Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
ACS Nano ; 18(33): 22298-22315, 2024 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-39117621

RESUMO

A series of successes in RNA interference (RNAi) therapies for liver diseases using lipid nanoparticles and N-acetylgalactosamine have heralded a current era of RNA therapeutics. However, alternative delivery strategies are required to take RNAi out of the comfort zone of hepatocytes. Here we report SIRPα IgV/anti-CD47 siRNA (vS-siCD47) conjugates that selectively and persistently disrupt the antiphagocytic CD47/SIRPα axis in solid tumors. Conjugation of the SIRPα IgV domain protein to siRNAs enables tumor dash through CD47-mediated erythrocyte piggyback, primarily blocking the physical interaction between CD47 on cancer cells and SIRPα on phagocytes. After internalization of the vS-siCD47 conjugates within cancer cells, the detached free-standing anti-CD47 siRNAs subsequently attack CD47 through the RNAi mechanism. The dual-action approach of the vS-siCD47 conjugate effectively overcomes the "don't eat me" barrier and stimulates phagocyte-mediated tumor destruction, demonstrating a highly selective and potent CD47-blocking immunotherapy. This delivery strategy, employing IgV domain protein-siRNA conjugates with a dual mode of target suppression, holds promise for expanding RNAi applications beyond hepatocytes and advancing RNAi-based cancer immunotherapies for solid tumors.


Assuntos
Antígeno CD47 , RNA Interferente Pequeno , Receptores Imunológicos , Antígeno CD47/metabolismo , Antígeno CD47/química , Humanos , RNA Interferente Pequeno/química , Animais , Camundongos , Receptores Imunológicos/metabolismo , Neoplasias/terapia , Neoplasias/genética , Neoplasias/patologia , Antígenos de Diferenciação , Linhagem Celular Tumoral
2.
Int J Mol Sci ; 24(14)2023 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-37511470

RESUMO

Neurodevelopmental disorders stemming from maternal immune activation can significantly affect a child's life. A major limitation in pre-clinical studies is the scarcity of valid animal models that accurately mimic these challenges. Among the available models, administration of lipopolysaccharide (LPS) to pregnant females is a widely used paradigm. Previous studies have reported that a model of 'emotional stress', involving chronic exposure of rodents to ultrasonic frequencies, induces neuroinflammation, aberrant neuroplasticity, and behavioral deficits. In this study, we explored whether this model is a suitable paradigm for maternal stress and promotes neurodevelopmental abnormalities in the offspring of stressed females. Pregnant dams were exposed to ultrasound stress for 21 days. A separate group was injected with LPS on embryonic days E11.5 and E12.5 to mimic prenatal infection. The behavior of the dams and their female offspring was assessed using the sucrose test, open field test, and elevated plus maze. Additionally, the three-chamber sociability test and Barnes maze were used in the offspring groups. ELISA and qPCR were used to examine pro-inflammatory changes in the blood and hippocampus of adult females. Ultrasound-exposed adult females developed a depressive-like syndrome, hippocampal overexpression of GSK-3ß, IL-1ß, and IL-6 and increased serum concentrations of IL-1ß, IL-6, IL-17, RANTES, and TNFα. The female offspring also displayed depressive-like behavior, as well as cognitive deficits. These abnormalities were comparable to the behavioral changes induced by LPS. The ultrasound stress model can be a promising animal paradigm of neurodevelopmental pathology associated with prenatal 'emotional stress'.


Assuntos
Comportamento Animal , Efeitos Tardios da Exposição Pré-Natal , Camundongos , Gravidez , Animais , Humanos , Feminino , Comportamento Animal/fisiologia , Efeitos Tardios da Exposição Pré-Natal/patologia , Lipopolissacarídeos/toxicidade , Glicogênio Sintase Quinase 3 beta , Interleucina-6/efeitos adversos , Citocinas , Modelos Animais de Doenças
3.
Breast Cancer Res Treat ; 190(3): 373-387, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34553295

RESUMO

PURPOSE: Tumor cells are dependent on the glutathione and thioredoxin antioxidant pathways to survive oxidative stress. Since the essential amino acid methionine is converted to glutathione, we hypothesized that methionine restriction (MR) would deplete glutathione and render tumors dependent on the thioredoxin pathway and its rate-limiting enzyme thioredoxin reductase (TXNRD). METHODS: Triple (ER/PR/HER2)-negative breast cancer (TNBC) cells were treated with control or MR media and the effects on reactive oxygen species (ROS) and antioxidant signaling were examined. To determine the role of TXNRD in MR-induced cell death, TXNRD1 was inhibited by RNAi or the pan-TXNRD inhibitor auranofin, an antirheumatic agent. Metastatic and PDX TNBC mouse models were utilized to evaluate in vivo antitumor activity. RESULTS: MR rapidly and transiently increased ROS, depleted glutathione, and decreased the ratio of reduced glutathione/oxidized glutathione in TNBC cells. TXNRD1 mRNA and protein levels were induced by MR via a ROS-dependent mechanism mediated by the transcriptional regulators NRF2 and ATF4. MR dramatically sensitized TNBC cells to TXNRD1 silencing and the TXNRD inhibitor auranofin, as determined by crystal violet staining and caspase activity; these effects were suppressed by the antioxidant N-acetylcysteine. H-Ras-transformed MCF-10A cells, but not untransformed MCF-10A cells, were highly sensitive to the combination of auranofin and MR. Furthermore, dietary MR induced TXNRD1 expression in mammary tumors and enhanced the antitumor effects of auranofin in metastatic and PDX TNBC murine models. CONCLUSION: MR exposes a vulnerability of TNBC cells to the TXNRD inhibitor auranofin by increasing expression of its molecular target and creating a dependency on the thioredoxin pathway.


