Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 109
Filter
1.
J Stud Alcohol Drugs ; 2024 Jun 02.
Article in English | MEDLINE | ID: mdl-38830016

ABSTRACT

OBJECTIVE: Measure prevalence and overlap of secondhand harms from other people's use of alcohol, cannabis, opioid, or other drugs and examine sociodemographic and other correlates of these secondhand harms. METHODS: This cross-sectional analysis used data from 7,799 respondents (51.6% female; 12.9% Black, 15.6% Hispanic/Latiné; mean age: 47.6) in the 2020 U.S. National Alcohol Survey. Secondhand harms included family/marriage difficulties, traffic accidents, vandalism, physical harm, and financial difficulties. Weighted prevalence estimates provided nationally representative estimates of these harms. Logistic regression assessed associations between individual characteristics and secondhand harms. RESULTS: Lifetime prevalence of secondhand harms from alcohol, cannabis, opioid, or other drugs was 34.2%, 5.5%, 7.6%, and 8.3%, respectively. There was substantial overlap among lifetime harms: almost 30% of those reporting secondhand alcohol harms also reported secondhand drug harms. Significant correlates of secondhand substance harms included female sex (alcohol, other drug); white (alcohol, opioid), American Indian/Alaska Native (opioid), and Black (cannabis) race/ethnicity; and separated/divorced/widowed marital status (opioid). Those reporting family history of alcohol problems had significantly higher odds of reporting secondhand harms across substance types. Individuals who reported frequent cannabis use had higher odds of reporting secondhand alcohol and opioid harms compared to those with no cannabis use, (aOR=1.55; aOR=2.38), but lower odds of reporting secondhand cannabis harms (aOR=0.51). CONCLUSIONS: Although less prevalent than secondhand alcohol harms, 14% of participants reported secondhand harms from someone else's drug use and frequently experienced secondhand harms attributed to multiple substances. Population-focused interventions are needed to reduce the total burden of alcohol and drug use.

2.
Hum Reprod ; 34(12): 2399-2409, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31887223

ABSTRACT

STUDY QUESTION: Are urinary levels of oxidative stress biomarkers associated with reproductive outcome success following fertility treatments? SUMMARY ANSWER: Levels of oxidative stress in the middle tertile for women are associated with the highest levels of reproductive success while no associations were noted for men. WHAT IS KNOWN ALREADY: Oxidative stress may contribute to adverse fertility outcomes in the general population, but findings from couples undergoing fertility treatments are sparse. STUDY DESIGN, SIZE, DURATION: This prospective cohort study included 481 women and 249 of their male partners undergoing fertility treatments from 2007 to 2015, from the Environment and Reproductive Health (EARTH) study in Boston, MA. PARTICIPANTS/MATERIALS, SETTING, METHODS: One urine sample per participant was collected at each cycle and analysed for two oxidative stress markers: 8-isoprostane-PGF2α (8-iso-PGF2α) and 8-isoprostane-PGF2α metabolite (F2-isoP-M). Reproductive outcomes were abstracted from medical records and included the fertilization rate, for IVF (oocytes fertilized/mature oocytes retrieved), and rates of implantation, clinical pregnancy and live birth, for both IVF and IUI. Cluster-weighted generalized estimating equations were used to analyse adjusted associations between exposure tertiles and outcomes. MAIN RESULTS AND THE ROLE OF CHANCE: Levels of F2-isoP-M in the middle tertile were associated with the most success among women. Women in the upper tertile of F2-isoP-M had an adjusted mean live birth rate after IVF and IUI of 23% (95% CI: 17, 29) compared to 38% (95% CI: 31, 45) for women in the middle tertile and 27% (95% CI: 21, 34) in the lower tertile. The fertilization rate during IVF was higher for women with 8-iso-PGF2α in the middle tertile (0.77 [95% CI: 0.73, 0.80]) compared to women in the lower (0.69 [95% CI: 0.64, 0.73]) or upper tertiles (0.66 [95% CI: 0.61, 0.71]). No significant associations were found for other measured outcomes with 8-iso-PGF2α, or between any oxidative stress biomarker in men and reproductive outcomes in their partners. LIMITATIONS, REASONS FOR CAUTION: Isoprostanes are short-lived biomarkers and this study may not have captured the most relevant window of susceptibility for oxidative stress on the outcomes of interest. Findings from this study may not be generalizable to couples attempting conception without fertility assistance. WIDER IMPLICATIONS OF THE FINDINGS: This study suggests that a non-linear association may exist between oxidative stress and reproductive outcomes in a population undergoing fertility treatment, a finding not previously identified in the literature. Oxidative stress may represent the mechanism through which environmental chemicals are associated with adverse reproductive outcomes. STUDY FUNDING/COMPETING INTEREST(S): This research was supported by the Intramural Research Program of the National Institutes of Environmental Health Sciences (NIEHS) (ZIA ES103314) and by NIEHS grants R01ES022955, R01ES009718 and R01ES00002. There are no competing interests to report. TRIAL REGISTRATION NUMBER: N/A.


