Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Genes (Basel) ; 14(10)2023 09 28.
Article in English | MEDLINE | ID: mdl-37895235

ABSTRACT

In the brain, cell adhesion molecules (CAMs) are critical for neurite outgrowth, axonal fasciculation, neuronal survival and migration, and synapse formation and maintenance. Among CAMs, the IgLON family comprises five members: Opioid Binding Protein/Cell Adhesion Molecule Like (OPCML or OBCAM), Limbic System Associated Membrane Protein (LSAMP), neurotrimin (NTM), Neuronal Growth Regulator 1 (NEGR1), and IgLON5. IgLONs exhibit three N-terminal C2 immunoglobulin domains; several glycosylation sites; and a glycosylphosphatidylinositol anchoring to the membrane. Interactions as homo- or heterodimers in cis and in trans, as well as binding to other molecules, appear critical for their functions. Shedding by metalloproteases generates soluble factors interacting with cellular receptors and activating signal transduction. The aim of this review was to analyse the available data implicating a role for IgLONs in neuropsychiatric disorders. Starting from the identification of a pathological role for antibodies against IgLON5 in an autoimmune neurodegenerative disease with a poorly understood mechanism of action, accumulating evidence links IgLONs to neuropsychiatric disorders, albeit with still undefined mechanisms which will require future thorough investigations.


Subject(s)
Neurodegenerative Diseases , Humans , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurons/metabolism , Cell Adhesion Molecules/metabolism , Immunoglobulins/genetics , Brain/metabolism , GPI-Linked Proteins/metabolism , Cell Adhesion Molecules, Neuronal/genetics
2.
Cells ; 12(9)2023 05 05.
Article in English | MEDLINE | ID: mdl-37174725

ABSTRACT

The metabotropic glutamate receptor 1 (mGlu1) plays a pivotal role in synaptic transmission and neuronal plasticity. Despite the fact that several interacting proteins involved in the mGlu1 subcellular trafficking and intracellular transduction mechanisms have been identified, the protein network associated with this receptor in specific brain areas remains largely unknown. To identify novel mGlu1-associated protein complexes in the mouse cerebellum, we used an unbiased tissue-specific proteomic approach, namely co-immunoprecipitation followed by liquid chromatography/tandem mass spectrometry analysis. Many well-known protein complexes as well as novel interactors were identified, including G-proteins, Homer, δ2 glutamate receptor, 14-3-3 proteins, and Na/K-ATPases. A novel putative interactor, KCTD12, was further investigated. Reverse co-immunoprecipitation with anti-KCTD12 antibodies revealed mGlu1 in wild-type but not in KCTD12-knock-out homogenates. Freeze-fracture replica immunogold labeling co-localization experiments showed that KCTD12 and mGlu1 are present in the same nanodomain in Purkinje cell spines, although at a distance that suggests that this interaction is mediated through interposed proteins. Consistently, mGlu1 could not be co-immunoprecipitated with KCTD12 from a recombinant mammalian cell line co-expressing the two proteins. The possibility that this interaction was mediated via GABAB receptors was excluded by showing that mGlu1 and KCTD12 still co-immunoprecipitated from GABAB receptor knock-out tissue. In conclusion, this study identifies tissue-specific mGlu1-associated protein clusters including KCTD12 at Purkinje cell synapses.


Subject(s)
Proteomics , Receptors, Metabotropic Glutamate , Mice , Animals , Purkinje Cells , Receptors, Metabotropic Glutamate/metabolism , Receptors, GABA-B/metabolism , gamma-Aminobutyric Acid/metabolism , Glutamates/metabolism , Mammals/metabolism
3.
Article in English | MEDLINE | ID: mdl-36901549

ABSTRACT

The association between Alzheimer's disease (AD) and type 2 diabetes mellitus (T2DM) has been extensively demonstrated, but despite this, the pathophysiological mechanisms underlying it are still unknown. In previous work, we discovered a central role for the autophagy pathway in the common alterations observed between AD and T2DM. In this study, we further investigate the role of genes belonging to this pathway, measuring their mRNA expression and protein levels in 3xTg-AD transgenic mice, an animal model of AD. Moreover, primary mouse cortical neurons derived from this model and the human H4Swe cell line were used as cellular models of insulin resistance in AD brains. Hippocampal mRNA expression showed significantly different levels for Atg16L1, Atg16L2, GabarapL1, GabarapL2, and Sqstm1 genes at different ages of 3xTg-AD mice. Significantly elevated expression of Atg16L1, Atg16L2, and GabarapL1 was also observed in H4Swe cell cultures, in the presence of insulin resistance. Gene expression analysis confirmed that Atg16L1 was significantly increased in cultures from transgenic mice when insulin resistance was induced. Taken together, these results emphasise the association of the autophagy pathway in AD-T2DM co-morbidity, providing new evidence about the pathophysiology of both diseases and their mutual interaction.