Assuntos
Tiorredoxina Dissulfeto Redutase , Neoplasias de Mama Triplo Negativas , Animais , Auranofina/farmacologia , Humanos , Metionina/metabolismo , Camundongos , Oxirredução , Tiorredoxina Redutase 1/genética , Tiorredoxina Redutase 1/metabolismo , Tiorredoxina Dissulfeto Redutase/genética , Tiorredoxina Dissulfeto Redutase/metabolismo , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/genética
4.
Mol Cancer Ther ; 19(11): 2278-2287, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32943543

RESUMO

The initiation of androgen-deprivation therapy (ADT) induces susceptibilities in prostate cancer cells that make them vulnerable to synergistic treatment and enhanced cell death. Senescence results in cell-cycle arrest, but cells remain viable. In this study, we investigated the mechanisms by which prostate cancer cells undergo senescence in response to ADT, and determined whether an FDA-approved antidiabetic drug metformin has a synergistic effect with ADT in prostate cancer both in vitro and in vivo Our results show that longer term exposure to ADT induced senescence associated with p16INK4a and/or p27kip2 induction. The activation of PI3K/AKT and inactivation of AMPK in senescent cells resulted in mTORC1 activation. In addition, the antiapoptotic protein XIAP expression was increased in response to ADT. The addition of metformin following ADT induced apoptosis, attenuated mTOR activation, reduced senescent cell number in vitro, and inhibited tumor growth in prostate cancer patient-derived xenograft models. This study suggests that combining ADT and metformin may be a feasible therapeutic approach to remove persistent prostate cancer cells after ADT.


Assuntos
Androgênios/metabolismo , Metabolismo Energético/efeitos dos fármacos , Metformina/farmacologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Mutações Sintéticas Letais , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Linhagem Celular Tumoral , Senescência Celular/genética , Modelos Animais de Doenças , Humanos , Masculino , Camundongos , Modelos Biológicos , Fosfatidilinositol 3-Quinases/metabolismo , Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Breast Cancer Res Treat ; 183(3): 549-564, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32696316

RESUMO

PURPOSE: Transformed cells are vulnerable to depletion of certain amino acids. Lysine oxidase (LO) catalyzes the oxidative deamination of lysine, resulting in lysine depletion and hydrogen peroxide production. Although LO has broad antitumor activity in preclinical models, the cytotoxic mechanisms of LO are poorly understood. METHODS: Triple (ER/PR/HER2)-negative breast cancer (TNBC) cells were treated with control media, lysine-free media or control media supplemented with LO and examined for cell viability, caspase activation, induction of reactive oxygen species (ROS) and antioxidant signaling. To determine the role of nuclear factor erythroid 2-related factor 2 (NRF2) and thioredoxin reductase-1 (TXNRD1) in LO-induced cell death, NRF2 and TXNRD1 were individually silenced by RNAi. Additionally, the pan-TXNRD inhibitor auranofin was used in combination with LO. RESULTS: LO activates caspase-independent cell death that is suppressed by necroptosis and ferroptosis inhibitors, which are inactive against lysine depletion, pointing to fundamental differences between LO and lysine depletion. LO rapidly induces ROS with a return to baseline levels within 24 h that coincides temporally with induction of TXNRD activity, the rate-limiting enzyme in the thioredoxin antioxidant pathway. ROS induction is required for LO-mediated cell death and NRF2-dependent induction of TXNRD1. Silencing NRF2 or TXNRD1 enhances the cytotoxicity of LO. The pan-TXNRD inhibitor auranofin is synergistic with LO against transformed breast epithelial cells, but not untransformed cells, underscoring the tumor-selectivity of this strategy. CONCLUSIONS: LO exposes a redox vulnerability of TNBC cells to TXNRD inhibition by rendering tumor cells dependent on the thioredoxin antioxidant pathway for survival.


