Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
EMBO Rep ; 25(3): 991-1021, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38243137

ABSTRACT

Neuronal maturation is the phase during which neurons acquire their final characteristics in terms of morphology, electrical activity, and metabolism. However, little is known about the metabolic pathways governing neuronal maturation. Here, we investigate the contribution of the main metabolic pathways, namely glucose, glutamine, and fatty acid oxidation, during the maturation of primary rat hippocampal neurons. Blunting glucose oxidation through the genetic and chemical inhibition of the mitochondrial pyruvate transporter reveals that this protein is critical for the production of glutamate, which is required for neuronal arborization, proper dendritic elongation, and spine formation. Glutamate supplementation in the early phase of differentiation restores morphological defects and synaptic function in mitochondrial pyruvate transporter-inhibited cells. Furthermore, the selective activation of metabotropic glutamate receptors restores the impairment of neuronal differentiation due to the reduced generation of glucose-derived glutamate and rescues synaptic local translation. Fatty acid oxidation does not impact neuronal maturation. Whereas glutamine metabolism is important for mitochondria, it is not for endogenous glutamate production. Our results provide insights into the role of glucose-derived glutamate as a key player in neuronal terminal differentiation.


Subject(s)
Glutamine , Monocarboxylic Acid Transporters , Rats , Animals , Glutamine/metabolism , Monocarboxylic Acid Transporters/metabolism , Neurons/metabolism , Glutamic Acid/metabolism , Glucose/metabolism , Fatty Acids/metabolism
2.
Aging Cell ; 22(9): e13918, 2023 09.
Article in English | MEDLINE | ID: mdl-37537790

ABSTRACT

Frailty is a geriatric syndrome that results from multisystem impairment caused by age-associated accumulation of deficits. The frailty index is used to define the level of frailty. Several studies have searched for molecular biomarkers associated with frailty, to meet the needs for personalized care. Cyclase-associated protein 2 (CAP2) is a multifunctional actin-binding protein involved in various physiological and pathological processes, that might reflect frailty's intrinsic complexity. This study aimed to investigate the association between frailty index and circulating CAP2 concentration in 467 community-dwelling older adults (median age: 79; range: 65-92 years) from Milan, Italy. The selected robust regression model showed that circulating CAP2 concentration was not associated with chronological age, as well as sex and education. However, circulating CAP2 concentration was significantly and inversely associated with the frailty index: a 0.1-unit increase in frailty index leads to ~0.5-point mean decrease in CAP2 concentration. Furthermore, mean CAP2 concentration was significantly lower in frail participants (i.e., frailty index ≥0.25) than in non-frail participants. This study shows the association between serum CAP2 concentration and frailty status for the first time, highlighting the potential of CAP2 as a biomarker for age-associated accumulation of deficits.


Subject(s)
Adaptor Proteins, Signal Transducing , Frailty , Membrane Proteins , Aged , Humans , Biomarkers/blood , Cross-Sectional Studies , Frail Elderly , Frailty/blood , Geriatric Assessment/methods , Independent Living , Membrane Proteins/blood , Adaptor Proteins, Signal Transducing/blood
3.
ACS Chem Neurosci ; 14(15): 2727-2742, 2023 08 02.
Article in English | MEDLINE | ID: mdl-37474114

ABSTRACT

Serotonergic psychedelics are described to have activation of the serotonin 2A receptor (5-HT2A) as their main pharmacological action. Despite their relevance, the molecular mechanisms underlying the psychedelic effects induced by certain 5-HT2A agonists remain elusive. One of the proposed hypotheses is the occurrence of biased agonism, defined as the preferential activation of certain signaling pathways over others. This study comparatively monitored the efficiency of a diverse panel of 4-position-substituted (and N-benzyl-derived) phenylalkylamines to induce recruitment of ß-arrestin2 (ßarr2) or miniGαq to the 5-HT2A, allowing us to assess structure-activity relationships and biased agonism. All test compounds exhibited agonist properties with a relatively large range of both EC50 and Emax values. Interestingly, the lipophilicity of the 2C-X phenethylamines was correlated with their efficacy in both assays but yielded a stronger correlation in the miniGαq- than in the ßarr2-assay. Molecular docking suggested that accommodation of the 4-substituent of the 2C-X analogues in a hydrophobic pocket between transmembrane helices 4 and 5 of 5-HT2A may contribute to this differential effect. Aside from previously used standard conditions (lysergic acid diethylamide (LSD) as a reference agonist and a 2 h activation profile to assess a compound's activity), serotonin was included as a second reference agonist, and the compounds' activities were also assessed using the first 30 min of the activation profile. Under all assessed circumstances, the qualitative structure-activity relationships remained unchanged. Furthermore, the use of two reference agonists allowed for the estimation of both "benchmark bias" (relative to LSD) and "physiology bias" (relative to serotonin).


Subject(s)
Hallucinogens , Serotonin , Receptor, Serotonin, 5-HT2A , Molecular Docking Simulation , Hallucinogens/pharmacology , Hallucinogens/chemistry , Phenethylamines/pharmacology , Serotonin 5-HT2 Receptor Agonists/pharmacology
4.
Commun Biol ; 6(1): 42, 2023 01 13.
Article in English | MEDLINE | ID: mdl-36639734

ABSTRACT

The Membrane Attack Complex (MAC) is responsible for forming large ß-barrel channels in the membranes of pathogens, such as gram-negative bacteria. Off-target MAC assembly on endogenous tissue is associated with inflammatory diseases and cancer. Accordingly, a human C5b-9 specific antibody, aE11, has been developed that detects a neoepitope exposed in C9 when it is incorporated into the C5b-9 complex, but not present in the plasma native C9. For nearly four decades aE11 has been routinely used to study complement, MAC-related inflammation, and pathophysiology. However, the identity of C9 neoepitope remains unknown. Here, we determined the cryo-EM structure of aE11 in complex with polyC9 at 3.2 Å resolution. The aE11 binding site is formed by two separate surfaces of the oligomeric C9 periphery and is therefore a discontinuous quaternary epitope. These surfaces are contributed by portions of the adjacent TSP1, LDLRA, and MACPF domains of two neighbouring C9 protomers. By substituting key antibody interacting residues to the murine orthologue, we validated the unusual binding modality of aE11. Furthermore, aE11 can recognise a partial epitope in purified monomeric C9 in vitro, albeit weakly. Taken together, our results reveal the structural basis for MAC recognition by aE11.


Subject(s)
Complement C9 , Complement Membrane Attack Complex , Humans , Animals , Mice , Complement Membrane Attack Complex/metabolism , Complement C5b , Complement C9/chemistry , Complement C9/metabolism , Complement System Proteins/metabolism , Epitopes
5.
J Med Chem ; 65(18): 12031-12043, 2022 09 22.
Article in English | MEDLINE | ID: mdl-36099411

ABSTRACT

The serotonin 2A receptor (5-HT2AR) is the mediator of the psychedelic effects of serotonergic psychedelics, which have shown promising results in clinical studies for several neuropsychiatric indications. The 5-HT2AR is able to signal through the Gαq and ß-arrestin effector proteins, but it is currently not known how the different signaling pathways contribute to the therapeutic effects mediated by serotonergic psychedelics. In the present work, we have evaluated the subtype-selective 5-HT2AR agonist 25CN-NBOH and a series of close analogues for biased signaling at this receptor. These ligands were designed to evaluate the role of interactions with Ser1593×36. The lack of interaction between this hydroxyl moiety and Ser1593×36 resulted in detrimental effects on potency and efficacy in both ßarr2 and miniGαq recruitment assays. Remarkably, Gαq-mediated signaling was considerably more affected. This led to the development of the first efficacious ßarr2-biased 5-HT2AR agonists 4a-b and 6e-f, ßarr2 preferring, relative to lysergic acid diethylamide (LSD).


Subject(s)
Hallucinogens , Lysergic Acid Diethylamide , Hallucinogens/pharmacology , Lysergic Acid Diethylamide/pharmacology , Receptor, Serotonin, 5-HT2A , Serotonin , Serotonin 5-HT2 Receptor Agonists/pharmacology , beta-Arrestins
6.
Nat Struct Mol Biol ; 29(8): 767-773, 2022 08.
Article in English | MEDLINE | ID: mdl-35864164

ABSTRACT

P-Rex (PI(3,4,5)P3-dependent Rac exchanger) guanine nucleotide exchange factors potently activate Rho GTPases. P-Rex guanine nucleotide exchange factors are autoinhibited, synergistically activated by Gßγ and PI(3,4,5)P3 binding and dysregulated in cancer. Here, we use X-ray crystallography, cryogenic electron microscopy and crosslinking mass spectrometry to determine the structural basis of human P-Rex1 autoinhibition. P-Rex1 has a bipartite structure of N- and C-terminal modules connected by a C-terminal four-helix bundle that binds the N-terminal Pleckstrin homology (PH) domain. In the N-terminal module, the Dbl homology (DH) domain catalytic surface is occluded by the compact arrangement of the DH-PH-DEP1 domains. Structural analysis reveals a remarkable conformational transition to release autoinhibition, requiring a 126° opening of the DH domain hinge helix. The off-axis position of Gßγ and PI(3,4,5)P3 binding sites further suggests a counter-rotation of the P-Rex1 halves by 90° facilitates PH domain uncoupling from the four-helix bundle, releasing the autoinhibited DH domain to drive Rho GTPase signaling.


Subject(s)
Guanine Nucleotide Exchange Factors/chemistry , Neoplasms , Binding Sites , Guanine Nucleotide Exchange Factors/metabolism , Humans , Neoplasm Metastasis , Neoplasms/metabolism , Protein Domains , Signal Transduction
7.
Cell Death Dis ; 13(7): 616, 2022 07 16.
Article in English | MEDLINE | ID: mdl-35842432

ABSTRACT

Interest in the function of ataxia-telangiectasia-mutated protein (ATM) is extensively growing as evidenced by preclinical studies that continuously link ATM with new intracellular pathways. Here, we exploited Atm+/- and Atm-/- mice and demonstrate that cognitive defects are rescued by the delivery of the antidepressant Fluoxetine (Fluox). Fluox increases levels of the chloride intruder NKCC1 exclusively at hippocampal level suggesting an ATM context-specificity. A deeper investigation of synaptic composition unveils increased Gluk-1 and Gluk-5 subunit-containing kainate receptors (KARs) levels in the hippocampus, but not in the cortex, of Atm+/- and Atm-/- mice. Analysis of postsynaptic fractions and confocal studies indicates that KARs are presynaptic while in vitro and ex vivo electrophysiology that are fully active. These changes are (i) linked to KCC2 activity, as the KCC2 blockade in Atm+/- developing neurons results in reduced KARs levels and (ii) developmental regulated. Indeed, the pharmacological inhibition of ATM kinase in adults produces different changes as identified by RNA-seq investigation. Our data display how ATM affects both inhibitory and excitatory neurotransmission, extending its role to a variety of neurological and psychiatric disorders.


Subject(s)
Ataxia Telangiectasia Mutated Proteins , Hippocampus , Symporters , Animals , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Hippocampus/metabolism , Humans , Mice , Neurons/metabolism , Receptors, Kainic Acid , Symporters/genetics , Symporters/metabolism , Synaptic Transmission/physiology
8.
Mol Ther ; 30(7): 2474-2490, 2022 07 06.
Article in English | MEDLINE | ID: mdl-35390543

ABSTRACT

The development of new therapeutic avenues that target the early stages of Alzheimer's disease (AD) is urgently necessary. A disintegrin and metalloproteinase domain 10 (ADAM10) is a sheddase that is involved in dendritic spine shaping and limits the generation of amyloid-ß. ADAM10 endocytosis increases in the hippocampus of AD patients, resulting in the decreased postsynaptic localization of the enzyme. To restore this altered pathway, we developed a cell-permeable peptide (PEP3) with a strong safety profile that is able to interfere with ADAM10 endocytosis, upregulating the postsynaptic localization and activity of ADAM10. After extensive validation, experiments in a relevant animal model clarified the optimal timing of the treatment window. PEP3 administration was effective for the rescue of cognitive defects in APP/PS1 mice only if administered at an early disease stage. Increased ADAM10 activity promoted synaptic plasticity, as revealed by changes in the molecular compositions of synapses and the spine morphology. Even though further studies are required to evaluate efficacy and safety issues of long-term administration of PEP3, these results provide preclinical evidence to support the therapeutic potential of PEP3 in AD.


Subject(s)
Alzheimer Disease , ADAM10 Protein/genetics , ADAM10 Protein/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Disease Models, Animal , Endocytosis , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Synapses/metabolism
9.
Cell Chem Biol ; 29(4): 586-596.e4, 2022 04 21.
Article in English | MEDLINE | ID: mdl-34699747

ABSTRACT

Harnessing the immunomodulatory activity of cytokines is a focus of therapies targeting inflammatory disease. The interleukin (IL)-1 superfamily contains pro-inflammatory and anti-inflammatory members that help orchestrate the immune response in adaptive and innate immunity. Of these molecules, IL-37 has robust anti-inflammatory activity across a range of disease models through inhibition of pro-inflammatory signaling cascades downstream of tumor necrosis factor, IL-1, and toll-like receptor pathways. We find that IL-37 is unstable with a poor pharmacokinetic and manufacturing profile. Here, we present the engineering of IL-37 from an unstable cytokine into an anti-inflammatory molecule with an excellent therapeutic likeness. We overcame these shortcomings through site-directed mutagenesis, the addition of a non-native disulfide bond, and the engineering of IL-37 as an Fc-fusion protein. Our results provide a platform for preclinical testing of IL-37 Fc-fusion proteins. The engineering approaches undertaken herein will apply to the conversion of similar potent yet short-acting cytokines into therapeutics.


Subject(s)
Anti-Inflammatory Agents , Cytokines , Cytokines/metabolism , Immunity, Innate , Immunomodulation , Protein Engineering
10.
Nat Struct Mol Biol ; 28(12): 982-988, 2021 12.
Article in English | MEDLINE | ID: mdl-34887559

ABSTRACT

Neurofibromin (NF1) mutations cause neurofibromatosis type 1 and drive numerous cancers, including breast and brain tumors. NF1 inhibits cellular proliferation through its guanosine triphosphatase-activating protein (GAP) activity against rat sarcoma (RAS). In the present study, cryo-electron microscope studies reveal that the human ~640-kDa NF1 homodimer features a gigantic 30 × 10 nm array of α-helices that form a core lemniscate-shaped scaffold. Three-dimensional variability analysis captured the catalytic GAP-related domain and lipid-binding SEC-PH domains positioned against the core scaffold in a closed, autoinhibited conformation. We postulate that interaction with the plasma membrane may release the closed conformation to promote RAS inactivation. Our structural data further allow us to map the location of disease-associated NF1 variants and provide a long-sought-after structural explanation for the extreme susceptibility of the molecule to loss-of-function mutations. Collectively these findings present potential new routes for therapeutic modulation of the RAS pathway.


Subject(s)
GTPase-Activating Proteins/metabolism , Neurofibromatosis 1/genetics , Neurofibromin 1/metabolism , ras Proteins/metabolism , Cell Membrane/metabolism , Cell Proliferation/genetics , Cryoelectron Microscopy , Humans , Loss of Function Mutation/genetics , Neurofibromatosis 1/pathology , Neurofibromin 1/genetics , Protein Conformation
11.
Biomolecules ; 11(9)2021 08 24.
Article in English | MEDLINE | ID: mdl-34572474

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disorder representing the most common form of dementia. It is biologically characterized by the deposition of extracellular amyloid-ß (Aß) senile plaques and intracellular neurofibrillary tangles, constituted by hyperphosphorylated tau protein. The key protein in AD pathogenesis is the amyloid precursor protein (APP), which is cleaved by secretases to produce several metabolites, including Aß and APP intracellular domain (AICD). The greatest genetic risk factor associated with AD is represented by the Apolipoprotein E ε4 (APOE ε4) allele. Importantly, all of the above-mentioned molecules that are strictly related to AD pathogenesis have also been described as playing roles in the cell nucleus. Accordingly, evidence suggests that nuclear functions are compromised in AD. Furthermore, modulation of transcription maintains cellular homeostasis, and alterations in transcriptomic profiles have been found in neurodegenerative diseases. This report reviews recent advancements in the AD players-mediated gene expression. Aß, tau, AICD, and APOE ε4 localize in the nucleus and regulate the transcription of several genes, part of which is involved in AD pathogenesis, thus suggesting that targeting nuclear functions might provide new therapeutic tools for the disease.


Subject(s)
Alzheimer Disease/pathology , Cell Nucleus/pathology , Amyloid beta-Peptides/metabolism , Animals , Apolipoprotein E4/metabolism , Humans , Models, Biological , tau Proteins/metabolism
12.
Sci Signal ; 14(681)2021 05 04.
Article in English | MEDLINE | ID: mdl-33947796

ABSTRACT

The dual-specificity phosphatase PTEN functions as a tumor suppressor by hydrolyzing PI(3,4,5)P3 to PI(4,5)P2 to inhibit PI3K-AKT signaling and cellular proliferation. P-Rex2 is a guanine nucleotide exchange factor for Rho GTPases and can be activated by Gßγ subunits downstream of G protein-coupled receptor signaling and by PI(3,4,5)P3 downstream of receptor tyrosine kinases. The PTEN:P-Rex2 complex is a commonly mutated signaling node in metastatic cancer. Assembly of the PTEN:P-Rex2 complex inhibits the activity of both proteins, and its dysregulation can drive PI3K-AKT signaling and cellular proliferation. Here, using cross-linking mass spectrometry and functional studies, we gained mechanistic insights into PTEN:P-Rex2 complex assembly and coinhibition. We found that PTEN was anchored to P-Rex2 by interactions between the PDZ-interacting motif in the PTEN C-terminal tail and the second PDZ domain of P-Rex2. This interaction bridged PTEN across the P-Rex2 surface, preventing PI(3,4,5)P3 hydrolysis. Conversely, PTEN both allosterically promoted an autoinhibited conformation of P-Rex2 and blocked its binding to Gßγ. In addition, we observed that the PTEN-deactivating mutations and P-Rex2 truncations combined to drive Rac1 activation to a greater extent than did either single variant alone. These insights enabled us to propose a class of gain-of-function, cancer-associated mutations within the PTEN:P-Rex2 interface that uncouple PTEN from the inhibition of Rac1 signaling.


Subject(s)
Guanine Nucleotide Exchange Factors , Neoplasms , PTEN Phosphohydrolase , rac1 GTP-Binding Protein , Guanine Nucleotide Exchange Factors/metabolism , Humans , Mutation , Neoplasms/genetics , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases , Signal Transduction , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism
13.
Oncogene ; 40(23): 4033-4049, 2021 06.
Article in English | MEDLINE | ID: mdl-34017076

ABSTRACT

Intrahepatic cholangiocarcinoma (iCCA) is a rare malignancy of the intrahepatic biliary tract with a very poor prognosis. Although some clinicopathological parameters can be prognostic factors for iCCA, the molecular prognostic markers and potential mechanisms of iCCA have not been well investigated. Here, we report that the Fragile X mental retardation protein (FMRP), a RNA binding protein functionally absent in patients with the Fragile X syndrome (FXS) and also involved in several types of cancers, is overexpressed in human iCCA and its expression is significantly increased in iCCA metastatic tissues. The silencing of FMRP in metastatic iCCA cell lines affects cell migration and invasion, suggesting a role of FMRP in iCCA progression. Moreover, we show evidence that FMRP is localized at the invasive front of human iCCA neoplastic nests and in pseudopodia and invadopodia protrusions of migrating and invading iCCA cancer cells. Here FMRP binds several mRNAs encoding key proteins involved in the formation and/or function of these protrusions. In particular, we find that FMRP binds to and regulates the expression of Cortactin, a critical regulator of invadopodia formation. Altogether, our findings suggest that FMRP could promote cell invasiveness modulating membrane plasticity and invadopodia formation at the leading edges of invading iCCA cells.


Subject(s)
Bile Duct Neoplasms/metabolism , Cholangiocarcinoma/metabolism , Fragile X Mental Retardation Protein/metabolism , Animals , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/pathology , Cell Line, Tumor , Cell Plasticity/physiology , Cholangiocarcinoma/genetics , Cholangiocarcinoma/pathology , Cortactin/metabolism , Humans , Male , Mice, Nude , Neoplasm Metastasis , Podosomes/metabolism , Xenograft Model Antitumor Assays
14.
Hum Genet ; 139(2): 227-245, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31919630

ABSTRACT

Fragile X-related disorders are due to a dynamic mutation of the CGG repeat at the 5' UTR of the FMR1 gene, coding for the RNA-binding protein FMRP. As the CGG sequence expands from premutation (PM, 56-200 CGGs) to full mutation (> 200 CGGs), FMRP synthesis decreases until it is practically abolished in fragile X syndrome (FXS) patients, mainly due to FMR1 methylation. Cells from rare individuals with no intellectual disability and carriers of an unmethylated full mutation (UFM) produce slightly elevated levels of FMR1-mRNA and relatively low levels of FMRP, like in PM carriers. With the aim of clarifying how UFM cells differ from CTRL and FXS cells, a comparative proteomic approach was undertaken, from which emerged an overexpression of SOD2 in UFM cells, also confirmed in PM but not in FXS. The SOD2-mRNA bound to FMRP in UFM more than in the other cell types. The high SOD2 levels in UFM and PM cells correlated with lower levels of superoxide and reactive oxygen species (ROS), and with morphological anomalies and depolarization of the mitochondrial membrane detected through confocal microscopy. The same effect was observed in CTRL and FXS after treatment with MC2791, causing SOD2 overexpression. These mitochondrial phenotypes reverted after knock-down with siRNA against SOD2-mRNA and FMR1-mRNA in UFM and PM. Overall, these data suggest that in PM and UFM carriers, which have high levels of FMR1 transcription and may develop FXTAS, SOD2 overexpression helps to maintain low levels of both superoxide and ROS with signs of mitochondrial degradation.


Subject(s)
Ataxia/pathology , DNA Methylation , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/pathology , Mitochondria/pathology , Mitochondrial Proteins/metabolism , Mutation , Proteome/analysis , Tremor/pathology , Ataxia/genetics , Ataxia/metabolism , Case-Control Studies , Cells, Cultured , Fibroblasts/metabolism , Fibroblasts/pathology , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Fragile X Syndrome/metabolism , Humans , Male , Mitochondria/metabolism , Mitochondrial Proteins/genetics , RNA, Small Interfering/genetics , Superoxide Dismutase/antagonists & inhibitors , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Tremor/genetics , Tremor/metabolism
15.
Cells ; 9(1)2020 01 10.
Article in English | MEDLINE | ID: mdl-31936823

ABSTRACT

Interleukin (IL)-37 is a member of the IL-1 family of cytokines. Although its broad anti-inflammatory properties are well described, the effects of IL-37 on inflammasome function remain poorly understood. Performing gene expression analyses, ASC oligomerization/speck assays and caspase-1 assays in bone marrow-derived macrophages (BMDM), and employing an in vivo endotoxemia model, we studied how IL-37 affects the expression and maturation of IL-1ß and IL-18, inflammasome activation, and pyroptosis in detail. IL-37 inhibited IL-1ß production by NLRP3 and AIM2 inflammasomes, and IL-18 production by the NLRP3 inflammasome. This inhibition was partially attributable to effects on gene expression: whereas IL-37 did not affect lipopolysaccharide (LPS)-induced mRNA expression of Il18 or inflammasome components, IL-37-transgenic BMDM displayed an up to 83% inhibition of baseline and LPS-stimulated Il1b compared to their wild-type counterparts. Importantly, we observed that IL-37 suppresses nigericin- and silica-induced ASC oligomerization/speck formation (a step in inflammasome activation and subsequent caspase-1 activation), and pyroptosis (-50%). In mice subjected to endotoxemia, IL-37 inhibited plasma IL-1ß (-78% compared to wild-type animals) and IL-18 (-61%). Thus, our study adds suppression of inflammasome activity to the portfolio of anti-inflammatory pathways employed by IL-37, highlighting this cytokine as a potential tool for treating inflammasome-driven diseases.


Subject(s)
Inflammasomes/metabolism , Interleukin-1/metabolism , Interleukins/metabolism , Animals , Cells, Cultured , Interleukin-1/analysis , Interleukins/analysis , Mice , Mice, Inbred C57BL , Mice, Transgenic
17.
Hum Mol Genet ; 27(12): 2039-2051, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29590342

ABSTRACT

Fragile X syndrome (FXS) is a monogenic form of intellectual disability and autism spectrum disorder caused by the absence of the fragile X mental retardation protein (FMRP). In biological models for the disease, this leads to upregulated mRNA translation and as a consequence, deficits in synaptic architecture and plasticity. Preclinical studies revealed that pharmacological interventions restore those deficits, which are thought to mediate the FXS cognitive and behavioral symptoms. Here, we characterized the de novo rate of protein synthesis in patients with FXS and their relationship with clinical severity. We measured the rate of protein synthesis in fibroblasts derived from 32 individuals with FXS and from 17 controls as well as in fibroblasts and primary neurons of 27 Fmr1 KO mice and 20 controls. Here, we show that levels of protein synthesis are increased in fibroblasts of individuals with FXS and Fmr1 KO mice. However, this cellular phenotype displays a broad distribution and a proportion of fragile X individuals and Fmr1 KO mice do not show increased levels of protein synthesis, having measures in the normal range. Because the same Fmr1 KO animal measures in fibroblasts predict those in neurons we suggest the validity of this peripheral biomarker. Our study offers a potential explanation for the comprehensive drug development program undertaken thus far yielding negative results and suggests that a significant proportion, but not all individuals with FXS, may benefit from the reduction of excessive levels of protein synthesis.


Subject(s)
Autism Spectrum Disorder/genetics , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Adolescent , Adult , Aged , Animals , Autism Spectrum Disorder/physiopathology , Child , Disease Models, Animal , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Fragile X Mental Retardation Protein/biosynthesis , Fragile X Syndrome/physiopathology , Hippocampus/metabolism , Hippocampus/physiopathology , Humans , Male , Mice , Mice, Knockout , Middle Aged , Neurons/metabolism , Neurons/pathology , Young Adult
18.
Sci Signal ; 10(504)2017 Nov 07.
Article in English | MEDLINE | ID: mdl-29114037

ABSTRACT

Fragile X syndrome (FXS) is the most common form of inherited intellectual disability and autism spectrum disorder. FXS is caused by silencing of the FMR1 gene, which encodes fragile X mental retardation protein (FMRP), an mRNA-binding protein that represses the translation of its target mRNAs. One mechanism by which FMRP represses translation is through its association with cytoplasmic FMRP-interacting protein 1 (CYFIP1), which subsequently sequesters and inhibits eukaryotic initiation factor 4E (eIF4E). CYFIP1 shuttles between the FMRP-eIF4E complex and the Rac1-Wave regulatory complex, thereby connecting translational regulation to actin dynamics and dendritic spine morphology, which are dysregulated in FXS model mice that lack FMRP. Treating FXS mice with 4EGI-1, which blocks interactions between eIF4E and eIF4G, a critical interaction partner for translational initiation, reversed defects in hippocampus-dependent memory and spine morphology. We also found that 4EGI-1 normalized the phenotypes of enhanced metabotropic glutamate receptor (mGluR)-mediated long-term depression (LTD), enhanced Rac1-p21-activated kinase (PAK)-cofilin signaling, altered actin dynamics, and dysregulated CYFIP1/eIF4E and CYFIP1/Rac1 interactions in FXS mice. Our findings are consistent with the idea that an imbalance in protein synthesis and actin dynamics contributes to pathophysiology in FXS mice, and suggest that targeting eIF4E may be a strategy for treating FXS.


Subject(s)
Actins/metabolism , Eukaryotic Initiation Factor-4E/metabolism , Eukaryotic Initiation Factor-4G/metabolism , Fragile X Syndrome/metabolism , Adaptor Proteins, Signal Transducing , Animals , Dendritic Spines/drug effects , Disease Models, Animal , Eukaryotic Initiation Factor-4E/antagonists & inhibitors , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4G/antagonists & inhibitors , Eukaryotic Initiation Factor-4G/genetics , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/drug therapy , Hippocampus/drug effects , Hippocampus/physiopathology , Hydrazones/pharmacology , Hydrazones/therapeutic use , Male , Memory Disorders/drug therapy , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Thiazoles/pharmacology , Thiazoles/therapeutic use
19.
Sci Immunol ; 2(8)2017 Feb 10.
Article in English | MEDLINE | ID: mdl-28783685

ABSTRACT

Dysregulation of the inflammatory response underlies numerous diseases. Although most interleukin-1 family cytokines are proinflammatory, human interleukin-37 (IL-37) is a powerful, broad-spectrum inhibitor of inflammation and immunity. We determined the crystal structure of IL-37 to establish the anti-inflammatory mechanism of this key cytokine in view of developing IL-37-based therapies. We found that two ß-trefoil fold IL-37 molecules form a head-to-head dimer that is stable in solution. IL-37 variants mutated to convert the cytokine into an obligate monomer were up to 13-fold more effective than the dimer in suppressing proinflammatory events both in primary human blood cells and in vivo in murine endotoxic shock. Therapeutic exploitation of the powerful anti-inflammatory properties of monomeric IL-37 may prove beneficial in treating a wide range of inflammatory and autoimmune disorders.

20.
Neuropharmacology ; 116: 71-81, 2017 04.
Article in English | MEDLINE | ID: mdl-28012946

ABSTRACT

Many clinical and molecular features of the fragile X syndrome, a common form of intellectual disability and autism, can be modeled by deletion of the Fmr1 protein (Fmrp) in mice. Previous studies showed a decreased expression of several components of the GABAergic system in Fmr1 knockout mice. Here, we used this mouse model to investigate the functional consequences of Fmrp deletion on hippocampal GABAergic inhibition in the CA1-region of the hippocampus. Whole-cell patch-clamp recordings demonstrated a significantly reduced amplitude of evoked inhibitory postsynaptic currents (eIPSCs) and a decrease in the amplitude and frequency of spontaneous IPSCs. In addition, miniature IPSCs were reduced in amplitude and frequency and decayed significantly slower than mIPSCs in controls. Quantitative real-time PCR revealed a significantly lower expression of α2, ß1 and δ GABAA receptor subunits in the hippocampus of the juvenile mice (P22) compared to wild-type littermates. Correspondingly, we found also at the protein level reduced amounts of α2, ß1 and δ subunits in Fmr1 knockout mice. Overall, these results demonstrate that the reduction in several components of the GABAergic system is already present at young age and that this reduction results in measurable abnormalities on GABAA receptor-mediated phasic inhibition. These abnormalities might contribute to the behavioral and cognitive deficits of this fragile X mouse model.


Subject(s)
CA1 Region, Hippocampal/metabolism , Fragile X Mental Retardation Protein/metabolism , Inhibitory Postsynaptic Potentials/physiology , Pyramidal Cells/metabolism , Receptors, GABA-A/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , CA1 Region, Hippocampal/drug effects , Disease Models, Animal , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/metabolism , GABA-A Receptor Antagonists/pharmacology , Inhibitory Postsynaptic Potentials/drug effects , Male , Mice, Inbred C57BL , Mice, Knockout , Miniature Postsynaptic Potentials/drug effects , Miniature Postsynaptic Potentials/physiology , Patch-Clamp Techniques , Pyramidal Cells/drug effects , RNA, Messenger/metabolism , Tissue Culture Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...