Subject(s)
Alzheimer Disease , Diabetes Mellitus, Type 2 , Insulin Resistance , Mice , Humans , Animals , Alzheimer Disease/epidemiology , Diabetes Mellitus, Type 2/epidemiology , Disease Models, Animal , Comorbidity , Mice, Transgenic , Autophagy , RNA, Messenger , Carrier Proteins
4.
Sci Rep ; 13(1): 222, 2023 01 05.
Article in English | MEDLINE | ID: mdl-36604493

ABSTRACT

Alzheimer's disease is the most common form of dementia. Notwithstanding the huge investments in drug development, only one disease-modifying treatment has been recently approved. Here we present a single-cell-led systems biology pipeline for the identification of drug repurposing candidates. Using single-cell RNA sequencing data of brain tissues from patients with Alzheimer's disease, genome-wide association study results, and multiple gene annotation resources, we built a multi-cellular Alzheimer's disease molecular network that we leveraged for gaining cell-specific insights into Alzheimer's disease pathophysiology and for the identification of drug repurposing candidates. Our computational approach pointed out 54 candidate drugs, mainly targeting MAPK and IGF1R signaling pathways, which could be further evaluated for their potential as Alzheimer's disease therapy.


Subject(s)
Alzheimer Disease , Humans , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Drug Repositioning/methods , Genome-Wide Association Study , Systems Biology
5.
Int J Mol Sci ; 23(22)2022 Nov 14.
Article in English | MEDLINE | ID: mdl-36430520

ABSTRACT

Trazodone is an efficacious atypical antidepressant acting both as an SSRI and a 5HT2A and 5HT2C antagonist. Antagonism to H1-histaminergic and alpha1-adrenergic receptors is responsible for a sleep-promoting action. We studied long-term gene expression modulations induced by chronic trazodone to investigate the molecular underpinning of trazodone efficacy. Rats received acute or chronic treatment with trazodone or citalopram. mRNA expression of growth factor and circadian rhythm genes was evaluated by qPCR in the prefrontal cortex (PFCx), hippocampus, Nucleus Accumbens (NAc), amygdala, and hypothalamus. CREB levels and phosphorylation state were evaluated using Western blotting. BDNF levels were significantly increased in PFCx and hippocampus by trazodone and in the NAc and hypothalamus by citalopram. Likewise, TrkB receptor levels augmented in the PFCx after trazodone and in the amygdala after citalopram. FGF-2 and FGFR2 levels were higher after trazodone in the PFCx. The CREB phosphorylation state was increased by chronic trazodone in the PFCx, hippocampus, and hypothalamus. Bmal1 and Per1 were increased by both antidepressants after acute and chronic treatments, while Per2 levels were specifically augmented by chronic trazodone in the PFCx and NAc, and by citalopram in the PFCx, amygdala, and NAc. These findings show that trazodone affects the expression of neurotrophic factors involved in antidepressant responses and alters circadian rhythm genes implicated in the pathophysiology of depression, thus shedding light on trazodone's molecular mechanism of action.


Subject(s)
Trazodone , Animals , Rats , Trazodone/pharmacology , Trazodone/metabolism , Citalopram/pharmacology , Circadian Rhythm , Antidepressive Agents/pharmacology , Brain/metabolism , Gene Expression
6.
Eur Neuropsychopharmacol ; 57: 1-14, 2022 04.
Article in English | MEDLINE | ID: mdl-35008014

ABSTRACT

Anxiety disorders are pervasive psychiatric disorders causing great suffering. The high (HAB) and low (LAB) anxiety-related behaviour rats were selectively bred to investigate neurobiological correlates of anxiety. We compared the level of neuropeptides relevant for anxiety- and depression-related behaviours in selected brain regions of HAB and LAB rats. Increased anxiety and depression-like behaviours of male and female HAB rats in the elevated plus-maze and forced swim tests were accompanied by elevated levels of neuropeptide Y (NPY) in the prefrontal (PFC), frontal (FC) and cingulate cortex (CCx), the striatum, and periaqueductal grey (PAG). Moreover, HAB rats displayed sex-dependent, elevated levels of calcitonin gene-related peptide (CGRP) in PFC, FC, CCx, hippocampus, and PAG. Higher neurokinin A (NKA) levels were detected in CCx, striatum, and PAG in HAB males and in CCx and hypothalamus in HAB females. Increased neurotensin was detected in CCx and PAG in HAB males and in hypothalamus in HAB females. Elevated corticotropin-releasing hormone (CRH) levels appeared in female HAB hypothalamus. Significant correlations were found between anxiety-like behaviour and NPY, CGRP, NKA, and neurotensin, particularly with NPY in CCx and striatum, CGRP in FC and hippocampus, and NKA in entorhinal cortex. This is the first report of NPY, CGRP, NKA, Neurotensin, and CRH measurements in brain regions of HAB and LAB rats, which showed widespread NPY and CGRP alterations in cortical regions, with NKA and neurotensin changes localised in sub-cortical areas. The results may contribute to elucidate pathophysiological mechanisms underlying anxiety and depression and should facilitate identifying novel therapeutic targets.


Subject(s)
Calcitonin Gene-Related Peptide , Neuropeptide Y , Animals , Anxiety , Anxiety Disorders , Brain/metabolism , Calcitonin Gene-Related Peptide/metabolism , Female , Male , Neurokinin A/metabolism , Neuropeptide Y/metabolism , Neurotensin , Rats
7.
Front Pharmacol ; 12: 733577, 2021.
Article in English | MEDLINE | ID: mdl-34621169

ABSTRACT

This study aimed to investigate DNA methylation levels in patients undergoing major breast surgery under opioid-based general anesthesia. Blood samples were collected from eleven enrolled patients, before, during and after anesthesia. PBMC were isolated and global DNA methylation levels as well as DNA methyltransferase (DNMT) and cytokine gene expression were assessed. DNA methylation levels significantly declined by 26%, reversing the direction after the end of surgery. Likewise, DNMT1a mRNA expression was significantly reduced at all time points, with lowest level of -68%. DNMT3a and DNMT3b decreased by 65 and 71%, respectively. Inflammatory cytokines IL6 and TNFα mRNA levels showed a trend for increased expression at early time-points to end with a significant decrease at 48 h after surgery. This exploratory study revealed for the first time intraoperative global DNA hypomethylation in patients undergoing major breast surgery under general anesthesia with fentanyl. The alterations of global DNA methylation here observed seem to be in agreement with DNMTs gene expression changes. Furthermore, based on perioperative variations of IL6 and TNFα gene expression, we hypothesize that DNA hypomethylation may occur as a response to surgical stress rather than to opiate exposure.

8.
Int J Mol Sci ; 22(2)2021 Jan 09.
Article in English | MEDLINE | ID: mdl-33435320

ABSTRACT

Nicotine addiction is a severe public health problem. The aim of this study was to investigate the alterations in key neurotransmissions after 60 days of withdrawal from seven weeks of intermittent cigarette smoke, e-cigarette vapours, or an e-cigarette vehicle. In the nicotine withdrawal groups, increased depressive and anxiety/obsessive-compulsive-like behaviours were demonstrated in the tail suspension, sucrose preference and marble burying tests. Cognitive impairments were detected in the spatial object recognition test. A significant increase in Corticotropin-releasing factor (Crf) and Crf1 mRNA levels was observed, specifically after cigarette withdrawal in the caudate-putamen nucleus (CPu). The nociceptin precursor levels were reduced by cigarette (80%) and e-cigarette (50%) withdrawal in the CPu. The delta opioid receptor showed a significant reduction in the hippocampus driven by the exposure to an e-cigarette solubilisation vehicle, while the mRNA levels doubled in the CPu of mice that had been exposed to e-cigarettes. Withdrawal after exposure to e-cigarette vapour induced a 35% Bdnf mRNA decrease in the hippocampus, whereas Bdnf was augmented by 118% by cigarette withdrawal in the CPu. This study shows that long-term withdrawal-induced affective and cognitive symptoms associated to lasting molecular alterations in peptidergic signalling may determine the impaired neuroplasticity in the hippocampal and striatal circuitry.


Subject(s)
E-Cigarette Vapor/adverse effects , Hippocampus/drug effects , RNA, Messenger/genetics , Substance Withdrawal Syndrome/genetics , Tobacco Smoke Pollution/adverse effects , Animals , Brain-Derived Neurotrophic Factor/genetics , Caudate Nucleus/drug effects , Caudate Nucleus/metabolism , Caudate Nucleus/physiopathology , Corticotropin-Releasing Hormone/genetics , Down-Regulation/drug effects , Hippocampus/metabolism , Hippocampus/physiopathology , Male , Mice , Mice, Inbred BALB C , Opioid Peptides/genetics , Orexins/genetics , Putamen/drug effects , Putamen/metabolism , Putamen/physiopathology , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Opioid/genetics , Substance Withdrawal Syndrome/etiology , Substance Withdrawal Syndrome/physiopathology , Up-Regulation/drug effects
9.
World J Biol Psychiatry ; 22(4): 315-321, 2021 04.
Article in English | MEDLINE | ID: mdl-32787676

ABSTRACT

OBJECTIVES: Based on the hypothesis of a role for folate and vitamin B12 in major depressive disorders (MDD), we aimed at validating the association between folate pathway biomarkers and depression or antidepressant response in clinical trial populations. METHODS: We investigated serum levels erythrocyte folate and serum levels of homocysteine, vitamin B12, and folate as disease and response biomarkers for MDD in two independent randomised, placebo-controlled clinical trials, where paroxetine or venlafaxine were used as active controls, for a total of 881 patients. RESULTS: Significant but weak correlations between depression severity and biomarker levels could be detected in the paroxetine study for serum folate and vitamin B12, with no correlations for any biomarker in the venlafaxine study. Besides a weak association for erythrocyte folate in the venlafaxine study, no significant associations were observed between treatment response and pre-treatment levels of any of the biomarkers tested. CONCLUSIONS: Notwithstanding the relatively large number of patients tested, we did not find consistent associations between folate biomarkers and MDD severity, or response to paroxetine and venlafaxine. Our results may be related to the particular study design or clinical population; however, our findings do not support the hypothesis of a dysfunction of one-carbon metabolism in MDD.


Subject(s)
Depressive Disorder, Major , Paroxetine , Biomarkers , Depressive Disorder, Major/drug therapy , Folic Acid , Humans , Venlafaxine Hydrochloride
10.
Cells ; 9(8)2020 07 31.
Article in English | MEDLINE | ID: mdl-32751911

ABSTRACT

The Negr1 gene has been significantly associated with major depression in genetic studies. Negr1 encodes for a cell adhesion molecule cleaved by the protease Adam10, thus activating Fgfr2 and promoting neuronal spine plasticity. We investigated whether antidepressants modulate the expression of genes belonging to Negr1-Fgfr2 pathway in Flinders sensitive line (FSL) rats, in a corticosterone-treated mouse model of depression, and in mouse primary neurons. Negr1 and Adam10 were the genes mostly affected by antidepressant treatment, and in opposite directions. Negr1 was down-regulated by escitalopram in the hypothalamus of FSL rats, by fluoxetine in the hippocampal dentate gyrus of corticosterone-treated mice, and by nortriptyline in hippocampal primary neurons. Adam10 mRNA was increased by nortriptyline administration in the hypothalamus, by escitalopram in the hippocampus of FSL rats, and by fluoxetine in mouse dorsal dentate gyrus. Similarly, nortriptyline increased Adam10 expression in hippocampal cultures. Fgfr2 expression was increased by nortriptyline in the hypothalamus of FSL rats and in hippocampal neurons. Lsamp, another IgLON family protein, increased in mouse dentate gyrus after fluoxetine treatment. These findings suggest that Negr1-Fgfr2 pathway plays a role in the modulation of synaptic plasticity induced by antidepressant treatment to promote therapeutic efficacy by rearranging connectivity in corticolimbic circuits impaired in depression.


Subject(s)
Adrenergic Uptake Inhibitors/therapeutic use , Antidepressive Agents, Tricyclic/therapeutic use , Cell Adhesion Molecules, Neuronal/metabolism , Citalopram/therapeutic use , Depression/drug therapy , Depression/metabolism , Nortriptyline/therapeutic use , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Selective Serotonin Reuptake Inhibitors/therapeutic use , Signal Transduction/drug effects , Adrenergic Uptake Inhibitors/pharmacology , Animals , Antidepressive Agents, Tricyclic/pharmacology , Cell Adhesion Molecules, Neuronal/genetics , Cells, Cultured , Citalopram/pharmacology , Depression/genetics , Disease Models, Animal , Down-Regulation/drug effects , Gene Expression/drug effects , Hippocampus/cytology , Mice , Neurons/metabolism , Nortriptyline/pharmacology , Rats , Receptor, Fibroblast Growth Factor, Type 2/genetics , Selective Serotonin Reuptake Inhibitors/pharmacology
11.
Pharmacol Res ; 158: 104941, 2020 08.
Article in English | MEDLINE | ID: mdl-32450347

ABSTRACT

Smoking cessation induces a withdrawal syndrome associated with anxiety, depression, and impaired neurocognitive functions, but much less is known about the withdrawal of e-cigarettes (e-CIG). We investigated in Balb/c mice the behavioural and neurochemical effects of withdrawal for up to 90 days after seven weeks' intermittent exposure to e-CIG vapour or cigarette smoke (CIG). The withdrawal of e-CIG and CIG induced early behavioural alterations such as spatial memory deficits (spatial object recognition task), increased anxiety (elevated plus maze test) and compulsive-like behaviour (marble burying test) that persisted for 60-90 days. Notably, attention-related (virtual object recognition task) and depression-like behaviours (tail suspension and sucrose preference tests) appeared only 15-30 days after withdrawal and persisted for as long as up to 90 days. At hippocampal level, the withdrawal-induced changes in the levels of AMPA receptor GluA1 and GluA2/3 subunits, PSD 95 protein, corticotropin-releasing factor (Crf) and Crf receptor 1 (CrfR1) mRNA were biphasic: AMPA receptor subunit and PSD95 protein levels initially remained unchanged and decreased after 60-90 days, whereas Crf/CrfR1 mRNA levels initially increased and then markedly decreased after 60 days. These late reductions correlated with the behavioural impairments, particularly the appearance of depression-like behaviours. Our findings show that major behavioural and neurochemical alterations persist or even first appear late after the withdrawal of chronic CIG smoke or e-CIG vapour exposure, and underline importance of conducting similar studies of humans, including e-CIG vapers.


Subject(s)
Affect/drug effects , Cigarette Smoking/adverse effects , Cognition/drug effects , E-Cigarette Vapor/adverse effects , Substance Withdrawal Syndrome/metabolism , Tobacco Smoke Pollution/adverse effects , Affect/physiology , Animals , Cigarette Smoking/metabolism , Cognition/physiology , E-Cigarette Vapor/administration & dosage , Hippocampus/chemistry , Hippocampus/drug effects , Hippocampus/metabolism , Inhalation Exposure/adverse effects , Male , Maze Learning/drug effects , Maze Learning/physiology , Mice , Mice, Inbred BALB C , Recognition, Psychology/drug effects , Recognition, Psychology/physiology , Substance Withdrawal Syndrome/psychology
12.
Eur Neuropsychopharmacol ; 31: 69-85, 2020 02.
Article in English | MEDLINE | ID: mdl-31813757

ABSTRACT

Neurobiological underpinnings of treatment-resistant depression, a debilitating condition associated with significant functional impairment, have not been elucidated. Consequently, the aim of this study was to use animal models of response and resistance to antidepressant treatment, in an attempt to identify differences in associated transcriptional responses. Flinders Sensitive Line rats were subjected to maternal separation (MS) and chronically treated with Escitalopram or Nortriptyline. Antidepressants reduced immobility time in the forced swim test in non-MS rats, while lack of antidepressant behavioural response was observed in MS animals. We developed a novel bioinformatic algorithm that enabled identification of transcriptional signatures in hippocampus and pre-frontal cortex that discriminate vehicle- and antidepressant-treated subjects in both MS and non-MS rats. Functional annotation analysis showed that in antidepressant-responder rats the most enriched pathways included IQGAPs activation, toll-like receptor trafficking, energy metabolism, and regulation of endopeptidase activity. The analysis of interacting proteins implicated synaptic vesicles and neurotransmitter release, ubiquitin regulation, cytoskeleton organisation and carbohydrate metabolism. In contrast, in treatment-resistant MS rats, main expression changes were revealed in ribosomal proteins, inflammatory responses, transcriptional/epigenetic regulation, and small GTPases. Susceptibility signature shared Rtn1, Zdhhc5, Igsf6, and Sim1 genes with the latest depression GWAS meta-analysis, while antidepressant resistance signature shared Ctnnd1, Rbms3, Atp1a3, and Pla2r1 genes. In conclusion, this study demonstrated that distinct transcriptional signatures are associated with behavioural response or non-response to antidepressant treatment. The identification of genes involved in antidepressant response will increase the comprehension of the neurobiological underpinnings of treatment-resistant depression, thus contributing to identification of novel therapeutic targets.


Subject(s)
Antidepressive Agents/therapeutic use , Depressive Disorder, Treatment-Resistant/drug therapy , Depressive Disorder, Treatment-Resistant/genetics , Disease Models, Animal , Maternal Deprivation , Animals , Antidepressive Agents/pharmacology , Citalopram/pharmacology , Citalopram/therapeutic use , Depressive Disorder, Treatment-Resistant/psychology , Female , Gene Expression , Hippocampus/drug effects , Male , Protein Interaction Maps/drug effects , Protein Interaction Maps/genetics , Rats , Rats, Transgenic , Transcriptome/drug effects , Transcriptome/genetics , Treatment Outcome
13.
Transl Psychiatry ; 9(1): 182, 2019 08 02.
Article in English | MEDLINE | ID: mdl-31375659

ABSTRACT

The identification of biomarkers of response might speed drug development and set the premises to assist clinical practice in psychiatry. In this work, we evaluated a panel of peripheral biomarkers (including IL-6, IL-10, TNF-α, TNFRII, BDNF, CRP, MMP9 and PAI1) in depressed patients receiving paroxetine, venlafaxine, or placebo. Samples were obtained from two randomised placebo-controlled studies evaluating the efficacy and tolerability of a novel drug candidate, using either paroxetine or venlafaxine as active comparators. In both studies, the biomarker candidates were analysed in plasma collected at randomization and after 10 weeks of treatment with either placebo or active comparator (for a total of 106 and 108 subjects in the paroxetine and venlafaxine study, respectively). Data were obtained by multiplexing sandwich-ELISA system. Data were subjected to statistical analysis to assess their correlation with baseline severity and with response outcome. Increases in biomarker levels were correlated with reduction in depression severity for TNF-α, IL-6 IL-10 and CRP. Response to paroxetine treatment correlated with baseline IL-10, IL-6 and TNF-α levels, with the strongest signal being observed in males. In the venlafaxine study, a correlation was observed only between CRP level at randomisation and response, suggesting differences between the two active treatments and the two studies. Our investigations suggest that a combination of pro- and anti-inflammatory cytokines may predict response outcome in patients treated with paroxetine. The potential for IL-10, IL-6 and TNF-α as response biomarkers for a wider range of antidepressants warrants further investigations in clinical trials with other monoamine reuptake inhibitors.


Subject(s)
C-Reactive Protein/analysis , Depressive Disorder, Major/drug therapy , Interleukin-10/blood , Interleukin-6/blood , Paroxetine/therapeutic use , Tumor Necrosis Factor-alpha/blood , Venlafaxine Hydrochloride/therapeutic use , Adult , Biomarkers/blood , Depressive Disorder, Major/blood , Female , Humans , Male , Middle Aged , Treatment Outcome , Young Adult
14.
Sci Rep ; 9(1): 3965, 2019 03 08.
Article in English | MEDLINE | ID: mdl-30850634

ABSTRACT

Evidence is accumulating that the main chronic diseases of aging Alzheimer's disease (AD) and type-2 diabetes mellitus (T2DM) share common pathophysiological mechanisms. This study aimed at applying systems biology approaches to increase the knowledge of the shared molecular pathways underpinnings of AD and T2DM. We analysed transcriptomic data of post-mortem AD and T2DM human brains to obtain disease signatures of AD and T2DM and combined them with protein-protein interaction information to construct two disease-specific networks. The overlapping AD/T2DM network proteins were then used to extract the most representative Gene Ontology biological process terms. The expression of genes identified as relevant was studied in two AD models, 3xTg-AD and ApoE3/ApoE4 targeted replacement mice. The present transcriptomic data analysis revealed a principal role for autophagy in the molecular basis of both AD and T2DM. Our experimental validation in mouse AD models confirmed the role of autophagy-related genes. Among modulated genes, Cyclin-Dependent Kinase Inhibitor 1B, Autophagy Related 16-Like 2, and insulin were highlighted. In conclusion, the present investigation revealed autophagy as the central dys-regulated pathway in highly co-morbid diseases such as AD and T2DM allowing the identification of specific genes potentially involved in disease pathophysiology which could become novel targets for therapeutic intervention.


Subject(s)
Alzheimer Disease/pathology , Autophagy/physiology , Diabetes Mellitus, Type 2/pathology , Alzheimer Disease/metabolism , Animals , Brain/metabolism , Brain/pathology , Comorbidity , Diabetes Mellitus, Type 2/metabolism , Disease Models, Animal , Humans , Insulin/metabolism , Male , Mice , Mice, Inbred C57BL , Transcriptome/physiology
15.
Neuropsychopharmacology ; 43(10): 2134-2145, 2018 09.
Article in English | MEDLINE | ID: mdl-29950584

ABSTRACT

An enhanced understanding of the pathophysiology of depression would facilitate the discovery of new efficacious medications. To this end, we examined hippocampal transcriptional changes in rat models of disease and in humans to identify common disease signatures by using a new algorithm for signature-based clustering of expression profiles. The tool identified a transcriptomic signature comprising 70 probesets able to discriminate depression models from controls in both Flinders Sensitive Line and Learned Helplessness animals. To identify disease-relevant pathways, we constructed an expanded protein network based on signature gene products and performed functional annotation analysis. We applied the same workflow to transcriptomic profiles of depressed patients. Remarkably, a 171-probesets transcriptional signature which discriminated depressed from healthy subjects was identified. Rat and human signatures shared the SCARA5 gene, while the respective networks derived from protein-based significant interactions with signature genes contained 25 overlapping genes. The comparison between the most enriched pathways in the rat and human signature networks identified a highly significant overlap (p-value: 3.85 × 10-6) of 67 terms including ErbB, neurotrophin, FGF, IGF, and VEGF signaling, immune responses and insulin and leptin signaling. In conclusion, this study allowed the identification of a hippocampal transcriptional signature of resilient or susceptible responses in rat MDD models which overlapped with gene expression alterations observed in depressed patients. These findings are consistent with a loss of hippocampal neural plasticity mediated by altered levels of growth factors and increased inflammatory responses causing metabolic impairments as crucial factors in the pathophysiology of MDD.


Subject(s)
Depressive Disorder, Major/genetics , Depressive Disorder, Major/physiopathology , Intercellular Signaling Peptides and Proteins/genetics , Signal Transduction/genetics , Transcriptome/genetics , Animals , Brain Chemistry/genetics , Computational Biology , Gene Expression Profiling , Gene Expression Regulation/genetics , Helplessness, Learned , Hippocampus/drug effects , Hippocampus/physiology , Humans , Male , Rats , Scavenger Receptors, Class A/genetics , Species Specificity
16.
Drug Alcohol Depend ; 189: 12-20, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29857328

ABSTRACT

BACKGROUND: Nicotine addiction supports tobacco smoking, a main preventable cause of disease and death in Western countries. It develops through long-term neuroadaptations in the brain reward circuit by modulating intracellular pathways and regulating gene expression. This study assesses the regional expression of the transcripts of the CRF transmission in a nicotine sensitization model, since it is hypothesised that the molecular neuroadaptations that mediate the development of sensitization contribute to the development of addiction. METHODS: Rats received intraperitoneal nicotine administrations (0.4 mg/kg) once daily for either 1 day or over 5 days. Locomotor activity was assessed to evaluate the development of sensitization. The mRNA expression of CRF and CRF1 and CRF2 receptors was measured by qPCR in the ventral mesencephalon, ventral striatum, dorsal striatum (DS), prefrontal cortex (PFCx), and hippocampus (Hip). RESULTS: Acute nicotine administration increased locomotor activity in rats. In the sub-chronic group, locomotor activity progressively increased and reached a clear sensitization. Significant effects of sensitization on CRF mRNA levels were detected in the DS (increasing effect). Significantly higher CRF1 and CRF2 receptor levels after sensitization were detected in the Hip. Additionally, CRF2 receptor levels were augmented by sensitization in the PFCx, and treatment and time-induced increases were detected in the DS. Nicotine treatment effects were observed on CRF1R levels in the DS. CONCLUSIONS: This study suggests that the CRF transmission, in addition to its role in increasing withdrawal-related anxiety, may be involved in the development of nicotine-habituated behaviours through reduced control of impulses and the aberrant memory plasticity characterising addiction.


Subject(s)
Central Nervous System Sensitization/physiology , Corpus Striatum/metabolism , Corticotropin-Releasing Hormone/physiology , Hippocampus/metabolism , Nicotine/pharmacology , Prefrontal Cortex/metabolism , Receptors, Corticotropin-Releasing Hormone/physiology , Animals , Corticotropin-Releasing Hormone/biosynthesis , Locomotion/drug effects , Male , Rats , Receptors, Corticotropin-Releasing Hormone/biosynthesis , Reward
17.
Am J Med Genet B Neuropsychiatr Genet ; 174(3): 235-250, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27696737

ABSTRACT

Response to antidepressant (AD) treatment may be a more polygenic trait than previously hypothesized, with many genetic variants interacting in yet unclear ways. In this study we used methods that can automatically learn to detect patterns of statistical regularity from a sparsely distributed signal across hippocampal transcriptome measurements in a large-scale animal pharmacogenomic study to uncover genomic variations associated with AD. The study used four inbred mouse strains of both sexes, two drug treatments, and a control group (escitalopram, nortriptyline, and saline). Multi-class and binary classification using Machine Learning (ML) and regularization algorithms using iterative and univariate feature selection methods, including InfoGain, mRMR, ANOVA, and Chi Square, were used to uncover genomic markers associated with AD response. Relevant genes were selected based on Jaccard distance and carried forward for gene-network analysis. Linear association methods uncovered only one gene associated with drug treatment response. The implementation of ML algorithms, together with feature reduction methods, revealed a set of 204 genes associated with SSRI and 241 genes associated with NRI response. Although only 10% of genes overlapped across the two drugs, network analysis shows that both drugs modulated the CREB pathway, through different molecular mechanisms. Through careful implementation and optimisations, the algorithms detected a weak signal used to predict whether an animal was treated with nortriptyline (77%) or escitalopram (67%) on an independent testing set. The results from this study indicate that the molecular signature of AD treatment may include a much broader range of genomic markers than previously hypothesized, suggesting that response to medication may be as complex as the pathology. The search for biomarkers of antidepressant treatment response could therefore consider a higher number of genetic markers and their interactions. Through predominately different molecular targets and mechanisms of action, the two drugs modulate the same Creb1 pathway which plays a key role in neurotrophic responses and in inflammatory processes. © 2016 The Authors. American Journal of Medical Genetics Part B: Neuropsychiatric Genetics Published by Wiley Periodicals, Inc.


Subject(s)
Antidepressive Agents/therapeutic use , Serotonin and Noradrenaline Reuptake Inhibitors/pharmacology , Animals , Citalopram/therapeutic use , Cyclic AMP Response Element-Binding Protein , Depression/drug therapy , Depressive Disorder/drug therapy , Depressive Disorder/genetics , Disease Models, Animal , Female , Hippocampus , Male , Mice , Multifactorial Inheritance/genetics , Nortriptyline/therapeutic use , Pharmacogenetics , Selective Serotonin Reuptake Inhibitors/therapeutic use , Serotonin and Noradrenaline Reuptake Inhibitors/therapeutic use , Transcriptome/genetics , Treatment Outcome
18.
Pharmacol Res ; 114: 209-218, 2016 12.
Article in English | MEDLINE | ID: mdl-27989838

ABSTRACT

The recreational drug of abuse 3,4-methylenedioxymethamphetamine (MDMA) has been shown to produce neurotoxic damage and long-lasting changes in several brain areas. In addition to the involvement of serotoninergic and dopaminergic systems, little information exists about the contribution of nociceptin/orphaninFQ (N/OFQ)-NOP and dynorphin (DYN)-KOP systems in neuronal adaptations evoked by MDMA. Here we investigated the behavioral and molecular effects induced by acute (8mg/kg) or repeated (8mg/kg twice daily for seven days) MDMA exposure. MDMA exposure affected body weight gain and induced hyperlocomotion; this latter effect progressively decreased after repeated administration. Gene expression analysis indicated a down-regulation of the N/OFQ system and an up-regulation of the DYN system in the nucleus accumbens (NAc), highlighting an opposite systems regulation in response to MDMA exposure. Since histone modifications have been strongly associated to the addiction-related maladaptive changes, we examined two permissive (acH3K9 and me3H3K4) and two repressive transcription marks (me3H3K27 and me2H3K9) at the pertinent opioid gene promoter regions. Chromatin immunoprecipitation assays revealed that acute MDMA increased me3H3K4 at the pN/OFQ, pDYN and NOP promoters. Following acute and repeated treatment a significant decrease of acH3K9 at the pN/OFQ promoter was observed, which correlated with gene expression results. Acute treatment caused an acH3K9 increase and a me2H3K9 decrease at the pDYN promoter which matched its mRNA up-regulation. Our data indicate that the activation of the DYNergic stress system together with the inactivation of the N/OFQergic anti-stress system contribute to the neuroadaptive actions of MDMA and offer novel epigenetic information associated with MDMA abuse.


Subject(s)
Dynorphins/genetics , Histone Code/drug effects , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , Nucleus Accumbens/drug effects , Opioid Peptides/genetics , Serotonin Agents/pharmacology , Adrenergic Uptake Inhibitors/administration & dosage , Adrenergic Uptake Inhibitors/pharmacology , Animals , Gene Expression Regulation/drug effects , Locomotion/drug effects , Male , N-Methyl-3,4-methylenedioxyamphetamine/administration & dosage , Nucleus Accumbens/metabolism , Promoter Regions, Genetic/drug effects , Rats, Sprague-Dawley , Serotonin Agents/administration & dosage , Nociceptin
19.
Proteomics Clin Appl ; 10(12): 1254-1263, 2016 12.
Article in English | MEDLINE | ID: mdl-27612656

ABSTRACT

PURPOSE: The pathophysiological basis of major depression is incompletely understood. Recently, numerous proteomic studies have been performed in rodent models of depression to investigate the molecular underpinnings of depressive-like behaviours with an unbiased approach. The objective of the study is to integrate the results of these proteomic studies in depression models to shed light on the most relevant molecular pathways involved in the disease. EXPERIMENTAL DESIGN: Network analysis is performed integrating preexisting proteomic data from rodent models of depression. The IntAct mouse and the HRPD are used as reference protein-protein interaction databases. The functionality analyses of the networks are then performed by testing overrepresented GO biological process terms and pathways. RESULTS: Functional enrichment analyses of the networks revealed an association with molecular processes related to depression in humans, such as those involved in the immune response. Pathways impacted by clinically effective antidepressants are modulated, including glutamatergic signaling and neurotrophic responses. Moreover, dysregulations of proteins regulating energy metabolism and circadian rhythms are implicated. The comparison with protein pathways modulated in depressive patients revealed significant overlapping. CONCLUSIONS AND CLINICAL RELEVANCE: This systems biology study supports the notion that animal models can contribute to the research into the biology and therapeutics of depression.


Subject(s)
Depressive Disorder, Major/immunology , Depressive Disorder, Major/pathology , Glutamic Acid/metabolism , Proteomics , Signal Transduction , Systems Biology , Animals , Depressive Disorder, Major/metabolism , Disease Models, Animal , Mice , Rats
20.
Drug Alcohol Depend ; 166: 150-8, 2016 Sep 01.
Article in English | MEDLINE | ID: mdl-27430399

ABSTRACT

BACKGROUND: Nicotine dependence is maintained by neurobiological adaptations in the dopaminergic brain reward pathway with the contribution of opioidergic circuits. This study assessed the role of opioid peptides and receptors on the molecular changes associated with nicotine dependence. To this aim we analysed nicotine effects on opioid gene and receptor expression in the reward pathway in a nicotine sensitization model. METHODS: Sprague-Dawley rats received nicotine administrations for five days and locomotor activity assessment showed the development of sensitization. The mRNA expression of prodynorphin (pdyn), pronociceptin (pnoc) and the respective receptors was measured by quantitative PCR in the ventral midbrain (VM), the nucleus accumbens (NAc), the caudate-putamen (CPu), the pre-frontal cortex (PFCx), and the hippocampus. RESULTS: A significant positive effect of sensitization on pdyn mRNA levels was detected in the CPu. This effect was supported by a significant and selective correlation between the two parameters in this region. Moreover, chronic but not acute nicotine treatment significantly decreased pdyn mRNA levels in the NAc and increased expression in the PFCx. Pnoc mRNA was significantly increased in the VM and the PFCx after sub-chronic administration of nicotine, whereas no alterations were observed after acute treatment. No treatment associated changes were detected in κ-opioid receptor or nociceptin receptor mRNAs. CONCLUSIONS: This experiment revealed an effect of nicotine administration that was distinguishable from the effect of nicotine sensitization. While several pnoc and pdyn changes were associated to nicotine administration, the only significant effect of sensitization was a significant increase in pdyn in the CPu.


Subject(s)
Brain/drug effects , Enkephalins/genetics , Gene Expression/drug effects , Gene Expression/genetics , Motivation/drug effects , Motivation/genetics , Nerve Net/drug effects , Nicotine/pharmacology , Nucleus Accumbens/drug effects , Protein Precursors/genetics , RNA, Messenger/genetics , Receptors, Opioid/genetics , Reward , Tobacco Use Disorder/genetics , Animals , Brain Mapping , Dopamine/metabolism , Motor Activity/drug effects , Opioid Peptides/genetics , Rats , Rats, Sprague-Dawley , Receptors, Opioid, kappa/genetics , Tobacco Use Disorder/metabolism , Nociceptin Receptor
SELECTION OF CITATIONS
SEARCH DETAIL
...