Assuntos
Neoplasias de Mama Triplo Negativas , Antioxidantes/farmacologia , Humanos , Lisina , Estresse Oxidativo , Oxirredutases , Espécies Reativas de Oxigênio , Tiorredoxinas/genética , Tiorredoxinas/metabolismo , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/genética
6.
Cancer Lett ; 481: 24-31, 2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32246957

RESUMO

Brain metastases manifest the advanced stage of breast cancer disease with poor prognosis for patient survival. Recent reports demonstrate that some therapeutic agents can activate the expression of several breast cancer-associated genes, whose products are involved in the onset and development of brain metastases. In this study, we discovered a functional link between KISS1 and E-cadherin that could be observed in both primary brain metastatic lesions and paired cell lines, such as parental CN34TGL and MDA-MB-231 and their respective brain metastatic subclones CN34Brm2Ctgl and MDA-MB-231Br. Remarkably, expression of KISS1 and E-cadherin genes consistently showed an inverse correlation in all of the above cell/tissue types. While E-cadherin expression was strongly upregulated in metastatic clones isolated from blood and brain, the levels of this protein in parental MDA-MB-231 cell line was low. Furthermore, E-cadherin upregulation can be artificially induced in MDA-MB-231Br and CN34Brm2Ctgl cell populations by knocking down KISS1 expression directly or through overexpressing the miR345 mimic. In the aggregate, our data suggest that the tumor microenvironment, which controls breast cancer spreading via miR345-regulated KISS1 expression, might modulate metastatic spreading by a mechanism(s) involving upregulation of E-cadherin production.


Assuntos
Antígenos CD/genética , Neoplasias Encefálicas/genética , Neoplasias da Mama/genética , Caderinas/genética , Kisspeptinas/genética , MicroRNAs/genética , Adulto , Carcinoma Ductal de Mama/genética , Linhagem Celular Tumoral , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Genes Supressores de Tumor/fisiologia , Humanos , Pessoa de Meia-Idade , Ativação Transcricional/genética , Microambiente Tumoral/genética , Regulação para Cima/genética
7.
Breast Cancer Res Treat ; 175(1): 39-50, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30712196

RESUMO

PURPOSE: Many transformed cells and embryonic stem cells are dependent on the biosynthesis of the universal methyl-donor S-adenosylmethionine (SAM) from methionine by the enzyme MAT2A to maintain their epigenome. We hypothesized that cancer stem cells (CSCs) rely on SAM biosynthesis and that the combination of methionine depletion and MAT2A inhibition would eradicate CSCs. METHODS: Human triple (ER/PR/HER2)-negative breast carcinoma (TNBC) cell lines were cultured as CSC-enriched mammospheres in control or methionine-free media. MAT2A was inhibited with siRNAs or cycloleucine. The effects of methionine restriction and/or MAT2A inhibition on the formation of mammospheres, the expression of CSC markers (CD44hi/C24low), MAT2A and CSC transcriptional regulators, apoptosis induction and histone modifications were determined. A murine model of metastatic TNBC was utilized to evaluate the effects of dietary methionine restriction, MAT2A inhibition and the combination. RESULTS: Methionine restriction inhibited mammosphere formation and reduced the CD44hi/C24low CSC population; these effects were partly rescued by SAM. Methionine depletion induced MAT2A expression (mRNA and protein) and sensitized CSCs to inhibition of MAT2A (siRNAs or cycloleucine). Cycloleucine enhanced the effects of methionine depletion on H3K4me3 demethylation and suppression of Sox9 expression. Dietary methionine restriction induced MAT2A expression in mammary tumors, and the combination of methionine restriction and cycloleucine was more effective than either alone at suppressing primary and lung metastatic tumor burden in a murine TNBC model. CONCLUSIONS: Our findings point to SAM biosynthesis as a unique metabolic vulnerability of CSCs that can be targeted by combining methionine depletion with MAT2A inhibition to eradicate drug-resistant CSCs.


Assuntos
Neoplasias/metabolismo , Células-Tronco Neoplásicas/metabolismo , S-Adenosilmetionina/metabolismo , Animais , Apoptose , Antígeno CD24 , Linhagem Celular Tumoral , Modelos Animais de Doenças , Inativação Gênica , Histonas/metabolismo , Humanos , Receptores de Hialuronatos , Espectrometria de Massas , Metionina/metabolismo , Metionina Adenosiltransferase/genética , Metionina Adenosiltransferase/metabolismo , Camundongos , Neoplasias/genética , Neoplasias/patologia
8.
Methods Mol Biol ; 1866: 61-73, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30725408

RESUMO

We have developed a novel therapeutic paradigm ("metabolic priming") for cancer whereby restriction of the essential amino acid methionine activates a number of cell-stress-response pathways that can be selectively targeted to enhance the therapeutic impact of methionine restriction. One example of metabolic priming is the combination of methionine restriction with proapoptotic TRAIL receptor-2 (TRAIL-R2) agonists. Methionine restriction enhances the cell surface expression of TRAIL-R2 selectively in transformed breast epithelial cells and renders them more susceptible to cell death induction by TRAIL-R2 agonists in cellular and murine models of breast cancer. This methods review focuses on preclinical models of breast cancer to investigate metabolic priming by methionine restriction. Multiple cell-based methods are detailed to measure cell viability, cell survival, caspase activity, apoptosis, and matrix detachment-induced cell death (anoikis). In addition, we describe an orthotopic model of metastatic breast cancer that utilizes mCherry-fluorescently-labeled human breast cancer cells. This model captures the entire metastatic cascade from the mammary gland to the lung and mimics key features of the human disease. These breast-cancer models can be readily adapted to other tumor types. Overall, we provide a stepwise, translationally-relevant approach to study metabolic priming in the context of cancer.


