Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Matern Child Nutr ; 13(4)2017 10.
Article in English | MEDLINE | ID: mdl-28133900

ABSTRACT

We investigated the long-term implications of infantile thiamine (vitamin B1) deficiency on motor function in preschoolers who had been fed during the first 2 years of life with a faulty milk substitute. In this retrospective cohort study, 39 children aged 5-6 years who had been exposed to a thiamine-deficient formula during infancy were compared with 30 age-matched healthy children with unremarkable infant nutritional history. The motor function of the participants was evaluated with The Movement Assessment Battery for Children (M-ABC) and the Zuk Assessment. Both evaluation tools revealed statistically significant differences between the exposed and unexposed groups for gross and fine motor development (p < .001, ball skills p = .01) and grapho-motor development (p = .004). The differences were especially noteworthy on M-ABC testing for balance control functioning (p < .001, OR 5.4; 95% CI 3.4-7.4) and fine motor skills (p < .001, OR 3.2; 95% CI 1.8-4.6). In the exposed group, both assessments concurred on the high rate of children exhibiting motor function difficulties in comparison to unexposed group (M-ABC: 56% vs. 10%, Zuk Assessment: 59% vs. 3%, p < .001). Thiamine deficiency in infancy has long-term implications on gross and fine motor function and balance skills in childhood, thiamine having a crucial role in normal motor development. The study emphasizes the importance of proper infant feeding and regulatory control of breast milk substitutes.


Subject(s)
Infant Nutritional Physiological Phenomena , Motor Skills , Thiamine Deficiency/epidemiology , Birth Weight , Case-Control Studies , Child , Child Development , Child, Preschool , Female , Humans , Infant , Infant Formula/chemistry , Male , Milk, Human/chemistry , Postural Balance , Retrospective Studies , Thiamine/administration & dosage , Thiamine/blood , Thiamine Deficiency/blood
2.
PLoS One ; 9(6): e101257, 2014.
Article in English | MEDLINE | ID: mdl-24979049

ABSTRACT

Post-streptococcal A (GAS) sequelae including movement and neuropsychiatric disorders have been associated with improvement in response to antibiotic therapy. Besides eradication of infection, the underlying basis of attenuation of neuropsychiatric symptoms following antibiotic treatment is not known. The aim of the present study was to test the efficacy of antibiotic treatment in a rat model of GAS-related neuropsychiatric disorders. In the model, rats were not infected but were exposed to GAS-antigen or to adjuvants only (Control rats) and treated continuously with the antibiotic ampicillin in their drinking water from the first day of GAS-antigen exposure. Two additional groups of rats (GAS and Control) did not receive ampicillin in their drinking water. Behavior of the four groups was assessed in the forced swim, marble burying and food manipulation assays. We assessed levels of D1 and D2 dopamine receptors and tyrosine hydroxylase in the prefrontal cortex and striatum, and IgG deposition in the prefrontal cortex, striatum and thalamus. Ampicillin treatment prevented emergence of the motor and some of the behavioral alterations induced by GAS-antigen exposure, reduced IgG deposition in the thalamus of GAS-exposed rats, and tended to attenuate the increase in the level of TH and D1 and D2 receptors in their striatum, without concomitantly reducing the level of sera anti-GAS antibodies. Our results reinforce the link between exposure to GAS antigen, dysfunction of central dopaminergic pathways and motor and behavioral alterations. Our data further show that some of these deleterious effects can be attenuated by antibiotic treatment, and supports the latter's possible efficacy as a prophylactic treatment in GAS-related neuropsychiatric disorders.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Antigens, Bacterial/immunology , Behavior, Animal/drug effects , Streptococcal Infections/drug therapy , Streptococcal Infections/immunology , Streptococcus pyogenes/immunology , Ampicillin/pharmacology , Ampicillin/therapeutic use , Animals , Anti-Bacterial Agents/pharmacology , Brain/drug effects , Brain/enzymology , Brain/pathology , Immunoglobulin G/immunology , Male , Rats, Inbred Lew , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism , Streptococcal Infections/microbiology , Swimming , Tyrosine 3-Monooxygenase/metabolism
3.
PLoS One ; 9(4): e91455, 2014.
Article in English | MEDLINE | ID: mdl-24690945

ABSTRACT

Anxiety disorders are a major public health concern worldwide. Studies indicate that repeated exposure to adverse experiences early in life can lead to anxiety disorders in adulthood. Current treatments for anxiety disorders are characterized by a low success rate and are associated with a wide variety of side effects. The aim of the present study was to evaluate the anxiolytic effects of a novel herbal treatment, in comparison to treatment with the selective serotonin reuptake inhibitor escitalopram. We recently demonstrated the anxiolytic effects of these treatments in BALB mice previously exposed to one week of stress. In the present study, ICR mice were exposed to post natal maternal separation and to 4 weeks of unpredictable chronic mild stress in adolescence, and treated during or following exposure to stress with the novel herbal treatment or with escitalopram. Anxiety-like behavior was evaluated in the elevated plus maze. Blood corticosterone levels were evaluated using radioimmunoassay. Brain derived neurotrophic factor levels in the hippocampus were evaluated using enzyme-linked immunosorbent assay. We found that (1) exposure to stress in childhood and adolescence increased anxiety-like behavior in adulthood; (2) the herbal treatment reduced anxiety-like behavior, both when treated during or following exposure to stress; (3) blood corticosterone levels were reduced following treatment with the herbal treatment or escitalopram, when treated during or following exposure to stress; (4) brain derived neurotrophic factor levels in the hippocampus of mice treated with the herbal treatment or escitalopram were increased, when treated either during or following exposure to stress. This study expands our previous findings and further points to the proposed herbal compound's potential to be highly efficacious in treating anxiety disorders in humans.


Subject(s)
Anxiety/drug therapy , Anxiety/etiology , Brain-Derived Neurotrophic Factor/blood , Citalopram/therapeutic use , Corticosterone/blood , Plant Extracts/therapeutic use , Stress, Psychological/complications , Animals , Behavior, Animal/drug effects , Citalopram/pharmacology , Mice, Inbred ICR , Motor Activity/drug effects
4.
Brain Behav Immun ; 38: 249-62, 2014 May.
Article in English | MEDLINE | ID: mdl-24561489

ABSTRACT

Group A ß-hemolytic streptococcal (GAS) infection is associated with a spectrum of neuropsychiatric disorders. The leading hypothesis regarding this association proposes that a GAS infection induces the production of auto-antibodies, which cross-react with neuronal determinants in the brain through the process of molecular mimicry. We have recently shown that exposure of rats to GAS antigen leads to the production of anti-neuronal antibodies concomitant with the development of behavioral alterations. The present study tested the causal role of the antibodies by assessing the behavior of naïve rats following passive transfer of purified antibodies from GAS-exposed rats. Immunoglobulin G (IgG) purified from the sera of GAS-exposed rats was infused directly into the striatum of naïve rats over a 21-day period. Their behavior in the induced-grooming, marble burying, food manipulation and beam walking assays was compared to that of naïve rats infused with IgG purified from adjuvant-exposed rats as well as of naïve rats. The pattern of in vivo antibody deposition in rat brain was evaluated using immunofluorescence and colocalization. Infusion of IgG from GAS-exposed rats to naïve rats led to behavioral and motor alterations partially mimicking those seen in GAS-exposed rats. IgG from GAS-exposed rats reacted with D1 and D2 dopamine receptors and 5HT-2A and 5HT-2C serotonin receptors in vitro. In vivo, IgG deposits in the striatum of infused rats colocalized with specific brain proteins such as dopamine receptors, the serotonin transporter and other neuronal proteins. Our results demonstrate the potential pathogenic role of autoantibodies produced following exposure to GAS in the induction of behavioral and motor alterations, and support a causal role for autoantibodies in GAS-related neuropsychiatric disorders.


Subject(s)
Corpus Striatum/immunology , Immunoglobulin G/immunology , Streptococcus pyogenes/immunology , Animals , Behavior, Animal , Corpus Striatum/metabolism , Male , Motor Activity , Rats , Rats, Inbred Lew , Receptors, Dopamine/metabolism , Receptors, Serotonin/metabolism
5.
Life Sci ; 94(2): 151-7, 2014 Jan 17.
Article in English | MEDLINE | ID: mdl-24184295

ABSTRACT

AIMS: Depression is a chronic, recurring and potentially life-threatening illness. Current treatments for depression are characterized by a low success rate and associated with a wide variety of side effects. The aim of the present study was to evaluate the behavioral and biological anti-depressant effects of a novel herbal treatment (NHT), as well as to assess its potential side effects, in comparison to treatment with the selective serotonin reuptake inhibitor escitalopram. MAIN METHODS: Depressive-like behavior was evaluated using the forced swim test (FST) and the tail suspension test (TST). Sexual behavior was evaluated following treatment by measuring latency before first mount and number of total mounts. Brain derived neurotrophic factor (BDNF) levels were evaluated using enzyme-linked immunosorbent assay. Serotonin transporter (SERT) levels in the pre-frontal cortex (PFC) and hypothalamus were evaluated using high affinity binding assay. KEY FINDINGS: (1) The NHT reduced depressive-like behavior in the FST and TST; (2) BDNF levels in the PFC of mice treated both with the NHT and escitalopram were increased; (3) SERT levels in the hypothalamus were significantly higher in the NHT group, in comparison to escitalopram and the control groups, and significantly lower in the PFC of the NHT group in comparison to the escitalopram group; and (4) the NHT led to less sexual dysfunction, compared to treatment with escitalopram. SIGNIFICANCE: Our NHT has the potential of being highly efficacious in treating depression in humans, while causing minimal to no influence on sexual function.


Subject(s)
Brain Chemistry/drug effects , Brain-Derived Neurotrophic Factor/analysis , Depression/drug therapy , Phytotherapy/methods , Stress, Psychological/drug therapy , Animals , Citalopram/therapeutic use , Crataegus , Depression/metabolism , Disease Models, Animal , Female , Hypothalamus/chemistry , Lilium , Male , Mice , Mice, Inbred ICR , Plant Preparations/therapeutic use , Prefrontal Cortex/chemistry , Serotonin Plasma Membrane Transport Proteins/analysis , Selective Serotonin Reuptake Inhibitors/therapeutic use , Sexual Behavior, Animal/drug effects , Sexual Behavior, Animal/physiology , Stress, Psychological/psychology , Triticum
6.
Life Sci ; 90(25-26): 995-1000, 2012 Jun 27.
Article in English | MEDLINE | ID: mdl-22683433

ABSTRACT

AIMS: Anxiety and stress disorders are currently among the ten most important public health concerns, and in recent years, have reached epidemic proportions. The current success rate of treatments for anxiety disorders is not high, reaching 50% at most. These treatments are also associated with a wide variety of side effects. The aim of the present study was to investigate the anxiolytic properties of a novel herbal treatment produced in our laboratory compared to a conventional treatment for anxiety disorders, namely SSRIs. MAIN METHODS: Anxiety-like behavior was evaluated in adult mice exposed to stress during childhood following 1, 2 and 3 weeks of treatment with the novel herbal treatment or escitalopram, using the novel open field and the elevated plus maze paradigms. The behavioral evaluation in these mice was followed by a biochemical assessment of their brain hippocampal BDNF levels and blood corticosterone levels. KEY FINDINGS: The study showed that (1) the novel herbal treatment reduced anxiety-like behaviors in both behavioral tests. Interestingly, this reduction was observed only following a 3-week treatment; (2) following the novel treatment, corticosterone levels in the plasma of treated mice were reduced and this reduction was similar to the one observed following escitalopram treatment; and (3) BDNF levels in the hippocampus of mice treated both with the novel herbal treatment and escitalopram were increased. SIGNIFICANCE: These behavioral and biological findings indicate that our novel herbal compound has the potential of being highly efficacious in treating anxiety disorders.


Subject(s)
Anti-Anxiety Agents/therapeutic use , Anxiety/drug therapy , Plant Preparations/therapeutic use , Animals , Anti-Anxiety Agents/pharmacology , Anxiety/blood , Anxiety/psychology , Corticosterone/blood , Disease Models, Animal , Male , Maze Learning/drug effects , Maze Learning/physiology , Mice , Mice, Inbred BALB C , Plant Preparations/pharmacology , Treatment Outcome
7.
Neuropsychopharmacology ; 37(9): 2076-87, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22534626

ABSTRACT

Group A streptococcal (GAS) infections and autoimmunity are associated with the onset of a spectrum of neuropsychiatric disorders in children, with the prototypical disorder being Sydenham chorea (SC). Our aim was to develop an animal model that resembled the behavioral, pharmacological, and immunological abnormalities of SC and other streptococcal-related neuropsychiatric disorders. Male Lewis rats exposed to GAS antigen exhibited motor symptoms (impaired food manipulation and beam walking) and compulsive behavior (increased induced-grooming). These symptoms were alleviated by the D2 blocker haloperidol and the selective serotonin reuptake inhibitor paroxetine, respectively, drugs that are used to treat motor symptoms and compulsions in streptococcal-related neuropsychiatric disorders. Streptococcal exposure resulted in antibody deposition in the striatum, thalamus, and frontal cortex, and concomitant alterations in dopamine and glutamate levels in cortex and basal ganglia, consistent with the known pathophysiology of SC and related neuropsychiatric disorders. Autoantibodies (IgG) of GAS rats reacted with tubulin and caused elevated calcium/calmodulin-dependent protein kinase II signaling in SK-N-SH neuronal cells, as previously found with sera from SC and related neuropsychiatric disorders. Our new animal model translates directly to human disease and led us to discover autoantibodies targeted against dopamine D1 and D2 receptors in the rat model as well as in SC and other streptococcal-related neuropsychiatric disorders.


Subject(s)
Chorea/psychology , Disease Models, Animal , Mental Disorders/psychology , Motor Activity/immunology , Streptococcal Infections/psychology , Streptococcus pyogenes , Adolescent , Animals , Autoantibodies/biosynthesis , Child , Child, Preschool , Chorea/immunology , Chorea/microbiology , Female , Grooming/drug effects , Grooming/physiology , Humans , Male , Mental Disorders/immunology , Mental Disorders/microbiology , Motor Activity/drug effects , Rats , Rats, Inbred Lew , Streptococcal Infections/drug therapy , Streptococcal Infections/immunology , Streptococcus pyogenes/drug effects , Streptococcus pyogenes/immunology
8.
Cancer Res ; 70(21): 8917-26, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20959490

ABSTRACT

Signal transducers and activators of transcription 3 (Stat3) is activated by cytokines and growth factors in lung cancers and regulates expression of genes implicated in cell growth, survival, and transformation. Previously, we found that mice with a deletion of the G protein-coupled receptor, family C, group 5, member a (Gprc5a) gene develop lung tumors, indicating that Gprc5a is a tumor suppressor. Herein, we show that epithelial cells from Gprc5a knockout mouse lung (Gprc5a(-/-) cells) survive better in vitro in medium deprived of exogenous growth factors and form more colonies in semisolid medium than their counterparts from wild-type mice (Gprc5a(+/+) cells). Stat3 tyrosine 705 phosphorylation and expression of several Stat3-regulated antiapoptotic genes were higher in Gprc5a(-/-) than in Gprc5a(+/+) cells. Both cell types secreted leukemia inhibitory factor (Lif); however, whereas Stat3 activation was persistent in Gprc5a(-/-) cells, it was transient in Gprc5a(+/+) cells. Lung adenocarcinoma cells isolated from Gprc5a(-/-) mice also exhibited autocrine Lif-mediated Stat3 activation. The level of Socs3, the endogenous Stat3 inhibitory protein, was higher in Gprc5a(+/+) than in Gprc5a(-/-) cells, and expression of the tumor suppressor stabilized Socs3. Inhibition of Stat3 signaling in Gprc5a(-/-) normal and cancer cells by the Janus-activated kinase 2 inhibitor AG490 or by a dominant negative Stat3(Y705F) increased starvation-induced apoptosis and inhibited colony formation. These results show that persistent Stat3 activation is important for the survival and transformation of Gprc5a(-/-) lung cells and suggest that the tumor suppressive effects of Gprc5a are mediated, at least in part, by inhibition of Stat3 signaling through Socs3 stabilization.


