Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Alcohol Clin Exp Res ; 41(1): 117-127, 2017 01.
Article in English | MEDLINE | ID: mdl-27987329

ABSTRACT

BACKGROUND: Prenatal alcohol exposure (PAE) can result in physical and neurocognitive deficits that are collectively termed "fetal alcohol spectrum disorders" (FASD). Although FASD is associated with lifelong intellectual disability, the mechanisms mediating the emergence of secondary mental health and physical disabilities are poorly understood. Based on our previous data showing that maternal ethanol (EtOH) exposure in mice resulted in an immediate reduction in cranially directed fetal blood flow, we hypothesized that such exposure would also result in persistent alterations in cranially directed blood flow in the prenatally alcohol-exposed (PAE) adult. We also hypothesized that PAE adults exposed to an acute cerebrovascular insult would exhibit more brain damage and neurobehavioral impairment compared to non-PAE adult controls. METHODS: Pregnant C57BL/6 mice were exposed to EtOH, 3 g/kg, or water by intragastric gavage. Blood flow in carotid, renal, and femoral arteries was assessed by ultrasound imaging in PAE and control adults at 3, 6, and 12 months of age. To mimic ischemic stroke in young adult populations, 3-month-old PAE and control animals were subject to transient middle cerebral artery occlusion (MCAo) and subsequently assessed for behavioral recovery, stroke infarct volume, and brain cytokine profiles. RESULTS: PAE resulted in a significant age-related decrease in blood acceleration in adult mice, specifically in the carotid artery. A unilateral transient MCAo resulted in equivalent cortico-striatal damage in both PAE and control adults. However, PAE adult mice exhibited significantly decreased poststroke behavioral recovery compared to controls. CONCLUSIONS: Our data collectively show that PAE adult mice exhibit a persistent, long-term loss of cranially directed blood flow, and decreased capacity to compensate for brain trauma due to acute-onset adult diseases like ischemic stroke.


Subject(s)
Blood Flow Velocity/physiology , Cerebrovascular Circulation/physiology , Ethanol/toxicity , Ischemic Attack, Transient/physiopathology , Prenatal Exposure Delayed Effects/physiopathology , Animals , Blood Flow Velocity/drug effects , Cerebrovascular Circulation/drug effects , Ethanol/administration & dosage , Female , Ischemic Attack, Transient/chemically induced , Ischemic Attack, Transient/diagnostic imaging , Male , Mice , Mice, Inbred C57BL , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/diagnostic imaging
2.
Alcohol Res ; 35(1): 18-24, 2013.
Article in English | MEDLINE | ID: mdl-24313161

ABSTRACT

MicroRNAs (miRNAs) are members of a large class of non-protein-coding RNA (ncRNA) molecules that represent a significant, but until recently unappreciated, layer of cellular regulation. Assessment of the generation and function of miRNAs suggests that these ncRNAs are vulnerable to interference from genetic, epigenetic, and environmental factors. A small but rapidly expanding body of studies using a variety of animal- and cell culture-based experimental models also has shown that miRNAs are important targets of alcohol during fetal development and that their dysregulation likely plays a significant role in the etiology of fetal alcohol spectrum disorders (FASD). Accordingly, an analysis of the regulation and function of these miRNAs may yield important clues to the management of FASD.


Subject(s)
Central Nervous System Depressants/adverse effects , Ethanol/adverse effects , Fetal Alcohol Spectrum Disorders/genetics , Gene Expression Regulation/drug effects , MicroRNAs , Female , Gene Expression , Humans , Pregnancy
3.
PLoS One ; 8(7): e69560, 2013.
Article in English | MEDLINE | ID: mdl-23894503

ABSTRACT

BACKGROUND: Ethanol is a potent teratogen. Its adverse neural effects are partly mediated by disrupting fetal neurogenesis. The teratogenic process is poorly understood, and vulnerable neurogenic stages have not been identified. Identifying these is a prerequisite for therapeutic interventions to mitigate effects of teratogen exposures. METHODS: We used flow cytometry and qRT-PCR to screen fetal mouse-derived neurosphere cultures for ethanol-sensitive neural stem cell (NSC) subpopulations, to study NSC renewal and differentiation. The identity of vulnerable NSC populations was validated in vivo, using a maternal ethanol exposure model. Finally, the effect of ethanol exposure on the ability of vulnerable NSC subpopulations to integrate into the fetal neurogenic environment was assessed following ultrasound guided, adoptive transfer. RESULTS: Ethanol decreased NSC mRNAs for c-kit, Musashi-1and GFAP. The CD24(+) NSC population, specifically the CD24(+)CD15(+) double-positive subpopulation, was selectively decreased by ethanol. Maternal ethanol exposure also resulted in decreased fetal forebrain CD24 expression. Ethanol pre-exposed CD24(+) cells exhibited increased proliferation, and deficits in cell-autonomous and cue-directed neuronal differentiation, and following orthotopic transplantation into naïve fetuses, were unable to integrate into neurogenic niches. CD24(depleted) cells retained neurosphere regeneration capacity, but following ethanol exposure, generated increased numbers of CD24(+) cells relative to controls. CONCLUSIONS: Neuronal lineage committed CD24(+) cells exhibit specific vulnerability, and ethanol exposure persistently impairs this population's cell-autonomous differentiation capacity. CD24(+) cells may additionally serve as quorum sensors within neurogenic niches; their loss, leading to compensatory NSC activation, perhaps depleting renewal capacity. These data collectively advance a mechanistic hypothesis for teratogenesis leading to microencephaly.


Subject(s)
CD24 Antigen/genetics , Ethanol/toxicity , Fetus/cytology , Gene Expression Regulation, Developmental/drug effects , Neural Stem Cells/drug effects , Neural Stem Cells/transplantation , Stem Cell Transplantation , Animals , Biomarkers/metabolism , CD24 Antigen/metabolism , Cell Count , Cell Differentiation/drug effects , Cell Lineage/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Lewis X Antigen/metabolism , Mice , Mice, Inbred C57BL , Neural Stem Cells/cytology , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Teratogenesis/drug effects , Teratogens/toxicity
4.
Alcohol ; 47(2): 109-20, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23317542

ABSTRACT

Identification of the transcriptional networks disrupted by prenatal ethanol exposure remains a core requirement to better understanding the molecular mechanisms of alcohol-induced teratogenesis. In this regard, quantitative reverse-transcriptase polymerase chain reaction (qPCR) has emerged as an essential technique in our efforts to characterize alterations in gene expression brought on by exposure to alcohol. However, many publications continue to report the utilization of inappropriate methods of qPCR normalization, and for many in vitro models, no consistent set of empirically tested normalization controls have been identified. In the present study, we sought to identify a group of candidate reference genes for use within studies of alcohol exposed embryonic, placental, and neurosphere stem cells under both conditions maintaining stemness as well as throughout in vitro differentiation. To this end, we surveyed the recent literature and compiled a short list of fourteen candidate genes commonly used as normalization controls in qPCR studies of gene expression. This list included: Actb, B2m, Gapdh, Gusb, H2afz, Hk2, Hmbs, Hprt, Mrpl1, Pgk1, Ppia, Sdha, Tbp, and Ywhaz. From these studies, we find no single candidate gene was consistently refractory to the influence of alcohol nor completely stable throughout in vitro differentiation. Accordingly, we propose normalizing qPCR measurements to the geometric mean C(T) values obtained for three independent reference mRNAs as a reliable method to accurately interpret qPCR data and assess alterations in gene expression within alcohol treated cultures. Highlighting the importance of careful and empirical reference gene selection, the commonly used reference gene Actb was often amongst the least stable candidate genes tested. In fact, it would not serve as a valid normalization control in many cases. Data presented here will aid in the design of future experiments using stem cells to study the transcriptional processes driving differentiation, and model the developmental impact of teratogens.


Subject(s)
Embryonic Stem Cells/drug effects , Ethanol/adverse effects , Gene Expression/drug effects , Neural Stem Cells/drug effects , Placenta/cytology , Reverse Transcriptase Polymerase Chain Reaction , Abnormalities, Drug-Induced , Animals , Cell Differentiation/genetics , Cells, Cultured , Female , Genetic Markers/drug effects , Maternal-Fetal Exchange , Mice , Mice, Inbred C57BL , Pregnancy , RNA, Messenger/analysis , Stem Cells/drug effects
5.
Alcohol Clin Exp Res ; 36(5): 748-58, 2012 May.
Article in English | MEDLINE | ID: mdl-22141380