Assuntos
Neoplasias da Mama/patologia , Metionina/deficiência , Animais , Anexina A5/metabolismo , Anoikis , Caspase 3/metabolismo , Caspase 7/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular , Modelos Animais de Doenças , Feminino , Fluorescência , Humanos , Camundongos , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo
9.
Autophagy ; 13(11): 1905-1923, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28981380

RESUMO

Formation of metastases, also known as cancer dissemination, is an important stage of breast cancer (BrCa) development. KISS1 expression is associated with inhibition of metastases development. Recently we have demonstrated that BrCa metastases to the brain exhibit low levels of KISS1 expression at both mRNA and protein levels. By using multicolor immunofluorescence and coculture techniques here we show that normal adult astrocytes in the brain are capable of promoting metastatic transformation of circulating breast cancer cells localized to the brain through secretion of chemokine CXCL12. The latter was found in this study to downregulate KISS1 expression at the post-transcriptional level via induction of microRNA-345 (MIR345). Furthermore, we demonstrated that ectopic expression of KISS1 downregulates ATG5 and ATG7, 2 key modulators of autophagy, and works concurrently with autophagy inhibitors, thereby implicating autophagy in the mechanism of KISS1-mediated BrCa metastatic transformation. We also found that expression of KISS1 in human breast tumor specimens inversely correlates with that of MMP9 and IL8, implicated in the mechanism of metastatic invasion, thereby supporting the role of KISS1 as a potential regulator of BrCa metastatic invasion in the brain. This conclusion is further supported by the ability of KISS1, ectopically overexpressed from an adenoviral vector in MDA-MB-231Br cells with silenced expression of the endogenous gene, to revert invasive phenotype of those cells. Taken together, our results strongly suggest that human adult astrocytes can promote brain invasion of the brain-localized circulating breast cancer cells by upregulating autophagy signaling pathways via the CXCL12-MIR345- KISS1 axis.


Assuntos
Astrócitos/patologia , Autofagia , Neoplasias Encefálicas/secundário , Neoplasias da Mama/patologia , Carcinoma Ductal de Mama/secundário , Quimiocina CXCL12/metabolismo , Kisspeptinas/metabolismo , MicroRNAs/metabolismo , Adulto , Idoso , Animais , Astrócitos/metabolismo , Proteína 5 Relacionada à Autofagia/metabolismo , Proteína 7 Relacionada à Autofagia/metabolismo , Linhagem Celular Tumoral , Feminino , Humanos , Interleucina-8/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Microglia/metabolismo , Microglia/patologia , Pessoa de Meia-Idade , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Breast Cancer Res Treat ; 163(3): 435-447, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28324269

RESUMO

PURPOSE: Despite robust antitumor activity in diverse preclinical models, TNF-related apoptosis-inducing ligand (TRAIL) receptor agonists have not demonstrated efficacy in clinical trials, underscoring the need to identify agents that enhance their activity. We postulated that the metabolic stress induced by the diabetes drug metformin would sensitize breast cancer cells to TRAIL receptor agonists. METHODS: Human triple (estrogen receptor, progesterone receptor, and HER2)-negative breast cancer (TNBC) cell lines were treated with TRAIL receptor agonists (monoclonal antibodies or TRAIL peptide), metformin, or the combination. The effects on cell survival, caspase activation, and expression of TRAIL receptors and the antiapoptotic protein XIAP were determined. In addition, XIAP was silenced by RNAi in TNBC cells and the effects on sensitivity to TRAIL were determined. The antitumor effects of metformin, TRAIL, or the combination were evaluated in an orthotopic model of metastatic TNBC. RESULTS: Metformin sensitized diverse TNBC cells to TRAIL receptor agonists. Metformin selectively enhanced the sensitivity of transformed breast epithelial cells to TRAIL receptor agonist-induced caspase activation and apoptosis with little effect on untransformed breast epithelial cells. These effects of metformin were accompanied by robust reductions in the protein levels of XIAP, a negative regulator of TRAIL-induced apoptosis. Silencing XIAP in TNBC cells mimicked the TRAIL-sensitizing effects of metformin. Metformin also enhanced the antitumor effects of TRAIL in a metastatic murine TNBC model. CONCLUSIONS: Our findings indicate that metformin enhances the activity of TRAIL receptor agonists, thereby supporting the rationale for additional translational studies combining these agents.