Subject(s)
Adenocarcinoma/pathology , Cell Transformation, Neoplastic/pathology , Leukemia Inhibitory Factor/pharmacology , Lung Neoplasms/pathology , Lung/pathology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/physiology , STAT3 Transcription Factor/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Animals , Apoptosis , Blotting, Western , Cell Adhesion , Cell Movement , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Humans , Kidney/cytology , Kidney/metabolism , Lung/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , RNA, Messenger/genetics , Receptors, G-Protein-Coupled/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Deletion , Signal Transduction
9.
PLoS One ; 5(7): e11847, 2010 Jul 29.
Article in English | MEDLINE | ID: mdl-20686609

ABSTRACT

BACKGROUND: Improved understanding of lung cancer development and progression, including insights from studies of animal models, are needed to combat this fatal disease. Previously, we found that mice with a knockout (KO) of G-protein coupled receptor 5A (Gprc5a) develop lung tumors after a long latent period (12 to 24 months). METHODOLOGY/PRINCIPAL FINDINGS: To determine whether a tobacco carcinogen will enhance tumorigenesis in this model, we administered 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) i.p. to 2-months old Gprc5a-KO mice and sacrificed groups (n=5) of mice at 6, 9, 12, and 18 months later. Compared to control Gprc5a-KO mice, NNK-treated mice developed lung tumors at least 6 months earlier, exhibited 2- to 4-fold increased tumor incidence and multiplicity, and showed a dramatic increase in lesion size. A gene expression signature, NNK-ADC, of differentially expressed genes derived by transcriptome analysis of epithelial cell lines from normal lungs of Gprc5a-KO mice and from NNK-induced adenocarcinoma was highly similar to differential expression patterns observed between normal and tumorigenic human lung cells. The NNK-ADC expression signature also separated both mouse and human adenocarcinomas from adjacent normal lung tissues based on publicly available microarray datasets. A key feature of the signature, up-regulation of Ube2c, Mcm2, and Fen1, was validated in mouse normal lung and adenocarcinoma tissues and cells by immunohistochemistry and western blotting, respectively. CONCLUSIONS/SIGNIFICANCE: Our findings demonstrate that lung tumorigenesis in the Gprc5a-KO mouse model is augmented by NNK and that gene expression changes induced by tobacco carcinogen(s) may be conserved between mouse and human lung epithelial cells. Further experimentation to prove the reliability of the Gprc5a knockout mouse model for the study of tobacco-induced lung carcinogenesis is warranted.


Subject(s)
Adenocarcinoma/genetics , Carcinogens/toxicity , Genomics/methods , Lung Neoplasms/genetics , Nitrosamines/toxicity , Receptors, G-Protein-Coupled/physiology , Animals , Blotting, Western , Cyclin D1/genetics , Cyclin D1/metabolism , Flap Endonucleases/genetics , Flap Endonucleases/metabolism , Humans , Immunohistochemistry , In Vitro Techniques , Mice , Mice, Knockout , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Receptors, G-Protein-Coupled/genetics , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism
10.
Neoplasia ; 12(6): 499-505, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20563252

ABSTRACT

Increasing the understanding of the impact of changes in oncogenes and tumor suppressor genes is essential for improving the management of lung cancer. Recently, we identified a new mouse lung-specific tumor suppressor-the G protein-coupled receptor 5A (Gprc5a). Microarray analysis of the transcriptomes of lung epithelial cells cultured from normal tracheas of Gprc5a knockout and wild-type mice defined a loss-of-Gprc5a gene signature, which revealed many aberrations in cancer-associated pathways. To assess the relevance of this mouse tumor suppressor to human lung cancer, the loss-of-Gprc5a signature was cross species compared with and integrated with publicly available gene expression data of human normal lung tissue and non-small cell lung cancers. The loss-of-Gprc5a signature was prevalent in human lung adenocarcinomas compared with squamous cell carcinomas or normal lung. Furthermore, it identified subsets of lung adenocarcinomas with poor outcome. These results demonstrate that gene expression patterns of Gprc5a loss in nontumorigenic mouse lung epithelial cells are evolutionarily conserved and important in human lung adenocarcinomas.


Subject(s)
Adenocarcinoma/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Squamous Cell/genetics , Lung Neoplasms/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/physiology , Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/mortality , Female , Gene Expression Profiling , Genes, Tumor Suppressor , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate
11.
Cancer Prev Res (Phila) ; 3(4): 424-37, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20354164

ABSTRACT

Mouse models can be useful for increasing the understanding of lung tumorigenesis and assessing the potential of chemopreventive agents. We explored the role of inflammation in lung tumor development in mice with knockout of the tumor suppressor Gprc5a. Examination of normal lung tissue and tumors from 51 Gprc5a(+/+) (adenoma incidence, 9.8%; adenocarcinoma, 0%) and 38 Gprc5a(-/-) mice (adenoma, 63%; adenocarcinoma, 21%) revealed macrophage infiltration into lungs of 45% of the Gprc5a(-/-) mice and 8% of Gprc5a(+/+) mice and the direct association of macrophages with 42% of adenomas and 88% of adenocarcinomas in the knockout mice. Gprc5a(-/-) mouse lungs contained higher constitutive levels of proinflammatory cytokines and chemokines and were more sensitive than lungs of Gprc5a(+/+) mice to stimulation of NF-kappaB activation by lipopolysaccharide in vivo. Studies with epithelial cells cultured from tracheas of Gprc5a(-/-) and Gprc5a(+/+) mice revealed that Gprc5a loss is associated with increased cell proliferation, resistance to cell death in suspension, and increased basal, tumor necrosis factor alpha-induced, and lipopolysaccharide-induced NF-kappaB activation, which were reversed partially in Gprc5a(-/-) adenocarcinoma cells by reexpression of Gprc5a. Compared with Gprc5a(+/+) cells, the Gprc5a(-/-) cells produced higher levels of chemokines and cytokines and their conditioned medium induced more extensive macrophage migration. Silencing Gprc5a and the p65 subunit of NF-kappaB in Gprc5a(+/+) and Gprc5a(-/-) cells, respectively, reversed these effects. Thus, Gprc5a loss enhances NF-kappaB activation in lung epithelial cells, leading to increased autocrine and paracrine interactions, cell autonomy, and enhanced inflammation, which may synergize in the creation of a tumor-promoting microenvironment.


Subject(s)
Adenocarcinoma/metabolism , Cell Transformation, Neoplastic/metabolism , Lung Neoplasms/metabolism , NF-kappa B/metabolism , Pneumonia/metabolism , Receptors, G-Protein-Coupled/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Animals , Cell Transformation, Neoplastic/genetics , Chemotaxis, Leukocyte/immunology , Electrophoretic Mobility Shift Assay , Enzyme Activation/genetics , Immunoblotting , Immunohistochemistry , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Pneumonia/genetics , Pneumonia/immunology , RNA, Messenger/analysis , RNA, Small Interfering , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/genetics , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Reverse Transcriptase Polymerase Chain Reaction
12.
Cancer Prev Res (Phila) ; 3(3): 331-8, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20145189

ABSTRACT

Curcumin has shown some promise in the prevention of oral carcinogenesis by mechanism(s) that are still not completely resolved. Messenger RNA translation is mediated in eukaryotes by the eIF4F complex composed of eukaryotic translation initiation factors eIF4E, eIF4G, and eIF4A. Overexpression of some of these components or the inactivation of initiation repressor proteins (4E-BP1) has been implicated in cancer development including oral carcinogenesis by affecting cell survival, angiogenesis, and tumor growth and invasion. In this study, we examined the possibility that curcumin affects the translational machinery differently in normal, immortalized normal, leukoplakia, and malignant cells. Curcumin treatment in vitro inhibited the growth of immortalized oral mucosa epithelial cells (NOM9-CT) and the leukoplakia cells (MSK-Leuk1s) as well as in the UMSCC22B and SCC4 cells derived from head and neck squamous cell carcinoma. Curcumin only exerted minor effects on the growth of normal oral epithelial cells (NOM9). In the immortalized, leukoplakia, and cancer cells, curcumin inhibited cap-dependent translation by suppressing the phosphorylation of 4E-BP1, eIF4G, eIF4B, and Mnk1, and also reduced the total levels of eIF4E and Mnk1. Our findings show that immortalized normal, leukoplakia, and malignant oral cells are more sensitive to curcumin and show greater modulation of protein translation machinery than the normal oral cells, indicating that targeting this process may be an important approach to chemoprevention in general and for curcumin in particular.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Line, Transformed/drug effects , Curcumin/pharmacology , Mouth Mucosa/drug effects , Peptide Chain Initiation, Translational/drug effects , Precancerous Conditions/drug therapy , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/metabolism , Blotting, Western , Cell Cycle Proteins , Cell Line, Transformed/cytology , Cell Proliferation/drug effects , Cells, Cultured , Eukaryotic Initiation Factor-4G/antagonists & inhibitors , Eukaryotic Initiation Factor-4G/metabolism , Eukaryotic Initiation Factors/antagonists & inhibitors , Eukaryotic Initiation Factors/metabolism , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/metabolism , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/metabolism , Phosphorylation/drug effects , Precancerous Conditions/pathology , Protein Biosynthesis/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism
13.
Cancer Prev Res (Phila) ; 2(8): 702-11, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19638491

ABSTRACT

Lung cancer continues to be a major deadly malignancy. The mortality of this disease could be reduced by improving the ability to predict cancer patients' survival. We hypothesized that genes differentially expressed among cells constituting an in vitro human lung carcinogenesis model consisting of normal, immortalized, transformed, and tumorigenic bronchial epithelial cells are relevant to the clinical outcome of non-small cell lung cancer (NSCLC). Multidimensional scaling, microarray, and functional pathways analyses of the transcriptomes of the above cells were done and combined with integrative genomics to incorporate the microarray data with published NSCLC data sets. Up-regulated (n = 301) and down-regulated genes (n = 358) displayed expression level variation across the in vitro model with progressive changes in cancer-related molecular functions. A subset of these genes (n = 584) separated lung adenocarcinoma clinical samples (n = 361) into two clusters with significant survival differences. Six genes, UBE2C, TPX2, MCM2, MCM6, FEN1, and SFN, selected by functional array analysis, were also effective in prognosis. The mRNA and protein levels of one these genes-UBE2C-were significantly up-regulated in NSCLC tissue relative to normal lung and increased progressively in lung lesions. Moreover, stage I NSCLC patients with positive UBE2C expression exhibited significantly poorer overall and progression-free survival than patients with negative expression. Our studies with this in vitro model have lead to the identification of a robust six-gene signature, which may be valuable for predicting the survival of lung adenocarcinoma patients. Moreover, one of those genes, UBE2C, seems to be a powerful biomarker for NSCLC survival prediction.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma, Non-Small-Cell Lung/diagnosis , Cell Transformation, Neoplastic/genetics , Gene Expression Profiling , Lung Neoplasms/diagnosis , Adenocarcinoma/diagnosis , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Biomarkers, Tumor/genetics , Biomarkers, Tumor/isolation & purification , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/mortality , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Oligonucleotide Array Sequence Analysis , Prognosis , Survival Analysis , Tumor Cells, Cultured , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism
14.
Cancer Biol Ther ; 8(9): 801-7, 2009 May.
Article in English | MEDLINE | ID: mdl-19333010

ABSTRACT

Phorbol 12-myristate 13-acetate (PMA) modulates cell proliferation and survival by activating several intracellular signaling pathways. Protein kinase C (PKC) plays a key role in PMA-induced growth arrest of non-small cell lung cancer (NSCLC) cells. Kruppel-like transcription factor 6 (KLF6), which is associated with negative control of cell proliferation, is downregulated in many cancers, including NSCLC. In this study, we found that KLF6 is downregulated in 17 lung cancer cell lines and in cells representing early stages of lung cancer development. Moreover, PMA induced cell growth arrest through KLF6 induction in H358 NSCLC cells. The increase in KLF6 by PMA was associated with upregulation of the cyclin-dependent kinase inhibitors (CDKIs) p21(WAF1/CIP1) and p27(KIP1). In addition, inhibition of PKC or JNK activation decreased PMA-induced KLF6 induction and activation of PKC alone by Bryostatin-1 and Thymeleatoxin increased KLF6 levels. Moreover, siRNA-mediated knockdown of KLF6 reduced PMA-induced cell growth inhibition concomitantly with decreased expression of both p21(WAF1/CIP1) and p27(KIP1), and in accordance, overexpression of KLF6 alone upregulated both CDKIs protein levels. Our results demonstrate the induction of the tumor suppressor KLF6 following PKC activation and its importance for PMA-mediated cancer cell growth arrest.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Kruppel-Like Transcription Factors/biosynthesis , Lung Neoplasms/metabolism , Protein Kinase C/metabolism , Proto-Oncogene Proteins/biosynthesis , Tetradecanoylphorbol Acetate/pharmacology , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Growth Processes/drug effects , Cell Growth Processes/physiology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Down-Regulation , Enzyme Activation/drug effects , G1 Phase/drug effects , Humans , Kruppel-Like Factor 6 , Kruppel-Like Transcription Factors/genetics , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Phosphorylation/drug effects , Proto-Oncogene Proteins/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , S Phase/drug effects
15.
Cancer Res ; 68(11): 4416-23, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18519704

ABSTRACT

The synthetic retinoid N-(4-hydroxyphenyl)retinamide (4HPR) has shown potential as a chemopreventive and therapeutic agent. The ability of 4HPR to enhance production of reactive oxygen species (ROS) leading to apoptosis has been suggested as a possible mechanism underlying these effects. We explored the possibility that ROS induction by 4HPR involves the small GTPase Ras-related C3 botulinum toxin substrate (Rac), a regulatory subunit of the NADPH oxidase complex. Rac was activated in human head and neck squamous cell carcinoma (HNSCC) cells as early as 5 minutes following 4HPR exposure. Moreover, inhibition of Rac activity or silencing of its expression by RNA interference decreased ROS generation in human head and neck, lung, and cervical cancer cells and murine melanoma cells. In HNSCC UMSCC-22B cells, this decrease correlated with reduction in apoptosis induction by 4HPR. Expression of a constitutive active mutant Rac increased basal and 4HPR-induced ROS generation and poly(ADP-ribose) polymerase cleavage. In addition, the metastatic DM14 cells exhibited higher Rac activation following 4HPR treatment compared with the primary Tu167-C2 cells. Furthermore, the metastatic cancer cells tested exhibited higher ROS generation and growth inhibition due to 4HPR exposure compared with their primary cancer cell counterparts. These findings show a preferential susceptibility of metastatic cells to the proapoptotic retinoid 4HPR through Rac activation and support the use of ROS-inducing agents such as 4HPR against metastatic cancer cells.


Subject(s)
Apoptosis/drug effects , Fenretinide/pharmacology , rac GTP-Binding Proteins/physiology , Aminoquinolines/pharmacology , Blotting, Western , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Division/drug effects , Cell Line, Tumor , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Pyrimidines/pharmacology , Reactive Oxygen Species/metabolism , rac GTP-Binding Proteins/antagonists & inhibitors
16.
J Natl Cancer Inst ; 99(22): 1668-82, 2007 Nov 21.
Article in English | MEDLINE | ID: mdl-18000218

ABSTRACT

BACKGROUND: Lung cancers develop via multiple genetic and epigenetic changes, including inactivation of tumor suppressor genes. We previously cloned human G protein-coupled receptor family C type 5A (GPRC5A), whose expression is suppressed in some human lung carcinoma cells, and its mouse homolog Gprc5a. METHODS: We generated Gprc5a knockout mice by homologous recombination and studied their phenotype by macroscopic observation and microscopic histologic analysis of embryos and lungs of 1- to 2-year-old mice. GPRC5A mRNA expression was analyzed by reverse transcription-polymerase chain reaction in surgical specimens of 18 human lung tumors and adjacent normal tissues and by analyzing previously published data from 186 lung tumor tissues of a variety of histologic types and 17 normal lung samples. Human embryonic kidney, human non-small-cell lung cancer, and mouse lung adenocarcinoma cells were transfected with a GPRC5A expression vector or a control vector, and colony formation in semisolid medium was assayed. Statistical tests were two-sided. RESULTS: Homozygous knockout mice developed many more lung tumors at 1-2 years of age (incidence: 76% adenomas and 17% adenocarcinomas) than heterozygous (11% adenomas) or wild-type (10% adenomas) mice. Human GPRC5A mRNA levels were lower in most (11 of 18 [61%]) human lung tumors than in adjacent normal tissues. The mean GPRC5A mRNA level in adenocarcinoma (n = 139), squamous cell carcinoma (n = 21), small-cell lung cancer (n = 6), and carcinoid (n = 20) tissues was 46.2% (P = .014), 7.5% (P<.001), 5.3% (P<.001), and 1.8% (P<.001), respectively, that in normal lung tissues (n = 17) GPRC5A transfection suppressed colony formation in semisolid medium of immortalized human embryonic kidney, human non-small-cell lung cancer, and mouse lung adenocarcinoma cells by 91%, 91%, and 68%, respectively, compared with vector controls (all P<.001). CONCLUSIONS: Gprc5a functions as a tumor suppressor in mouse lung, and human GPRC5A may share this property. The Gprc5a-deficient mouse is a novel model to study lung carcinogenesis and chemoprevention.


Subject(s)
Adenocarcinoma/chemistry , Genes, Tumor Suppressor , Lung Neoplasms/chemistry , Lung/chemistry , Neoplastic Stem Cells/chemistry , Receptors, G-Protein-Coupled/genetics , Respiratory Mucosa/pathology , Adenocarcinoma/pathology , Animals , Blotting, Northern , Carcinoma, Non-Small-Cell Lung/chemistry , Carcinoma, Small Cell/chemistry , Cell Line, Tumor , Cell Transformation, Neoplastic , Disease Models, Animal , Embryonic Stem Cells , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic , Genes, ras , Genetic Predisposition to Disease , Homozygote , Humans , Immunoblotting , Immunohistochemistry , In Situ Hybridization , Lung Neoplasms/pathology , Mice , Mice, Knockout , Mutation , Neoplasm Proteins/genetics , Phenotype , Protein Array Analysis , RNA, Messenger/analysis , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Tumor Stem Cell Assay
17.
Cancer Biol Ther ; 6(5): 705-11, 2007 May.
Article in English | MEDLINE | ID: mdl-17404501

ABSTRACT

N-(4-hydroxyphenyl)retinamide (4HPR), which has shown efficacy in cancer chemopreventionand therapy, induces the mitochondrial apoptosis pathway via increased generation of reactive oxygen species (ROS). ROS is also known to be able to induce an endoplasmic reticulum (ER) stress response, which can contribute to apoptosis but may also antagonize it. Therefore, we used human head and neck squamous cell carcinoma (HNSCC) cells to determine whether 4HPR affects ER stress. Different experimental approaches have indicated that 4HPR induces ER stress response: electron microscopy, which showed extensive ER dilation; splicing of the X-box binding protein 1 (XBP-1), a marker of unfolded protein response (UPR) activation; and quantitative real-time PCR and immunoblotting, which revealed the upregulation of several ER-stress associated mRNAs and proteins, including the chaperone heat shock protein HSPA1A. Most of these effects of 4HPR were abrogated by cotreatment of cells with the antioxidant 3-tert-butyl-4-hydroxyanisole (BHA) indicating that they were downstream of the increase in ROS. Furthermore, siRNA-mediated silencing and chemical inhibition of HSPA1A, which exerts either pro- or anti-apoptotic effects, decreased 4HPR-induced apoptosis. These results demonstrate that 4HPR induces ER stress and uncovered a pro-apoptotic role for HSPA1A in 4HPR-induced apoptosis.


Subject(s)
Anticarcinogenic Agents/pharmacology , Endoplasmic Reticulum/drug effects , Fenretinide/pharmacology , Head and Neck Neoplasms/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism , Antioxidants/pharmacology , Apoptosis/drug effects , Blotting, Western , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Caspases/metabolism , Cytochromes c/metabolism , DNA-Binding Proteins/genetics , Endoplasmic Reticulum/metabolism , Enzyme Activation , Enzyme Inhibitors/pharmacology , HSP70 Heat-Shock Proteins/antagonists & inhibitors , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Head and Neck Neoplasms/pathology , Humans , Nuclear Proteins/genetics , Poly(ADP-ribose) Polymerases , RNA Splicing , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Regulatory Factor X Transcription Factors , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors , Tumor Cells, Cultured , X-Box Binding Protein 1
18.
Cancer Res ; 66(19): 9762-70, 2006 Oct 01.
Article in English | MEDLINE | ID: mdl-17018636

ABSTRACT

Celecoxib is being evaluated as a chemopreventive agent. However, its mechanism of action is not clear because high doses were used for in vitro studies to obtain antitumor effects. We found that celecoxib inhibited the growth of premalignant and malignant human bronchial epithelial cells with IC(50) values between 8.9 and 32.7 micromol/L, irrespective of cyclooxygenase-2 (COX-2) expression. Normal human bronchial epithelial cells were less sensitive to celecoxib. Because these concentrations were higher than those attainable in vivo (

Subject(s)
Anticarcinogenic Agents/pharmacology , Apoptosis/physiology , Bronchi/drug effects , Bronchial Diseases/pathology , Cyclooxygenase 2 Inhibitors/pharmacology , Fenretinide/pharmacology , Precancerous Conditions/pathology , Proto-Oncogene Proteins c-akt/physiology , Pyrazoles/pharmacology , Sulfonamides/pharmacology , Apoptosis/drug effects , Bronchi/cytology , Celecoxib , Cell Line, Tumor/cytology , Cell Line, Tumor/drug effects , Cells, Cultured/cytology , Cells, Cultured/drug effects , Drug Screening Assays, Antitumor , Drug Synergism , Epithelial Cells/cytology , Epithelial Cells/drug effects , Humans , Lung Neoplasms/pathology , Lung Neoplasms/prevention & control , Mitochondria/physiology , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Proto-Oncogene Proteins c-akt/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/physiology
19.
Cancer Biol Ther ; 5(5): 518-22, 2006 May.
Article in English | MEDLINE | ID: mdl-16582595

ABSTRACT

Growth and Differentiation Factor-15 (GDF-15, NAG-1, MIC-1) is induced by several apoptosis-inducing agents including the retinoid-related molecule (RRM) 6-[3-(1-adamantyl-4-hydroxyphenyl]-2-naphthalene carboxylic acid (CD437). It has been suggested that GDF-15 may be involved in the induction of apoptosis by CD437 in H460 lung cancer cells. The present study was designed to probe this hypothesis more directly. Several RRMs (CD437, ST1926 and MX3350-1) but not the retinoids all-trans- retinoic acid and 4HPR were able to induce GDF-15 in H460 cells. A similar differential effect of these retinoids was observed for the induction of p53, which has been reported to regulate GDF-15 expression. In H460 cells transfected with a neo vector control (H460-Neo), treatment with RRMs but not ATRA or 4HPR resulted in increases in p53, GDF-15 and apoptosis evidenced by poly(ADP ribose) polymerase (PARP) cleavage. In contrast, RRMs failed to increase p53 or induce apoptosis in H460 cells in which p53 was inactivated by transfection of the human papillomavirus E6-6 (H460-E6-6). The increase in GDF-15 by RRMs was also compromised in the H460-E6-6 cells. Because PARP cleavage was only evident when GDF-15 levels where elevated it appeared that GDF-15 was mediating the pro-apoptotic effects of RRMs. However, silencing of GDF-15 induction by RNA interference failed to decrease the ability of CD437 and ST1926 to induce apoptosis. These results demonstrate that GDF-15 is dispensable for the pro-apoptotic activity of CD437 and ST1926.


Subject(s)
Adamantane/analogs & derivatives , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cinnamates/pharmacology , Cytokines/metabolism , Lung Neoplasms/metabolism , Retinoids/pharmacology , Adamantane/pharmacology , Blotting, Western , Carcinoma, Large Cell/metabolism , Carcinoma, Large Cell/pathology , Cytokines/antagonists & inhibitors , Cytokines/genetics , Gene Expression Regulation, Neoplastic/drug effects , Growth Differentiation Factor 15 , Humans , Lung Neoplasms/pathology , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/metabolism , Poly(ADP-ribose) Polymerases/metabolism , RNA, Small Interfering/pharmacology , Repressor Proteins/genetics , Repressor Proteins/metabolism , Tretinoin/pharmacology , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
20.
Cancer Biol Ther ; 4(4): 407-13, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15846100

ABSTRACT

Celecoxib exhibits cancer preventive and therapeutic effects in animal models and clinical trials. It presumably acts through selective inhibition of cyclooxygenase-2 (COX-2) and subsequent reduction of prostaglandin (PG) synthesis. However, the concentrations of celecoxib required for growth inhibition and apoptosis induction in vitro are higher than those needed for suppression of PGs. Moreover, those concentrations are not achievable in humans raising a controversy regarding the clinical relevance of in vitro data. We investigated the activity of celecoxib alone and in combination with the pro-apoptotic retinoid N-(4-hydroxyphenyl)retinamide (4HPR) on growth and apoptosis of human nonsmall cell lung cancer (NSCLC) cell lines. Celecoxib inhibited growth of thirteen NSCLC cell lines with IC50 values ranging from 19 to 33 microM regardless of their COX-2 expression. Apoptosis was induced in cells with high (A549) as well as low (H1792) COX-2 levels but only at a concentration of 75 microM celecoxib. However, treatment with pharmacologically feasible concentrations of celecoxib (< or = 10 microM) in combination with 4HPR (< or = 2 microM) resulted in a marked suppression of NSCLC cell growth and colony formation. Apoptosis mediated by activation of caspase-3, cleavage of PARP and lamin A was suppressed by addition of antioxidants, suggesting that the generation of reactive oxygen species was partially involved. This study indicates, that celecoxib combined with 4HPR is more effective than treatment with either agent alone in inhibition of growth and induction of apoptosis in NSCLC cells. It suggests further investigations of this combination for lung cancer treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Proliferation/drug effects , Cyclooxygenase Inhibitors/pharmacology , Fenretinide/pharmacology , Lung Neoplasms/drug therapy , Pyrazoles/pharmacology , Sulfonamides/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Caspase 3 , Caspases/metabolism , Celecoxib , Cell Line, Tumor , Drug Synergism , Enzyme Activation/drug effects , Humans , Inhibitory Concentration 50 , Lung Neoplasms/metabolism , Lung Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...