Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Curr Neuropharmacol ; 17(5): 459-471, 2019.
Article in English | MEDLINE | ID: mdl-29956632

ABSTRACT

BACKGROUND: In women, changes in estrogen levels may increase the incidence and/or symptomatology of depression and affect the response to antidepressant treatments. Estrogen therapy in females may provide some mood benefits as a single treatment or might augment clinical response to antidepressants that inhibit serotonin reuptake. OBJECTIVE: We analyzed the mechanisms of estradiol action involved in the regulation of gene expression that modulates serotonin neurotransmission implicated in depression. METHOD: Publications were identified by a literature search on PubMed. RESULTS: The participation of estradiol in depression may include regulation of the expression of tryptophan hydroxylase-2, monoamine oxidase A and B, serotonin transporter and serotonin-1A receptor. This effect is mediated by estradiol binding to intracellular estrogen receptor that interacts with estrogen response elements in the promoter sequences of tryptophan hydroxylase-2, serotonin transporter and monoamine oxidase-B. In addition to directly binding deoxyribonucleic acid, estrogen receptor can tether to other transcription factors, including activator protein 1, specificity protein 1, CCAAT/enhancer binding protein ß and nuclear factor kappa B to regulate gene promoters that lack estrogen response elements, such as monoamine oxidase-A and serotonin 1A receptor. CONCLUSION: Estradiol increases tryptophan hydroxylase-2 and serotonin transporter expression and decreases the expression of serotonin 1A receptor and monoamine oxidase A and B through the interaction with its intracellular receptors. The understanding of molecular mechanisms of estradiol regulation on the protein expression that modulates serotonin neurotransmission will be helpful for the development of new and more effective treatment for women with depression.


Subject(s)
Depression/physiopathology , Estradiol/physiology , Gene Expression Regulation/physiology , Serotonergic Neurons/physiology , Animals , Depression/genetics , Depression/metabolism , Estradiol/metabolism , Estradiol/pharmacology , Female , Gene Expression Regulation/drug effects , Humans , Serotonergic Neurons/drug effects , Serotonergic Neurons/enzymology , Serotonergic Neurons/metabolism , Serotonin/physiology , Serotonin Plasma Membrane Transport Proteins/metabolism , Synaptic Transmission/drug effects , Tryptophan Hydroxylase/metabolism
2.
World J Gastroenterol ; 22(19): 4662-72, 2016 May 21.
Article in English | MEDLINE | ID: mdl-27217698

ABSTRACT

AIM: To determine if expression of colonic tryptophan hydroxylase-2 (TPH2), a surrogate marker of neuronal 5-hydroxytryptamine, is altered in Hirschsprung's-associated enterocolitis. METHODS: Entire resected colonic specimens were collected at the time of pull-through operation in children with Hirschsprung's disease (HSCR, n = 12). Five of these patients had a history of pre-operative Hirschsprung's-associated enterocolitis (HAEC). Controls were collected at colostomy closure in children with anorectal malformation (n = 10). The distribution of expression of TPH2 was evaluated using immunofluorescence and confocal microscopy. Protein expression of TPH2 was quantified using western blot analysis in the deep smooth muscle layers. RESULTS: TPH2 was co-expressed in nitrergic and cholinergic ganglia in the myenteric and submucosal plexuses in ganglionic colon in HSCR and healthy controls. Co-expression was also seen in submucosal interstitial cells of Cajal and PDGFRα(+) cells. The density of TPH2 immuno-positive fibers decreased incrementally from ganglionic bowel to transition zone bowel to aganglionic bowel in the myenteric plexus. Expression of TPH2 was reduced in ganglionic bowel in those affected by pre-operative HAEC compared to those without HAEC and healthy controls. However, expression of TPH2 was similar or high compared to controls in the colons of children who had undergone diverting colostomy for medically refractory HAEC. CONCLUSION: Altered TPH2 expression in colonic serotonergic nerves of patients with HSCR complicated by HAEC may contribute to intestinal secretory and motor disturbances, including recurrent HAEC.


Subject(s)
Colon/innervation , Enteric Nervous System/enzymology , Enterocolitis/enzymology , Hirschsprung Disease/enzymology , Serotonergic Neurons/enzymology , Tryptophan Hydroxylase/analysis , Anoctamin-1 , Biomarkers/analysis , Blotting, Western , Case-Control Studies , Chloride Channels/analysis , Colon/pathology , Colon/surgery , Enteric Nervous System/pathology , Enteric Nervous System/physiopathology , Enterocolitis/pathology , Enterocolitis/physiopathology , Enterocolitis/surgery , Female , Fluorescent Antibody Technique , Hirschsprung Disease/pathology , Hirschsprung Disease/physiopathology , Hirschsprung Disease/surgery , Humans , Infant , Male , Microscopy, Confocal , Neoplasm Proteins/analysis , Receptors, Platelet-Derived Growth Factor/analysis
3.
Eur Neuropsychopharmacol ; 24(4): 595-607, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24139910

ABSTRACT

Exposure to prenatal stress (PS) can predispose individuals to the development of psychopathology later in life. We examined the effects of unpredictable chronic mild stress (CMS) exposure during adolescence on a background of PS in male and female Sprague-Dawley rats. PS induced more anxiety-like behavior in the elevated zero maze in both sexes, an effect that was normalized by subsequent exposure to CMS. Moreover, PS was associated with increased depression-like behavior in the forced swim test in males only. Conversely, sucrose intake was increased in PS males, whilst being decreased in females when consecutively exposed to PS and CMS. Hypothalamo-pituitary-adrenal (HPA) axis reactivity was affected in males only, with higher stress-induced plasma corticosterone levels after PS. Markedly, CMS normalized the effects of PS on elevated zero maze behavior as well as basal and stress-induced plasma corticosterone secretion. At the neurochemical level, both PS and CMS induced various sex-specific alterations in serotonin (5-HT) and tryptophan hydroxylase 2 (TPH2) immunoreactivity in the dorsal raphe nucleus, hippocampus and prefrontal cortex with, in line with the behavioral observations, more profound effects in male offspring. In conclusion, these findings show that prenatal maternal stress in Sprague-Dawley rats induces various anxiety- and depression-related behavioral and neuroendocrine changes, as well as alterations in central 5-HT and TPH2 function, predominantly in male offspring. Moreover, CMS exposure partially normalized the effects of previous PS experience, suggesting that the outcome of developmental stress exposure largely depends on the environmental conditions later in life and vice versa.


Subject(s)
Allostasis , Anxiety/etiology , Depression/etiology , Disease Models, Animal , Prenatal Exposure Delayed Effects/physiopathology , Serotonergic Neurons/metabolism , Stress, Physiological , Animals , Anxiety/blood , Anxiety/prevention & control , Behavior, Animal , Depression/blood , Depression/prevention & control , Female , Hippocampus/enzymology , Hippocampus/metabolism , Hippocampus/pathology , Hypothalamo-Hypophyseal System/metabolism , Hypothalamo-Hypophyseal System/physiopathology , Male , Nerve Tissue Proteins/metabolism , Pituitary-Adrenal System/metabolism , Pituitary-Adrenal System/physiopathology , Prefrontal Cortex/enzymology , Prefrontal Cortex/metabolism , Prefrontal Cortex/pathology , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/psychology , Raphe Nuclei/enzymology , Raphe Nuclei/metabolism , Raphe Nuclei/pathology , Rats , Rats, Sprague-Dawley , Serotonergic Neurons/enzymology , Serotonergic Neurons/pathology , Sex Characteristics , Tryptophan Hydroxylase/metabolism
4.
Brain Struct Funct ; 218(2): 563-73, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22532304

ABSTRACT

The effects of epigenetics on brain functions are not completely understood, but histone deacetylases (HDACs) are known to affect brain function and dysfunction by mediating the acetylation status of target proteins, thereby affecting gene expression. The current study used immunochemistry to illuminate the regional distribution of one member of the HDAC family, HDAC2, in the C57BL/6J mouse brain. Our data show that HDAC2 is ubiquitously expressed throughout the mouse brain and is localized primarily within the cell nucleus. Using double-immunofluorescence, we demonstrated HDAC2 expression in neuronal cells, including cholinergic, serotonergic and catecholaminergic neurons, as well as postsynaptic glutamatergic and GABAergic neurons. HDAC2 was also observed in oligodendrocytes, but not in astrocytes or microglia. These detailed immunological studies illuminate the distribution of HDAC2 throughout the mouse brain and will facilitate investigation of the roles of HDAC2 in brain function and neurological disorders.


