Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
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
2.
Apoptosis ; 28(3-4): 263-276, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36536258

RESUMEN

Tumor cells always have the need to produce an increased amount of proteins in the cells. This elevated amount of proteins increases the pressure on the organelles of the cell such as the endoplasmic reticulum and compels it to increase its protein folding efficiency. However, it is by a matter of fact, that the amount of proteins synthesized outweighs the protein folding capacity of the ER which in turn switches on the UPR pathway by activating the three major molecular sensors and other signaling cascades, which helps in cell survival instead of instant death. However, if this pathway is active for a prolonged period of time the tumor cells heads toward apoptosis. Again, interestingly this is not the same as in case of non- tumorogenic cells. This exhibit a straight natural pathway for tumor cells-specific destruction which has a great implication in today's world where hormone therapies and chemo-therapies are non-effective for various types of breast cancer, a major type being Triple Negative Breast Cancer. Thus a detailed elucidation of the molecular involvement of the UPR pathway in breast cancer may open new avenues for management and attract novel chemotherapeutic targets providing better hopes to patients worldwide.


Asunto(s)
Neoplasias de la Mama , Humanos , Femenino , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Estrés del Retículo Endoplásmico , Apoptosis/genética , Respuesta de Proteína Desplegada , Transducción de Señal
3.
Environ Toxicol ; 37(1): 52-68, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34581487

RESUMEN

Although comprehensive exertions have been made in late decades for treating advanced lung cancer with inclusive therapies but efficient anti-lung cancer therapeutics are statically inadequate in the clinics. Hence, compelling novel anti-lung cancer drugs are considerably desired. This backdrop enticed us to unveil anticancer efficacy of astrakurkurol, derivative of wild edible mushroom against lung cancer, whose effects have not yet been described. Mechanistic analysis disclosed that sensitizing effect of astrakurkurol is due to cell cycle arrest at G0/G1 phase, increased level of Fas, FADD, decreased ratio of Bax/Bcl-2, and increased cleaved form of caspase 9, 8, and 3. Apart from the induction of apoptosis, it was demonstrated for the first time that astrakurkurol induced an autophagic response as evidenced by the development of acidic vesicular organelles (AVOs) with up-regulation of beclin-1, Atg7, and downregulated p62. Apoptosis and autophagy can be sparked by the same stimuli, which was as evident from the astrakurkurol-induced inactivation of PI3K/AKT signaling. The thorough scanning of the mechanism of crosstalk between apoptosis and autophagy is requisite for prosperous anticancer remedy. Triterpenoid has evidently intensified cytotoxicity, induced apoptosis and autophagy on A549 cells. Besides astrakurkurol could also curb migration and regress the size of tumor in ex ovo xenograft model. All these findings put forth astrakurkurol as a convincing novel anti-cancer agent, for scrutinizing the lung cancer therapies and as a robust contender for future in vitro and in vivo analysis.


Asunto(s)
Adenocarcinoma del Pulmón , Agaricales , Neoplasias Pulmonares , Células A549 , Apoptosis , Autofagia , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt
4.
J Water Health ; 18(3): 306-313, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32589617

RESUMEN

A novel reactor was designed and implemented for water purification using deep ultraviolet light emitting diodes (LEDs). The focus was on minimizing the number of LEDs required for effective germicidal action. Simulation studies were carried out on the flow of water as well as the irradiance of UV. Variation was made in the beam divergence of the UV sources and reflectivity of optical coatings used for photon recycling. Based on optimized reactor designs, water purification was carried out both in the static and flow-through configuration. Water from various sources was spiked with a known bacterial strain, exposure studies were carried out and germicidal effect was determined. Our results indicate that under optimal design, a 3 mL volume of water shows a three order inactivation using a single UV-LED in a static reactor in 180 s. For a flow-through geometry, only three LEDs were used in the reactor implementation, and a multi-pass procedure was used to purify 150 mL of water from an Escherichia coli CFU count of 4.3 × 104/mL to 12/mL. While slow, this process requires less than 2 W, and can be powered from rechargeable sources. Faster processes can be implanted using multiple such reactor units in parallel, and can be optimized to the requirement and power levels.


Asunto(s)
Desinfección , Rayos Ultravioleta , Purificación del Agua , Escherichia coli , Microbiología del Agua
5.
Apoptosis ; 24(11-12): 958-971, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31641961

RESUMEN

Anoikis resistance is an essential property of cancer cells that allow the extra-cellular matrix-detached cells to survive in a suspended state in body fluid in order to metastasize and invade to distant organs. It is known that integrins play an important role in anoikis resistance, but detailed mechanisms are not well understood. Here we report that highly metastatic colon cancer cells showed a higher degree of anoikis resistance than the normal intestinal epithelial cells. These anoikis-resistant cancer cells express high-levels of integrin-α2, ß1, and activated EGFR in the anchorage-independent state than the anchorage-dependent state. In contrast, normal intestinal epithelial cells failed to elevate these proteins. Interestingly, a higher co-association of EGFR with integrin-α2ß1/-α5ß1 was observed on the surface of anoikis-resistant cells. Thus, in the absence of extra-cellular matrix, integrins in association with EGFR activates downstream effectors ERK and AKT and suppress Caspase-3 activation to induce anoikis resistance as was confirmed from the gene-ablation and pharmacological inhibitor studies. Interestingly, these anoikis-resistant cancer cells express high-level of cancer stem cell signatures (CD24, CD44, CD133, EpCAM) and pluripotent stem cell markers (OCT-4, SOX-2, Nanog) as well as drug-resistant pumps (ABCG2, MDR1, MRP1). Altogether, our findings unravel the interplay between integrin-α2ß1/-α5ß1 and EGFR in anoikis resistance and suggest that the resistant cells are cancer initiating or cancer stem cells, which may serve as a promising target to combat metastasis of cancer.


Asunto(s)
Anoicis , Neoplasias del Colon/metabolismo , Integrina alfa2beta1/metabolismo , Células Madre Neoplásicas/metabolismo , Antígeno AC133/metabolismo , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Anoicis/genética , Antígeno CD24/metabolismo , Línea Celular Tumoral , Neoplasias del Colon/genética , Células Epiteliales/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Humanos , Receptores de Hialuranos/metabolismo , Integrina alfa2beta1/genética , Sistema de Señalización de MAP Quinasas/genética , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Proteína Homeótica Nanog/metabolismo , Proteínas de Neoplasias/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factores de Transcripción SOXB1/metabolismo
6.
Mol Carcinog ; 56(9): 2086-2103, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28418078

RESUMEN

Triple negative breast cancer (TNBC) tends to form aggressive tumors associated with high mortality and morbidity which urge the need for development of new therapeutic strategies. Recently, the normal metabolite Methylglyoxal (MG) has been documented for its anti-proliferative activity against human breast cancer. However, the mode of action of MG against TNBC remains open to question. In our study, we investigated the anticancer activity of MG in MDA MB 231 and 4T1 TNBC cell lines and elucidated the underlying mechanisms. MG dose-dependently caused cell death, induced apoptosis, and generated ROS in both the TNBC cell lines. Furthermore, such effects were attenuated in presence of ROS scavenger N-Acetyl cysteine. MG triggered mitochondrial cytochrome c release in the cytosol and up-regulated Bax while down-regulated anti-apoptotic protein Bcl-2. Additionally, MG treatment down-regulated phospho-akt and inhibited the nuclear translocation of the p65 subunit of NF-κB. MG exhibited a tumor suppressive effect in BALB/c mouse 4T1 breast tumor model as well. The cytotoxic effect was studied using MTT assay. Apoptosis, ROS generation, and mitochondrial dysfunction was evaluated by flow cytometry as well as fluorescence microscopy. Western blot assay was performed to analyze proteins responsible for apoptosis. This study demonstrated MG as a potent anticancer agent against TNBC both in vitro and in vivo. The findings will furnish fresh insights into the treatment of this subgroup of breast cancer.


Asunto(s)
Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Piruvaldehído/uso terapéutico , Especies Reactivas de Oxígeno/metabolismo , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Citocromos c/metabolismo , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Ratones , Ratones Endogámicos BALB C , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factor de Transcripción ReIA/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
7.
J Biol Chem ; 290(7): 3936-49, 2015 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-25527500

RESUMEN

Triple negative breast cancers (TNBC) are among the most aggressive and therapy-resistant breast tumors and currently possess almost no molecular targets for therapeutic options in this horizon. In the present study we discerned the molecular mechanisms of potential interaction between the endoplasmic reticulum (ER) stress response and the MEK/ERK pathway in inducing apoptosis in TNBC cells. Here we observed that induction of ER stress alone was not sufficient to trigger significant apoptosis but simultaneous inhibition of the MEK/ERK pathway enhanced ER stress-induced apoptosis via a caspase-dependent mechanism. Our study also demonstrated nifetepimine, a dihydropyrimidone derivative as a potent anti-cancer agent in TNBC cells. Nifetepimine down-regulated the MEK/ERK pathway in MDAMB-231 and MDAMB-468 cells and resulted in blockage of ER stress-mediated GRP78 up-regulation. Detailed mechanistic studies also revealed that nifetepimine by down-regulating pERK expression also declined the promoter binding activity of TFII-I to the GRP78 promoter and in turn regulated GRP78 transcription. Studies further extended to in vivo Swiss albino and SCID mice models also revalidated the anti-carcinogenic property of nifetepimine. Thus our findings cumulatively suggest that nifetepimine couples two distinct signaling pathways to induce the apoptotic death cascade in TNBC cells and raises the possibility for the use of nifetepimine as a potent anti-cancer agent with strong immune-restoring properties for therapeutic intervention for this group of cancer bearers.


Asunto(s)
Apoptosis/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Quinasas Quinasa Quinasa PAM/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Pirimidinonas/farmacología , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/patología , Animales , Western Blotting , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Chaperón BiP del Retículo Endoplásmico , Femenino , Proteínas de Choque Térmico/antagonistas & inhibidores , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Humanos , Técnicas para Inmunoenzimas , Quinasas Quinasa Quinasa PAM/genética , Masculino , Ratones , Ratones Desnudos , Ratones SCID , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/genética , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Neoplasias de la Mama Triple Negativas/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Apoptosis ; 21(10): 1106-24, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27392939

RESUMEN

The efficacy of cancer chemotherapeutics is limited by side effects resulting from narrow therapeutic windows between the anticancer activity of a drug and its cytotoxicity. Thus identification of small molecules that can selectively target cancer cells has gained major interest. Cancer cells under stress utilize the Unfolded protein response (UPR) as an effective cell adaptation mechanism. The purpose of the UPR is to balance the ER folding environment and calcium homeostasis under stress. If ER stress is prolonged, tumor cells undergo apoptosis. In the present study we demonstrated an 3,3'-(Arylmethylene)-bis-1H-indole (AMBI) derivative 3,3'-[(4-Methoxyphenyl) methylene]-bis-(5-bromo-1H-indole), named as Mephebrindole (MPB) as an effective anti-cancer agent in breast cancer cells. MPB disrupted calcium homeostasis in MCF7 cells which triggered ER stress development. Detailed evaluations revealed that mephebrindole by activating p38MAPK also regulated GRP78 and eIF2α/ATF4 downstream to promote apoptosis. Studies extended to in vivo allograft mice models revalidated its anti-carcinogenic property thus highlighting the role of MPB as an improved chemotherapeutic option.


Asunto(s)
Factor de Transcripción Activador 4/metabolismo , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Carcinoma/tratamiento farmacológico , Factor 2 Eucariótico de Iniciación/metabolismo , Indoles/administración & dosificación , Factor de Transcripción CHOP/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Factor de Transcripción Activador 4/genética , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/fisiopatología , Carcinoma/genética , Carcinoma/metabolismo , Carcinoma/fisiopatología , Línea Celular Tumoral , Chaperón BiP del Retículo Endoplásmico , Factor 2 Eucariótico de Iniciación/genética , Femenino , Humanos , Indoles/síntesis química , Indoles/química , Ratones , Receptor Cross-Talk/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factor de Transcripción CHOP/genética , Proteínas Quinasas p38 Activadas por Mitógenos/genética
9.
J Biol Chem ; 289(37): 25431-44, 2014 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-25086032

RESUMEN

The evolution of the cancer cell into a metastatic entity is the major cause of death in patients with cancer. It has been acknowledged that aberrant activation of a latent embryonic program, known as the epithelial-mesenchymal transition (EMT), can endow cancer cells with the migratory and invasive capabilities associated with metastatic competence for which E-cadherin switch is a well-established hallmark. Discerning the molecular mechanisms that regulate E-cadherin expression is therefore critical for understanding tumor invasiveness and metastasis. Here we report that SMAR1 overexpression inhibits EMT and decelerates the migratory potential of breast cancer cells by up-regulating E-cadherin in a bidirectional manner. While SMAR1-dependent transcriptional repression of Slug by direct recruitment of SMAR1/HDAC1 complex to the matrix attachment region site present in the Slug promoter restores E-cadherin expression, SMAR1 also hinders E-cadherin-MDM2 interaction thereby reducing ubiquitination and degradation of E-cadherin protein. Consistently, siRNA knockdown of SMAR1 expression in these breast cancer cells results in a coordinative action of Slug-mediated repression of E-cadherin transcription, as well as degradation of E-cadherin protein through MDM2, up-regulating breast cancer cell migration. These results indicate a crucial role for SMAR1 in restraining breast cancer cell migration and suggest the candidature of this scaffold matrix-associated region-binding protein as a tumor suppressor.


Asunto(s)
Neoplasias de la Mama/genética , Cadherinas/biosíntesis , Proteínas de Ciclo Celular/biosíntesis , Proteínas de Unión al ADN/biosíntesis , Transición Epitelial-Mesenquimal/genética , Proteínas Nucleares/biosíntesis , Neoplasias de la Mama/patología , Cadherinas/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Histona Desacetilasa 1/genética , Histona Desacetilasa 1/metabolismo , Humanos , Metástasis de la Neoplasia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteolisis , Proteínas Proto-Oncogénicas c-mdm2/genética , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética
10.
J Biol Chem ; 289(42): 29074-85, 2014 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-25157104

RESUMEN

Matrix attachment region (MAR)-binding proteins have been implicated in the transcriptional regulation of host as well as viral genes, but their precise role in HPV-infected cervical cancer remains unclear. Here we show that HPV18 promoter contains consensus MAR element in the LCR and E6 sequences where SMAR1 binds and reinforces HPV18 E6 transcriptional silencing. In fact, curcumin-induced up-regulation of SMAR1 ensures recruitment of SMAR1-HDAC1 repressor complex at the LCR and E6 MAR sequences, thereby decreasing histone acetylation at H3K9 and H3K18, leading to reorientation of the chromatin. As a consequence, c-Fos binding at the putative AP-1 sites on E6 promoter is inhibited. E6 depletion interrupts degradation of E6-mediated p53 and lysine acetyl transferase, Tip60. Tip60, in turn, acetylates p53, thereby restoring p53-mediated transactivation of proapoptotic genes to ensure apoptosis. This hitherto unexplained function of SMAR1 signifies the potential of this unique scaffold matrix-associated region-binding protein as a critical regulator of E6-mediated anti-apoptotic network in HPV18-infected cervical adenocarcinoma. These results also justify the candidature of curcumin for the treatment of HPV18-infected cervical carcinoma.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Cromatina/metabolismo , Proteínas de Unión al ADN/metabolismo , Regulación Viral de la Expresión Génica , Proteínas Nucleares/metabolismo , Proteínas Oncogénicas Virales/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Transcripción Genética , Acetilación , Apoptosis , Células HeLa , Histonas/metabolismo , Humanos , Regiones Promotoras Genéticas , Unión Proteica , Factor de Transcripción AP-1/metabolismo
11.
J Phys Chem Lett ; 15(11): 3078-3088, 2024 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-38467015

RESUMEN

A biomimetic cell-based carrier system based on monocyte membranes and liposomes has been designed to create a hybrid "Monocyte-LP" which inherits the surface antigens of the monocytes along with the drug encapsulation property of the liposome. Förster resonance energy transfer (FRET) and polarization gated anisotropy measurements show the stiffness of the vesicles obtained from monocyte membranes (Mons), phosphatidylcholine membranes (LP), and Monocyte-LP to follow an increasing order of Mons > Monocyte-LP > LP. The dynamics of interface bound water molecules plays a key role in the elasticity of the vesicles, which in turn imparts higher delivery efficacy to the hybrid Monocyte-LP for a model anticancer drug doxorubicin than the other two vesicles, indicating a critical balance between flexibility and rigidity for an efficient cellular uptake. The present work provides insight on the influence of elasticity of delivery vehicles for enhanced drug delivery.


Asunto(s)
Antineoplásicos , Liposomas , Liposomas/metabolismo , Monocitos/metabolismo , Doxorrubicina , Sistemas de Liberación de Medicamentos
12.
J Biol Chem ; 287(39): 32881-96, 2012 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-22851172

RESUMEN

Multiple mechanisms have been proposed by which tumors induce T cell apoptosis to circumvent tumor immune-surveillance. Although sarco/endoplasmic reticulum Ca(2+)-ATPase (SERCA) have long been known to regulate intracellular Ca(2+) homeostasis, few studies have examined the role of SERCA in processes of T lymphocyte survival and activation. In this context it remains largely unexplored as to how tumors jeopardize SERCA function to disable T cell-mediated anti-tumor immunity. Here, we show that human CD4(+) T cells in the presence of tumor conditions manifested an up-regulation of SERCA3 expression that resulted in development of endoplasmic reticulum stress leading to CD4(+) T cell apoptosis. Prostaglandin E(2) produced by the tumor cell plays a critical role in up-regulating SERCA3 by enhancing the binding of its transcription factor Sp1. Gene manipulation and pharmacological approaches further established that an increase in SERCA expression also resulted in subsequent inhibition of PKCα and -θ and retention of NFκB in the cytosol; however, down-modulation of SERCA3 expression by a dihydropyrimidone derivative, ethyl-4-(3-nitro)-phenyl-6-methyl-2-oxo-1,2,3,4-tetrahydropyrimidine-5 carboxylate (nifetepimine), protected the CD4(+) T cells from tumor-induced apoptosis. In fact, nifetepimine-mediated restoration of PKC activity resulted in nuclear translocation of p65NFκB, thereby ensuring its survival. Studies further undertaken in a tumor-bearing mice model revalidated the immunoprotective role of nifetepimine. Our present study thus strongly suggests that imbalance in cellular calcium homeostasis is an important factor leading to CD4(+) T cell death during cancer and holds promise that nifetepimine may have the potential to be used as an immunorestoring agent in cancer bearers.


Asunto(s)
Neoplasias de la Mama/enzimología , Linfocitos T CD4-Positivos/metabolismo , Calcio/metabolismo , Factores Inmunológicos/farmacología , Proteínas de Neoplasias/metabolismo , Pirimidinonas/farmacología , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/biosíntesis , Microambiente Tumoral/efectos de los fármacos , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/inmunología , Linfocitos T CD4-Positivos/inmunología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Dinoprostona/genética , Dinoprostona/inmunología , Dinoprostona/metabolismo , Estrés del Retículo Endoplásmico/efectos de los fármacos , Estrés del Retículo Endoplásmico/genética , Estrés del Retículo Endoplásmico/inmunología , Femenino , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/genética , Regulación Enzimológica de la Expresión Génica/inmunología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Masculino , Ratones , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/inmunología , Trasplante de Neoplasias , Proteína Quinasa C-alfa/genética , Proteína Quinasa C-alfa/inmunología , Proteína Quinasa C-alfa/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/inmunología , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp1/inmunología , Factor de Transcripción Sp1/metabolismo , Trasplante Heterólogo , Microambiente Tumoral/genética , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética , Regulación hacia Arriba/inmunología
13.
Apoptosis ; 18(5): 589-604, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23329180

RESUMEN

Mutations in REarranged during Transfection (RET) receptor tyrosine, followed by the oncogenic activation of RET kinase is responsible for the development of medullary thyroid carcinoma (MTC) that responds poorly to conventional chemotherapy. Targeting RET, therefore, might be useful in tailoring surveillance of MTC patients. Here we showed that theaflavins, the bioactive components of black tea, successfully induced apoptosis in human MTC cell line, TT, by inversely modulating two molecular pathways: (i) stalling PI3K/Akt/Bad pathway that resulted in mitochondrial transmembrane potential (MTP) loss, cytochrome-c release and activation of the executioner caspases-9 and -3, and (ii) upholding p38MAPK/caspase-8/caspase-3 pathway via inhibition of Ras/Raf/ERK. Over-expression of either constitutively active myristoylated-Akt-cDNA (Myr-Akt-cDNA) or dominant-negative-caspase-8-cDNA (Dn-caspase-8-cDNA) partially blocked theaflavin-induced apoptosis, while co-transfection of Myr-Akt-cDNA and Dn-caspase-8-cDNA completely eradicated the effect of theaflavins thereby negating the possibility of existence of other pathways. A search for the upstream signaling revealed that theaflavin-induced disruption of lipid raft caused interference in anchorage of RET in lipid raft that in turn stalled phosphorylation of Ras and PI3Kinase. In such anti-survival cellular micro-environment, pro-apoptotic signals were triggered to culminate into programmed death of MTC cell. These findings not only unveil a hitherto unexplained mechanism underlying theaflavin-induced MTC death, but also validate RET as a promising and potential target for MTC therapy.


Asunto(s)
Caspasa 8/genética , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-ret/genética , Neoplasias de la Tiroides/genética , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Antineoplásicos Fitogénicos/farmacología , Apoptosis , Biflavonoides/farmacología , Carcinoma Neuroendocrino , Caspasa 8/metabolismo , Catequina/farmacología , Línea Celular Tumoral , Citocromos c/metabolismo , ADN Complementario , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Microdominios de Membrana/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Proteína Oncogénica p21(ras)/genética , Proteína Oncogénica p21(ras)/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-ret/metabolismo , Transducción de Señal/efectos de los fármacos , Neoplasias de la Tiroides/metabolismo , Neoplasias de la Tiroides/patología , Transfección , Proteína Letal Asociada a bcl/genética , Proteína Letal Asociada a bcl/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
14.
Biometals ; 26(3): 517-34, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23733180

RESUMEN

Drug induced toxicity and drug resistance are the major impediments to successful application of cancer chemotherapy. Therefore, selective targeting of the key biochemical events of the malignant cells may have a great therapeutic potential in specifically kill the cancer cells. We have evaluated in vitro the cytotoxic efficacy of a previously reported copper complex viz. copper N-(2-hydroxy acetophenone) glycinate (CuNG) on different drug sensitive and resistant cancer cell lines by MTT, annexin V positivity and caspase 3 activation assays. We have also investigated the underlying signalling events in CuNG mediated apoptosis of cancer cells by Western blotting technique. We have found that CuNG preferentially induces apoptosis to malignant cells irrespective of drug sensitivity and spares the normal cells. Our studies disclose that CuNG causes cellular redox imbalance in cancer cells through depletion of intracellular GSH level. CuNG mediated depletion of intracellular GSH level induces mitochondrial superoxide generation, which detaches cyto C from mitochondrial membrane through lipid peroxidation. The detached cyto C then release into the extra mitochondrial milieu in Bax mediated pathway where CuNG facilitates the binding of Bax through dissociation of hexokinase II from mitochondrial membrane. The present study opens the possibility of developing effective chemotherapeutic drugs by synthesizing numerous chemical compounds capable of targeting cellular redox environment and thus specifically kills cancer cells of broad spectrum.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Quelantes/farmacología , Glutatión/metabolismo , Glicina/análogos & derivados , Compuestos Organometálicos/farmacología , Animales , Antineoplásicos/química , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Quelantes/química , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Glicina/química , Glicina/farmacología , Células HCT116 , Células HEK293 , Células HeLa , Humanos , Células K562 , Ratones , Células 3T3 NIH , Compuestos Organometálicos/química , Relación Estructura-Actividad
15.
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
16.
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
17.
Mol Cell Biochem ; 364(1-2): 309-20, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22258826

RESUMEN

One of the major reasons for multidrug resistance (MDR) in cancer is the overexpression of P-glycoprotein (P-gp, ABCB1), a drug efflux pump. A novel copper complex, namely, copper (II) N-(2-hydroxyacetophenone) glycinate (CuNG) previously synthesized and characterized by the authors had been tested in this study. In a cell-based assay system with human MDR1 cDNA overexpressed mouse fibroblast NIH MDR1-G185 cell line, we demonstrated that this metal complex can directly interact with this transporter. As CuNG increased cellular accumulation of doxorubicin in P-gp-expressing cells, we presumed that of CuNG may be potential to reverse P-gp-mediated drug resistance probably by lowering the P-gp expression at the protein as well as mRNA level. Interestingly, our studies on UIC2 (a conformation sensitive monoclonal antibody) binding assay indicated the direct interaction of CuNG with P-gp. However, CuNG did not compete for the substrate binding as photoaffinity labeling of P-gp with a substrate analog [(125)I] iodoarylazidoprazosin ([(125)I] IAAP) showed approximately twofold increase in [(125)I] IAAP binding in presence of CuNG. In vitro study showed that CuNG significantly stimulated P-gp-specific ATPase activity in isolated membrane preparations from NIH MDR1-G185 cells. This result further confirmed the CuNG-P-gp direct interaction. This study also demonstrated that CuNG has a drug interaction site different from verapamil-, vinblastine- and progesterone-binding sites on P-gp. Taken together, this is the first report of molecular interaction of any Schiff's base metal chelate CuNG with human P-gp. This information may be useful to design more efficacious nontoxic metal-based drugs as MDR-reversing agents.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Quelantes/farmacología , Cobre , Subfamilia B de Transportador de Casetes de Unión a ATP , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/biosíntesis , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Adenosina Trifosfatasas/metabolismo , Animales , Línea Celular , Quelantes/química , Doxorrubicina/farmacología , Fibroblastos/metabolismo , Glicina/análogos & derivados , Glicina/química , Glicina/farmacología , Humanos , Ratones , Compuestos Organometálicos/química , Compuestos Organometálicos/farmacología
18.
J Food Biochem ; 46(1): e14021, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34811765

RESUMEN

Astraeus hygrometricus extensively been utilized by tribal people for long time. A triterpene, astrakurkurol has been isolated from A. hygrometricus but anticancer effect of this novel triterpene has imperceptibly been investigated. Motive of this research was to scrutinize its underlying apoptotic mechanism in HepG2 cells. Cytotoxicity studies demonstrated a selective effect of astrakurkurol with towering influence in HepG2 than Thle2 cells. The exposure of these triterpene-induced marked apoptotic morphological changes enhanced the rate of cell apoptosis and arrest cell cycle at G0/G1. Furthermore, these results are aided by decline in the expression of Bcl-2, Bcl-xL with an increase in the expression of p53, Bax, Fas, FADD together with the activation of caspase cascade. Astrakurkurol also displayed a remarkable anti-migratory capacity at a lower concentration. Altogether, studies explained anti-proliferative, pro-apoptotic, and anti-migratory efficacy of astrakurkurol on HepG2, composing a gripping challenge in the advancement of novel treatments against hepatocellular carcinoma. PRACTICAL APPLICATIONS: Mushrooms, the minuscule pharmaceutical factory, bear hundreds of novel elements with incredible biological attributes. Triterpenoids from mushrooms has been proven to bear potentials of curing cancer. This study highlights the cytotoxic and anti-migratory effects of novel triterpene in vitro in HepG2 cell, an HCC cell line. Astrakurkurol mediated cell death via both extrinsic and intrinsic apoptotic signaling. Utilization of astrakurkurol will provide a non-toxic substitute of chemotherapy and also uplift the value of forsaken taxon, Astraeus and boost the rural acceptance.


Asunto(s)
Agaricales , Carcinoma Hepatocelular , Neoplasias Hepáticas , Apoptosis , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/genética , Puntos de Control del Ciclo Celular , Línea Celular , Folclore , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/genética
19.
J Biol Chem ; 285(43): 33104-33112, 2010 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-20675383

RESUMEN

Tumor suppressor p53 plays the central role in regulating apoptosis in response to genotoxic stress. From an evolutionary perspective, the activity of p53 has to be backed up by other protein(s) in case of any functional impairment of this protein, to trigger DNA damage-induced apoptosis in cancer cells. We adopted multiple experimental approaches to demonstrate that in p53-impaired cancer cells, DNA damage caused accumulation of p53 paralogue p73 via Chk-1 that strongly impacted Bax expression and p53-independent apoptosis. On the contrary, when p53 function was restored by ectopic expression, Chk-2 induced p53 accumulation that in turn overshadowed p73 activity, suggesting an antagonistic interaction between p53 family members. To understand such interaction better, p53-expressing cells were impaired differentially for p53 activity. In wild-type p53-expressing cancer cells that were silenced for p53 for several generations, p73 was activated, whereas no such trend was observed when p53 was transiently silenced. Prolonged p53 interference, even in functional p53 settings, therefore, leads to the "gain of cellular adaptation" in a way that alters the cellular microenvironment in favor of p73 activation by altering p73-regulatory proteins, e.g. Chk1 activation and dominant negative p73 down-regulation. These findings not only unveil a hitherto unexplained mechanism underlying the functional switchover from p53 to p73, but also validate p73 as a promising and potential target for cancer therapy in the absence of functional p53.


Asunto(s)
Daño del ADN , Proteínas de Unión al ADN/metabolismo , Leucemia Mieloide Aguda/metabolismo , Proteínas Nucleares/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Apoptosis/genética , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Proteínas de Unión al ADN/genética , Regulación Leucémica de la Expresión Génica/genética , Silenciador del Gen , Células HL-60 , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/terapia , Proteínas Nucleares/genética , Proteínas Quinasas/biosíntesis , Proteínas Quinasas/genética , Factores de Tiempo , Proteína Tumoral p73 , Proteína p53 Supresora de Tumor/genética , Proteínas Supresoras de Tumor/genética , Células U937 , Proteína X Asociada a bcl-2/biosíntesis , Proteína X Asociada a bcl-2/genética
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA