Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Sci Total Environ ; 839: 156379, 2022 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-35654185

RESUMO

Cytotoxicity, molecular function disorder, mitophagy, and apoptosis were studied in loach fin cells in vitro after exposure to doxycycline (DOX). The semi-lethal concentration of DOX in loach cells was calculated as 668.96 ± 2.83 mol/L. Loss of cell viability and increases in vacuoles and autolysosomes were evident in cells exposed to DOX at 200 and 400 µmol/L, and apoptotic bodies occurred at 600 µmol/L. In addition, Superoxide Dismutase (SOD), catalase (CAT), Na+-K+-ATPase, and Ca2+-ATPase activities increased significantly in cells exposed to 200 µmol/L DOX, and dose-dependent inhibitory effects on activities were observed in cells exposed to 400 and 600 µmol/L DOX. Quantitative gene expression showed that 400 and 600 µmol/L DOX could induce caspase-3- and caspase-8-mediated apoptosis as well as caspase-activated DNase in loach cells. Transcriptome sequencing in DOX vs. control groups found 16,288 differentially expressed genes, among which protein binding (2633, 31.91%) was the most significant in Gene Ontology terms. Furthermore, 11,930 genes were enriched in 298 Kyoto Encyclopedia of Genes and Genomes (KEGG)pathways. The top three upregulated pathways included "lysosome", "protein processing in endoplasmic reticulum", and "proteasome". FPKM analysis indicated that most genes associated with autophagy and in "protein processing in the endoplasmic reticulum", "TNF signaling pathway", and "NF-kappa B signaling pathway" were upregulated. This suggests that at lower concentrations, DOX induces reactive oxidative species (ROS) in loach fin cells to reduce cell proliferation. ROS in turn stimulate oxidant stress, ion excretion capability and mitophagy to maintain cell homeostasis. Apoptosis was induced in cells subjected to higher concentrations of DOX. The transcriptome data and pathways determined in this study will provide a foundation for the analysis of DOX toxicity in loach cells, which must be examined thoroughly to further understand the cytotoxic mechanism of antibiotics in fish cells.


Assuntos
Doxiciclina , Estresse Oxidativo , Adenosina Trifosfatases/metabolismo , Adenosina Trifosfatases/farmacologia , Animais , Apoptose , Autofagia , Doxiciclina/toxicidade , Espécies Reativas de Oxigênio/metabolismo
2.
Environ Res ; 196: 110962, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33675800

RESUMO

Perfluorooctane sulfonate (PFOS) is a persistent organic pollutant that has been widely detected in the environment and has caused growing international concern. The liver is the main target organ of PFOS exposure. Animal experiments have shown that PFOS exposure can increase the risk of liver tumorigenesis. However, whether PFOS can accelerate liver tumor progression is still unclear. In this study, transgenic zebrafish Tg(fabp10:rtTA2s-M2; TRE2:EGFP-KRASG12V), a hepatocellular carcinoma (HCC) model that can cause liver tumorigenesis by doxycycline (DOX) induction, was used to investigate the effect of PFOS exposure in HCC progression. The male krasV12 transgenic zebrafish were exposed to 20 mg/L DOX, 500 µg/L PFOS or combined 20 mg/L DOX and 500 µg/L PFOS for 10 d. The results showed that co-treated with PFOS and DOX caused oncogenic Kras-induced liver enlargement, increased the percentages of zebrafish with HCC, and aggravated metabolic reprogramming of liver. To the best of our knowledge, this study for the first proved that PFOS could promote liver tumor progression. Decreased vitamin D level and increased fatty acid intake caused by PFOS might be responsible for the tumor-promoting effects. The results suggest that attention should be paid to the tumor-promoting effects of PFOS when assessing its environmental health risks, and these findings provide new insights into the toxicity of PFOS.


Assuntos
Ácidos Alcanossulfônicos , Carcinoma Hepatocelular , Fluorocarbonos , Neoplasias Hepáticas , Ácidos Alcanossulfônicos/toxicidade , Animais , Doxiciclina/toxicidade , Fluorocarbonos/toxicidade , Fígado , Neoplasias Hepáticas/induzido quimicamente , Neoplasias Hepáticas/genética , Masculino , Proteínas Proto-Oncogênicas p21(ras) , Peixe-Zebra/genética
3.
Cancer Biol Ther ; 21(5): 400-411, 2020 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-32037955

RESUMO

B Cell-Specific Moloney Murine Leukemia Virus Integration Site 1 (Bmi-1, Bmi1), an epigenetic protein, is necessary for normal stem cell self-renewal in adult animals and for cancer stem cell (CSC) functions in adult animals. To elucidate the functions of Bmi-1 in the oral cavity we created a transgenic mouse line (KrTBmi-1) that expresses ectopic, Flag-tagged Bmi-1 in tongue basal epithelial stem cells only upon doxycycline (DOX) treatment. Genome wide transcriptomics and Ingenuity Pathway Analysis identified several pathways altered by exogenous Bmi-1 expression in the normal tongue epithelium, including EIF2 signaling (P value = 1.58 x 10-49), mTOR signaling (P value = 2.45 x 10-12), oxidative phosphorylation (P = 6.61 x 10-3) and glutathione redox reactions I (P = 1.74 x 10-2). Overall, our data indicate that ectopic Bmi-1 expression has an impact on normal tongue epithelial homeostasis. We then assessed the KrTBmi-1 mice in the 4-nitroquinoline 1-oxide (4-NQO) model of oral carcinogenesis. We found that 80% of mice expressing exogenous Bmi-1 (+DOX, +4-NQO KrTBmi-1; N = 10) developed tumors classified as grade 3 or higher, compared to 60% and 40% of mice expressing just endogenous Bmi-1 (+DOX, +4-NQO Kr and -DOX, +4-NQO KrTBmi-1 groups, respectively; N = 10/group; P value = <0.0001); and 30% of mice expressing ectopic Bmi-1 mice developed 20 or more lesions compared to 10% of mice expressing only endogenous Bmi-1 (P = .009). This demonstrates that exogenous Bmi-1 expression increases the susceptibility of mice to 4-NQO-induced oral carcinogenesis, strengthening the evidence for Bmi-1 as a therapeutic target in human oral squamous cell carcinoma.


Assuntos
4-Nitroquinolina-1-Óxido/toxicidade , Doxiciclina/toxicidade , Regulação Neoplásica da Expressão Gênica , Neoplasias de Cabeça e Pescoço/patologia , Complexo Repressor Polycomb 1/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço/patologia , Animais , Antibacterianos/toxicidade , Carcinogênese , Carcinógenos/toxicidade , Modelos Animais de Doenças , Neoplasias de Cabeça e Pescoço/etiologia , Neoplasias de Cabeça e Pescoço/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Complexo Repressor Polycomb 1/genética , Proteínas Proto-Oncogênicas/genética , Carcinoma de Células Escamosas de Cabeça e Pescoço/etiologia , Carcinoma de Células Escamosas de Cabeça e Pescoço/metabolismo
4.
BMC Vet Res ; 14(1): 86, 2018 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-29530019

RESUMO

BACKGROUND: The teratogenic effects of immunomodulatory and certain antimicrobial therapies are described in small rodents and humans. While the described teratogenic effects in small rodents have been extrapolated to make conclusions about its use in the pregnant dam, teratogenic effects of prednisone and doxycycline have not yet been reported in the dog. Here we report and describe midline defects observed in a litter of golden retriever puppies exposed to mid-gestational immunosuppressive and antimicrobial therapy. CASE PRESENTATION: Twenty-one days into gestation, the dam of a litter of eight golden retriever puppies was administered prednisone, doxycycline, and tramadol as treatment for immune-mediated polyarthritis. The individuals in the litter were subsequently diagnosed with a variety of midline defects and congenital cardiac defects. This case series describes the variety of identified defects and presents a descriptive account of complex congenital abnormalities that are likely secondary to teratogenic effects of one or more drugs administered during gestation. The available puppies, dam, and grand dam underwent thorough physical examination, complete echocardiogram, and where indicated, advanced imaging with various surgical corrections when possible. Numerous midline congenital defects and congenital heart disease were identified in the puppies evaluated. Ultimately 5 of 8 puppies born to the dam were presented for thorough evaluation. The midline defects include: gastroschisis (1), peritoneopericardial diaphragmatic hernias (4, PPDH), umbilical hernia (4), unilateral cryptorchidism (1 of 4 males), cleft palate (1), renal agenesis (1), renal abnormalities (1), sternal and vertebral abnormalities (3), remnant liver lobe (1) and malformations consistent with ductal plate malformations with congenital hepatic fibrosis (1). The congenital cardiac defects include: ventricular septal defect (4, VSD) and subaortic stenosis (4, SAS). The presence of greater than one congenital defect was noted in all 5 of the dogs evaluated. Surgical correction was necessary for PPDH in 4 puppies. Medical intervention was recommended for congenital cardiac disease in 1 puppy. CONCLUSION: This case report is the first to describe midline defects in dogs that have been exposed to immunomodulatory therapy during gestation. A causative relationship between mid-gestational immunomodulatory exposure and midline defects cannot be proven, however, this case supports a clear association and provides case-based evidence to support its avoidance when possible.


Assuntos
Anormalidades Induzidas por Medicamentos/veterinária , Antibacterianos/toxicidade , Anti-Inflamatórios/toxicidade , Cães/anormalidades , Doxiciclina/toxicidade , Cardiopatias Congênitas/veterinária , Prednisona/toxicidade , Anormalidades Induzidas por Medicamentos/etiologia , Anormalidades Induzidas por Medicamentos/patologia , Animais , Antibacterianos/uso terapêutico , Anti-Inflamatórios/uso terapêutico , Artrite/complicações , Artrite/tratamento farmacológico , Artrite/veterinária , Doxiciclina/uso terapêutico , Ecocardiografia/veterinária , Feminino , Cardiopatias Congênitas/induzido quimicamente , Masculino , Defeitos do Tubo Neural/induzido quimicamente , Defeitos do Tubo Neural/veterinária , Prednisona/uso terapêutico , Gravidez , Complicações na Gravidez/tratamento farmacológico , Complicações na Gravidez/veterinária
5.
Proc Natl Acad Sci U S A ; 115(3): E428-E437, 2018 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-29282319

RESUMO

Fibrous dysplasia (FD) is a disease caused by postzygotic activating mutations of GNAS (R201C and R201H) that encode the α-subunit of the Gs stimulatory protein. FD is characterized by the development of areas of abnormal fibroosseous tissue in the bones, resulting in skeletal deformities, fractures, and pain. Despite the well-defined genetic alterations underlying FD, whether GNAS activation is sufficient for FD initiation and the molecular and cellular consequences of GNAS mutations remains largely unresolved, and there are no currently available targeted therapeutic options for FD. Here, we have developed a conditional tetracycline (Tet)-inducible animal model expressing the GαsR201C in the skeletal stem cell (SSC) lineage (Tet-GαsR201C/Prrx1-Cre/LSL-rtTA-IRES-GFP mice), which develops typical FD bone lesions in both embryos and adult mice in less than 2 weeks following doxycycline (Dox) administration. Conditional GαsR201C expression promoted PKA activation and proliferation of SSCs along the osteogenic lineage but halted their differentiation to mature osteoblasts. Rather, as is seen clinically, areas of woven bone admixed with fibrous tissue were formed. GαsR201C caused the concomitant expression of receptor activator of nuclear factor kappa-B ligand (Rankl) that led to marked osteoclastogenesis and bone resorption. GαsR201C expression ablation by Dox withdrawal resulted in FD-like lesion regression, supporting the rationale for Gαs-targeted drugs to attempt FD cure. This model, which develops FD-like lesions that can form rapidly and revert on cessation of mutant Gαs expression, provides an opportunity to identify the molecular mechanism underlying FD initiation and progression and accelerate the development of new treatment options.


Assuntos
Displasia Fibrosa Óssea/metabolismo , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Células-Tronco Mesenquimais/metabolismo , Animais , Antibacterianos/toxicidade , Desenvolvimento Ósseo/efeitos dos fármacos , Osso e Ossos/patologia , Diferenciação Celular , Doxiciclina/toxicidade , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Regulação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Camundongos , Mutação
6.
J Cardiovasc Pharmacol Ther ; 22(5): 458-466, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28793824

RESUMO

Cardiomyopathy related to the absence of dystrophin is an important feature in Duchenne muscular dystrophy (DMD) and in the mdx mouse. Doxycycline (DOX) could be a potential therapy for mdx skeletal muscles dystrophy. We investigated whether the corticoid deflazacort (DFZ) plus DOX could improve cardiac mdx dystrophy better than DFZ alone, later (17 months) in dystrophy. Mdx mice (8 months old) received DFZ/DOX or DFZ for 9 months. The combined therapy was greater than DFZ in reducing fibrosis (60% decrease with DFZ/DOX and 40% with DFZ alone) in the right ventricle and transforming growth factor ß levels (6.8 ± 3.2 in untreated mdx mice, 2.8 ± 1.4 in combined therapy, and 4.6 ± 1.7 in DFZ; P < .05). Combined therapy more effectively ameliorated cardiac dysfunction (electrocardiogram [ECG]) than DFZ. Improvements were seen in the cardiomyopathy index (0.8 ± 0.1 in combined therapy and 1.0 ± 0.2 in DFZ), heart rate (418 ± 46 bpm in combined therapy and 457 ± 29 bpm in DFZ), QRS interval (11.3 ± 2 in combined therapy and 13.6 ± 1 in DFZ), and Q wave amplitude (-40.7 ± 21 in combined therapy and -90.9 ± 36 in DFZ). Both therapies decreased markers of inflammation (tumor necrosis factor α, nuclear factor κB, and metalloproteinase 9). DFZ/DOX improved mdx cardiomyopathy at this stage of the disease, supporting further clinical investigations.


Assuntos
Cardiomiopatias/tratamento farmacológico , Doxiciclina/administração & dosagem , Distrofina/deficiência , Distrofia Muscular de Duchenne/complicações , Pregnenodionas/uso terapêutico , Animais , Cardiomiopatias/etiologia , Doxiciclina/toxicidade , Quimioterapia Combinada , Eletrocardiografia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pregnenodionas/administração & dosagem , Pregnenodionas/toxicidade
7.
Cell Physiol Biochem ; 42(1): 407-415, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28558368

RESUMO

BACKGROUND: Tumor derived vascular endothelial growth factor (VEGF) can stimulate proliferation and migration of endothelial cells and recruit endothelial progenitor cells into tumors for vascular formation via a paracrine manner. Now increasing evidence suggests that VEGF also serves as an autocrine factor promoting cell survival and tumor angiogenesis. Real time visualization of VEGF activity in the early stages of tumor formation using molecular imaging will provide unprecedented insight into the biological processes of cancer. METHODS: The mouse breast cancer cell line 4T1 was transfected with an inducible, bidirectional tetracycline (Bi-Tet) promoter driving VEGF and renilla luciferase (Rluc). This was used to quantitatively image conditional switching of VEGF by bioluminescence imaging (BLI) under the control of systemic administration of doxycycline. Simultaneously, 4T1 cells were labelled with the double fusion reporter gene (Fluc-eGFP) to establish a breast cancer model. RESULTS: We found that inducible VEGF could promote proliferation and attenuate apoptosis due to oxidative stress in an autocrine manner in vitro. In vivo studies revealed that induction of VEGF expression during early tumor development not only dramatically enhanced tumor growth but also increased tumor angiogenesis as visualized by BLI. Finally, immunohistochemistry staining confirmed that inducing VEGF expression promoted cell survival and tumor neovascularization. CONCLUSION: Together the inducible bidirectional tetracycline (Bi-Tet) co-expression system combined with the dual bioluminescence imaging (BLI) system provides a platform to investigate a target gene's role in the pathologic process of cancer and facilitates noninvasive monitoring of biological responses in real time.


Assuntos
Neoplasias da Mama/diagnóstico , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Apoptose/efeitos dos fármacos , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Progressão da Doença , Doxiciclina/toxicidade , Feminino , Regulação Neoplásica da Expressão Gênica , Genes Reporter , Humanos , Peróxido de Hidrogênio/toxicidade , Camundongos , Camundongos Endogâmicos BALB C , Microscopia de Fluorescência , Neovascularização Patológica/prevenção & controle , Imagem Óptica , Estresse Oxidativo/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/genética
8.
BMB Rep ; 49(7): 370-5, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26818088

RESUMO

Ras oncoproteins are small molecular weight GTPases known for their involvement in oncogenesis, which operate in a complex signaling network with multiple effectors. Approximately 25% of human tumors possess mutations in a member of this family. The Raf1/MEK/Erk1/2 pathway is one of the most intensively studied signaling mechanisms. Different levels of regulation account for the inactivation of MAP kinases by MAPK phosphatases in a cell type- and stimuli-dependent manner. In the present study, using three inducible Ras-expressing NIH/3T3 cell lines, we demonstrated that MKP3 upregulation requires the activation of the Erk1/2 pathway, which correlates with the shutdown of this pathway. We also demonstrated, by applying pharmacological inhibitors and effector mutants of Ras, that induction of MKP3 at the protein level is positively regulated by the oncogenic Ras/Raf/MEK/Erk1/2 signaling pathway. [BMB Reports 2016; 49(7): 370-375].


Assuntos
Fosfatase 6 de Especificidade Dupla/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteínas ras/metabolismo , Animais , Butadienos/farmacologia , Cromonas/farmacologia , Doxiciclina/toxicidade , Fator 2 de Crescimento de Fibroblastos/farmacologia , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Immunoblotting , Camundongos , Morfolinas/farmacologia , Mutagênese , Células NIH 3T3 , Nitrilas/farmacologia , Plasmídeos/genética , Plasmídeos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Proteínas ras/genética
9.
J Neurosci ; 35(5): 2191-9, 2015 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-25653374

RESUMO

Seizures are accompanied by cardiovascular changes that are a major cause of sudden unexpected death in epilepsy (SUDEP). Seizures activate inflammatory responses in the cardiovascular nuclei of the medulla oblongata and increase neuronal excitability. Pituitary adenylate cyclase-activating polypeptide (PACAP) is a neuropeptide with autocrine and paracrine neuroprotective properties. Microglia are key players in inflammatory responses in the CNS. We sought to determine whether PACAP and microglia mitigate the adverse effects of seizure on cardiovascular function in a rat model of temporal lobe epilepsy. Kainic acid (KA)-induced seizures increased splanchnic sympathetic nerve activity by 97%, accompanied by increase in heart rate (HR) but not blood pressure (BP). Intrathecal infusion of the PACAP antagonist PACAP(6-38) or the microglia antagonists minocycline and doxycycline augmented sympathetic responses to KA-induced seizures. PACAP(6-38) caused a 161% increase, whereas minocycline and doxycycline caused a 225% and 215% increase, respectively. In intrathecal PACAP-antagonist-treated rats, both BP and HR increased, whereas after treatment with microglial antagonists, only BP was significantly increased compared with control. Our findings support the idea that PACAP and its action on microglia at the level of the spinal cord elicit cardioprotective effects during seizure. However, intrathecal PACAP did not show additive effects, suggesting that the agonist effect was at maximum. The protective effect of microglia may occur by adoption of an M2 phenotype and expression of factors such as TGF-ß and IL-10 that promote neuronal quiescence. In summary, therapeutic interventions targeting PACAP and microglia could be a promising strategy for preventing SUDEP.


Assuntos
Arritmias Cardíacas/etiologia , Coração/efeitos dos fármacos , Microglia/efeitos dos fármacos , Fragmentos de Peptídeos/toxicidade , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/toxicidade , Convulsões/fisiopatologia , Animais , Arritmias Cardíacas/fisiopatologia , Pressão Sanguínea , Doxiciclina/toxicidade , Epilepsia do Lobo Temporal/complicações , Epilepsia do Lobo Temporal/metabolismo , Epilepsia do Lobo Temporal/fisiopatologia , Frequência Cardíaca , Masculino , Minociclina/toxicidade , Ratos , Ratos Sprague-Dawley , Convulsões/complicações , Convulsões/metabolismo , Medula Espinal/efeitos dos fármacos , Medula Espinal/fisiopatologia , Sistema Nervoso Simpático/efeitos dos fármacos , Sistema Nervoso Simpático/fisiopatologia
10.
Cell Metab ; 21(1): 117-25, 2015 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-25533479

RESUMO

Dietary effects on tumor biology can be exploited to unravel cancer vulnerabilities. Here, we present surprising evidence for anti-proliferative action of high-calorie-diet (HCD) feeding on KRAS-driven lung tumors. Tumors of mice that commenced HCD feeding before tumor onset displayed defective unfolded protein response (UPR) and unresolved endoplasmic reticulum (ER) stress. Unresolved ER stress and reduced proliferation are reversed by chemical chaperone treatment. Whole-genome transcriptional analyses revealed FKBP10 as one of the most downregulated chaperones in tumors of the HCD-pre-tumor-onset group. FKBP10 downregulation dampens tumor growth in vitro and in vivo. Providing translational value to these results, we report that FKBP10 is expressed in human KRAS-positive and -negative lung cancers, but not in healthy parenchyma. Collectively, our data shed light on an unexpected anti-tumor action of HCD imposed before tumor onset and identify FKBP10 as a putative therapeutic target to selectively hinder lung cancer.


Assuntos
Dieta , Neoplasias Pulmonares/patologia , Fenilbutiratos/toxicidade , Proteínas ras/metabolismo , Animais , Linhagem Celular Tumoral , Transformação Celular Neoplásica/efeitos dos fármacos , Regulação para Baixo , Doxiciclina/toxicidade , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/metabolismo , Masculino , Camundongos , Camundongos SCID , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Proteínas de Ligação a Tacrolimo/antagonistas & inibidores , Proteínas de Ligação a Tacrolimo/genética , Proteínas de Ligação a Tacrolimo/metabolismo , Transplante Heterólogo , Resposta a Proteínas não Dobradas/efeitos dos fármacos , Proteínas ras/genética
11.
Cell Death Dis ; 5: e1401, 2014 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-25188511

RESUMO

Neuroblastoma (NBL) is the most common solid tumor in infants and accounts for 15% of all pediatric cancer deaths. Several risk factors predict NBL outcome: age at the time of diagnosis, stage, chromosome alterations and MYCN (V-Myc Avian Myelocytomatosis Viral Oncogene Neuroblastoma-Derived Homolog) amplification, which characterizes the subset of the most aggressive NBLs with an overall survival below 30%. MYCN-amplified tumors develop exceptional chemoresistance and metastatic capacity. These properties have been linked to defects in the apoptotic machinery, either by silencing components of the extrinsic apoptotic pathway (e.g. caspase-8) or by overexpression of antiapoptotic regulators (e.g. Bcl-2, Mcl-1 or FLIP). Very little is known on the implication of death receptors and their antagonists in NBL. In this work, the expression levels of several death receptor antagonists were analyzed in multiple human NBL data sets. We report that Lifeguard (LFG/FAIM2 (Fas apoptosis inhibitory molecule 2)/NMP35) is downregulated in the most aggressive and undifferentiated tumors. Intringuingly, although LFG has been initially characterized as an antiapoptotic protein, we have found a new association with NBL differentiation. Moreover, LFG repression resulted in reduced cell adhesion, increased sphere growth and enhanced migration, thus conferring a higher metastatic capacity to NBL cells. Furthermore, LFG expression was found to be directly repressed by MYCN at the transcriptional level. Our data, which support a new functional role for a hitherto undiscovered MYCN target, provide a new link between MYCN overexpression and increased NBL metastatic properties.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Proteínas de Membrana/metabolismo , Neuroblastoma/patologia , Proteínas Nucleares/metabolismo , Proteínas Oncogênicas/metabolismo , Animais , Antibacterianos/toxicidade , Proteínas Reguladoras de Apoptose/antagonistas & inibidores , Proteínas Reguladoras de Apoptose/genética , Adesão Celular , Diferenciação Celular , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Doxiciclina/toxicidade , Feminino , Humanos , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/genética , Camundongos , Camundongos Nus , Proteína Proto-Oncogênica N-Myc , Metástase Neoplásica , Estadiamento de Neoplasias , Neuroblastoma/metabolismo , Proteínas Nucleares/genética , Proteínas Oncogênicas/genética , Interferência de RNA , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Receptores de Morte Celular/antagonistas & inibidores , Receptores de Morte Celular/metabolismo , Transplante Heterólogo , Tretinoína/farmacologia , Regulação para Cima/efeitos dos fármacos
12.
Am J Physiol Gastrointest Liver Physiol ; 307(5): G499-507, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-24994859

RESUMO

Resistin-like molecule (Relm)-α is a secreted, cysteine-rich protein belonging to a newly defined family of proteins, including resistin, Relm-ß, and Relm-γ. Although resistin was initially defined based on its insulin-resistance activity, the family members are highly induced in various inflammatory states. Earlier studies implicated Relm-α in insulin resistance, asthmatic responses, and intestinal inflammation; however, its function still remains an enigma. We now report that Relm-α is strongly induced in the esophagus in an allergen-challenged murine model of eosinophilic esophagitis (EoE). Furthermore, to understand the in vivo role of Relm-α, we generated Relm-α gene-inducible bitransgenic mice by using lung-specific CC-10 promoter (CC10-rtTA-Relm-α). We found Relm-α protein is significantly induced in the esophagus of CC10-rtTA-Relm-α bitransgenic mice exposed to doxycycline food. The most prominent effect observed by the induction of Relm-α is epithelial cell hyperplasia, basal layer thickness, accumulation of activated CD4(+) and CD4(-) T cell subsets, and eosinophilic inflammation in the esophagus. The in vitro experiments further confirm that Relm-α promotes primary epithelial cell proliferation but has no chemotactic activity for eosinophils. Taken together, our studies report for the first time that Relm-α induction in the esophagus has a major role in promoting epithelial cell hyperplasia and basal layer thickness, and the accumulation of activated CD4(+) and CD4(-) T cell subsets may be responsible for partial esophageal eosinophilia in the mouse models of EoE. Notably, the epithelial cell hyperplasia and basal layer thickness are the characteristic features commonly observed in human EoE.


Assuntos
Alérgenos/toxicidade , Doxiciclina/toxicidade , Esofagite Eosinofílica/metabolismo , Células Epiteliais/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Animais , Contagem de Linfócito CD4 , Proliferação de Células , Células Cultivadas , Quimiotaxia , Esofagite Eosinofílica/imunologia , Esofagite Eosinofílica/patologia , Eosinófilos/fisiologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Esôfago/efeitos dos fármacos , Esôfago/metabolismo , Esôfago/patologia , Hiperplasia/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/genética , Camundongos , Camundongos Endogâmicos BALB C
13.
Am J Physiol Lung Cell Mol Physiol ; 306(8): L726-35, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24508732

RESUMO

A number of growth factors and signaling pathways regulate matrix deposition and fibroblast proliferation in the lung. The epidermal growth factor receptor (EGFR) family of receptors and the transforming growth factor-ß (TGF-ß) family are active in diverse biological processes and are central mediators in the initiation and maintenance of fibrosis in many diseases. Transforming growth factor-α (TGF-α) is a ligand for the EGFR, and doxycycline (Dox)-inducible transgenic mice conditionally expressing TGF-α specifically in the lung epithelium develop progressive fibrosis accompanied with cachexia, changes in lung mechanics, and marked pleural thickening. Although recent studies demonstrate that EGFR activation modulates the fibroproliferative effects involved in the pathogenesis of TGF-ß induced pulmonary fibrosis, in converse, the direct role of EGFR induction of the TGF-ß pathway in the lung is unknown. The αvß6 integrin is an important in vivo activator of TGF-ß activation in the lung. Immunohistochemical analysis of αvß6 protein expression and bronchoalveolar analysis of TGF-ß pathway signaling indicates activation of the αvß6/TGF-ß pathway only at later time points after lung fibrosis was already established in the TGF-α model. To determine the contribution of the αvß6/TGF-ß pathway on the progression of established fibrotic disease, TGF-α transgenic mice were administered Dox for 4 wk, which leads to extensive fibrosis; these mice were then treated with a function-blocking anti-αvß6 antibody with continued administration of Dox for an additional 4 wk. Compared with TGF-α transgenic mice treated with control antibody, αvß6 inhibition significantly attenuated pleural thickening and altered the decline in lung mechanics. To test the effects of genetic loss of the ß6 integrin, TGF-α transgenic mice were mated with ß6-null mice and the degree of fibrosis was compared in adult mice following 8 wk of Dox administration. Genetic ablation of the ß6 integrin attenuated histological and physiological changes in the lungs of TGF-α transgenic mice although a significant degree of fibrosis still developed. In summary, inhibition of the ß6 integrin led to a modest, albeit significant, effect on pleural thickening and lung function decline observed with TGF-α-induced pulmonary fibrosis. These data support activation of the αvß6/TGF-ß pathway as a secondary effect contributing to TGF-α-induced pleural fibrosis and suggest a complex contribution of multiple mediators to the maintenance of progressive fibrosis in the lung.


Assuntos
Integrinas/antagonistas & inibidores , Fibrose Pulmonar/patologia , Fator de Crescimento Transformador alfa/farmacologia , Animais , Antibacterianos/toxicidade , Anticorpos Neutralizantes , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Lavagem Broncoalveolar , Colágeno , Doxiciclina/toxicidade , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Técnicas Imunoenzimáticas , Integrinas/genética , Integrinas/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Crescimento Transformador beta/farmacologia , Uteroglobina/fisiologia
14.
Hum Gene Ther Clin Dev ; 24(3): 116-26, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24007469

RESUMO

Glioblastoma multiforme (GBM) is the most common malignant primary brain cancer in adults; it carries a dismal prognosis despite improvements in standard of care. We developed a combined gene therapy strategy using (1) herpes simplex type 1-thymidine kinase in conjunction with the cytotoxic prodrug ganciclovir to kill actively proliferating tumor cells and (2) doxycycline (DOX)-inducible Fms-like tyrosine kinase 3 ligand (Flt3L), an immune stimulatory molecule that induces anti-GBM immunity. As a prelude to a phase I clinical trial, we examined the efficacy and safety of this approach (Muhammad et al., 2010, 2012). In the present article, we investigated the efficacy and safety of the "off-label" use of the antibiotic DOX to turn on the high-capacity adenoviral vector (HC-Ad) encoding therapeutic Flt3L expression. DOX-inducible Flt3L expression in male Lewis rats was assessed using DOX doses of 30.8 mg/kg/day (low-DOX) or 46.2 mg/kg/day (high-DOX), which are allometrically equivalent (Voisin et al., 1990) to the human doses that are recommended for the treatment of infections: 200 or 300 mg/day. Naïve rats were intracranially injected with 1×10(9) viral particles of HC-Ad-TetOn-Flt3L, and expression of the therapeutic transgene, that is, Flt3L, was assessed using immunohistochemistry in brain sections after 2 weeks of DOX administration via oral gavage. The results show robust expression of Flt3L in the rat brain parenchyma in areas near the injection site in both the low-DOX and the high-DOX groups, suggesting that Flt3L will be expressed in human glioma patients at a DOX dose of 200 or 300 mg/day. These doses have been approved by the U.S. Food and Drug Administration to treat infections in humans and would thus be considered safe for an off-label use to treat GBM patients undergoing HC-Ad-mediated gene therapy in a phase I clinical trial.


