Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Pathol ; 245(1): 85-100, 2018 05.
Article in English | MEDLINE | ID: mdl-29435980

ABSTRACT

Alzheimer's disease (AD) is a devastating neurological disorder that still lacks an effective treatment, and this has stimulated an intense pursuit of disease-modifying therapeutics. Given the increasingly recognized link between AD and defective brain insulin signaling, we investigated the actions of liraglutide, a glucagon-like peptide-1 (GLP-1) analog marketed for treatment of type 2 diabetes, in experimental models of AD. Insulin receptor pathology is an important feature of AD brains that impairs the neuroprotective actions of central insulin signaling. Here, we show that liraglutide prevented the loss of brain insulin receptors and synapses, and reversed memory impairment induced by AD-linked amyloid-ß oligomers (AßOs) in mice. Using hippocampal neuronal cultures, we determined that the mechanism of neuroprotection by liraglutide involves activation of the PKA signaling pathway. Infusion of AßOs into the lateral cerebral ventricle of non-human primates (NHPs) led to marked loss of insulin receptors and synapses in brain regions related to memory. Systemic treatment of NHPs with liraglutide provided partial protection, decreasing AD-related insulin receptor, synaptic, and tau pathology in specific brain regions. Synapse damage and elimination are amongst the earliest known pathological changes and the best correlates of memory impairment in AD. The results illuminate mechanisms of neuroprotection by liraglutide, and indicate that GLP-1 receptor activation may be harnessed to protect brain insulin receptors and synapses in AD. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Subject(s)
Cognitive Dysfunction/drug therapy , Liraglutide/pharmacology , Memory/drug effects , Receptor, Insulin/drug effects , Synapses/pathology , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Disease Models, Animal , Hippocampus/drug effects , Hypoglycemic Agents/pharmacology , Male , Mice , Receptor, Insulin/metabolism , Synapses/drug effects
2.
J Biol Chem ; 292(18): 7327-7337, 2017 05 05.
Article in English | MEDLINE | ID: mdl-28283575

ABSTRACT

Brain accumulation of the amyloid-ß protein (Aß) and synapse loss are neuropathological hallmarks of Alzheimer disease (AD). Aß oligomers (AßOs) are synaptotoxins that build up in the brains of patients and are thought to contribute to memory impairment in AD. Thus, identification of novel synaptic components that are targeted by AßOs may contribute to the elucidation of disease-relevant mechanisms. Trans-synaptic interactions between neurexins (Nrxs) and neuroligins (NLs) are essential for synapse structure, stability, and function, and reduced NL levels have been associated recently with AD. Here we investigated whether the interaction of AßOs with Nrxs or NLs mediates synapse damage and cognitive impairment in AD models. We found that AßOs interact with different isoforms of Nrx and NL, including Nrx2α and NL1. Anti-Nrx2α and anti-NL1 antibodies reduced AßO binding to hippocampal neurons and prevented AßO-induced neuronal oxidative stress and synapse loss. Anti-Nrx2α and anti-NL1 antibodies further blocked memory impairment induced by AßOs in mice. The results indicate that Nrx2α and NL1 are targets of AßOs and that prevention of this interaction reduces the deleterious impact of AßOs on synapses and cognition. Identification of Nrx2α and NL1 as synaptic components that interact with AßOs may pave the way for development of novel approaches aimed at halting synapse failure and cognitive loss in AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Brain/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Nerve Tissue Proteins/metabolism , Peptide Fragments/metabolism , Protein Aggregation, Pathological/metabolism , Synapses/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/genetics , Animals , Brain/pathology , Cell Adhesion Molecules, Neuronal/genetics , Cells, Cultured , Disease Models, Animal , Humans , Male , Mice , Nerve Tissue Proteins/genetics , Peptide Fragments/genetics , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/pathology , Rats , Rats, Wistar , Synapses/genetics
3.
Front Cell Neurosci ; 9: 97, 2015.
Article in English | MEDLINE | ID: mdl-25904842

ABSTRACT

Parkinson's disease (PD) is an incurable progressive neurodegenerative disorder. Clinical presentation of PD stems largely from the loss of dopaminergic neurons in the nigrostriatal dopaminergic pathway, motivating experimental strategies of replacement based on cell therapy. Transplantation of dopaminergic neurons derived from embryonic stem cells significantly improves motor functions in rodent and non-human primate models of PD. However, protocols to generate dopaminergic neurons from embryonic stem cells generally meet with low efficacy and high risk of teratoma formation upon transplantation. To address these issues, we have pre-treated undifferentiated mouse embryonic stem cells (mESCs) with the DNA alkylating agent mitomycin C (MMC) before transplantation. MMC treatment of cultures prevented tumorigenesis in a 12 week follow-up after mESCs were injected in nude mice. In 6-OH-dopamine-lesioned mice, intrastriatal injection of MMC-treated mESCs markedly improved motor function without tumor formation for as long as 15 months. Furthermore, we show that halting mitotic activity of undifferentiated mESCs induces a four-fold increase in dopamine release following in vitro differentiation. Our findings indicate that treating mESCs with MMC prior to intrastriatal transplant is an effective to strategy that could be further investigated as a novel alternative for treatment of PD.

4.
J Neurosci ; 34(41): 13629-43, 2014 Oct 08.
Article in English | MEDLINE | ID: mdl-25297091

ABSTRACT

Alzheimer's disease (AD) is a devastating neurodegenerative disorder and a major medical problem. Here, we have investigated the impact of amyloid-ß (Aß) oligomers, AD-related neurotoxins, in the brains of rats and adult nonhuman primates (cynomolgus macaques). Soluble Aß oligomers are known to accumulate in the brains of AD patients and correlate with disease-associated cognitive dysfunction. When injected into the lateral ventricle of rats and macaques, Aß oligomers diffused into the brain and accumulated in several regions associated with memory and cognitive functions. Cardinal features of AD pathology, including synapse loss, tau hyperphosphorylation, astrocyte and microglial activation, were observed in regions of the macaque brain where Aß oligomers were abundantly detected. Most importantly, oligomer injections induced AD-type neurofibrillary tangle formation in the macaque brain. These outcomes were specifically associated with Aß oligomers, as fibrillar amyloid deposits were not detected in oligomer-injected brains. Human and macaque brains share significant similarities in terms of overall architecture and functional networks. Thus, generation of a macaque model of AD that links Aß oligomers to tau and synaptic pathology has the potential to greatly advance our understanding of mechanisms centrally implicated in AD pathogenesis. Furthermore, development of disease-modifying therapeutics for AD has been hampered by the difficulty in translating therapies that work in rodents to humans. This new approach may be a highly relevant nonhuman primate model for testing therapeutic interventions for AD.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/toxicity , Peptide Fragments/toxicity , Alzheimer Disease/chemically induced , Amyloid beta-Peptides/administration & dosage , Animals , Apoptosis/drug effects , Astrocytes/pathology , Injections, Intraventricular , Macaca fascicularis , Male , Microglia/pathology , Microinjections , Neurofibrillary Tangles/pathology , Peptide Fragments/administration & dosage , Rats , Rats, Wistar , Synapses/pathology , Synapses/physiology , Synapses/ultrastructure
5.
Cell Metab ; 18(6): 831-43, 2013 Dec 03.
Article in English | MEDLINE | ID: mdl-24315369

ABSTRACT

Alzheimer's disease (AD) and type 2 diabetes appear to share similar pathogenic mechanisms. dsRNA-dependent protein kinase (PKR) underlies peripheral insulin resistance in metabolic disorders. PKR phosphorylates eukaryotic translation initiation factor 2α (eIF2α-P), and AD brains exhibit elevated phospho-PKR and eIF2α-P levels. Whether and how PKR and eIF2α-P participate in defective brain insulin signaling and cognitive impairment in AD are unknown. We report that ß-amyloid oligomers, AD-associated toxins, activate PKR in a tumor necrosis factor α (TNF-α)-dependent manner, resulting in eIF2α-P, neuronal insulin receptor substrate (IRS-1) inhibition, synapse loss, and memory impairment. Brain phospho-PKR and eIF2α-P were elevated in AD animal models, including monkeys given intracerebroventricular oligomer infusions. Oligomers failed to trigger eIF2α-P and cognitive impairment in PKR(-/-) and TNFR1(-/-) mice. Bolstering insulin signaling rescued phospho-PKR and eIF2α-P. Results reveal pathogenic mechanisms shared by AD and diabetes and establish that proinflammatory signaling mediates oligomer-induced IRS-1 inhibition and PKR-dependent synapse and memory loss.


Subject(s)
Amyloid beta-Peptides/toxicity , Brain/drug effects , Insulin Receptor Substrate Proteins/metabolism , Polymers/toxicity , Tumor Necrosis Factor-alpha/metabolism , eIF-2 Kinase/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/chemistry , Animals , Brain/metabolism , Disease Models, Animal , Haplorhini/metabolism , Hypoglycemic Agents/pharmacology , Insulin Receptor Substrate Proteins/antagonists & inhibitors , Memory Disorders/metabolism , Memory Disorders/pathology , Mice , Mice, Knockout , Neurons/drug effects , Neurons/metabolism , Phosphorylation/drug effects , Polymers/chemistry , Receptors, Tumor Necrosis Factor, Type I/deficiency , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction/drug effects , Synapses/drug effects , Synapses/metabolism , Tumor Necrosis Factor-alpha/antagonists & inhibitors , eIF-2 Kinase/deficiency , eIF-2 Kinase/genetics
6.
J Neurochem ; 126(2): 191-202, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23668663

ABSTRACT

Alzheimer's disease (AD) is the most common form of dementia in the elderly. Memory loss in AD is increasingly attributed to soluble oligomers of the amyloid-ß peptide (AßOs), toxins that accumulate in AD brains and target particular synapses. Glutamate receptors appear to be centrally involved in synaptic targeting by AßOs. Once bound to neurons, AßOs dysregulate the activity and reduce the surface expression of both N-methyl-D-aspartate (NMDA) and 2-amino-3-(3-hydroxy-5-methyl-isoxazol-4-yl)propanoic acid (AMPA) types of glutamate receptors, impairing signaling pathways involved in synaptic plasticity. In the extracellular milieu, AßOs promote accumulation of the excitatory amino acids, glutamate and D-serine. This leads to overactivation of glutamate receptors, triggering abnormal calcium signals with noxious impacts on neurons. Here, we review key findings linking AßOs to deregulated glutamate neurotransmission and implicating this as a primary mechanism of synapse failure in AD. We also discuss strategies to counteract the impact of AßOs on excitatory neurotransmission. In particular, we review evidence showing that inducing neuronal hyperpolarization via activation of inhibitory GABA(A) receptors prevents AßO-induced excitotoxicity, suggesting that this could comprise a possible therapeutic approach in AD.


Subject(s)
Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Synapses/metabolism , Synaptic Transmission/physiology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/metabolism , Brain/pathology , Humans , Long-Term Potentiation/physiology , Receptors, Glutamate/metabolism
7.
J Clin Invest ; 122(4): 1339-53, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22476196

ABSTRACT

Defective brain insulin signaling has been suggested to contribute to the cognitive deficits in patients with Alzheimer's disease (AD). Although a connection between AD and diabetes has been suggested, a major unknown is the mechanism(s) by which insulin resistance in the brain arises in individuals with AD. Here, we show that serine phosphorylation of IRS-1 (IRS-1pSer) is common to both diseases. Brain tissue from humans with AD had elevated levels of IRS-1pSer and activated JNK, analogous to what occurs in peripheral tissue in patients with diabetes. We found that amyloid-ß peptide (Aß) oligomers, synaptotoxins that accumulate in the brains of AD patients, activated the JNK/TNF-α pathway, induced IRS-1 phosphorylation at multiple serine residues, and inhibited physiological IRS-1pTyr in mature cultured hippocampal neurons. Impaired IRS-1 signaling was also present in the hippocampi of Tg mice with a brain condition that models AD. Importantly, intracerebroventricular injection of Aß oligomers triggered hippocampal IRS-1pSer and JNK activation in cynomolgus monkeys. The oligomer-induced neuronal pathologies observed in vitro, including impaired axonal transport, were prevented by exposure to exendin-4 (exenatide), an anti-diabetes agent. In Tg mice, exendin-4 decreased levels of hippocampal IRS-1pSer and activated JNK and improved behavioral measures of cognition. By establishing molecular links between the dysregulated insulin signaling in AD and diabetes, our results open avenues for the investigation of new therapeutics in AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/toxicity , Hippocampus/drug effects , Hypoglycemic Agents/therapeutic use , Insulin Receptor Substrate Proteins/metabolism , Insulin Resistance , Insulin/physiology , Peptides/therapeutic use , Venoms/therapeutic use , Aged , Aged, 80 and over , Alzheimer Disease/genetics , Alzheimer Disease/prevention & control , Alzheimer Disease/psychology , Animals , Antibodies, Monoclonal/pharmacology , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Exenatide , Female , Hippocampus/cytology , Hippocampus/metabolism , Hippocampus/pathology , Humans , Hypoglycemic Agents/pharmacology , Infliximab , MAP Kinase Signaling System/drug effects , Macaca fascicularis , Male , Maze Learning/drug effects , Memory Disorders/etiology , Memory Disorders/metabolism , Memory Disorders/prevention & control , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Neurons/drug effects , Neurons/metabolism , Peptides/pharmacology , Phosphorylation , Protein Processing, Post-Translational , Rats , Venoms/pharmacology
8.
J Biol Chem ; 287(23): 19377-85, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22505713

ABSTRACT

Dysregulated cholinergic signaling is an early hallmark of Alzheimer disease (AD), usually ascribed to degeneration of cholinergic neurons induced by the amyloid-ß peptide (Aß). It is now generally accepted that neuronal dysfunction and memory deficits in the early stages of AD are caused by the neuronal impact of soluble Aß oligomers (AßOs). AßOs build up in AD brain and specifically attach to excitatory synapses, leading to synapse dysfunction. Here, we have investigated the possibility that AßOs could impact cholinergic signaling. The activity of choline acetyltransferase (ChAT, the enzyme that carries out ACh production) was inhibited by ~50% in cultured cholinergic neurons exposed to low nanomolar concentrations of AßOs. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) reduction, lactate dehydrogenase release, and [(3)H]choline uptake assays showed no evidence of neuronal damage or loss of viability that could account for reduced ChAT activity under these conditions. Glutamate receptor antagonists fully blocked ChAT inhibition and oxidative stress induced by AßOs. Antioxidant polyunsaturated fatty acids had similar effects, indicating that oxidative damage may be involved in ChAT inhibition. Treatment with insulin, previously shown to down-regulate neuronal AßO binding sites, fully prevented AßO-induced inhibition of ChAT. Interestingly, we found that AßOs selectively bind to ~50% of cultured cholinergic neurons, suggesting that ChAT is fully inhibited in AßO-targeted neurons. Reduction in ChAT activity instigated by AßOs may thus be a relevant event in early stage AD pathology, preceding the loss of cholinergic neurons commonly observed in AD brains.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Avian Proteins/metabolism , Choline O-Acetyltransferase/metabolism , Neurons/metabolism , Oxidative Stress , Alzheimer Disease/pathology , Animals , Antioxidants/metabolism , Avian Proteins/genetics , Cell Culture Techniques , Cells, Cultured , Chickens , Excitatory Amino Acid Antagonists/pharmacology , Neurons/pathology
9.
J Biol Chem ; 286(5): 3270-6, 2011 Feb 04.
Article in English | MEDLINE | ID: mdl-21115476

ABSTRACT

Soluble oligomers of the amyloid-ß peptide (AßOs) accumulate in the brains of Alzheimer disease (AD) patients and are implicated in synapse failure and early memory loss in AD. AßOs have been shown to impact synapse function by inhibiting long term potentiation, facilitating the induction of long term depression and inducing internalization of both AMPA and NMDA glutamate receptors, critical players in plasticity mechanisms. Because activation of dopamine D1/D5 receptors plays important roles in memory circuits by increasing the insertion of AMPA and NMDA receptors at synapses, we hypothesized that selective activation of D1/D5 receptors could protect synapses from the deleterious action of AßOs. We show that SKF81297, a selective D1/D5 receptor agonist, prevented the reduction in surface levels of AMPA and NMDA receptors induced by AßOs in hippocampal neurons in culture. Protection by SKF81297 was abrogated by the specific D1/D5 antagonist, SCH23390. Levels of AMPA receptor subunit GluR1 phosphorylated at Ser(845), which regulates AMPA receptor association with the plasma membrane, were reduced in a calcineurin-dependent manner in the presence of AßOs, and treatment with SKF81297 prevented this reduction. Establishing the functional relevance of these findings, SKF81297 blocked the impairment of long term potentiation induced by AßOs in hippocampal slices. Results suggest that D1/D5 receptors may be relevant targets for development of novel pharmacological approaches to prevent synapse failure in AD.


Subject(s)
Amyloid beta-Peptides/pharmacology , Neurons , Receptors, Dopamine D1/physiology , Receptors, Dopamine D5/physiology , Synapses/pathology , Animals , Benzazepines , Cells, Cultured , Hippocampus , Humans , Memory , Protective Agents , Rats , Receptors, AMPA/metabolism , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D5/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism
10.
J Neurochem ; 115(6): 1520-9, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20950339

ABSTRACT

Soluble amyloid-ß peptide (Aß) oligomers, known to accumulate in Alzheimer's disease brains, target excitatory post-synaptic terminals. This is thought to trigger synapse deterioration, a mechanism possibly underlying memory loss in early stage Alzheimer's disease. A major unknown is the identity of the receptor(s) targeted by oligomers at synapses. Because oligomers have been shown to interfere with N-methyl-d-aspartate receptor (NMDAR) function and trafficking, we hypothesized that NMDARs might be required for oligomer binding to synapses. An amplicon vector was used to knock-down NMDARs in mature hippocampal neurons in culture, yielding 90% reduction in dendritic NMDAR expression and blocking neuronal oxidative stress induced by Aß oligomers, a pathological response that has been shown to be mediated by NMDARs. Remarkably, NMDAR knock-down abolished oligomer binding to dendrites, indicating that NMDARs are required for synaptic targeting of oligomers. Nevertheless, oligomers do not appear to bind directly to NMDARs as indicated by the fact that both oligomer-attacked and non-attacked neurons exhibit similar surface levels of NMDARs. Furthermore, pre-treatment of neurons with insulin down-regulates oligomer-binding sites in the absence of a parallel reduction in surface levels of NMDARs. Establishing that NMDARs are key components of the synaptic oligomer binding complex may illuminate the development of novel approaches to prevent synapse failure triggered by Aß oligomers.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Peptide Fragments/metabolism , Receptors, N-Methyl-D-Aspartate/physiology , Synapses/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/toxicity , Animals , Cells, Cultured , Gene Knockdown Techniques , Hippocampus/metabolism , Hippocampus/pathology , Peptide Fragments/chemistry , Peptide Fragments/toxicity , Protein Binding/physiology , Rats , Receptors, N-Methyl-D-Aspartate/deficiency , Synapses/pathology
11.
Int J Biochem Cell Biol ; 41(6): 1361-70, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19130896

ABSTRACT

Aggregates of the amyloid-beta peptide (Abeta) play a central role in the pathogenesis of Alzheimer's disease (AD). Identification of proteins that physiologically bind Abeta and modulate its aggregation and neurotoxicity could lead to the development of novel disease-modifying approaches in AD. By screening a phage display peptide library for high affinity ligands of aggregated Abeta(1-42), we isolated a peptide homologous to a highly conserved amino acid sequence present in the N-terminus of apolipoprotein A-I (apoA-I). We show that purified human apoA-I and Abeta form non-covalent complexes and that interaction with apoA-I affects the morphology of amyloid aggregates formed by Abeta. Significantly, Abeta/apoA-I complexes were also detected in cerebrospinal fluid from AD patients. Interestingly, apoA-I and apoA-I-containing reconstituted high density lipoprotein particles protect hippocampal neuronal cultures from Abeta-induced oxidative stress and neurodegeneration. These results suggest that human apoA-I modulates Abeta aggregation and Abeta-induced neuronal damage and that the Abeta-binding domain in apoA-I may constitute a novel framework for the design of inhibitors of Abeta toxicity.


Subject(s)
Amyloid beta-Peptides/metabolism , Apolipoprotein A-I/metabolism , Peptide Fragments/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amino Acid Sequence , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/toxicity , Animals , Apolipoprotein A-I/genetics , Electrophoresis, Polyacrylamide Gel , Female , Humans , Neurons/metabolism , Neurons/pathology , Oxidative Stress , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/toxicity , Pregnancy , Rats , Rats, Sprague-Dawley
12.
Neuropharmacology ; 49(8): 1140-8, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16150468

ABSTRACT

The beta-amyloid peptide (Abeta) is centrally related to the pathogenesis of Alzheimer's disease (AD) and is potently neurotoxic to central nervous system neurons. The neurotoxicity of Abeta has been partially related to the over activation of glutamatergic transmission and excitotoxicity. Taurine is a naturally occurring beta-amino acid present in the mammalian brain. Due to its safety and tolerability, taurine has been clinically used in humans in the treatment of a number of non-neurological disorders. Here, we show that micromolar doses of taurine block the neurotoxicity of Abeta to rat hippocampal and cortical neurons in culture. Moreover, taurine also rescues central neurons from the excitotoxicity induced by high concentrations of extracellular glutamate. Neuroprotection by taurine is abrogated by picrotoxin, a GABA(A) receptor antagonist. GABA and muscimol, an agonist of the GABA(A) receptor, also block neuronal death induced by Abeta in rat hippocampal and cortical neurons. These results suggest that activation of GABA(A) receptors protects neurons against Abeta toxicity in AD-affected regions of the mammalian brain and that taurine should be investigated as a novel therapeutic tool in the treatment of AD and of other neurological disorders in which excitotoxicity plays a relevant role.


Subject(s)
Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/toxicity , Cerebral Cortex/cytology , GABA Agonists/pharmacology , GABA-A Receptor Agonists , Hippocampus/drug effects , Muscimol/pharmacology , Neurons/drug effects , Taurine/pharmacology , Animals , Cell Survival , Cells, Cultured , Cerebral Cortex/drug effects , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...