ABSTRACT

BACKGROUND: Ethanol (EtOH) consumption during pregnancy can lead to fetal growth retardation, mental retardation, and neurodevelopmental delay. The fetal brain initiates neurogenesis and vasculogenesis during the second trimester, and depends on maternal-fetal circulation for nutrition and growth signals. We used high-resolution in vivo ultrasound imaging to test the hypothesis that EtOH interferes with fetal brain-directed blood flow during this critical developmental period. METHODS: Pregnant mice were lightly anesthetized on gestational day 12 with an isoflurane/oxygen mixture. We assessed the effect of single and repeated binge-like maternal EtOH exposures at 3 g/kg, administered by intragastric gavage or intraperitoneal injection, on maternal circulation and fetal umbilical, aortic, internal carotid, and middle cerebral arterial circulation. RESULTS: Binge maternal EtOH exposure, regardless of exposure route, significantly reduced fetal arterial blood acceleration and velocity time integral (VTI), from umbilical to cerebral arteries, without a change in fetal heart rate and resistivity indices. Importantly a single maternal binge EtOH exposure induced persistent suppression of fetal arterial VTI for at least 24 hours. Repeated binge episodes resulted in a continuing and persistent suppression of fetal VTI. Qualitative assessments showed that maternal EtOH exposure induced oscillatory, nondirectional blood flow in fetal cerebral arteries. Maternal cardiac and other physiological parameters remained unaltered. CONCLUSIONS: These data show that binge-type maternal EtOH exposure results in rapid and persistent loss of blood flow from the umbilical artery to the fetal brain, potentially compromising nutrition and the maternal/fetal endocrine environment during a critical period for neuron formation and angiogenesis in the maturing brain.


Subject(s)
Central Nervous System Depressants/adverse effects , Cerebrovascular Circulation/drug effects , Ethanol/adverse effects , Fetal Alcohol Spectrum Disorders/etiology , Fetus/drug effects , Adaptation, Physiological , Alcohol Drinking/adverse effects , Animals , Central Nervous System Depressants/administration & dosage , Central Nervous System Depressants/blood , Ethanol/administration & dosage , Ethanol/blood , Female , Fetus/blood supply , Head/blood supply , Hemodynamics/drug effects , Injections, Intraperitoneal , Maternal Exposure/adverse effects , Mice , Pregnancy , Pregnancy Trimester, Second , Respiration/drug effects , Ultrasonography, Prenatal
6.
BMC Neurosci ; 6: 59, 2005 Sep 13.
Article in English | MEDLINE | ID: mdl-16159388

ABSTRACT

BACKGROUND: The fetal cortical neuroepithelium is a mosaic of distinct progenitor populations that elaborate diverse cellular fates. Ethanol induces apoptosis and interferes with the survival of differentiating neurons. However, we know little about ethanol's effects on neuronal progenitors. We therefore exposed neurosphere cultures from fetal rat cerebral cortex, to varying ethanol concentrations, to examine the impact of ethanol on stem cell fate. RESULTS: Ethanol promoted cell cycle progression, increased neurosphere number and increased diversity in neurosphere size, without inducing apoptosis. Unlike controls, dissociated cortical progenitors exposed to ethanol exhibited morphological evidence for asymmetric cell division, and cells derived from ethanol pre-treated neurospheres exhibited decreased proliferation capacity. Ethanol significantly reduced the numbers of cells expressing the stem cell markers CD117, CD133, Sca-1 and ABCG2, without decreasing nestin expression. Furthermore, ethanol-induced neurosphere proliferation was not accompanied by a commensurate increase in telomerase activity. Finally, cells derived from ethanol-pretreated neurospheres exhibited decreased differentiation in response to retinoic acid. CONCLUSION: The reduction in stem cell number along with a transient ethanol-driven increase in cell proliferation, suggests that ethanol promotes stem to blast cell maturation, ultimately depleting the reserve proliferation capacity of neuroepithelial cells. However, the lack of a concomitant change in telomerase activity suggests that neuroepithelial maturation is accompanied by an increased potential for genomic instability. Finally, the cellular phenotype that emerges from ethanol pre-treated, stem cell depleted neurospheres is refractory to additional differentiation stimuli, suggesting that ethanol exposure ablates or delays subsequent neuronal differentiation.


Subject(s)
Cell Cycle/drug effects , Cell Differentiation/drug effects , Ethanol/pharmacology , Nerve Regeneration/drug effects , Neuroepithelial Cells/drug effects , Stem Cells/drug effects , Animals , Cell Cycle/physiology , Cell Differentiation/physiology , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cerebral Cortex/physiology , Female , Fetus , Nerve Regeneration/physiology , Neuroepithelial Cells/cytology , Neuroepithelial Cells/physiology , Pregnancy , Rats , Rats, Sprague-Dawley , Stem Cells/cytology , Stem Cells/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...