Assuntos
Metformina/administração & dosagem , Ligante Indutor de Apoptose Relacionado a TNF/genética , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/genética , Animais , Anticorpos Monoclonais/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Apoptose/efeitos dos fármacos , Caspases/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Humanos , Camundongos , Interferência de RNA , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/agonistas , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/genética , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/patologia , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/antagonistas & inibidores , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Pharmacol Ther ; 160: 1-10, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26820756

RESUMO

αB-crystallin is a widely expressed member of the small heat shock protein family that protects cells from stress by its dual function as a molecular chaperone to preserve proteostasis and as a cell death antagonist that negatively regulates components of the conserved apoptotic cell death machinery. Deregulated expression of αB-crystallin occurs in a broad array of solid tumors and has been linked to tumor progression and poor clinical outcomes. This review will focus on new insights into the molecular mechanisms by which oncogenes, oxidative stress, matrix detachment and other tumor microenvironmental stressors deregulate αB-crystallin expression. We will also review accumulating evidence pointing to an essential role for αB-crystallin in the multi-step metastatic cascade whereby tumor cells colonize distant organs by circumventing a multitude of barriers to cell migration and survival. Finally, we will evaluate emerging strategies to therapeutically target αB-crystallin and/or interacting proteins to selectively activate apoptosis and/or derail the metastatic cascade in an effort to improve outcomes for patients with metastatic disease.


Assuntos
Cristalinas/metabolismo , Chaperonas Moleculares/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia , Apoptose/fisiologia , Humanos , Metástase Neoplásica/patologia , Estresse Oxidativo/fisiologia , Microambiente Tumoral/fisiologia
12.
Clin Cancer Res ; 21(12): 2780-91, 2015 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-25724522

RESUMO

PURPOSE: Many neoplasms are vulnerable to methionine deficiency by mechanisms that are poorly understood. Because gene profiling studies have revealed that methionine depletion increases TNF-related apoptosis-inducing ligand receptor-2 (TRAIL-R2) mRNA, we postulated that methionine stress sensitizes breast cancer cells to proapoptotic TRAIL-R2 agonists. EXPERIMENTAL DESIGN: Human triple (ER/PR/HER2)-negative breast carcinoma cell lines were cultured in control or methionine-free media. The effects of methionine depletion on TRAIL receptor expression and sensitivity to chemotherapy or a humanized agonistic TRAIL-R2 monoclonal antibody (lexatumumab) were determined. The melanoma-associated antigen MAGED2 was silenced to delineate its functional role in sensitizing TNBC cells to methionine stress. An orthotopic TNBC model was utilized to evaluate the effects of dietary methionine deficiency, lexatumumab, or the combination. RESULTS: Methionine depletion sensitized TNBC cells to lexatumumab-induced caspase activation and apoptosis by increasing TRAIL-R2 mRNA and cell surface expression. MCF-10A cells transformed by oncogenic H-Ras, but not untransformed cells, and matrix-detached TNBC cells were highly sensitive to the combination of lexatumumab and methionine depletion. Proteomics analyses revealed that MAGED2, which has been reported to reduce TRAIL-R2 expression, was suppressed by methionine stress. Silencing MAGED2 recapitulated features of methionine deprivation, including enhanced mRNA and cell surface expression of TRAIL receptors and increased sensitivity to TRAIL receptor agonists. Dietary methionine deprivation enhanced the antitumor effects of lexatumumab in an orthotopic metastatic TNBC model. CONCLUSIONS: Methionine depletion exposes a targetable defect in TNBC cells by increasing TRAIL-R2 expression. Our findings provide the foundation for a clinical trial combining dietary methionine restriction and TRAIL-R2 agonists. Clin Cancer Res; 21(12); 2780-91. ©2015 AACR.


Assuntos
Metionina/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Neoplasias de Mama Triplo Negativas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Anticorpos Monoclonais/farmacologia , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Apoptose/efeitos dos fármacos , Caspases/metabolismo , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Resistencia a Medicamentos Antineoplásicos , Feminino , Inativação Gênica , Humanos , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/agonistas , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/genética , Neoplasias de Mama Triplo Negativas/genética
13.
Nanoscale ; 7(16): 7178-83, 2015 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-25820516

RESUMO

Gating of mesoporous silica nanoparticles (MSNs) with the stimuli-responsive poly(ß-amino ester) has been achieved. This hybrid nanocarrier releases doxorubicin (DOX) under acidic conditions or in the presence of porcine liver esterase. The DOX loaded poly(ß-amino ester)-capped MSNs reduce cell viability when tested on MDA-MB-231 human breast cancer cells.


Assuntos
Portadores de Fármacos/química , Esterases/metabolismo , Nanopartículas/química , Polímeros/química , Dióxido de Silício/química , Animais , Antibióticos Antineoplásicos/química , Antibióticos Antineoplásicos/toxicidade , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Doxorrubicina/química , Doxorrubicina/toxicidade , Esterases/química , Humanos , Concentração de Íons de Hidrogênio , Porosidade , Suínos
14.
Behav Brain Res ; 276: 118-29, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24786329

RESUMO

Antioxidant enzymes and lipid peroxidation in the brain are involved in neuropsychiatric pathologies, including depression. 14- or 28-day chronic stress model induced a depressive syndrome defined by lowered reward sensitivity in C57BL/6J mice and changed gene expression of peroxidation enzymes as shown in microarray assays. We studied how susceptibility or resilience to anhedonia is related to lipid peroxidation in the prefrontal cortex (PFC). With 14-day stress, a comparison of the activities of catalase (CAT), superoxide dismutase (SOD), glutathione peroxidase (GPX) and accumulation of malondialdehyde (MDA) revealed a decrease of the first two measures in susceptible, but not in resilient animals or in stressed mice chronically dosed with imipramine (7mg/kg/day). Acute stress elevated activity of CAT and SOD and dynamics of MDA accumulation in the PFC that was prevented by imipramine (30mg/kg). 28-day stress evoked anhedonia lasting two but not five weeks while behavioural invigoration was detected at the latter time point in anhedonic but not non-anhedonic mice; enhanced aggressive traits were observed in both groups. After two weeks of a stress-free period, CAT and SOD activity levels in the PFC were reduced in anhedonic animals; after five weeks, only CAT was diminished. Thus, in the present chronic stress depression paradigm, lasting alterations in brain peroxidation occur not only during anhedonia but also in the recovery period and are accompanied by behavioural abnormalities in mice. This mimics behavioural and neurochemical deficits observed in depressed patients during remission which could be used to develop remedies preventing their relapse.


Assuntos
Anedonia , Regulação para Baixo , Peroxidação de Lipídeos/genética , Córtex Pré-Frontal/enzimologia , Estresse Psicológico/enzimologia , Agressão/efeitos dos fármacos , Anedonia/efeitos dos fármacos , Animais , Catalase/genética , Catalase/metabolismo , Preferências Alimentares , Perfilação da Expressão Gênica , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Glutationa Peroxidase/genética , Glutationa Peroxidase/metabolismo , Hipocampo/metabolismo , Imipramina/farmacologia , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Malondialdeído/metabolismo , Camundongos , Atividade Motora/efeitos dos fármacos , Córtex Pré-Frontal/efeitos dos fármacos , Resiliência Psicológica , Estresse Psicológico/genética , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo
15.
Proc Natl Acad Sci U S A ; 111(48): 17104-9, 2014 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-25404304

RESUMO

Metastasis portends a poor prognosis for cancer patients. Primary tumor cells disseminate through the bloodstream before the appearance of detectable metastatic lesions. The analysis of cancer cells in blood­so-called circulating tumor cells (CTCs)­may provide unprecedented opportunities for metastatic risk assessment and investigation. NanoFlares are nanoconstructs that enable live-cell detection of intracellular mRNA. NanoFlares, when coupled with flow cytometry, can be used to fluorescently detect genetic markers of CTCs in the context of whole blood. They allow one to detect as few as 100 live cancer cells per mL of blood and subsequently culture those cells. This technique can also be used to detect CTCs in a murine model of metastatic breast cancer. As such, NanoFlares provide, to our knowledge, the first genetic-based approach for detecting, isolating, and characterizing live cancer cells from blood and may provide new opportunities for cancer diagnosis, prognosis, and personalized therapy.


Assuntos
Carbocianinas/química , DNA Antissenso/química , Ouro/química , Nanopartículas Metálicas/química , Células Neoplásicas Circulantes/química , Sequência de Bases , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/sangue , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Caderinas/genética , Caderinas/metabolismo , Carbocianinas/metabolismo , Linhagem Celular Tumoral , DNA Antissenso/genética , DNA Antissenso/metabolismo , Feminino , Fibronectinas/genética , Fibronectinas/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Microscopia de Fluorescência , Nanotecnologia/métodos , Células Neoplásicas Circulantes/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transplante Heterólogo , Vimentina/genética , Vimentina/metabolismo , Proteína Vermelha Fluorescente
16.
Clin Cancer Res ; 20(1): 56-67, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24132917

RESUMO

PURPOSE: Basal-like breast tumors are typically (ER/PR/HER2) triple-negative and are associated with a high incidence of brain metastases and poor clinical outcomes. The molecular chaperone αB-crystallin is predominantly expressed in triple-negative breast cancer (TNBC) and contributes to an aggressive tumor phenotype in preclinical models. We investigated the potential role of αB-crystallin in brain metastasis in TNBCs. EXPERIMENTAL DESIGN: αB-crystallin expression in primary breast carcinomas and brain metastases was analyzed by immunohistochemistry among patients with breast cancer with brain metastases. αB-crystallin was overexpressed or silenced in two different TNBC cell lines. The effects on cell adhesion to human brain microvascular endothelial cells (HBMEC) or extracellular matrix proteins, transendothelial migration, and transmigration across a HBMEC/astrocyte coculture blood-brain barrier (BBB) model were examined. In addition, the effects of overexpressing or silencing αB-crystallin on brain metastasis in vivo were investigated using orthotopic TNBC models. RESULTS: In a cohort of women with breast cancer brain metastasis, αB-crystallin expression in primary breast carcinomas was associated with poor overall survival and poor survival after brain metastasis, even among patients with TNBC. Stable overexpression of αB-crystallin in TNBC cells enhanced adhesion to HBMECs, transendothelial migration, and BBB transmigration in vitro, whereas silencing αB-crystallin inhibited these events. αB-crystallin promoted adhesion of TNBC cells to HBMECs, at least in part, through an α3ß1 integrin-dependent mechanism. αB-crystallin overexpression promoted brain metastasis, whereas silencing αB-crystallin inhibited brain metastasis in orthotopic TNBC models. CONCLUSION: αB-crystallin is a novel regulator of brain metastasis in TNBC and represents a potential biomarker and drug target for this aggressive disease.


Assuntos
Neoplasias Encefálicas/metabolismo , Neoplasias de Mama Triplo Negativas/metabolismo , Cadeia B de alfa-Cristalina/fisiologia , Animais , Barreira Hematoencefálica/patologia , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/secundário , Adesão Celular , Linhagem Celular Tumoral , Endotélio Vascular/patologia , Feminino , Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Estimativa de Kaplan-Meier , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Transplante de Neoplasias , Migração Transendotelial e Transepitelial , Neoplasias de Mama Triplo Negativas/mortalidade , Neoplasias de Mama Triplo Negativas/patologia
17.
J Am Chem Soc ; 135(31): 11603-13, 2013 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-23815127

RESUMO

Ruthenium(II) polypyridyl complexes have emerged both as promising probes of DNA structure and as anticancer agents because of their unique photophysical and cytotoxic properties. A key consideration in the administration of those therapeutic agents is the optimization of their chemical reactivities to allow facile attack on the target sites, yet avoid unwanted side effects. Here, we present a drug delivery platform technology, obtained by grafting the surface of mesoporous silica nanoparticles (MSNPs) with ruthenium(II) dipyridophenazine (dppz) complexes. This hybrid nanomaterial displays enhanced luminescent properties relative to that of the ruthenium(II) dppz complex in a homogeneous phase. Since the coordination between the ruthenium(II) complex and a monodentate ligand linked covalently to the nanoparticles can be cleaved under irradiation with visible light, the ruthenium complex can be released from the surface of the nanoparticles by selective substitution of this ligand with a water molecule. Indeed, the modified MSNPs undergo rapid cellular uptake, and after activation with light, the release of an aqua ruthenium(II) complex is observed. We have delivered, in combination, the ruthenium(II) complex and paclitaxel, loaded in the mesoporous structure, to breast cancer cells. This hybrid material represents a promising candidate as one of the so-called theranostic agents that possess both diagnostic and therapeutic functions.


Assuntos
Antineoplásicos/administração & dosagem , Nanopartículas/química , Compostos Organometálicos/administração & dosagem , Paclitaxel/administração & dosagem , Dióxido de Silício/química , Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , DNA/metabolismo , Sistemas de Liberação de Medicamentos , Feminino , Humanos , Luz , Modelos Moleculares , Compostos Organometálicos/farmacologia , Paclitaxel/farmacologia
18.
Breast Cancer Res Treat ; 138(2): 415-25, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23471649

RESUMO

The retinoblastoma (Rb) tumor suppressor gene is frequently inactivated in cancer, resulting in deregulated activation of E2F transcription factors, which promote S-phase entry, p53-dependent and p53-independent apoptosis. Transformed cells evade p53-dependent apoptosis initiated by Rb inactivation by TP53 mutation. However, the mechanisms by which cancer cells circumvent p53-independent apoptosis in this context are poorly understood. Because Rb inactivation primes cells for apoptosis by p53-independent induction of procaspases, we postulated that αB-crystallin, an inhibitor of procaspase-3 activation, would suppress caspase activation in cells with combined Rb and p53 inactivation. Notably, αB-crystallin is commonly expressed in ER/PR/HER2 "triple-negative" breast carcinomas characterized by frequent Rb loss and TP53 mutation. We report that αB-crystallin (-/-) knock out (KO) MEFs immortalized by dominant negative (DN) p53 are resistant to transformation by the adenovirus E1A oncoprotein, which inactivates Rb, while wild-type (WT) MEFs are readily transformed by DN p53 and E1A. αB-crystallin (-/-) KO MEFs stably expressing DN p53 and E1A were more sensitive to chemotherapy-induced caspase-3 activation and apoptosis than the corresponding WT MEFs, despite comparable induction of procaspases by E1A. Similarly, silencing Rb in WT and αB-crystallin (-/-) KO MEFs immortalized by DN p53 increased procaspase levels and sensitized αB-crystallin (-/-) KO MEFs to chemotherapy. Furthermore, silencing αB-crystallin in triple-negative breast cancer cells, which lack Rb and express mutant p53, enhanced chemotherapy sensitivity compared to non-silencing controls. Our results indicate that αB-crystallin inhibits caspase activation in cells primed for apoptosis by Rb inactivation and plays a novel oncogenic role in the context of combined Rb and p53 inactivation.


