Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Sci Adv ; 9(16): eadg3200, 2023 04 21.
Article in English | MEDLINE | ID: mdl-37075109

ABSTRACT

Echinoderm mass mortality events shape marine ecosystems by altering the dynamics among major benthic groups. The sea urchin Diadema antillarum, virtually extirpated in the Caribbean in the early 1980s by an unknown cause, recently experienced another mass mortality beginning in January 2022. We investigated the cause of this mass mortality event through combined molecular biological and veterinary pathologic approaches comparing grossly normal and abnormal animals collected from 23 sites, representing locations that were either affected or unaffected at the time of sampling. Here, we report that a scuticociliate most similar to Philaster apodigitiformis was consistently associated with abnormal urchins at affected sites but was absent from unaffected sites. Experimentally challenging naïve urchins with a Philaster culture isolated from an abnormal, field-collected specimen resulted in gross signs consistent with those of the mortality event. The same ciliate was recovered from treated specimens postmortem, thus fulfilling Koch's postulates for this microorganism. We term this condition D. antillarum scuticociliatosis.


Subject(s)
Ecosystem , Sea Urchins , Animals , Caribbean Region
2.
Biomark Res ; 10(1): 26, 2022 Apr 25.
Article in English | MEDLINE | ID: mdl-35468853

ABSTRACT

INTRODUCTION: PD-L1 expression in non-small cell lung cancer (NSCLC) predicts response to immune checkpoint blockade, however is an imperfect biomarker given tumor heterogeneity, and the antigen presentation pathway requiring other components including HLA I expression. HLA I downregulation may contribute to resistance, warranting its evaluation in attempts to guide patient selection. In addition, earlier detection of acquired resistance could prompt earlier change in treatment and prolong patient survival. Analysis of circulating tumor cells (CTCs) captures heterogeneity across multiple sites of metastases, enables detection of changes in tumor burden that precede radiographic response, and can be obtained in serial fashion. METHODS: To quantify the expression of both PD-L1 and HLA I on CTCs, we developed exclusion-based sample preparation technology, achieving high-yield with gentle magnetic movement of antibody-labeled cells through virtual barriers of surface tension. To achieve clinical-grade quantification of rare cells, we employ high quality fluorescence microscopy image acquisition and automated image analysis together termed quantitative microscopy. RESULTS: In preparation for clinical laboratory implementation, we demonstrate high precision and accuracy of these methodologies using a diverse set of control materials. Preliminary testing of CTCs isolated from patients with NSCLC demonstrate heterogeneity in PD-L1 and HLA I expression and promising clinical value in predicting PFS in response to PD-L1 targeted therapies. CONCLUSIONS: By confirming high performance, we ensure compatibility for clinical laboratory implementation and future application to better predict and detect resistance to PD-L1 targeted therapy in patients with NSCLC.

3.
Psychopharmacology (Berl) ; 233(4): 549-69, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26758282

ABSTRACT

RATIONALE: For several decades, elite athletes and a growing number of recreational consumers have used anabolic androgenic steroids (AAS) as performance enhancing drugs. Despite mounting evidence that illicit use of these synthetic steroids has detrimental effects on affective states, information available on sex-specific actions of these drugs is lacking. OBJECTIVES: The focus of this review is to assess information to date on the importance of sex and its interaction with other environmental factors on affective behaviors, with an emphasis on data derived from non-human studies. METHODS: The PubMed database was searched for relevant studies in both sexes. RESULTS: Studies examining AAS use in females are limited, reflecting the lower prevalence of use in this sex. Data, however, indicate significant sex-specific differences in AAS effects on anxiety-like and aggressive behaviors, interactions with other drugs of abuse, and the interplay of AAS with other environmental factors such as diet and exercise. CONCLUSIONS: Current methods for assessing AAS use have limitations that suggest biases of both under- and over-reporting, which may be amplified for females who are poorly represented in self-report studies of human subjects and are rarely used in animal studies. Data from animal literature suggest that there are significant sex-specific differences in the impact of AAS on aggression, anxiety, and concomitant use of other abused substances. These results have relevance for human females who take these drugs as performance-enhancing substances and for transgender XX individuals who may illicitly self-administer AAS as they transition to a male gender identity.


Subject(s)
Anabolic Agents/adverse effects , Androgens/adverse effects , Mood Disorders/chemically induced , Mood Disorders/psychology , Sex Characteristics , Aggression/drug effects , Aggression/psychology , Anabolic Agents/administration & dosage , Androgens/administration & dosage , Animals , Anxiety/chemically induced , Anxiety/psychology , Female , Humans , Male , Self Administration , Steroids/administration & dosage , Steroids/adverse effects , Treatment Outcome
4.
Horm Behav ; 66(2): 283-97, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24768711

ABSTRACT

Anabolic androgenic steroids (AAS) are taken by both sexes to enhance athletic performance and body image, nearly always in conjunction with an exercise regime. Although taken to improve physical attributes, chronic AAS use can promote negative behavior, including anxiety. Few studies have directly compared the impact of AAS use in males versus females or assessed the interaction of exercise and AAS. We show that AAS increase anxiety-like behaviors in female but not male mice and that voluntary exercise accentuates these sex-specific differences. We also show that levels of the anxiogenic peptide corticotrophin releasing factor (CRF) are significantly greater in males, but that AAS selectively increase CRF levels in females, thus abrogating this sex-specific difference. Exercise did not ameliorate AAS-induced anxiety or alter CRF levels in females. Exercise was anxiolytic in males, but this behavioral outcome did not correlate with CRF levels. Brain-derived neurotrophic factor (BDNF) has also been implicated in the expression of anxiety. As with CRF, levels of hippocampal BDNF mRNA were significantly greater in males than females. AAS and exercise were without effect on BDNF mRNA in females. In males, anxiolytic effects of exercise correlated with increased BDNF mRNA, however AAS-induced changes in BDNF mRNA and anxiety did not. In sum, we find that AAS elicit sex-specific differences in anxiety and that voluntary exercise accentuates these differences. In addition, our data suggest that these behavioral outcomes may reflect convergent actions of AAS and exercise on a sexually differentiated CRF signaling system within the extended amygdala.


Subject(s)
Anabolic Agents/toxicity , Androgens/toxicity , Anxiety/chemically induced , Anxiety/psychology , Physical Conditioning, Animal/physiology , Sexual Behavior, Animal/physiology , Steroids/toxicity , Animals , Body Weight/drug effects , Choice Behavior , Corticotropin-Releasing Hormone/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Reflex, Startle/drug effects , Sex Characteristics , Social Behavior
5.
Clin Vaccine Immunol ; 19(9): 1539-48, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22855390

ABSTRACT

Human babesiosis, a blood-borne infection caused by several species of Babesia, including B. microti, is an emerging disease that is endemic in the Northeast, upper Midwest, and Pacific Northwest regions of the United States. Risk factors for babesiosis include exposure to the infected tick vector and blood transfusions from infected donors. In this work, we cloned and expressed two of the immunodominant antigens from B. microti and used them in a multiplex bead format assay (MBA) to detect parasite-specific IgG responses in human sera. The MBA using recombinant B. microti secreted antigen 1 (BmSA1) protein was more specific (100%) and slightly more sensitive (98.7%) than the assay using a truncated recombinant BMN1-17 construct (97.6% and 97.4%, respectively). Although some antibody reactivity was observed among sera from confirmed-malaria patients, only one Plasmodium falciparum sample was simultaneously positive for IgG antibodies to both antigens. Neither antigen reacted with sera from babesiosis patients who were infected with Babesia species other than B. microti. Both positive and negative MBA results were reproducible between assays and between instruments. Additional studies of these recombinant antigens and of the multiplex bead assay using blood samples from clinically defined babesiosis patients and from blood donors are needed to more clearly define their usefulness as a blood screening assay.


Subject(s)
Antibodies, Protozoan/blood , Antigens, Protozoan , Babesia microti/isolation & purification , Babesiosis/diagnosis , Clinical Laboratory Techniques/methods , Immunoglobulin G/blood , Adult , Antigens, Protozoan/genetics , Babesia microti/genetics , Babesia microti/immunology , Cloning, Molecular , Female , Gene Expression , Humans , Immunoassay/methods , Microspheres , Recombinant Proteins/genetics , Sensitivity and Specificity , United States
6.
Neuroendocrinology ; 96(2): 141-51, 2012.
Article in English | MEDLINE | ID: mdl-22576754

ABSTRACT

Anabolic androgenic steroids (AAS) comprise a large and growing class of synthetic androgens used clinically to promote tissue-building in individuals suffering from genetic disorders, injuries, and diseases. Despite these beneficial therapeutic applications, the predominant use of AAS is illicit: these steroids are self-administered to promote athletic performance and body image. Hand in hand with the desired anabolic actions of the AAS are untoward effects on the brain and behavior. While the signaling routes by which the AAS impose both beneficial and harmful actions may be quite diverse, key endpoints are likely to include ligand-gated and voltage-dependent ion channels that govern the activity of electrically excitable tissues. Here, we review the known effects of AAS on molecular targets that play critical roles in controlling electrical activity, with a specific focus on the effects of AAS on neurotransmission mediated by GABA(A) receptors in the central nervous system.


Subject(s)
Anabolic Agents/pharmacology , Neurons/metabolism , Synaptic Transmission/physiology , Testosterone Congeners/pharmacology , Animals , Brain/cytology , Humans , Ion Channels/metabolism , Neurons/drug effects , Receptors, GABA-A/metabolism , Steroids/metabolism , Synaptic Transmission/drug effects
7.
Trends Neurosci ; 35(6): 382-92, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22516619

ABSTRACT

Anabolic androgenic steroids (AAS) are illicitly administered to enhance athletic performance and body image. Although conferring positive actions on performance, steroid abuse is associated with changes in anxiety and aggression. AAS users are often keenly invested in understanding the biological actions of these drugs. Thus, mechanistic information on AAS actions is important not only for the biomedical community, but also for steroid users. Here we review findings from animal studies on the impact of AAS exposure on neural systems that are crucial for the production of anxiety and aggression, and compare the effects of the different classes of AAS and their potential signaling mechanisms, as well as context-, age- and sex-dependent aspects of their actions.


Subject(s)
Aggression/drug effects , Anabolic Agents/adverse effects , Anxiety/physiopathology , Brain/physiopathology , Fear/drug effects , Performance-Enhancing Substances/adverse effects , Steroids/adverse effects , Anxiety/chemically induced , Humans , Models, Neurological
8.
Neuropsychopharmacology ; 37(6): 1483-99, 2012 May.
Article in English | MEDLINE | ID: mdl-22298120

ABSTRACT

Increased anxiety is commonly observed in individuals who illicitly administer anabolic androgenic steroids (AAS). Behavioral effects of steroid abuse have become an increasing concern in adults and adolescents of both sexes. The dorsolateral bed nucleus of the stria terminalis (dlBnST) has a critical role in the expression of diffuse anxiety and is a key site of action for the anxiogenic neuromodulator, corticotropin releasing factor (CRF). Here we demonstrate that chronic, but not acute, exposure of female mice during adolescence to AAS augments anxiety-like behaviors; effects that were blocked by central infusion of the CRF receptor type 1 antagonist, antalarmin. AAS treatment selectively increased action potential (AP) firing in neurons of the central amygdala (CeA) that project to the dlBnST, increased the frequency of GABA(A) receptor-mediated spontaneous inhibitory postsynaptic currents (sIPSCs) in dlBnST target neurons, and decreased both c-FOS immunoreactivity (IR) and AP frequency in these postsynaptic cells. Acute application of antalarmin abrogated the enhancement of GABAergic inhibition induced by chronic AAS exposure whereas application of CRF to brain slices of naïve mice mimicked the actions of this treatment. These results, in concert with previous data demonstrating that chronic AAS treatment results in enhanced levels of CRF mRNA in the CeA and increased CRF-IR in the dlBnST neuropil, are consistent with a mechanism in which the enhanced anxiety elicited by chronic AAS exposure involves augmented inhibitory activity of CeA afferents to the dlBnST and CRF-dependent enhancement of GABAergic inhibition in this brain region.


Subject(s)
Androgens/toxicity , Anxiety/pathology , Corticotropin-Releasing Hormone/pharmacology , Prosencephalon/drug effects , Acoustic Stimulation , Action Potentials/drug effects , Amygdala/drug effects , Amygdala/metabolism , Amygdala/pathology , Analysis of Variance , Animals , Anxiety/chemically induced , Anxiety/drug therapy , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/therapeutic use , Disease Models, Animal , Dose-Response Relationship, Drug , Female , GABAergic Neurons/drug effects , In Vitro Techniques , Inhibitory Postsynaptic Potentials/drug effects , Injections, Intraventricular , Mice , Mice, Inbred C57BL , Patch-Clamp Techniques , Prosencephalon/metabolism , Prosencephalon/pathology , Proto-Oncogene Proteins c-fos/metabolism , Pyrimidines/pharmacology , Pyrroles/pharmacology , RNA, Messenger/metabolism , Sensory Gating/drug effects , Time Factors
9.
Hum Mol Genet ; 21(4): 730-50, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22048958

ABSTRACT

We have identified a point mutation in Npc1 that creates a novel mouse model (Npc1(nmf164)) of Niemann-Pick type C1 (NPC) disease: a single nucleotide change (A to G at cDNA bp 3163) that results in an aspartate to glycine change at position 1005 (D1005G). This change is in the cysteine-rich luminal loop of the NPC1 protein and is highly similar to commonly occurring human mutations. Genetic and molecular biological analyses, including sequencing the Npc1(spm) allele and identifying a truncating mutation, confirm that the mutation in Npc1(nmf164) mice is distinct from those in other existing mouse models of NPC disease (Npc1(nih), Npc1(spm)). Analyses of lifespan, body and spleen weight, gait and other motor activities, as well as acoustic startle responses all reveal a more slowly developing phenotype in Npc1(nmf164) mutant mice than in mice with the null mutations (Npc1(nih), Npc1(spm)). Although Npc1 mRNA levels appear relatively normal, Npc1(nmf164) brain and liver display dramatic reductions in Npc1 protein, as well as abnormal cholesterol metabolism and altered glycolipid expression. Furthermore, histological analyses of liver, spleen, hippocampus, cortex and cerebellum reveal abnormal cholesterol accumulation, glial activation and Purkinje cell loss at a slower rate than in the Npc1(nih) mouse model. Magnetic resonance imaging studies also reveal significantly less demyelination/dysmyelination than in the null alleles. Thus, although prior mouse models may correspond to the severe infantile onset forms of NPC disease, Npc1(nmf164) mice offer many advantages as a model for the late-onset, more slowly progressing forms of NPC disease that comprise the large majority of human cases.


Subject(s)
Carrier Proteins/genetics , Disease Models, Animal , Membrane Glycoproteins/genetics , Niemann-Pick Disease, Type C/genetics , Point Mutation/genetics , Age of Onset , Alleles , Animals , Astrocytes/pathology , Brain/metabolism , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Cholesterol/metabolism , DNA Mutational Analysis , Disease Progression , Endoplasmic Reticulum Stress , Gangliosides/metabolism , Homozygote , Humans , Intracellular Signaling Peptides and Proteins , Lipid Metabolism , Lung/cytology , Macrophages/metabolism , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/metabolism , Mice , Microglia/pathology , Myelin Sheath , Niemann-Pick C1 Protein , Niemann-Pick Disease, Type C/metabolism , Niemann-Pick Disease, Type C/pathology , Niemann-Pick Disease, Type C/physiopathology , Phenotype , Proteostasis Deficiencies , Purkinje Cells/pathology , RNA, Messenger/analysis , RNA, Messenger/genetics , Reflex, Startle , Survival Rate
10.
Neuropharmacology ; 61(4): 653-64, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21645530

ABSTRACT

Disruption of reproductive function is a hallmark of abuse of anabolic androgenic steroids (AAS) in female subjects. To understand the central actions of AAS, patch clamp recordings were made in estrous, diestrous and AAS-treated mice from gonadotropin releasing hormone (GnRH) neurons, neurons in the medial preoptic area (mPOA) and neurons in the anteroventroperiventricular nucleus (AVPV); regions known to provide GABAergic and kisspeptin inputs to the GnRH cells. Action potential (AP) frequency was significantly higher in GnRH neurons of estrous mice than in AAS-treated or diestrous animals. No significant differences in AAS-treated, estrous or diestrous mice were evident in the amplitude or kinetics of spontaneous postsynaptic currents (sPSCs), miniature PSCs or tonic currents mediated by GABA(A) receptors or in GABA(A) receptor subunit expression in GnRH neurons. In contrast, the frequency of GABA(A) receptor-mediated sPSCs in GnRH neurons showed an inverse correlation with AP frequency across the three hormonal states. Surprisingly, AP activity in the medial preoptic area (mPOA), a likely source of GABAergic afferents to GnRH cells, did not vary in concert with the sPSCs in the GnRH neurons. Furthermore, pharmacological blockade of GABA(A) receptors did not alter the pattern in which there was lower AP frequency in GnRH neurons of AAS-treated and diestrous versus estrous mice. These data suggest that AAS do not impose their effects either directly on GnRH neurons or on putative GABAergic afferents in the mPOA. AP activity recorded from neurons in kisspeptin-rich regions of the AVPV and the expression of kisspeptin mRNA and peptide did vary coordinately with AP activity in GnRH neurons. Our data demonstrate that AAS treatment imposes a "diestrous-like" pattern of activity in GnRH neurons and suggest that this effect may arise from suppression of presynaptic kisspeptin-mediated excitatory drive arising from the AVPV. The actions of AAS on neuroendocrine regulatory circuits may contribute the disruption of reproductive function observed in steroid abuse.


Subject(s)
Anabolic Agents/administration & dosage , Methyltestosterone/administration & dosage , Midline Thalamic Nuclei/drug effects , Preoptic Area/drug effects , Synapses/drug effects , Synaptic Potentials/drug effects , Animals , Female , Gonadotropin-Releasing Hormone/biosynthesis , Gonadotropin-Releasing Hormone/physiology , Mice , Mice, Transgenic , Midline Thalamic Nuclei/physiology , Neurons/metabolism , Preoptic Area/physiology , Synapses/physiology , Synaptic Potentials/physiology
11.
Eur J Med Genet ; 54(2): 161-4, 2011.
Article in English | MEDLINE | ID: mdl-21078420

ABSTRACT

Abnormalities involving sex chromosomes account for approximately 0.5% of live births. The phenotypes of individuals with mosaic cell lines having structural aberrations of the X and Y chromosomes are variable and hard to accurately predict. Phenotypes associated with sex chromosome mosaicism range from Turner syndrome to males with infertility, and often present with ambiguous genitalia. Previous studies of individuals with an 45,X/46,X,idic(Y)(p11) karyotype suggest that the presence of both cell lines should result from an intermediate, 46,XY cell line. Here we report a 2.5 year old female with phenotypic features of Turner syndrome with an isodicentric Y chromosome and a cell line with a deleted Y with a final karyotype of 45,X/46,X,idic(Y)(p11.31)/46,X,del(Y)(p11.31). Fluorescence in situ hybridization (FISH) mapping of the Y chromosome breakpoint revealed very low percentages of the deleted Y cells, but suggested a potential mechanism for the formation of the isodicentric Y chromosome. To our knowledge, the 46,X,del(Y) intermediate cell line in our patient has not been previously reported in individuals with mosaic sex chromosome structural abnormalities.


Subject(s)
Chromosomes, Human, Y/genetics , Mosaicism , Sex Chromosome Aberrations , Child, Preschool , Chromosome Aberrations , Female , Humans , Karyotyping , Turner Syndrome
12.
Psychoneuroendocrinology ; 35(10): 1473-85, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20537804

ABSTRACT

In the past several decades, the therapeutic use of anabolic androgenic steroids (AAS) has been overshadowed by illicit use of these drugs by elite athletes and a growing number of adolescents to enhance performance and body image. As with adults, AAS use by adolescents is associated with a range of behavioral effects, including increased anxiety and altered responses to stress. It has been suggested that adolescents, especially adolescent females, may be particularly susceptible to the effects of these steroids, but few experiments in animal models have been performed to test this assertion. Here we show that chronic exposure of adolescent female mice to a mixture of three commonly abused AAS (testosterone cypionate, nandrolone decanoate and methandrostenolone; 7.5 mg/kg/day for 5 days) significantly enhanced anxiety-like behavior as assessed by the acoustic startle response (ASR), but did not augment the fear-potentiated startle response (FPS) or alter sensorimotor gating as assessed by prepulse inhibition of the acoustic startle response (PPI). AAS treatment also significantly increased the levels of corticotropin releasing factor (CRF) mRNA and somal-associated CRF immunoreactivity in the central nucleus of the amygdala (CeA), as well as neuropil-associated immunoreactivity in the dorsal aspect of the anterolateral division of the bed nucleus of the stria terminalis (dBnST). AAS treatment did not alter CRF receptor 1 or 2 mRNA in either the CeA or the dBnST; CRF immunoreactivity in the ventral BnST, the paraventricular nucleus (PVN) or the median eminence (ME); or peripheral levels of corticosterone. These results suggest that chronic AAS treatment of adolescent female mice may enhance generalized anxiety, but not sensorimotor gating or learned fear, via a mechanism that involves increased CRF-mediated signaling from CeA neurons projecting to the dBnST.


Subject(s)
Anabolic Agents/pharmacology , Androgens/pharmacology , Anxiety/psychology , Corticotropin-Releasing Hormone/biosynthesis , Steroids/pharmacology , Amygdala/drug effects , Animals , Behavior, Animal/drug effects , Corticosterone/blood , Fear/psychology , Female , Habituation, Psychophysiologic/drug effects , Immunohistochemistry , Methandrostenolone/pharmacology , Mice , Mice, Inbred C57BL , Nandrolone/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reflex, Startle/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Septal Nuclei/drug effects
13.
J Neurosci ; 30(19): 6497-506, 2010 May 12.
Article in English | MEDLINE | ID: mdl-20463213

ABSTRACT

Gonadotropin-releasing hormone (GnRH) neurons are the central regulators of reproduction. GABAergic transmission plays a critical role in pubertal activation of pulsatile GnRH secretion. Self-administration of excessive doses of anabolic androgenic steroids (AAS) disrupts reproductive function and may have critical repercussions for pubertal onset in adolescent users. Here, we demonstrate that chronic treatment of adolescent male mice with the AAS 17alpha-methyltestosterone significantly decreased action potential frequency in GnRH neurons, reduced the serum gonadotropin levels, and decreased testes mass. AAS treatment did not induce significant changes in GABAA receptor subunit mRNA levels or alter the amplitude or decay kinetics of GABAA receptor-mediated spontaneous postsynaptic currents (sPSCs) or tonic currents in GnRH neurons. However, AAS treatment significantly increased action potential frequency in neighboring medial preoptic area (mPOA) neurons and GABAA receptor-mediated sPSC frequency in GnRH neurons. In addition, physical isolation of the more lateral aspects of the mPOA from the medially localized GnRH neurons abrogated the AAS-induced increase in GABAA receptor-mediated sPSC frequency and the decrease in action potential firing in the GnRH cells. Our results indicate that AAS act predominantly on steroid-sensitive presynaptic neurons within the mPOA to impart significant increases in GABAA receptor-mediated inhibitory tone onto downstream GnRH neurons, resulting in diminished activity of these pivotal mediators of reproductive function. These AAS-induced changes in central GABAergic circuits of the forebrain may significantly contribute to the disruptive actions of these drugs on pubertal maturation and the development of reproductive competence in male steroid abusers.


Subject(s)
Anabolic Agents/toxicity , Gonadotropin-Releasing Hormone/metabolism , Methyltestosterone/toxicity , Neurons/drug effects , Receptors, GABA-A/metabolism , Substance-Related Disorders/physiopathology , Synaptic Transmission/drug effects , Action Potentials/drug effects , Action Potentials/physiology , Androgens/toxicity , Animals , Gonadotropins/blood , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/physiopathology , Male , Mice , Mice, Transgenic , Neurons/physiology , Preoptic Area/drug effects , Preoptic Area/physiopathology , Presynaptic Terminals/drug effects , Presynaptic Terminals/physiology , RNA, Messenger/metabolism , Substance-Related Disorders/blood , Substance-Related Disorders/pathology , Synaptic Potentials/drug effects , Synaptic Potentials/physiology , Synaptic Transmission/physiology , Testis/drug effects , Testis/pathology
14.
J Neurosci ; 30(4): 1463-70, 2010 Jan 27.
Article in English | MEDLINE | ID: mdl-20107073

ABSTRACT

AMPA receptors (AMPARs) are tetrameric ligand-gated ion channels that couple the energy of glutamate binding to the opening of a transmembrane channel. Crystallographic and electrophysiological analysis of AMPARs has suggested a coupling between (1) cleft closure in the bilobate ligand-binding domain (LBD), (2) the resulting separation of transmembrane helix attachment points across subunit dimers, and (3) agonist efficacy. In general, more efficacious agonists induce greater degrees of cleft closure and transmembrane separation than partial agonists. Several apparent violations of the cleft-closure/efficacy paradigm have emerged, although in all cases, intradimer separation remains as the driving force for channel opening. Here, we examine the structural basis of partial agonism in GluA4 AMPARs. We find that the L651V substitution enhances the relative efficacy of kainate without increasing either LBD cleft closure or transmembrane separation. Instead, the conformational change relative to the wild-type:kainate complex involves a twisting motion with the efficacy contribution opposite from that expected based on previous analyses. As a result, channel opening may involve transmembrane rearrangements with a significant rotational component. Furthermore, a two-dimensional analysis of agonist-induced GluA2 LBD motions suggests that efficacy is not a linearly varying function of lobe 2 displacement vectors, but is rather determined by specific conformational requirements of the transmembrane domains.


Subject(s)
Excitatory Amino Acid Agonists/chemistry , Excitatory Amino Acid Agonists/pharmacology , Glutamic Acid/metabolism , Receptors, AMPA/agonists , Receptors, AMPA/chemistry , Synaptic Transmission/physiology , Binding Sites , Cell Line , Cell Membrane/chemistry , Cell Membrane/drug effects , Cell Membrane/metabolism , Crystallography, X-Ray , Humans , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , Ion Channels/agonists , Ion Channels/chemistry , Kainic Acid/pharmacology , Ligands , Patch-Clamp Techniques , Protein Structure, Tertiary/drug effects , Protein Structure, Tertiary/physiology , Proteomics , Receptors, AMPA/metabolism
15.
J Neurosci ; 29(40): 12484-96, 2009 Oct 07.
Article in English | MEDLINE | ID: mdl-19812324

ABSTRACT

Anabolic androgenic steroids (AAS) can promote detrimental effects on social behaviors for which GABA type A (GABA(A)) receptor-mediated circuits in the forebrain play a critical role. While all AAS bind to androgen receptors (AR), they may also be aromatized to estrogens and thus potentially impart effects via estrogen receptors (ER). Chronic exposure of wild-type male mice to a combination of chemically distinct AAS increased action potential (AP) frequency, selective GABA(A) receptor subunit mRNAs, and GABAergic synaptic current decay in the medial preoptic area (mPOA). Experiments performed with pharmacological agents and in AR-deficient Tfm mutant mice suggest that the AAS-dependent enhancement of GABAergic transmission in wild-type mice is AR-mediated. In AR-deficient mice, the AAS elicited dramatically different effects, decreasing AP frequency, spontaneous IPSC amplitude and frequency and the expression of selective GABA(A) receptor subunit mRNAs. Surprisingly, in the absence of AR signaling, the data indicate that the AAS do not act as ER agonists, but rather suggest a novel in vivo action in which the AAS inhibit aromatase and impair endogenous ER signaling. These results show that the AAS have the capacity to alter neuronal function in the forebrain via multiple steroid signaling mechanisms and suggest that effects of these steroids in the brain will depend not only on the balance of AR- versus ER-mediated regulation for different target genes, but also on the ability of these drugs to alter steroid metabolism and thus the endogenous steroid milieu.


Subject(s)
Methyltestosterone/administration & dosage , Nandrolone/analogs & derivatives , Neurons/drug effects , Preoptic Area/drug effects , Receptors, Estrogen/metabolism , Receptors, GABA/drug effects , Testosterone/analogs & derivatives , Action Potentials/drug effects , Anabolic Agents/administration & dosage , Androgen Receptor Antagonists , Androgens/administration & dosage , Animals , Aromatase Inhibitors/pharmacology , Drug Combinations , Estradiol/metabolism , Estrogens/metabolism , Estrogens/pharmacology , Injections, Intraperitoneal , Male , Mice , Mice, Transgenic , Nandrolone/administration & dosage , Nandrolone Decanoate , Neurons/metabolism , Preoptic Area/cytology , Receptors, Androgen/administration & dosage , Receptors, Androgen/deficiency , Synaptic Transmission/drug effects , Testosterone/administration & dosage , Testosterone/metabolism
16.
J Nutr Elder ; 28(2): 112-26, 2009 Apr.
Article in English | MEDLINE | ID: mdl-21184361

ABSTRACT

Adults aged 60 years and older are more likely than younger adults to experience complications, hospitalization, and death because of food-borne infections. Recognizing this risk, we conducted a nationally representative survey (n = 1,140) to characterize older adults' food safety knowledge, attitudes, and practices as well as the demographic characteristics of older adults with risky food handling practices. The survey was conducted using a Web-enabled panel. We found that although older adults consider themselves to be knowledgeable about food safety, many are not following recommended food safety practices. Areas for improvement include the following: reheating deli meats to steaming hot, not eating store-bought deli salads, cooking eggs properly, monitoring refrigerator temperature using a thermometer, using a food thermometer to check doneness of meat/poultry/egg dishes, and storing leftovers properly. The survey results also suggest that food safety education targeting older adults is needed and that such initiatives should emphasize practices to prevent listeriosis, a potentially fatal illness among older adults. Our findings suggest that, in particular, men, individuals with higher incomes, and college-educated individuals would benefit from food safety education.


Subject(s)
Consumer Product Safety , Food Handling/methods , Foodborne Diseases/epidemiology , Health Education/organization & administration , Health Knowledge, Attitudes, Practice , Aged , Educational Status , Female , Food Handling/standards , Food Microbiology , Food Safety , Health Education/standards , Humans , Male , Middle Aged , Risk-Taking , Sex Distribution
17.
Biochemistry ; 47(52): 13831-41, 2008 Dec 30.
Article in English | MEDLINE | ID: mdl-19102704

ABSTRACT

AMPA receptors are glutamate-gated ion channels that are essential mediators of synaptic signals in the central nervous system. They form tetramers that are assembled as combinations of subunits GluR1-4, each of which contains a ligand-binding domain (LBD). Crystal structures of the GluR2 LBD have revealed an agonist-binding cleft, which is located between two lobes and which acts like a Venus flytrap. In general, agonist efficacy is correlated with the extent of cleft closure. However, recent observations show that cleft closure is not the sole determinant of the relative efficacy for glutamate receptors. In addition, these studies have focused on the GluR2 subunit, which is the specific target of a physiologically important RNA-editing modification in vivo. We therefore sought to test the generality of the cleft closure-efficacy correlation for other AMPA-R subunits. Here, we present crystal structures of the GluR4(flip) LBD in complex with both full and partial agonists. As for GluR2, both agonists stabilize a closed-cleft conformation, and the partial agonist induces a smaller cleft closure than the full agonist. However, a detailed analysis of LBD-kainate interactions reveals the importance of subtle backbone conformational changes in the ligand-binding pocket in determining the magnitude of agonist-associated conformational changes. Furthermore, the GluR4 subunit exhibits a different correlation between receptor activation and LBD cleft closure than does GluR2.


Subject(s)
Receptors, AMPA/agonists , Receptors, AMPA/chemistry , Animals , Binding Sites , Crystallography, X-Ray , Ligands , Protein Binding , Protein Conformation , Rats , Receptors, AMPA/metabolism
18.
Phys Ther ; 88(2): 286-94, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18073266

ABSTRACT

BACKGROUND AND PURPOSE: The Movement Assessment Battery for Children (M-ABC) is a widely used, standardized assessment of motor performance in children. The total score obtained on this test often is used to identify children who are either definitely impaired or at risk for motor impairment. The purpose of this study was to determine the interrater reliability of data for the M-ABC when scored by pediatric physical therapists working in routine clinical settings. SUBJECTS AND METHODS: For 9 children who were referred to clinical settings for an assessment of possible movement difficulties, performance on the appropriate age band of the M-ABC was videotaped. The 9 children, one at each age from 4 through 12 years, represented all ages covered by the test. The videotaped performances were rated according to the test instructions by 131 pediatric physical therapists with a range of experience and by an expert rater who developed the Dutch version of the test. RESULTS: The average agreement between therapists in their classification of the children was very high. The kappa coefficients for the 9 videos ranged from .95 to 1.00. DISCUSSION AND CONCLUSION: Errors made by the therapists could be classified as those that might be common to all tests and those that are specific to the M-ABC.


Subject(s)
Disabled Children/classification , Motor Skills Disorders/diagnosis , Motor Skills , Child , Child, Preschool , Female , Humans , Male , Motor Skills Disorders/classification , Reproducibility of Results , Videotape Recording
19.
J Neurochem ; 103(3): 1258-71, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17714454

ABSTRACT

Incorporation of the epsilon subunit into the GABAA receptor has been suggested to confer unusual, but variable, biophysical and pharmacological characteristics to both recombinant and native receptors. Due to their structural similarity with the gamma subunits, epsilon subunits have been assumed to substitute at the single position of the gamma subunit in assembled receptors. However, prior work suggests that functional variability in epsilon-containing receptors may reflect alternative sites of incorporation and of not just one, but possibly multiple epsilon subunits in the pentameric receptor complex. Here we present data indicating that increased expression of epsilon, in conjunction with alpha2 and beta3 subunits, results in expression of GABAA receptors with correspondingly altered rectification, deactivation and levels of spontaneous openings, but not increased total current density. We also provide data that the epsilon subunit, like the beta3 subunit, can self-export and data from chimeric receptors suggesting that similarities between the assembly domains of the beta3 and the epsilon subunits may allow the epsilon subunit to replace the beta, as well as the gamma, subunit. The substitution of an epsilon for a beta, as well as the gamma subunit and formation of receptors with alternative patterns of assembly with respect to epsilon incorporation may underlie the observed variability in both biophysical and pharmacological properties noted not only in recombinant, but also in native receptors.


Subject(s)
Cell Membrane/metabolism , Receptors, GABA-A/biosynthesis , Cell Line , Humans , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , Macromolecular Substances/chemistry , Macromolecular Substances/metabolism , Membrane Potentials/drug effects , Membrane Potentials/genetics , Models, Molecular , Neural Inhibition/genetics , Protein Structure, Tertiary/drug effects , Protein Structure, Tertiary/physiology , Protein Subunits/drug effects , Protein Subunits/genetics , Protein Subunits/metabolism , Protein Transport/physiology , Receptors, GABA-A/chemistry , Receptors, GABA-A/drug effects , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Synaptic Transmission/genetics , Transfection , Zinc/metabolism , Zinc/pharmacology , gamma-Aminobutyric Acid/metabolism , gamma-Aminobutyric Acid/pharmacology
20.
Neuropharmacology ; 52(7): 1439-53, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17433821

ABSTRACT

The hypothalamus, the seat of neuroendocrine control, is exquisitely sensitive to gonadal steroids. For decades it has been known that androgens, estrogens and progestins, acting through nuclear hormone receptors, elicit both organizational and activational effects in the hypothalamus and basal forebrain that are essential for reproductive function. While changes in gene expression mediated by these classical hormone pathways are paramount in governing both sexual differentiation and the neural control of reproduction, it is also clear that steroids impart critical control of neuroendocrine functions through non-genomic mechanisms. Specifically, endogenous neurosteroid derivatives of deoxycorticosterone, progesterone and testosterone, as well and synthetic anabolic androgenic steroids that are self-administered as drugs of abuse, elicit acute effects via allosteric modulation of gamma-aminobutyric acid type A receptors. GABAergic transmission within the hypothalamus and basal forebrain is a key regulator of pubertal onset, the expression of sexual behaviors, pregnancy and parturition. Summarized here are the known actions of steroid modulators on GABAergic transmission within the hypothalamus/basal forebrain, with a focus on the medial preoptic area and the supraoptic/paraventricular nuclei that are known to be central players in the control of reproduction.


Subject(s)
Hypothalamus/physiology , Receptors, GABA-B/physiology , Reproduction/physiology , Steroids/metabolism , Synaptic Transmission/physiology , Animals , Humans , Hypothalamus/drug effects , Reproduction/drug effects , Steroids/pharmacology , Synaptic Transmission/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL