Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 229
Filtrar
1.
Int J Mol Sci ; 25(11)2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38892407

RESUMO

Breast cancer is influenced by factors such as diet, a sedentary lifestyle, obesity, and postmenopausal status, which are all linked to prolonged hormonal and inflammatory exposure. Physical activity offers protection against breast cancer by modulating hormones, immune responses, and oxidative defenses. This study aimed to assess how a prolonged high-fat diet (HFD) affects the effectiveness of physical activity in preventing and managing mammary tumorigenesis. Ovariectomised C57BL/6 mice were provided with an enriched environment to induce spontaneous physical activity while being fed HFD. After 44 days (short-term, ST HFD) or 88 days (long-term, LT HFD), syngenic EO771 cells were implanted into mammary glands, and tumour growth was monitored until sacrifice. Despite similar physical activity and food intake, the LT HFD group exhibited higher visceral adipose tissue mass and reduced skeletal muscle mass. In the tumour microenvironment, the LT HFD group showed decreased NK cells and TCD8+ cells, with a trend toward increased T regulatory cells, leading to a collapse of the T8/Treg ratio. Additionally, the LT HFD group displayed decreased tumour triglyceride content and altered enzyme activities indicative of oxidative stress. Prolonged exposure to HFD was associated with tumour growth despite elevated physical activity, promoting a tolerogenic tumour microenvironment. Future studies should explore inter-organ exchanges between tumour and tissues.


Assuntos
Dieta Hiperlipídica , Camundongos Endogâmicos C57BL , Condicionamento Físico Animal , Microambiente Tumoral , Animais , Dieta Hiperlipídica/efeitos adversos , Feminino , Camundongos , Estresse Oxidativo , Carcinogênese , Neoplasias Mamárias Experimentais/patologia , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/prevenção & controle , Linhagem Celular Tumoral , Neoplasias Mamárias Animais/patologia , Neoplasias Mamárias Animais/metabolismo , Neoplasias Mamárias Animais/prevenção & controle , Gordura Intra-Abdominal/metabolismo , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo
2.
Sci Rep ; 11(1): 23121, 2021 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-34848739

RESUMO

In our previous study, immunoinformatic tools were used to design a novel multiepitope cancer vaccine based on the most immunodominant regions of BORIS cancer-testis antigen. The final vaccine construct was an immunogenic, non-allergenic, and stable protein consisted of multiple cytotoxic T lymphocytes epitopes, IFN-γ inducing epitopes, and B cell epitopes according to bioinformatic analyzes. Herein, the DNA sequence of the final vaccine construct was placed into the pcDNA3.1 vector as a DNA vaccine (pcDNA3.1-VAC). Also, the recombinant multiepitope peptide vaccine (MPV) was produced by a transfected BL21 E. coli strain using a recombinant pET-28a vector and then, purified and screened by Fast protein liquid chromatography technique (FPLC) and Western blot, respectively. The anti-tumor effects of prophylactic co-immunization with these DNA and protein cancer vaccines were evaluated in the metastatic non-immunogenic 4T1 mammary carcinoma in BALB/c mice. Co-immunization with the pcDNA3.1-VAC and MPV significantly (P < 0.001) increased the serum levels of the MPV-specific IgG total, IgG2a, and IgG1. The splenocytes of co-immunized mice exhibited a significantly higher efficacy to produce interleukin-4 and interferon-γ and proliferation in response to MPV in comparison with the control. The prophylactic co-immunization regime caused significant breast tumors' growth inhibition, tumors' weight decrease, inhibition of metastasis formation, and enlarging tumor-bearing mice survival time, without any considerable side effects. Taking together, this cancer vaccine can evoke strong immune response against breast tumor and inhibits its growth and metastasis.


Assuntos
Vacinas Anticâncer/imunologia , Proteínas de Ligação a DNA/biossíntese , Neoplasias Mamárias Animais/imunologia , Neoplasias Mamárias Animais/prevenção & controle , Animais , Vacinas Anticâncer/química , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células , Cromatografia Líquida , Biologia Computacional , Simulação por Computador , Modelos Animais de Doenças , Epitopos , Feminino , Imunidade Humoral , Interferon gama/química , Neoplasias Mamárias Animais/terapia , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/prevenção & controle , Neoplasias Mamárias Experimentais/terapia , Camundongos , Camundongos Endogâmicos BALB C , Metástase Neoplásica , Linfócitos T Citotóxicos/imunologia , Vacinas de Subunidades Antigênicas
3.
J Nutr ; 151(1): 73-84, 2021 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-33188406

RESUMO

BACKGROUND: Aberrations in the regulation of cell proliferation perturb cellular homeostasis and lead to malignancies in which dysregulation of the cell cycle and suppressed apoptosis are 2 common phenomena. Combinatorial nutritional approaches could be efficacious in ameliorating these aberrations. OBJECTIVES: We sought to investigate the effect of dietary broccoli sprouts (BSp) and green tea polyphenol (GTP) administration on cell cycle progression and apoptosis in mammary tumors. METHODS: Forty female HER2/neu transgenic mice were randomly divided into 4 groups and treated with control, 26% BSp (wt:wt) in food, 0.5% GTPs (wt:vol) in drinking water, or combined BSp and GTPs from dams' conception until their pups were killed at 29 wk of age. Pups' tumor growth was monitored weekly for 27 wk. Tumor cell cycle- and apoptosis-related protein expression was measured. Data were analyzed with 2-factor or 3-factor (repeated-measures) ANOVA. RESULTS: Compared with the control group, BSp and/or GTPs decreased tumor incidence (P < 0.05) and combined BSp and GTPs synergistically [combination index (CIn) < 1] reduced tumor volume over time (P-time < 0.01). BSp and/or GTPs upregulated the expression of phosphatase and tension homolog, P16, and P53 (P < 0.05) and downregulated myelocytomatosis oncogene, Bmi1 polycomb ring finger oncogene, and telomerase reverse transcriptase (P < 0.05) compared with the control group. Combined BSp and GTPs synergistically (CIn < 1) downregulated the expression of cyclin B1, D1, and E1 and cyclin-dependent kinase 1, 2, and 4 (P < 0.05) compared with the control group. Moreover, combined BSp and GTPs induced apoptosis by regulating Bcl-2-associated X protein and B-cell lymphoma 2 (P < 0.05). BSp and/or GTPs also reduced the expression of DNA methyltransferase 1, 3A, and 3B and histone deacetylase 1 compared with the control group (P < 0.05). CONCLUSIONS: Collectively, lifelong BSp and GTP administration can prevent estrogen receptor-negative mammary tumorigenesis through cell cycle arrest and inducing apoptosis in HER2/neu mice.


Assuntos
Brassica , Neoplasias Mamárias Animais/prevenção & controle , Polifenóis , Receptores de Estrogênio/metabolismo , Chá/química , Animais , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genótipo , Camundongos , Camundongos Transgênicos , Distribuição Aleatória , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Receptores de Estrogênio/genética
4.
Nutrients ; 12(8)2020 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-32751496

RESUMO

Elevated levels of estrogen are a risk factor for breast cancer. In addition to inducing DNA damage, estrogens can enhance cell proliferation as well as modulate fatty acid metabolism that collectively contributes to mammary tumorigenesis. Sulforaphane (SFN) is an isothiocyanate derived from broccoli that is currently under evaluation in multiple clinical trials for prevention of several diseases, including cancer. Previous studies showed that SFN suppressed DNA damage and lipogenesis pathways. Therefore, we hypothesized that administering SFN to animals that are co-exposed to 17ß-estradiol (E2) would prevent mammary tumor formation. In our study, 4-6 week old female August Copenhagen Irish rats were implanted with slow-release E2 pellets (3 mg x 3 times) and gavaged 3x/week with either vehicle or 100 µmol/kg SFN for 56 weeks. SFN-treated rats were protected significantly against mammary tumor formation compared to vehicle controls. Mammary glands of SFN-treated rats showed decreased DNA damage while serum free fatty acids and triglyceride species were 1.5 to 2-fold lower in SFN-treated rats. Further characterization also showed that SFN diminished expression of enzymes involved in mammary gland lipogenesis. This study indicated that SFN protects against breast cancer development through multiple potential mechanisms in a clinically relevant hormonal carcinogenesis model.


Assuntos
Anticarcinógenos/farmacologia , Isotiocianatos/farmacologia , Neoplasias Mamárias Animais/prevenção & controle , Neoplasias Mamárias Experimentais/prevenção & controle , Animais , Proliferação de Células/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Estradiol , Ácidos Graxos/sangue , Feminino , Lipogênese/efeitos dos fármacos , Neoplasias Mamárias Animais/induzido quimicamente , Neoplasias Mamárias Experimentais/induzido quimicamente , Ratos , Sulfóxidos , Triglicerídeos/sangue
5.
Pesqui. vet. bras ; 40(6): 466-473, June 2020. tab, graf, ilus
Artigo em Inglês | VETINDEX, LILACS | ID: biblio-1135647

RESUMO

Mammary tumors in female dogs are the most frequent and corresponds to half of the canine tumors. The objectives of this study were to determine the risk factors associated with the occurrence of mammary tumors in female dogs and to evaluate the macroscopic characteristics of these neoformations, using 386 dogs from the "Outubro Rosa Pets" events done within the cities of Uberlândia and Patos de Minas, Minas Gerais State, Brazil, in 2015 (n=194), 2016 (n=105) and 2017 (n=87). For the determination of risk factors, the binary logistic regression test (P<0.05) was performed. The occurrence of mammary tumors was 23.6% (91/386). The significant risk factors identified were increased age (P<0.001), overweight (P=0.048) and non-castration (P<0.001) with a chance of, respectively, 1.6, 2.3 and 9.3 times for the development of mammary tumors. In dogs with mammary tumors (n=91), 153 lesions were present, of which 39 female dogs had two or more lesions (42.8%). Most of the lesions were at the caudal abdominal (M4) and inguinal (M5) mammary glands (60.13%, 92/153). Relative to the size of the lesions, it was observed that in 78% of the female dogs the lesions were determined asT1 (<3cm), 16.5% were T2 (3-5cm) and 5.5% T3 (>5cm). At least 15.4% (14/91) of the dogs had one of the regional lymph nodes increased. In conclusion, the occurrence of mammary tumors in the evaluated population was 23.6% and that age, overweight and non-realization of ovariohysterectomy are risk factors associated with the development of mammary tumors.(AU)


Em cadelas os tumores mamários são os mais frequentes e correspondem a aproximadamente metade dos tumores em cães. Este estudo teve os objetivos de determinar os fatores de risco envolvidos na ocorrência de tumores mamários em cadelas e avaliar as características macroscópicas destas neoformações, utilizando 386 cadelas do evento "Outubro Rosa Pets" nos municípios de Uberlândia e Patos de Minas, Minas Gerais, Brasil, em 2015 (n=194), 2016 (n=105) e 2017 (n=87). Para a determinação dos fatores de risco utilizou-se o teste de Regressão logística binária (P<0,05). A ocorrência de tumores mamários foi de 23,6% (91/386). Os fatores de risco significativos identificados foram aumento da idade (P<0,001), sobrepeso (P=0,048) e não-castração (P<0,001) com a chance de, respectivamente, 1,6, 2,3 e 9,3 vezes de desenvolvimento de tumores mamários. Nas cadelas com tumores mamários (n=91), constatou-se a presença de 153 lesões, sendo que 39 cadelas apresentaram duas ou mais lesões (42,8%). A maioria das lesões localizaram-se nas mamas abdominais caudais (M4) e inguinais (M5) (60,13%; 92/153). Em relação ao tamanho das lesões, observou-se que 78% das cadelas eram T1 (<3cm), 16,5% T2 (3-5cm) e 5,5% T3 (>5cm). Pelo menos 15,4% (14/91) das cadelas apresentaram um dos linfonodos regionais aumentados. Conclui-se que a ocorrência dos tumores mamários na população avaliada foi de 23,6% e que a idade, sobrepeso e não ovariohisterectomia são fatores de risco para o desenvolvimento de tumores mamários.(AU)


Assuntos
Animais , Feminino , Cães , Neoplasias Mamárias Animais/etiologia , Neoplasias Mamárias Animais/epidemiologia , Doenças do Cão , Neoplasias Mamárias Animais/prevenção & controle , Fatores de Risco
6.
Oxid Med Cell Longev ; 2020: 4014838, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32184916

RESUMO

Currently, one of the central problems in cancer management is the relapse of disease following conventional treatments, yet few therapeutic agents targeting resistance and tolerance exist. Propolis is known as a healing agent since ancient times. Therefore, over time, its curative properties have kept the interest of scientists, thus leading permanently to investigations of its other possible undiscovered effects. In this context, current experiments were performed to establish the chemopreventive potential of propolis extract (PE) (1.05 mg/kg BW/day) in N-methyl-N-nitrosourea- (MNU-) induced rat mammary tumors. MNU-inoculated/PE-treated rats had tumors of different physical attributes compared with control rats MNU-inoculated. The number of developed tumors (mean 49% versus 100%), incidence (mean 49% versus 100%), multiplicity (1.8 versus 3.7 (p < 0.001)), tumor volume (mean 10 cm3 versus 16 cm3 (p < 0.001)), and weight of the tumor mass (mean 7.42 g versus 9.00 g (p < 0.05)) were noted. The numbers of grade I tumors recorded for MNU-inoculated rats were 24 (Group 1) and 7 (Group 2) for MNU-induced/PE-treated rats. In the serum of rats MNU-inoculated/PE-treated were found higher levels of antioxidative enzymes (SOD, CAT, and GPx) than in MNU-induced. Taken together, these data indicate that propolis could be a chemopreventive agent against MNU-induced mammary carcinogenesis.


Assuntos
Neoplasias Mamárias Animais/tratamento farmacológico , Neoplasias Mamárias Animais/prevenção & controle , Própole/uso terapêutico , Animais , Antioxidantes/uso terapêutico , Biomarcadores Tumorais/metabolismo , Quimioprevenção , Dieta , Feminino , Neoplasias Mamárias Animais/sangue , Metilnitrosoureia , Ratos Sprague-Dawley
7.
Nutrients ; 11(11)2019 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-31689992

RESUMO

Breast cancer is the most common cancer among women worldwide. We previously showed that early-life exposure to flaxseed (FS) or its components, FS oil (FSO) and secoisolariciresinol diglucoside (SDG), affects the mammary gland (MG) and is associated with the reduction of breast cancer risk during adulthood. However, the underlying mechanisms are not understood. This study aimed to investigate the effect of FS, FSO, and SDG on the MG miRNA signature at a late stage of development. Female C57BL/6 mice, 4-5 weeks of age, were randomized into four groups to receive: (i) basal AIN-93G, (ii) 10% FS, (iii) 3.67% FSO, or (iv) 0.15% SDG. After 21 days, the mice were sacrificed and MG miRNAs were profiled. Diet-specific MG miRNA signatures were identified. Deregulated miRNAs were associated with breast cancer and targeted genes involved in MG development, growth, and cancer. The study allowed for the identification of potential biomarkers or novel therapeutic targets to prevent and/or reduce the risk of breast cancer.


Assuntos
Linho , Neoplasias Mamárias Animais/prevenção & controle , Animais , Biomarcadores/sangue , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , MicroRNAs/metabolismo , Distribuição Aleatória
8.
J Vet Sci ; 20(5): e48, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31565891

RESUMO

Breast cancer is among the most common malignancies affecting women and reproductively intact female dogs, resulting in death from metastatic disease if not treated effectively. To better manage the disease progression, canine mammary tumor (CMT) cells derived from malignant canine mammary cancers were fused to autologous dendritic cells (DCs) to produce living hybrid-cell fusion vaccines for canine patients diagnosed with spontaneous mammary carcinoma. The high-speed sorting of rare autologous canine patient DCs from the peripheral blood provides the autologous component of fusion vaccines, and fusion to major histocompatibility complex-unmatched CMT cells were produced at high rates. The vaccinations were delivered to each patient following a surgical resection 3 times at 3-week intervals in combination with immuno-stimulatory oligonucleotides and Gemcitabine adjunct therapy. The immunized patient animals survived 3.3-times longer (median survival 611 days) than the control patients (median survival 184 days) and also appeared to exhibit an enhanced quality of life. A comparison of vaccinated patients diagnosed with inflammatory mammary carcinoma resulted in a very short median survival (42 days), suggesting no effect of vaccination. The data showed that the development of autologous living DC-based vaccine strategies in patient animals designed to improve the management of canine mammary carcinoma can be successful and may allow an identification of the antigens that can be translatable to promote effective immunity in canine and human patients.


Assuntos
Vacinas Anticâncer/administração & dosagem , Carcinoma/veterinária , Células Dendríticas/fisiologia , Neoplasias Mamárias Animais/prevenção & controle , Animais , Carcinoma/prevenção & controle , Fusão Celular , Linhagem Celular Tumoral , Modelos Animais de Doenças , Cães , Feminino
9.
Theranostics ; 9(12): 3580-3594, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31281499

RESUMO

Tumor microenvironment, such as hypoxia and presence of immune cells, plays a critical role in cancer initiation, growth as well as progression, and seriously affects antitumor effect. Accordingly, we constructed a kind of multifunctional nanoparticles (NPs) with macrophage transformation and oxygen (O2) generation characteristics, to regulate the tumor microenvironment. Methods: In this study, we synthesized mesoporous Prussian blue (MPB) NPs with low molecular weight hyaluronic acid (LMWHA) surface modification (LMWHA-MPB), and discovered that LMWHA-MPB could be used as an in situ macrophages converter and O2 generator. Results:In vitro results showed after uptake by M2 macrophages, LMWHA-MPB displayed the potential in remodeling tumor-associated macrophages (TAMs) phenotype (pro-tumor M2→anti-tumor M1), and anti-metastatic effect on 4T1 cells. Furthermore, in vivo visualized near-infrared (NIR) imaging data proved IR783 labeled LMWHA-MPB NPs could selectively accumulate in tumor sites. Then plenty of O2 generated to alleviate tumor hypoxia via catalytic decomposition of endogenous hydrogen peroxide (H2O2). Based on these outstanding characteristics, LMWHA-MPB NPs were adopted as multifunctional nanocarriers to load sonosensitizer hematoporphyrin monomethyl ether (HMME) for O2 self-provided sonodynamic therapy (SDT). In vivo anti-tumor results showed LMWHA-MPB/HMME could effectively inhibit the proliferation and metastasis of 4T1 tumors by improving tumor microenvironment. Conclusion: The multifunctional NPs can be used as in situ microenvironmental nano-regulators to inhibit the proliferation and metastasis of 4T1 tumor.


Assuntos
Fatores Imunológicos/metabolismo , Neoplasias Mamárias Animais/prevenção & controle , Nanopartículas/metabolismo , Metástase Neoplásica/prevenção & controle , Oxidantes/metabolismo , Microambiente Tumoral/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Ferrocianetos/síntese química , Ferrocianetos/metabolismo , Ácido Hialurônico/metabolismo , Fatores Imunológicos/síntese química , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Camundongos , Modelos Biológicos , Oxidantes/síntese química , Resultado do Tratamento
10.
J Dev Orig Health Dis ; 10(4): 479-487, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30626462

RESUMO

Environmental factors during perinatal life can lead to changes in the mammary gland, making it susceptible to cancer in adulthood. Breastfeeding has a special importance since it takes place at a critical period of growth and development of the newborn. We aimed to analyze if an appropriate lactation protects the offspring against mammary carcinogenesis during adult life and explore the mechanisms involved in the protective effect. One-day-old Sprague-Dawley female rats were randomly distributed in litters of three (L3), eight (L8) or 12 (L12) pups per dam, to induce a differential consumption of breast milk. At 55 days of age, the animals were treated with a single dose of dimethylbenzanthracene to study tumor latency, incidence and progression. Histological, immunohistochemical and Western blot studies were performed. We observed lower incidence and higher latency in L3 compared to the other groups. The mitotic index and expression of proliferating cell nuclear antigen (PCNA) was significantly augmented in tumors of L12 rats compared to L3 and L8, while the apoptotic index was augmented in tumors of L3 v. L12. Cleaved caspase 8 was significantly higher in tumors from L3 compared to L12. Tumors developed in L3 have a greater number of apoptotic bodies and a greater expression of caspase 8. These results demonstrate that the animals that maintained a higher intake of maternal milk (L3) presented lower incidence and greater tumor latency. Lower consumption of breast milk (L12) would increase tumor mitosis and the expression of PCNA, explaining the higher tumor incidence observed in this group.


Assuntos
Aleitamento Materno/estatística & dados numéricos , Neoplasias Mamárias Animais/prevenção & controle , Leite/química , Envelhecimento , Animais , Apoptose , Feminino , Incidência , Lactação , Neoplasias Mamárias Animais/epidemiologia , Leite/estatística & dados numéricos , Mitose , Gravidez , Ratos , Ratos Sprague-Dawley
11.
Biochem Biophys Res Commun ; 508(2): 536-542, 2019 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-30509497

RESUMO

WNT/ß-catenin signaling plays pivotal roles in mammary development and tumorigenesis; and aberrant activation of this pathway is frequently observed in human breast cancer, correlating with poor outcome. However, the mechanisms underlying WNT-driven mammary tumorigenesis remain incompletely understood. Here, we used mouse mammary tumor virus (MMTV)-Wnt1 transgenic mice, which develop aggressive mammary adenocarcinomas, to examine whether Limb-Bud-and-Heart (LBH) - a WNT/ß-catenin target transcription co-factor overexpressed in human triple-negative breast cancers with WNT pathway hyperactivation, contributes to WNT-induced tumorigenesis. We found LBH is specifically overexpressed in basal epithelial tumor cells of MMTV-Wnt1 mammary tumors reminiscent of its basal cell-restricted expression in the normal postnatal mammary gland. To determine the role of LBH in mammary tumorigenesis, we crossed MMTV-Wnt1 mice with basal epithelial-specific Keratin 14/K14-Cre;LbhloxP knockout mice. Mammary glands from virgin LBH-deficient MMTV-Wnt1 mice exhibited reduced hyperplasia, cell proliferation and increased apoptosis. Importantly, LBH inactivation in mammary epithelium significantly delayed tumor onset in MMTV-Wnt1 transgenic mice, with a median tumor-free survival of 32.5 weeks compared to 22.5 weeks in control LBH wild type MMTV-Wnt1 mice (p < 0.05). This data provides the first evidence that LBH plays an essential role in WNT-induced mammary tumorigenesis by promoting hyperplastic growth and tumor formation.


Assuntos
Carcinogênese/induzido quimicamente , Hiperplasia/prevenção & controle , Glândulas Mamárias Animais/patologia , Neoplasias Mamárias Animais/prevenção & controle , Proteínas Nucleares/deficiência , Animais , Proteínas de Ciclo Celular , Feminino , Vírus do Tumor Mamário do Camundongo/genética , Camundongos , Camundongos Transgênicos , Fatores de Transcrição , Proteína Wnt1/genética
12.
Cancer Immunol Res ; 6(8): 910-920, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29907693

RESUMO

Radiotherapy (RT) used at immunogenic doses leads to accumulation of cytosolic double-stranded DNA (dsDNA) in cancer cells, which activates type I IFN (IFN-I) via the cGAS/STING pathway. Cancer cell-derived IFN-I is required to recruit BATF3-dependent dendritic cells (DC) to poorly immunogenic tumors and trigger antitumor T-cell responses in combination with immune checkpoint blockade. We have previously demonstrated that the exonuclease TREX1 regulates radiation immunogenicity by degrading cytosolic dsDNA. Tumor-derived DNA can also activate cGAS/STING-mediated production of IFN-I by DCs infiltrating immunogenic tumors. However, how DNA from cancer cells is transferred to the cytoplasm of DCs remains unclear. Here, we showed that tumor-derived exosomes (TEX) produced by irradiated mouse breast cancer cells (RT-TEX) transfer dsDNA to DCs and stimulate DC upregulation of costimulatory molecules and STING-dependent activation of IFN-I. In vivo, RT-TEX elicited tumor-specific CD8+ T-cell responses and protected mice from tumor development significantly better than TEX from untreated cancer cells in a prophylactic vaccination experiment. We demonstrated that the IFN-stimulatory dsDNA cargo of RT-TEX is regulated by TREX1 expression in the parent cells. Overall, these results identify RT-TEX as a mechanism whereby IFN-stimulatory dsDNA is transferred from irradiated cancer cells to DCs. We have previously shown that the expression of TREX1 is dependent on the RT dose size. Thus, these data have important implications for the use of RT with immunotherapy. Cancer Immunol Res; 6(8); 910-20. ©2018 AACR.


Assuntos
DNA de Neoplasias/imunologia , Células Dendríticas/imunologia , Exodesoxirribonucleases/imunologia , Exossomos/genética , Neoplasias Mamárias Animais/imunologia , Fosfoproteínas/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/imunologia , Exossomos/imunologia , Feminino , Interferon Tipo I/imunologia , Linfócitos do Interstício Tumoral/imunologia , Neoplasias Mamárias Animais/genética , Neoplasias Mamárias Animais/prevenção & controle , Neoplasias Mamárias Animais/radioterapia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Baço/imunologia , Células Tumorais Cultivadas
13.
Anim Sci J ; 89(8): 1169-1177, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29770537

RESUMO

Human epigenetic studies suggest that consumption of seaweed prevents mammary cancer, which possibly is explained by iodine daily intake. In this study, we evaluated the efficacy of dietary intake of iodine-enriched eggs on mammary tumor incidence caused by the expression of activated type ErbB2. Female transgenic mice were divided into three groups, and fed a basic diet, a diet supplemented with ordinary eggs, or with iodine-enriched eggs. The number of mammary tumors greater than 5 mm in diameter was recorded in mice at 6 months of age. We report that the average number of mammary tumors per mouse was significantly lower in the iodine-enriched egg-added diet group than in either the basic diet or ordinary egg diet groups. These results indicate that iodine intake through livestock-derived products can reduce the incidence of mammary cancers caused by the expression of activated type ErbB2.


Assuntos
Ração Animal , Dieta/veterinária , Suplementos Nutricionais , Ovos , Expressão Gênica , Iodo/administração & dosagem , Neoplasias Mamárias Animais/genética , Neoplasias Mamárias Animais/prevenção & controle , Receptor ErbB-2/genética , Ativação Transcricional , Animais , Feminino , Incidência , Camundongos Transgênicos
14.
Nat Commun ; 9(1): 1216, 2018 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-29572483

RESUMO

Transforming growth factor-ß (TGFß) signaling is initiated by the type I, II TGFß receptor (TßRI/TßRII) complex. Here we report the formation of an alternative complex between TßRI and the orphan GPR50, belonging to the G protein-coupled receptor super-family. The interaction of GPR50 with TßRI induces spontaneous TßRI-dependent Smad and non-Smad signaling by stabilizing the active TßRI conformation and competing for the binding of the negative regulator FKBP12 to TßRI. GPR50 overexpression in MDA-MB-231 cells mimics the anti-proliferative effect of TßRI and decreases tumor growth in a xenograft mouse model. Inversely, targeted deletion of GPR50 in the MMTV/Neu spontaneous mammary cancer model shows decreased survival after tumor onset and increased tumor growth. Low GPR50 expression is associated with poor survival prognosis in human breast cancer irrespective of the breast cancer subtype. This describes a previously unappreciated spontaneous TGFß-independent activation mode of TßRI and identifies GPR50 as a TßRI co-receptor with potential impact on cancer development.


Assuntos
Neoplasias Mamárias Animais/prevenção & controle , Proteínas do Tecido Nervoso/fisiologia , Receptor do Fator de Crescimento Transformador beta Tipo I/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Animais , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Endossomos/metabolismo , Feminino , Perfilação da Expressão Gênica , Células HEK293 , Células HeLa , Humanos , Neoplasias Mamárias Animais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Proteínas do Tecido Nervoso/genética , Análise de Sequência com Séries de Oligonucleotídeos , Receptores Acoplados a Proteínas G/genética , Transdução de Sinais , Proteínas Smad/metabolismo , Proteína 1A de Ligação a Tacrolimo/metabolismo
15.
J Nanobiotechnology ; 16(1): 9, 2018 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-29382332

RESUMO

BACKGROUND: In the photodynamic therapy (PDT), the photosensitizer absorbs light and transfers the energy of the excited state to the oxygen in the cell environment producing reactive oxygen species (ROS), that in its turn, may cause cell damage. In the photothermal therapy (PTT), light also is responsible for activating the photothermal agent, which converts the absorbed energy in heat. Graphene oxide is a carbon-based material that presents photothermal activity. Its physical properties allow the association with the photosensitizer methylene blue and consequently the production of ROS when submitted to light irradiation. Therefore, the association between nanographene oxide and methylene blue could represent a strategy to enhance therapeutic actions. In this work, we report the nanographene oxide-methylene blue platform (NanoGO-MB) used to promote tumor ablation in combination with photodynamic and photothermal therapies against a syngeneic orthotopic murine breast cancer model. RESULTS: In vitro, NanoGO-MB presented 50% of the reactive oxygen species production compared to the free MB after LED light irradiation, and a temperature increase of ~ 40 °C followed by laser irradiation. On cells, the ROS production by the nanoplatform displayed higher values in tumor than normal cells. In vivo assays demonstrated a synergistic effect obtained by the combined PDT/PTT therapies using NanoGO-MB, which promoted complete tumor ablation in 5/5 animals. Up to 30 days after the last treatment, there was no tumor regrowth compared with only PDT or PTT groups, which displayed tumoral bioluminescence 63-fold higher than the combined treatment group. Histological studies confirmed that the combined therapies were able to prevent tumor regrowth and liver, lung and spleen metastasis. In addition, low systemic toxicity was observed in pathologic examinations of liver, spleen, lungs, and kidneys. CONCLUSIONS: The treatment with combined PDT/PTT therapies using NanoGO-MB induced more toxicity on breast carcinoma cells than on normal cells. In vivo, the combined therapies promoted complete tumor ablation and metastasis prevention while only PDT or PTT were unable to stop tumor development. The results show the potential of NanoGO-MB in combination with the phototherapies in the treatment of the breast cancer and metastasis prevention.


Assuntos
Técnicas de Ablação , Grafite/química , Neoplasias Mamárias Animais/tratamento farmacológico , Neoplasias Mamárias Animais/patologia , Azul de Metileno/química , Nanopartículas/química , Fototerapia , Animais , Apoptose , Peso Corporal , Linhagem Celular Tumoral , Sobrevivência Celular , Modelos Animais de Doenças , Feminino , Imuno-Histoquímica , Luminescência , Neoplasias Mamárias Animais/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Células NIH 3T3 , Nanopartículas/ultraestrutura , Metástase Neoplásica , Fotoquimioterapia , Espécies Reativas de Oxigênio , Carga Tumoral
16.
Int J Cancer ; 143(2): 368-382, 2018 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-29441580

RESUMO

Human epidemiological studies suggest that 1,25(OH)2 D3 deficiency might increase cancer incidence, but no spontaneous tumors have been reported in mice lacking 1,25(OH)2 D3 or deficient in its receptor. In our study, we detected, for the first time, diverse types of spontaneous tumors in l,25(OH)2 D3 deficient mice more than 1 year of age. This was associated with increased oxidative stress, cellular senescence and senescence-associated secretory phenotype molecules, such as hepatocyte growth factor, mediated via its receptor c-Met. Furthermore, 1,25(OH)2 D3 prevented spontaneous tumor development. We also demonstrated that l,25(OH)2 D3 deficiency accelerates allograft tumor initiation and growth by increasing oxidative stress and DNA damage, activating oncogenes, inactivating tumor suppressor genes, stimulating malignant cell proliferation and inhibiting their senescence; in contrast, supplementation with exogenous l,25(OH)2 D3 or antioxidant, or knock-down of the Bmi1 or c-Met oncogene, largely rescued the phenotypes of allograft tumors. Results from our study suggest that 1,25(OH)2 D3 deficiency enhances tumorigenesis by increasing malignant cell oxidative stress and DNA damage, stimulating microenvironmental cell senescence and a senescence-associated secretory phenotype, and activating oncogenes and inactivating tumor suppressor genes, thus increasing malignant cell proliferation. Our study provides direct evidence supporting the role of vitamin D deficiency in increasing cancer incidence. Conversely, 1,25(OH)2 D3 prevented spontaneous tumor development, suggesting that this inhibitory effect prevents the initiation and progression of tumorigenesis, thus provides a mechanistic basis for 1,25(OH)2 D3 to prevent tumorigenesis in an aging organism.


Assuntos
Calcitriol/administração & dosagem , Transformação Celular Neoplásica/efeitos dos fármacos , Neoplasias Mamárias Animais/prevenção & controle , Estresse Oxidativo/efeitos dos fármacos , Deficiência de Vitamina D/tratamento farmacológico , Animais , Calcitriol/farmacologia , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Senescência Celular , Dano ao DNA/efeitos dos fármacos , Feminino , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Neoplasias Mamárias Animais/genética , Neoplasias Mamárias Animais/metabolismo , Camundongos , Proteínas Proto-Oncogênicas c-met/metabolismo , Deficiência de Vitamina D/complicações , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Am J Pathol ; 188(2): 515-524, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29169987

RESUMO

Epidemiologic studies report improved breast cancer survival in women who receive ketorolac (Toradol) for postoperative pain relief compared with other analgesic agents. Ketorolac is a racemic drug. The S-enantiomer inhibits cyclooxygenases; R-ketorolac is a selective inhibitor of the small GTPases Ras-related C3 botulinum toxin substrate 1 (Rac1) and cell division control protein 42 (Cdc42), which are signaling molecules up-regulated during breast cancer progression and metastasis. The goal of this study was to determine whether R-ketorolac altered breast cancer development in the mouse mammary tumor virus-polyoma middle T-antigen model. Mice were administered ketorolac orally at 1 mg/kg twice daily to approximate the typical human dose. Mammary glands were analyzed for tumor number and immunohistochemical markers of proliferation and differentiation. R-ketorolac treatment significantly reduced mammary epithelial proliferation, based on Ki67 staining, and suppressed tumor development. Proliferative mammary epithelium from R-ketorolac-treated mice displayed greater differentiation, based on significantly higher total E-cadherin and decreased keratin 5 staining than epithelium of placebo-treated mice. No differences were detected in estrogen receptor, progesterone receptor, ß-catenin, or vimentin expression between placebo and R-ketorolac treatment groups. These findings indicate that R-ketorolac treatment slows tumor progression in an aggressive model of breast cancer. R-ketorolac may thus represent a novel therapeutic approach for breast cancer prevention or treatment based on its pharmacologic activity as a Rac1 and Cdc42 inhibitor.


Assuntos
Antineoplásicos/uso terapêutico , Cetorolaco de Trometamina/uso terapêutico , Neoplasias Mamárias Animais/prevenção & controle , Administração Oral , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacologia , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Progressão da Doença , Esquema de Medicação , Avaliação Pré-Clínica de Medicamentos/métodos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Feminino , Cetorolaco de Trometamina/administração & dosagem , Cetorolaco de Trometamina/farmacologia , Neoplasias Mamárias Animais/patologia , Vírus do Tumor Mamário do Camundongo , Camundongos Transgênicos , Polyomavirus
18.
Anticancer Res ; 37(11): 6041-6046, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-29061783

RESUMO

BACKGROUND/AIM: Fucoidans are gaining popularity as natural immunomodulators. The aim of this study was to compare the immunological activities or both purified samples and commercially available mixtures containing fucoidan. MATERIALS AND METHODS: We evaluated the effects of various samples on phagocytosis, mitogenic response, natural killer (NK) activity, antibody formation and inhibition of breast cancer growth. RESULTS: We found significant immunostimulating activity, but the strength of these effects was different among individual samples. CONCLUSION: Fucoidans have strong immunostimulating potential, including inhibition of cancer, with isolated samples offering better activity than commercial mixtures.


Assuntos
Inflamação/prevenção & controle , Células Matadoras Naturais/imunologia , Macrófagos/imunologia , Neoplasias Mamárias Animais/prevenção & controle , Polissacarídeos/farmacologia , Linfócitos T/imunologia , Adjuvantes Imunológicos/farmacologia , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Feminino , Fatores Imunológicos , Inflamação/imunologia , Inflamação/patologia , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/patologia , Macrófagos/efeitos dos fármacos , Macrófagos/patologia , Neoplasias Mamárias Animais/imunologia , Neoplasias Mamárias Animais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Fagocitose/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos , Linfócitos T/patologia
19.
Cancer Prev Res (Phila) ; 10(10): 553-562, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28904060

RESUMO

Obesity and alterations in metabolic programming from early diet exposures can affect the propensity to disease in later life. Through dietary manipulation, developing mouse pups were exposed to a hyperinsulinemic, hyperglycemic milieu during three developmental phases: gestation, lactation, and postweaning. Analyses showed that a postweaning high fat/high sugar (HF/HS) diet had the main negative effect on adult body weight, glucose tolerance, and insulin resistance. However, dimethylbenz[a]anthracene (DMBA)-induced carcinogenesis revealed that animals born to a mother fed a HF/HS gestation diet, nursed by a mother on a mildly diet-restricted, low fat/low sugar diet (DR) and weaned onto a HF/HS diet (HF/DR/HF) had the highest mammary tumor incidence, while HF/HF/DR had the lowest tumor incidence. Cox proportional hazards analysis showed that a HF/HS postweaning diet doubled mammary cancer risk, and a HF/HS diet during gestation and postweaning increased risk 5.5 times. Exposure to a HF/HS diet during gestation, when combined with a postweaning DR diet, had a protective effect, reducing mammary tumor risk by 86% (HR = 0.142). Serum adipocytokine analysis revealed significant diet-dependent differences in leptin/adiponectin ratio and IGF-1. Flow cytometry analysis of cells isolated from mammary glands from a high tumor incidence group, DR/HF/HF, showed a significant increase in the size of the mammary stem cell compartment compared with a low tumor group, HF/HF/DR. These results indicate that dietary reprogramming induces an expansion of the mammary stem cell compartment during mammary development, increasing likely carcinogen targets and mammary cancer risk. Cancer Prev Res; 10(10); 553-62. ©2017 AACRSee related editorial by Freedland, p. 551-2.


Assuntos
Carcinogênese/metabolismo , Dieta Hiperlipídica/efeitos adversos , Açúcares da Dieta/efeitos adversos , Glândulas Mamárias Animais/embriologia , Neoplasias Mamárias Animais/etiologia , Neoplasias Mamárias Experimentais/metabolismo , Células-Tronco/patologia , 9,10-Dimetil-1,2-benzantraceno/toxicidade , Adiponectina/sangue , Animais , Peso Corporal/fisiologia , Dieta com Restrição de Gorduras , Comportamento Alimentar , Feminino , Resistência à Insulina/fisiologia , Fator de Crescimento Insulin-Like I/análise , Lactação/metabolismo , Leptina/sangue , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/patologia , Neoplasias Mamárias Animais/sangue , Neoplasias Mamárias Animais/metabolismo , Neoplasias Mamárias Animais/prevenção & controle , Neoplasias Mamárias Experimentais/sangue , Neoplasias Mamárias Experimentais/induzido quimicamente , Neoplasias Mamárias Experimentais/prevenção & controle , Exposição Materna/efeitos adversos , Camundongos , Camundongos Endogâmicos SENCAR , Obesidade/etiologia , Obesidade/metabolismo , Fatores de Risco , Fatores de Tempo
20.
J Pathol ; 241(4): 511-521, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27943283

RESUMO

Women with heterozygous germline mutations in the BRCA1 tumour suppressor gene are strongly predisposed to developing early-onset breast cancer through loss of the remaining wild-type BRCA1 allele and inactivation of TP53. Although tumour prevention strategies in BRCA1-mutation carriers are still limited to prophylactic surgery, several therapeutic strategies have been developed to target the DNA repair defects (also known as 'BRCAness') of BRCA1-deficient tumours. In particular, DNA-damaging agents such as platinum drugs and poly(ADP-ribose) polymerase (PARP) inhibitors show strong activity against BRCA1-mutated tumours. However, it is unclear whether drugs that target BRCAness can also be used to prevent tumour formation in BRCA1-mutation carriers, especially as loss of wild-type BRCA1 may not be the first event in BRCA1-associated tumourigenesis. We performed prophylactic treatments in a genetically engineered mouse model in which de novo development of BRCA1-deficient mammary tumours is induced by stochastic loss of BRCA1 and p53. We found that prophylactic window therapy with nimustine, cisplatin or olaparib reduced the amount and size of mammary gland lesions, and significantly increased the median tumour latency. Similar results were obtained with intermittent prophylactic treatment with olaparib. Importantly, prophylactic window therapy with nimustine and cisplatin resulted in an increased fraction of BRCA1-proficient mammary tumours, suggesting selective survival and malignant transformation of BRCA1-proficient lesions upon prophylactic treatment with DNA-damaging agents. Prophylactic therapy with olaparib significantly prolonged mammary tumour-free survival without any significant increase in the fraction of BRCA1-proficient tumours, warranting the evaluation of this PARP inhibitor in prophylactic trials in BRCA1-mutation carriers. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Mamárias Animais/tratamento farmacológico , Ftalazinas/farmacologia , Piperazinas/farmacologia , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Proteínas Supressoras de Tumor/genética , Animais , Antineoplásicos/uso terapêutico , Proteína BRCA1 , Cisplatino/farmacologia , Reparo do DNA , Modelos Animais de Doenças , Feminino , Mutação em Linhagem Germinativa , Humanos , Neoplasias Mamárias Animais/patologia , Neoplasias Mamárias Animais/prevenção & controle , Camundongos , Nimustina/farmacologia , Ftalazinas/uso terapêutico , Piperazinas/uso terapêutico , Inibidores de Poli(ADP-Ribose) Polimerases/uso terapêutico , Poli(ADP-Ribose) Polimerases/genética , Poli(ADP-Ribose) Polimerases/metabolismo , Proteínas Supressoras de Tumor/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...