Assuntos
Neoplasias Encefálicas/terapia , Doxiciclina/toxicidade , Terapia Genética , Glioblastoma/terapia , Transgenes/efeitos dos fármacos , Tirosina Quinase 3 Semelhante a fms/genética , Animais , Ensaios Clínicos Fase I como Assunto , Doxiciclina/farmacologia , Avaliação Pré-Clínica de Medicamentos , Expressão Gênica/efeitos dos fármacos , Masculino , Ratos , Ratos Endogâmicos Lew , Tirosina Quinase 3 Semelhante a fms/metabolismo
15.
Genesis ; 51(1): 41-9, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22927121

RESUMO

Macrophages play an essential role in tissue homeostasis, innate immunity, inflammation, and wound repair. Macrophages are also essential during development, severely limiting the use of mouse models in which these cells have been constitutively deleted. Consequently, we have developed a transgenic model of inducible macrophage depletion in which macrophage-specific induction of the cytotoxic diphtheria toxin A chain (DTA) is achieved by administration of doxycycline. Induction of the DTA protein in transgenic animals resulted in a significant 50% reduction in CD68+ macrophages of the liver, spleen, and bone over a period of 6 weeks. Pertinently, the macrophages remaining after doxycycline treatment were substantially smaller and are functionally impaired as shown by reduced inflammatory cytokine production in response to lipopolysaccharide. This inducible model of macrophage depletion can now be utilized to determine the role of macrophages in both development and animal models of chronic inflammatory diseases.


Assuntos
Macrófagos/fisiologia , Camundongos Transgênicos , Modelos Animais , Animais , Antígenos CD/imunologia , Antígenos de Diferenciação Mielomonocítica/imunologia , Osso e Ossos/citologia , Citocinas/imunologia , Toxina Diftérica/genética , Doxiciclina/toxicidade , Terapia de Imunossupressão , Lipopolissacarídeos/imunologia , Fígado/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Camundongos , Fragmentos de Peptídeos/genética , Baço/citologia
16.
Braz. dent. j ; 23(5): 527-533, Sept.-Oct. 2012. ilus, graf, tab
Artigo em Inglês | LILACS | ID: lil-660355

RESUMO

The aim of the present study was to evaluate the capacity of some root canal irrigants to induce genetic damage and/or cellular death in vitro. Murine fibroblast cells were exposed to ethylenediaminetetraacetic acid (EDTA), sodium hypochlorite (NaOCl), MTAD™ and citric acid in increasing concentrations for 3 h at 37ºC. The negative control group was treated with vehicle control (phosphate buffer solution - PBS) for 3 h at 37°C, and the positive control group was treated with methylmetanesulfonate, 1 μM. for 3 h at 37°C. Cytotoxicity was assessed by the trypan blue test and genotoxicity was evaluated by the single cell gel (comet) assay. The results showed that exposure to 2.5% and 5% NaOCl and 8.5% citric acid resulted in a significant cytotoxic effect. NaOCl, EDTA and citric acid did not produce genotoxic effects with respect to the comet assay data for all evaluated concentrations. Although MTAD was not a cytotoxic agent, it showed significant genotoxic effects at all tested concentrations (ANOVA and Tukey's test; p<0.05). NaOCl, EDTA and citric acid were found to be cytotoxic in a dose-dependent manner, but they were not genotoxic. MTAD did not cause cell death, but presented genotoxic effects.


O objetivo do presente estudo foi avaliar a capacidade de alguns irrigantes endodônticos em induzir danos genéticos e/ou morte celular in vitro. Células de fibroblastos murinos foram expostas ao ácido etilenodiaminotetracético (EDTA), hipoclorito de sódio (NaOCl), MTAD™ e ácido cítrico em concentrações crescentes durante 3 h a 37°C. O grupo controle negativo foi tratado com solução tampão fosfato - PBS por 3 h a 37° C e o grupo controle positivo foi tratado com metilmetanesulfonato a 1 μM por 3 h a 37° C. A citotoxicidade foi testada pelo azul de tripan e a genotoxicidade foi avaliada pelo teste do cometa. Os resultados apontaram que a exposição ao NaOCl a 2,5% e 5%, e ácido cítrico a 21% resultou em efeitos citotóxicos significativos. O NaOCl, EDTA e o ácido cítrico não produziram efeitos genotóxicos no que diz respeito aos dados obtidos pelo ensaio do Cometa em todas as concentrações testadas. Embora o MTAD não tenha sido um agente citotóxico, mostrou efeitos genotóxicos significativos em todas as concentrações testadas (ANOVA e teste de Tuckey; p<0,05). O NaOCl, o EDTA e o ácido cítrico mostraram-se citotóxicos de maneira dose-dependente, mas não genotóxicos. Por outro lado, apesar do MTAD não ter causado a morte celular, foi genotóxico em todas as concentrações testadas.


Assuntos
Animais , Camundongos , Morte Celular/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Dentina/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Mutagênicos , Irrigantes do Canal Radicular/toxicidade , Análise de Variância , Linhagem Celular , Ensaio Cometa , Ácido Cítrico/toxicidade , Doxiciclina/toxicidade , Ácido Edético/toxicidade , Fibroblastos/citologia , Polissorbatos/toxicidade , Hipoclorito de Sódio/toxicidade , Azul Tripano/química
17.
Drug Chem Toxicol ; 35(3): 334-40, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22289567

RESUMO

Doxycycline (DOX) is a broad-spectrum tetracycline antibiotic used in the treatment of many infections. In this study, the genotoxic and cytotoxic effects of DOX in cultured human peripheral blood lymphocytes were investigated by measuring chromosome aberrations (CAs), cytokinesis-block micronucleus (CBMN) assay, mitotic index (MI), and nuclear division index (NDI). Cultures were treated with DOX at three concentrations (2, 4, and 6 µg/mL) for 48 hours. Mitomycin C (MMC) was used as a positive control. All the tested concentrations of DOX for MI and the higher concentrations (4 and 6 µg/mL) for NDI significantly decreased mitotic activity. However, there are no significant differences between negative control and all the tested concentrations of DOX for CA and MN frequencies. In conclusion, our results indicate that DOX has a cytotoxic effect, but not a genotoxic effect, on human peripheral blood lymphocyte cultures. Further detailed studies, especially about the cell-cycle kinetics of DOX, are required to elucidate the decreases in dividing cells and make a possible risk assessment on cells of patients receiving therapy with this drug. Further, if the specific cytostatic and cytotoxic potential of DOX to different types of cancer cells is investigated in detail, it may also have been used as an antitumoral drug.


Assuntos
Aberrações Cromossômicas/efeitos dos fármacos , Doxiciclina/análogos & derivados , Linfócitos/efeitos dos fármacos , Análise de Variância , Células Cultivadas , Relação Dose-Resposta a Droga , Doxiciclina/química , Doxiciclina/toxicidade , Humanos , Testes para Micronúcleos , Índice Mitótico , Estrutura Molecular , Testes de Toxicidade
18.
Oncogene ; 30(12): 1402-12, 2011 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-21076466

RESUMO

The transcription factor nuclear factor kappa B (NF-κB) is activated in human breast cancer tissues and cell lines. However, it is unclear whether NF-κB activation is a consequence of tumor formation or a contributor to tumor development. We developed a doxycycline (dox)-inducible mouse model, termed DNMP, to inhibit NF-κB activity specifically within the mammary epithelium during tumor development in the polyoma middle T oncogene (PyVT) mouse mammary tumor model. DNMP females and PyVT littermate controls were treated with dox from 4 to 12 weeks of age. We observed an increase in tumor latency and a decrease in final tumor burden in DNMP mice compared with PyVT controls. A similar effect with treatment from 8 to 12 weeks indicates that outcome is independent of effects on postnatal virgin ductal development. In both cases, DNMP mice were less likely to develop lung metastases than controls. Treatment from 8 to 9 weeks was sufficient to impact primary tumor formation. Inhibition of NF-κB increases apoptosis in hyperplastic stages of tumor development and decreases proliferation at least in part by reducing Cyclin D1 expression. To test the therapeutic potential of NF-κB inhibition, we generated palpable tumors by orthotopic injection of PyVT cells and then treated systemically with the NF-κB inhibitor thymoquinone (TQ). TQ treatment resulted in a reduction in tumor volume and weight as compared with vehicle-treated control. These data indicate that epithelial NF-κB is an active contributor to tumor progression and demonstrate that inhibition of NF-κB could have a significant therapeutic impact even at later stages of mammary tumor progression.


Assuntos
Epitélio/metabolismo , Epitélio/patologia , Neoplasias Mamárias Animais/metabolismo , Neoplasias Mamárias Animais/patologia , NF-kappa B/metabolismo , Carga Tumoral , Animais , Apoptose , Linhagem Celular Tumoral , Modelos Animais de Doenças , Doxiciclina/toxicidade , Epitélio/efeitos dos fármacos , Feminino , Glândulas Mamárias Animais/efeitos dos fármacos , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Animais/patologia , Neoplasias Mamárias Animais/induzido quimicamente , Camundongos , Camundongos Transgênicos , NF-kappa B/antagonistas & inibidores
19.
Cancer Res ; 69(10): 4286-93, 2009 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-19401449

RESUMO

The RAS pathway is one of the most frequently deregulated pathways in cancer. RAS signals through multiple effector pathways, including the RAF/mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) kinase (MEK)/ERK MAPK and phosphatidylinositol 3-kinase (PI3K)-AKT signaling cascades. The oncogenic potential of these effector pathways is illustrated by the frequent occurrence of activating mutations in BRAF and PIK3CA as well as loss-of-function mutations in the tumor suppressor PTEN, a negative regulator of PI3K. Previous studies have found that whereas BRAF mutant cancers are highly sensitive to MEK inhibition, RAS mutant cancers exhibit a more variable response. The molecular mechanisms responsible for this heterogeneous response remain unclear. In this study, we show that PI3K pathway activation strongly influences the sensitivity of RAS mutant cells to MEK inhibitors. Activating mutations in PIK3CA reduce the sensitivity to MEK inhibition, whereas PTEN mutations seem to cause complete resistance. We further show that down-regulation of PIK3CA resensitizes cells with co-occurring KRAS and PIK3CA mutations to MEK inhibition. At the molecular level, the dual inhibition of both pathways seems to be required for complete inhibition of the downstream mammalian target of rapamycin effector pathway and results in the induction of cell death. Finally, we show that whereas inactivation of either the MEK or PI3K pathway leads to partial tumor growth inhibition, targeted inhibition of both pathways is required to achieve tumor stasis. Our study provides molecular insights that help explain the heterogeneous response of KRAS mutant cancers to MEK pathway inhibition and presents a strong rationale for the clinical testing of combination MEK and PI3K targeted therapies.


Assuntos
Benzamidas/toxicidade , Difenilamina/análogos & derivados , Doxiciclina/toxicidade , Genes ras/genética , Mutação , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Animais , Antineoplásicos/toxicidade , Apoptose/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Neoplasias do Colo/enzimologia , Neoplasias do Colo/genética , Difenilamina/toxicidade , Ativação Enzimática , Feminino , Células HCT116 , Humanos , Camundongos , Camundongos Nus , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transplante Heterólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA