad, along with upregulated vimentin expression, as compared to the control A549 cells. Moreover, the invasive nature and migration potential are more amplified in A549/GR cells. Pre-incubation of A549 cells with TGFß1 also initiated EMT, leading to drug resistance. Conversely, treatment of A549 or A549/GR cells with NP-dual-3 effectively retrieved the sensitivity to gefitinib, restricted the EMT phenomenon, and impaired the TGFß1-induced EMT. On unveiling the underlying mechanism of therapeutic action, we found that STAT3 and miR-21 were individually overexpressed in the A549/GR cells by transfection, and followed by treatment with NP-dual-3. Simultaneously, NP-dual-3 fragmented HIF1-α induced EMT in A549/GR cells and reduced the CSCs markers, viz., Oct-4, Sox-2, Nanog, and Aldh1. These data are self-sufficient to suggest that NP-dual-3 re-sensitizes the drug-resistant A549/GR cells to gefitinib, possibly by retrieving MET phenomena via modulation of STAT3/mir-21/Akt/PTEN/HIF1-α axis. Thus, TQ nanoparticles combined with TKI gefitinib may provide an effective platform to treat NSCLC.


Asunto(s)
Antineoplásicos , Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Nanopartículas , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Benzoquinonas , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Línea Celular Tumoral , Proliferación Celular , Resistencia a Antineoplásicos , Gefitinib/farmacología , Gefitinib/uso terapéutico , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Transferrina
12.
Toxicol Rep ; 8: 1428-1436, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34354930

RESUMEN

Several precedents have confirmed numerous infirmities caused by arsenic poisoning, including immune suppression and cancer. Exposure to arsenic leads to alterations of the cellular machinery and eventually cell death, depending on the dose and duration of exposure. Oxidative stress induced by arsenic is the major mechanism by which it inflicts cellular toxicity, challenging the survival-support - autophagy and culminating in apoptosis in the thymus and spleen of mice. Curcumin, a potent dietary anti-oxidant with known anti-apoptotic and anti-inflammatory properties, was assessed for therapeutic benefits. However, the major caveat of this polyphenol is its low water solubility and limited bioavailability. Therefore, Self Nano-Emulsifying Curcumin (SNEC30) was used to treat mice exposed to arsenic. When administered, SNEC30 effectively ameliorated the adverse effects of arsenic in mice, by restoring structural alterations and reducing ROS-mediated cell death, thereby endorsing the importance of nutraceuticals in counteracting heavy metal-induced cellular toxicity.

13.
Biomater Sci ; 9(16): 5665-5690, 2021 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-34259681

RESUMEN

To date, most of the accessible therapeutic options are virtually non-responsive towards triple-negative breast cancer (TNBC) due to its highly aggressive and metastatic nature. Interestingly, chemotherapy reacts soundly in many TNBC cases compared to other types of breast cancer. However, the side effects of many chemotherapeutic agents are still under cross-examination, and thus prohibit their extensive uses. In this present study, we have developed a series of coumarin-dihydropyrimidinone conjugates (CDHPs) and subsequently their poly(lactic-co-glycolic acid) (PLGA)-PEG4000 mixed copolymer nanoparticles as excellent chemotherapeutic nanomedicine to control TNBC. Among all the synthesized CDHPs, CDHP-4 (prepared by the combination of EDCO with 3,4-difluorobenzaldehyde) showed excellent therapeutic effect on a wide variety of cancer cell lines, including TNBC. Besides, it can control the metastasis and stemness property of TNBC. Furthermore, the nano-encapsulation of CDHP-4 in a mixed polymer nanoparticle system (CDHP-4@PP-NPs) and simultaneous delivery showed much improved therapeutic efficacy at a much lower dose, and almost negligible side effects in normal healthy cells or organs. The effectiveness of the present therapeutic agent was observed both in intravenous and oral mode of administration in in vivo experiments. Moreover, on elucidating the molecular mechanism, we found that CDHP-4@PP-NPs could exhibit apoptotic, anti-migratory, as well as anti-stemness activity against TNBC cell lines through the downregulation of miR-138. We validated our findings in MDA-MB-231 xenograft chick embryos, as well as in 4T1-induced mammary tumor-bearing BALB/c mice models, and studied the bio-distribution of CDHP-4@PP-NPs on the basis of the photoluminescence property of nanoparticles. Our recent study, hence for the first time, unravels the synthesis of CDHP-4@PP-NPs and the molecular mechanism behind the anti-migration, anti-stemness and anti-tumor efficacy of the nanoparticles against the TNBC cells through the miR-138/p65/TUSC2 axis.


Asunto(s)
Cumarinas , Nanopartículas , Neoplasias de la Mama Triple Negativas , Animales , Línea Celular Tumoral , Embrión de Pollo , Humanos , Ratones , Ratones Endogámicos BALB C , MicroARNs , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Proteínas Supresoras de Tumor
14.
J Nutr Biochem ; 97: 108812, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34224820

RESUMEN

It has been widely reported that cancer, along with its treatment regimens, cause severe toxicity in the host. A suitable agent having chemopreventive properties as well as capabilities of ameliorating tumor- and drug-induced toxicities is of imminent need. Pomegranate has been projected as an excellent anti-tumor, anti-inflammatory and anti-oxidant agent. In this study, for the first time, we delineated the exact signaling cascade by which dietary supplementation of pomegranate fruit extract (PFE) protects tumor-bearing mice from tumor-induced hepatotoxicity. Increased activities of serum Alanine transaminase, Aspartate transaminase, Lactate dehydrogenase and Alkaline phosphatase, as well as histological studies confirmed the establishment of a state of hepatic dysfunction in tumor-bearers. Further investigations revealed that increased hepatic reactive oxygen species content and glutathione depletion-initiated apoptosis in these hepatocytes as we observed an alteration in the apoptotic proteins. PFE supplementation in tumor-bearing mice, on the other hand, differentially modulated redox-sensitive transcription factors Nrf2 and NF-κB, ultimately decreasing tumor-induced hepatic oxidative damage and cell death. siRNA-mediated inhibition of Nrf2 and NF-κB completely abolished the hepato-protective activities of PFE while pre-treatment of tumor-conditioned hepatocytes with N-acetyl cysteine augmented the cyto-protective properties of PFE. The present study clearly identified Nrf2/NF-κB/glutathione axis as the key factor behind the hepatoprotective potential of PFE. These findings would add to the existing knowledge about cancer chemoprevention by dietary polyphenols and might lead to the application of pomegranate polyphenols as supplement to escalate the effectiveness of cancer therapy by protecting normal cells from cancer related toxicities.


Asunto(s)
Carcinoma de Ehrlich/complicaciones , Glutatión/metabolismo , Hepatopatías/prevención & control , Factor 2 Relacionado con NF-E2/metabolismo , FN-kappa B/metabolismo , Polifenoles/administración & dosificación , Granada (Fruta) , Animales , Antioxidantes/metabolismo , Carcinoma de Ehrlich/metabolismo , Citocinas/metabolismo , Suplementos Dietéticos , Femenino , Hepatocitos/fisiología , Inflamación , Hígado/metabolismo , Hepatopatías/etiología , Hepatopatías/metabolismo , Hepatopatías/patología , Ratones , Estrés Oxidativo , Extractos Vegetales/administración & dosificación
15.
Heliyon ; 7(3): e06476, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33768175

RESUMEN

The role of oleic acid as a protective antioxidant has recently been recognized. The present study is aimed to explore whether oleic acid can afford protection to rat gastric tissue when challenged with adrenaline. Sixty adult healthy male albino rats were divided into 10 groups comprising of 6 animals each. First group constituted the control. Rats of the second group were injected sub-cutaneously with adrenaline bitartrate at the dose of 0.3mg/kg body weight, every day for a period of 17 days. Rats of the third, to the sixth groups were orally fed with different doses of oleic acid (2.5, 5, 10, 20 mg/kg body weight/day) respectively. The rats of seventh to tenth groups were orally fed with doses of oleic acid as mentioned above and subsequently injected with adrenaline bitartrate at 0.3mg/kg body weight sub-cutaneously. After the treatment period, the animals were euthanized through cervical dislocation following light ether anaesthesia and gastric tissues were collected for morphological and biochemical studies. Subcutaneously administered pharmacological dose of adrenaline bitartrate caused oxidative stress inducing gastric lesion in male albino rats as evident from the altered levels of biomarkers of oxidative stress, activities of antioxidant and mitochondrial enzymes related to energy metabolism with changes in tissue morphology. Pre-treatment of rats with oleic acid dose-dependently protected against these gastric injuries induced by adrenaline indicating the potentiality of oleic acid in protecting against adrenaline induced gastric injury in male albino rats where antioxidant mechanisms appear to play a pivotal role in mediating such protection.

16.
Biochim Biophys Acta Gen Subj ; 1865(3): 129841, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33412224

RESUMEN

BACKGROUND: Triple negative breast cancer (TNBC) is the most aggressive form of breast cancer with limited treatment modalities. It is associated with high propensity of cancer recurrence. METHODS: UV Spectroscopy, FTIR, DLS, Zeta potential, TEM and SEM were employed to characterize nanoparticles. MTT assay, Wound healing assay, SEM, Immunocytochemistry analysis, Western blot, RT-PCR, mammosphere formation assay were employed to study apoptosis, cell migration and stemness. Tumor regression was studied in chick embryo xenograft and BALB/c mice model. RESULTS: Hylaluronic acid engrafted metformin loaded graphene oxide (HA-GO-Met) nanoparticles exhibited an anti-cancer efficacy at much lower dosage as compared to metformin alone. HA-GO-Met nanoparticles induced apoptosis and inhibited cell migration of TNBC cells by targeting miR-10b/PTEN axis via NFkB-p65. Upregulation of PTEN affected pAKT(473) expression that induced apoptosis. Cell migration was inhibited by reduction of pFAK/integrinß1 expressions. Treatment inhibited epithelial mesenchymal transition (EMT) and reduced stemness as evident from the increase in E-cadherin expression, inhibition of mammosphere formation and low expression levels of stemness markers including nanog, oct4 and sox2 as compared to control. Moreover, tumor regression was studied in chick embryo xenograft and BALB/c mice model. HA-GO-Met nanoparticle treatment reduced tumor load and nullified toxicity in peripheral organs imparted by tumor. CONCLUSIONS: HA-GO-Met nanoparticles exhibited an enormous anti-cancer efficacy in TNBC in vitro and in vivo. GENERAL SIGNIFICANCE: HA-GO-Met nanoparticles induced apoptosis and attenuated cell migration in TNBC. It nullified overall toxicity imparted by tumor load. It inhibited EMT and reduced stemness and thereby addressed the issue of cancer recurrence.


Asunto(s)
Antineoplásicos/farmacología , Grafito/química , Receptores de Hialuranos/genética , Ácido Hialurónico/química , Metformina/farmacología , Nanopartículas/química , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Animales , Antineoplásicos/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Embrión de Pollo , Membrana Corioalantoides/efectos de los fármacos , Membrana Corioalantoides/metabolismo , Membrana Corioalantoides/patología , Portadores de Fármacos , Femenino , Quinasa 1 de Adhesión Focal/genética , Quinasa 1 de Adhesión Focal/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Receptores de Hialuranos/metabolismo , Metformina/metabolismo , Ratones , Ratones Endogámicos BALB C , MicroARNs/genética , MicroARNs/metabolismo , Terapia Molecular Dirigida , Nanopartículas/administración & dosificación , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Biomater Sci ; 8(10): 2939-2954, 2020 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-32319481

RESUMEN

The development of new therapeutic strategies to target triple-negative breast cancer (TNBC) is in much demand to overcome the roadblocks associated with the existing treatment procedures. In this regard, therapies targeting the CD44 receptor have drawn attention for more than a decade. MicroRNAs (miRNAs) modulate post-transcriptional gene regulation and thus, the correction of specific miRNA alterations using miRNA mimics or antagomiRs is an emerging strategy to normalize the genetic regulation in the tumor microenvironment. It has been acknowledged that miR-34a is downregulated and miR-10b is upregulated in TNBC, which promotes tumorigenesis and metastatic dissemination. However, there are a few barriers related to miRNA delivery. Herein, we have introduced tailored mesoporous silica nanoparticles (MSNs) for the co-delivery of miR-34a-mimic and antisense-miR-10b. MSN was functionalized with a cationic basic side chain and then loaded with the dual combination to overexpress miR-34a and downregulate miR-10b simultaneously. Finally, the loaded MSNs were coated with an hyaluronic acid-appended PEG-PLGA polymer for specific targeting. The cellular uptake, release profile, and subsequent effect in TNBC cells were evaluated. In vitro and in vivo studies demonstrated high specificity in TNBC tumor targeting, leading to efficient tumor growth inhibition as well as the retardation of metastasis, which affirmed the clinical application potential of the system.


Asunto(s)
Técnicas de Transferencia de Gen , MicroARNs/genética , Neoplasias de la Mama Triple Negativas/terapia , Animales , Femenino , Humanos , Receptores de Hialuranos/genética , Receptores de Hialuranos/metabolismo , Ratones , Ratones Endogámicos BALB C , MicroARNs/metabolismo , Nanopartículas/química , Tamaño de la Partícula , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Porosidad , Dióxido de Silicio/química , Propiedades de Superficie , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/metabolismo , Células Tumorales Cultivadas
18.
J Control Release ; 322: 357-374, 2020 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-32243981

RESUMEN

Triple-negative breast cancer (TNBC) is a highly aggressive and metastatic subtype of breast cancer showing non-responsiveness to most available therapeutic options. Therefore, smart therapeutic approaches to selectively transport and target TNBCs are required. Herein, we developed thymoquinone (TQ)-loaded, hyaluronic acid (HA)-conjugated Pluronic® P123 and F127 copolymer nanoparticles (HA-TQ-Nps) as a selective drug-carrying vehicle to deliver anticancer phytochemical TQ to TNBC cells. The mean size of nanoparticles was around 19.3 ± 3.2 nm. and they were stable at room temperature up to 4 months. HA-TQ-Nps were immensely cytotoxic towards TNBC cells but did not show the toxic effect on normal cells. Detailed investigations also demonstrated its pro-apoptotic, anti-metastatic and anti-angiogenic activity. In-depth mechanistic studies highlighted that HA-TQ-Nps retarded cell migration of TNBC cells through up-regulation of microRNA-361 which in turn down-regulated Rac1 and RhoA mediated cell migration and also perturbed the cancer cell migration under the influence of the autocrine effect of VEGF-A. Moreover, HA-TQ-Np-treatment also perturbed tumor-induced vascularization by reducing the secretion of VEGF-A. The anti-metastatic and anti-angiogenic activity of HA-TQ-Nps was found to be evident in both MDA-MB-231 xenograft chick embryos and 4T1-mammary solid tumor model in syngeneic mice. Thus, an innovative targeted nano-therapeutic approach is being established to reduce the tumor burden and inhibit metastasis and angiogenesis simultaneously for better management of TNBC.


Asunto(s)
MicroARNs , Nanopartículas , Neoplasias de la Mama Triple Negativas , Animales , Benzoquinonas , Línea Celular Tumoral , Embrión de Pollo , Humanos , Ácido Hialurónico , Ratones , Poloxámero , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico
19.
J Ethnopharmacol ; 250: 112463, 2020 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-31838178

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Bamboo shoots (BS) are consumed in various forms and used largely in naturopathy for curing ailments since ancient times to present days. It is eaten in South East Asian countries in several indigenous preparations. In north east India, it is consumed predominantly and used as natural cure to treat various diseases. Although known for its beneficial effects, adverse effects including goitrogenic/antithyroidal potential are emerging. AIM OF THE STUDY: Endemic goiter exists in Manipur, India even after adequate iodine intake for consumption of BS. It is thus important to study the impact of this goitrogenic food on certain thyroid hormone synthesizing regulatory factors at cellular and molecular level in thyrocytes. MATERIALS AND METHODS: Phytochemical analysis of BS - Bambusa balcooa Roxb (BSBR) extract conducted. IC50 of the extract on thyrocytes in culture was determined. To study the antithyroid effects of this goitrogenic food, activity status of Na+-K+-ATPase, TPO and Deiodinase, mRNA and protein expressions of NIS, TPO and PAX8 were investigated with and without extra iodine in culture media. Simultaneously ROS generation in terms of H2O2 and antioxidant status, NO, LPO were assayed. RESULTS: Activities of the studied enzymes decreased depending on dose and time with increased H2O2, decreased antioxidants followed by increased NO with LPO. DNA damage and LDH also increased while mRNA and protein expression of NIS, TPO and PAX8 were downregulated. Extra iodine ameliorated all such effects partially. CONCLUSIONS: Bioactive constituents of the extract imbalances oxidative status of thyrocytes impairing action of hormone synthesizing elements at cellular and molecular level.


Asunto(s)
Bambusa , Extractos Vegetales/farmacología , Células Epiteliales Tiroideas/efectos de los fármacos , Animales , Células Cultivadas , Daño del ADN , Femenino , Peróxido de Hidrógeno/metabolismo , Yoduro Peroxidasa/genética , Fitoquímicos/análisis , Fitoquímicos/farmacología , Extractos Vegetales/química , Brotes de la Planta , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/genética , Células Epiteliales Tiroideas/metabolismo , Hormonas Tiroideas/metabolismo
20.
Cell Signal ; 68: 109521, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31881324

RESUMEN

Inflammation has been associated with the progression of many neurological diseases. Peripheral inflammation has also been vaguely linked to depression-like symptoms in animal models, but the underlying pathways that orchestrate inflammation-induced behavioral or molecular changes in the brain are still elusive. We have recently shown that intraperitoneal injections of lipopolysaccharide (LPS) to Swiss albino mice triggers systemic inflammation, leading to an activated immune response along with changes in monoamine levels in the brain. Herein we pinpoint the fundamental pathways linking peripheral inflammation and depression-like behavior in a mouse model, thereby identifying suitable targets of intervention to combat the situation. We show that LPS-induced peripheral inflammation provoked a depression-like behavior in mice and a distinct pro-inflammatory bias in the hippocampus, as evident from increased microglial activation and elevated levels of pro-inflammatory cytokines IL-6 and TNF-α, and activation of NFκB-p65 pathway. Significant alterations in Nrf2-dependent cellular redox status, coupled with altered autophagy and increased apoptosis were noticed in the hippocampus of LPS-exposed mice. We and others have previously shown that, fluoxetine (an anti-depressant) has effective anti-inflammatory and antioxidant properties by virtue of its abilities to regulate NFκB and Nrf2 signaling. We observed that treatment with fluoxetine or the Nrf2 activator tBHQ (tert-butyl hydroquinone), could reverse depression-like-symptoms and mitigate alterations in autophagy and cell death pathways in the hippocampus by activating Nrf2-dependent gene expressions. Taken together, the data suggests that systemic inflammation potentiates Nrf2-dependent changes in cell death and autophagy pathway in the hippocampus, eventually leading to major pathologic sequelae associated with depression. Therefore, targeting Nrf2 could be a novel approach in combatting depression and ameliorating its associated pathogenesis.


Asunto(s)
Apoptosis , Autofagia , Conducta Animal , Fluoxetina/farmacología , Hipocampo/patología , Inflamación/patología , Animales , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Catalasa/metabolismo , Depresión/patología , Glutatión/metabolismo , Hidroquinonas , Lipopolisacáridos , Masculino , Ratones , Microglía/efectos de los fármacos , Microglía/patología , Factor 2 Relacionado con NF-E2/metabolismo , FN-kappa B/metabolismo , Óxido Nítrico/metabolismo , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Superóxido Dismutasa/metabolismo , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...