Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Acta Histochem ; 113(2): 150-5, 2011 Feb.
Article in English | MEDLINE | ID: mdl-19853285

ABSTRACT

PRDM16 is a member of the PR domain-containing protein family and is associated with various disease states including myelodysplastic syndrome and adult T-cell leukemia, as well as developmental abnormalities such as cleft palate. It is also known to act as a regulator of cell differentiation. Expression analysis of PRDM16 is limited, especially within the developing embryo. The current study evaluated the temporal and spatial localization of PRDM16 during early mouse development (embryonic days 8.5-14.5). PRDM16 was first detected on E9.5 in a limited number of tissues and by E14.5, was expressed in a broad range of developing tissues including those of the brain, lung, kidney, and gastrointestinal tract. The expression pattern is consistent with a role for PRDM16 in the development of multiple tissues. Collectively, these studies are the first to characterize the expression of the PRDM16 gene during early murine development.


Subject(s)
DNA-Binding Proteins/biosynthesis , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Transcription Factors/biosynthesis , Animals , DNA-Binding Proteins/genetics , Female , Male , Mice , Mice, Inbred ICR , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics
2.
Reprod Toxicol ; 29(2): 164-75, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19969065

ABSTRACT

BACKGROUND: Little is known about the effects of passive smoke exposures on the developing brain. OBJECTIVE: The purpose of the current study was to identify changes in gene expression in the murine hippocampus as a consequence of in utero exposure to sidestream cigarette smoke (an experimental equivalent of environmental tobacco smoke (ETS)) at exposure levels that do not result in fetal growth inhibition. METHODS: A whole body smoke inhalation exposure system was utilized to deliver ETS to pregnant C57BL/6J mice for 6 h/day from gestational days 6-17 (gd 6-17) [for microarray] or gd 6-18.5 [for fetal phenotyping]. RESULTS: There were no significant effects of ETS exposure on fetal phenotype. However, 61 "expressed" genes in the gd 18.5 fetal hippocampus were differentially regulated (up- or down-regulated by 1.5-fold or greater) by maternal exposure to ETS. Of these 61 genes, 25 genes were upregulated while 36 genes were down-regulated. A systems biology approach, including computational methodologies, identified cellular response pathways, and biological themes, underlying altered fetal programming of the embryonic hippocampus by in utero cigarette smoke exposure. CONCLUSIONS: Results from the present study suggest that even in the absence of effects on fetal growth, prenatal smoke exposure can alter gene expression during the "early" period of hippocampal growth and may result in abnormal hippocampal morphology, connectivity, and function.


Subject(s)
Gene Expression Regulation, Developmental/drug effects , Gene Expression/drug effects , Hippocampus/drug effects , Maternal Exposure/adverse effects , Tobacco Smoke Pollution/adverse effects , Abnormalities, Drug-Induced/genetics , Animals , Down-Regulation/drug effects , Female , Fetal Development/drug effects , Fetal Development/genetics , Gene Expression Profiling , Gene Regulatory Networks , Gestational Age , Hippocampus/abnormalities , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Pregnancy , Prenatal Exposure Delayed Effects , RNA, Messenger/metabolism , Up-Regulation/drug effects
3.
Int J Dev Biol ; 53(7): 1097-104, 2009.
Article in English | MEDLINE | ID: mdl-19598128

ABSTRACT

Mutations in each of the transcriptional co-activator genes - CBP, p300, Cited2, Cart1 and Carm1 - result in neural tube defects in mice. The present study thus furnishes a complete and comparative temporal and spatial expression map of CBP/p300 and associated transcriptional co-activators, Cited2, Cart1 and Carm1 during the period of murine neural tube development (embryonic days 8.5 to 10.5). Each co-activator except Cart1 was expressed in the dorsal neural folds on E8.5. Although CBP and p300 are functionally interchangeable in vitro, their respective expression patterns diverge during embryogenesis before neural fold fusion is complete. CBP gene expression was lost from the neural folds by E8.75 and was thereafter weakly expressed in the maxillary region and limb buds, while p300 exhibited strong expression in the first branchial arch, limb bud and telencephalic regions on E9.5. Cart1 exhibited strong expression in the forebrain mesenchyme from E9.0 through E10.5. Although CBP, p300, Carm1 and Cited2 share temporal expression on E8.5, these co-activators have different spatial expression in mesenchyme and/or the neuroepithelium. Nevertheless, co-localization to the dorsal neural folds on E8.5 suggests a functional role in elevation and/or fusion of the neural folds. Target genes, and pathways that promote cranial neural tube fusion that are activated by CBP/p300/Carm1/Cited2/Cart1-containing transcriptional complexes await elucidation.


Subject(s)
Facial Bones/embryology , Facial Bones/metabolism , Neural Tube/embryology , Neural Tube/metabolism , Skull/embryology , Skull/metabolism , p300-CBP Transcription Factors/genetics , Animals , Base Sequence , CREB-Binding Protein/genetics , DNA Primers/genetics , DNA-Binding Proteins/genetics , E1A-Associated p300 Protein/genetics , Female , Gene Expression Regulation, Developmental , Gestational Age , Homeodomain Proteins , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Inbred ICR , Pregnancy , Protein-Arginine N-Methyltransferases/genetics , Repressor Proteins/genetics , Trans-Activators/genetics , p300-CBP Transcription Factors/metabolism
4.
Reprod Toxicol ; 26(3-4): 203-9, 2008.
Article in English | MEDLINE | ID: mdl-18992323

ABSTRACT

Environmental tobacco smoke exposures have been linked to adverse health effects. Folate is essential for normal development, with deficiencies often causing fetal growth restriction. Mice lacking the folate binding protein-2 receptor (Folr2) exhibit increased susceptibility to teratogens. The purpose of the current study was to determine if the loss of Folr2 would increase sensitivity to cigarette smoke-induced effects on development. Pregnant Folr2(-/-), Folr2(+/+), and C57BL/6J mice were exposed to sidestream cigarette smoke during gestation. Exposure to sidestream smoke on gd 6-9 had no adverse effects on fetal outcomes. However, cigarette smoke exposure on gd 6-18.5 increased the number of fetal resorptions (Folr2(-/-) cohort) and decreased crown-rump length (Folr2(+/+) fetuses). These data confirm an association between sidestream smoke exposure and fetal growth restriction, but do not suggest that loss of Folr2 increased susceptibility to these effects.


Subject(s)
Carrier Proteins/physiology , Fetal Development , Receptors, Cell Surface/physiology , Tobacco Smoke Pollution/adverse effects , Animals , Congenital Abnormalities/etiology , Cotinine/blood , Female , Fetal Resorption/etiology , Folate Receptors, GPI-Anchored , Mice , Mice, Inbred C57BL , Pregnancy , Pregnancy Rate , Weight Gain
5.
Toxicology ; 246(2-3): 193-202, 2008 Apr 18.
Article in English | MEDLINE | ID: mdl-18316152

ABSTRACT

Maternal/fetal genetic constitution and environmental factors are vital to delivery of a healthy baby. In the United States (US), a low birth weight (LBW) baby is born every minute and a half. LBW, defined as weighing less than 5.5 lbs at birth, affects nearly 1 in 12 infants born in the US with resultant costs for the nation of more than 15 billion dollars annually. Infant birth weight is the single most important factor affecting neonatal mortality. Various environmental and genetic risk factors for LBW have been identified. Several risks are preventable, such as cigarette smoking during pregnancy. Over one million babies are exposed prenatally to cigarette smoke accounting for over 20% of the LBW incidence in the US. Cigarette smoke exposure in utero results in a variety of adverse developmental outcomes with intrauterine growth restriction and infant LBW being the most well documented. However, the mechanisms underlying the causes of LBW remain poorly understood. The purpose of this study was: (1) to establish an animal model of cigarette smoke-induced in utero growth retardation and LBW using physiologically relevant inhalation exposure conditions which simulate "active" and "passive" tobacco smoke exposures, and (2) to determine whether particular stages of development are more susceptible than others to the adverse effects of in utero smoke exposure on embryo/fetal growth. Pregnant C57BL/6J mice were exposed to cigarette smoke during three periods of gestation: pre-/peri-implantation (gestational days [gds] 1-5), post-implantation (gds 6-18), and throughout gestation (gds 1-17). Reproductive and fetal outcomes were assessed on gd 18.5. Exposure of dams to mainstream/sidestream cigarette smoke, simulating "active" maternal smoking, resulted in decreases in fetal weight and crown-rump length when exposed throughout gestation (gds 1-17). Similar results were seen when dams were exposed only during the first 5 days of gestation (pre-/peri-implantation period gds 1-5). Exposure of dams from the post-implantation period through gestation (gds 6-18) did not result in reduced fetal weight, although a significant reduction in crown-rump length remained evident. Interestingly, maternal sidestream smoke exposure, simulating exposure to environmental tobacco smoke (ETS), during the pre-/peri-implantation period of development also produced significant decreases in fetal weight and crown-rump length. Collectively, results from the present study confirm an association between prenatal exposure to either "active" or "passive" cigarette smoke and in utero growth retardation. The data also identify a period of susceptibility to in utero cigarette smoke exposure-induced growth retardation and LBW during pre-/peri-implantation embryonic development.


Subject(s)
Disease Models, Animal , Embryonic Development/drug effects , Fetal Development/drug effects , Fetal Growth Retardation/chemically induced , Fetal Weight/drug effects , Smoke/adverse effects , Animals , Crown-Rump Length , Female , Gestational Age , Inhalation Exposure , Maternal Exposure , Mice , Mice, Inbred C57BL , Pregnancy , Time Factors
6.
Biochim Biophys Acta ; 1773(6): 814-20, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17467076

ABSTRACT

TGFbeta signaling regulates central cellular processes such as proliferation and extracellular matrix production during development of the orofacial region. Extracellular TGFbeta binds to cell surface receptors to activate the nucleocytoplasmic Smad proteins that, along with other transcription factors and cofactors, bind specific DNA sequences in the promoters of target genes to regulate their expression. To determine the identity of Smad binding proteins that regulate TGFbeta signaling in developing murine orofacial tissue, a yeast two-hybrid screening approach was employed. The PR-domain containing protein, PRDM16/MEL1 was identified as a novel Smad binding protein. The interaction between PRDM16/MEL1 and Smad 3 was confirmed by GST pull-down assays. The expression of PRDM16/MEL1 was detected in developing orofacial tissue by both Northern blot and in situ hybridization. PRDM16/MEL1 was constitutively expressed in orofacial tissue on E12.5-E14.5 as well as other embryonic tissues such as heart, brain, liver, and limb buds. Taken together, these results demonstrate that PRDM16/MEL1 is a Smad binding protein that may be important for development of orofacial structures through modulation of the TGFbeta signaling pathway.


Subject(s)
DNA-Binding Proteins/biosynthesis , Mouth/embryology , Organogenesis/physiology , Signal Transduction/physiology , Smad3 Protein/metabolism , Transcription Factors/biosynthesis , Transforming Growth Factor beta/metabolism , Animals , DNA-Binding Proteins/genetics , Mice , Mice, Inbred ICR , Organ Specificity , Protein Binding/physiology , Smad3 Protein/genetics , Transcription Factors/genetics
7.
Exp Biol Med (Maywood) ; 230(6): 394-406, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15956769

ABSTRACT

There are multiple mechanisms by which alcohol can damage the developing brain, but the type of damage induced will depend on the amount and developmental timing of exposure, along with other maternal and genetic factors. This article reviews current perspectives on how ethanol can produce neuroteratogenic effects by its interactions with molecular regulators of brain development. The current evidence suggests that alcohol produces many of its damaging effects by exerting specific actions on molecules that regulate key developmental processes (e.g., L1 cell adhesion molecule, alcohol dehydrogenase, catalase), interfering with the early development of midline serotonergic neurons and disrupting their regulatory-signaling function for other target brain structures, interfering with trophic factors that regulate neurogenesis and cell survival, or inducing excessive cell death via oxidative stress or activation of caspase-3 proteases. The current understanding of pathogenesis mechanisms suggests several strategic approaches to develop rational molecular prevention. However, the development of behavioral and biologic treatments for alcohol-affected children is crucial because it is unlikely that effective delivery of preventative interventions can realistically be achieved in ways to prevent prenatal damage in at-risk pregnancies. Toward that end, behavioral training that promotes experience-dependent neuroplasticity has been effective in a rat model of cerebellar damage induced by alcohol exposure during the period of brain development that is comparable to that of the human third trimester.


Subject(s)
Brain Damage, Chronic/etiology , Ethanol/toxicity , Prenatal Exposure Delayed Effects , Teratogens/toxicity , Alcohol-Induced Disorders, Nervous System/drug therapy , Alcohol-Induced Disorders, Nervous System/etiology , Alcohol-Induced Disorders, Nervous System/prevention & control , Animals , Antioxidants/therapeutic use , Brain Damage, Chronic/drug therapy , Brain Damage, Chronic/prevention & control , Ethanol/antagonists & inhibitors , Female , Humans , Neuronal Plasticity , Neuroprotective Agents/therapeutic use , Pregnancy , Serotonin 5-HT1 Receptor Agonists
8.
Alcohol Clin Exp Res ; 29(1): 117-29, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15654300

ABSTRACT

BACKGROUND: Rodent studies have shown that heavy binge-like ethanol (EtOH) exposure during the brain growth spurt [postnatal days (PD) 4-9] causes cerebellar neuronal loss and deficits in cerebellar-mediated eyeblink classical conditioning (ECC). Oxidative stress has been implicated in EtOH-mediated brain damage, and studies using vitamin E have reported amelioration of EtOH-induced tissue damage, including protection in rats against EtOH-induced cerebellar Purkinje cell (PC) loss on PD 4 to 5. The purpose of this study was to determine whether dietary supplementation with vitamin E concurrent with binge EtOH exposure on PD 4 to 9 in rats would attenuate the cerebellar cell death and ECC deficits. METHODS: Rat pups were given one of five different neonatal treatments: (1) intubation with EtOH in milk formula (twice daily, total dose 5.25 g/kg/day), (2) intubation with EtOH in milk formula supplemented with vitamin E (12.26 mg/kg/feeding), (3) intubation with milk formula that contained vitamin E only, (4) sham intubations, or (5) normally reared unintubated controls. Between PD 26 and 33, subjects received short-delay ECC for 3 consecutive days. Unbiased stereological cell counts were performed on cerebellar PCs of left cerebellar lobules I to VI and neurons of the interpositus nucleus. In a separate study with PD 4 pups, the effects of vitamin E on EtOH-induced expression of caspase-3 active subunits were assessed using Western blot analysis. RESULTS: EtOH-treated groups showed significant deficits in acquisition of conditioned eyeblink responses and reductions in cerebellar PCs and interpositus nucleus neurons compared with controls. Vitamin E supplementation failed to protect against these deficits. Vitamin E also failed to protect against increases in caspase-3 active subunit expression induced by acute binge EtOH exposure on PD 4. CONCLUSIONS: In contrast to the previously reported neuroprotective potential of antioxidants on EtOH-mediated cerebellar damage, vitamin E supplementation did not diminish EtOH-induced structural and functional damage to the cerebellum in this model of binge EtOH exposure during the brain growth spurt in rats.


Subject(s)
Cerebellum/drug effects , Cerebellum/pathology , Conditioning, Eyelid/drug effects , Ethanol/toxicity , Neuroprotective Agents/pharmacology , Prenatal Exposure Delayed Effects , Vitamin E/pharmacology , Animals , Animals, Newborn , Cell Count , Cerebellum/metabolism , Conditioning, Eyelid/physiology , Female , Male , Pregnancy , Rats , Rats, Long-Evans
SELECTION OF CITATIONS
SEARCH DETAIL
...