Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
Int J Mol Sci ; 24(11)2023 May 27.
Article in English | MEDLINE | ID: mdl-37298310

ABSTRACT

Multiple sclerosis (MS) is an autoimmune disorder of the central nervous system (CNS) whose aetiology is only partly understood. Investigating the intricate transcriptional changes occurring in MS brains is critical to unravel novel pathogenic mechanisms and therapeutic targets. Unfortunately, this process is often hindered by the difficulty in retrieving an adequate number of samples. However, by merging data from publicly available datasets, it is possible to identify alterations in gene expression profiles and regulatory pathways that were previously overlooked. Here, we merged microarray gene expression profiles obtained from CNS white matter samples taken from MS donors to identify novel differentially expressed genes (DEGs) linked with MS. Data from three independent datasets (GSE38010, GSE32915, and GSE108000) were combined and used to detect novel DEGs using the Stouffer's Z-score method. Corresponding regulatory pathways were analysed using the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway databases. Finally, top up- and down-regulated transcripts were validated by real-time quantitative PCR (qPCR) using an independent set of white matter tissue samples obtained from MS donors with different disease subtypes. There were a total of 1446 DEGs, of which 742 were up-regulated and 704 genes were down-regulated. DEGs were associated with several myelin-related pathways and protein metabolism pathways. Validation studies of selected top up- or down-regulated genes highlighted MS subtype-specific differences in the expression of some of the identified genes, underlining a more complex scenario of white matter pathology amongst people afflicted by this devastating disease.


Subject(s)
Multiple Sclerosis , Humans , Multiple Sclerosis/genetics , Multiple Sclerosis/metabolism , Gene Expression Profiling/methods , Brain/metabolism , Microarray Analysis , Central Nervous System/metabolism , Computational Biology/methods , Gene Regulatory Networks
2.
Int J Mol Sci ; 24(4)2023 Feb 11.
Article in English | MEDLINE | ID: mdl-36835076

ABSTRACT

Physiological aging triggers a cascade of negative effects on the human body and the human joint is only one of the several compartments affected by this irreversible and natural process. Osteoarthritis and cartilage degeneration can cause pain and disability; therefore, identifying the molecular processes underlying these phenomena and the biomarkers produced during physical activity is of critical importance. In the present review, the main goal was to identify and discuss the articular cartilage biomarkers analyzed in studies in which physical or sports activities were adopted and eventually to propose a standard operating procedure for the assessment. Articles collected from Pubmed, Web of Science, and Scopus were scrutinized to detect reliable cartilage biomarkers. The principal articular cartilage biomarkers detected in these studies were cartilage oligomeric matrix protein, matrix metalloproteinases, interleukins, and carboxy-terminal telopeptide. The articular cartilage biomarkers identified in this scoping review may aid in a better comprehension of where research on the topic is heading and offer a viable instrument for streamlining investigations on cartilage biomarker discovery.


Subject(s)
Cartilage, Articular , Osteoarthritis , Humans , Aging/metabolism , Aging/pathology , Biomarkers/metabolism , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Exercise/physiology , Osteoarthritis/metabolism
3.
Int J Mol Sci ; 24(13)2023 Jul 05.
Article in English | MEDLINE | ID: mdl-37446298

ABSTRACT

Neuropsychiatric systemic lupus erythematosus (NPSLE) is one of the most common and severe manifestations of lupus; however, its pathogenesis is still poorly understood. While there is sparse evidence suggesting that the ongoing autoimmunity may trigger pathogenic changes to the central nervous system (CNS) microvasculature, culminating in inflammatory/ischemic damage, further evidence is still needed. In this study, we used the spontaneous mouse model of SLE (NZBWF1 mice) to investigate the expression of genes and proteins associated with endothelial (dys)function: tissue and urokinase plasminogen activators (tPA and uPA), intercellular and vascular adhesion molecules 1 (ICAM-1 and VCAM-1), brain derived neurotrophic factor (BDNF), endothelial nitric oxide synthase (eNOS) and Krüppel-like factor 4 (KLF4) and neuroprotection/immune modulation: pituitary adenylate cyclase-activating peptide (PACAP), vasoactive intestinal peptide (VIP), PACAP receptor (PAC1), VIP receptors 1 and 2 (VPAC1 and VPAC2). Analyses were carried out both in the hippocampus and striatum of SLE mice of two different age groups (2 and 7 months old), since age correlates with disease severity. In the hippocampus, we identified a gene/protein expression profile indicative of mild endothelial dysfunction, which increased in severity in aged SLE mice. These alterations were paralleled by moderate alterations in the expression of VIP, PACAP and related receptors. In contrast, we report a robust upregulation of endothelial activation markers in the striatum of both young and aged mice, concurrent with significant induction of the VIP/PACAP system. These data identify molecular signatures of endothelial alterations in the hippocampus and striatum of NZBWF1 mice, which are accompanied by a heightened expression of endogenous protective/immune-modulatory neuropeptides. Collectively, our results support the idea that NPSLE may cause alterations of the CNS micro-vascular compartment that cannot be effectively counteracted by the endogenous activity of the neuropeptides PACAP and VIP.


Subject(s)
Lupus Erythematosus, Systemic , Vasoactive Intestinal Peptide , Mice , Animals , Vasoactive Intestinal Peptide/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Receptors, Vasoactive Intestinal Polypeptide, Type I , Receptors, Vasoactive Intestinal Peptide, Type II
4.
Molecules ; 28(12)2023 Jun 16.
Article in English | MEDLINE | ID: mdl-37375363

ABSTRACT

L-Azetidine-2-carboxylic acid (AZE) is a non-protein amino acid that shares structural similarities with its proteogenic L-proline amino acid counterpart. For this reason, AZE can be misincorporated in place of L-proline, contributing to AZE toxicity. In previous work, we have shown that AZE induces both polarization and apoptosis in BV2 microglial cells. However, it is still unknown if these detrimental effects involve endoplasmic reticulum (ER) stress and whether L-proline co-administration prevents AZE-induced damage to microglia. Here, we investigated the gene expression of ER stress markers in BV2 microglial cells treated with AZE alone (1000 µM), or co-treated with L-proline (50 µM), for 6 or 24 h. AZE reduced cell viability, nitric oxide (NO) secretion and caused a robust activation of the unfolded protein response (UPR) genes (ATF4, ATF6, ERN1, PERK, XBP1, DDIT3, GADD34). These results were confirmed by immunofluorescence in BV2 and primary microglial cultures. AZE also altered the expression of microglial M1 phenotypic markers (increased IL-6, decreased CD206 and TREM2 expression). These effects were almost completely prevented upon L-proline co-administration. Finally, triple/quadrupole mass spectrometry demonstrated a robust increase in AZE-bound proteins after AZE treatment, which was reduced by 84% upon L-proline co-supplementation. This study identified ER stress as a pathogenic mechanism for AZE-induced microglial activation and death, which is reversed by co-administration of L-proline.


Subject(s)
Microglia , Proline , Proline/pharmacology , Proline/chemistry , Azetidinecarboxylic Acid/pharmacology , Azetidinecarboxylic Acid/chemistry , Amino Acids , Endoplasmic Reticulum Stress
5.
Curr Issues Mol Biol ; 44(10): 4500-4516, 2022 Sep 28.
Article in English | MEDLINE | ID: mdl-36286023

ABSTRACT

L-Azetidine-2-carboxylic acid (AZE) is a toxic non-protein coding amino acid (npAA) that is highly abundant in sugar and table beets. Due to its structural similarity with the amino acid L-proline, AZE can evade the editing process during protein assembly in eukaryotic cells and be misincorporated into L-proline-rich proteins, potentially causing protein misfolding and other detrimental effects to cells. In this study, we sought to determine if AZE treatment triggered pro-inflammatory and pro-apoptotic responses in BV2 microglial cells. BV2 microglial cells exposed to AZE at increasing concentrations (0−2000 µM) at 0, 3, 6, 12 and 24 h were assayed for cell viability (MTT) and nitric oxide release (Griess assay). Annexin V-FITC/propidium iodide (PI) staining was used to assess apoptosis. Real-time qPCR, Western blot and immunocytochemistry were used to interrogate relevant pro- and anti-inflammatory and other molecular targets of cell survival response. AZE (at concentrations > 1000 µM) significantly reduced cell viability, increased BAX/Bcl2 ratio and caused cell death. Results were mirrored by a robust increase in nitric oxide release, percentage of activated/polarised cells and expression of pro-inflammatory markers (IL-1ß, IL-6, NOS2, CD68 and MHC-2a). Additionally, we found that AZE induced the expression of the extracellular matrix degrading enzyme matrix metalloproteinase 9 (MMP-9) and brain derived neurotrophic factor (BDNF), two critical regulators of microglial motility and structural plasticity. Collectively, these data indicate that AZE-induced toxicity is associated with increased pro-inflammatory activity and reduced survival in BV2 microglia. This evidence may prompt for an increased monitoring of AZE consumption by humans.

6.
J Integr Neurosci ; 21(1): 33, 2022 Jan 28.
Article in English | MEDLINE | ID: mdl-35164469

ABSTRACT

Vasoactive intestinal peptide (VIP) and pituitary adenylate cyclase-activating polypeptide (PACAP) are two widely expressed neuropeptides with important immunomodulatory and neuroprotective properties in the central nervous system (CNS). Both VIP and PACAP have been implicated in several neurological diseases and have shown favourable effects in different animal models of multiple sclerosis (MS). MS is a chronic inflammatory and neurodegenerative disease of the CNS affecting over 2.5 million people worldwide. The disease is characterised by extensive neuroinflammation, demyelination and axonal loss. Currently, there is no cure for MS, with treatment options only displaying partial efficacy. Importantly, epidemiological studies in the MS population have demonstrated that there is a high incidence of neurological and psychological comorbidities such as depression, anxiety, epilepsy and stroke among afflicted people. Hence, given the widespread protective effects of the VIP/PACAP system in the CNS, this review will aim at exploring the beneficial roles of VIP and PACAP in ameliorating some of the most common neurological comorbidities associated with MS. The final scope of the review is to put more emphasis on how targeting the VIP/PACAP system may be an effective therapeutic strategy to modify MS disease course and its associated comorbidities.


Subject(s)
Mental Disorders/metabolism , Multiple Sclerosis/metabolism , Nervous System Diseases/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Vasoactive Intestinal Peptide/metabolism , Animals , Comorbidity , Humans , Mental Disorders/drug therapy , Mental Disorders/epidemiology , Multiple Sclerosis/drug therapy , Multiple Sclerosis/epidemiology , Nervous System Diseases/drug therapy , Nervous System Diseases/epidemiology
7.
Int J Mol Sci ; 23(3)2022 Feb 06.
Article in English | MEDLINE | ID: mdl-35163768

ABSTRACT

A pharmacological and genetic blockade of the dopamine D3 receptor (D3R) has shown to be neuroprotective in models of Parkinson's disease (PD). The anxiolytic drug buspirone, a serotonin receptor 1A agonist, also functions as a potent D3R antagonist. To test if buspirone elicited neuroprotective activities, C57BL/6 mice were subjected to rotenone treatment (10mg/kg i.p for 21 days) to induce PD-like pathology and were co-treated with increasing dosages of buspirone (1, 3, or 10 mg/kg i.p.) to determine if the drug could prevent rotenone-induced damage to the central nervous system (CNS). We found that high dosages of buspirone prevented the behavioural deficits caused by rotenone in the open field test. Molecular and histological analyses confirmed that 10 mg/kg of buspirone prevented the degeneration of TH-positive neurons. Buspirone attenuated the induction of interleukin-1ß and interleukin-6 expression by rotenone, and this was paralleled by the upregulation of arginase-1, brain-derived neurotrophic factor (BDNF), and activity-dependent neuroprotective protein (ADNP) in the midbrain, striatum, prefrontal cortex, amygdala, and hippocampus. Buspirone treatment also improved mitochondrial function and antioxidant activities. Lastly, the drug prevented the disruptions in the expression of two neuroprotective peptides, pituitary adenylate cyclase-activating polypeptide (PACAP) and vasoactive intestinal peptide (VIP). These results pinpoint the neuroprotective efficacy of buspirone against rotenone toxicity, suggesting its potential use as a therapeutic agent in neurodegenerative and neuroinflammatory diseases, such as PD.


Subject(s)
Buspirone/administration & dosage , Neuroprotective Agents/administration & dosage , Parkinson Disease/drug therapy , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Rotenone/toxicity , Vasoactive Intestinal Peptide/metabolism , Animals , Buspirone/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Injections, Intraperitoneal , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Male , Mice , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology , Parkinson Disease/etiology , Parkinson Disease/genetics , Parkinson Disease/psychology , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Vasoactive Intestinal Peptide/genetics
8.
Int J Mol Sci ; 23(9)2022 Apr 26.
Article in English | MEDLINE | ID: mdl-35563181

ABSTRACT

Multiple sclerosis (MS) is a chronic neuroinflammatory and demyelinating disease of the central nervous system (CNS), characterised by the infiltration of peripheral immune cells, multifocal white-matter lesions, and neurodegeneration. In recent years, microglia have emerged as key contributors to MS pathology, acting as scavengers of toxic myelin/cell debris and modulating the inflammatory microenvironment to promote myelin repair. In this review, we explore the role of two neuropeptides, pituitary adenylate cyclase-activating polypeptide (PACAP) and vasoactive intestinal peptide (VIP), as important regulators of microglial functioning during demyelination, myelin phagocytosis, and remyelination, emphasising the potential of these neuropeptides as therapeutic targets for the treatment of MS.


Subject(s)
Multiple Sclerosis , Vasoactive Intestinal Peptide , Humans , Microglia , Pituitary Adenylate Cyclase-Activating Polypeptide
9.
Int J Mol Sci ; 22(20)2021 Oct 11.
Article in English | MEDLINE | ID: mdl-34681607

ABSTRACT

Pituitary adenylate cyclase-activating polypeptide (PACAP) and vasoactive intestinal peptide (VIP) are two structurally related immunosuppressive peptides. However, the underlying mechanisms through which these peptides regulate microglial activity are not fully understood. Using lipopolysaccharide (LPS) to induce an inflammatory challenge, we tested whether PACAP or VIP differentially affected microglial activation, morphology and cell migration. We found that both peptides attenuated LPS-induced expression of the microglial activation markers Iba1 and iNOS (### p < 0.001), as well as the pro-inflammatory mediators IL-1ß, IL-6, Itgam and CD68 (### p < 0.001). In contrast, treatment with PACAP or VIP exerted distinct effects on microglial morphology and migration. PACAP reversed LPS-induced soma enlargement and increased the percentage of small-sized, rounded cells (54.09% vs. 12.05% in LPS-treated cells), whereas VIP promoted a phenotypic shift towards cell subpopulations with mid-sized, spindle-shaped somata (48.41% vs. 31.36% in LPS-treated cells). Additionally, PACAP was more efficient than VIP in restoring LPS-induced impairment of cell migration and the expression of urokinase plasminogen activator (uPA) in BV2 cells compared with VIP. These results suggest that whilst both PACAP and VIP exert similar immunosuppressive effects in activated BV2 microglia, each peptide triggers distinctive shifts towards phenotypes of differing morphologies and with differing migration capacities.


Subject(s)
Microglia/drug effects , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Vasoactive Intestinal Peptide/pharmacology , Animals , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Line , Cell Movement/drug effects , Gene Expression/drug effects , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Lipopolysaccharides/pharmacology , Mice , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Microglia/cytology , Microglia/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Nitrites/metabolism , Phenotype , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Urokinase-Type Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/metabolism
10.
Int J Mol Sci ; 22(24)2021 Dec 20.
Article in English | MEDLINE | ID: mdl-34948457

ABSTRACT

High-fat diet (HFD)-induced comorbid cognitive and behavioural impairments are thought to be the result of persistent low-grade neuroinflammation. Metformin, a first-line medication for the treatment of type-2 diabetes, seems to ameliorate these comorbidities, but the underlying mechanism(s) are not clear. Pituitary adenylate cyclase-activating peptide (PACAP) and vasoactive intestinal peptide (VIP) are neuroprotective peptides endowed with anti-inflammatory properties. Alterations to the PACAP/VIP system could be pivotal during the development of HFD-induced neuroinflammation. To unveil the pathogenic mechanisms underlying HFD-induced neuroinflammation and assess metformin's therapeutic activities, (1) we determined if HFD-induced proinflammatory activity was present in vulnerable brain regions associated with the development of comorbid behaviors, (2) investigated if the PACAP/VIP system is altered by HFD, and (3) assessed if metformin rescues such diet-induced neurochemical alterations. C57BL/6J male mice were divided into two groups to receive either standard chow (SC) or HFD for 16 weeks. A further HFD group received metformin (HFD + M) (300 mg/kg BW daily for 5 weeks) via oral gavage. Body weight, fasting glucose, and insulin levels were measured. After 16 weeks, the proinflammatory profile, glial activation markers, and changes within the PI3K/AKT intracellular pathway and the PACAP/VIP system were evaluated by real-time qPCR and/or Western blot in the hypothalamus, hippocampus, prefrontal cortex, and amygdala. Our data showed that HFD causes widespread low-grade neuroinflammation and gliosis, with regional-specific differences across brain regions. HFD also diminished phospho-AKT(Ser473) expression and caused significant disruptions to the PACAP/VIP system. Treatment with metformin attenuated these neuroinflammatory signatures and reversed PI3K/AKT and PACAP/VIP alterations caused by HFD. Altogether, our findings demonstrate that metformin treatment rescues HFD-induced neuroinflammation in vulnerable brain regions, most likely by a mechanism involving the reinstatement of PACAP/VIP system homeostasis. Data also suggests that the PI3K/AKT pathway, at least in part, mediates some of metformin's beneficial effects.


Subject(s)
Diet, High-Fat/adverse effects , Encephalitis/drug therapy , Metformin/administration & dosage , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Vasoactive Intestinal Peptide/metabolism , Amygdala/drug effects , Amygdala/metabolism , Animals , Case-Control Studies , Down-Regulation , Encephalitis/chemically induced , Encephalitis/genetics , Encephalitis/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Hypothalamus/drug effects , Hypothalamus/metabolism , Male , Metformin/pharmacology , Mice , Mice, Inbred C57BL , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Signal Transduction/drug effects , Vasoactive Intestinal Peptide/genetics
11.
Pharmacol Res ; 141: 384-391, 2019 03.
Article in English | MEDLINE | ID: mdl-30648615

ABSTRACT

The dopamine D3 receptor (D3R), in the nucleus accumbens (NAc), plays an important role in alcohol reward mechanisms. The major neuronal type within the NAc is the GABAergic medium spiny neuron (MSN), whose activity is regulated by dopaminergic inputs. We previously reported that genetic deletion or pharmacological blockade of D3R increases GABAA α6 subunit in the ventral striatum. Here we tested the hypothesis that D3R-dependent changes in GABAA α6 subunit in the NAc affect voluntary alcohol intake, by influencing the inhibitory transmission of MSNs. We performed in vivo and ex vivo experiments in D3R knockout (D3R -/-) mice and wild type littermates (D3R +/+). Ro 15-4513, a high affinity α6-GABAA ligand was used to study α6 activity. At baseline, NAc α6 expression was negligible in D3R+/+, whereas it was robust in D3R-/-; other relevant GABAA subunits were not changed. In situ hybridization and qPCR confirmed α6 subunit mRNA expression especially in the NAc. In the drinking-in-the-dark paradigm, systemic administration of Ro 15-4513 inhibited alcohol intake in D3R+/+, but increased it in D3R-/-; this was confirmed by intra-NAc administration of Ro 15-4513 and furosemide, a selective α6-GABAA antagonist. Whole-cell patch-clamp showed peak amplitudes of miniature inhibitory postsynaptic currents in NAc medium spiny neurons higher in D3R-/- compared to D3R+/+; Ro 15-4513 reduced the peak amplitude in the NAc of D3R-/-, but not in D3R+/+. We conclude that D3R-dependent enhanced expression of α6 GABAA subunit inhibits voluntary alcohol intake by increasing GABA inhibition in the NAc.


Subject(s)
Binge Drinking/genetics , GABAergic Neurons/pathology , Receptors, Dopamine D3/genetics , Receptors, GABA-A/genetics , Animals , Binge Drinking/pathology , GABAergic Neurons/metabolism , Gene Expression Regulation , Male , Mice , Mice, Knockout , Nucleus Accumbens/metabolism , Nucleus Accumbens/pathology , Protein Subunits/genetics , RNA, Messenger/genetics
12.
Int J Mol Sci ; 20(3)2019 01 25.
Article in English | MEDLINE | ID: mdl-30691048

ABSTRACT

The purpose of this study was to investigate the influence of moderate physical activity (MPA) on the expression of osteoarthritis (OA)-related (IL-1ß, IL-6, TNF-α, MMP-13) and anti-inflammatory and chondroprotective (IL-4, IL-10, lubricin) biomarkers in the synovium of an OA-induced rat model. A total of 32 rats were divided into four groups: Control rats (Group 1); rats performing MPA (Group 2); anterior cruciate ligament transection (ACLT)-rats with OA (Group 3); and, ACLT-rats performing MPA (Group 4). Analyses were performed using Hematoxylin & Eosin (H & E) staining, histomorphometry and immunohistochemistry. In Group 3, OA biomarkers were significantly increased, whereas, IL-4, IL-10, and lubricin were significantly lower than in the other experimental groups. We hypothesize that MPA might partake in rescuing type B synoviocyte dysfunction at the early stages of OA, delaying the progression of the disease.


Subject(s)
Anterior Cruciate Ligament Injuries/complications , Cytokines/metabolism , Osteoarthritis, Knee/prevention & control , Physical Conditioning, Animal/methods , Synoviocytes/metabolism , Animals , Disease Models, Animal , Interleukin-10/metabolism , Interleukin-1beta/metabolism , Interleukin-4/metabolism , Interleukin-6/metabolism , Male , Osteoarthritis, Knee/metabolism , Rats , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/metabolism
13.
Int J Mol Sci ; 19(4)2018 Mar 25.
Article in English | MEDLINE | ID: mdl-29587404

ABSTRACT

Following peripheral nerve injury, dysregulations of certain non-coding microRNAs (miRNAs) occur in Schwann cells. Whether these alterations are the result of local inflammation and/or correlate with perturbations in the expression profile of the protective vasoactive intestinal peptide (VIP)/pituitary adenylate cyclase-activating polypeptide (PACAP) system is currently unknown. To address these issues, we aimed at profiling the expression of selected miRNAs in the rat RT4 Schwann cell line. Cells exposed to lipopolysaccharide (LPS), to mimic the local inflammatory milieu, were appraised by real-time qPCR, Western blot and ELISAs. We found that upon LPS treatment, levels of pro-inflammatory cytokines (IL-1ß, -6, -18, -17A, MCP-1 and TNFα) increased in a time-dependent manner. Unexpectedly, the expression levels of VIP and PACAP were also increased. Conversely, levels of VPAC1 and VPAC2 receptors were reduced. Downregulated miRNAs included miR-181b, -145, -27a, -340 and -132 whereas upregulated ones were miR-21, -206, -146a, -34a, -155, -204 and -29a, respectively. Regression analyses revealed that a subset of the identified miRNAs inversely correlated with the expression of VPAC1 and VPAC2 receptors. In conclusion, these findings identified a novel subset of miRNAs that are dysregulated by immune challenge whose activities might elicit a regulatory function on the VIP/PACAP system.


Subject(s)
Inflammation/metabolism , MicroRNAs/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Schwann Cells/metabolism , Vasoactive Intestinal Peptide/metabolism , Animals , Cell Line , Cell Survival/drug effects , Cytokines/analysis , Dose-Response Relationship, Drug , Lipopolysaccharides/pharmacology , Rats , Receptors, Vasoactive Intestinal Peptide, Type II/metabolism , Receptors, Vasoactive Intestinal Polypeptide, Type I/metabolism , Regression Analysis , Schwann Cells/drug effects , Transcriptome/drug effects
14.
Exp Cell Res ; 346(1): 91-8, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27349291

ABSTRACT

Angiogenesis plays a crucial role in progression of pleural malignant mesothelioma. A significantly increased incidence of pleural mesothelioma has been attributed to exposure to fluoro-edenite, a fibrous amphibole extracted from a local stone quarry. In this study, we have investigated the expression of CD68-positive macrophages, tryptase-positive mast cells and CD31 positive areas, as expression of microvascular density, in lung tissue of sheeps exposed to fluoro-edenite fibers vs controls, by immunohistochemical, morphometric and Western blot analysis. The result have evidenced a significant increase in the expression of CD68-positive macrophages, tryptase-positive mast cells as well as a significant increase in microvascular density evaluated as CD31 positive areas in lung tissue of of sheeps exposed to fluoro-edenite fibers vs controls. These data confirmed the important role played by tumor microenvironment components, including macrophages and mast cells, in favour of angiogenesis in pleural mesothelioma induced by fluoro-edenite exposure.


Subject(s)
Asbestos, Amphibole/toxicity , Lung/pathology , Macrophages/metabolism , Mast Cells/metabolism , Neovascularization, Physiologic , Animals , Antigens, CD/metabolism , Blotting, Western , Densitometry , Female , Immunohistochemistry , Lung/drug effects , Macrophages/drug effects , Male , Mast Cells/drug effects , Mast Cells/enzymology , Neovascularization, Physiologic/drug effects , Sheep , Staining and Labeling , Tryptases/metabolism , Tubulin/metabolism
15.
Exp Cell Res ; 339(2): 407-16, 2015 Dec 10.
Article in English | MEDLINE | ID: mdl-26376118

ABSTRACT

Glioblastoma multiforme (GBM) is characterized by numerous abnormal blood vessels, which rapidly proliferate and invade brain tissue and express different angiogenic factors. In this study we have investigated whether the expression levels of CD31/ PECAM1 are deregulated in human GBM tissue specimens and we have also correlated the expression levels of CD31/PECAM1 with those of HIF-1α. Finally, we have established a correlation between the expression levels of CD31/PECAM1 and HIF-1α, and those of two other biomarkers, namely N-cadherin and ADAM-10, of aggressiveness in the same tumors. Results have shown an increased expression of CD31/PECAM1 correlated to HIF-1α expression, confirming evidence demonstrating that different types of tumor are able to trigger aberrant angiogenesis through HIF-1α. Moreover, we also established a further correlation among CD31/PECAM1 and HIF-1α and N-cadherin and ADAM-10, two other markers of aggressiveness in the same tumors.


Subject(s)
Brain Neoplasms/genetics , Glioblastoma/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Aged , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Middle Aged , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism
16.
Cell Tissue Res ; 362(1): 45-60, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25948484

ABSTRACT

Glioblastoma multiforme (GBM) is the most common and aggressive malignant primary brain tumor in humans, whose invasiveness and proliferation are associated with poor prognosis. Matrix metalloproteinases (MMPs) and the related family of "a disintegrin and metalloproteinase" (ADAM) both contribute to increase cell invasion, and its substrate N-cadherin is involved in proliferation and metastatic capacities of tumor cells. However, these molecular determinants of aggressiveness have not been adequately characterized in GBM. In an attempt to better define these pathogenetic signatures, in the present study we evaluated the comparative expression of two main MMPs (MMP-2 and -9), as well as of ADAM-10 and N-cadherin in surgical samples from patients diagnosed with WHO grade IV GBM (n = 25) and in cortical tissue specimens obtained from untreatable epileptic patients (controls, n = 8) through a series of histopathological, immunohistochemical and biochemical tests. Our studies revealed that both MMP-2 and -9 immunoreactivities (IRs) were upregulated in 13 of 25 (52 %) and 19 of 25 (76 %) GBMs, respectively, and the extent of the increase was highly significant with respect to controls (p < 0.001). ADAM-10 IR was also found to be increased (p < 0.001) in 16 of 25 GBM specimens (64 %). Conversely, N-cadherin IR was remarkably decreased (p < 0.001) in almost the totality of tumor samples (22 of 25, 88 %). A similar trend was also obtained at the mRNA and protein level by qPCR and western blot analyses, respectively. Collectively, the current study provides a comprehensive molecular portrayal of some of the major pathological hallmarks of GBM aggressiveness, which could be exploitable as potential targets for a new therapeutic approach.


Subject(s)
ADAM Proteins/biosynthesis , Amyloid Precursor Protein Secretases/biosynthesis , Antigens, CD/biosynthesis , Biomarkers, Tumor/biosynthesis , Brain Neoplasms/metabolism , Cadherins/biosynthesis , Glioblastoma/metabolism , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 9/biosynthesis , Membrane Proteins/biosynthesis , ADAM Proteins/genetics , ADAM10 Protein , Amyloid Precursor Protein Secretases/genetics , Antigens, CD/genetics , Biomarkers, Tumor/genetics , Brain Neoplasms/enzymology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cadherins/genetics , Case-Control Studies , Cell Line, Tumor , Glioblastoma/enzymology , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Immunohistochemistry , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/genetics , Membrane Proteins/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics
17.
Exp Cell Res ; 322(1): 108-21, 2014 Mar 10.
Article in English | MEDLINE | ID: mdl-24246222

ABSTRACT

PACAP and its cognate peptide VIP participate in various biological functions, including myelin maturation and synthesis. However, defining whether these peptides affect peripheral expression of myelin proteins still remains unanswered. To address this issue, we assessed whether PACAP or VIP contribute to regulate the expression of three myelin proteins (MAG, MBP and MPZ, respectively) using the rat schwannoma cell line (RT4-P6D2T), a well-established model to study myelin gene expression. In addition, we endeavored to partly unravel the underlying molecular mechanisms involved. Expression of myelin-specific proteins was assessed in cells grown either in normal serum (10% FBS) or serum starved and treated with or without 100 nM PACAP or VIP. Furthermore, through pharmacological approach using the PACAP/VIP receptor antagonist (PACAP6-38) or specific pathway (MAPK or PI3K) inhibitors we defined the relative contribution of receptors and/or signaling pathways on the expression of myelin proteins. Our data show that serum starvation (24h) significantly increased both MAG, MBP and MPZ expression. Concurrently, we observed increased expression of endogenous PACAP and related receptors. Treatment with PACAP or VIP further exacerbated starvation-induced expression of myelin markers, suggesting that serum withdrawal might sensitize cells to peptide activity. Stimulation with either peptides increased phosphorylation of Akt at Ser473 residue but had no effect on phosphorylated Erk-1/2. PACAP6-38 (10 µM) impeded starvation- or peptide-induced expression of myelin markers. Similar effects were obtained after pretreatment with the PI3K inhibitor (wortmannin, 10 µM) but not the MAPKK inhibitor (PD98059, 50 µM). Together, the present finding corroborate the hypothesis that PACAP and VIP might contribute to the myelinating process preferentially via the canonical PI3K/Akt signaling pathway, providing the basis for future studies on the role of these peptides in demyelinating diseases.


Subject(s)
Myelin Proteins/genetics , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/physiology , Receptors, Vasoactive Intestinal Peptide, Type II/physiology , Schwann Cells/drug effects , Vasoactive Intestinal Peptide/pharmacology , Animals , Cell Line, Tumor , Enzyme Activation/physiology , Gene Expression Regulation, Neoplastic/drug effects , Myelin Basic Protein/genetics , Myelin Basic Protein/metabolism , Myelin P0 Protein/genetics , Myelin P0 Protein/metabolism , Myelin Proteins/metabolism , Myelin-Associated Glycoprotein/genetics , Myelin-Associated Glycoprotein/metabolism , Oncogene Protein v-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Rats , Schwann Cells/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology
18.
Int J Mol Sci ; 16(3): 5922-44, 2015 Mar 13.
Article in English | MEDLINE | ID: mdl-25782157

ABSTRACT

Osteoarthritis (OA); the most common form of degenerative joint disease, is associated with variations in pro-inflammatory growth factor levels, inflammation and hypocellularity resulting from chondrocyte apoptosis. Pituitary adenylate cyclase-activating polypeptide (PACAP) is a neuropeptide endowed with a range of trophic effects in several cell types; including chondrocytes. However; its role in OA has not been studied. To address this issue, we investigated whether PACAP expression is affected in OA cartilage obtained from experimentally-induced OA rat models, and then studied the effects of PACAP in isolated chondrocytes exposed to IL-1ß in vitro to mimic the inflammatory milieu of OA cartilage. OA induction was established by histomorphometric and histochemical analyses. Changes in PACAP distribution in cartilage, or its concentration in synovial fluid (SF), were assessed by immunohistochemistry and ELISA. Results showed that PACAP abundance in cartilage tissue and SF was high in healthy controls. OA induction decreased PACAP levels both in affected cartilage and SF. In vitro, PACAP prevented IL-1ß-induced chondrocyte apoptosis, as determined by MTT assay; Hoechst staining and western blots of apoptotic-related proteins. These changes were also accompanied by decreased i-NOS and COX-2 levels, suggesting an anti-inflammatory effect. Altogether, these findings support a potential role for PACAP as a chondroprotective agent for the treatment of OA.


Subject(s)
Cartilage, Articular/metabolism , Osteoarthritis/pathology , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Cartilage, Articular/pathology , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/drug effects , Chondrocytes/metabolism , Cyclooxygenase 2/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Interleukin-1beta/analysis , Interleukin-1beta/pharmacology , Male , Nitric Oxide Synthase Type II/metabolism , Osteoarthritis/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/analysis , Rats , Rats, Sprague-Dawley , Synovial Fluid/metabolism
19.
Biometals ; 27(6): 1149-58, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25053108

ABSTRACT

We have previously reported that nickel acetate (Ni(2+)), a well-known human carcinogenic agents, differentially affected apoptosis in two different airway epithelial cell lines derived from the human respiratory tract (A549 and Beas-2B, respectively), suggesting a potential involvement of epidermal growth factor receptor (EGFR)/Neu receptors in mediating this effect. Since ErbBs are closely associated to Mucin 1 (MUC1), a glycoprotein component of airway mucus that is overexpressed in lung tumors, we have investigated the role of this signaling system in the survival response of airway epithelial cells against Ni(2+)-induced cell death. We found that A549 cells exposed to Ni(2+) do not show any significant increase of MUC1 levels. Conversely, Beas-2B cells exposed to equivalent concentrations of Ni(2+) showed increased expression of MUC1 levels and this correlated with increased phosphorylation of both EGFR and of the extracellular-regulated kinase 1/2 (ERK1/2) and increase resistance to apoptosis, as indicated by cell viability assessments and DNA damage. Interestingly, suppression of MUC1 by small interfering RNA inhibited the EGFR activation in Beas-2B cells, leading to a significant decrease of survival and enhancement of DNA fragmentation and cleaved Caspase-3 expression. These results strongly suggest a role for MUC1 in Ni(2+)-induced neoplastic transformation, which likely involves the activation of the EGFR-mediated cell survival pathway, highlighting new avenues in the molecular approach to lung cancer prevention.


Subject(s)
Acetates/toxicity , Bronchi/cytology , Carcinogens/toxicity , Epithelial Cells/drug effects , Mucin-1/physiology , Organometallic Compounds/toxicity , Apoptosis/drug effects , Cell Line , ErbB Receptors/physiology , Humans , MAP Kinase Signaling System/drug effects , Mucin-1/genetics , Nucleosomes/metabolism , Pulmonary Alveoli/cytology , RNA Interference , RNA, Small Interfering/pharmacology , Signal Transduction/physiology , Transcriptional Activation/drug effects
20.
J Funct Morphol Kinesiol ; 9(1)2024 Jan 03.
Article in English | MEDLINE | ID: mdl-38249091

ABSTRACT

The habenular complex is a diencephalic structure divided into the medial and lateral divisions that lie within the epithalamus of most vertebrates. This brain structure, whose activities are mainly regulated via inputs/outputs from and to the stria medullaris and the fasciculus retroflexus, plays a significant role in the modulation of anti-reward behaviors in both the rodent and human brain. Such anti-reward circuits are regulated by dopaminergic and serotonergic projections with several other subcortical and cortical regions; therefore, it is plausible that impairment to this key subcortical structure or its connections contributes to the pathogenesis of affective disorders. Current literature reveals the existence of structural changes in the habenula complex in individuals afflicted by such disorders; however, there is a need for more comprehensive investigations to elucidate the underlying neuroanatomical connections that underpin disease development. In this review article, we aim to provide a comprehensive view of the neuroanatomical differences between the rodent and human habenular complex, the main circuitries, and provide an update on the emerging roles of this understudied subcortical structure in the control of affective behaviors, with special emphasis to morbid conditions of the affective sphere.

SELECTION OF CITATIONS
SEARCH DETAIL