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.
Ear Hear ; 45(1): 198-206, 2024.
Article in English | MEDLINE | ID: mdl-37563758

ABSTRACT

OBJECTIVES: Congenital cytomegalovirus (cCMV) is the most common cause of nongenetic sensorineural hearing loss (SNHL) in children. We examined the longitudinal hearing outcomes of children with cCMV in relation to their newborn hearing screening findings, and their use of antiviral therapy. DESIGN: The study was based on a retrospective chart review using a database of pediatric patients (N = 445) seen at the University of Minnesota Lions clinic. Chart review identified infants with cCMV, and records were reviewed for information about universal newborn hearing screen (UNHS) results, the clinical course of SNHL, and the use of antiviral therapy. RESULTS: A total of 44 children were identified with cCMV. In this group, 33 (75%) had SNHL of varying degree and age at onset. Notably, 17 (39%) children passed UNHS bilaterally. Of those children, 6 (35%) ultimately acquired bilateral or unilateral SNHL, detected at a mean age of 20 months (median age, 12 months). Five out of 10 children (50%) that did not pass UNHS in one ear acquired late-onset hearing loss in the contralateral ear, identified at a mean age of 24 months (median age, 4 months). Eleven (25%) children passed UNHS bilaterally and continued to demonstrate normal hearing in both ears at their most recent follow-up visit at a mean age of 19 months (SD, 18 months). Of the 33 children with cCMV and SNHL, 18 (55%) received antiviral medication (ganciclovir and/or valganciclovir). While, on average, both treated and untreated ears experienced a progression of hearing loss over time, the group that received antiviral treatment experienced less overall hearing change compared with the untreated group (baseline-adjusted expected mean difference, -10.5 dB; 95% confidence interval, -28.1 to 7.2 dB). CONCLUSIONS: Among children with cCMV included in this study who passed UNHS in both ears, 35% demonstrated delayed-onset SNHL. Notably, of those children who referred unilaterally, 50% later demonstrated SNHL in the contralateral ear. These findings have implications for audiological monitoring, and potentially antiviral therapy, of children with cCMV. As implementation of universal cCMV screening moves forward, a key aspect of follow-up will be appropriate long-term audiologic monitoring.


Subject(s)
Cytomegalovirus Infections , Deafness , Hearing Loss, Sensorineural , Infant , Infant, Newborn , Humans , Child , Child, Preschool , Cytomegalovirus , Retrospective Studies , Hearing Loss, Sensorineural/diagnosis , Hearing , Cytomegalovirus Infections/complications , Cytomegalovirus Infections/drug therapy , Cytomegalovirus Infections/congenital , Deafness/complications , Antiviral Agents/therapeutic use , Neonatal Screening/methods
2.
Front Mol Neurosci ; 15: 1032302, 2022.
Article in English | MEDLINE | ID: mdl-36523606

ABSTRACT

2,3,7,8-tetrachlorodibenzo-[p]-dioxin (TCDD) is a persistent global pollutant that exhibits a high affinity for the aryl hydrocarbon receptor (AHR), a ligand activated transcription factor. Epidemiological studies have associated AHR agonist exposure with multiple human neuropathologies. Consistent with the human data, research studies using laboratory models have linked pollutant-induced AHR activation to disruptions in learning and memory as well as motor impairments. Our understanding of endogenous AHR functions in brain development is limited and, correspondingly, scientists are still determining which cell types and brain regions are sensitive to AHR modulation. To identify novel phenotypes resulting from pollutant-induced AHR activation and ahr2 loss of function, we utilized the optically transparent zebrafish model. Early embryonic TCDD exposure impaired embryonic brain morphogenesis, resulted in ventriculomegaly, and disrupted neural connectivity in the optic tectum, habenula, cerebellum, and olfactory bulb. Altered neural network formation was accompanied by reduced expression of synaptic vesicle 2. Loss of ahr2 function also impaired nascent network development, but did not affect gross brain or ventricular morphology. To determine whether neural AHR activation was sufficient to disrupt connectivity, we used the Gal4/UAS system to express a constitutively active AHR specifically in differentiated neurons and observed disruptions only in the cerebellum; thus, suggesting that the phenotypes resulting from global AHR activation likely involve multiple cell types. Consistent with this hypothesis, we found that TCDD exposure reduced the number of oligodendrocyte precursor cells and their derivatives. Together, our findings indicate that proper modulation of AHR signaling is necessary for the growth and maturation of the embryonic zebrafish brain.

3.
Aquat Toxicol ; 234: 105786, 2021 May.
Article in English | MEDLINE | ID: mdl-33735685

ABSTRACT

The aryl hydrocarbon receptor (AHR) has endogenous functions in mammalian vascular development and is necessary for mediating the toxic effects of a number of environmental contaminants. Studies in mice have demonstrated that AHR is necessary for the formation of the renal, retinal, and hepatic vasculature. In fish, exposure to the prototypic AHR agonist 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) induces expression of the AHR biomarker cyp1a throughout the developing vasculature and produces vascular malformations in the head and heart. However, it is not known whether the vascular structures that are sensitive to loss of AHR function are also disrupted by aberrant AHR activation. Here, we report that TCDD-exposure in zebrafish disrupts development of 1) the subintestinal venous plexus (SIVP), which vascularizes the developing liver, kidney, gut, and pancreas, and 2) the superficial annular vessel (SAV), an essential component of the retinal vasculature. Furthermore, we determined that TCDD exposure increased the expression of bmp4, a key molecular mediator of SIVP morphogenesis. We hypothesize that the observed SIVP phenotypes contribute to one of the hallmarks of TCDD exposure in fish - the failure of the yolk sac to absorb. Together, our data describe novel TCDD-induced vascular phenotypes and provide molecular insight into critical factors producing the observed vascular malformations.


Subject(s)
Polychlorinated Dibenzodioxins/toxicity , Retinal Vein/drug effects , Water Pollutants, Chemical/toxicity , Zebrafish/metabolism , Animals , Animals, Genetically Modified/metabolism , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein 4/metabolism , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Liver/blood supply , Retinal Vein/growth & development , Veins/drug effects , Zebrafish/growth & development , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
4.
Sci Rep ; 8(1): 13906, 2018 09 17.
Article in English | MEDLINE | ID: mdl-30224706

ABSTRACT

The high mobility group transcription factor SOX9 is expressed in stem cells, progenitor cells, and differentiated cell-types in developing and mature organs. Exposure to a variety of toxicants including dioxin, di(2-ethylhexyl) phthalate, 6:2 chlorinated polyfluorinated ether sulfonate, and chlorpyrifos results in the downregulation of tetrapod Sox9 and/or zebrafish sox9b. Disruption of Sox9/sox9b function through environmental exposures or genetic mutations produce a wide range of phenotypes and adversely affect organ development and health. We generated a dominant-negative sox9b (dnsox9b) to inhibit sox9b target gene expression and used the Gal4/UAS system to drive dnsox9b specifically in cardiomyocytes. Cardiomyocyte-specific inhibition of sox9b function resulted in a decrease in ventricular cardiomyocytes, an increase in atrial cardiomyocytes, hypoplastic endothelial cushions, and impaired epicardial development, ultimately culminating in heart failure. Cardiomyocyte-specific dnsox9b expression significantly reduced end diastolic volume, which corresponded with a decrease in stroke volume, ejection fraction, and cardiac output. Further analysis of isolated cardiac tissue by RT-qPCR revealed cardiomyocyte-specific inhibition of sox9b function significantly decreased the expression of the critical cardiac development genes nkx2.5, nkx2.7, and myl7, as well as c-fos, an immediate early gene necessary for cardiomyocyte progenitor differentiation. Together our studies indicate sox9b transcriptional regulation is necessary for cardiomyocyte development and function.


Subject(s)
Heart/embryology , Morphogenesis , Myocytes, Cardiac/metabolism , SOX9 Transcription Factor/genetics , Animals , Gene Expression Regulation, Developmental , Genes, Dominant , HEK293 Cells , Humans , Mice , SOX9 Transcription Factor/metabolism , Transcription, Genetic , Zebrafish
5.
BMC Dev Biol ; 15: 50, 2015 Dec 29.
Article in English | MEDLINE | ID: mdl-26715205

ABSTRACT

BACKGROUND: The vertebrate heart consists of three cell layers: the innermost endothelium, the contractile myocardium and the outermost epicardium. The epicardium is vital for heart development and function, and forms from epicardial progenitor cells (EPCs), which migrate to the myocardium during early development. Disruptions in EPC migration and epicardium formation result in a number of cardiac malformations, many of which resemble congenital heart diseases in humans. Hence, it is important to understand the mechanisms that influence EPC migration and spreading in the developing heart. In vitro approaches heretofore have been limited to monolayer epicardial cell cultures, which may not fully capture the complex interactions that can occur between epicardial and myocardial cells in vivo. RESULTS: Here we describe a novel in vitro co-culture assay for assessing epicardial cell migration using embryonic zebrafish hearts. We isolated donor hearts from embryonic zebrafish carrying an epicardial-specific fluorescent reporter after epicardial cells were present on the heart. These were co-cultured with recipient hearts expressing a myocardial-specific fluorescent reporter, isolated prior to EPC migration. Using this method, we can clearly visualize the movement of epicardial cells from the donor heart onto the myocardium of the recipient heart. We demonstrate the utility of this method by showing that epicardial cell migration is significantly delayed or absent when myocardial cells lack contractility and when myocardial cells are deficient in tbx5 expression. CONCLUSIONS: We present a method to assess the migration of epicardial cells in an in vitro assay, wherein the migration of epicardial cells from a donor heart onto the myocardium of a recipient heart in co-culture is monitored and scored. The donor and recipient hearts can be independently manipulated, using either genetic tools or pharmacological agents. This allows flexibility in experimental design for determining the role that target genes/signaling pathways in specific cell types may have on epicardial cell migration.


Subject(s)
Cell Movement/physiology , Heart/embryology , Organogenesis/physiology , Pericardium/physiology , Zebrafish/embryology , Animals , Cell Proliferation , Coculture Techniques , Embryo, Nonmammalian/embryology , Heart Defects, Congenital/embryology , Heart Transplantation/methods , Myocardium/metabolism , Organ Culture Techniques , Pericardium/cytology , T-Box Domain Proteins/genetics
6.
Aquat Toxicol ; 162: 10-17, 2015 May.
Article in English | MEDLINE | ID: mdl-25766903

ABSTRACT

The swim bladder is a gas-filled organ that is used for regulating buoyancy and is essential for survival in most teleost species. In zebrafish, swim bladder development begins during embryogenesis and inflation occurs within 5 days post fertilization (dpf). Embryos exposed to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) before 96 h post fertilization (hpf) developed swim bladders normally until the growth/elongation phase, at which point growth was arrested. It is known that TCDD exposure causes heart malformations that lead to heart failure in zebrafish larvae, and that blood circulation is a key factor in normal development of the swim bladder. The adverse effects of TCDD exposure on the heart occur during the same period of time that swim bladder development and growth occurs. Based on this coincident timing, and the dependence of swim bladder development on proper circulatory development, we hypothesized that the adverse effects of TCDD on swim bladder development were secondary to heart failure. We compared swim bladder development in TCDD-exposed embryos to: (1) silent heart morphants, which lack cardiac contractility, and (2) transiently transgenic cmlc2:caAHR-2AtRFP embryos, which mimic TCDD-induced heart failure via heart-specific, constitutive activation of AHR signaling. Both of these treatment groups, which were not exposed to TCDD, developed hypoplastic swim bladders of comparable size and morphology to those found in TCDD-exposed embryos. Furthermore, in all treatment groups swim bladder development was arrested during the growth/elongation phase. Together, these findings support a potential role for heart failure in the inhibition of swim bladder development caused by TCDD.


Subject(s)
Air Sacs/drug effects , Heart Failure/chemically induced , Heart/drug effects , Organogenesis/drug effects , Polychlorinated Dibenzodioxins/toxicity , Water Pollutants, Chemical/toxicity , Zebrafish/embryology , Air Sacs/embryology , Animals , Embryo, Nonmammalian/drug effects , Heart/embryology , Heart Failure/embryology , Zebrafish/physiology
7.
BMC Dev Biol ; 14: 18, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24885804

ABSTRACT

BACKGROUND: The outermost layer of the vertebrate heart, the epicardium, forms from a cluster of progenitor cells termed the proepicardium (PE). PE cells migrate onto the myocardium to give rise to the epicardium. Impaired epicardial development has been associated with defects in valve development, cardiomyocyte proliferation and alignment, cardiac conduction system maturation and adult heart regeneration. Zebrafish are an excellent model for studying cardiac development and regeneration; however, little is known about how the zebrafish epicardium forms. RESULTS: We report that PE migration occurs through multiple mechanisms and that the zebrafish epicardium is composed of a heterogeneous population of cells. Heterogeneity is first observed within the PE and persists through epicardium formation. Using in vivo imaging, histology and confocal microscopy, we show that PE cells migrate through a cellular bridge that forms between the pericardial mesothelium and the heart. We also observed the formation of PE aggregates on the pericardial surface, which were released into the pericardial cavity. It was previously reported that heartbeat-induced pericardiac fluid advections are necessary for PE cluster formation and subsequent epicardium development. We manipulated heartbeat genetically and pharmacologically and found that PE clusters clearly form in the absence of heartbeat. However, when heartbeat was inhibited the PE failed to migrate to the myocardium and the epicardium did not form. We isolated and cultured hearts with only a few epicardial progenitor cells and found a complete epicardial layer formed. However, pharmacologically inhibiting contraction in culture prevented epicardium formation. Furthermore, we isolated control and silent heart (sih) morpholino (MO) injected hearts prior to epicardium formation (60 hpf) and co-cultured these hearts with "donor" hearts that had an epicardium forming (108 hpf). Epicardial cells from donor hearts migrated on to control but not sih MO injected hearts. CONCLUSIONS: Epicardial cells stem from a heterogeneous population of progenitors, suggesting that the progenitors in the PE have distinct identities. PE cells attach to the heart via a cellular bridge and free-floating cell clusters. Pericardiac fluid advections are not necessary for the development of the PE cluster, however heartbeat is required for epicardium formation. Epicardium formation can occur in culture without normal hydrodynamic and hemodynamic forces, but not without contraction.


Subject(s)
Cell Movement , Models, Biological , Pericardium/cytology , Stem Cells/cytology , Animals , Animals, Genetically Modified , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Heart Rate/physiology , Immunohistochemistry , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microscopy, Confocal , Organogenesis , Pericardium/embryology , Pericardium/physiology , Stem Cells/metabolism , Time Factors , Tissue Culture Techniques , Zebrafish/embryology , Zebrafish/metabolism , Zebrafish/physiology
8.
Rev Environ Contam Toxicol ; 222: 93-109, 2013.
Article in English | MEDLINE | ID: mdl-22990946

ABSTRACT

The insecticide methomyl, an oxime carbamate, was first introduced in 1968 for broad spectrum control of several insect classes, including Lepidoptera, Hemiptera, Homoptera, Diptera, and Coleoptera. Like other carbamates, it inhibits AChE activity, resulting in nerve and/or tissue failure and possibly death. Considered highly toxic to insects (larval and adult stages), methomyl is thought to be metabolically degraded via mixed-function oxidase(s). Methomyl has both a low vapor pressure and Henry's law constant; hence, volatilization is not a major dissipation route from either water or moist or dry soils. Photolysis represents a minor dissipation pathway; however, under catalytic conditions, degradation via photolysis does occur. Methomyl possesses a moderate-to-high water solubility; thus hydrolysis, under alkaline conditions, represents a major degradation pathway. Methomyl has a low-to-moderate sorption capacity to soil. Although results may vary with soil type and organic matter content, methomyl is unlikely to persist in complex soils. Methomyl is more rapidly degraded by microbes, and bacterial species have been identified that are capable of using methomyl as a carbon and/or nitrogen source. The main degradation products of methomyl from both abiotic and biotic processes are methomyl oxime, acetonitrile, and CO2. Methomyl is moderately to highly toxic to fishes and very highly toxic to aquatic invertebrates. Methomyl is highly toxic orally to birds and mammals. Methomyl is classed as being highly toxic to humans via oral exposures, moderately toxic via inhalation, and slightly toxic via dermal exposure. At relatively high doses, it can be fatal to humans. Although methomyl has been widely used to treat field crops and has high water solubility, it has only infrequently been detected as a contaminant of water bodies in the USA. It is classified as a restricted-use insecticide because of its toxicity to multiple nontarget species. To prevent nontarget species toxicity or the possibility of contamination, as with all pesticides, great care should be taken when applying methomyl-containing products for agricultural, residential, or other uses.


Subject(s)
Environmental Pollutants/toxicity , Insecticides/toxicity , Methomyl/toxicity , Animals , Environmental Pollutants/analysis , Insecticides/analysis , Methomyl/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...