Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
J Biomed Opt ; 28(7): 076002, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37469831

RESUMEN

Significance: Maternal exposure to drugs during pregnancy is known to have detrimental effects on the fetus. Alcohol (ethanol) and nicotine are two of the most commonly co-abused substances during pregnancy, and prenatal poly-drug exposure is common due, in part, to the prevalence of unplanned pregnancies. The second trimester is a critical period for fetal neurogenesis and angiogenesis. When drug exposure occurs during this time, fetal brain development is affected. Several behavioral, morphological, and functional studies have evaluated the changes in fetal brain development due to exposure to these drugs individually. However, research on the combined effects of ethanol and nicotine is far more limited, specifically on fetal vasculature changes and development. Aim: We use correlation mapping optical coherence angiography (cm-OCA) to evaluate acute changes in fetal brain vasculature caused by maternal exposure to a combination of ethanol and nicotine. Approach: Ethanol (16.6% v/v, at a dose of 0.75g/kg) and nicotine (at a dose of 0.1 mg/kg) were administered to pregnant mice after initial cm-OCA measurements in utero. Subsequent measurements were taken at 5-min intervals for a total period of 45 min. Results from these experiments were compared to results from our previous studies in which the mother was exposed to only ethanol (dose: 0.75 g/kg) or nicotine (dose: 0.1 mg/kg). Results: While results from exposure to ethanol or nicotine independently showed vasoconstriction, no significant change in vasculature was observed with combined exposure. Conclusion: Results suggested antagonistic effects of ethanol and nicotine on fetal brain vasculature.


Asunto(s)
Etanol , Nicotina , Animales , Femenino , Ratones , Embarazo , Angiografía , Encéfalo/diagnóstico por imagen , Encéfalo/irrigación sanguínea , Etanol/efectos adversos , Feto/diagnóstico por imagen , Feto/irrigación sanguínea , Nicotina/efectos adversos
2.
Aging Dis ; 12(6): 1516-1535, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34527425

RESUMEN

The developmental origins of health and disease (DOHaD) is a paradigm that links prenatal and early life exposures that occur during crucial periods of development to health outcome and risk of disease later in life. Maternal exposures to stress, some psychoactive drugs and alcohol, and environmental chemicals, among others, may result in functional changes in developing fetal tissues, creating a predisposition for disease in the individual as they age. Extracellular vesicles (EVs) may be mediators of both the immediate effects of exposure during development and early childhood as well as the long-term consequences of exposure that lead to increased risk and disease severity later in life. Given the prevalence of diseases with developmental origins, such as cardiovascular disease, neurodegenerative disorders, osteoporosis, metabolic dysfunction, and cancer, it is important to identify persistent mediators of disease risk. In this review, we take this approach, viewing diseases typically associated with aging in light of early life exposures and discuss the potential role of EVs as mediators of lasting consequences.

3.
Sci Rep ; 11(1): 1429, 2021 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-33446819

RESUMEN

Prenatal alcohol exposure (PAE) can result in cognitive and behavioral disabilities and growth deficits. Because alcohol-related neurobehavioral deficits may occur in the absence of overt dysmorphic features or growth deficits, there is a need to identify biomarkers of PAE that can predict neurobehavioral impairment. In this study, we assessed infant plasma extracellular, circulating miRNAs (exmiRNAs) obtained from a heavily exposed Cape Town cohort to determine whether these can be used to predict PAE-related growth restriction and cognitive impairment. PAE, controlling for smoking as a covariate, altered 27% of expressed exmiRNAs with clinically-relevant effect sizes (Cohen's d ≥ 0.4). Moreover, at 2 weeks, PAE increased correlated expression of exmiRNAs across chromosomes, suggesting potential co-regulation. In confirmatory factor analysis, the variance in expression for PAE-altered exmiRNAs at 2 weeks and 6.5 months was best described by three-factor models. Pathway analysis found that factors at 2 weeks were associated with (F1) cell maturation, cell cycle inhibition, and somatic growth, (F2) cell survival, apoptosis, cardiac development, and metabolism, and (F3) cell proliferation, skeletal development, hematopoiesis, and inflammation, and at 6.5 months with (F1) neurodevelopment, neural crest/mesoderm-derivative development and growth, (F2) immune system and inflammation, and (F3) somatic growth and cardiovascular development. Factors F3 at 2 weeks and F2 at 6.5 months partially mediated PAE-induced growth deficits, and factor F3 at 2 weeks partially mediated effects of PAE on infant recognition memory at 6.5 months. These findings indicate that infant exmiRNAs can help identify infants who will exhibit PAE-related deficits in growth and cognition.


Asunto(s)
MicroARN Circulante/sangre , Trastornos del Espectro Alcohólico Fetal/sangre , Adolescente , Adulto , Biomarcadores/sangre , Femenino , Humanos , Recién Nacido , Masculino
4.
Biomed Opt Express ; 11(7): 3618-3632, 2020 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-33014555

RESUMEN

Maternal smoking causes several defects ranging from intrauterine growth restriction to sudden infant death syndrome and spontaneous abortion. While several studies have documented the effects of prenatal nicotine exposure in development and behavior, acute vasculature changes in the fetal brain due to prenatal nicotine exposure have not been evaluated yet. This study uses correlation mapping optical coherence angiography to evaluate changes in fetal brain vasculature flow caused by maternal exposure to nicotine during the second trimester-equivalent of gestation in a mouse model. The effects of two different doses of nicotine were evaluated. Results showed a decrease in the vasculature for both doses of nicotine, which was not seen in the case of the sham group.

5.
Birth Defects Res ; 111(17): 1308-1319, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31356004

RESUMEN

Completion of the Human Genome Project has led to the identification of a large number of transcription start sites that are not paired with protein-coding genes, supporting the growing recognition of the abundance of encoded nonprotein-coding RNAs (ncRNAs) and their importance for speciation and species-specific development. Present in both plants and animals, ncRNAs vary in size, function, primary sequence, and secondary structure. While microRNAs (miRNAs) are the best known, there are a number of other ncRNAs (long[er] nonprotein-coding RNA, pseudogenes, circular RNAs, and so on) that have been shown to play an important role in the development either directly or via networks of proteins and other ncRNAs, including modulating the impact of miRNAs. Furthermore, these ncRNAs and their developmental regulatory networks are sensitive to teratogens such as ethanol, cannabis, cocaine, and nicotine. A better understanding of the developmental role of ncRNAs and their capacity to mediate teratogenesis is a necessary step in efforts to minimize the long-term consequences of developmental exposures to drugs-of-abuse. Moreover, with increasing awareness of the prevalence of polydrug use, experimental models will need to incorporate more complex drug exposure paradigms into meaningful assessments of developmental ncRNA function.


Asunto(s)
Anomalías Inducidas por Medicamentos/genética , Anomalías Congénitas/genética , Redes Reguladoras de Genes/efectos de los fármacos , ARN no Traducido/genética , Animales , Cannabis/efectos adversos , Cocaína/efectos adversos , Etanol/efectos adversos , Humanos , Nicotina/efectos adversos , Teratología/métodos
6.
Life Sci Alliance ; 2(2)2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30833415

RESUMEN

Prenatal alcohol exposure (PAE), like other pregnancy complications, can result in placental insufficiency and fetal growth restriction, although the linking causal mechanisms are unclear. We previously identified 11 gestationally elevated maternal circulating miRNAs (HEamiRNAs) that predicted infant growth deficits following PAE. Here, we investigated whether these HEamiRNAs contribute to the pathology of PAE, by inhibiting trophoblast epithelial-mesenchymal transition (EMT), a pathway critical for placental development. We now report for the first time that PAE inhibits expression of placental pro-EMT pathway members in both rodents and primates, and that HEamiRNAs collectively, but not individually, mediate placental EMT inhibition. HEamiRNAs collectively, but not individually, also inhibited cell proliferation and the EMT pathway in cultured trophoblasts, while inducing cell stress, and following trophoblast syncytialization, aberrant endocrine maturation. Moreover, a single intravascular administration of the pooled murine-expressed HEamiRNAs, to pregnant mice, decreased placental and fetal growth and inhibited the expression of pro-EMT transcripts in the placenta. Our data suggest that HEamiRNAs collectively interfere with placental development, contributing to the pathology of PAE, and perhaps also, to other causes of fetal growth restriction.


Asunto(s)
MicroARN Circulante/metabolismo , Etanol/efectos adversos , Trastornos del Espectro Alcohólico Fetal/metabolismo , Placentación/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal/metabolismo , Alcoholismo/complicaciones , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Etanol/administración & dosificación , Femenino , Trastornos del Espectro Alcohólico Fetal/etiología , Retardo del Crecimiento Fetal/etiología , Retardo del Crecimiento Fetal/metabolismo , Humanos , Macaca mulatta , Ratones , Ratones Endogámicos C57BL , Embarazo , Ratas , Ratas Sprague-Dawley , Trofoblastos/metabolismo
7.
J Neurotrauma ; 34(10): 1873-1890, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-27788626

RESUMEN

Spinal cord injury (SCI) is often accompanied by other tissue damage (polytrauma) that provides a source of pain (nociceptive) input. Recent findings are reviewed that show SCI places the caudal tissue in a vulnerable state that exaggerates the effects nociceptive stimuli and promotes the development of nociceptive sensitization. Stimulation that is both unpredictable and uncontrollable induces a form of maladaptive plasticity that enhances nociceptive sensitization and impairs spinally mediated learning. In contrast, relational learning induces a form of adaptive plasticity that counters these adverse effects. SCI sets the stage for nociceptive sensitization by disrupting serotonergic (5HT) fibers that quell overexcitation. The loss of 5HT can enhance neural excitability by reducing membrane-bound K+-Cl- cotransporter 2, a cotransporter that regulates the outward flow of Cl-. This increases the intracellular concentration of Cl-, which reduces the hyperpolarizing (inhibitory) effect of gamma-aminobutyric acid. Uncontrollable noxious stimulation also undermines the recovery of locomotor function, and increases behavioral signs of chronic pain, after a contusion injury. Nociceptive stimulation has a greater effect if experienced soon after SCI. This adverse effect has been linked to a downregulation in brain-derived neurotrophic factor and an upregulation in the cytokine, tumor necrosis factor. Noxious input enhances tissue loss at the site of injury by increasing the extent of hemorrhage and apoptotic/pyroptotic cell death. Intrathecal lidocaine blocks nociception-induced hemorrhage, cellular indices of cell death, and its adverse effect on behavioral recovery. Clinical implications are discussed.


Asunto(s)
Plasticidad Neuronal/fisiología , Dimensión del Dolor/métodos , Dolor/patología , Recuperación de la Función/fisiología , Traumatismos de la Médula Espinal/patología , Animales , Humanos , Dolor/etiología , Dolor/fisiopatología , Traumatismos de la Médula Espinal/complicaciones , Traumatismos de la Médula Espinal/fisiopatología
8.
PLoS One ; 11(11): e0165081, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27828986

RESUMEN

Fetal alcohol spectrum disorders (FASD) are difficult to diagnose since many heavily exposed infants, at risk for intellectual disability, do not exhibit craniofacial dysmorphology or growth deficits. Consequently, there is a need for biomarkers that predict disability. In both animal models and human studies, alcohol exposure during pregnancy resulted in significant alterations in circulating microRNAs (miRNAs) in maternal blood. In the current study, we asked if changes in plasma miRNAs in alcohol-exposed pregnant mothers, either alone or in conjunction with other clinical variables, could predict infant outcomes. Sixty-eight pregnant women at two perinatal care clinics in western Ukraine were recruited into the study. Detailed health and alcohol consumption histories, and 2nd and 3rd trimester blood samples were obtained. Birth cohort infants were assessed by a geneticist and classified as unexposed (UE), heavily prenatally exposed and affected (HEa) or heavily exposed but apparently unaffected (HEua). MiRNAs were assessed in plasma samples using qRT-PCR arrays. ANOVA models identified 11 miRNAs that were all significantly elevated in maternal plasma from the HEa group relative to HEua and UE groups. In a random forest analysis classification model, a combination of high variance miRNAs, smoking history and socioeconomic status classified membership in HEa and UE groups, with a misclassification rate of 13%. The RFA model also classified 17% of the HEua group as UE-like, whereas 83% were HEa-like, at least at one stage of pregnancy. Collectively our data indicate that maternal plasma miRNAs predict infant outcomes, and may be useful to classify difficult-to-diagnose FASD subpopulations.


Asunto(s)
Consumo de Bebidas Alcohólicas/efectos adversos , Trastornos del Espectro Alcohólico Fetal/genética , MicroARNs/genética , Efectos Tardíos de la Exposición Prenatal/genética , Adulto , Animales , Biomarcadores/sangre , Estudios de Cohortes , Femenino , Trastornos del Espectro Alcohólico Fetal/diagnóstico , Trastornos del Espectro Alcohólico Fetal/etiología , Humanos , Lactante , Recién Nacido , Masculino , MicroARNs/sangre , MicroARNs/clasificación , Atención Perinatal , Valor Predictivo de las Pruebas , Embarazo , Efectos Tardíos de la Exposición Prenatal/diagnóstico , Efectos Tardíos de la Exposición Prenatal/etiología , Pronóstico , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sensibilidad y Especificidad , Ucrania , Adulto Joven
9.
PLoS One ; 10(12): e0145061, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26661278

RESUMEN

Hereditary, hormonal, and behavioral factors contribute to the development of breast cancer. Alcohol consumption is a modifiable behavior that is linked to increased breast cancer risks and is associated with the development of hormone-dependent breast cancers as well as disease progression and recurrence following endocrine treatment. In this study we examined the molecular mechanisms of action of alcohol by applying molecular, genetic, and genomic approaches in characterizing its effects on estrogen receptor (ER)-positive breast cancer cells. Treatments with alcohol promoted cell proliferation, increased growth factor signaling, and up-regulated the transcription of the ER target gene GREB1 but not the canonical target TFF1/pS2. Microarray analysis following alcohol treatment identified a large number of alcohol-responsive genes, including those which function in apoptotic and cell proliferation pathways. Furthermore, expression profiles of the responsive gene sets in tumors were strongly associated with clinical outcomes in patients who received endocrine therapy. Correspondingly, alcohol treatment attenuated the anti-proliferative effects of the endocrine therapeutic drug tamoxifen in ER-positive breast cancer cells. To determine the contribution and functions of responsive genes, their differential expression in tumors were assessed between outcome groups. The proto-oncogene BRAF was identified as a novel alcohol- and estrogen-induced gene that showed higher expression in patients with poor outcomes. Knock-down of BRAF, moreover, prevented the proliferation of breast cancer cells. These findings not only highlight the mechanistic basis of the effects of alcohol on breast cancer cells and increased risks for disease incidents and recurrence, but may facilitate the discovery and characterization of novel oncogenic pathways and markers in breast cancer research and therapeutics.


Asunto(s)
Antineoplásicos Hormonales/toxicidad , Etanol/farmacología , Tamoxifeno/toxicidad , Antineoplásicos Hormonales/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Estradiol/toxicidad , Femenino , Humanos , Células MCF-7 , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , ARN Interferente Pequeño/metabolismo , Receptores de Estrógenos/metabolismo , Transducción de Señal/efectos de los fármacos , Tamoxifeno/uso terapéutico , Factor Trefoil-1 , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Regulación hacia Arriba/efectos de los fármacos
10.
Pain ; 155(11): 2344-59, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25180012

RESUMEN

We previously showed that peripheral noxious input after spinal cord injury (SCI) inhibits beneficial spinal plasticity and impairs recovery of locomotor and bladder functions. These observations suggest that noxious input may similarly affect the development and maintenance of chronic neuropathic pain, an important consequence of SCI. In adult rats with a moderate contusion SCI, we investigated the effect of noxious tail stimulation, administered 1 day after SCI on mechanical withdrawal responses to von Frey stimuli from 1 to 28 days after treatment. In addition, because the proinflammatory cytokine tumor necrosis factor alpha (TNFα) is implicated in numerous injury-induced processes including pain hypersensitivity, we assessed the temporal and spatial expression of TNFα, TNF receptors, and several downstream signaling targets after stimulation. Our results showed that unlike sham surgery or SCI only, nociceptive stimulation after SCI induced mechanical sensitivity by 24h. These behavioral changes were accompanied by increased expression of TNFα. Cellular assessments of downstream targets of TNFα revealed that nociceptive stimulation increased the expression of caspase 8 and the active subunit (12 kDa) of caspase 3, indicative of active apoptosis at a time point consistent with the onset of mechanical allodynia. In addition, immunohistochemical analysis revealed distinct morphological signs of apoptosis in neurons and microglia at 24h after stimulation. Interestingly, expression of the inflammatory mediator NFκB was unaltered by nociceptive stimulation. These results suggest that noxious input caudal to the level of SCI can increase the onset and expression of behavioral responses indicative of pain, potentially involving TNFα signaling.


Asunto(s)
Apoptosis/fisiología , Regulación de la Expresión Génica/fisiología , Hiperalgesia/etiología , Umbral del Dolor/fisiología , Traumatismos de la Médula Espinal/complicaciones , Factor de Necrosis Tumoral alfa/metabolismo , Análisis de Varianza , Animales , Caspasa 3/metabolismo , Modelos Animales de Enfermedad , Locomoción/fisiología , Masculino , Microglía/metabolismo , Microglía/patología , Neuronas/metabolismo , Neuronas/patología , Dimensión del Dolor , Ratas , Ratas Sprague-Dawley , Transducción de Señal/fisiología , Factores de Tiempo , Factor de Necrosis Tumoral alfa/genética
11.
PLoS One ; 8(7): e69560, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23894503

RESUMEN

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.


Asunto(s)
Antígeno CD24/genética , Etanol/toxicidad , Feto/citología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/trasplante , Trasplante de Células Madre , Animales , Biomarcadores/metabolismo , Antígeno CD24/metabolismo , Recuento de Células , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Femenino , Antígeno Lewis X/metabolismo , Ratones , Ratones Endogámicos C57BL , Células-Madre Neurales/citología , Embarazo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Teratogénesis/efectos de los fármacos , Teratógenos/toxicidad
12.
Alcohol Clin Exp Res ; 36(10): 1669-77, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22458409

RESUMEN

BACKGROUND: Ethanol (EtOH) and nicotine are often co-abused. However, their combined effects on fetal neural development, particularly on fetal neural stem cells (NSCs), which generate most neurons of the adult brain during the second trimester of pregnancy, are poorly understood. We previously showed that EtOH influenced NSC maturation in part, by suppressing the expression of specific microRNAs (miRNAs). Here, we tested in fetal NSCs the extent to which EtOH and nicotine coregulated known EtOH-sensitive (miR-9, miR-21, miR-153, and miR-335), a nicotine-sensitive miRNA (miR-140-3p), and mRNAs for nicotinic acetylcholine receptor (nAChR) subunits. Additionally, we tested the extent to which these effects were nAChR dependent. METHODS: Gestational day 12.5 mouse fetal murine cerebral cortical-derived neurosphere cultures were exposed to EtOH, nicotine, and mecamylamine, a noncompetitive nAChR antagonist, individually or in combination, for short (24 hour) and long (5 day) periods, to mimic exposure during the in vivo period of neurogenesis. Levels of miRNAs, miRNA-regulated transcripts, and nAChR subunit mRNAs were assessed by quantitative reverse transcription polymerase chain reaction. RESULTS: EtOH suppressed the expression of known EtOH-sensitive miRNAs and miR-140-3p, while nicotine at concentrations attained by cigarette smokers induced a dose-related increase in these miRNAs. Nicotine's effect was blocked by EtOH and by mecamylamine. Finally, EtOH decreased the expression of nAChR subunit mRNAs and, like mecamylamine, prevented the nicotine-associated increase in α4 and ß2 nAChR transcripts. CONCLUSIONS: EtOH and nicotine exert mutually antagonistic, nAChR-mediated effects on teratogen-sensitive miRNAs in fetal NSCs. These data suggest that concurrent exposure to EtOH and nicotine disrupts miRNA regulatory networks that are important for NSC maturation.


Asunto(s)
Corteza Cerebral/fisiología , Etanol/farmacología , Células Madre Fetales/fisiología , MicroARNs/fisiología , Células-Madre Neurales/fisiología , Nicotina/farmacología , Receptores Nicotínicos/fisiología , Animales , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Células Madre Fetales/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , MicroARNs/antagonistas & inhibidores , Células-Madre Neurales/efectos de los fármacos , Embarazo
13.
Alcohol Clin Exp Res ; 34(4): 575-87, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20102566

RESUMEN

Ethanol exerts complex effects on human physiology and health. Ethanol is not only addictive, but it is also a fetal teratogen, an adult neurotoxin, and an etiologic agent in hepatic and cardiovascular disease, inflammation, bone loss, and fracture susceptibility. A large number of genes and signaling mechanisms have been implicated in ethanol's deleterious effects leading to the suggestion that ethanol is a "dirty drug." An important question is, are there cellular "master-switches" that can explain these pleiotropic effects of ethanol? MicroRNAs (miRNAs) have been recently identified as master regulators of the cellular transcriptome and proteome. miRNAs play an increasingly appreciated and crucial role in shaping the differentiation and function of tissues and organs in both health and disease. This critical review discusses new evidence showing that ethanol-sensitive miRNAs are indeed regulatory master-switches. More specifically, miRNAs control the development of tolerance, a crucial component of ethanol addiction. Other drugs of abuse also target some ethanol-sensitive miRNAs suggesting that common biochemical mechanisms underlie addiction. This review also discusses evidence that miRNAs mediate several ethanol pathologies, including disruption of neural stem cell proliferation and differentiation in the exposed fetus, gut leakiness that contributes to endotoxemia and alcoholic liver disease, and possibly also hepatocellular carcinomas and other gastrointestinal cancers. Finally, this review provides a perspective on emerging investigations into potential roles of miRNAs as mediators of ethanol's effects on inflammation and fracture healing, as well as the potential for miRNAs as diagnostic biomarkers and as targets for therapeutic interventions for alcohol-related disorders.


Asunto(s)
Alcoholismo/genética , Alcoholismo/metabolismo , Etanol/metabolismo , Etanol/toxicidad , MicroARNs/fisiología , Alcoholismo/patología , Animales , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Humanos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
14.
Alcohol Clin Exp Res ; 31(4): 694-703, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17374049

RESUMEN

BACKGROUND: In utero exposure to ethanol can result in severe fetal brain defects. Previous studies showed that ethanol induces apoptosis in differentiated cortical neurons. However, we know little about ethanol's effects on proliferating embryonic cortical progenitors. This study investigated the impact of ethanol exposure on the Fas/Apo-1/CD95 suicide receptor pathway, and on the survival of proliferating cortical neuroepithelial progenitors. METHODS: Murine embryonic-derived primary cortical neuroepithelial cells were maintained as neurosphere cultures and exposed to a dose range of ethanol for periods ranging from 1 to 5 days. Programmed cell death was measured by 4 independent means (Annexin-V staining, caspase activation, DNA fragmentation, and autophagic vacuole formation). Surface Fas/Apo-1 suicide receptor expression was measured by flow cytometry. Expression of Fas/Apo-1-associated DISC-complex genes was measured by quantitative polymerase chain reaction. RESULTS: Ethanol exposure did not substantially increase apoptosis, necrosis, or surface Fas/Apo-1 expression. Moreover, ethanol significantly decreased caspase activation and autophagic activity. Finally, ethanol exposure induced mRNA expression of genes that constitute the death receptor complex. CONCLUSIONS: This study provides surprising evidence that ethanol does not induce either programmed cell death or necrosis of immature progenitors during neurogenesis, although ethanol may render neural progenitors susceptible to future apoptotic insults. Furthermore, our novel observation that ethanol suppresses autophagy is consistent with a hypothesis that ethanol promotes premature neural progenitor maturation. Taken together with our previous data regarding the role of the Fas/Apo-1 receptor in neural development, we conclude that ethanol disrupts basic proliferation and differentiation machinery rather than initiating cell death per se.


Asunto(s)
Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Depresores del Sistema Nervioso Central/toxicidad , Corteza Cerebral/citología , Corteza Cerebral/embriología , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte/genética , Etanol/toxicidad , Células Madre/efectos de los fármacos , Animales , Anexina A5/metabolismo , Caspasas/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Fragmentación del ADN , Activación Enzimática/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Células Epiteliales/efectos de los fármacos , Femenino , Citometría de Flujo , Fluorometría , Ratones , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Embarazo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Receptor fas/metabolismo
15.
Alcohol Clin Exp Res ; 31(2): 324-35, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17250626

RESUMEN

BACKGROUND: Heavy alcohol consumption during pregnancy can cause significant mental retardation and brain damage. We recently showed that ethanol depletes reserve cerebral cortical stem cell capacity. Moreover, proliferating neuroepithelial cells exposed to ethanol were resistant to subsequent retinoic acid-induced differentiation. Emerging evidence suggests that cytokines play a crucial growth-promoting role in the developing neural tube. METHODS: We cultured murine cortical neurosphere cultures in control or ethanol-supplemented mitogenic medium, to mimic alcohol exposure during the period of neuroepithelial proliferation. Cultures were then treated with a step-wise mitogen-withdrawal, integrin-activation model to mimic subsequent phases of neuronal migration and early differentiation. We examined the impact of alcohol exposure during neurogenesis on the secretion of inflammatory and growth-promoting cytokines. RESULTS: Cortical neurosphere cultures exhibit increasingly complex differentiation phenotypes in response to step-wise mitogen-withdrawal and laminin exposure. Some inflammation-modulating cytokines were secreted independent of differentiation state. However, chemotactic cytokines were specifically secreted at high levels, as a function of differentiation stage. monocyte chemotactic protein-1, vascular endothelial growth factor-A, and interleukin (IL)-10 were coordinately decreased during differentiation compared with neuroepithelial proliferation, while granulocyte macrophage-colony stimulating factor (GM-CSF) was induced during differentiation, compared with the neuroepithelial proliferation period. Ethanol exposure during the period of neuroepithelial proliferation prevented the early differentiation-induced increase in GM-CSF while inducing differentiation-associated increase in IL-12 secretion. CONCLUSION: Embryonic cerebral cortical neuroepithelial-derived precursors secrete high levels of several angiogenic and neural-growth-promoting cytokines as they differentiate into neurons. Our data collectively suggest that ethanol exposure during the period of neuroepithelial proliferation significantly disrupts cytokine signals that are required for the support of emerging neurovascular networks, and the maintenance of neural stem cell beds.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Depresores del Sistema Nervioso Central/toxicidad , Corteza Cerebral/citología , Citocinas/metabolismo , Etanol/toxicidad , Sistema Nervioso/embriología , Células Neuroepiteliales/citología , Proteínas Angiogénicas/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/efectos de los fármacos , Quimiocina CCL2/metabolismo , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Interleucina-10/metabolismo , Interleucina-12/metabolismo , Ratones , Ratones Endogámicos C57BL , Sistema Nervioso/efectos de los fármacos , Células Neuroepiteliales/efectos de los fármacos , Embarazo , Factor A de Crecimiento Endotelial Vascular/metabolismo
16.
Am J Med Genet C Semin Med Genet ; 135C(1): 48-58, 2005 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-15800851

RESUMEN

Folic acid is essential for many cellular reactions, including synthesis of nucleotides and regulation of cell cycle. Folic acid-binding protein one (Folbp1), a membrane-bounded protein, is the primary mediator of folic acid transport. Mice deficient in Folbp1 gene die in utero with multiple malformations, including severe exencephaly and craniofacial defects. Fusion of the neural tube and craniofacies require precisely regulated interactions of apoptosis, cell proliferation, and differentiation. To understand the role of Folbp1 in regulating the fusions of these primordia, levels of dead and proliferating precursor cells from Folbp1 embryos were quantified before the fusion processes. Massive apoptosis was detected in the Folbp1-/- defective tissues, with Bax and activated caspase-3 distributed evenly across the apico-basal axis of the lateral neural plate. 5-Bromodeoxyuridine (BrdU) and PCNA labeling assays revealed a reduced cell proliferation as well. However, telomerase activity was unaltered, arguing against telomere shortening and consequently, chromosomal instability, as the cause of the apoptosis. Notably, Islet-1 and 2H3 immunohistochemistry demonstrated the presence of differentiating neuronal cells, albeit in decreased numbers. Interestingly, Folbp1-/- embryos also elaborated novel neural structures that sprouted orthogonally from the embryonic neuraxis. Assays on the defective craniofacies exhibited similar phenomena, suggesting the neural crest precursor population that gives rise to both these structures is selectively vulnerable to Folbp1 inactivation. The results demonstrate a prominent role of Folbp1 in the regional regulation of apoptosis and cell proliferation that underlies the aberrant neural tube and craniofacial defects.


Asunto(s)
Apoptosis/fisiología , Proteínas Portadoras/fisiología , Receptores de Superficie Celular/fisiología , Animales , Bromodesoxiuridina/metabolismo , Caspasa 3 , Caspasas/metabolismo , Diferenciación Celular , Proliferación Celular , Inestabilidad Cromosómica , Anomalías Craneofaciales/etiología , Desarrollo Embrionario , Activación Enzimática , Femenino , Receptores de Folato Anclados a GPI , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas de Homeodominio/metabolismo , Proteínas con Homeodominio LIM , Masculino , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/metabolismo , Fenómenos Fisiológicos del Sistema Nervioso , Defectos del Tubo Neural/etiología , Defectos del Tubo Neural/genética , Defectos del Tubo Neural/patología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Receptores de Superficie Celular/deficiencia , Telomerasa/metabolismo , Factores de Transcripción , Proteína X Asociada a bcl-2
17.
BMC Neurosci ; 5: 11, 2004 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-15038834

RESUMEN

BACKGROUND: Apoptosis is important for normal cerebral cortical development. We previously showed that the Fas suicide receptor was expressed within the developing cerebral cortex, and that in vitro Fas activation resulted in caspase-dependent death. Alterations in cell-surface Fas expression may significantly influence cortical development. Therefore, in the following studies, we sought to identify developmentally relevant cell biological processes that regulate cell-surface Fas expression and reciprocal consequences of Fas receptor activation. RESULTS: Flow-cytometric analyses identified two distinct neural sub-populations that expressed Fas on their cell surface at high (FasHi) or moderate (FasMod) levels. The anti-apoptotic protein FLIP further delineated a subset of Fas-expressing cells with potential apoptosis-resistance. FasMod precursors were mainly in G0, while FasHi precursors were largely apoptotic. However, birth-date analysis indicated that neuroblasts express the highest levels of cell-surface Fas at the end of S-phase, or after their final round of mitosis, suggesting that Fas expression is induced at cell cycle checkpoints or during interkinetic nuclear movements. FasHi expression was associated with loss of cell-matrix adhesion and anoikis. Activation of the transcription factor p53 was associated with induction of Fas expression, while the gonadal hormone estrogen antagonistically suppressed cell-surface Fas expression. Estrogen also induced entry into S-phase and decreased the number of Fas-expressing neuroblasts that were apoptotic. Concurrent exposure to estrogen and to soluble Fas-ligand (sFasL) suppressed p21/waf-1 and PCNA. In contrast, estrogen and sFasL, individually and together, induced cyclin-A expression, suggesting activation of compensatory survival mechanisms. CONCLUSIONS: Embryonic cortical neuronal precursors are intrinsically heterogeneous with respect to Fas suicide-sensitivity. Competing intrinsic (p53, cell cycle, FLIP expression), proximal (extra-cellular matrix) and extrinsic factors (gonadal hormones) collectively regulate Fas suicide-sensitivity either during neurogenesis, or possibly during neuronal migration, and may ultimately determine which neuroblasts successfully contribute neurons to the differentiating cortical plate.


Asunto(s)
Estrógenos/fisiología , Matriz Extracelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Glicoproteínas de Membrana/fisiología , Células Madre/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Receptor fas/metabolismo , Animales , Anoicis/fisiología , Apoptosis/fisiología , Western Blotting , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD , Proteínas Portadoras/biosíntesis , Ciclo Celular/fisiología , Proteínas de Ciclo Celular/metabolismo , Núcleo Celular/metabolismo , Uniones Célula-Matriz/metabolismo , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/embriología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/metabolismo , Estrógenos/farmacología , Proteína Ligando Fas , Citometría de Flujo , Neuronas/clasificación , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Ratas , Ratas Sprague-Dawley , Células Madre/citología , Células Madre/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA