Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
2.
Front Immunol ; 12: 641311, 2021.
Article in English | MEDLINE | ID: mdl-34305885

ABSTRACT

PTX3 is a unique member of the long pentraxins family and plays an indispensable role in regulating the immune system. We previously showed that PTX3 deletion aggravates allergic inflammation via a Th17 -dominant phenotype and enhanced CD4 T cell survival using a murine model of ovalbumin (OVA) induced allergic inflammation. In this study, we identified that upon OVA exposure, increased infiltration of CD11c+CD11b+ dendritic cells (DCs) was observed in the lungs of PTX3-/- mice compared to wild type littermate. Further analysis showed that a short-term OVA exposure led to an increased number of bone marrow common myeloid progenitors (CMP) population concomitantly with increased Ly6Chigh CCR2high monocytes and CD11c+CD11b+ DCs in the lungs. Also, pulmonary CD11c+CD11b+ DCs from OVA-exposed PTX3-/- mice exhibited enhanced expression of maturation markers, chemokines receptors CCR2, and increased OVA uptake and processing compared to wild type controls. Taken together, our data suggest that PTX3 deficiency heightened lung CD11c+CD11b+DC numbers and function, hence exacerbating airway inflammatory response.


Subject(s)
C-Reactive Protein/deficiency , C-Reactive Protein/immunology , Dendritic Cells/immunology , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/immunology , Respiratory Hypersensitivity/immunology , Allergens/immunology , Allergens/toxicity , Animals , CD11b Antigen/immunology , CD11c Antigen/immunology , Disease Models, Animal , Female , Mice , Mice, Knockout , Ovalbumin/immunology , Ovalbumin/toxicity
3.
Front Immunol ; 12: 666198, 2021.
Article in English | MEDLINE | ID: mdl-34093560

ABSTRACT

Klebsiella pneumoniae is a common pathogen in human sepsis. The emergence of multidrug-resistant K. pneumoniae strains represents a major clinical challenge in nosocomial and community acquired infections. The long pentraxin PTX3, a key component of humoral innate immunity, is involved in resistance to selected pathogens by promoting opsonophagocytosis. We investigated the relevance of PTX3 in innate immunity against K. pneumoniae infections using Ptx3-/- mice and mouse models of severe K. pneumoniae infections. Local and systemic PTX3 expression was induced following K. pneumoniae pulmonary infection, in association with the up-regulation of TNF-α and IL-1ß. PTX3 deficiency in mice was associated with higher bacterial burden and mortality, release of pro-inflammatory cytokines as well as IL-10 in the lung and systemically. The analysis of the mechanisms responsible of PTX3-dependent control of K. pneumoniae infection revealed that PTX3 did not interact with K. pneumoniae, or promote opsonophagocytosis. The comparison of susceptibility of wild-type, Ptx3-/-, C3-/- and Ptx3-/- /C3-/- mice to the infection showed that PTX3 acted in a complement-independent manner. Lung histopathological analysis showed more severe lesions in Ptx3-/- mice with fibrinosuppurative, necrotizing and haemorrhagic bronchopneumonia, associated with increased fibrin deposition in the lung and circulating fibrinogen consumption. These findings indicate that PTX3 contributes to the control of K. pneumoniae infection by modulating inflammatory responses and tissue damage. Thus, this study emphasizes the relevance of the role of PTX3 as regulator of inflammation and orchestrator of tissue repair in innate responses to infections.


Subject(s)
C-Reactive Protein/immunology , Klebsiella Infections/immunology , Klebsiella pneumoniae/pathogenicity , Serum Amyloid P-Component/immunology , Animals , Bacterial Load/immunology , C-Reactive Protein/deficiency , C-Reactive Protein/metabolism , Cytokines/metabolism , Fibrin/metabolism , Fibrinogen/metabolism , Immunity, Innate , Inflammation , Klebsiella Infections/metabolism , Klebsiella Infections/microbiology , Klebsiella Infections/pathology , Klebsiella pneumoniae/immunology , Lung/immunology , Lung/metabolism , Lung/microbiology , Lung/pathology , Mice , Sepsis/immunology , Sepsis/metabolism , Sepsis/microbiology , Sepsis/pathology , Serum Amyloid P-Component/deficiency , Serum Amyloid P-Component/metabolism , Stromal Cells/metabolism
4.
J Neuroinflammation ; 17(1): 350, 2020 Nov 22.
Article in English | MEDLINE | ID: mdl-33222690

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is one of the most prevalent neurodegenerative disorders characterized by gradual memory loss and neuropsychiatric symptoms. We have previously demonstrated that the 2-({3-[2-(1-cyclohexene-1-yl)ethyl]-6,7-dimethoxy-4-oxo-3,4-dihydro-2-quinazolinyl}sulfanyl)-N-(4-ethylphenyl)butanamide (K284-6111), the inhibitor of CHI3L1, has the inhibitory effect on memory impairment in Αß infusion mouse model and on LPS-induced neuroinflammation in the murine BV-2 microglia and primary cultured astrocyte. METHODS: In the present study, we investigated the inhibitory effect of K284-6111 on memory dysfunction and neuroinflammation in Tg2576 transgenic mice, and a more detailed correlation of CHI3L1 and AD. To investigate the effects of K284-6111 on memory dysfunction, we administered K284-6111 (3 mg/kg, p.o.) daily for 4 weeks to Tg2576 mice, followed by behavioral tests of water maze test, probe test, and passive avoidance test. RESULTS: Administration of K284-6111 alleviated memory impairment in Tg2576 mice and had the effect of reducing the accumulation of Aß and neuroinflammatory responses in the mouse brain. K284-6111 treatment also selectively inactivated ERK and NF-κB pathways, which were activated when CHI3L1 was overexpressed, in the mouse brain and in BV-2 cells. Web-based gene network analysis and our results of gene expression level in BV-2 cells showed that CHI3L1 is closely correlated with PTX3. Our result revealed that knockdown of PTX3 has an inhibitory effect on the production of inflammatory proteins and cytokines, and on the phosphorylation of ERK and IκBα. CONCLUSION: These results suggest that K284-6111 could improve memory dysfunction by alleviating neuroinflammation through inhibiting CHI3L1 enhancing ERK-dependent PTX3 pathway.


Subject(s)
C-Reactive Protein/deficiency , Chitinase-3-Like Protein 1/antagonists & inhibitors , Inflammation Mediators/antagonists & inhibitors , MAP Kinase Signaling System/drug effects , Memory Disorders/drug therapy , Nerve Tissue Proteins/deficiency , Quinazolines/therapeutic use , Animals , Brain/drug effects , Brain/metabolism , C-Reactive Protein/genetics , Cell Line , Chitinase-3-Like Protein 1/metabolism , Gene Knockdown Techniques/methods , Inflammation Mediators/metabolism , MAP Kinase Signaling System/physiology , Maze Learning/drug effects , Maze Learning/physiology , Memory Disorders/metabolism , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Quinazolines/pharmacology
5.
Int J Obes (Lond) ; 44(2): 525-538, 2020 02.
Article in English | MEDLINE | ID: mdl-31209269

ABSTRACT

BACKGROUND/OBJECTIVES: Pentraxin 3 (PTX3) has been characterized as a soluble and multifunctional pattern recognition protein in the regulation of innate immune response. However, little is known about its role in adipose tissue inflammation and obesity. Herein, we investigated the role of PTX3 in the regulation of lipopolysaccharide (LPS)-induced inflammation in adipocytes and adipose tissue, as well as high-fat diet (HFD)-induced metabolic inflammation in obesity. METHODS: Ptx3 knockdown 3T3-L1 Cells were generated using shRNA for Ptx3 gene and treated with different inflammatory stimuli. For the in vivo studies, Ptx3 knockout mice were treated with 0.3 mg/kg of LPS for 6 h. Adipose tissues were collected for gene and protein expression by qPCR and western blotting, respectively. Ptx3 knockout mice were fed with HFD for 12 week since 6 week of age. RESULTS: We observed that the expression of PTX3 in adipose tissue and serum PTX3 were markedly increased in response to LPS administration. Knocking down Ptx3 in 3T3-L1 cells reduced adipogenesis and caused a more profound and sustained upregulation of proinflammatory gene expression and signaling pathway activation during LPS-stimulated inflammation in 3T3-L1 adipocytes. In vivo studies showed that PTX3 deficiency significantly exacerbated the LPS-induced upregulation of inflammatory genes and downregulation of adipogeneic genes in visceral and subcutaneous adipose tissue of mice. Accordingly, LPS stimulation elicited increased activation of nuclear factor-κB (NF-κB) and p44/42 MAPK (Erk1/2) signaling pathways in visceral and subcutaneous adipose tissue. The expression of PTX3 in adipose tissue was also induced by HFD, and PTX3 deficiency led to the upregulation of proinflammatory genes in visceral adipose tissue of HFD-induced obese mice. CONCLUSIONS: Our results suggest a protective role of PTX3 in LPS- and HFD-induced sustained inflammation in adipose tissue.


Subject(s)
Adipose Tissue/metabolism , C-Reactive Protein , Inflammation/metabolism , Nerve Tissue Proteins , 3T3-L1 Cells , Animals , C-Reactive Protein/deficiency , C-Reactive Protein/genetics , C-Reactive Protein/metabolism , Diet, High-Fat , Female , Inflammation/chemically induced , Lipopolysaccharides/adverse effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism
6.
Cardiovasc Res ; 115(13): 1861-1872, 2019 Nov 01.
Article in English | MEDLINE | ID: mdl-30859179

ABSTRACT

AIMS: Low-grade chronic inflammation characterizes obesity and metabolic syndrome. Here, we aim at investigating the impact of the acute-phase protein long pentraxin 3 (PTX3) on the immune-inflammatory response occurring during diet-induced obesity. METHODS AND RESULTS: PTX3 deficiency in mice fed a high-fat diet for 20 weeks protects from weight gain and adipose tissue deposition in visceral and subcutaneous depots. This effect is not related to changes in glucose homeostasis and lipid metabolism but is associated with an improved immune cell phenotype in the adipose tissue of Ptx3 deficient animals, which is characterized by M2-macrophages polarization and increased angiogenesis. These findings are recapitulated in humans where carriers of a PTX3 haplotype (PTX3 h2/h2 haplotype), resulting in lower PTX3 plasma levels, presented with a reduced prevalence of obesity and decreased abdominal adiposity compared with non-carriers. CONCLUSION: Our results support a critical role for PTX3 in the onset of obesity by promoting inflammation and limiting adipose tissue vascularization and delineate PTX3 targeting as a valuable strategy for the treatment of adipose tissue-associated inflammatory response.


Subject(s)
C-Reactive Protein/deficiency , Diet, High-Fat , Energy Metabolism , Immunity, Innate , Inflammation Mediators/metabolism , Inflammation/prevention & control , Intra-Abdominal Fat/blood supply , Intra-Abdominal Fat/metabolism , Nerve Tissue Proteins/deficiency , Obesity/metabolism , Subcutaneous Fat/blood supply , Subcutaneous Fat/metabolism , Adipogenesis , Adiposity , Aged , Animals , C-Reactive Protein/genetics , Cell Plasticity , Cells, Cultured , Disease Models, Animal , Female , Haplotypes , Humans , Inflammation/immunology , Inflammation/metabolism , Inflammation/physiopathology , Inflammation Mediators/immunology , Intra-Abdominal Fat/immunology , Macrophages/immunology , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Neovascularization, Physiologic , Nerve Tissue Proteins/genetics , Obesity/immunology , Obesity/physiopathology , Obesity, Abdominal/epidemiology , Obesity, Abdominal/genetics , Obesity, Abdominal/physiopathology , Phenotype , Serum Amyloid P-Component/genetics , Signal Transduction , Subcutaneous Fat/immunology , Weight Gain
7.
J Pathol ; 240(4): 495-506, 2016 12.
Article in English | MEDLINE | ID: mdl-27659908

ABSTRACT

The discovery that genetic abnormalities in complement factor H (FH) are associated with an increased risk for age-related macular degeneration (AMD), the most common cause of blindness among the elderly, raised hope of new treatments for this vision-threatening disease. Nonetheless, over a decade after the identification of this important association, how innate immunity contributes to AMD remains unresolved. Pentraxin 3 (PTX3), an essential component of the innate immunity system that plays a non-redundant role in controlling inflammation, regulates complement by interacting with complement components. Here, we show that PTX3 is induced by oxidative stress, a known cause of AMD, in the retinal pigmented epithelium (RPE). PTX3 deficiency in vitro and in vivo magnified complement activation induced by oxidative stress, leading to increased C3a, FB, and C3d, but not C5b-9 complex formation. Increased C3a levels, resulting from PTX3 deficiency, raised the levels of Il1b mRNA and secretion of activated interleukin (IL)-1ß by interacting with C3aR. Importantly, PTX3 deficiency augmented NLRP3 inflammasome activation, resulting in enhanced IL-1ß, but not IL-18, production by the RPE. Thus, in the presence of PTX3 deficiency, the complement and inflammasome pathways worked in concert to produce IL-1ß in sufficient abundance to, importantly, result in macrophages accumulating in the choroid. These results demonstrate that PTX3 acts as an essential brake for complement and inflammasome activation by regulating the abundance of FH in the RPE, and provide critical insights into the complex interplay between oxidative stress and innate immunity in the early stages of AMD development. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
C-Reactive Protein/immunology , Complement Factor H/immunology , Inflammasomes/immunology , Macular Degeneration/immunology , Nerve Tissue Proteins/immunology , Oxidative Stress/immunology , Aldehydes/pharmacology , Animals , C-Reactive Protein/deficiency , Cells, Cultured , Choroid/immunology , Complement Activation/drug effects , Complement Activation/immunology , Complement C3a/immunology , Cysteine Proteinase Inhibitors/pharmacology , Humans , Immunity, Innate/immunology , Interleukin-1beta/biosynthesis , Macrophages/immunology , Mice, Inbred C57BL , Nerve Tissue Proteins/deficiency , Oxidative Stress/drug effects , Retinal Pigment Epithelium/immunology
8.
Eur J Immunol ; 46(3): 619-33, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26578407

ABSTRACT

Cardiomyocyte death as a result of viral infection is an excellent model for dissecting the inflammatory stress response that occurs in heart tissue. We reported earlier that a specific proteasome isoform, the immunoproteasome, prevents exacerbation of coxsackievirus B3 (CVB3)-induced myocardial destruction and preserves cell vitality in heart tissue inflammation. Following the aim to decipher molecular targets of immunoproteasome-dependent proteolysis, we investigated the function and regulation of the soluble PRR Pentraxin3 (PTX3). We show that the ablation of PTX3 in mice aggravated CVB3-triggered inflammatory injury of heart tissue, without having any significant effect on viral titers. Thus, there might be a role of PTX3 in preventing damage-associated molecular pattern-induced cell death. We found that the catalytic activity of the immunoproteasome subunit LMP7 regulates the timely availability of factors controlling PTX3 production. We report on immunoproteasome-dependent alteration of ERK1/2 and p38MAPKs, which were both found to be involved in PTX3 expression control. Our finding of a cardioprotective function of immunoproteasome-dependent PTX3 expression revealed a crucial mechanism of the stress-induced damage response in myocardial inflammation. In addition to antigen presentation and cytokine production, proteolysis by the immunoproteasome can also regulate the innate immune response during viral infection.


Subject(s)
C-Reactive Protein/immunology , C-Reactive Protein/metabolism , Nerve Tissue Proteins/immunology , Nerve Tissue Proteins/metabolism , Proteasome Endopeptidase Complex/immunology , Proteasome Endopeptidase Complex/physiology , Animals , C-Reactive Protein/deficiency , C-Reactive Protein/genetics , Cells, Cultured , Coxsackievirus Infections/immunology , Coxsackievirus Infections/virology , Disease Models, Animal , Heart/anatomy & histology , Heart/virology , Immunity, Innate , Inflammation/virology , MAP Kinase Signaling System/genetics , Mice , Myocarditis/immunology , Myocarditis/virology , Myocardium/immunology , Myocytes, Cardiac/pathology , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Proteasome Endopeptidase Complex/genetics , Proteolysis , Stress, Physiological , Viral Load
9.
PLoS One ; 10(3): e0120807, 2015.
Article in English | MEDLINE | ID: mdl-25786110

ABSTRACT

Long pentraxin 3 (PTX3) is a non-redundant component of the humoral arm of innate immunity. The present study was designed to investigate the interaction of PTX3 with Neisseria meningitidis. PTX3 bound acapsular meningococcus, Neisseria-derived outer membrane vesicles (OMV) and 3 selected meningococcal antigens (GNA0667, GNA1030 and GNA2091). PTX3-recognized microbial moieties are conserved structures which fulfil essential microbial functions. Ptx3-deficient mice had a lower antibody response in vaccination protocols with OMV and co-administration of PTX3 increased the antibody response, particularly in Ptx3-deficient mice. Administration of PTX3 reduced the bacterial load in infant rats challenged with Neisseria meningitidis. These results suggest that PTX3 recognizes a set of conserved structures from Neisseria meningitidis and acts as an amplifier/endogenous adjuvant of responses to this bacterium.


Subject(s)
Adjuvants, Immunologic/genetics , Antibodies, Bacterial/biosynthesis , Antigens, Bacterial/immunology , C-Reactive Protein/immunology , Meningitis, Meningococcal/prevention & control , Meningococcal Vaccines/immunology , Neisseria meningitidis/immunology , Serum Amyloid P-Component/immunology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/deficiency , Animals , Animals, Newborn , Antibodies, Bacterial/immunology , Antigens, Bacterial/genetics , Bacterial Load/drug effects , C-Reactive Protein/administration & dosage , C-Reactive Protein/deficiency , C-Reactive Protein/genetics , Female , Gene Expression , Immunity, Humoral/drug effects , Immunity, Innate/drug effects , Male , Meningitis, Meningococcal/immunology , Meningitis, Meningococcal/virology , Meningococcal Vaccines/administration & dosage , Meningococcal Vaccines/genetics , Mice , Mice, Knockout , Neisseria meningitidis/drug effects , Neisseria meningitidis/genetics , Ovalbumin/administration & dosage , Rats , Rats, Wistar , Serum Amyloid P-Component/administration & dosage , Serum Amyloid P-Component/deficiency , Serum Amyloid P-Component/genetics , Vaccination
10.
Neuron ; 85(6): 1257-72, 2015 03 18.
Article in English | MEDLINE | ID: mdl-25754824

ABSTRACT

Circuit computation requires precision in the timing, extent, and synchrony of principal cell (PC) firing that is largely enforced by parvalbumin-expressing, fast-spiking interneurons (PVFSIs). To reliably coordinate network activity, PVFSIs exhibit specialized synaptic and membrane properties that promote efficient afferent recruitment such as expression of high-conductance, rapidly gating, GluA4-containing AMPA receptors (AMPARs). We found that PVFSIs upregulate GluA4 during the second postnatal week coincident with increases in the AMPAR clustering proteins NPTX2 and NPTXR. Moreover, GluA4 is dramatically reduced in NPTX2(-/-)/NPTXR(-/-) mice with consequent reductions in PVFSI AMPAR function. Early postnatal NPTX2(-/-)/NPTXR(-/-) mice exhibit delayed circuit maturation with a prolonged critical period permissive for giant depolarizing potentials. Juvenile NPTX2(-/-)/NPTXR(-/-) mice display reduced feedforward inhibition yielding a circuit deficient in rhythmogenesis and prone to epileptiform discharges. Our findings demonstrate an essential role for NPTXs in controlling network dynamics highlighting potential therapeutic targets for disorders with inhibition/excitation imbalances such as schizophrenia.


Subject(s)
Action Potentials/physiology , C-Reactive Protein/metabolism , Interneurons/metabolism , Nerve Net/growth & development , Nerve Tissue Proteins/metabolism , Parvalbumins/metabolism , Synapses/metabolism , Animals , Animals, Newborn , C-Reactive Protein/deficiency , Disease Models, Animal , Mice , Mice, Knockout , Nerve Tissue Proteins/deficiency
11.
Neurobiol Dis ; 75: 15-30, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25554688

ABSTRACT

Neonatal hypoxic-ischemic (HI) brain injury is a leading cause of mortality and morbidity in infants and children for which there is no promising therapy at present. Previously, we reported induction of neuronal pentraxin 1 (NP1), a novel neuronal protein of the long-pentraxin family, following HI injury in neonatal brain. Here, we report that genetic deletion of NP1 expression prevents HI injury in neonatal brain. Elevated expression of NP1 was observed in neurons, not in astrocytes, of the ipsilateral cortical layers (I-IV) and in the hippocampal CA1 and CA3 areas of WT brains following hypoxia-ischemia; brain areas that developed infarcts (at 24-48 h), showed significantly increased numbers of TUNEL-(+) cells and tissue loss (at 7 days). In contrast, NP1-KO mice showed no evidence of brain infarction and tissue loss after HI. The immunofluorescence staining of brain sections with mitochondrial protein COX IV and subcellular fractionation analysis showed increased accumulation of NP1 in mitochondria, pro-death protein Bax activation and NP1 co-localization with activated caspase-3 in WT, but not in the NP1-KO brains; corroborating NP1 interactions with the mitochondria-derived pro-death pathways. Disruption of NP1 translocation to mitochondria by NP1-siRNA in primary cortical cultures significantly reduced ischemic neuronal death. NP1 was immunoprecipitated with activated Bax [6A7] proteins; HI caused increased interactions of NP1 with Bax, thereby, facilitating Bax translocation to mitochondrial and neuronal death. To further delineate the specificity of NPs, we found that NP1 but not the NP2 induction is specifically involved in brain injury mechanisms and that knockdown of NP1 only results in neuroprotection. Furthermore, live in vivo T2-weighted magnetic resonance imaging (MRI) including fractional anisotropy (FA) mapping showed no sign of delayed brain injury or tissue loss in the NP1-KO mice as compared to the WT at different post-HI periods (4-24 weeks) examined; indicating a long-term neuroprotective efficacy of NP1 gene deletion. Collectively, our results demonstrate a novel mechanism of neuronal death and predict that inhibition of NP1 expression is a promising strategy to prevent hypoxic-ischemic injury in immature brain.


Subject(s)
Brain/metabolism , C-Reactive Protein/deficiency , Hypoxia-Ischemia, Brain/metabolism , Nerve Tissue Proteins/deficiency , Animals , Animals, Newborn , Astrocytes/metabolism , Astrocytes/pathology , Brain/pathology , C-Reactive Protein/genetics , Caspase 3/metabolism , Cell Death/physiology , Cell Hypoxia/physiology , Cells, Cultured , Disease Models, Animal , Disease Progression , Glucose/deficiency , Hypoxia-Ischemia, Brain/pathology , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Nerve Tissue Proteins/genetics , Neurons/metabolism , Neurons/pathology , RNA, Small Interfering , bcl-2-Associated X Protein/metabolism
12.
FASEB J ; 29(4): 1220-34, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25466900

ABSTRACT

Neuronal-activity-regulated pentraxin (NARP/NPTX2/NP2) is a secreted synaptic protein that regulates the trafficking of glutamate receptors and mediates learning, memory, and drug addiction. The role of NPTX2 in regulating structural synaptic plasticity and behavior in a developing vertebrate is indefinite. We characterized the expression of nptx2a in larvae and adult zebrafish and established a transcription activator-like effector nuclease (TALEN)-mediated nptx2a mutant (nptx2a(-/-)) to study the role of Nptx2a in regulating structural synaptic plasticity and behavior. Similar to mammals, the zebrafish nptx2a was expressed in excitatory neurons in the brain and spinal cord. Its expression was induced in response to a mechanosensory stimulus but did not change during day and night. Behavioral assays showed that loss of Nptx2a results in reduced locomotor response to light-to-dark transition states and to a sound stimulus. Live imaging of synapses using the transgenic nptx2a:GAL4VP16 zebrafish and a fluorescent presynaptic synaptophysin (SYP) marker revealed reduced synaptic density in the axons of the spinal motor neurons and the anterodorsal lateral-line ganglion (gAD), which regulate locomotor activity and locomotor response to mechanosensory stimuli, respectively. These results suggest that Nptx2a affects locomotor response to external stimuli by mediating structural synaptic plasticity in excitatory neuronal circuits.


Subject(s)
C-Reactive Protein/physiology , Nerve Tissue Proteins/physiology , Neuronal Plasticity/physiology , Zebrafish Proteins/physiology , Zebrafish/anatomy & histology , Zebrafish/physiology , Acoustic Stimulation , Animals , Animals, Genetically Modified , C-Reactive Protein/deficiency , C-Reactive Protein/genetics , Evolution, Molecular , Gene Expression Regulation, Developmental , Gene Knockout Techniques , Humans , Larva/cytology , Larva/physiology , Mechanotransduction, Cellular , Motor Activity , Motor Neurons/physiology , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Neuronal Plasticity/genetics , Photic Stimulation , Phylogeny , RNA, Messenger/genetics , RNA, Messenger/metabolism , Zebrafish/genetics , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics
13.
Trans R Soc Trop Med Hyg ; 108(9): 591-3, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25002461

ABSTRACT

BACKGROUND: C-reactive protein (CRP) level correlates with parasitemia and severity of malaria, but whether this reflects causality remains unknown. METHODS: Using CRP-transgenic and CRP-deficient mice we compared the onset and severity of experimental cerebral malaria (ECM) induced by Plasmodium berghei ANKA (PbA). RESULTS: CRP-deficient mice were most resistant to ECM. CONCLUSIONS: CRP might contribute to the development of cerebral malaria, rather than protect against it.


Subject(s)
C-Reactive Protein/metabolism , Malaria, Cerebral/physiopathology , Plasmodium berghei/isolation & purification , Animals , C-Reactive Protein/deficiency , C-Reactive Protein/genetics , Carrier Proteins , Malaria, Cerebral/immunology , Malaria, Cerebral/parasitology , Mice , Mice, Inbred C57BL , Plasmodium berghei/immunology , Sequence Deletion , Severity of Illness Index , Up-Regulation
17.
Immunology ; 142(3): 414-20, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24673624

ABSTRACT

No deficiency of human C-reactive protein (CRP), or even structural polymorphism of the protein, has yet been reported so its physiological role is not known. Here we show for the first time that CRP-deficient mice are remarkably susceptible to Streptococcus pneumoniae infection and are protected by reconstitution with isolated pure human CRP, or by anti-pneumococcal antibodies. Autologous mouse CRP is evidently essential for innate resistance to pneumococcal infection before antibodies are produced. Our findings are consistent with the significant association between clinical pneumococcal infection and non-coding human CRP gene polymorphisms which affect CRP expression. Deficiency or loss of function variation in CRP may therefore be lethal at the first early-life encounter with this ubiquitous virulent pathogen, explaining the invariant presence and structure of CRP in human adults.


Subject(s)
C-Reactive Protein/immunology , Immunity, Innate , Pneumococcal Infections/immunology , Streptococcus pneumoniae/immunology , Animals , C-Reactive Protein/deficiency , C-Reactive Protein/genetics , Humans , Mice , Mice, Knockout , Phenotype
18.
N Engl J Med ; 370(5): 421-32, 2014 Jan 30.
Article in English | MEDLINE | ID: mdl-24476432

ABSTRACT

BACKGROUND: The soluble pattern-recognition receptor known as long pentraxin 3 (PTX3) has a nonredundant role in antifungal immunity. The contribution of single-nucleotide polymorphisms (SNPs) in PTX3 to the development of invasive aspergillosis is unknown. METHODS: We screened an initial cohort of 268 patients undergoing hematopoietic stem-cell transplantation (HSCT) and their donors for PTX3 SNPs modifying the risk of invasive aspergillosis. The analysis was also performed in a multicenter study involving 107 patients with invasive aspergillosis and 223 matched controls. The functional consequences of PTX3 SNPs were investigated in vitro and in lung specimens from transplant recipients. RESULTS: Receipt of a transplant from a donor with a homozygous haplotype (h2/h2) in PTX3 was associated with an increased risk of infection, in both the discovery study (cumulative incidence, 37% vs. 15%; adjusted hazard ratio, 3.08; P=0.003) and the confirmation study (adjusted odds ratio, 2.78; P=0.03), as well as with defective expression of PTX3. Functionally, PTX3 deficiency in h2/h2 neutrophils, presumably due to messenger RNA instability, led to impaired phagocytosis and clearance of the fungus. CONCLUSIONS: Genetic deficiency of PTX3 affects the antifungal capacity of neutrophils and may contribute to the risk of invasive aspergillosis in patients treated with HSCT. (Funded by the European Society of Clinical Microbiology and Infectious Diseases and others.).


Subject(s)
Aspergillosis/genetics , C-Reactive Protein/deficiency , Hematopoietic Stem Cell Transplantation , Immunity, Innate/genetics , Neutrophils/immunology , Polymorphism, Single Nucleotide , Serum Amyloid P-Component/deficiency , Adult , Aspergillosis/immunology , C-Reactive Protein/genetics , Female , Genetic Predisposition to Disease , Haplotypes , Humans , Male , Serum Amyloid P-Component/genetics
19.
Behav Brain Res ; 257: 178-81, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24120400

ABSTRACT

Narp knockout (KO) mice demonstrate cognitive inflexibility and addictive behavior, which are associated with abnormal reactivity to a novel stimulus. To assess reactivity to novelty, we tested Narp KO and wild-type (WT) mice on a neophobia procedure. Both Narp KO and WT mice showed a similar decrease in consumption upon initial exposure to a novel flavor, but Narp KO mice did not increase consumption with subsequent exposures to the novel flavor like the WT mice. Therefore, Narp KO mice do not have abnormal reactivity to novelty but show deficits in adapting behavior to reflect the updated value of a stimulus.


Subject(s)
Adaptation, Psychological/physiology , C-Reactive Protein/deficiency , Exploratory Behavior/physiology , Nerve Tissue Proteins/deficiency , Phobic Disorders/physiopathology , Recovery of Function/genetics , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/genetics , Phobic Disorders/genetics , Time Factors
20.
Neuron ; 79(2): 335-46, 2013 Jul 24.
Article in English | MEDLINE | ID: mdl-23889936

ABSTRACT

The immediate early gene neuronal activity-regulated pentraxin (NARP) is an α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) binding protein that is specifically enriched at excitatory synapses onto fast-spiking parvalbumin-positive interneurons (FS [PV] INs). Here, we show that transgenic deletion of NARP decreases the number of excitatory synaptic inputs onto FS (PV) INs and reduces net excitatory synaptic drive onto FS (PV) INs. Accordingly, the visual cortex of NARP(-/-) mice is hyperexcitable and unable to express ocular dominance plasticity, although many aspects of visual function are unimpaired. Importantly, the number and strength of inhibitory synaptic contacts from FS (PV) INs onto principle neurons in the visual cortex is normal in NARP(-/-) mice, and enhancement of this output recovers the expression of experience-dependent synaptic plasticity. Thus the recruitment of inhibition from FS (PV) INs plays a central role in enabling the critical period for ocular dominance plasticity.


Subject(s)
C-Reactive Protein/deficiency , C-Reactive Protein/genetics , Critical Period, Psychological , Genes, Immediate-Early/physiology , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Neuronal Plasticity/genetics , Visual Cortex/physiology , Visual Pathways/physiology , Action Potentials/genetics , Animals , Dominance, Ocular/genetics , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL
...