Subject(s)
Biomarkers/urine , Birth Rate , Oxidative Stress , Reproductive Techniques, Assisted/statistics & numerical data , Adult , Female , Humans , Male , Pregnancy , Prospective Studies
3.
Oncogene ; 27(6): 794-802, 2008 Jan 31.
Article in English | MEDLINE | ID: mdl-17653086

ABSTRACT

BRCA1 can regulate estrogen receptor-alpha (ERalpha) activity. This study tested the hypotheses that Brca1 loss in mammary epithelium alters the estrogenic growth response and that exposure to increased estrogen or ERalpha collaborates with Brca1 deficiency to accelerate preneoplasia and cancer development. Longer ductal extension was found in mammary glands of Brca1(f/f;MMTV-Cre) mice during puberty as compared to wild-type mice. Terminal end bud differentiation was impaired in Brca1 mutant mice with preservation of prolactin-induced alveolar differentiation. Exogenous estrogen stimulated an abnormal sustained increase in mammary epithelial cell proliferation and the appearance of ERalpha-negative preneoplasia in postpubertal Brca1 mutant mice. Carcinogenesis was investigated using Brca1(f/f;MMTV-Cre) mice hemizygous for p53. Exogenous estrogen increased the percentage of mice with multiple hyperplastic alveolar nodules. Targeted conditional ERalpha overexpression in mammary epithelial cells of mice that were Brca1 mutant and hemizygous for p53 increased the percentage of mice exhibiting multiple hyperplastic nodules, invasive mammary cancers and cancer multiplicity. Significantly more than half of the preneoplasia and cancers were ERalpha negative even as their initiation was promoted by ERalpha overexpression.


Subject(s)
BRCA1 Protein/genetics , Estrogen Receptor alpha/metabolism , Estrogens/metabolism , Mammary Neoplasms, Experimental/pathology , Precancerous Conditions/pathology , Animals , Cell Proliferation , Estrogen Receptor alpha/analysis , Estrogens/pharmacology , Female , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Mutant Strains , Neoplasm Invasiveness , Precancerous Conditions/genetics , Precancerous Conditions/metabolism
4.
Oncogene ; 26(33): 4774-96, 2007 Jul 19.
Article in English | MEDLINE | ID: mdl-17297451

ABSTRACT

Scatter factor (SF) (hepatocyte growth factor) is a pleiotrophic cytokine that accumulates within tumors in vivo and protects tumor cells against cytotoxicity and apoptosis due to DNA damaging agents in vitro. Previous studies have established that SF-mediated cell protection involves antiapoptotic signaling from its receptor (c-Met) to PI3 kinase --> c-Akt --> Pak1 (p21-activated kinase -1) --> NF-kappaB (nuclear factor-kappa B). Here, we found that Ras proteins (H-Ras and R-Ras) enhance SF-mediated activation of NF-kappaB and protection of DU-145 and MDCK (Madin-Darby canine kidney) cells against the topoisomerase IIalpha inhibitor adriamycin. Studies of Ras effector loop mutants and their downstream effectors suggest that Ras/PI3 kinase and Ras/Raf1 pathways contribute to SF stimulation of NF-kappaB signaling and cell protection. Further studies revealed that Raf1 positively regulates the ability of SF to stimulate NF-kappaB activity and cell protection. The ability of Raf1 to stimulate NF-kappaB activity was not due to the classical Raf1 --> MEK1/2 --> ERK1/2 pathway. However, we found that a MEK3/6 --> p38 pathway contributes to SF-mediated activation of NF-kappaB. In contrast, RalA, a target of the Ras/RalGDS pathway negatively regulated the ability of SF to stimulate NF-kappaB activity and cell protection. Ras, Raf1 and RalA modulate SF stimulation of NF-kappaB activity, in part, by regulating IkappaB kinase (IKK)-beta kinase activity. These findings suggest that Ras/Raf1/RalA pathways may converge to modulate NF-kappaB activation and SF-mediated survival signaling at the IKK complex and/or a kinase upstream of this complex.


Subject(s)
DNA Damage , Hepatocyte Growth Factor/metabolism , NF-kappa B/metabolism , Signal Transduction/physiology , ras Proteins/metabolism , Animals , Antibiotics, Antineoplastic/pharmacology , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Doxorubicin/pharmacology , Genetic Vectors/genetics , Hepatocyte Growth Factor/genetics , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Mutation , NF-kappa B/genetics , Phosphorylation , Protein Binding , Proto-Oncogene Proteins c-raf/genetics , Proto-Oncogene Proteins c-raf/metabolism , RNA Interference , Signal Transduction/genetics , Transfection , ral GTP-Binding Proteins/genetics , ral GTP-Binding Proteins/metabolism , ras Proteins/genetics
5.
Oncogene ; 26(20): 2925-38, 2007 May 03.
Article in English | MEDLINE | ID: mdl-17099727

ABSTRACT

The cytokine scatter factor (SF) (hepatocyte growth factor) transduces various biologic actions, including cell motility, invasion, angiogenesis and apoptosis inhibition. The latter is relevant to understanding the role of SF in promoting tumor cell survival in different contexts, for example, detachment from basement membrane, growth in metastatic sites and responses to chemo- and radiotherapy. Previously, we showed that SF protects cells against apoptosis owing to DNA damage, by a mechanism involving phosphoinositol-3-kinase/c-Akt signaling. Here, we used DNA microarray assays to identify c-Akt-regulated genes that might contribute to cell protection. DU-145 human prostate cancer cells were transfected+/-a dominant-negative mutant Akt, treated+/-SF and analysed for gene expression using Affymetrix arrays. These studies identified SF-regulated genes for which induction was c-Akt-dependent vs -independent. Selected microarray findings were confirmed by semiquantitative and quantitative reverse transcription-polymerase chain reaction. We tested the contribution of four SF-inducible/c-Akt-dependent genes (AMPD3, EPHB2, MX1 and WNT4) to protection against adriamycin (a DNA topoisomerase IIalpha inhibitor) using RNA interference. Knockdown of each gene except EPHB2 caused a small but significant reduction in the SF cell protection. The lack of effect of EPHB2 knockdown may be due to the fact that DU-145 cells contain a single-mutant EPHB2 allele. A combination of three small interfering RNAs blocked most of the protection by SF in both DU-145 and T47D cells. These findings identify novel c-Akt-regulated genes, some of which contribute to SF-mediated cytoprotection.


Subject(s)
Gene Expression Regulation, Neoplastic/drug effects , Hepatocyte Growth Factor/physiology , Oncogene Protein v-akt/antagonists & inhibitors , Prostatic Neoplasms/genetics , RNA, Small Interfering/pharmacology , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/genetics , Gene Expression Profiling , Humans , Male , NF-kappa B/genetics , NF-kappa B/physiology , Oligonucleotide Array Sequence Analysis , Oncogene Protein v-akt/genetics , Oncogene Protein v-akt/physiology , Prostatic Neoplasms/pathology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/physiology , Tumor Cells, Cultured , p21-Activated Kinases
6.
Br J Cancer ; 94(3): 407-26, 2006 Feb 13.
Article in English | MEDLINE | ID: mdl-16434996

ABSTRACT

Indole-3-carbinol (I3C) and genistein are naturally occurring chemicals derived from cruciferous vegetables and soy, respectively, with potential cancer prevention activity for hormone-responsive tumours (e.g., breast and prostate cancers). Previously, we showed that I3C induces BRCA1 expression and that both I3C and BRCA1 inhibit oestrogen (E2)-stimulated oestrogen receptor (ER-alpha) activity in human breast cancer cells. We now report that both I3C and genistein induce the expression of both breast cancer susceptibility genes (BRCA1 and BRCA2) in breast (MCF-7 and T47D) and prostate (DU-145 and LNCaP) cancer cell types, in a time- and dose-dependent fashion. Induction of the BRCA genes occurred at low doses of I3C (20 microM) and genistein (0.5-1.0 microM), suggesting potential relevance to cancer prevention. A combination of I3C and genistein gave greater than expected induction of BRCA expression. Studies using small interfering RNAs (siRNAs) and BRCA expression vectors suggest that the phytochemical induction of BRCA2 is due, in part, to BRCA1. Functional studies suggest that I3C-mediated cytoxicity is, in part, dependent upon BRCA1 and BRCA2. Inhibition of E2-stimulated ER-alpha activity by I3C and genistein was dependent upon BRCA1; and inhibition of ligand-inducible androgen receptor (AR) activity by I3C and genistein was partially reversed by BRCA1-siRNA. Finally, we provide evidence suggesting that the phytochemical induction of BRCA1 expression is due, in part, to endoplasmic reticulum stress response signalling. These findings suggest that the BRCA genes are molecular targets for some of the activities of I3C and genistein.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , BRCA1 Protein/agonists , BRCA2 Protein/agonists , Breast Neoplasms/drug therapy , Genistein/therapeutic use , Indoles/therapeutic use , Prostatic Neoplasms/drug therapy , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Combined Chemotherapy Protocols , BRCA1 Protein/genetics , BRCA2 Protein/genetics , Breast Neoplasms/metabolism , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Estrogen Receptor alpha/antagonists & inhibitors , Female , Gene Expression/drug effects , Genistein/pharmacology , Humans , Indoles/pharmacology , Male , Prostatic Neoplasms/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Tumor Cells, Cultured , Up-Regulation
7.
Endocr Relat Cancer ; 12(3): 533-48, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16172191

ABSTRACT

The breast and ovarian cancer susceptibility gene-1 (BRCA1) located on chromosome 17q21 encodes a tumor suppressor gene that functions, in part, as a caretaker gene in preserving chromosomal stability. The observation that most BRCA1 mutant breast cancers are hormone receptor negative has led some to question whether hormonal factors contribute to the etiology of BRCA1-mutant breast cancers. Nevertheless, the caretaker function of BRCA1 is a generic one and does not explain why BRCA1 mutations confer a specific risk for tumor types that are hormone-responsive or that hormonal factors contribute to the etiology, including those of the breast, uterus, cervix, and prostate. An accumulating body of research indicates that in addition to its well-established roles in regulation of the DNA damage response, the BRCA1 protein interacts with steroid hormone receptors (estrogen receptor (ER-alpha) and androgen receptor (AR)) and regulates their activity, inhibiting ER-alpha activity and stimulating AR activity. The ability of BRCA1 to regulate steroid hormone action is consistent with clinical-epidemiological research suggesting that: (i) hormonal factors contribute to breast cancer risk in BRCA1 mutation carriers; and (ii) the spectrum of risk-modifying effects of hormonal factors in BRCA1 carriers is not identical to that observed in the general population. These data suggest a model for BRCA1 carcinogenesis in which genomic instability leads to the initiation of cancerous cell clones, while loss of normal restraint on hormonal stimulation of mammary epithelial cell proliferation allows amplification of these pre-existing clones. Further research will be required to substantiate this hypothesis.


Subject(s)
Breast Neoplasms/genetics , Genes, BRCA1 , Ovarian Neoplasms/genetics , Carrier State , Chromosome Mapping , Chromosomes, Human, Pair 17 , Female , Humans , Mutation
8.
Cancer Res ; 61(21): 8022-31, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11691828

ABSTRACT

The cytokine hepatocyte growth factor/scatter factor (HGF/SF) protects epithelial and cancer cells against DNA-damaging agents via a pathway involving signaling from c-Met --> phosphatidylinositol-3- kinase --> c-Akt. However, the downstream alterations in gene expression resulting from this pathway have not been established. On the basis of cDNA microarray and semiquantitative RT-PCR assays, we found that MDA-MB-453 human breast cancer cells preincubated with HGF/SF and then exposed to Adriamycin (ADR), a DNA topoisomerase II inhibitor, exhibit an altered pattern of gene expression, as compared with cells treated with ADR only. [HGF/SF+ADR]-treated cells showed altered expression of genes involved in the DNA damage response, cell cycle regulation, signal transduction, metabolism, and development. Some of these alterations suggest mechanisms by which HGF/SF may exert its protective activity, e.g., up-regulation of polycystic kidney disease-1 (a survival-promoting component of cadherin-catenin complexes), down-regulation of 51C (an inositol polyphosphate-5-phosphatase), and down-regulation of TOPBP1 (a topoisomerase IIB binding protein). We showed that enforced expression of the cdc42-interacting protein CIP4, a cytoskeleton-associated protein for which expression was decreased in [HGF/SF+ADR]-treated cells, inhibited HGF/SF-mediated protection against ADR. The cDNA microarray approach may open up new avenues for investigation of the DNA damage response and its regulation by HGF/SF.


Subject(s)
Breast Neoplasms/genetics , DNA Damage/physiology , Gene Expression Regulation, Neoplastic/drug effects , Hepatocyte Growth Factor/pharmacology , Antibiotics, Antineoplastic/pharmacology , Breast Neoplasms/metabolism , Doxorubicin/pharmacology , Gene Expression Profiling , Humans , Microtubule-Associated Proteins/physiology , Minor Histocompatibility Antigens , Oligonucleotide Array Sequence Analysis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured/drug effects
9.
J Biol Chem ; 276(50): 47257-65, 2001 Dec 14.
Article in English | MEDLINE | ID: mdl-11571297

ABSTRACT

The cytokine hepatocyte growth factor/scatter factor (HGF/SF) has been found to protect a variety of epithelial and cancer cell types against cytotoxicity and apoptosis induced by DNA damage, but the specific apoptotic signaling events and the levels at which they are blocked by HGF/SF have not been identified. We found that treatment of MDA-MB-453 human breast cancer cells with adriamycin (also known as doxorubicin, a DNA topoisomerase IIalpha inhibitor) induced a series of time-dependent events, including the mitochondrial release of cytochrome c and apoptosis-inducing factor, mitochondrial membrane depolarization, activation of a set of caspases (caspase-9, -3, -7, -2, and -8), cleavage of poly(ADP-ribose) polymerase (PARP), and up-regulation of expression of the Fas ligand. All of these events were blocked by preincubation of the cells with HGF/SF. In contrast, the pan-caspase inhibitor benzyloxycarbonyl-VAD-fluoromethylketone blocked some of these events (e.g. caspase-3 activation and PARP cleavage) but did not block cytochrome c release or mitochondrial depolarization. These findings suggest that HGF/SF functions, in part, upstream of the mitochondria to block mitochondrial apoptosis signaling, prevent activation of multiple caspases, and protect breast cancer cells against apoptosis.


Subject(s)
Apoptosis , Breast Neoplasms/metabolism , Hepatocyte Growth Factor/metabolism , Mitochondria/metabolism , Amino Acid Chloromethyl Ketones/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis Inducing Factor , Caspase 3 , Caspases/metabolism , Coloring Agents/pharmacology , Cysteine Proteinase Inhibitors/pharmacology , Cytochrome c Group/metabolism , Doxorubicin/pharmacology , Enzyme Activation , Enzyme Inhibitors/pharmacology , Fas Ligand Protein , Flavoproteins/metabolism , Humans , Membrane Glycoproteins/metabolism , Membrane Potentials , Membrane Proteins/metabolism , Recombinant Proteins/metabolism , Signal Transduction , Tetrazolium Salts/pharmacology , Thiazoles/pharmacology , Time Factors , Tumor Cells, Cultured , Up-Regulation
10.
Oncogene ; 20(35): 4827-41, 2001 Aug 09.
Article in English | MEDLINE | ID: mdl-11521194

ABSTRACT

The tumor suppressor activity of the BRCA1 gene product is due, in part, to functional interactions with other tumor suppressors, including p53 and the retinoblastoma (RB) protein. RB binding sites on BRCA1 were identified in the C-terminal BRCT domain (Yarden and Brody, 1999) and in the N-terminus (aa 304-394) (Aprelikova et al., 1999). The N-terminal site contains a consensus RB binding motif, LXCXE (aa 358-362), but the role of this motif in RB binding and BRCA1 functional activity is unclear. In both in vitro and in vivo assays, we found that the BRCA1:RB interaction does not require the BRCA1 LXCXE motif, nor does it require an intact A/B binding pocket of RB. In addition, nuclear co-localization of the endogenous BRCA1 and RB proteins was observed. Over-expression of wild-type BRCA1 (wtBRCA1) did not cause cell cycle arrest but did cause down-regulation of expression of RB, p107, p130, and other proteins (e.g., p300), associated with increased sensitivity to DNA-damaging agents. In contrast, expression of a full-length BRCA1 with an LXCXE inactivating mutation (LXCXE-->RXRXH) failed to down-regulate RB, blocked the down-regulation of RB by wtBRCA1, induced chemoresistance, and abrogated the ability of BRCA1 to mediate tumor growth suppression of DU-145 prostate cancer cells. wtBRCA1-induced chemosensitivity was partially reversed by expression of either Rb or p300 and fully reversed by co-expression of Rb plus p300. Our findings suggest that: (1) disruption of the LXCXE motif within the N-terminal RB binding region alters the biologic function of BRCA1; and (2) over-expression of BRCA1 inhibits the expression of RB and RB family (p107 and p130) proteins.


Subject(s)
BRCA1 Protein/physiology , Retinoblastoma Protein/metabolism , Amino Acid Motifs , Amino Acid Sequence , BRCA1 Protein/chemistry , Binding Sites , Down-Regulation , Genes, Retinoblastoma , Humans , Molecular Sequence Data , Nuclear Proteins/genetics , Trans-Activators/genetics , Tumor Cells, Cultured
11.
Cell Mol Biol (Noisy-le-grand) ; 47(3): 485-93, 2001 May.
Article in English | MEDLINE | ID: mdl-11441956

ABSTRACT

Duck embryo was studied as a model for assessing the effects of microbeam radiation therapy (MRT) on the human infant brain. Because of the high risk of radiation-induced disruption of the developmental process in the immature brain, conventional wide-beam radiotherapy of brain tumors is seldom carried out in infants under the age of three. Other types of treatment for pediatric brain tumors are frequently ineffective. Recent findings from studies in Grenoble on the brain of suckling rats indicate that MRT could be of benefit for the treatment of early childhood tumors. In our studies, duck embryos were irradiated at 3-4 days prior to hatching. Irradiation was carried out using a single exposure of synchrotron-generated X-rays, either in the form of parallel microplanar beams (microbeams), or as non-segmented broad beam. The individual microplanar beams had a width of 27 microm and height of 11 mm, and a center-to-center spacing of 100 microm. Doses to the exposed areas of embryo brain were 40, 80, 160 and 450 Gy (in-slice dose) for the microbeam, and 6, 12 and 18 Gy for the broad beam. The biological end point employed in the study was ataxia. This neurological symptom of radiation damage to the brain developed within 75 days of hatching. Histopathological analysis of brain tissue did not reveal any radiation induced lesions for microbeam doses of 40-160 Gy (in-slice), although some incidences of ataxia were observed in that dose group. However, severe brain lesions did occur in animals in the 450 Gy microbeam dose groups, and mild lesions in the 18 Gy broad beam dose group. These results indicate that embryonic duck brain has an appreciably higher tolerance to the microbeam modality, as compared to the broad beam modality. When the microbeam dose was normalized to the full volume of the irradiated tissue. i.e., the dose averaged over microbeams and the space between the microbeams, brain tolerance was estimated to be about three times higher to microbeam irradiation as compared with broad beam irradiation.


Subject(s)
Brain/embryology , Brain/radiation effects , Ducks , Radiation Injuries/embryology , X-Ray Therapy/adverse effects , Animals , Ataxia/physiopathology , Body Weight/radiation effects , Brain/pathology , Dose-Response Relationship, Radiation , Ducks/embryology , Models, Animal , Monte Carlo Method , Radiation Dosage , Radiation Injuries/pathology , Radiation Injuries/physiopathology , Survival Rate , Synchrotrons , Time Factors
12.
Mol Cell Biol ; 21(15): 4968-84, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11438654

ABSTRACT

Hepatocyte growth factor (scatter factor) (HGF/SF) is a pleiotrophic mediator of epithelial cell motility, morphogenesis, angiogenesis, and tumorigenesis. HGF/SF protects cells against DNA damage by a pathway from its receptor c-Met to phosphatidylinositol 3-kinase (PI3K) to c-Akt, resulting in enhanced DNA repair and decreased apoptosis. We now show that protection against the DNA-damaging agent adriamycin (ADR; topoisomerase IIalpha inhibitor) requires the Grb2-binding site of c-Met, and overexpression of the Grb2-associated binder Gab1 (a multisubstrate adapter required for epithelial morphogenesis) inhibits the ability of HGF/SF to protect MDCK epithelial cells against ADR. In contrast to Gab1 and its homolog Gab2, overexpression of c-Cb1, another multisubstrate adapter that associates with c-Met, did not affect protection. Gab1 blocked the ability of HGF/SF to cause the sustained activation of c-Akt and c-Akt signaling (FKHR phosphorylation). The Gab1 inhibition of sustained c-Akt activation and of cell protection did not require the Gab1 pleckstrin homology or SHP2 phosphatase-binding domain but did require the PI3K-binding domain. HGF/SF protection of parental MDCK cells was blocked by wortmannin, expression of PTEN, and dominant negative mutants of p85 (regulatory subunit of PI3K), Akt, and Pak1; the protection of cells overexpressing Gab1 was restored by wild-type or activated mutants of p85, Akt, and Pak1. These findings suggest that the adapter Gab1 may redirect c-Met signaling through PI3K away from a c-Akt/Pak1 cell survival pathway.


Subject(s)
Adaptor Proteins, Signal Transducing , Hepatocyte Growth Factor/metabolism , Phosphoproteins/metabolism , Phosphoproteins/physiology , Proto-Oncogene Proteins c-met/metabolism , Androstadienes/pharmacology , Animals , Apoptosis , Binding Sites , Blotting, Western , Caspase 3 , Caspases/metabolism , Caspases/physiology , Cell Division , Cell Line , Cell Movement , Cell Survival , DNA/metabolism , DNA Damage , DNA Fragmentation , DNA Repair , Dogs , Doxorubicin/pharmacology , Enzyme Inhibitors/pharmacology , Epithelial Cells/metabolism , GRB2 Adaptor Protein , Genetic Vectors , Humans , Models, Biological , Mutation , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Precipitin Tests , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Recombinant Proteins/metabolism , Signal Transduction , Tetrazolium Salts/pharmacology , Thiazoles/pharmacology , Time Factors , Transfection , Wortmannin , p21-Activated Kinases
13.
Cancer Invest ; 19(4): 396-412, 2001.
Article in English | MEDLINE | ID: mdl-11405179

ABSTRACT

The breast cancer susceptibility gene BRCA1 on chromosome 17q21 encodes an 1863 amino acid protein that is important for normal embryonic development. Germline mutations of this gene are linked to a significantly increased lifetime risk for breast and/or ovarian cancer, and recent studies suggest that the same may be true for prostate cancer. Several activities that may contribute to the tumor suppressor function of BRCA1 have been identified via in vitro and experimental animal studies. These include (i) regulation of cell proliferation; (ii) participation in DNA repair/recombination processes related to the maintenance of genomic integrity; (iii) induction of apoptosis in damaged cells; and (iv) regulation of transcription. A second breast cancer susceptibility gene (BRCA2) operates in some of the same molecular pathways as BRCA1, and mutations of this gene predispose to breast and ovarian cancer and probably to other tumor types, including prostate cancer. Finally, recent studies from our laboratory suggest that BRCA1 modulates proliferation, chemosensitivity, repair of DNA strand breaks, apoptosis induction, and expression of certain key cellular regulatory proteins (including BRCA2 and p300) in human prostate cancer cells. These activities are consistent with a putative prostate tumor suppressor function of BRCA1.


Subject(s)
Adenocarcinoma/genetics , Genes, BRCA1 , Prostatic Neoplasms/genetics , Adenocarcinoma/epidemiology , Adenocarcinoma/pathology , Amino Acid Motifs , Animals , Apoptosis/genetics , BRCA1 Protein/chemistry , BRCA1 Protein/physiology , BRCA2 Protein , Breast Neoplasms/ethnology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Cycle , Cell Division , Chromosomes, Human, Pair 13/genetics , Chromosomes, Human, Pair 17/genetics , DNA Damage , Embryonic and Fetal Development/genetics , Estrogens , Female , Gene Expression Regulation , Genes, Tumor Suppressor , Genetic Predisposition to Disease , Humans , Jews/genetics , Male , Mice , Mice, Knockout , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/pathology , Neoplastic Syndromes, Hereditary/genetics , Ovarian Neoplasms/genetics , Prostatic Neoplasms/epidemiology , Prostatic Neoplasms/pathology , Protein Structure, Tertiary , Risk Factors , Transcription Factors/genetics , Transcription Factors/physiology , Transcriptional Activation , Tumor Cells, Cultured
14.
Oncogene ; 20(1): 77-87, 2001 Jan 04.
Article in English | MEDLINE | ID: mdl-11244506

ABSTRACT

The BRCA1 gene was previously found to inhibit the transcriptional activity of the estrogen receptor [ER-alpha] in human breast and prostate cancer cell lines. In this study, we found that breast cancer-associated mutations of BRCA1 abolish or reduce its ability to inhibit ER-alpha activity and that domains within the amino- and carboxyl-termini of the BRCA1 protein are required for the inhibition. BRCA1 inhibition of ER-alpha activity was demonstrated under conditions in which a BRCA1 transgene was transiently or stably over-expressed in cell lines with endogenous wild-type BRCA1 and in a breast cancer cell line that lacks endogenous functional BRCA1 (HCC1937). In addition, BRCA1 blocked the expression of two endogenous estrogen-regulated gene products in human breast cancer cells: pS2 and cathepsin D. The BRCA1 protein was found to associate with ER-alpha in vivo and to bind to ER-alpha in vitro, by an estrogen-independent interaction that mapped to the amino-terminal region of BRCA1 (ca. amino acid 1-300) and the conserved carboxyl-terminal activation function [AF-2] domain of ER-alpha. Furthermore, several truncated BRCA1 proteins containing the amino-terminal ER-alpha binding region blocked the ability of the full-length BRCA1 protein to inhibit ER-alpha activity. Our findings suggest that the amino-terminus of BRCA1 interacts with ER-alpha, while the carboxyl-terminus of BRCA1 may function as a transcriptional repression domain. Oncogene (2001) 20, 77 - 87.


Subject(s)
BRCA1 Protein/genetics , BRCA1 Protein/metabolism , Genes, BRCA1/physiology , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/metabolism , BRCA1 Protein/deficiency , BRCA1 Protein/physiology , Binding Sites/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Chromosome Mapping , Estrogen Receptor alpha , Genes, BRCA1/genetics , Glutathione Transferase/genetics , Humans , Mutation , Peptide Fragments/genetics , Peptide Fragments/metabolism , Receptors, Estrogen/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Deletion , Transcription, Genetic , Tumor Cells, Cultured
15.
Cancer Chemother Pharmacol ; 47(2): 161-9, 2001.
Article in English | MEDLINE | ID: mdl-11269743

ABSTRACT

UNLABELLED: P73, a new p53 homologue, has been recently identified as a candidate tumor suppressor gene. PURPOSE: We studied the alterations in p73 in a panel of human cancer cell lines treated with the chemotherapeutic agent, Adriamycin (ADR), in comparison with the changes in p53. METHODS: P73 and p53 mRNA and protein were determined using semiquantitative reverse transcription-polymerase chain reaction (RT-PCR) and Western blotting, respectively. ADR cytotoxicity was examined by a trypan blue dye exclusion assay. RESULTS: The cell lines bearing wild-type p53 were more susceptible to ADR than the cell lines bearing mutant p53. ADR treatment resulted in a significant accumulation of p53 protein and mRNA expression in the wild-type p53 cell lines and caused little (slight increase) or no influence on p53 expression in the cell lines with p53 mutation and deletion. However, in striking contrast to the alterations in p53, a decline in p73 at both the protein and mRNA levels was observed in all the cell lines examined following ADR treatment. Further studies indicated that this p53-independent downregulation of p73 was induced by ADR in a dose- and time-dependent manner. Moreover, the p73 protein decline was abrogated by the presence of proteasome inhibitors. CONCLUSIONS: Our findings revealed that although p73 shares a similar structural and functional composition with p53, there is a significant difference in the mechanisms that govern the responses of p53 and p73 to ADR-induced DNA damage.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , DNA-Binding Proteins/genetics , Doxorubicin/pharmacology , Genes, p53/physiology , Nuclear Proteins/genetics , Cysteine Endopeptidases , Dose-Response Relationship, Drug , Down-Regulation , Genes, Tumor Suppressor , Humans , Multienzyme Complexes/antagonists & inhibitors , Proteasome Endopeptidase Complex , RNA, Messenger/analysis , Time Factors , Tumor Cells, Cultured , Tumor Protein p73 , Tumor Suppressor Proteins
16.
FEBS Lett ; 488(3): 179-84, 2001 Jan 19.
Article in English | MEDLINE | ID: mdl-11163768

ABSTRACT

Germ-line mutations of the breast cancer susceptibility gene 1 (BRCA1) confer a high risk for breast and ovarian cancer in women and prostate cancer in men. The BRCA1 protein contributes to cell proliferation, cell cycle regulation, DNA repair and apoptosis; however, the mechanisms underlying these functions of BRCA1 remain largely unknown. Here, we showed that, in Du-145 human prostate cancer cells, enhanced expression of BRCA1 resulted in constitutive activation of signal transducer and activator transcription factor 3 (STAT3) tyrosine and serine phosphorylation. Moreover, Janus kinase 1 (JAK1) and JAK2, the upstream activators of STAT3, were also activated by BRCA1. Immunoprecipitation assay showed that BRCA1 interacted with JAK1 and JAK2. Blocking STAT3 activation using antisense oligonucleotides significantly inhibited cell proliferation and triggered apoptosis in Du-145 cells with enhanced expression of BRCA1. These findings indicate that BRCA1 interacts with the components of the JAK-STAT signaling cascade and modulates its activation, which may provide a new critical survival signal for the growth of breast, ovarian and prostate cancers in the presence of normal BRCA1.


Subject(s)
BRCA1 Protein/metabolism , DNA-Binding Proteins/metabolism , Prostatic Neoplasms/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins , Trans-Activators/metabolism , Apoptosis , BRCA1 Protein/genetics , Blotting, Western , Cell Division , Cell Survival , DNA-Binding Proteins/genetics , Enzyme Activation , Genes, BRCA1/genetics , Humans , Interleukin-6/pharmacology , Janus Kinase 1 , Janus Kinase 2 , Male , Oligonucleotides, Antisense/genetics , Phosphorylation , Precipitin Tests , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/genetics , Protein Binding , STAT3 Transcription Factor , Signal Transduction/drug effects , Trans-Activators/genetics , Transfection , Tumor Cells, Cultured
17.
Shi Yan Sheng Wu Xue Bao ; 34(1): 55-64, 2001 Mar.
Article in English | MEDLINE | ID: mdl-12549011

ABSTRACT

Mutations of the human breast cancer susceptibility gene 1 (BRCA1) confers a risk for breast, ovarian and prostate cancers and BRCA1 exerts multiple biological functions. Using Western blot and semi-quantitative reverse transcription-polymerase chain reaction (RT-PCR) assays, we have determined the expression of endogenous BRCA1 protein and mRNA in forty-three human tumor cell lines established from eleven types of human tumor tissues. BRCA1 was differentially expressed in tumor cell lines. No significant association was found between BRCA1 expression and the p53 gene status of cell lines. The disruption of wild-type p53 by either the human papillomavirus E6 oncogene or the mutant p53 gene (143Ala-->Val) did not cause any significant alteration in basal level of BRCA1 expression, while the knockout of p21 (-/-) by homologous recombination assay and Blocking Gadd45 expression by constitutive antisense expression slightly increased BRCA1 protein expression. Therefore, although the functional significance of the differential expression in human tumor cells is currently unknown, the present data provide a valuable background for further study of BRCA1 in tumor cell lines.


Subject(s)
BRCA1 Protein/genetics , Genes, p53/genetics , BRCA1 Protein/biosynthesis , Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Humans , Mutation , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
18.
Oncogene ; 20(57): 8215-35, 2001 Dec 13.
Article in English | MEDLINE | ID: mdl-11781837

ABSTRACT

Unregulated expression of wild-type BRCA1 (wtBRCA1) confers an altered phenotype in cultured human prostate cancer cells, characterized by chemosensitivity, susceptibility to apoptosis, decreased DNA repair activity, and alterations of key cell regulatory proteins. We now report that the expression of truncated or mutant full-length BRCA1 genes can abrogate certain phenotypic characteristics and/or confer the opposite phenotype to the wild-type BRCA1 gene. In particular, several carboxyl-terminal truncated BRCA1 proteins conferred chemoresistance, decreased susceptibility to apoptosis, and decreased ability to suppress in vivo tumor growth. These truncated BRCA1 proteins also blocked the ability of ectopically expressed wtBRCA1 to induce chemosensitivity and to inhibit estrogen receptor transcriptional activity. Studies using epitope-tagged truncated proteins confirmed their expression, nuclear localization, and functionality. On the other hand, in cells with no endogenous wild-type BRCA1 (HCC1937 human breast cancer cells), the wtBRCA1 gene enhanced cellular DNA repair activity and rendered the cells resistant to DNA damage; while truncated BRCA1 proteins blocked the wtBRCA1-induced chemoresistance. Our findings suggest that truncated BRCA1 proteins can inhibit the function of wild-type BRCA1. They raise the possibility that some inherited BRCA1 mutations may actively promote oncogenesis by blocking the function of the remaining wild-type BRCA1 allele, although this hypothesis remains to be proved.


Subject(s)
BRCA1 Protein/antagonists & inhibitors , Genes, BRCA1 , Mutation , Prostatic Neoplasms/pathology , Apoptosis , BRCA1 Protein/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Division , DNA Damage , DNA Repair , DNA, Neoplasm/analysis , Drug Resistance, Neoplasm , Estrogen Receptor alpha , Female , Humans , Male , Neoplasm Proteins/metabolism , Phenotype , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Receptors, Estrogen/antagonists & inhibitors , Transfection , Tumor Cells, Cultured
19.
J Nutr ; 130(12): 2927-31, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11110848

ABSTRACT

Estrogen, via its binding to the estrogen receptor (ER), plays an important role in breast cancer cell proliferation and tumor development. Indole-3-carbinol (I3C), a compound occurring naturally in cruciferous vegetables, exhibits a potent antitumor activity via its regulation of estrogen activity and metabolism. This study was designed to determine the effect of I3C on the potential to inhibit the ER-alpha. Using a reporter gene driven by the estrogen receptor, I3C (10-125 micromol/L) significantly repressed the 17ss-estradiol (E2)-activated ER-alpha signaling in a dose-dependent manner. I3C and breast cancer susceptibility gene 1 (BRCA1) synergistically inhibited transcriptional activity of ER-alpha. Moreover, I3C down-regulated the expression of the estrogen-responsive genes, pS2 and cathepsin-D, and up-regulated BRCA1. The inhibitory effects of I3C did not contribute to its cytotoxic effects because these activities were observed at less than toxic concentrations. These results further suggest that antitumor activities of I3C are associated not only with its regulation of estrogen activity and metabolism, but also its modulation of ER transcription activity.


Subject(s)
Breast Neoplasms/genetics , Estrogen Antagonists/pharmacology , Indoles/pharmacology , Receptors, Estrogen/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/etiology , Cathepsin D/drug effects , Cathepsin D/genetics , Cathepsin D/metabolism , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Estrogen Receptor alpha , Female , Gene Expression Regulation, Neoplastic/drug effects , Genes, BRCA1/drug effects , Growth Inhibitors/metabolism , Humans , Proteins/drug effects , Proteins/genetics , Proteins/metabolism , Receptors, Estrogen/genetics , Sp1 Transcription Factor/drug effects , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/physiology , Transcription, Genetic/drug effects , Trefoil Factor-1 , Tumor Cells, Cultured , Tumor Suppressor Proteins
20.
Breast Cancer Res Treat ; 63(2): 147-52, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11097090

ABSTRACT

Indole-3-carbinol (I3C) is a promising phytochemical agent in chemoprevention of breast cancer. Our present study is the first description of I3C that significantly inhibits the cell adhesion, spreading and invasion associated with an up-regulation of PTEN (a tumor suppressor gene) and E-cadherin (a regulator of cell-cell adhesion) expression in T47-D human breast cancer cells. Therefore, I3C exhibits anti-cancer activities by suppressing breast tumor cell growth and metastatic spread. Metastatic breast cancer is a devastating problem, clinical application of I3C as a potent chemopreventive agent may be helpful in limiting breast cancer invasion and metastasis.


Subject(s)
Anticarcinogenic Agents/pharmacology , Breast Neoplasms/pathology , Indoles/pharmacology , Tumor Suppressor Proteins , Breast Neoplasms/prevention & control , Cadherins/analysis , Cell Adhesion , Cell Movement , Female , Humans , Neoplasm Invasiveness , PTEN Phosphohydrolase , Phosphoric Monoester Hydrolases/genetics , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...