Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Transplant ; 30: 9636897211033275, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34338573

RESUMO

Myocardial infarction (MI) is identified as the myocardial necrosis due to myocardial ischemia/reperfusion (I/R) injury and remains a leading cause of mortality. C1q/TNF-related protein 13 (CTRP13) is a member of CTRP family that has been found to be involved in coronary artery disease (CAD). However, the role of CTRP13 in MI remains unclear. We aimed to explore the functional role of CTRP13 in H9c2 cells exposed to hypoxia/reoxygenation (H/R). Our results demonstrated that H/R stimulation significantly decreased the expression of CTRP13 in H9c2 cells. H/R-induced an increase in ROS production and reductions in activities of SOD and CAT were prevented by CTRP13 overexpression but were aggravated by CTRP13 silencing. Moreover, CTRP13 overexpression could reverse the inductive effect of H/R on caspase-3 activity and bax expression, as well as the inhibitory effect of H/R on bcl-2 expression in H9c2 cells. However, CTRP13 silencing presented opposite effects with CTRP13 overexpression. Furthermore, CTRP13 overexpression enhanced the H/R-stimulated the expression levels of p-AMPK and nuclear Nrf2, and Nrf2 transcriptional activity. However, inhibition of AMPK reversed the CTRP13-mediated activation of Nrf2/ARE signaling and the cardiac-protective effect in H/R-exposed H9c2 cells. Additionally, silencing of Nrf2 reversed the protective effects of CTRP13 against H/R-stimulated oxidative stress and apoptosis in H9c2 cells. Finally, recombinant CTRP13 protein attenuated myocardial I/R-induced injury in rats. Taken together, these findings indicated that CTRP13 protected H9c2 cells from H/R-stimulated oxidative stress and apoptosis via regulating the AMPK/Nrf2/ARE signaling pathway. Our results provided evidence for the therapeutic potential of CTRP13 in myocardial I/R injury.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Adipocinas/metabolismo , Complemento C1q/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Miócitos Cardíacos/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Adipocinas/biossíntese , Adipocinas/genética , Animais , Hidrolases de Éster Carboxílico/genética , Hidrolases de Éster Carboxílico/metabolismo , Hipóxia Celular/fisiologia , Linhagem Celular , Complemento C1q/biossíntese , Complemento C1q/genética , Traumatismo por Reperfusão Miocárdica/patologia , Miócitos Cardíacos/patologia , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Ratos , Transdução de Sinais
2.
J Chemother ; 33(7): 476-485, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33825671

RESUMO

This study aims to investigate the effects of C1q-like 1 (C1QL1) on the growth and migration of lung adenocarcinoma (LUAD) cells and the underlying mechanism. The expression of C1QL1 in LUAD tissues and its prognostic value were analyzed using the data from The Cancer Genome Atlas (TCGA) database. To investigate the function of C1QL1, loss-of-function and gain-of-function assays were conducted in Calu-3 cells and LTEP-a-2 cells, respectively. Cell growth was evaluated by CCK-8 and colony formation assays. Transwell assays were performed to assess cell invasive and migratory abilities. qRT-PCR and Western blotting were performed to detect RNA and protein expression, respectively. Firstly, we found that C1QL1 was highly expressed and predicted poor outcomes in LUAD patients from TCGA database. Moreover, the mRNA and protein expression levels of C1QL1 were higher in LUAD cells than that in normal lung cells. Results of functional experiments illustrated that depletion of C1QL1 restrained the growth, invasion and migration of Calu-3 cells, meanwhile over-expression of C1QL1 presented the opposite results in LTEP-a-2 cells. Furthermore, we discovered that down-regulation of C1QL1 elevated the protein level of E-cadherin and reduced the protein levels of N-cadherin, Vimentin and Snail in Calu-3 cells, whereas over-expression of C1QL1 led to the opposite outcomes in LTEP-a-2 cells. Our data indicated that C1QL1 functioned as a crucial driver in LUAD cell growth and motility, which might be achieved by modulating epithelial-mesenchymal transition (EMT). These consequences are of important relevance for the design of therapeutic strategies for LUAD.


Assuntos
Adenocarcinoma de Pulmão/patologia , Complemento C1q/biossíntese , Neoplasias Pulmonares/patologia , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Biologia Computacional , Humanos , Invasividade Neoplásica/fisiopatologia , Prognóstico , Regulação para Cima
3.
Mol Neurobiol ; 57(5): 2290-2300, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32008166

RESUMO

The complement C1q plays a critical role in microglial phagocytosis of glutamatergic synapses and in the pathogenesis of neuroinflammation in Alzheimer's disease (AD). We recently reported that upregulation of metabotropic glutamate receptor signaling is associated with increased synaptic C1q production and subsequent microglial phagocytosis of synapses in the rodent models of AD. Here, we explored the role of astrocytic glutamate transporter in the synaptic C1q production and microglial phagocytosis of hippocampal glutamatergic synapses in a rat model of AD. Activation of astrocyte and reduction glutamate transporter 1 (GLT1) were noted after bilateral microinjection of amyloid-beta (Aß1-40) fibrils into the hippocampal CA1 area of rats. Ceftriaxone is a ß-lactam antibiotic that upregulates GLT1 expression. Bilateral microinjection of ceftriaxone recovered GLT1 expression, decreased synaptic C1q production, suppressed microglial phagocytosis of glutamatergic synapses in the hippocampal CA1, and attenuated synaptic and cognitive deficits in rats microinjected with Aß1-40. In contrast, artificial suppression of GLT1 activity by DL-threo-beta-benzyloxyaspartate (DL-TBOA) in naïve rats induced synaptic C1q expression and microglial phagocytosis of glutamatergic synapses in the hippocampal CA1 area, resulting in synaptic and cognitive dysfunction. These findings demonstrated that impairment of astrocytic glutamate transporter plays a role in the pathogenesis of AD.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides/toxicidade , Astrócitos/efeitos dos fármacos , Região CA1 Hipocampal/efeitos dos fármacos , Transtornos Cognitivos/induzido quimicamente , Complemento C1q/fisiologia , Transportador 2 de Aminoácido Excitatório/antagonistas & inibidores , Ácido Glutâmico/fisiologia , Microglia/fisiologia , Neurônios/metabolismo , Fragmentos de Peptídeos/toxicidade , Animais , Ácido Aspártico/farmacologia , Astrócitos/metabolismo , Região CA1 Hipocampal/metabolismo , Região CA1 Hipocampal/patologia , Ceftriaxona/farmacologia , Transtornos Cognitivos/metabolismo , Transtornos Cognitivos/patologia , Complemento C1q/biossíntese , Complemento C1q/genética , Modelos Animais de Doenças , Transportador 2 de Aminoácido Excitatório/biossíntese , Transportador 2 de Aminoácido Excitatório/genética , Transportador 2 de Aminoácido Excitatório/fisiologia , Masculino , Teste do Labirinto Aquático de Morris/efeitos dos fármacos , Teste do Labirinto Aquático de Morris/fisiologia , Técnicas de Patch-Clamp , Fagocitose/efeitos dos fármacos , Fagocitose/fisiologia , Ratos , Ratos Sprague-Dawley , Receptores de Glutamato/fisiologia , Sinapses/metabolismo , Regulação para Cima/efeitos dos fármacos
4.
Cell Biochem Funct ; 38(4): 392-400, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31904875

RESUMO

The circumventricular organs (CVOs) are the brain regions that lack the blood-brain barrier and allow free entry of blood-derived molecules, offering specialized niche to initiate rapid and early neuroinflammatory responses in the brain. Complement component 1q (C1q) is shown to be the first recognition component of the complement pathway and has a crucial function in the brain under pathological conditions. In the present study, we found that C1q expression in CX3CR1-positive microglia was increased in the CVOs and their neighbouring brain regions of adult mice at 1 day after a single administration of 1 mg/kg lipopolysaccharide (LPS), whereas it returned to control levels at 3 days after LPS stimulation. C1q expression was also seen to localize at synapsin-positive presynaptic axonal terminals in various brain regions. Thus, the present study demonstrates a transient upregulation of microglial C1q expression in the CVOs and their adjacent brain regions, indicating that a transient upregulation of C1q is possibly concerned with physiological responses at early phase of brain inflammation. SIGNIFICANCE OF THE STUDY: The circumventricular organs (CVOs) are specialized brain regions that lack the blood-brain barrier (BBB) and initiate neuroinflammatory responses in the brains. The present study showed that the expression of complement protein C1q was highly increased in microglia of the CVOs and their adjacent brain regions. Moreover, C1q expression was observed to localize specifically at presynaptic axonal terminals in the CVOs and their neighbouring brain regions. Thus, the present study indicates that C1q is possibly correlated with physiological responses at early phase of brain inflammation.


Assuntos
Encéfalo/metabolismo , Órgãos Circunventriculares/metabolismo , Complemento C1q/biossíntese , Regulação da Expressão Gênica/efeitos dos fármacos , Lipopolissacarídeos/toxicidade , Microglia/metabolismo , Animais , Axônios/metabolismo , Axônios/patologia , Encéfalo/patologia , Órgãos Circunventriculares/patologia , Inflamação/induzido quimicamente , Inflamação/metabolismo , Inflamação/patologia , Camundongos , Microglia/patologia
5.
Mediators Inflamm ; 2020: 8873152, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33424438

RESUMO

Atrial fibrillation (AF) is a highly prevalent cardiac arrhythmia characterized by atrial remodeling. Complement C1q tumor necrosis factor-related protein 3 (CTRP3) is one of the adipokines associated with obesity, diabetes, and coronary heart disease. The association between plasma CTRP3 levels and AF is uncertain. The aim of this study was to investigate whether plasma CTRP3 concentrations were correlated with AF. Our study included 75 AF patients who underwent catheter ablation at our hospital and 47 sinus rhythm patients to determine the difference in plasma CTRP3 concentrations. Blood samples before the ablation were collected, and ELISA was used to measure the concentrations of CTRP3. Plasma CTRP3 concentrations were significantly lower in AF patients compared with control group (366.9 ± 105.2 ng/ml vs. 429.1 ± 100.1 ng/ml, p = 0.002). In subgroup studies, patients with persistent AF had lower plasma CTRP3 concentrations than those with paroxysmal AF (328.3 ± 83.3 ng/ml vs. 380.0 ± 109.2 ng/ml, p = 0.037). The concentrations of plasma CTRP3 in the recurrence group after radiofrequency catheter ablation of AF were lower than those in the nonrecurrence group (337.9 ± 77.3 ng/ml vs. 386.6 ± 108.1 ng/ml, p = 0.045). Multivariate regression analysis revealed the independent correlation between plasma CTRP3 level and AF. Plasma CTRP3 concentrations were correlated with the presence of AF and AF recurrence.


Assuntos
Fibrilação Atrial/sangue , Complemento C1q/biossíntese , Fatores de Necrose Tumoral/sangue , Idoso , Estudos de Casos e Controles , Ablação por Cateter , Ecocardiografia , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Prognóstico , Recidiva
6.
Mol Neurodegener ; 13(1): 45, 2018 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-30126455

RESUMO

BACKGROUND: The role of the alternative complement pathway and its mediation by retinal microglia and macrophages, is well-established in the pathogenesis of Age-Related Macular Degeneration (AMD). However, the contribution of the classical complement pathway towards the progression of retinal degenerations is not fully understood, including the role of complement component 1q (C1q) as a critical activator molecule of the classical pathway. Here, we investigated the contribution of C1q to progressive photoreceptor loss and neuroinflammation in retinal degenerations. METHODS: Wild-type (WT), C1qa knockout (C1qa-/-) and mice treated with a C1q inhibitor (ANX-M1; Annexon Biosciences), were exposed to photo-oxidative damage (PD) and were observed for progressive lesion development. Retinal function was assessed by electroretinography, followed by histological analyses to assess photoreceptor degeneration. Retinal inflammation was investigated through complement activation, macrophage recruitment and inflammasome expression using western blotting, qPCR and immunofluorescence. C1q was localised in human AMD donor retinas using immunohistochemistry. RESULTS: PD mice had increased levels of C1qa which correlated with increasing photoreceptor cell death and macrophage recruitment. C1qa-/- mice did not show any differences in photoreceptor loss or inflammation at 7 days compared to WT, however at 14 days after the onset of damage, C1qa-/- retinas displayed less photoreceptor cell death, reduced microglia/macrophage recruitment to the photoreceptor lesion, and higher visual function. C1qa-/- mice displayed reduced inflammasome and IL-1ß expression in microglia and macrophages in the degenerating retina. Retinal neutralisation of C1q, using an intravitreally-delivered anti-C1q antibody, reduced the progression of retinal degeneration following PD, while systemic delivery had no effect. Finally, retinal C1q was found to be expressed by subretinal microglia/macrophages located in the outer retina of early AMD donor eyes, and in mouse PD retinas. CONCLUSIONS: Our data implicate subretinal macrophages, C1q and the classical pathway in progressive retinal degeneration. We demonstrate a role of local C1q produced by microglia/macrophages as an instigator of inflammasome activation and inflammation. Crucially, we have shown that retinal C1q neutralisation during disease progression may slow retinal atrophy, providing a novel strategy for the treatment of complement-mediated retinal degenerations including AMD.


Assuntos
Complemento C1q/biossíntese , Macrófagos/metabolismo , Degeneração Retiniana/metabolismo , Degeneração Retiniana/patologia , Animais , Progressão da Doença , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
7.
J Immunol ; 199(3): 1069-1085, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28687659

RESUMO

Inflammatory processes play a key role in pathophysiology of many neurologic diseases/trauma, but the effect of immune cells and factors on neurotransplantation strategies remains unclear. We hypothesized that cellular and humoral components of innate immunity alter fate and migration of human neural stem cells (hNSC). In these experiments, conditioned media collected from polymorphonuclear leukocytes (PMN) selectively increased hNSC astrogliogenesis and promoted cell migration in vitro. PMN were shown to generate C1q and C3a; exposure of hNSC to PMN-synthesized concentrations of these complement proteins promoted astrogliogenesis and cell migration. Furthermore, in vitro, Abs directed against C1q and C3a reversed the fate and migration effects observed. In a proof-of-concept in vivo experiment, blockade of C1q and C3a transiently altered hNSC migration and reversed astroglial fate after spinal cord injury. Collectively, these data suggest that modulation of the innate/humoral inflammatory microenvironment may impact the potential of cell-based therapies for recovery and repair following CNS pathology.


Assuntos
Astrócitos/fisiologia , Diferenciação Celular/fisiologia , Complemento C1q/biossíntese , Complemento C3a/biossíntese , Células-Tronco Neurais/fisiologia , Neutrófilos/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Movimento Celular , Células Cultivadas , Complemento C1q/antagonistas & inibidores , Complemento C1q/genética , Complemento C1q/imunologia , Complemento C3a/antagonistas & inibidores , Complemento C3a/genética , Complemento C3a/imunologia , Meios de Cultivo Condicionados , Humanos , Imunidade Inata , Camundongos , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/imunologia , Neutrófilos/imunologia , Traumatismos da Medula Espinal/imunologia , Traumatismos da Medula Espinal/fisiopatologia
8.
Exp Neurol ; 295: 184-193, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28601603

RESUMO

Microglia-mediated neuroinflammation is widely associated with seizures and epilepsy. Although microglial cells are professional phagocytes, less is known about the status of this phenotype in epilepsy. Recent evidence supports that phagocytosis-associated molecules from the classical complement (C1q-C3) play novel roles in microglia-mediated synaptic pruning. Interestingly, in human and experimental epilepsy, altered mRNA levels of complement molecules were reported. Therefore, to identify a potential role for complement and microglia in the synaptodendritic pathology of epilepsy, we determined the protein levels of classical complement proteins (C1q-C3) along with other phagocytosis signaling molecules in human epilepsy. Cortical brain samples surgically resected from patients with refractory epilepsy (RE) and non-epileptic lesions (NE) were examined. Western blotting was used to determine the levels of phagocytosis signaling proteins such as the complements C1q and C3, MerTK, Trem2, and Pros1 along with cleaved-caspase 3. In addition, immunostaining was used to determine the distribution of C1q and co-localization to microglia and dendrites. We found that the RE samples had significantly increased protein levels of C1q (p=0.034) along with those of its downstream activation product iC3b (p=0.027), and decreased levels of Trem2 (p=0.045) and Pros1 (p=0.005) when compared to the NE group. Protein levels of cleaved-caspase 3 were not different between the groups (p=0.695). In parallel, we found C1q localization to microglia and dendrites in both NE and RE samples, and also observed substantial microglia-dendritic interactions in the RE tissue. These data suggest that aberrant phagocytic signaling occurs in human refractory epilepsy. It is likely that alteration of phagocytic pathways may contribute to unwanted elimination of cells/synapses and/or impaired clearance of dead cells. Future studies will investigate whether altered complement signaling contributes to the hyperexcitability that result in epilepsy.


Assuntos
Ativação do Complemento/genética , Via Clássica do Complemento , Epilepsia/genética , Fagocitose/genética , Células Cultivadas , Córtex Cerebral/patologia , Complemento C1q/biossíntese , Complemento C1q/genética , Complemento C3/biossíntese , Complemento C3/genética , Via Clássica do Complemento/genética , Dendritos/genética , Dendritos/metabolismo , Epilepsia Resistente a Medicamentos/genética , Epilepsia Resistente a Medicamentos/patologia , Humanos , Microglia/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/biossíntese , Proteína Quinase 1 Ativada por Mitógeno/genética
9.
J Leukoc Biol ; 101(2): 481-491, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27630215

RESUMO

Antibodies against C1q (anti-C1q) are frequently found in patients with systemic lupus erythematosus (SLE). The anti-C1q antibodies strongly correlate with the occurrence of lupus nephritis and low-circulating C1q levels. Previous studies have demonstrated that myeloid cells, i.e., dendritic cells and macrophages, are a major source of C1q. However, a direct effect of anti-C1q on C1q secretion by macrophages has not yet been established. In the present study, we investigated the C1q secretion profile of in vitro human monocyte-derived macrophages (HMDMs) obtained from healthy donors and from patients with SLE. The effect of SLE patient-derived anti-C1q bound to immobilized C1q (imC1q) and imC1q alone on HMDMs was investigated by C1q secretion levels, the expression of membrane-bound and intracellular C1q using flow cytometry and ImageStreamX technology, and testing the ability of secreted C1q to activate the classical pathway (CP) of the complement. Bound anti-C1q induced significantly greater C1q secretion levels as compared with imC1q alone or healthy donor IgG. The extent of C1q secretion by HMDMs correlated with IgG anti-C1q levels of patients with SLE but not of healthy controls. Furthermore, bound autoantibodies and imC1q induced continuous and de novo C1q synthesis as evident by the intracellular C1q content, which correlated with C1q secretion levels. Finally, secreted C1q was able to activate the CP, as reflected by C4b deposition. Interestingly, anti-C1q-dependent C1q secretion could also be observed in SLE patient-derived cells. In conclusion, our data indicate that imC1q-bound anti-C1q strongly stimulate the C1q production by HMDMs. Anti-C1q-induced C1q secretion might be an important immune-modulatory factor in SLE.


Assuntos
Autoanticorpos/imunologia , Complemento C1q/biossíntese , Complemento C1q/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Macrófagos/metabolismo , Adulto , Idoso , Feminino , Citometria de Fluxo , Humanos , Proteínas Imobilizadas/metabolismo , Imunoglobulina G/metabolismo , Espaço Intracelular/metabolismo , Cinética , Receptores de Lipopolissacarídeos/metabolismo , Macrófagos/patologia , Masculino , Pessoa de Meia-Idade , Monócitos/patologia
10.
Mol Immunol ; 78: 164-170, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27648858

RESUMO

C1q is the initiation molecule of the classical pathway of the complement system and is produced by macrophages and immature dendritic cells. As mast cells share the same myeloid progenitor cells, we have studied whether also mast cells can produce and secrete C1q. Mast cells were generated in vitro from CD34+ progenitor cells from buffy coats or cord blood. Fully differentiated mast cells were shown by both RNA sequencing and qPCR to express C1QA, C1QB and C1QC. C1q produced by mast cells has a similar molecular make-up as serum C1q. Reconstituting C1q depleted serum with mast cell supernatant in haemolytic assays, indicated that C1q secreted by mast cells is functionally active. The level of C1q in supernatants produced under basal conditions was considerably enhanced upon stimulation with LPS, dexamethasone in combination with IFN- γ or via FcεRI triggering. Mast cells in human tissues stained positive for C1q in both healthy and in inflamed tissue. Moreover, mast cells in healthy and diseased skin appear to be the predominant C1q positive cells. Together, our data reveal that mast cells are able to produce and secrete functional active C1q and indicate mast cells as a local source of C1q in human tissue.


Assuntos
Complemento C1q/biossíntese , Mastócitos/imunologia , Western Blotting , Separação Celular , Células Cultivadas , Complemento C1q/metabolismo , Ensaio de Imunoadsorção Enzimática , Imunofluorescência , Humanos , Mastócitos/metabolismo
11.
Neuron ; 91(5): 1034-1051, 2016 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-27478018

RESUMO

C1ql3 is a secreted neuronal protein that binds to BAI3, an adhesion-class GPCR. C1ql3 is homologous to other gC1q-domain proteins that control synapse numbers, but a role for C1ql3 in regulating synapse density has not been demonstrated. We show in cultured neurons that C1ql3 expression is activity dependent and supports excitatory synapse density. Using newly generated conditional and constitutive C1ql3 knockout mice, we found that C1ql3-deficient mice exhibited fewer excitatory synapses and diverse behavioral abnormalities, including marked impairments in fear memories. Using circuit-tracing tools and conditional ablation of C1ql3 targeted to specific brain regions, we demonstrate that C1ql3-expressing neurons in the basolateral amygdala project to the medial prefrontal cortex, that these efferents contribute to fear memory behavior, and that C1ql3 is required for formation and/or maintenance of these synapses. Our results suggest that C1ql3 is a signaling protein essential for subsets of synaptic projections and the behaviors controlled by these projections.


Assuntos
Tonsila do Cerebelo/fisiologia , Complemento C1q/fisiologia , Memória/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Núcleo Accumbens/fisiologia , Córtex Pré-Frontal/fisiologia , Sinapses/fisiologia , Animais , Células Cultivadas , Complemento C1q/biossíntese , Complemento C1q/genética , Masculino , Camundongos , Camundongos Knockout , Mutação , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Vias Neurais/fisiologia , Neurônios/metabolismo , Neurônios/fisiologia , Sinapses/metabolismo
12.
Nanomedicine ; 12(4): 1033-1043, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26767511

RESUMO

When inhaled nanoparticles deposit in the lungs, they transit through respiratory tract lining fluid (RTLF) acquiring a biomolecular corona reflecting the interaction of the RTLF with the nanomaterial surface. Label-free snapshot proteomics was used to generate semi-quantitative profiles of corona proteins formed around silica (SiO2) and poly(vinyl) acetate (PVAc) nanoparticles in RTLF, the latter employed as an archetype drug delivery vehicle. The evolved PVAc corona was significantly enriched compared to that observed on SiO2 nanoparticles (698 vs. 429 proteins identified); however both coronas contained a substantial contribution from innate immunity proteins, including surfactant protein A, napsin A and complement (C1q and C3) proteins. Functional protein classification supports the hypothesis that corona formation in RTLF constitutes opsonisation, preparing particles for phagocytosis and clearance from the lungs. These data highlight how an understanding of the evolved corona is necessary for the design of inhaled nanomedicines with acceptable safety and tailored clearance profiles. FROM THE CLINICAL EDITOR: Inhaled nanoparticles often acquire a layer of protein corona while they go through the respiratory tract. Here, the authors investigated the identity of these proteins. The proper identification would improve the understanding of the use of inhaled nanoparticles in future therapeutics.


Assuntos
Sistemas de Liberação de Medicamentos , Nanopartículas/administração & dosagem , Coroa de Proteína , Sistema Respiratório/metabolismo , Adulto , Ácido Aspártico Endopeptidases/biossíntese , Ácido Aspártico Endopeptidases/isolamento & purificação , Líquidos Corporais/metabolismo , Complemento C1q/biossíntese , Complemento C1q/isolamento & purificação , Complemento C3/biossíntese , Complemento C3/isolamento & purificação , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Nanopartículas/efeitos adversos , Proteômica , Proteína A Associada a Surfactante Pulmonar/biossíntese , Proteína A Associada a Surfactante Pulmonar/isolamento & purificação , Sistema Respiratório/efeitos dos fármacos , Dióxido de Silício/administração & dosagem , Dióxido de Silício/química
13.
J Biol Chem ; 290(38): 23039-49, 2015 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-26231212

RESUMO

Calcium-binding proteins (CaBPs) such as parvalbumin are part of the cellular calcium buffering system that determines intracellular calcium diffusion and influences the spatiotemporal dynamics of calcium signals. In neurons, CaBPs are primarily localized to the cytosol and function, for example, in nerve terminals in short-term synaptic plasticity. However, CaBPs are also expressed in the cell nucleus, suggesting that they modulate nuclear calcium signals, which are key regulators of neuronal gene expression. Here we show that the calcium buffering capacity of the cell nucleus in mouse hippocampal neurons regulates neuronal architecture by modulating the expression levels of VEGFD and the complement factor C1q-c, two nuclear calcium-regulated genes that control dendrite geometry and spine density, respectively. Increasing the levels of nuclear calcium buffers by means of expression of a nuclearly targeted form of parvalbumin fused to mCherry (PV.NLS-mC) led to a reduction in VEGFD expression and, as a result, to a decrease in total dendritic length and complexity. In contrast, mRNA levels of the synapse pruning factor C1q-c were increased in neurons expressing PV.NLS-mC, causing a reduction in the density and size of dendritic spines. Our results establish a close link between nuclear calcium buffering capacity and the transcription of genes that determine neuronal structure. They suggest that the development of cognitive deficits observed in neurological conditions associated with CaBP deregulation may reflect the loss of necessary structural features of dendrites and spines.


Assuntos
Cálcio/metabolismo , Espinhas Dendríticas/metabolismo , Hipocampo/metabolismo , Rede Nervosa/metabolismo , Animais , Complemento C1q/biossíntese , Regulação da Expressão Gênica/fisiologia , Hipocampo/citologia , Camundongos , Rede Nervosa/citologia , Ratos , Ratos Sprague-Dawley , Fator D de Crescimento do Endotélio Vascular/biossíntese
14.
Dev Comp Immunol ; 48(1): 151-63, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25312696

RESUMO

The C1q family includes many proteins that contain a globular (gC1q) domain, and this family is widely conserved from bacteria to mammals. The family is divided into three subgroups: C1q, C1q-like and ghC1q. In this study, a novel C1q family member, sghC1q, was cloned and identified from Cynoglossus semilaevis (named CssghC1q). The full-length CssghC1q cDNA spans 905 bp, including an open reading frame (ORF) of 768 bp, a 5'-untranslated region (UTR) of 25 bp and a 3'-UTR of 112 bp. The ORF encodes a putative protein of 255 amino acids (aa) with a deduced molecular weight of 28 kDa. The predicted protein contains a signal peptide (aa 1-19), a coiled-coil region (aa 61-102) and a globular C1q (gC1q) domain (aa 117-255). Protein sequence alignment indicated that the C-terminus of CssghC1q is highly conserved across several species. Phylogenetic analysis indicated that CssghC1q is most closely related to Maylandia zebra C1q-like-2-like. The CssghC1q genomic sequence spanned 1562 bp, with three exons and two introns. CssghC1q is constitutively expressed in all evaluated tissues, with the highest expression in the liver and the weakest in the heart. After a challenge with Vibrio anguillarum, CssghC1q transcript levels exhibited distinct time-dependent response patterns in the blood, head kidney, skin, spleen, intestine and liver. Recombinant CssghC1q protein exhibited antimicrobial activities against Gram-negative bacteria, Gram-positive bacteria and viruses. The minimum inhibitory concentration (MIC) values against Vibrio harveyi, Vibrio anguillarum, Pseudomonas aeruginosa and Staphylococcus aureus were 0.043 mg/mL, 0.087 mg/mL, 0.174 mg/mL and 0.025 mg/mL, respectively. A low concentration (0.06 mg/mL) of CssghC1q showed significant antiviral activity in vitro against nervous necrosis virus (NNV). These results suggest that CssghC1q plays a vital role in immune defense against bacteria and viruses.


Assuntos
Antibacterianos/imunologia , Antivirais/imunologia , Complemento C1q/genética , Complemento C1q/imunologia , Doenças dos Peixes/imunologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Clonagem Molecular , Complemento C1q/biossíntese , Sequência Conservada , Doenças dos Peixes/microbiologia , Doenças dos Peixes/virologia , Linguados , Perfilação da Expressão Gênica , Testes de Sensibilidade Microbiana , Dados de Sequência Molecular , Nodaviridae/imunologia , Infecções por Pseudomonas/imunologia , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/imunologia , Alinhamento de Sequência , Análise de Sequência de DNA , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/imunologia , Vibrio/imunologia , Vibrioses/imunologia , Vibrioses/microbiologia
15.
J Immunol ; 192(3): 1138-53, 2014 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-24353269

RESUMO

The complement system is activated in a wide spectrum of CNS diseases and is suggested to play a role in degenerative phenomena such as elimination of synaptic terminals. Still, little is known of mechanisms regulating complement activation in the CNS. Loss of synaptic terminals in the spinal cord after an experimental nerve injury is increased in the inbred DA strain compared with the PVG strain and is associated with expression of the upstream complement components C1q and C3, in the absence of membrane attack complex activation and neutrophil infiltration. To further dissect pathways regulating complement expression, we performed genome-wide expression profiling and linkage analysis in a large F2(DA × PVG) intercross, which identified quantitative trait loci regulating expression of C1qa, C1qb, C3, and C9. Unlike C1qa, C1qb, and C9, which all displayed distinct coregulation with different cis-regulated C-type lectins, C3 was regulated in a coexpression network immediately downstream of butyrylcholinesterase. Butyrylcholinesterase hydrolyses acetylcholine, which exerts immunoregulatory effects partly through TNF-α pathways. Accordingly, increased C3, but not C1q, expression was demonstrated in rat and mouse glia following TNF-α stimulation, which was abrogated in a dose-dependent manner by acetylcholine. These findings demonstrate new pathways regulating CNS complement expression using unbiased mapping in an experimental in vivo system. A direct link between cholinergic activity and complement activation is supported by in vitro experiments. The identification of distinct pathways subjected to regulation by naturally occurring genetic variability is of relevance for the understanding of disease mechanisms in neurologic conditions characterized by neuronal injury and complement activation.


Assuntos
Sistema Nervoso Central/metabolismo , Fibras Colinérgicas/fisiologia , Ativação do Complemento , Complemento C3/biossíntese , Regulação da Expressão Gênica/imunologia , Redes Reguladoras de Genes , Acetilcolina/farmacologia , Acetilcolina/fisiologia , Animais , Animais Congênicos , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Lesões Encefálicas/imunologia , Lesões Encefálicas/fisiopatologia , Butirilcolinesterase/fisiologia , Células Cultivadas , Sistema Nervoso Central/química , Sistema Nervoso Central/patologia , Complemento C1q/biossíntese , Complemento C1q/genética , Complemento C3/genética , Denervação , Fatores de Transcrição Forkhead/metabolismo , Ligação Genética , Estudo de Associação Genômica Ampla , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Microglia/metabolismo , Locos de Características Quantitativas , Ratos , Rizotomia , Organismos Livres de Patógenos Específicos , Raízes Nervosas Espinhais/cirurgia , Sinaptofisina/análise , Fator de Necrose Tumoral alfa/farmacologia , Fator de Necrose Tumoral alfa/fisiologia
16.
J Neurosci ; 33(33): 13460-74, 2013 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-23946404

RESUMO

The decline of cognitive function has emerged as one of the greatest health threats of old age. Age-related cognitive decline is caused by an impacted neuronal circuitry, yet the molecular mechanisms responsible are unknown. C1q, the initiating protein of the classical complement cascade and powerful effector of the peripheral immune response, mediates synapse elimination in the developing CNS. Here we show that C1q protein levels dramatically increase in the normal aging mouse and human brain, by as much as 300-fold. This increase was predominantly localized in close proximity to synapses and occurred earliest and most dramatically in certain regions of the brain, including some but not all regions known to be selectively vulnerable in neurodegenerative diseases, i.e., the hippocampus, substantia nigra, and piriform cortex. C1q-deficient mice exhibited enhanced synaptic plasticity in the adult and reorganization of the circuitry in the aging hippocampal dentate gyrus. Moreover, aged C1q-deficient mice exhibited significantly less cognitive and memory decline in certain hippocampus-dependent behavior tests compared with their wild-type littermates. Unlike in the developing CNS, the complement cascade effector C3 was only present at very low levels in the adult and aging brain. In addition, the aging-dependent effect of C1q on the hippocampal circuitry was independent of C3 and unaccompanied by detectable synapse loss, providing evidence for a novel, complement- and synapse elimination-independent role for C1q in CNS aging.


Assuntos
Envelhecimento/metabolismo , Encéfalo/metabolismo , Complemento C1q/biossíntese , Animais , Comportamento Animal , Western Blotting , Encéfalo/fisiologia , Eletrofisiologia , Potenciais Pós-Sinápticos Excitadores , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Imunoeletrônica
17.
Biochem J ; 447(2): 229-37, 2012 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-22812635

RESUMO

C1q deficiency is the strongest known risk factor for SLE (systemic lupus erythematosus) but its endogenous cellular origin remains limitedly understood. In the present study we investigate the production of C1q by both cultured and endogenous bone osteoclasts. Blood monocytes were cultured with RANKL (receptor activator of nuclear factor κB ligand) and M-CSF (macrophage colony-stimulating factor) to generate osteoclasts and these cells expressed C1Q mRNA and also secreted C1q protein. Intracellular C1q was detectable in developing osteoclasts at day 3 by Western blotting and was also detectable by flow cytometry. By immunofluorescence microscopy, C1q was preferentially detected in immature osteoclasts. By multiple detection methods, C1q expression was markedly increased after IFNγ (interferon γ) treatment. By immunohistochemistry, C1q was also detected in endogenous bone osteoclasts. When osteoclasts were cultured on immobilized C1q, these cells exhibited 2-7-fold increases in the expression of signature osteoclast genes [TRAP (tartrate-resistant acid phosphatase), cathepsin K, calcitonin receptor, carbonic anhydrase II and NFATc1 (nuclear factor of activated T-cells, cytoplasmic, calcineurin-dependent 1)], suggesting an osteoclastogenic capability. This is the first report of C1q production by osteoclasts. Its ability to enhance osteoclast development implies reduced osteoclastogenesis in patients with SLE as they often experience decreased C1q levels. This is consistent with the non-erosive nature of lupus arthritis.


Assuntos
Complemento C1q/biossíntese , Osteoclastos/metabolismo , Catepsina K/biossíntese , Diferenciação Celular , Células Cultivadas , Humanos , Interferon gama/farmacologia , Lúpus Eritematoso Sistêmico/fisiopatologia , Monócitos/metabolismo , Monócitos/fisiologia , Osteoclastos/citologia , RNA Mensageiro/metabolismo
18.
Neurosci Res ; 71(3): 289-93, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21801761

RESUMO

Schizophrenia (SZ) is a psychiatric disease with plausible neurodevelopmental etiology. Although genetic studies show significant association of immune molecules loci such as major histocompatibility complex (MHC) class I with SZ, it is not clear whether these immune molecules are involved in the pathology observed in SZ brains. MHC class I and the classical pathway components of complement system (C1q and C3) have been shown to regulate brain neuronal maturation and function. We have examined the expression of MHC class I and complement protein C3 in two frontal cortical regions of postmortem brains of SZ patients. Since cigarette smoking may modulate MHC class I protein expression and a higher rate of smoking is observed in SZ patients, we studied the expression of MHC class I and C3 in relation to the presence of smoking. We found that MHC class I protein expression is reduced in the dorsolateral prefrontal cortex (DLPFC) but not in the orbitofrontal cortex (OFC) of nonsmoker SZ patients. We did not observe SZ-associated changes in C3 mRNA expression. Our exploratory research suggests a potential involvement of MHC class I in SZ and implies that smoking might modulate its expression.


Assuntos
Antígenos de Histocompatibilidade Classe I/biossíntese , Antígenos de Histocompatibilidade Classe I/genética , Córtex Pré-Frontal/metabolismo , Esquizofrenia/metabolismo , Fumar/metabolismo , Complemento C1q/biossíntese , Complemento C1q/genética , Complemento C1q/fisiologia , Complemento C3/biossíntese , Complemento C3/genética , Complemento C3/fisiologia , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/imunologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/imunologia , Humanos , Nicotina/farmacologia , Agonistas Nicotínicos/farmacologia , Córtex Pré-Frontal/imunologia , Córtex Pré-Frontal/patologia , Esquizofrenia/imunologia , Esquizofrenia/patologia , Fumar/imunologia
19.
J Neuroimmunol ; 236(1-2): 39-46, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21640391

RESUMO

Here we analyzed C1q, the initial recognition subcomponent of classical complement activation cascade, in an experimental model of Parkinson disease (PD). Nigrostriatal dopaminergic pathway injury was induced by treatment of wildtype mice subchronically with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Constitutive expression of C1q was restricted to microglia throughout the brain, and microglial C1q expression was early and transiently upregulated after MPTP in the substantia nigra (SN) and striatum, as analyzed by immunohistochemistry and in situ hybridization. C1q-positive microglia exhibited morphological characteristics of activated macrophage-type of cells, co-stained for MHCII, proliferated and were in close contact with degenerating dopaminergic neurons and fibers in the MPTP-lesioned SN. However, mice deficient in functional C1q protein were not significantly different in MPTP-induced loss of nigral dopaminergic neurons, striatal dopaminergic fibers and dopamine levels than their control littermates. In conclusion, C1q is upregulated and considered to be a marker of microglial activation in the nigrostriatal system after subchronic MPTP, but nigrostriatal dopaminergic injury may be not affected by C1q in this model.


Assuntos
Complemento C1q/biossíntese , Corpo Estriado/metabolismo , Dopamina/fisiologia , Intoxicação por MPTP/metabolismo , Microglia/metabolismo , Doença de Parkinson/metabolismo , Substância Negra/metabolismo , Regulação para Cima/fisiologia , Animais , Corpo Estriado/patologia , Modelos Animais de Doenças , Intoxicação por MPTP/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/patologia , Doença de Parkinson/patologia , Substância Negra/patologia
20.
J Gastrointest Surg ; 14(8): 1207-13, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20496011

RESUMO

AIMS: C1q, an element of the first component of complement, is known to be expressed by interdigitating and follicular dendritic cells in the spleen, where it has been suggested that C1q is involved in capturing immune complexes. The present study investigated whether C1q is expressed in Barrett's esophagus and esophageal adenocarcinoma and, if so, whether its expression is associated with dendritic cells. MATERIAL AND METHODS: Endoscopic biopsy or operative surgical specimens were obtained from 15 patients with Barrett's esophagus, 13 patients with esophageal adenocarcinoma and 12 patients whose biopsy specimens did not show the presence of specialized intestinal metaplasia or adenocarcinoma. Barrett's esophagus was diagnosed by the presence of a macroscopic area of columnar-lined esophagus as well as microscopic intestinal metaplasia with goblet cells. Immunohistochemistry utilizing anti-C1q and markers for dendritic cells and macrophages was performed on sections of tissue samples embedded in paraffin. Double immunostaining with C1q/CD83 and C1q/CD68 was used to analyze the possible co-localization of C1q with dendritic cells and macrophages. The expression of C1q by dendritic cells and macrophages was also examined in in vitro studies using reverse transcriptase polymerase chain reaction (RT-PCR) and Western blotting. RESULTS: In all specimens studied, C1q expression was detected as being distributed irregularly throughout the lamina propria. A computerized quantitative analysis showed that C1q expression was significantly higher in tissue specimens without specialized intestinal-type metaplasia than in Barrett's esophagus specimens and specimens with adenocarcinoma. Double immunostaining revealed that dendritic cells and macrophages expressed C1q in all analyzed esophageal specimens. The expression of C1q by dendritic cells and macrophages was also demonstrated in in vitro studies using RT-PCR and Western blotting. CONCLUSION: The findings suggest that reduced levels of the expression of C1q by dendritic cells and macrophages in the esophagus may play a role in the formation of immune responses associated with the formation of specialized intestinal metaplasia and the development of adenocarcinoma.


Assuntos
Adenocarcinoma/genética , Esôfago de Barrett/genética , Complemento C1q/genética , Neoplasias Esofágicas/genética , Regulação Neoplásica da Expressão Gênica , Lesões Pré-Cancerosas/genética , RNA Neoplásico/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Esôfago de Barrett/metabolismo , Esôfago de Barrett/patologia , Biópsia , Western Blotting , Complemento C1q/biossíntese , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patologia , Humanos , Imuno-Histoquímica , Lesões Pré-Cancerosas/metabolismo , Lesões Pré-Cancerosas/patologia , RNA Neoplásico/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...