Assuntos
Apoptose , Caspase 3/metabolismo , Transformação Celular Neoplásica/metabolismo , Proteína do Retinoblastoma/genética , Cadeia B de alfa-Cristalina/genética , Proteínas E1A de Adenovirus/metabolismo , Animais , Antineoplásicos/farmacologia , Proliferação de Células , Células Cultivadas , Doxorrubicina/farmacologia , Ativação Enzimática , Células Epiteliais/metabolismo , Feminino , Técnicas de Silenciamento de Genes , Humanos , Glândulas Mamárias Humanas/patologia , Camundongos , Camundongos Knockout , Paclitaxel/farmacologia , RNA Interferente Pequeno/genética , Proteína do Retinoblastoma/metabolismo , Proteína Supressora de Tumor p53/genética , Cadeia B de alfa-Cristalina/metabolismo
19.
Biomed Res Int ; 2013: 565218, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24386638

RESUMO

Central thyroid hormone signaling is important in brain function/dysfunction, including affective disorders and depression. In contrast to 3,3',5-triiodo-L-thyronine (T3), the role of 3,5-diiodo-L-thyronine (T2), which until recently was considered an inactive metabolite of T3, has not been studied in these pathologies. However, both T3 and T2 stimulate mitochondrial respiration, a factor counteracting the pathogenesis of depressive disorder, but the cellular origins in the CNS, mechanisms, and kinetics of the cellular action for these two hormones are distinct and independent of each other. Here, Illumina and RT PCR assays showed that hippocampal gene expression of deiodinases 2 and 3, enzymes involved in thyroid hormone regulation, is increased in resilience to stress-induced depressive syndrome and after antidepressant treatment in mice that might suggest elevated T2 and T3 turnover in these phenotypes. In a separate experiment, bolus administration of T2 at the doses 750 and 1,500 mcg/kg but not 250 mcg/kg in naive mice reduced immobility in a two-day tail suspension test in various settings without changing locomotion or anxiety. This demonstrates an antidepressant-like effect of T2 that could be exploited clinically. In a wider context, the current study suggests important central functions of T2, whose biological role only lately is becoming to be elucidated.


Assuntos
Transtorno Depressivo/metabolismo , Di-Iodotironinas/genética , Iodeto Peroxidase/genética , Animais , Transtorno Depressivo/patologia , Di-Iodotironinas/metabolismo , Di-Iodotironinas/farmacologia , Regulação Enzimológica da Expressão Gênica , Hipocampo/metabolismo , Hipocampo/patologia , Iodeto Peroxidase/biossíntese , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tri-Iodotironina/genética , Tri-Iodotironina/metabolismo , Iodotironina Desiodinase Tipo II
20.
BMC Neurosci ; 13: 110, 2012 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-22989159

RESUMO

BACKGROUND: A number of epidemiological studies have established a link between insulin resistance and the prevalence of depression. The occurrence of depression was found to precede the onset of diabetes and was hypothesized to be associated with inherited inter-related insufficiency of the peripheral and central insulin receptors. Recently, dicholine succinate, a sensitizer of the neuronal insulin receptor, was shown to stimulate insulin-dependent H2O2 production of the mitochondrial respiratory chain leading to an enhancement of insulin receptor autophosphorylation in neurons. As such, this mechanism can be a novel target for the elevation of insulin signaling. RESULTS: Administration of DS (25 mg/kg/day, intraperitoneal) in CD1 mice for 7 days prior to the onset of stress procedure, diminished manifestations of anhedonia defined in a sucrose test and behavioral despair in the forced swim test. Treatment with dicholine succinate reduced the anxiety scores of stressed mice in the dark/light box paradigm, precluded stress-induced decreases of long-term contextual memory in the step-down avoidance test and hippocampal gene expression of IGF2. CONCLUSIONS: Our data suggest that dicholine succinate has an antidepressant-like effect, which might be mediated via the up-regulation of hippocampal expression of IGF2, and implicate the neuronal insulin receptor in the pathogenesis of stress-induced depressive syndrome.


Assuntos
Antidepressivos/uso terapêutico , Depressão/tratamento farmacológico , Fator de Crescimento Insulin-Like II/fisiologia , Transtornos da Memória/tratamento farmacológico , Estresse Psicológico/tratamento farmacológico , Succinilcolina/uso terapêutico , Animais , Antidepressivos/farmacologia , Ansiedade/complicações , Ansiedade/tratamento farmacológico , Ansiedade/psicologia , Depressão/complicações , Depressão/psicologia , Modelos Animais de Doenças , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Imipramina/farmacologia , Imipramina/uso terapêutico , Fator de Crescimento Insulin-Like II/biossíntese , Masculino , Transtornos da Memória/complicações , Transtornos da Memória/psicologia , Camundongos , Camundongos Endogâmicos ICR , Neurônios/metabolismo , Receptor de Insulina/metabolismo , Estresse Psicológico/complicações , Estresse Psicológico/psicologia , Succinilcolina/farmacologia , Regulação para Cima/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA