Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Nat Immunol ; 23(2): 275-286, 2022 02.
Article in English | MEDLINE | ID: mdl-35102342

ABSTRACT

The humoral arm of innate immunity includes diverse molecules with antibody-like functions, some of which serve as disease severity biomarkers in coronavirus disease 2019 (COVID-19). The present study was designed to conduct a systematic investigation of the interaction of human humoral fluid-phase pattern recognition molecules (PRMs) with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Of 12 PRMs tested, the long pentraxin 3 (PTX3) and mannose-binding lectin (MBL) bound the viral nucleocapsid and spike proteins, respectively. MBL bound trimeric spike protein, including that of variants of concern (VoC), in a glycan-dependent manner and inhibited SARS-CoV-2 in three in vitro models. Moreover, after binding to spike protein, MBL activated the lectin pathway of complement activation. Based on retention of glycosylation sites and modeling, MBL was predicted to recognize the Omicron VoC. Genetic polymorphisms at the MBL2 locus were associated with disease severity. These results suggest that selected humoral fluid-phase PRMs can play an important role in resistance to, and pathogenesis of, COVID-19, a finding with translational implications.


Subject(s)
COVID-19/immunology , Immunity, Humoral , Receptors, Pattern Recognition/immunology , SARS-CoV-2/immunology , Animals , C-Reactive Protein/immunology , C-Reactive Protein/metabolism , COVID-19/metabolism , COVID-19/virology , Case-Control Studies , Chlorocebus aethiops , Complement Activation , Coronavirus Nucleocapsid Proteins/genetics , Coronavirus Nucleocapsid Proteins/immunology , Coronavirus Nucleocapsid Proteins/metabolism , Female , Glycosylation , HEK293 Cells , Host-Pathogen Interactions , Humans , Male , Mannose-Binding Lectin/genetics , Mannose-Binding Lectin/immunology , Mannose-Binding Lectin/metabolism , Phosphoproteins/genetics , Phosphoproteins/immunology , Phosphoproteins/metabolism , Polymorphism, Genetic , Protein Binding , Receptors, Pattern Recognition/genetics , Receptors, Pattern Recognition/metabolism , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , SARS-CoV-2/pathogenicity , Serum Amyloid P-Component/immunology , Serum Amyloid P-Component/metabolism , Signal Transduction , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/metabolism , Vero Cells
2.
Nat Immunol ; 20(8): 1012-1022, 2019 08.
Article in English | MEDLINE | ID: mdl-31263276

ABSTRACT

The plasma membrane tetraspan molecule MS4A4A is selectively expressed by macrophage-lineage cells, but its function is unknown. Here we report that MS4A4A was restricted to murine and human mononuclear phagocytes and was induced during monocyte-to-macrophage differentiation in the presence of interleukin 4 or dexamethasone. Human MS4A4A was co-expressed with M2/M2-like molecules in subsets of normal tissue-resident macrophages, infiltrating macrophages from inflamed synovium and tumor-associated macrophages. MS4A4A interacted and colocalized with the ß-glucan receptor dectin-1 in lipid rafts. In response to dectin-1 ligands, Ms4a4a-deficient macrophages showed defective signaling and defective production of effector molecules. In experimental models of tumor progression and metastasis, Ms4a4a deficiency in macrophages had no impact on primary tumor growth, but was essential for dectin-1-mediated activation of macrophages and natural killer (NK) cell-mediated metastasis control. Thus, MS4A4A is a tetraspan molecule selectively expressed in macrophages during differentiation and polarization, essential for dectin-1-dependent activation of NK cell-mediated resistance to metastasis.


Subject(s)
Killer Cells, Natural/immunology , Lectins, C-Type/metabolism , Macrophages/immunology , Membrane Proteins/metabolism , Neoplasm Metastasis/immunology , Neoplasms/immunology , Animals , Cell Differentiation/immunology , Cell Lineage , Dexamethasone/pharmacology , Humans , Interleukin-4/metabolism , Lymphocyte Activation/immunology , Macrophage Activation/immunology , Macrophages/cytology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Neoplasm Metastasis/prevention & control , Neoplasms/pathology
4.
Int J Mol Sci ; 23(2)2022 Jan 08.
Article in English | MEDLINE | ID: mdl-35054869

ABSTRACT

The retinal pigmented epithelium (RPE) plays a pivotal role in retinal homeostasis. It is therefore an interesting target to fill the unmet medical need of different retinal diseases, including age-related macular degeneration and Stargardt disease. RPE replacement therapy may use different cellular sources: induced pluripotent stem cells or embryonic stem cells. Cells can be transferred as suspension on a patch with different surgical approaches. Results are promising although based on very limited samples. In this review, we summarize the current progress of RPE replacement and provide a comparative assessment of different published approaches which may become standard of care in the future.


Subject(s)
Ophthalmologists , Retinal Pigment Epithelium/pathology , Translational Research, Biomedical , Clinical Trials as Topic , Humans , Macular Degeneration/therapy , Stargardt Disease/therapy
5.
Stem Cells ; 37(7): 973-987, 2019 07.
Article in English | MEDLINE | ID: mdl-30942926

ABSTRACT

Mesenchymal stem cells (MSCs) are well established to have promising therapeutic properties. TNF-stimulated gene-6 (TSG-6), a potent tissue-protective and anti-inflammatory factor, has been demonstrated to be responsible for a significant part of the tissue-protecting properties mediated by MSCs. Nevertheless, current knowledge about the biological function of TSG-6 in MSCs is limited. Here, we demonstrated that TSG-6 is a crucial factor that influences many functional properties of MSCs. The transcriptomic sequencing analysis of wild-type (WT) and TSG-6-/- -MSCs shows that the loss of TSG-6 expression leads to the perturbation of several transcription factors, cytokines, and other key biological pathways. TSG-6-/- -MSCs appeared morphologically different with dissimilar cytoskeleton organization, significantly reduced size of extracellular vesicles, decreased cell proliferative rate, and loss of differentiation abilities compared with the WT cells. These cellular effects may be due to TSG-6-mediated changes in the extracellular matrix (ECM) environment. The supplementation of ECM with exogenous TSG-6, in fact, rescued cell proliferation and changes in morphology. Importantly, TSG-6-deficient MSCs displayed an increased capacity to release interleukin-6 conferring pro-inflammatory and pro-tumorigenic properties to the MSCs. Overall, our data provide strong evidence that TSG-6 is crucial for the maintenance of stemness and other biological properties of murine MSCs.


Subject(s)
Cell Adhesion Molecules/genetics , Cell Transformation, Neoplastic/genetics , Interleukin-6/genetics , Mesenchymal Stem Cells/metabolism , Transcriptome , Animals , Autocrine Communication/genetics , Cell Adhesion Molecules/deficiency , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Cytokines/genetics , Cytokines/metabolism , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , Extracellular Matrix/chemistry , Extracellular Matrix/genetics , Extracellular Vesicles/chemistry , Extracellular Vesicles/genetics , Female , Gene Expression Profiling , Humans , Interleukin-6/metabolism , Male , Mesenchymal Stem Cells/cytology , Metabolic Networks and Pathways/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Transcription Factors/genetics , Transcription Factors/metabolism
6.
J Biol Chem ; 291(13): 6958-66, 2016 Mar 25.
Article in English | MEDLINE | ID: mdl-26884339

ABSTRACT

The aggregation of amyloid ß protein (Aß) is a fundamental pathogenic mechanism leading to the neuronal damage present in Alzheimer disease, and soluble Aß oligomers are thought to be a major toxic culprit. Thus, better knowledge and specific targeting of the pathways that lead to these noxious species may result in valuable therapeutic strategies. We characterized some effects of the molecular chaperone clusterin, providing new and more detailed evidence of its potential neuroprotective effects. Using a classical thioflavin T assay, we observed a dose-dependent inhibition of the aggregation process. The global analysis of time courses under different conditions demonstrated that clusterin has no effect on the elongation rate but mainly interferes with the nucleation processes (both primary and secondary), reducing the number of nuclei available for further fibril growth. Then, using a recently developed immunoassay based on surface plasmon resonance, we obtained direct evidence of a high-affinity (KD= 1 nm) interaction of clusterin with biologically relevant Aß1-42oligomers, selectively captured on the sensor chip. Moreover, with the same technology, we observed that substoichiometric concentrations of clusterin prevent oligomer interaction with the antibody 4G8, suggesting that the chaperone shields hydrophobic residues exposed on the oligomeric assemblies. Finally, we found that preincubation with clusterin antagonizes the toxic effects of Aß1-42oligomers, as evaluated in a recently developedin vivomodel inCaenorhabditis elegans.These data substantiate the interaction of clusterin with biologically active regions exposed on nuclei/oligomers of Aß1-42, providing a molecular basis for the neuroprotective effects of the chaperone.


Subject(s)
Amyloid beta-Peptides/antagonists & inhibitors , Caenorhabditis elegans/drug effects , Clusterin/pharmacology , Neuroprotective Agents/pharmacology , Peptide Fragments/antagonists & inhibitors , Pharynx/drug effects , Protein Aggregation, Pathological/prevention & control , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/chemical synthesis , Amyloid beta-Peptides/toxicity , Animals , Biological Assay , Caenorhabditis elegans/physiology , Clusterin/isolation & purification , Humans , Kinetics , Larva/drug effects , Larva/physiology , Neuroprotective Agents/isolation & purification , Peptide Fragments/chemical synthesis , Peptide Fragments/toxicity , Pharynx/physiology , Protein Aggregates/drug effects , Protein Aggregation, Pathological/pathology , Protein Binding
7.
Neurobiol Dis ; 88: 75-84, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26792398

ABSTRACT

One attractive pharmacological strategy for Alzheimer's disease (AD) is to design small peptides to interact with amyloid-ß (Aß) protein reducing its aggregation and toxicity. Starting from clinical observations indicating that patients coding a mutated Aß variant (AßA2V) in the heterozygous state do not develop AD, we developed AßA2V synthetic peptides, as well as a small peptide homologous to residues 1-6. These hindered the amyloidogenesis of Aß and its neurotoxicity in vitro, suggesting a basis for the design of a new small peptide in D-isomeric form, linked to the arginine-rich TAT sequence [Aß1-6A2V-TAT(D)], to allow translocation across biological membranes and the blood-brain barrier. Aß1-6A2V-TAT(D) was resistant to protease degradation, stable in serum and specifically able to interfere with Aß aggregation in vitro, reducing the appearance of toxic soluble species and protecting transgenic C. elegans from toxicity related to the muscular expression of human Aß. These observations offer a proof of concept for future pharmacological studies in mouse models of AD, providing a foundation for the design of AßA2V-based peptidomimetic molecules for therapeutic purposes.


Subject(s)
Amyloid beta-Peptides/metabolism , Mutation/genetics , Neurotoxicity Syndromes , Peptide Fragments/metabolism , Amino Acid Sequence , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/toxicity , Animals , Animals, Genetically Modified , Caenorhabditis elegans , Disease Models, Animal , Dose-Response Relationship, Drug , Gene Products, tat/genetics , Gene Products, tat/metabolism , Humans , In Vitro Techniques , Movement Disorders/etiology , Neuromuscular Junction/physiopathology , Neurotoxicity Syndromes/complications , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/genetics , Paralysis/etiology , Peptide Fragments/genetics , Peptide Fragments/toxicity , Protein Binding , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Surface Plasmon Resonance
8.
Chemistry ; 22(11): 3686-91, 2016 Mar 07.
Article in English | MEDLINE | ID: mdl-26696414

ABSTRACT

Antagonists of mannose binding lectin (MBL) have shown a protective role against brain reperfusion damage after acute ischemic stroke. Here we describe the design and streamlined synthesis of glycomimetic MBL antagonists based on a new tetravalent dendron scaffold. The dendron was developed by optimisation of a known polyester structure previously demonstrated to be very efficient for ligand presentation to MBL. Replacement of a labile succinyl ester bond with a more robust amide functionality, use of a longer and more hydrophilic linker, fast modular synthesis and orthogonal functionalisation at the focal point are the main features of the new scaffold. The glycoconjugate constructs become stable to silica gel chromatography and to water solutions at physiological pH, while preserving water solubility and activity in an SPR assay against the murine MBL-C isoform. Higher-order constructs were easily assembled, as demonstrated by the synthesis of a 16-valent dendrimer, which leads to two orders of magnitude increase in activity over the tetravalent version against MBL-C.


Subject(s)
Brain Ischemia/physiopathology , Dendrimers/chemistry , Glycoconjugates/chemistry , Mannose-Binding Lectin/deficiency , Stroke/pathology , Animals , Brain Ischemia/genetics , Brain Ischemia/metabolism , Glycoconjugates/metabolism , Ligands , Mannose-Binding Lectin/physiology , Mice
9.
J Biol Chem ; 288(11): 7857-7866, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23362282

ABSTRACT

A hallmark of Alzheimer disease (AD) is the accumulation of the amyloid-ß (Aß) peptide in the brain. Considerable evidence suggests that soluble Aß oligomers are responsible for the synaptic dysfunction and cognitive deficit observed in AD. However, the mechanism by which these oligomers exert their neurotoxic effect remains unknown. Recently, it was reported that Aß oligomers bind to the cellular prion protein with high affinity. Here, we show that N1, the main physiological cleavage fragment of the cellular prion protein, is necessary and sufficient for binding early oligomeric intermediates during Aß polymerization into amyloid fibrils. The ability of N1 to bind Aß oligomers is influenced by positively charged residues in two sites (positions 23-31 and 95-105) and is dependent on the length of the sequence between them. Importantly, we also show that N1 strongly suppresses Aß oligomer toxicity in cultured murine hippocampal neurons, in a Caenorhabditis elegans-based assay, and in vivo in a mouse model of Aß-induced memory dysfunction. These data suggest that N1, or small peptides derived from it, could be potent inhibitors of Aß oligomer toxicity and represent an entirely new class of therapeutic agents for AD.


Subject(s)
Amyloid beta-Peptides/chemistry , Prions/chemistry , Alzheimer Disease/metabolism , Amyloidogenic Proteins/chemistry , Animals , Benzothiazoles , Caenorhabditis elegans/metabolism , Hippocampus/metabolism , Mice , Mice, Inbred C57BL , Models, Biological , Neurodegenerative Diseases/metabolism , Neurons/metabolism , Peptides/chemistry , Protein Binding , Protein Structure, Tertiary , Surface Plasmon Resonance , Synapses/metabolism , Thiazoles/chemistry
10.
Neurobiol Dis ; 62: 521-32, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24184799

ABSTRACT

Although Alzheimer's disease (AD) is usually sporadic, in a small proportion of cases it is familial and can be linked to mutations in ß-amyloid precursor protein (APP). Unlike the other genetic defects, the mutation [alanine-673→valine-673] (A673V) causes the disease only in the homozygous condition with enhanced amyloid ß (Aß) production and aggregation; heterozygous carriers remain unaffected. It is not clear how misfolding and aggregation of Aß is affected in vivo by this mutation and whether this correlates with its toxic effects. No animal models over-expressing the A673V-APP gene or alanine-2-valine (A2V) mutated human Aß protein are currently available. Using the invertebrate Caenorhabditis elegans, we generated the first transgenic animal model to express the human Aß1-40 wild-type (WT) in neurons or possess the A2V mutation (Aß1-40A2V). Insertion of an Aß-mutated gene into this nematode reproduced the homozygous state of the human pathology. Functional and biochemical characteristics found in the A2V strain were compared to those of transgenic C. elegans expressing Aß1-40WT. The expression of both WT and A2V Aß1-40 specifically reduced the nematode's lifespan, causing behavioral defects and neurotransmission impairment which were worse in A2V worms. Mutant animals were more resistant than WT to paralysis induced by the cholinergic agonist levamisole, indicating that the locomotor defect was specifically linked to postsynaptic dysfunctions. The toxicity caused by the mutated protein was associated with a high propensity to form oligomeric assemblies which accumulate in the neurons, suggesting this to be the central event involved in the postsynaptic damage and early onset of the disease in homozygous human A673V carriers.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/toxicity , Neurons/metabolism , Peptide Fragments/metabolism , Peptide Fragments/toxicity , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/genetics , Animals , Animals, Genetically Modified , Caenorhabditis elegans , Humans , Locomotion/drug effects , Mutation , Neurons/drug effects , Peptide Fragments/chemistry , Peptide Fragments/genetics
11.
Biochem J ; 454(3): 417-25, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23808898

ABSTRACT

Genetic Creutzfeldt-Jakob disease, Gerstmann-Sträussler-Scheinker syndrome, fatal familial insomnia and prion protein cerebral amyloid angiopathy are clinically and neuropathologically distinct neurodegenerative diseases linked to mutations in the PRNP gene encoding the cellular prion protein (PrPC). How sequence variants of PRNP encode the information to specify these disease phenotypes is not known. It is suggested that each mutation produces a misfolded variant of PrPC with specific neurotoxic properties. However, structural studies of recombinant PrP did not detect major differences between wild-type and mutant molecules, pointing to the importance of investigating mutant PrPs from mammalian brains. We used surface plasmon resonance and a slot-blot immunoassay to analyse the antibody-binding profiles of soluble and insoluble PrP molecules extracted from the brains of transgenic mice modelling different prion diseases. By measuring the reactivity of monoclonal antibodies against different PrP epitopes, we obtained evidence of conformational differences between wild-type and mutant PrPs, and among different mutants. We detected structural heterogeneity in both monomeric and aggregated PrP, supporting the hypothesis that the phenotype of genetic prion diseases is encoded by mutant PrP conformation and assembly state.


Subject(s)
Brain/metabolism , Creutzfeldt-Jakob Syndrome/metabolism , Gerstmann-Straussler-Scheinker Disease/metabolism , Prions/metabolism , Animals , Antibodies, Monoclonal/chemistry , Creutzfeldt-Jakob Syndrome/genetics , Detergents/chemistry , Epitope Mapping , Gerstmann-Straussler-Scheinker Disease/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Mutation, Missense , Polymorphism, Genetic , Prions/chemistry , Prions/genetics , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Sodium Dodecyl Sulfate/chemistry , Surface Plasmon Resonance
12.
Sensors (Basel) ; 14(6): 10864-75, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24949642

ABSTRACT

A new immunoassay based on surface plasmon resonance (SPR) for the rapid, reproducible and sensitive determination of pentraxin-3 (PTX3) levels in human plasma has been developed and characterized. The method involves a 3-min flow of plasma over a sensor chip pre-coated with a monoclonal anti-PTX3 antibody (MNB4), followed by a 3-min flow of a polyclonal anti-PTX3 antibody (pAb), required for specific recognition of captured PTX3. The SPR signal generated with this secondary antibody linearly correlates with the plasma PTX3 concentration, in the range of 5-1500 ng/mL, with a lowest limit of detection of 5 ng/mL. The PTX3 concentrations determined with the SPR-based immunoassay in the plasma of 21 patients with sepsis, ranging 15-1600 ng/mL, were superimposable to those found in a classic ELISA immunoassay. Since the PTX3 concentration in the plasma of healthy subjects is <2 ng/mL, but markedly rises in certain medical conditions, the method is useful to quantify pathological levels of this important biomarker, with important diagnostic applications. In comparison with the classic ELISA, the SPR-based approach is much faster (30 min versus 4-5 h) and could be exploited for the development of new cost-effective SPR devices for point-of-care diagnosis.


Subject(s)
Biosensing Techniques/instrumentation , Blood Chemical Analysis/instrumentation , C-Reactive Protein/analysis , Immunoassay/instrumentation , Serum Amyloid P-Component/analysis , Surface Plasmon Resonance/instrumentation , Computer Systems , Equipment Design , Equipment Failure Analysis , Humans , Reproducibility of Results , Sensitivity and Specificity
13.
EBioMedicine ; 105: 105213, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38908098

ABSTRACT

BACKGROUND: COVID-19 clinical course is highly variable and secondary infections contribute to COVID-19 complexity. Early detection of secondary infections is clinically relevant for patient outcome. Procalcitonin (PCT) and C-reactive protein (CRP) are the most used biomarkers of infections. Pentraxin 3 (PTX3) is an acute phase protein with promising performance as early biomarker in infections. In patients with COVID-19, PTX3 plasma concentrations at hospital admission are independent predictor of poor outcome. In this study, we assessed whether PTX3 contributes to early identification of co-infections during the course of COVID-19. METHODS: We analyzed PTX3 levels in patients affected by COVID-19 with (n = 101) or without (n = 179) community or hospital-acquired fungal or bacterial secondary infections (CAIs or HAIs). FINDINGS: PTX3 plasma concentrations at diagnosis of CAI or HAI were significantly higher than those in patients without secondary infections. Compared to PCT and CRP, the increase of PTX3 plasma levels was associated with the highest hazard ratio for CAIs and HAIs (aHR 11.68 and 24.90). In multivariable Cox regression analysis, PTX3 was also the most significant predictor of 28-days mortality or intensive care unit admission of patients with potential co-infections, faring more pronounced than CRP and PCT. INTERPRETATION: PTX3 is a promising predictive biomarker for early identification and risk stratification of patients with COVID-19 and co-infections. FUNDING: Dolce & Gabbana fashion house donation; Ministero della Salute for COVID-19; EU funding within the MUR PNRR Extended Partnership initiative on Emerging Infectious Diseases (Project no. PE00000007, INF-ACT) and MUR PNRR Italian network of excellence for advanced diagnosis (Project no. PNC-E3-2022-23683266 PNC-HLS-DA); EU MSCA (project CORVOS 860044).


Subject(s)
Biomarkers , C-Reactive Protein , COVID-19 , Coinfection , SARS-CoV-2 , Serum Amyloid P-Component , Humans , COVID-19/blood , COVID-19/diagnosis , C-Reactive Protein/metabolism , C-Reactive Protein/analysis , Serum Amyloid P-Component/metabolism , Biomarkers/blood , Male , Female , Aged , Middle Aged , SARS-CoV-2/isolation & purification , Bacterial Infections/blood , Bacterial Infections/diagnosis , Procalcitonin/blood , Prognosis , Mycoses/blood , Mycoses/diagnosis , Aged, 80 and over
14.
J Biol Chem ; 287(33): 27796-805, 2012 Aug 10.
Article in English | MEDLINE | ID: mdl-22736768

ABSTRACT

Soluble oligomers of the amyloid-ß (Aß) peptide play a key role in the pathogenesis of Alzheimer's disease, but their elusive nature makes their detection challenging. Here we describe a novel immunoassay based on surface plasmon resonance (SPR) that specifically recognizes biologically active Aß oligomers. As a capturing agent, we immobilized on the sensor chip the monoclonal antibody 4G8, which targets a central hydrophobic region of Aß. This SPR assay allows specific recognition of oligomeric intermediates that rapidly appear and disappear during the incubation of synthetic Aß(1-42), discriminating them from monomers and higher order aggregates. The species recognized by SPR generate ionic currents in artificial lipid bilayers and inhibit the physiological pharyngeal contractions in Caenorhabditis elegans, a new method for testing the toxic potential of Aß oligomers. With these assays we found that the formation of biologically relevant Aß oligomers is inhibited by epigallocatechin gallate and increased by the A2V mutation, previously reported to induce early onset dementia. The SPR-based immunoassay provides new opportunities for detection of toxic Aß oligomers in biological samples and could be adapted to study misfolding proteins in other neurodegenerative disorders.


Subject(s)
Amyloid beta-Peptides/chemistry , Amyloid/chemistry , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans/chemistry , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid/genetics , Amyloid/metabolism , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Animals , Antibodies, Monoclonal, Murine-Derived/chemistry , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Surface Plasmon Resonance
15.
Circulation ; 126(12): 1484-94, 2012 Sep 18.
Article in English | MEDLINE | ID: mdl-22879370

ABSTRACT

BACKGROUND: The involvement of the complement system in brain injury has been scarcely investigated. Here, we document the pivotal role of mannose-binding lectin (MBL), one of the recognition molecules of the lectin complement pathway, in brain ischemic injury. METHODS AND RESULTS: Focal cerebral ischemia was induced in mice (by permanent or transient middle cerebral artery occlusion) and rats (by 3-vessel occlusion). We first observed that MBL is deposited on ischemic vessels up to 48 hours after injury and that functional MBL/MBL-associated serine protease 2 complexes are increased. Next, we demonstrated that (1) MBL(-/-) mice are protected from both transient and permanent ischemic injury; (2) Polyman2, the newly synthesized mannosylated molecule selected for its binding to MBL, improves neurological deficits and infarct volume when given up to 24 hours after ischemia in mice; (3) anti-MBL-A antibody improves neurological deficits and infarct volume when given up to 18 hours after ischemia, as assessed after 28 days in rats. CONCLUSIONS: Our data show an important role for MBL in the pathogenesis of brain ischemic injury and provide a strong support to the concept that MBL inhibition may be a relevant therapeutic target in humans, one with a wide therapeutic window of application.


Subject(s)
Brain Ischemia/physiopathology , Infarction, Middle Cerebral Artery/physiopathology , Mannose-Binding Lectin/genetics , Mannose-Binding Lectin/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Brain Edema/drug therapy , Brain Edema/genetics , Brain Edema/physiopathology , Brain Ischemia/drug therapy , Brain Ischemia/genetics , Disease Models, Animal , Humans , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/genetics , Male , Mannans/metabolism , Mannans/pharmacology , Mannose-Binding Lectin/immunology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Rats , Rats, Inbred Strains
16.
J Immunol ; 187(1): 172-80, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21642543

ABSTRACT

Shiga toxin (Stx)-producing E.coli O157:H7 has become a global threat to public health; it is a primary cause of diarrhea-associated hemolytic uremic syndrome (HUS), a disorder of thrombocytopenia, microangiopathic hemolytic anemia, and acute renal failure with thrombi occluding renal microcirculation. In this study, we explored whether Stx triggers complement-dependent microvascular thrombosis in in vitro and in vivo experimental settings of HUS. Stx induced on human microvascular endothelial cell surface the expression of P-selectin, which bound and activated C3 via the alternative pathway, leading to thrombus formation under flow. In the search for mechanisms linking complement activation and thrombosis, we found that exuberant complement activation in response to Stx generated an increased amount of C3a that caused further endothelial P-selectin expression, thrombomodulin (TM) loss, and thrombus formation. In a murine model of HUS obtained by coinjection of Stx2 and LPS and characterized by thrombocytopenia and renal dysfunction, upregulation of glomerular endothelial P-selectin was associated with C3 and fibrin(ogen) deposits, platelet clumps, and reduced TM expression. Treatment with anti-P-selectin Ab limited glomerular C3 accumulation. Factor B-deficient mice after Stx2/LPS exhibited less thrombocytopenia and were protected against glomerular abnormalities and renal function impairment, indicating the involvement of complement activation via the alternative pathway in the glomerular thrombotic process in HUS mice. The functional role of C3a was documented by data showing that glomerular fibrin(ogen), platelet clumps, and TM loss were markedly decreased in HUS mice receiving C3aR antagonist. These results identify Stx-induced complement activation, via P-selectin, as a key mechanism of C3a-dependent microvascular thrombosis in diarrhea-associated HUS.


Subject(s)
Complement C3a/toxicity , Complement Pathway, Alternative/immunology , Endothelium, Vascular/immunology , Endothelium, Vascular/pathology , Hemolytic-Uremic Syndrome/immunology , Hemolytic-Uremic Syndrome/pathology , Shiga Toxin 1/toxicity , Shiga Toxin 2/toxicity , Animals , Cell Line , Complement C3a/biosynthesis , Complement C3a/metabolism , Complement Factor B/deficiency , Complement Factor B/genetics , Disease Models, Animal , Endothelium, Vascular/metabolism , Escherichia coli Infections/immunology , Escherichia coli Infections/metabolism , Escherichia coli Infections/pathology , Escherichia coli O157/immunology , Escherichia coli O157/pathogenicity , Hemolytic-Uremic Syndrome/metabolism , Humans , Kidney Glomerulus/blood supply , Kidney Glomerulus/immunology , Kidney Glomerulus/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microcirculation/immunology , P-Selectin/physiology , Protein Binding/immunology
17.
Proc Natl Acad Sci U S A ; 107(5): 2295-300, 2010 Feb 02.
Article in English | MEDLINE | ID: mdl-20133875

ABSTRACT

Inability to form new memories is an early clinical sign of Alzheimer's disease (AD). There is ample evidence that the amyloid-beta (Abeta) peptide plays a key role in the pathogenesis of this disorder. Soluble, bio-derived oligomers of Abeta are proposed as the key mediators of synaptic and cognitive dysfunction, but more tractable models of Abeta-mediated cognitive impairment are needed. Here we report that, in mice, acute intracerebroventricular injections of synthetic Abeta(1-42) oligomers impaired consolidation of the long-term recognition memory, whereas mature Abeta(1-42) fibrils and freshly dissolved peptide did not. The deficit induced by oligomers was reversible and was prevented by an anti-Abeta antibody. It has been suggested that the cellular prion protein (PrP(C)) mediates the impairment of synaptic plasticity induced by Abeta. We confirmed that Abeta(1-42) oligomers interact with PrP(C), with nanomolar affinity. However, PrP-expressing and PrP knock-out mice were equally susceptible to this impairment. These data suggest that Abeta(1-42) oligomers are responsible for cognitive impairment in AD and that PrP(C) is not required.


Subject(s)
Amyloid beta-Peptides/pharmacology , Memory/drug effects , Peptide Fragments/pharmacology , PrPC Proteins/metabolism , Alzheimer Disease/etiology , Amyloid beta-Peptides/chemical synthesis , Amyloid beta-Peptides/chemistry , Animals , Cognition Disorders/etiology , Cognition Disorders/metabolism , Humans , Injections, Intraventricular , Male , Memory/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Peptide Fragments/chemical synthesis , Peptide Fragments/chemistry , Prion Proteins , Prions/genetics , Prions/metabolism , Protein Binding , Surface Plasmon Resonance
18.
Sensors (Basel) ; 12(12): 16420-32, 2012 Nov 27.
Article in English | MEDLINE | ID: mdl-23443386

ABSTRACT

Great interest is currently being devoted to the development of nanoparticles (NPs) for biomedical purposes, designed to improve the pharmacokinetic profile of their cargos (either imaging probes or drugs) and to enhance the specific targeting at the disease site. Recent works suggest that Surface Plasmon Resonance (SPR), widely used for the analysis of biomolecular interactions, represents a technique of choice for rapid and quantitative analyses of the interaction between NPs--functionalized with specific ligands--and their putative biological targets. Moreover, SPR can provide important details on the formation and the role of the protein "corona", i.e., the protein layer which coats NPs once they come into contact with biological fluids. These novel applications of SPR sensors may be very useful to characterize, screen and develop nanodevices for biomedical purposes.


Subject(s)
Biosensing Techniques , Nanoparticles/chemistry , Surface Plasmon Resonance , Humans , Ligands
19.
J Leukoc Biol ; 111(4): 817-836, 2022 04.
Article in English | MEDLINE | ID: mdl-34346525

ABSTRACT

The MS4A gene family encodes 18 tetraspanin-like proteins, most of which with unknown function. MS4A1 (CD20), MS4A2 (FcεRIß), MS4A3 (HTm4), and MS4A4A play important roles in immunity, whereas expression and function of other members of the family are unknown. The present investigation was designed to obtain an expression fingerprint of MS4A family members, using bioinformatics analysis of public databases, RT-PCR, and protein analysis when possible. MS4A3, MS4A4A, MS4A4E, MS4A6A, MS4A7, and MS4A14 were expressed by myeloid cells. MS4A6A and MS4A14 were expressed in circulating monocytes and decreased during monocyte-to-Mϕ differentiation in parallel with an increase in MS4A4A expression. Analysis of gene expression regulation revealed a strong induction of MS4A4A, MS4A6A, MS4A7, and MS4A4E by glucocorticoid hormones. Consistently with in vitro findings, MS4A4A and MS4A7 were expressed in tissue Mϕs from COVID-19 and rheumatoid arthritis patients. Interestingly, MS4A3, selectively expressed in myeloid precursors, was found to be a marker of immature circulating neutrophils, a cellular population associated to COVID-19 severe disease. The results reported here show that members of the MS4A family are differentially expressed and regulated during myelomonocytic differentiation, and call for assessment of their functional role and value as therapeutic targets.


Subject(s)
COVID-19 , Membrane Proteins , Antigens, CD20 , Family , Humans , Membrane Proteins/genetics , Monocytes/metabolism
20.
Front Immunol ; 13: 979232, 2022.
Article in English | MEDLINE | ID: mdl-36189302

ABSTRACT

Background: Early prognostic stratification of patients with sepsis is a difficult clinical challenge. Aim of this study was to evaluate novel molecules in association with clinical parameters as predictors of 90-days mortality in patients admitted with sepsis at Humanitas Research Hospital. Methods: Plasma samples were collected from 178 patients, diagnosed based on Sepsis-3 criteria, at admission to the Emergency Department and after 5 days of hospitalization. Levels of pentraxin 3 (PTX3), soluble IL-1 type 2 receptor (sIL-1R2), and of a panel of pro- and anti-inflammatory cytokines were measured by ELISA. Cox proportional-hazard models were used to evaluate predictors of 90-days mortality. Results: Circulating levels of PTX3, sIL-1R2, IL-1ß, IL-6, IL-8, IL-10, IL-18, IL-1ra, TNF-α increased significantly in sepsis patients on admission, with the highest levels measured in shock patients, and correlated with SOFA score (PTX3: r=0.44, p<0.0001; sIL-1R2: r=0.35, p<0.0001), as well as with 90-days mortality. After 5 days of hospitalization, PTX3 and cytokines, but not sIL-1R2 levels, decreased significantly, in parallel with a general improvement of clinical parameters. The combination of age, blood urea nitrogen, PTX3, IL-6 and IL-18, defined a prognostic index predicting 90-days mortality in Sepsis-3 patients and showing better apparent discrimination capacity than the SOFA score (AUC=0.863, 95% CI: 0.780-0.945 vs. AUC=0.727, 95% CI: 0.613-0.840; p=0.021 respectively). Conclusion: These data suggest that a prognostic index based on selected cytokines, PTX3 and clinical parameters, and hence easily adoptable in clinical practice, performs in predicting 90-days mortality better than SOFA. An independent validation is required.


Subject(s)
Interleukin-10 , Sepsis , Biomarkers , C-Reactive Protein , Cytokines , Humans , Infant, Newborn , Interleukin 1 Receptor Antagonist Protein , Interleukin-1 , Interleukin-18 , Interleukin-6 , Interleukin-8 , Prognosis , ROC Curve , Serum Amyloid P-Component , Tumor Necrosis Factor-alpha
SELECTION OF CITATIONS
SEARCH DETAIL