Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Nanomedicine ; 13(6): 1983-1992, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28434932

ABSTRACT

Inhalation delivery of prostaglandin E (PGE2) in combination with selected siRNA(s) was proposed for the efficient treatment of idiopathic pulmonary fibrosis (IPF). Nanostructured lipid carriers (NLC) were used as a delivery system for PGE2 with and without siRNAs targeted to MMP3, CCL12, and HIF1Alpha mRNAs. The model of IPF was developed in SKH1 mice by intratracheal administration of bleomycin at a dose of 1.5U/kg. Results showed that NLC-PGE2 in combination with three siRNAs delivered locally to the lungs by inhalation markedly reduced mouse body mass, substantially limited hydroxyproline content in the lungs and disturbances of the mRNAs and protein expression, restricted lung tissue damage and prevented animal mortality. Our data provide evidence that IPF can be effectively treated by inhalation of the NLC-PGE2 in combination with siRNAs delivered locally into the lungs. This effect could not be achieved by using NLC containing just PGE2 or siRNA(s) alone.


Subject(s)
Drug Delivery Systems/methods , Idiopathic Pulmonary Fibrosis/therapy , Nanoparticles/administration & dosage , Prostaglandins/therapeutic use , RNA, Small Interfering/administration & dosage , Administration, Inhalation , Animals , Antibiotics, Antineoplastic/pharmacology , Bleomycin/pharmacology , Combined Modality Therapy , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/pathology , Lipids/chemistry , Mice , Mice, Hairless , RNA, Small Interfering/genetics
2.
Neurotoxicology ; 54: 72-80, 2016 05.
Article in English | MEDLINE | ID: mdl-27045884

ABSTRACT

Following the postnatal decline of cell proliferation in the mammalian central nervous system, the adult brain retains progenitor cells with stem cell-like properties in the subventricular zone (SVZ) and the subgranular zone (SGZ) of the hippocampus. Brain injury can stimulate proliferation and redirect the migration pattern of SVZ precursor cells to the injury site. Sublethal exposure to the neurotoxicant trimethyltin (TMT) causes dose-dependent necrosis and apoptosis in the hippocampus dentate gyrus and increases SGZ stem cell proliferation to generate new granule cells. To determine whether SVZ cells also contribute to the repopulation of the TMT-damaged dentate gyrus, 6-8 week old male C3H mice were injected with the carbocyanine dye spDiI and bromodeoxyuridine (80mg/kg; ip.) to label ventricular cells prior to TMT exposure. The presence of labeled cells in hippocampus was determined 7 and 28days after TMT exposure. No significant change in the number of BrdU(+) and spDiI(+) cells was observed in the dentate gyrus 7days after TMT treatment. However, 28days after TMT treatment there was a 3-4 fold increase in the number of spDiI-labeled cells in the hippocampal hilus and dentate gyrus. Few spDiI(+) cells stained positive for the mature phenotypic markers NeuN or GFAP, suggesting they may represent undifferentiated cells. A small percentage of migrating cells were BrdU(+)/spDiI(+), indicating some newly produced, SVZ- derived precursors migrated to the hippocampus. Taken together, these data suggest that TMT-induced injury of the hippocampus can stimulate the migration of ventricular zone-derived cells to injured dentate gyrus.


Subject(s)
Brain Injuries/pathology , Cell Movement/drug effects , Hippocampus/pathology , Lateral Ventricles/pathology , Trimethyltin Compounds/toxicity , Amino Acids/metabolism , Animals , Bromodeoxyuridine/metabolism , Functional Laterality , Glial Fibrillary Acidic Protein/metabolism , Lateral Ventricles/drug effects , Male , Mice , Mice, Inbred C3H , Neurogenesis/drug effects , Neurogenesis/physiology
3.
Breast Cancer Res Treat ; 151(1): 57-73, 2015 May.
Article in English | MEDLINE | ID: mdl-25859923

ABSTRACT

Metabotropic glutamate receptor 1 (mGluR1/Grm1) is a member of the G-protein-coupled receptor superfamily, which was once thought to only participate in synaptic transmission and neuronal excitability, but has more recently been implicated in non-neuronal tissue functions. We previously described the oncogenic properties of Grm1 in cultured melanocytes in vitro and in spontaneous melanoma development with 100 % penetrance in vivo. Aberrant mGluR1 expression was detected in 60-80 % of human melanoma cell lines and biopsy samples. As most human cancers are of epithelial origin, we utilized immortalized mouse mammary epithelial cells (iMMECs) as a model system to study the transformative properties of Grm1. We introduced Grm1 into iMMECs and isolated several stable mGluR1-expressing clones. Phenotypic alterations in mammary acinar architecture were assessed using three-dimensional morphogenesis assays. We found that mGluR1-expressing iMMECs exhibited delayed lumen formation in association with decreased central acinar cell death, disrupted cell polarity, and a dramatic increase in the activation of the mitogen-activated protein kinase pathway. Orthotopic implantation of mGluR1-expressing iMMEC clones into mammary fat pads of immunodeficient nude mice resulted in mammary tumor formation in vivo. Persistent mGluR1 expression was required for the maintenance of the tumorigenic phenotypes in vitro and in vivo, as demonstrated by an inducible Grm1-silencing RNA system. Furthermore, mGluR1 was found be expressed in human breast cancer cell lines and breast tumor biopsies. Elevated levels of extracellular glutamate were observed in mGluR1-expressing breast cancer cell lines and concurrent treatment of MCF7 xenografts with glutamate release inhibitor, riluzole, and an AKT inhibitor led to suppression of tumor progression. Our results are likely relevant to human breast cancer, highlighting a putative role of mGluR1 in the pathophysiology of breast cancer and the potential of mGluR1 as a novel therapeutic target.


Subject(s)
Breast Neoplasms/genetics , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Receptors, Metabotropic Glutamate/genetics , Animals , Apoptosis/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic/drug effects , Heterocyclic Compounds, 3-Ring/administration & dosage , Humans , MCF-7 Cells , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/pathology , Mice , Receptors, Metabotropic Glutamate/biosynthesis , Riluzole/administration & dosage , Xenograft Model Antitumor Assays
4.
Urology ; 84(5): 1249.e9-15, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25443947

ABSTRACT

OBJECTIVE: To assess the effectiveness of l-cystine dimethyl ester (CDME), an inhibitor of cystine crystal growth, for the treatment of cystine urolithiasis in an Slc3a1 knockout mouse model of cystinuria. MATERIALS AND METHODS: CDME (200 µg per mouse) or water was delivered by gavage daily for 4 weeks. Higher doses by gavage or in the water supply were administered to assess organ toxicity. Urinary amino acids and cystine stones were analyzed to assess drug efficacy using several analytical methods. RESULTS: Treatment with CDME led to a significant decrease in stone size compared with that of the water group (P = .0002), but the number of stones was greater (P = .005). The change in stone size distribution between the 2 groups was evident by micro computed tomography. Overall, cystine excretion in urine was the same between the 2 groups (P = .23), indicating that CDME did not interfere with cystine metabolism. Scanning electron microscopy analysis of cystine stones from the CDME group demonstrated a change in crystal habit, with numerous small crystals. l-cysteine methyl ester was detected by ultra-performance liquid chromatography-mass spectrometer in stones from the CDME group only, indicating that a CDME metabolite was incorporated into the crystal structure. No pathologic changes were observed at the doses tested. CONCLUSION: These data demonstrate that CDME promotes formation of small stones but does not prevent stone formation, consistent with the hypothesis that CDME inhibits cystine crystal growth. Combined with the lack of observed adverse effects, our findings support the use of CDME as a viable treatment for cystine urolithiasis.


Subject(s)
Cystine/analogs & derivatives , Cystinuria/drug therapy , Urolithiasis/drug therapy , Amino Acid Transport Systems, Basic/genetics , Amino Acid Transport Systems, Neutral/genetics , Animals , Chromatography, Liquid , Cystine/chemistry , Cystinuria/urine , Male , Mass Spectrometry , Mice , Mice, Knockout , Microscopy, Electron, Scanning , X-Ray Microtomography
5.
Mol Ther ; 22(3): 547-553, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24394185

ABSTRACT

The blood-brain barrier (BBB) presents a major challenge to effective treatment of neurological disorders, including lysosomal storage diseases (LSDs), which frequently present with life-shortening and untreatable neurodegeneration. There is considerable interest in methods for intravenous delivery of lysosomal proteins across the BBB but for the most part, levels achievable in the brain of mouse models are modest and increased lifespan remains to be demonstrated. In this study, we have investigated delivery across the BBB using a mouse model of late-infantile neuronal ceroid lipofuscinosis (LINCL), a neurodegenerative LSD caused by loss of tripeptidyl peptidase I (TPP1). We have achieved supraphysiological levels of TPP1 throughout the brain of LINCL mice by intravenous (IV) coadministration of recombinant TPP1 with a 36-residue peptide that contains polylysine and a low-density lipoprotein receptor binding sequence from apolipoprotein E. Importantly, IV administration of TPP1 with the peptide significantly reduces brain lysosomal storage, increases lifespan and improves neurological function. This simple "mix and inject" method is immediately applicable towards evaluation of enzyme replacement therapy to the brain in preclinical models and further exploration of its clinical potential is warranted.


Subject(s)
Aminopeptidases/metabolism , Apolipoproteins E/metabolism , Blood-Brain Barrier/metabolism , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Neuronal Ceroid-Lipofuscinoses/drug therapy , Neuronal Ceroid-Lipofuscinoses/physiopathology , Peptides/administration & dosage , Serine Proteases/metabolism , Administration, Intravenous , Animals , CHO Cells , Cricetulus , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Replacement Therapy , Humans , Lysosomes/metabolism , Mice , Neuronal Ceroid-Lipofuscinoses/pathology , Recombinant Proteins , Tripeptidyl-Peptidase 1
6.
Mol Ther Nucleic Acids ; 2: e92, 2013 May 14.
Article in English | MEDLINE | ID: mdl-23673539

ABSTRACT

U1 Adaptor is a recently discovered oligonucleotide-based gene-silencing technology with a unique mechanism of action that targets nuclear pre-mRNA processing. U1 Adaptors have two distinct functional domains, both of which must be present on the same oligonucleotide to exert their gene-silencing function. Here, we present the first in vivo use of U1 Adaptors by targeting two different human genes implicated in melanomagenesis, B-cell lymphoma 2 (BCL2) and metabotropic glutamate receptor 1 (GRM1), in a human melanoma cell xenograft mouse model system. Using a newly developed dendrimer delivery system, anti-BCL2 U1 Adaptors were very potent and suppressed tumor growth at doses as low as 34 µg/kg with twice weekly intravenous (iv) administration. Anti-GRM1 U1 Adaptors suppressed tumor xenograft growth with similar potency. Mechanism of action was demonstrated by showing target gene suppression in tumors and by observing that negative control U1 Adaptors with just one functional domain show no tumor suppression activity. The anti-BCL2 and anti-GRM1 treatments were equally effective against cell lines harboring either wild-type or a mutant V600E B-RAF allele, the most common mutation in melanoma. Treatment of normal immune-competent mice (C57BL6) indicated no organ toxicity or immune stimulation. These proof-of-concept studies represent an in-depth (over 800 mice in ~108 treatment groups) validation that U1 Adaptors are a highly potent gene-silencing therapeutic and open the way for their further development to treat other human diseases.Molecular Therapy - Nucleic Acids (2013) 2, e92; doi:10.1038/mtna.2013.24; published online 14 May 2013.

7.
Carcinogenesis ; 34(9): 2184-91, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23671133

ABSTRACT

Each enantiomer of the diastereomeric pair of bay-region dibenz[a,h]anthracene 3,4-diol-1,2-epoxides in which the benzylic 4-hydroxyl group and epoxide oxygen are either cis (isomer 1) or trans (isomer 2) were evaluated for mutagenic activity. In strains TA 98 and TA 100 of Salmonella typhimurium, the diol epoxide with (1S,2R,3S,4R) absolute configuration [(-)-diol epoxide-1] had the highest mutagenic activity. In Chinese hamster V-79 cells, the diol epoxide with (1R,2S,3S,4R) absolute configuration [(+)-diol epoxide-2] had the highest mutagenic activity. The (1R,2S,3R,4S) diol epoxide [(+)-diol epoxide-1] also had appreciable activity, whereas the other two bay-region diol epoxide enantiomers had very low activity. In tumor studies, the (1R,2S,3S,4R) enantiomer was the only diol epoxide isomer tested that had strong activity as a tumor initiator on mouse skin and in causing lung and liver tumors when injected into newborn mice. This stereoisomer was about one-third as active as the parent hydrocarbon, dibenz[a,h]anthracene as a tumor initiator on mouse skin; it was several-fold more active than dibenz[a,h]anthracene as a lung and liver carcinogen when injected into newborn mice. (-)-(3R,4R)-3ß,4α-dihydroxy-3,4-dihydro-dibenz[a,h]anthracene [(-)-3,4-dihydrodiol] was slightly more active than dibenz[a,h]anthracene as a tumor initiator on mouse skin, whereas (+)-(3S,4S)-3α,4ß-dihydroxy-3,4-dihydro-dibenz[a,h]anthracene [(+)-3,4-dihydrodiol] had only very weak activity. The present investigation and previous studies with the corresponding four possible enantiopure bay-region diol epoxide enantiomers/diastereomers of benzo[a]pyrene, benz[a]anthracene, chrysene, benzo[c]phenanthrene, dibenz[c,h]acridine, dibenz[a,h]acridine and dibenz[a,h]anthracene indicate that the bay-region diol epoxide enantiomer with [R,S,S,R] absolute stereochemistry has high tumorigenic activity on mouse skin and in newborn mice.


Subject(s)
Carcinogenesis/pathology , Chrysenes/pharmacology , Epoxy Compounds/pharmacology , Skin Neoplasms/chemically induced , Animals , Carcinogenesis/chemically induced , Carcinogenesis/chemistry , Chrysenes/chemistry , Chrysenes/toxicity , Cricetinae , Epoxy Compounds/toxicity , Humans , Mice , Mutagenesis/drug effects , Mutagens/pharmacology , Mutagens/toxicity , Salmonella typhimurium/drug effects , Salmonella typhimurium/genetics , Skin Neoplasms/pathology , Stereoisomerism , Structure-Activity Relationship
8.
Eur J Pharm Biopharm ; 84(2): 335-44, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23228437

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, and often fatal form of interstitial lung disease. We hypothesized that the local pulmonary delivery of prostaglandin E2 (PGE2) by liposomes can be used for the effective treatment of IPF. To test this hypothesis, we used a murine model of bleomycin-induced IPF to evaluate liposomal delivery of PGE2 topically to the lungs. Animal survival, body weight, hydroxyproline content in the lungs, lung histology, mRNA, and protein expression were studied. After inhalation delivery, liposomes accumulated predominately in the lungs. In contrast, intravenous administration led to the accumulation of liposomes mainly in kidney, liver, and spleen. Liposomal PGE2 prevented the disturbances in the expression of many genes associated with the development of IPF, substantially restricted inflammation and fibrotic injury in the lung tissues, prevented decrease in body weight, limited hydroxyproline accumulation in the lungs, and virtually eliminated mortality of animals after intratracheal instillation of bleomycin. In summary, our data provide evidence that pulmonary fibrosis can be effectively treated by the inhalation administration of liposomal form of PGE2 into the lungs. The results of the present investigations make the liposomal form of PGE2 an attractive drug for the effective inhalation treatment of idiopathic pulmonary fibrosis.


Subject(s)
Dinoprostone/administration & dosage , Idiopathic Pulmonary Fibrosis/drug therapy , Administration, Intravenous , Animals , Bleomycin/toxicity , Body Weight/drug effects , Disease Models, Animal , Hydroxyproline/metabolism , Idiopathic Pulmonary Fibrosis/chemically induced , Immunohistochemistry , Kidney/drug effects , Liposomes/administration & dosage , Liposomes/metabolism , Liver/drug effects , Lung/drug effects , Lung/metabolism , Mice , RNA, Messenger/metabolism , Spleen/drug effects , Time Factors , Tissue Distribution
9.
Cancer Prev Res (Phila) ; 5(7): 963-72, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22581815

ABSTRACT

To develop a relevant mouse model for prostate cancer prevention research, we administered a dietary carcinogen, 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP), to CYP1A-humanized mice. In comparison with mouse Cyp1a2, human CYP1A2 preferentially activates PhIP to a proximate carcinogen. Following a single oral dose of PhIP (200 mg/kg body weight), we observed inflammation, atrophy of acini, low-grade prostatic intraepithelial neoplasia (PIN; after 20 weeks), and high-grade PIN (HgPIN; after 30 to 50 weeks) in dorsolateral, ventral, and coagulating anterior prostate glands of these mice. These lesions were androgen receptor positive and featured the loss of expression of the basal cell marker p63 and the tumor suppressor PTEN. Similar to human prostate carcinogenesis, glutathione S-transferase P1 (GSTP1) expression was lost or partially lost in HgPIN. E-Cadherin expression was also lost in HgPIN. The expression of DNA methyltransferase 1 was elevated, possibly to enhance promoter hypermethylation for the silencing of GSTP1 and E-cadherin. Prostate carcinogenesis was promoted by a high-fat stress diet, resulting in HgPIN that developed earlier and in advanced lesions displayed features consistent with carcinoma in situ. This dietary carcinogen-induced prostate cancer model, recapitulating important features of early human prostate carcinogenesis, constitutes a new experimental system for prostate cancer research.


Subject(s)
Cell Transformation, Neoplastic/pathology , Cytochrome P-450 CYP1A2/physiology , Diet, High-Fat/adverse effects , Disease Models, Animal , Imidazoles/toxicity , Prostatic Intraepithelial Neoplasia/pathology , Prostatic Neoplasms/pathology , Animals , Cadherins/metabolism , Carcinogens/toxicity , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/metabolism , Female , Glutathione S-Transferase pi/metabolism , Humans , Immunoenzyme Techniques , Male , Mice , Mice, Inbred C57BL , Prostatic Intraepithelial Neoplasia/chemically induced , Prostatic Intraepithelial Neoplasia/metabolism , Prostatic Neoplasms/chemically induced , Prostatic Neoplasms/metabolism
10.
Nanomedicine (Lond) ; 7(2): 185-97, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22339132

ABSTRACT

AIMS: To develop an approach for the effective treatment of ovarian tumor, prevention of metastases and limitation of side effects. MATERIALS & METHODS: In order to combine chemotherapy with genotherapy, we constructed a nanoscale-based tumor-targeted system containing two anticancer drugs, two antisense oligonucleotides (suppressors of cellular resistance) and a ligand specific to receptors overexpressed in cancer cells. The system was tested in a mouse metastatic xenograft model using tumor isolates from patients with ovarian carcinoma. RESULTS: Treatment with the proposed nanosystem decreased primary tumor, prevented intraperitoneal metastases and limited adverse side effects. CONCLUSIONS: The advantages of the proposed targeted complex treatment of primary aggressive ovarian tumor and prevention of metastases make the proposed approach promising for clinical applications.


Subject(s)
Antineoplastic Agents/administration & dosage , Carcinoma/secondary , Carcinoma/therapy , Genetic Therapy/methods , Molecular Targeted Therapy/methods , Nanocapsules/therapeutic use , Ovarian Neoplasms/therapy , Carcinoma/pathology , Combined Modality Therapy , Female , Humans , Nanocapsules/chemistry , Ovarian Neoplasms/pathology , Treatment Outcome , Tumor Cells, Cultured
12.
J Agric Food Chem ; 59(21): 11862-71, 2011 Nov 09.
Article in English | MEDLINE | ID: mdl-21932846

ABSTRACT

The aim of this study was to investigate the effects of (-)-epigallocatechin-3-gallate (EGCG) on newly developed high-fat/Western-style diet-induced obesity and symptoms of metabolic syndrome. Male C57BL/6J mice were fed a high fat/Western-style (HFW; 60% energy as fat and lower levels of calcium, vitamin D(3), folic acid, choline bitartrate, and fiber) or HFW with EGCG (HFWE; HFW with 0.32% EGCG) diet for 17 wks. As a comparison, two other groups of mice fed a low-fat diet (LF; 10% energy as fat) and high-fat diet (HF; 60% energy as fat) were also included. The HFW group developed more body weight gain and severe symptoms of metabolic syndrome than the HF group. The EGCG treatment significantly reduced body weight gain associated with increased fecal lipids and decreased blood glucose and alanine aminotransferase (ALT) levels compared to those of the HFW group. Fatty liver incidence, liver damage, and liver triglyceride levels were also decreased by the EGCG treatment. Moreover, the EGCG treatment attenuated insulin resistance and levels of plasma cholesterol, monocyte chemoattractant protein-1 (MCP-1), C-reactive protein (CRP), interlukin-6 (IL-6), and granulocyte colony-stimulating factor (G-CSF). Our results demonstrate that the HFW diet produces more severe symptoms of metabolic syndrome than the HF diet and that the EGCG treatment can alleviate these symptoms and body fat accumulation. The beneficial effects of EGCG are associated with decreased lipid absorption and reduced levels of inflammatory cytokines.


Subject(s)
Catechin/analogs & derivatives , Diet, High-Fat/adverse effects , Metabolic Syndrome/drug therapy , Obesity/drug therapy , Plant Extracts/administration & dosage , Polyphenols/administration & dosage , Animals , Blood Glucose/analysis , Body Weight/drug effects , Catechin/administration & dosage , Disease Models, Animal , Humans , Male , Metabolic Syndrome/metabolism , Metabolic Syndrome/physiopathology , Mice , Mice, Inbred C57BL , Obesity/metabolism , Obesity/physiopathology
13.
Cancer Res ; 71(19): 6282-91, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21835894

ABSTRACT

Neurobehavioral stress has been shown to promote tumor growth and progression and dampen the immune system. In this study, we investigated whether inhibiting stress hormone production could inhibit the development of mammary carcinoma and metastasis in a rat model of breast carcinogenesis. To enhance ß-endorphin (BEP), the endogenous opioid polypeptide that boosts immune activity and decreases stress, we generated BEP neurons by in vitro differentiation from fetal neuronal stem cells and transplanted them into the hypothalami of rats subjected to breast carcinogenesis. BEP-transplanted rats displayed a reduction in mammary tumor incidence, growth, malignancy rate, and metastasis compared with cortical cells-transplanted rats. BEP neuron transplants also reduced inflammation and epithelial to mesenchymal transition in the tumor tissues. In addition, BEP neuron transplants increased peripheral natural killer (NK) cell and macrophage activities, elevated plasma levels of antiinflammatory cytokines, and reduced plasma levels of inflammatory cytokines. Antimetastatic effects along with stimulation of NK cells and macrophages could be reversed by treatment with the opiate antagonist naloxone, the ß-receptor agonist metaproterenol, or the nicotine acetylcholine receptor antagonist methyllycaconitine. Together, our findings establish a protective role for BEP against the growth and metastasis of mammary tumor cells by altering autonomic nervous system activities that enhance innate immune function.


Subject(s)
Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/prevention & control , Neurons/transplantation , beta-Endorphin/metabolism , Adrenal Cortex/cytology , Animals , Autonomic Nervous System , Cell Differentiation , Cells, Cultured , Epithelial-Mesenchymal Transition , Female , Hypothalamus/immunology , Hypothalamus/metabolism , Hypothalamus/pathology , Immunity, Innate , Killer Cells, Natural/immunology , Macrophage Activation , Male , Mammary Neoplasms, Experimental/pathology , Neoplasm Grading , Neoplasm Metastasis , Neural Stem Cells/cytology , Neurons/immunology , Neurons/metabolism , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley
14.
Alcohol Clin Exp Res ; 34(11): 1879-87, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20662802

ABSTRACT

BACKGROUND: Altered fetal programming because of a suboptimal in utero environment has been shown to increase susceptibility to many diseases later in life. This study examined the effect of alcohol exposure in utero on N-nitroso-N-methylurea (NMU)-induced mammary cancer risk during adulthood. METHODS: Study 1: Pregnant Sprague Dawley rats were fed a liquid diet containing 6.7% ethanol (alcohol-fed), an isocaloric liquid diet (pair-fed), or rat chow ad libitum (ad lib-fed) from day 11 to 21 of gestation. At birth, female pups were cross-fostered to ad lib-fed control dams. Adult offspring were given an I.P. injection of NMU at a dose of 50 mg/kg body weight. Mammary glands were palpated for tumors twice a week, and rats were euthanized at 23 weeks postinjection. Study 2: To investigate the role of estradiol (E2), animals were exposed to the same in utero treatments but were not given NMU. Serum was collected during the preovulatory phase of the estrous cycle. RESULTS: At 16 weeks postinjection, overall tumor multiplicity was greater in the offspring from the alcohol-fed group compared to the control groups, indicating a decrease in tumor latency. At study termination, 70% of all animals possessed tumors. Alcohol-exposed animals developed more malignant tumors and more estrogen receptor-α-negative tumors relative to the control groups. In addition, IGF-binding protein-5 (IGFBP-5) mRNA and protein were decreased in tumors of alcohol-exposed animals. Study 2 showed that alcohol-fed animals had significantly increased circulating E2 when compared to either control group. CONCLUSIONS: These data indicate that alcohol exposure in utero increases susceptibility to mammary tumorigenesis in adulthood and suggest that alterations in the IGF and E2 systems may play a role in the underlying mechanism.


Subject(s)
Central Nervous System Depressants/toxicity , Ethanol/toxicity , Mammary Neoplasms, Animal/chemically induced , Mammary Neoplasms, Animal/pathology , Prenatal Exposure Delayed Effects/pathology , Animals , Blotting, Western , Disease Progression , Estradiol/physiology , Female , Immunohistochemistry , Insulin-Like Growth Factor Binding Protein 5/biosynthesis , Insulin-Like Growth Factor I/metabolism , Phenotype , Pregnancy , RNA/biosynthesis , RNA/genetics , Rats , Rats, Sprague-Dawley , Somatomedins/metabolism , Somatomedins/physiology
15.
Proc Natl Acad Sci U S A ; 107(23): 10737-42, 2010 Jun 08.
Article in English | MEDLINE | ID: mdl-20498076

ABSTRACT

Development of cancer cell resistance, low accumulation of therapeutic drug in the lungs, and severe adverse treatment side effects represent main obstacles to efficient chemotherapy of lung cancer. To overcome these difficulties, we propose inhalation local delivery of anticancer drugs in combination with suppressors of pump and nonpump cellular resistance. To test this approach, nanoscale-based delivery systems containing doxorubicin as a cell death inducer, antisense oligonucleotides targeted to MRP1 mRNA as a suppressor of pump resistance and to BCL2 mRNA as a suppressor of nonpump resistance, were developed and examined on an orthotopic murine model of human lung carcinoma. The experimental results show high antitumor activity and low adverse side effects of proposed complex inhalatory treatment that cannot be achieved by individual components applied separately. The present work potentially contributes to the treatment of lung cancer by describing a unique combinatorial local inhalation delivery of drugs and suppressors of pump and nonpump cellular resistance.


Subject(s)
Doxorubicin/therapeutic use , Drug Resistance, Neoplasm , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Oligonucleotides/pharmacology , Administration, Inhalation , Animals , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Down-Regulation/drug effects , Doxorubicin/administration & dosage , Drug Delivery Systems , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/genetics , Mice , Mice, Nude , Multidrug Resistance-Associated Proteins/genetics , Oligonucleotides/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2
16.
Food Chem Toxicol ; 48(1): 409-16, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19883714

ABSTRACT

The tea polyphenol (-)-epigallocatechin-3-gallate (EGCG) has been studied for chronic disease preventive effects, and is marketed as part of many dietary supplements. However, case-reports have associated the use of green tea-based supplements with liver toxicity. We studied the hepatotoxic effects of high dose EGCG in male CF-1 mice. A single dose of EGCG (1500 mg/kg, i.g.) increased plasma alanine aminotransferase (ALT) by 138-fold and reduced survival by 85%. Once-daily dosing with EGCG increased hepatotoxic response. Plasma ALT levels were increased 184-fold following two once-daily doses of 750 mg/kg, i.g. EGCG. Moderate to severe hepatic necrosis was observed following treatment with EGCG. EGCG hepatotoxicity was associated with oxidative stress including increased hepatic lipid peroxidation (5-fold increase), plasma 8-isoprostane (9.5-fold increase) and increased hepatic metallothionein and gamma-histone 2AX protein expression. EGCG also increased plasma interleukin-6 and monocyte chemoattractant protein-1. Our results indicate that higher bolus doses of EGCG are hepatotoxic to mice. Further studies on the dose-dependent hepatotoxic effects of EGCG and the underlying mechanisms are important given the increasing use of green tea dietary supplements, which may deliver much higher plasma and tissue concentrations of EGCG than tea beverages.


Subject(s)
Antioxidants/toxicity , Catechin/analogs & derivatives , Chemical and Drug Induced Liver Injury/pathology , Aldehydes/pharmacology , Animals , Antioxidants/pharmacokinetics , Biomarkers , Catechin/pharmacokinetics , Catechin/toxicity , Chromatography, High Pressure Liquid , Cysteine/urine , Cytokines/metabolism , Dose-Response Relationship, Drug , Immunohistochemistry , Lipid Peroxidation/drug effects , Liver Function Tests , Male , Metallothionein/metabolism , Mice , Oxidants/toxicity , Oxidative Stress/drug effects , Spectrometry, Mass, Electrospray Ionization , Survival Analysis
17.
Int J Cancer ; 124(6): 1270-5, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19058177

ABSTRACT

We previously demonstrated that oxidative stress subsequent to gastroesophageal reflux is an important driving force of esophageal adenocarcinoma (EAC) formation in the esophagogastroduodenal anastomosis (EGDA) rat model. This study investigated the possible tumor inhibitory effects of 2 antioxidants, alpha-tocopherol (389 and 778 ppm), N-acetylcysteine (NAC, 500 and 1,000 ppm), and their combination (389 and 500 ppm, respectively), as well as an antacid therapeutic agent, omeprazole (1,400 ppm). The rats were fed experimental diets 2 weeks after EGDA. All the animals were sacrificed 40 weeks after EGDA and the esophagi were harvested for histopathological examination. alpha-Tocopherol dose-dependently decreased the incidence of EAC (p = 0.03), with 778 ppm alpha-tocopherol reducing the incidence of EAC to 59% (16/27) in comparison with 84% (26/31) in the control group (p = 0.04). Supplementation of alpha-tocopherol also increased the serum concentration of alpha-tocopherol. NAC at 500 and 1,000 ppm did not significantly decrease EAC incidence; however, the combination of alpha-tocopherol 389 ppm and NAC 500 ppm significantly reduced the incidence of EAC to 55% (15/27) (p = 0.02). alpha-Tocopherol alone or in combination with NAC significantly reduced the number of infiltrating cells positively stained for 4-hydroxynonenal. Omeprazole showed only a slight nonsignificant inhibitory effect at the dose given. Our results suggest that supplementation with alpha-tocopherol inhibits the development of EAC in the rat EGDA model and similar inhibitory effect can be achieved when a lower dose of alpha-tocopherol is used in combination with NAC.


Subject(s)
Acetylcysteine/therapeutic use , Adenocarcinoma/prevention & control , Esophageal Neoplasms/prevention & control , Omeprazole/therapeutic use , alpha-Tocopherol/therapeutic use , Adenocarcinoma/etiology , Adenocarcinoma/pathology , Adenocarcinoma/surgery , Animals , Disease Models, Animal , Esophageal Neoplasms/etiology , Esophageal Neoplasms/pathology , Esophageal Neoplasms/surgery , Gastroesophageal Reflux/complications , Hydrogen-Ion Concentration , Male , Rats , Rats, Sprague-Dawley , Vitamin A/blood , alpha-Tocopherol/blood
18.
J Nutr ; 138(9): 1677-83, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18716169

ABSTRACT

In this study, we investigated the effects of the major green tea polyphenol, (-)-epigallocatechin-3-gallate (EGCG), on high-fat-induced obesity, symptoms of the metabolic syndrome, and fatty liver in mice. In mice fed a high-fat diet (60% energy as fat), supplementation with dietary EGCG treatment (3.2 g/kg diet) for 16 wk reduced body weight (BW) gain, percent body fat, and visceral fat weight (P < 0.05) compared with mice without EGCG treatment. The BW decrease was associated with increased fecal lipids in the high-fat-fed groups (r(2) = 0.521; P < 0.05). EGCG treatment attenuated insulin resistance, plasma cholesterol, and monocyte chemoattractant protein concentrations in high-fat-fed mice (P < 0.05). EGCG treatment also decreased liver weight, liver triglycerides, and plasma alanine aminotransferase concentrations in high-fat-fed mice (P < 0.05). Histological analyses of liver samples revealed decreased lipid accumulation in hepatocytes in mice treated with EGCG compared with high-fat diet-fed mice without EGCG treatment. In another experiment, 3-mo-old high-fat-induced obese mice receiving short-term EGCG treatment (3.2 g/kg diet, 4 wk) had decreased mesenteric fat weight and blood glucose compared with high-fat-fed control mice (P < 0.05). Our results indicate that long-term EGCG treatment attenuated the development of obesity, symptoms associated with the metabolic syndrome, and fatty liver. Short-term EGCG treatment appeared to reverse preexisting high-fat-induced metabolic pathologies in obese mice. These effects may be mediated by decreased lipid absorption, decreased inflammation, and other mechanisms.


Subject(s)
Catechin/analogs & derivatives , Dietary Fats/adverse effects , Fatty Liver/prevention & control , Metabolic Syndrome/prevention & control , Obesity/prevention & control , Tea/chemistry , Adipose Tissue/drug effects , Animals , Biomarkers , Body Weight , Catechin/administration & dosage , Catechin/pharmacology , Diet , Eating , Fatty Liver/drug therapy , Feces/chemistry , Lipids/analysis , Male , Metabolic Syndrome/drug therapy , Mice , Mice, Inbred C57BL , Obesity/drug therapy
19.
Brain Res ; 1203: 126-32, 2008 Apr 08.
Article in English | MEDLINE | ID: mdl-18316065

ABSTRACT

Sodium valproate (VPA) administered to neonatal mice causes cognitive and motor deficits similar to those observed in humans with autism. In an effort to further evaluate similarities between early VPA exposure and autism, the present study examined treated mice for deficits in social behavior and neuronal damage. BALB/c mice injected on P14 with 400 mg/kg VPA engaged in fewer social interactions (including ano-genital sniffs, allogrooming, and crawl-under/over behaviors) than control mice. Treated mice also exhibited reduced motor activity in a social context but were not significantly different from controls when motor activity was assessed in non-social settings. A second set of BALB/c mice were treated with VPA on P14 and sacrificed at different times thereafter for histopathological analysis. At time-points 12 and 24 h following VPA, treated mice had up to a 30-fold increase in the number of TUNEL-positive cells in the external granule cell layer of the cerebellum and a 10-fold increase in TUNEL-positive cells in the dentate gyrus of the hippocampus. These observations may provide a histopathological correlate for the social deficits observed following post-natal VPA exposure and supports the use of early VPA administration as an animal model for the study of autism.


Subject(s)
Anticonvulsants/toxicity , Apoptosis/drug effects , Behavioral Symptoms/chemically induced , Behavioral Symptoms/pathology , Valproic Acid/toxicity , Analysis of Variance , Animals , Animals, Newborn , Behavior, Animal/drug effects , Brain/drug effects , Brain/pathology , Cell Count , Female , In Situ Nick-End Labeling/methods , Male , Mice , Mice, Inbred BALB C , Motor Activity/drug effects , Sex Factors , Social Behavior , Time Factors
20.
Toxicology ; 246(1): 63-72, 2008 Apr 03.
Article in English | MEDLINE | ID: mdl-18280627

ABSTRACT

Administration of 0.4% clofibrate in the diet stimulated estradiol (E(2))-induced mammary carcinogenesis in the August-Copenhagen Irish (ACI) rat without having an effect on serum levels of E(2). This treatment stimulated by several-fold the NAD(P)H-dependent oxidative metabolism of E(2) and oleyl-CoA-dependent esterification of E(2) to 17beta-oleyl-estradiol by liver microsomes. Glucuronidation of E(2) by microsomal glucuronosyltransferase was increased moderately. In contrast, the activity of NAD(P)H quinone reductase 1 (NQO1), a representative monofunctional phase 2 enzyme, was significantly decreased in liver cytosol of rats fed clofibrate. Decreases in hepatic NQO1 in livers of animals fed clofibrate were noted before the appearance of mammary tumors. E(2) was delivered in cholesterol pellets implanted in 7-8-week-old female ACI rats. The animals received AIN-76A diet containing 0.4% clofibrate for 6, 12 or 28 weeks. Control animals received AIN-76A diet. Dietary clofibrate increased the number and size of palpable mammary tumors but did not alter the histopathology of the E(2)-induced mammary adenocarcinomas. Collectively, these results suggest that the stimulatory effect of clofibrate on hepatic esterification of E(2) with fatty acids coupled with the inhibition of protective phase 2 enzymes, may in part, enhance E(2)-dependent mammary carcinogenesis in the ACI rat model.


Subject(s)
Clofibrate/toxicity , Estradiol/agonists , Hypolipidemic Agents/toxicity , Mammary Neoplasms, Animal/chemically induced , Microsomes, Liver/drug effects , Animals , Clofibrate/administration & dosage , Diet , Estradiol/blood , Estradiol/metabolism , Female , Hypolipidemic Agents/administration & dosage , Mammary Neoplasms, Animal/pathology , Microsomes, Liver/enzymology , NAD(P)H Dehydrogenase (Quinone)/metabolism , Rats , Rats, Inbred ACI
SELECTION OF CITATIONS
SEARCH DETAIL
...