Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Metab Brain Dis ; 37(8): 2735-2750, 2022 12.
Article in English | MEDLINE | ID: mdl-35951206

ABSTRACT

Alzheimer disease's (AD) is a neurodegenerative disorder characterized by cognitive and behavioral impairment. The central nervous system is an important target of thyroid hormones (TH). An inverse association between serum triiodothyronine (T3) levels and the risk of AD symptoms and progression has been reported. We investigated the effects of T3 treatment on the depression-like behavior in male transgenic 3xTg-AD mice. Animals were divided into 2 groups treated with daily intraperitoneal injections of 20 ng/g of body weight (b.w.) L-T3 (T3 group) or saline (vehicle, control group). The experimental protocol lasted 21 days, and behavioral tests were conducted on days 18-20. At the end of the experiment, the TH profile and hippocampal gene expression were evaluated. The T3-treated group significantly increased serum T3 and decreased thyroxine (T4) levels. When compared to control hippocampal samples, the T3 group exhibited attenuated glycogen synthase kinase-3 (GSK3), metalloproteinase 10 (ADAM10), amyloid-beta precursor-protein (APP), serotonin transporter (SERT), 5HT1A receptor, monocarboxylate transporter 8 (MCT8) and bone morphogenetic protein 7 (BMP-7) gene expression, whereas augmented superoxide dismutase 2 (SOD2) and Hairless gene expression. T3-treated animals also displayed reduced immobility time in both the tail suspension and forced swim tests, and in the latter presented a higher latency time compared to the control group. Therefore, our findings suggest that in an AD mouse model, T3 supplementation promotes improvements in depression-like behavior, through the modulation of the serotonergic related genes involved in the transmission mediated by 5HT1A receptors and serotonin reuptake, and attenuated disease progression.


Subject(s)
Alzheimer Disease , Triiodothyronine , Animals , Mice , Male , Triiodothyronine/pharmacology , Triiodothyronine/therapeutic use , Alzheimer Disease/metabolism , Depression/drug therapy , Glycogen Synthase Kinase 3 , Mice, Transgenic , Thyroid Hormones/metabolism , Disease Models, Animal
2.
Arq Neuropsiquiatr ; 74(4): 329-36, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27097007

ABSTRACT

Pathology of the rhinencephalon has been a subject of interest in the fields of neurodegenerative diseases, trauma, epilepsy and other neurological conditions. Most of what is known about the human rhinencephalon comes from comparative anatomy studies in other mammals and histological studies in primates. Functional imaging studies can provide new and important insight into the function of the rhinencephalon in humans but have limited spatial resolution, limiting its contribution to the study of the anatomy of the human rhinencephalon. In this study we aim to provide a brief and objective review of the anatomy of this important and often overlooked area of the nervous system.


Subject(s)
Olfactory Cortex/anatomy & histology , Humans , Medical Illustration , Olfactory Bulb/anatomy & histology , Olfactory Mucosa/anatomy & histology , Olfactory Receptor Neurons
3.
Arq. neuropsiquiatr ; 74(4): 329-336, Apr. 2016. tab, graf
Article in English | LILACS | ID: lil-779808

ABSTRACT

ABSTRACT Pathology of the rhinencephalon has been a subject of interest in the fields of neurodegenerative diseases, trauma, epilepsy and other neurological conditions. Most of what is known about the human rhinencephalon comes from comparative anatomy studies in other mammals and histological studies in primates. Functional imaging studies can provide new and important insight into the function of the rhinencephalon in humans but have limited spatial resolution, limiting its contribution to the study of the anatomy of the human rhinencephalon. In this study we aim to provide a brief and objective review of the anatomy of this important and often overlooked area of the nervous system.


RESUMO As patologias do rinencéfalo tem sido assunto de interesse para os estudiosos das doenças neurodegenerativas, do traumatismo cranio-encefálico, epilepsia e outras doenças neurológicas. A maior parte do conhecimento sobre a anatomia do rinencéfalo vem de estudos de anatomia comparativa com outros mamíferos e estudos histológicos em primatas. Estudos de imagem funcional, apesar de proporcionarem informações úteis e interessantes a respeito do funcionamento do rinencéfalo em humanos, sofrem de resolução espacial limitada, e portanto contribuem de maneira restrita ao estudo dos limites das áreas anatômicas. Neste artigo buscamos proporcionar ao neurologista e neurocientista interessado uma revisão prática e objetiva da anatomia desta área importante e muitas vezes esquecida do sistema nervoso.


Subject(s)
Humans , Olfactory Cortex/anatomy & histology , Medical Illustration , Olfactory Receptor Neurons , Olfactory Bulb/anatomy & histology , Olfactory Mucosa/anatomy & histology
4.
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
5.
Horm Behav ; 61(1): 67-75, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22057031

ABSTRACT

Humans self-administer anabolic androgenic steroids (AAS) at superphysiological doses for the purpose of building muscle mass and enhancing physique whereas considerably lower doses of AAS are prescribed in the clinic to treat a variety of disorders. A number of studies have demonstrated that individual AAS influence aggressive behavior in rats and mice, but few studies have examined the aggression-enhancing effects of combinations of AAS. Using the resident-intruder paradigm, Experiment 1 determined whether a cocktail of commonly abused AAS increased aggressive behavior in gonadally-intact male C57BL/6J mice and examined whether the androgen receptor (AR) was involved. Mice given either AAS cocktail or the cocktail and the AR antagonist, flutamide, for 6 weeks were subject to three weekly tests in which the percentage of mice that fought, the latency to initiate an aggressive event and the number of aggressive events per 5-min-fight session were recorded. In C57BL/6J mice, 6 weeks of AAS administration increased the likelihood of fighting, however, within the subset of mice that engaged in aggression, AAS did not specifically modulate the latency to fight or the number of aggressive events per fight. In addition, co-administration of flutamide only slightly altered the likelihood that mice given AAS will initiate a fight. Experiment 2 examined the aggression-promoting effects of AAS in gonadally-intact adult testicular feminization mutant (Tfm) mice, which are deficient in functional ARs. Overall, fewer Tfm mice compared to C57BL/6J mice fought in both drug conditions (vehicle or AAS). Taken together, these data suggest that given the presence of AR during development, AAS enhance adult male aggression in C57BL/6J mice through AR-independent and AR-dependent pathways. In contrast, in adult Tfm mice, the likelihood of AAS-enhanced aggression in adulthood is significantly reduced.


Subject(s)
Aggression/drug effects , Anabolic Agents/pharmacology , Androgen-Insensitivity Syndrome/psychology , Receptors, Androgen/physiology , Androgen Receptor Antagonists/pharmacology , Androgen-Insensitivity Syndrome/drug therapy , Animals , Female , Flutamide/pharmacology , Male , Mice , Mice, Inbred C57BL
6.
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
7.
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
8.
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
9.
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
10.
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
11.
Article in English | MEDLINE | ID: mdl-16920403

ABSTRACT

Amino metabolites with potential prooxidant properties, particularly alpha-aminocarbonyls, are the focus of this review. Among them we emphasize 5-aminolevulinic acid (a heme precursor formed from succinyl-CoA and glycine), aminoacetone (a threonine and glycine metabolite), and hexosamines and hexosimines, formed by Schiff condensation of hexoses with basic amino acid residues of proteins. All these metabolites were shown, in vitro, to undergo enolization and subsequent aerobic oxidation, yielding oxyradicals and highly cyto- and genotoxic alpha-oxoaldehydes. Their metabolic roles in health and disease are examined here and compared in humans and experimental animals, including rats, quail, and octopus. In the past two decades, we have concentrated on two endogenous alpha-aminoketones: (i) 5-aminolevulinic acid (ALA), accumulated in acquired (e.g., lead poisoning) and inborn (e.g., intermittent acute porphyria) porphyric disorders, and (ii) aminoacetone (AA), putatively overproduced in diabetes mellitus and cri-du-chat syndrome. ALA and AA have been implicated as contributing sources of oxyradicals and oxidative stress in these diseases. The end product of ALA oxidation, 4,5-dioxovaleric acid (DOVA), is able to alkylate DNA guanine moieties, promote protein cross-linking, and damage GABAergic receptors of rat brain synaptosome preparations. In turn, methylglyoxal (MG), the end product of AA oxidation, is also highly cytotoxic and able to release iron from ferritin and copper from ceruloplasmin, and to aggregate proteins. This review covers chemical and biochemical aspects of these alpha-aminoketones and their putative roles in the oxidative stress associated with porphyrias, tyrosinosis, diabetes, and cri-du-chat. In addition, we comment briefly on a side prooxidant behaviour of hexosamines, that are known to constitute building blocks of several glycoproteins and to be involved in Schiff base-mediated enzymatic reactions.


Subject(s)
Acetone/analogs & derivatives , Aminolevulinic Acid/metabolism , Hexosamines/metabolism , Imino Sugars/metabolism , Oxidants/metabolism , Acetone/metabolism , Animals , Cri-du-Chat Syndrome/metabolism , Diabetes Mellitus/metabolism , Humans , Lead Poisoning/metabolism , Oxidation-Reduction , Oxidative Stress/physiology , Porphyria, Acute Intermittent/metabolism , Rats
12.
Brain Res ; 1126(1): 122-38, 2006 Dec 18.
Article in English | MEDLINE | ID: mdl-17010954

ABSTRACT

Illicit use of anabolic androgenic steroids (AAS) has become a prevalent health concern not only among male professional athletes, but, disturbingly, among a growing number of women and adolescent girls. Despite the increasing use of AAS among women and adolescents, few studies have focused on the effects of these steroids in females, and female adolescent subjects are particularly underrepresented. Among the hallmarks of AAS abuse are changes in reproductive behaviors. Here, we discuss work from our laboratories on the actions of AAS on the onset of puberty and sexual behaviors in female rodents, AAS interactions and sex- and age-specific effects of these steroids on neural transmission mediated by gamma-aminobutyric acid receptors within forebrain neuroendocrine control regions that may underlie AAS-induced changes in these behaviors.


Subject(s)
Neurosecretory Systems/drug effects , Prosencephalon/drug effects , Reproduction/drug effects , Sexual Behavior, Animal/drug effects , Testosterone Congeners/adverse effects , gamma-Aminobutyric Acid/metabolism , Age Factors , Animals , Female , Humans , Neurosecretory Systems/growth & development , Neurosecretory Systems/metabolism , Prosencephalon/growth & development , Prosencephalon/metabolism , Puberty/drug effects , Puberty/metabolism , Receptors, GABA-A/drug effects , Receptors, GABA-A/metabolism , Reproduction/physiology , Sex Differentiation/drug effects , Sex Differentiation/physiology , Sexual Behavior, Animal/physiology
13.
Brain Res ; 1093(1): 95-104, 2006 Jun 06.
Article in English | MEDLINE | ID: mdl-16701578

ABSTRACT

Porphyrias are heme-associated metabolic disorders such as intermittent acute porphyria (IAP) and lead poisoning, where 5-aminolevulinate (ALA) accumulates. Effects of ALA on the CNS have been explained by ALA binding to GABA(A) receptors, followed by receptor lesions from oxyradicals and 4, 5-dioxovalerate (DOVA) generated from metal-catalyzed ALA oxidation by oxygen. We have characterized the effects of ALA and DOVA on GABA(A) receptor density in synaptosomes and neurons in vitro and also in brains of rats treated with ALA or succinylacetone methyl ester (SAME), a tyrosine catabolite derivative able to induce ALA accumulation. Radiolabeling assays revealed that following exposure to DOVA the concentration of synaptosomal GABAergic sites decreased by approximately 50%. Pretreatment with DOVA resulted in less GABA(A) receptor density in P19 and WERI cells and altered cell morphology. Furthermore, exposure to DOVA also induced a 5-fold increase in WERI cell mortality rate. Treatment with ALA resulted in loss of neuronal morphology and decrease of GABA(A) density in P19 neuronal cells. ALA and SAME treatment diminished the density of GABAergic receptors in the habenular complex and the parabigeminal nucleus of rat brain as studied by immunohistochemical procedures. Our results strongly suggest that ALA- and DOVA-promoted damage to GABA(A) receptors may contribute to the neurological manifestations of AIP and plumbism.


Subject(s)
Aminolevulinic Acid/toxicity , Brain/drug effects , Neurons/drug effects , Receptors, GABA-A/drug effects , Synaptosomes/drug effects , Valerates/toxicity , Aminolevulinic Acid/metabolism , Animals , Brain/metabolism , Cell Line , Cell Survival/drug effects , Humans , Immunohistochemistry , Ions , Male , Neurons/metabolism , Porphyrias/physiopathology , Rats , Rats, Wistar , Receptors, GABA-A/metabolism , Synaptosomes/metabolism , Valerates/metabolism
14.
Ciênc. cult. (Säo Paulo) ; 47(5/6): 346-57, Sept.-Oct. 1995. ilus
Article in English | LILACS | ID: lil-186431

ABSTRACT

Highly reactive oxyradicals and electronically excited triplet carbonyls can be generated in vitro by iron complexes and heme enzyme-catalyzed aerobic oxidation of synthetic or naturally occurring substances capable of enolization in aqueous medium. Monoenols and enamines, obtained by (alpha-methyne-carbonyl and -imine enolization, undergo dioxygen insertion and ultimately originate triplet species; e.g., isobutanal, 3-methylacetoacetone, Schiff bases. In turn, (alpha-hydroxy- and (alpha-aminocarbonyls (e.g., carbohydrates, 5-aminolevulinic acid) tautomerize to enediols and enolamines and yield oxyradicals, initiated by electron transfer to dioxygen, as polyphenols (e.g., 6-hydroxydopamine) and polyphenolamines do. Free radicals and excited species have been implicated in several normal and pathological processes. We here briefly review our contributions to this research area, emphasizing a possible in vivo prooxidant role for 5-aminolevulinic acid, the heme precursor accumulated in several porphyric disorders (e.g., lead poisoning, acut intermittent porphyria, tyrosinosis).


Subject(s)
Animals , Humans , Aminolevulinic Acid/chemistry , In Vitro Techniques , Reactive Oxygen Species , Free Radicals , Imino Acids/metabolism , Iron/metabolism , Lead/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...