Subject(s)
Brain/enzymology , Histone Deacetylase 2/analysis , Neurons/enzymology , Adrenergic Neurons/enzymology , Age Factors , Animals , Brain/cytology , Cell Nucleus/enzymology , Cholinergic Neurons/enzymology , GABAergic Neurons/enzymology , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Oligodendroglia/enzymology , Serotonergic Neurons/enzymology
5.
Brain Struct Funct ; 218(2): 587-93, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22527120

ABSTRACT

Recent research has shown that at least two tryptophan hydroxylase (Tph) genes are present in gnathostome vertebrates, but it is not known when the duplication of the ancestral Tph gene took place during evolution. By their position as an out-group of gnathostomes, lampreys (agnathans) are key models to understand molecular evolution in vertebrates. Here, we report the cloning of a Tph cDNA of the sea lamprey and the pattern of Tph mRNA expression in larval and postmetamorphic (young adult) sea lampreys using in situ hybridization. Phylogenetic analysis indicated that the lamprey Tph is an orthologue of Tphs of other vertebrates and suggested that the duplication of the ancestral Tph gene occurred before the separation of agnathans and gnathostomes, although alternative hypothesis are also discussed in the present study. In the sea lamprey brain, the Tph transcript was expressed in perikarya of the pineal organ, the retina, the diencephalic and rhombencephalic nuclei reported previously with serotonin immunohistochemistry and in small cells of the spinal cord, with a pattern similar to that observed with anti-serotonin antibodies. This suggests that expression of this Tph gene is shared by all lamprey serotonergic brain populations, unlike that reported in zebrafish and mammals for their different Tph genes. However, no Tph expression was observed in peripheral serotonergic cells, which, unlike in other vertebrates, are widely distributed in lampreys. Our results suggest that the selection of Tph2 to be expressed in raphe neurons may have occurred along the line leading to gnathostomes.


Subject(s)
Evolution, Molecular , Fish Proteins/genetics , Petromyzon/metabolism , Serotonergic Neurons/enzymology , Serotonin/metabolism , Tryptophan Hydroxylase/genetics , Animals , Biomarkers/metabolism , Cloning, Molecular , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Immunohistochemistry , In Situ Hybridization , Larva/enzymology , Petromyzon/embryology , Phylogeny , RNA, Messenger/metabolism
6.
Cell Metab ; 16(1): 113-21, 2012 Jul 03.
Article in English | MEDLINE | ID: mdl-22768843

ABSTRACT

Serotonergic regulation of feeding behavior has been studied intensively, both for an understanding of the basic neurocircuitry of energy balance in various organisms and as a therapeutic target for human obesity. However, its underlying molecular mechanisms remain poorly understood. Here, we show that neural serotonin signaling in C. elegans modulates feeding behavior through inhibition of AMP-activated kinase (AMPK) in interneurons expressing the C. elegans counterpart of human SIM1, a transcription factor associated with obesity. In turn, glutamatergic signaling links these interneurons to pharyngeal neurons implicated in feeding behavior. We show that AMPK-mediated regulation of glutamatergic release is conserved in rat hippocampal neurons. These findings reveal cellular and molecular mediators of serotonergic signaling.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/enzymology , Feeding Behavior , Glutamic Acid/metabolism , Protein Serine-Threonine Kinases/metabolism , Synaptic Transmission , AMP-Activated Protein Kinases , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Caenorhabditis elegans/cytology , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/genetics , Cells, Cultured , Chemoreceptor Cells/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Gastrointestinal Motility , Hippocampus/cytology , Pharynx/innervation , Pharynx/metabolism , Pharynx/physiology , Protein Serine-Threonine Kinases/genetics , Rats , Receptors, Serotonin/metabolism , Serotonergic Neurons/enzymology , Serotonergic Neurons/metabolism , Serotonin/metabolism
7.
Alcohol ; 46(7): 635-42, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22796363

ABSTRACT

Previous studies have demonstrated that alcohol use disorders (AUDs) are regulated by multiple mechanisms such as neurotransmitters and enzymes. The neurotransmitter, serotonin (5-hydroxytryptamine, 5-HT) may contribute to alcohol effects and serotonin receptors, including 5-HT3, play an important role in AUDs. Recent studies have also implicated histone deacetylases (HDACs) and acetyltransferases (HATS) in regulation of drug addiction, and HDAC inhibitors (HDACi) have been reported as transcriptional modulators of monoaminergic neurotransmission. Therefore, we hypothesize that HDACs may play a role in ethanol-induced serotonergic modulation. The effects of ethanol on serotonin and 5-HT3, and the role HDACs, HDAC activity and the HDACi, trichostatin A (TSA), play in alcohol-induced serotonergic effects were studied. Human SK-N-MC and neurons, were treated with ethanol (0.05, 0.1 and 0.2%), and/or TSA (50 nM), and 5-HT3 levels were assessed at 24-72 h. Gene expression was evaluated by qRT-PCR and protein by western blot and flow cytometry. Serotonin release was assessed by ELISA and HDAC activity by fluorometric assay. Our results show an increase in 5-HT3 gene after ethanol treatment. Further, ethanol significantly increased HDACs 1 and 3 genes accompanied by an increased in HDAC activity while TSA significantly inhibited HDACs. Studies with TSA show a significant upregulation of ethanol effects on 5-HT3, while surprisingly TSA inhibited ethanol-induced serotonin production. These results suggest that ethanol affects 5-HT3 and serotonin through mechanisms involving HDACs and HATs. In summary, our studies demonstrate some of the novel properties of HDAC inhibitors and contribute to the understanding of the mechanisms involve in alcohol-serotonergic modulation in the CNS.


Subject(s)
Ethanol/toxicity , Histone Deacetylases/metabolism , Serotonergic Neurons/drug effects , Blotting, Western , Cell Line , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fluorometry , Gene Expression Regulation, Enzymologic/drug effects , Histone Acetyltransferases/metabolism , Histone Deacetylase 1/genetics , Histone Deacetylase 1/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/genetics , Humans , Hydroxamic Acids/pharmacology , Ondansetron/pharmacology , Real-Time Polymerase Chain Reaction , Receptors, Serotonin, 5-HT3/drug effects , Receptors, Serotonin, 5-HT3/genetics , Receptors, Serotonin, 5-HT3/metabolism , Serotonergic Neurons/enzymology , Serotonin/metabolism , Serotonin 5-HT3 Receptor Antagonists/pharmacology , Time Factors , Up-Regulation
8.
Psychopharmacology (Berl) ; 222(4): 709-19, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22451092

ABSTRACT

RATIONALE: The transition to menopause is associated with an increased risk of depressed mood. OBJECTIVES: This study was conducted to investigate whether diphenyl diselenide [(PhSe)2] treatment could reduce the effects of postmenopausal depression-like behavior in ovariectomized female mice submitted to subchronic stress exposure. METHODS: Mice were divided into four groups: sham, (PhSe)2, ovariectomy (OVX), and OVX + (PhSe)2. Animals were ovariectomized/sham-operated and subjected to stress session once a day for 7 days from the fifth to the 11th day after OVX. The behavioral tests (open field, tail suspension (TST), and forced swimming (FST)) were performed on the 14th day after OVX. Mice were treated orally once a day with vehicle (canola oil, 10 ml/kg) or (PhSe)2 (10 mg/kg; 10 ml/kg) 30 min before being exposed to subchronic stress, or from the 11th to the 14th day. Paroxetine (8 mg/kg i.p.) and pargyline (30 mg/kg i.p.) were used as positive controls. The involvement of serotonergic receptor subtypes in the antidepressant-like effect of (PhSe)2 was assessed in the FST using WAY 100635 (0.1 mg/kg s.c.), ritanserin (1 mg/kg i.p.), and ondansetron (1 mg/kg i.p.) as serotonergic antagonists. Monoamine oxidase (MAO) A and B activities were also determined. RESULTS: The prolongation of immobility time in TST and FST in OVX mice submitted to subchronic stress was prevented by (PhSe)2 treatment. Ritanserin and ondansetron blocked the antidepressive-like effect of (PhSe)2, suggesting the involvement of 5-HT(2A/2C) and 5-HT3 receptor subtypes. Both paroxetine and pargyline were effective in reducing the immobility time of stressed OVX mice in the FST. No alterations in locomotor activity were observed. Although (PhSe)2 had inhibited in vitro MAO-A and MAO-B activities, none of the groups presented alterations neither in ex vivo MAO-A nor in MAO-B activity. CONCLUSIONS: (PhSe)2 treatment could influence mood and behavior, indicating a promising role of this organoselenium compound in the management of postmenopausal depressive symptoms.


Subject(s)
Benzene Derivatives/therapeutic use , Depression/enzymology , Menopause/physiology , Organoselenium Compounds/therapeutic use , Ovariectomy/psychology , Serotonergic Neurons/drug effects , Serotonergic Neurons/enzymology , Stress, Psychological/drug therapy , Animals , Antidepressive Agents/therapeutic use , Benzene Derivatives/antagonists & inhibitors , Benzene Derivatives/pharmacology , Cerebral Cortex/drug effects , Cerebral Cortex/enzymology , Depression/complications , Depression/drug therapy , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Female , Hippocampus/drug effects , Hippocampus/enzymology , Immobility Response, Tonic/drug effects , Mice , Mice, Inbred Strains , Mitochondria/enzymology , Monoamine Oxidase/metabolism , Monoamine Oxidase Inhibitors/pharmacology , Monoamine Oxidase Inhibitors/therapeutic use , Ondansetron/pharmacology , Organoselenium Compounds/antagonists & inhibitors , Organoselenium Compounds/pharmacology , Pargyline/pharmacology , Pargyline/therapeutic use , Paroxetine/pharmacology , Paroxetine/therapeutic use , Piperazines/pharmacology , Pyridines/pharmacology , Ritanserin , Serotonin Antagonists/pharmacology , Stress, Psychological/complications , Stress, Psychological/enzymology
9.
Mol Psychiatry ; 17(7): 694-704, 2012 Jul.
Article in English | MEDLINE | ID: mdl-21537332

ABSTRACT

Probably the foremost hypothesis of depression is the 5-hydroxytryptamine (5-HT, serotonin) deficiency hypothesis. Accordingly, anomalies in putative 5-HT biomarkers have repeatedly been reported in depression patients. However, whether such anomalies in fact reflect deficient central 5-HT neurotransmission remains unresolved. We employed a naturalistic model of 5-HT deficiency, the tryptophan hydroxylase 2 (Tph2) R439H knockin mouse, to address this question. We report that Tph2 knockin mice have reduced basal and stimulated levels of extracellular 5-HT (5-HT(Ext)). Interestingly, cerebrospinal fluid (CSF) 5-hydroxyindoleacetic acid (5-HIAA) and fenfluramine-induced plasma prolactin levels are markedly diminished in the Tph2 knockin mice. These data seemingly confirm that low CSF 5-HIAA and fenfluramine-induced plasma prolactin reflects chronic, endogenous central nervous system (CNS) 5-HT deficiency. Moreover, 5-HT(1A) receptor agonist-induced hypothermia is blunted and frontal cortex 5-HT(2A) receptors are increased in the Tph2 knockin mice. These data likewise parallel core findings in depression, but are usually attributed to anomalies in the respective receptors rather than resulting from CNS 5-HT deficiency. Further, 5-HT(2A) receptor function is enhanced in the Tph2 knockin mice. In contrast, 5-HT(1A) receptor levels and G-protein coupling is normal in Tph2 knockin mice, indicating that the blunted hypothermic response relates directly to the low 5-HT(Ext). Thus, we show that not only low CSF 5-HIAA and a blunted fenfluramine-induced prolactin response, but also blunted 5-HT(1A) agonist-induced hypothermia and increased 5-HT(2A) receptor levels are bona fide biomarkers of chronic, endogenous 5-HT deficiency. Potentially, some of these biomarkers could identify patients likely to have 5-HT deficiency. This could have clinical research utility or even guide pharmacotherapy.


Subject(s)
Depression/blood , Hydroxyindoleacetic Acid/cerebrospinal fluid , Receptor, Serotonin, 5-HT2A/metabolism , Serotonergic Neurons/physiology , Serotonin/deficiency , Synaptic Transmission/physiology , Tryptophan Hydroxylase/physiology , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Biomarkers/blood , Biomarkers/cerebrospinal fluid , Biomarkers/metabolism , Corticosterone/blood , Depression/cerebrospinal fluid , Depression/genetics , Disease Models, Animal , Extracellular Fluid/metabolism , Female , Fenfluramine/pharmacology , Frontal Lobe/metabolism , Gene Knock-In Techniques/methods , Gene Knock-In Techniques/psychology , Hippocampus/metabolism , Hypothermia/chemically induced , Hypothermia/physiopathology , Male , Mice , Mice, Inbred C57BL , Prolactin/blood , Receptor, Serotonin, 5-HT1A/genetics , Receptor, Serotonin, 5-HT1A/metabolism , Receptor, Serotonin, 5-HT2A/genetics , Serotonergic Neurons/drug effects , Serotonergic Neurons/enzymology , Serotonin/metabolism , Serotonin 5-HT1 Receptor Agonists/pharmacology , Synaptic Transmission/drug effects , Synaptic Transmission/genetics , Tryptophan Hydroxylase/genetics
10.
J Comp Neurol ; 520(5): 933-51, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-21830219

ABSTRACT

Monoaminergic innervation of the spinal cord has important modulatory functions for locomotion. Here we performed a quantitative study to determine the plastic changes of tyrosine hydroxylase-positive (TH1(+); mainly dopaminergic), and serotonergic (5-HT(+)) terminals and cells during successful spinal cord regeneration in adult zebrafish. TH1(+) innervation in the spinal cord is derived from the brain. After spinal cord transection, TH1(+) immunoreactivity is completely lost from the caudal spinal cord. Terminal varicosities increase in density rostral to the lesion site compared with unlesioned controls and are re-established in the caudal spinal cord at 6 weeks post lesion. Interestingly, axons mostly fail to re-innervate more caudal levels of the spinal cord even after prolonged survival times. However, densities of terminal varicosities correlate with recovery of swimming behavior, which is completely lost again after re-lesion of the spinal cord. Similar observations were made for terminals derived from descending 5-HT(+) axons from the brain. In addition, spinal 5-HT(+) neurons were newly generated after a lesion and transiently increased in number up to fivefold, which depended in part on hedgehog signaling. Overall, TH1(+) and 5-HT(+) innervation is massively altered in the successfully regenerated spinal cord of adult zebrafish. Despite these changes in TH and 5-HT systems, a remarkable recovery of swimming capability is achieved, suggesting significant plasticity of the adult spinal network during regeneration.


Subject(s)
Neuronal Plasticity/physiology , Serotonergic Neurons/metabolism , Spinal Cord Injuries/metabolism , Spinal Cord Regeneration , Tyrosine 3-Monooxygenase/metabolism , Age Factors , Animals , Animals, Genetically Modified , Serotonergic Neurons/enzymology , Serotonergic Neurons/pathology , Spinal Cord Injuries/enzymology , Spinal Cord Injuries/pathology , Spinal Cord Regeneration/genetics , Swimming/physiology , Tyrosine 3-Monooxygenase/genetics , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL
...