Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 118
Filtrar
1.
J Neuroinflammation ; 18(1): 177, 2021 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-34399786

RESUMO

BACKGROUND: Synucleinopathies are characterized by neurodegeneration and deposition of the presynaptic protein α-synuclein in pathological protein inclusions. Growing evidence suggests the complement system not only has physiological functions in the central nervous system, but also is involved in mediating the pathological loss of synapses in Alzheimer's disease. However, it is not established whether the complement system has a similar role in the diseases Parkinson's disease, Dementia with Lewy bodies, and multiple system atrophy (MSA) that are associated with α-synuclein aggregate pathology. METHODS: To investigate if the complement system has a pathological role in synucleinopathies, we assessed the effect of the complement system on the viability of an α-synuclein expressing cell model and examined direct activation of the complement system by α-synuclein in a plate-based activation assay. Finally, we investigated the levels of the initiator of the classical pathway, C1q, in postmortem brain samples from MSA patients. RESULTS: We demonstrate that α-synuclein activates the classical complement pathway and mediates complement-dependent toxicity in α-synuclein expressing SH-SY5Y cells. The α-synuclein-dependent cellular toxicity was rescued by the complement inhibitors RaCI (inhibiting C5) and Cp20 (inhibiting C3). Furthermore, we observed a trend for higher levels of C1q in the putamen of MSA subjects than that of controls. CONCLUSION: α-Synuclein can activate the classical complement pathway, and the complement system is involved in α-synuclein-dependent cellular cytotoxicity suggesting the system could play a prodegenerative role in synucleinopathies.


Assuntos
Via Clássica do Complemento/fisiologia , Corpos de Inclusão/metabolismo , Córtex Visual/metabolismo , alfa-Sinucleína/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Linhagem Celular Tumoral , Feminino , Humanos , Corpos de Inclusão/patologia , Masculino , Pessoa de Meia-Idade , Córtex Visual/patologia
2.
Front Immunol ; 12: 649882, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33868287

RESUMO

Intestinal ischemia reperfusion (IR)-induced tissue injury represents an acute inflammatory response with significant morbidity and mortality. The mechanism of IR-induced injury is not fully elucidated, but recent studies suggest a critical role for complement activation and for differences between sexes. To test the hypothesis that complement initiation differs by sex in intestinal IR, we performed intestinal IR on male and female WT C57B6L/, C1q-/-, MBL-/-, or properdin (P)-/- mice. Intestinal injury, C3b and C5a production and ex vivo secretions were analyzed. Initial studies demonstrated a difference in complement mRNA and protein in male and female WT mice. In response to IR, male C1q-, MBL- and P-deficient mice sustained less injury than male WT mice. In contrast, only female MBL-/- mice sustained significantly less injury than female wildtype mice. Importantly, wildtype, C1q-/- and P-/- female mice sustained significant less injury than the corresponding male mice. In addition, both C1q and MBL expression and deposition increased in WT male mice, while only elevated MBL expression and deposition occurred in WT female mice. These data suggested that males use both C1q and MBL pathways, while females tend to depend on lectin pathway during intestinal IR. Females produced significantly less serum C5a in MBL-/- and P-/- mice. Our findings suggested that complement activation plays a critical role in intestinal IR in a sex-dependent manner.


Assuntos
Complemento C1q/metabolismo , Via Clássica do Complemento/fisiologia , Lectina de Ligação a Manose da Via do Complemento/fisiologia , Lectina de Ligação a Manose/metabolismo , Traumatismo por Reperfusão/imunologia , Animais , Complemento C1q/genética , Modelos Animais de Doenças , Feminino , Humanos , Intestinos/irrigação sanguínea , Intestinos/imunologia , Intestinos/patologia , Masculino , Lectina de Ligação a Manose/genética , Camundongos , Camundongos Knockout , Properdina/genética , Properdina/metabolismo , Traumatismo por Reperfusão/patologia , Fatores Sexuais
3.
Exp Eye Res ; 203: 108402, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33326809

RESUMO

PURPOSE: To develop and characterize a new type of plasma rich in growth factors (PRGF) membrane for patients in which immune system is involved in the disease etiology. METHODS: Blood from three healthy donors was collected to obtain the different fibrin membranes by PRGF technology. PRGF obtained volumes were activated and divided into two groups: PRGF membrane (mPRGF) obtained after incubation at 37 °C for 30 min (control); and is-mPRGF: mPRGF obtained after incubation for 30 min at 56 °C. The concentration of several growth factors, proteins, immunoglobulin E and the complement activity was determined in the different mPRGF. The proliferative potential of heat-inactivated mPRGF were assayed on keratocytes (HK) and conjunctival fibroblasts (HConF). In addition, morphological and physical features of the inactivated mPRGF were evaluated in contrast to the control mPRGF. RESULTS: Heat-inactivation of the mPRGF preserves the content of most of the growth factors involved in the ocular wound healing while reducing drastically the content of IgE and the complement activity. The heat-inactivated mPRGF conserve the morphological and physical characteristics of the fibrin meshwork in comparison with the control mPRGF. Furthermore, no significant differences were found in the biological activity of the control mPRGF regarding the heat-inactivated mPRGF (is-mPRGF) in any of both ocular cell types evaluated. CONCLUSIONS: The heat-inactivation of the PRGF membranes (is-mPRGF) reduces drastically the content of IgE and complement activity while preserving the content of most of the proteins and morphogens involved in ocular wound healing. Furthermore, the morphological and physical features of the immunosafe mPRGF were also preserved after heat-inactivation.


Assuntos
Membrana Celular/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Fibrina Rica em Plaquetas/metabolismo , Tecnologia Biomédica , Doadores de Sangue , Células Cultivadas , Via Clássica do Complemento/fisiologia , Túnica Conjuntiva/citologia , Ceratócitos da Córnea/metabolismo , Ensaio de Imunoadsorção Enzimática , Fator de Crescimento Epidérmico/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Fibroblastos/metabolismo , Técnica Indireta de Fluorescência para Anticorpo , Humanos , Imunoglobulina E/imunologia , Microscopia Eletrônica de Varredura , Fator de Crescimento Derivado de Plaquetas/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
4.
Cell Rep ; 29(10): 3087-3100.e7, 2019 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-31801075

RESUMO

Movement is an essential behavior requiring the assembly and refinement of spinal motor circuits. However, the mechanisms responsible for circuit refinement and synapse maintenance are poorly understood. Similarly, the molecular mechanisms by which gene mutations cause dysfunction and elimination of synapses in neurodegenerative diseases that occur during development are unknown. Here, we demonstrate that the complement protein C1q is required for the refinement of sensory-motor circuits during normal development, as well as for synaptic dysfunction and elimination in spinal muscular atrophy (SMA). C1q tags vulnerable SMA synapses, which triggers activation of the classical complement pathway leading to microglia-mediated elimination. Pharmacological inhibition of C1q or depletion of microglia rescues the number and function of synapses, conferring significant behavioral benefit in SMA mice. Thus, the classical complement pathway plays critical roles in the refinement of developing motor circuits, while its aberrant activation contributes to motor neuron disease.


Assuntos
Via Clássica do Complemento/fisiologia , Microglia/metabolismo , Atrofia Muscular Espinal/metabolismo , Animais , Pré-Escolar , Complemento C1q/metabolismo , Modelos Animais de Doenças , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios Motores/metabolismo , Sinapses/metabolismo
5.
Annu Rev Genet ; 53: 263-288, 2019 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-31518519

RESUMO

Advances in human genetics have implicated a growing number of genes in neurodegenerative diseases, providing insight into pathological processes. For Alzheimer disease in particular, genome-wide association studies and gene expression studies have emphasized the pathogenic contributions from microglial cells and motivated studies of microglial function/dysfunction. Here, we summarize recent genetic evidence for microglial involvement in neurodegenerative disease with a focus on Alzheimer disease, for which the evidence is most compelling. To provide context for these genetic discoveries, we discuss how microglia influence brain development and homeostasis, how microglial characteristics change in disease, and which microglial activities likely influence the course of neurodegeneration. In all, we aim to synthesize varied aspects of microglial biology and highlight microglia as possible targets for therapeutic interventions in neurodegenerative disease.


Assuntos
Encéfalo/crescimento & desenvolvimento , Microglia/patologia , Microglia/fisiologia , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/fisiopatologia , Envelhecimento/fisiologia , Doença de Alzheimer/genética , Doença de Alzheimer/fisiopatologia , Animais , Encéfalo/fisiologia , Sistema Nervoso Central/metabolismo , Via Clássica do Complemento/fisiologia , Regulação da Expressão Gênica , Predisposição Genética para Doença , Homeostase , Humanos , Macrófagos/fisiologia , Placa Amiloide/fisiopatologia , Fator de Crescimento Transformador beta/metabolismo
6.
PLoS Pathog ; 15(3): e1007659, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30897158

RESUMO

The carboxy-terminal domain of the BBK32 protein from Borrelia burgdorferi sensu stricto, termed BBK32-C, binds and inhibits the initiating serine protease of the human classical complement pathway, C1r. In this study we investigated the function of BBK32 orthologues of the Lyme-associated Borrelia burgdorferi sensu lato complex, designated BAD16 from B. afzelii strain PGau and BGD19 from B. garinii strain IP90. Our data show that B. afzelii BAD16-C exhibits BBK32-C-like activities in all assays tested, including high-affinity binding to purified C1r protease and C1 complex, and potent inhibition of the classical complement pathway. Recombinant B. garinii BGD19-C also bound C1 and C1r with high-affinity yet exhibited significantly reduced in vitro complement inhibitory activities relative to BBK32-C or BAD16-C. Interestingly, natively produced BGD19 weakly recognized C1r relative to BBK32 and BAD16 and, unlike these proteins, BGD19 did not confer significant protection from serum killing. Site-directed mutagenesis was performed to convert BBK32-C to resemble BGD19-C at three residue positions that are identical between BBK32 and BAD16 but different in BGD19. The resulting chimeric protein was designated BXK32-C and this BBK32-C variant mimicked the properties observed for BGD19-C. To query the disparate complement inhibitory activities of BBK32 orthologues, the crystal structure of BBK32-C was solved to 1.7Å limiting resolution. BBK32-C adopts an anti-parallel four-helix bundle fold with a fifth alpha-helix protruding from the helical core. The structure revealed that the three residues targeted in the BXK32-C chimera are surface-exposed, further supporting their potential relevance in C1r binding and inhibition. Additional binding assays showed that BBK32-C only recognized C1r fragments containing the serine protease domain. The structure-function studies reported here improve our understanding of how BBK32 recognizes and inhibits C1r and provide new insight into complement evasion mechanisms of Lyme-associated spirochetes of the B. burgdorferi sensu lato complex.


Assuntos
Proteínas de Bactérias/genética , Borrelia burgdorferi/genética , Via Clássica do Complemento/genética , Proteínas de Bactérias/imunologia , Proteínas de Bactérias/ultraestrutura , Borrelia burgdorferi/imunologia , Grupo Borrelia Burgdorferi , Complemento C1r/metabolismo , Via Clássica do Complemento/fisiologia , Proteínas do Sistema Complemento/metabolismo , Humanos , Doença de Lyme/fisiopatologia , Domínios Proteicos/fisiologia , Proteínas Recombinantes , Análise de Sequência de Proteína
7.
PLoS One ; 13(6): e0198644, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29874282

RESUMO

The complement system is an intricate defense network that rapidly removes invading pathogens. Although many complement regulators are present to protect host cells under homeostasis, the impairment of Factor H (FH) regulatory mechanism has been associated with several autoimmune and inflammatory diseases. To understand the dynamics involved in the pivotal balance between activation and regulation, we have developed a comprehensive computational model of the alternative and classical pathways of the complement system. The model is composed of 290 ordinary differential equations with 142 kinetic parameters that describe the state of complement system under homeostasis and disorder through FH impairment. We have evaluated the state of the system by generating concentration-time profiles for the biomarkers C3, C3a-desArg, C5, C5a-desArg, Factor B (FB), Ba, Bb, and fC5b-9 that are influenced by complement dysregulation. We show that FH-mediated disorder induces substantial levels of complement activation compared to homeostasis, by generating reduced levels of C3 and FB, and to a lesser extent C5, and elevated levels of C3a-desArg, Ba, Bb, C5a-desArg, and fC5b-9. These trends are consistent with clinically observed biomarkers associated with complement-mediated diseases. Furthermore, we introduced therapy states by modeling known inhibitors of the complement system, a compstatin variant (C3 inhibitor) and eculizumab (C5 inhibitor). Compstatin demonstrates strong restorative effects for early-stage biomarkers, such as C3a-desArg, FB, Ba, and Bb, and milder restorative effects for late-stage biomarkers, such as C5a-desArg and fC5b-9, whereas eculizumab has strong restorative effects on late-stage biomarkers, and negligible effects on early-stage biomarkers. These results highlight the need for patient-tailored therapies that target early complement activation at the C3 level, or late-stage propagation of the terminal cascade at the C5 level, depending on the specific FH-mediated disease and the manifestations of a patient's genetic profile in complement regulatory function.


Assuntos
Via Alternativa do Complemento/fisiologia , Via Clássica do Complemento/fisiologia , Proteínas do Sistema Complemento/metabolismo , Modelos Biológicos , Síndrome Hemolítico-Urêmica Atípica/diagnóstico , Síndrome Hemolítico-Urêmica Atípica/imunologia , Biomarcadores/análise , Biomarcadores/metabolismo , Inativadores do Complemento/farmacologia , Inativadores do Complemento/uso terapêutico , Via Alternativa do Complemento/efeitos dos fármacos , Via Clássica do Complemento/efeitos dos fármacos , Proteínas do Sistema Complemento/genética , Proteínas do Sistema Complemento/imunologia , Biologia Computacional , Glomerulonefrite Membranoproliferativa/diagnóstico , Glomerulonefrite Membranoproliferativa/imunologia , Homeostase/efeitos dos fármacos , Homeostase/fisiologia , Humanos , Imunidade Inata/efeitos dos fármacos , Imunidade Inata/fisiologia , Degeneração Macular/diagnóstico , Degeneração Macular/imunologia
8.
Mol Psychiatry ; 22(11): 1554-1561, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28761078

RESUMO

The pathogenesis of schizophrenia is considered to be multi-factorial, with likely gene-environment interactions (GEI). Genetic and environmental risk factors are being identified with increasing frequency, yet their very number vastly increases the scope of possible GEI, making it difficult to identify them with certainty. Accumulating evidence suggests a dysregulated complement pathway among the pathogenic processes of schizophrenia. The complement pathway mediates innate and acquired immunity, and its activation drives the removal of damaged cells, autoantigens and environmentally derived antigens. Abnormalities in complement functions occur in many infectious and autoimmune disorders that have been linked to schizophrenia. Many older reports indicate altered serum complement activity in schizophrenia, though the data are inconclusive. Compellingly, recent genome-wide association studies suggest repeat polymorphisms incorporating the complement 4A (C4A) and 4B (C4B) genes as risk factors for schizophrenia. The C4A/C4B genetic associations have re-ignited interest not only in inflammation-related models for schizophrenia pathogenesis, but also in neurodevelopmental theories, because rodent models indicate a role for complement proteins in synaptic pruning and neurodevelopment. Thus, the complement system could be used as one of the 'staging posts' for a variety of focused studies of schizophrenia pathogenesis. They include GEI studies of the C4A/C4B repeat polymorphisms in relation to inflammation-related or infectious processes, animal model studies and tests of hypotheses linked to autoimmune diseases that can co-segregate with schizophrenia. If they can be replicated, such studies would vastly improve our understanding of pathogenic processes in schizophrenia through GEI analyses and open new avenues for therapy.


Assuntos
Ativação do Complemento/imunologia , Esquizofrenia/etiologia , Esquizofrenia/imunologia , Animais , Encéfalo/imunologia , Ativação do Complemento/genética , Complemento C4a/genética , Complemento C4a/metabolismo , Complemento C4b/genética , Complemento C4b/metabolismo , Via Clássica do Complemento/imunologia , Via Clássica do Complemento/fisiologia , Interação Gene-Ambiente , Estudo de Associação Genômica Ampla/métodos , Humanos , Herança Multifatorial , Polimorfismo Genético/genética , Esquizofrenia/genética
9.
Proc Natl Acad Sci U S A ; 114(5): 986-991, 2017 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-28104818

RESUMO

The complement system is an important antimicrobial and inflammation-generating component of the innate immune system. The classical pathway of complement is activated upon binding of the 774-kDa C1 complex, consisting of the recognition molecule C1q and the tetrameric protease complex C1r2s2, to a variety of activators presenting specific molecular patterns such as IgG- and IgM-containing immune complexes. A canonical model entails a C1r2s2 with its serine protease domains tightly packed together in the center of C1 and an intricate intramolecular reaction mechanism for activation of C1r and C1s, induced upon C1 binding to the activator. Here, we show that the serine protease domains of C1r and C1s are located at the periphery of the C1r2s2 tetramer both when alone or within the nonactivated C1 complex. Our structural studies indicate that the C1 complex adopts a conformation incompatible with intramolecular activation of C1, suggesting instead that intermolecular proteolytic activation between neighboring C1 complexes bound to a complement activating surface occurs. Our results rationalize how a multitude of structurally unrelated molecular patterns can activate C1 and suggests a conserved mechanism for complement activation through the classical and the related lectin pathway.


Assuntos
Complemento C1r/química , Complemento C1s/química , Via Clássica do Complemento/fisiologia , Complemento C1r/genética , Complemento C1r/metabolismo , Complemento C1s/genética , Complemento C1s/metabolismo , Ativação Enzimática , Genes Sintéticos , Células HEK293 , Humanos , Imunidade Inata , Microscopia Eletrônica , Modelos Moleculares , Conformação Proteica , Proteínas Recombinantes/química , Espalhamento a Baixo Ângulo , Relação Estrutura-Atividade , Difração de Raios X
10.
Acta Neuropathol Commun ; 4: 23, 2016 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-26936605

RESUMO

INTRODUCTION: Guillain-Barré syndrome (GBS) is an autoimmune disease that results in acute paralysis through inflammatory attack on peripheral nerves, and currently has limited, non-specific treatment options. The pathogenesis of the acute motor axonal neuropathy (AMAN) variant is mediated by complement-fixing anti-ganglioside antibodies that directly bind and injure the axon at sites of vulnerability such as nodes of Ranvier and nerve terminals. Consequently, the complement cascade is an attractive target to reduce disease severity. Recently, C5 complement component inhibitors that block the formation of the membrane attack complex and subsequent downstream injury have been shown to be efficacious in an in vivo anti-GQ1b antibody-mediated mouse model of the GBS variant Miller Fisher syndrome (MFS). However, since gangliosides are widely expressed in neurons and glial cells, injury in this model was not targeted exclusively to the axon and there are currently no pure mouse models for AMAN. Additionally, C5 inhibition does not prevent the production of early complement fragments such as C3a and C3b that can be deleterious via their known role in immune cell and macrophage recruitment to sites of neuronal damage. RESULTS AND CONCLUSIONS: In this study, we first developed a new in vivo transgenic mouse model of AMAN using mice that express complex gangliosides exclusively in neurons, thereby enabling specific targeting of axons with anti-ganglioside antibodies. Secondly, we have evaluated the efficacy of a novel anti-C1q antibody (M1) that blocks initiation of the classical complement cascade, in both the newly developed anti-GM1 antibody-mediated AMAN model and our established MFS model in vivo. Anti-C1q monoclonal antibody treatment attenuated complement cascade activation and deposition, reduced immune cell recruitment and axonal injury, in both mouse models of GBS, along with improvement in respiratory function. These results demonstrate that neutralising C1q function attenuates injury with a consequent neuroprotective effect in acute GBS models and promises to be a useful new target for human therapy.


Assuntos
Complemento C1q/metabolismo , Via Clássica do Complemento/fisiologia , Gangliosídeos/metabolismo , Doenças do Sistema Nervoso Periférico/patologia , Animais , Anticorpos/farmacologia , Anticorpos/uso terapêutico , Complemento C1q/genética , Via Clássica do Complemento/genética , Diafragma/metabolismo , Diafragma/patologia , Transportadores de Ácidos Dicarboxílicos/genética , Modelos Animais de Doenças , Gangliosídeos/classificação , Gangliosídeos/imunologia , Síndrome de Guillain-Barré/metabolismo , Síndrome de Guillain-Barré/patologia , Humanos , Infiltração Leucêmica , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Atividade Motora/efeitos dos fármacos , Atividade Motora/genética , Doenças do Sistema Nervoso Periférico/tratamento farmacológico , Doenças do Sistema Nervoso Periférico/genética , Doenças do Sistema Nervoso Periférico/fisiopatologia , Receptores Nicotínicos/metabolismo , Respiração/efeitos dos fármacos , Respiração/genética , Especificidade da Espécie , Simportadores/genética , Volume de Ventilação Pulmonar/efeitos dos fármacos , Volume de Ventilação Pulmonar/genética
11.
Sci Rep ; 6: 23533, 2016 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-27020276

RESUMO

The transcription factor Runx1 has essential roles throughout hematopoiesis. Here, we demonstrate that Runx1 is critical for T cell maturation. Peripheral naïve CD4(+) T cells from CD4-cre Runx1 cKO mice are phenotypically and functionally immature as shown by decreased production of TNF-α upon TCR stimulation. The loss of peripheral CD4(+) T cells in CD4-cre Runx1 cKO mice is not due to defects in homeostasis or decreased expression of IL-7Rα, as transgenic expression of IL-7Rα does not rescue the loss of CD4(+) T cells. Rather, immature Runx1-deficient CD4(+) T cells are eliminated in the periphery by the activation and fixation of the classical complement pathway. In the thymus, there is a severe block in all aspects of intrathymic T cell maturation, although both positive and negative selection are unaltered. Thus, loss of Runx1 leads to the earliest characterized block in post-positive selection intrathymic maturation of CD4 T cells.


Assuntos
Linfócitos T CD4-Positivos/fisiologia , Diferenciação Celular/fisiologia , Subunidade alfa 2 de Fator de Ligação ao Core/fisiologia , Subpopulações de Linfócitos T/fisiologia , Animais , Linfócitos T CD4-Positivos/metabolismo , Diferenciação Celular/genética , Células Cultivadas , Via Clássica do Complemento/fisiologia , Proteínas do Sistema Complemento/metabolismo , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Citometria de Fluxo , Glicosilação , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Ácido N-Acetilneuramínico/metabolismo , Receptores de Interleucina-7/genética , Receptores de Interleucina-7/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Subpopulações de Linfócitos T/metabolismo , Timócitos/citologia , Timócitos/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
12.
Adv Exp Med Biol ; 772: 229-62, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24272362

RESUMO

Complement is a central part of the immune system that has developed as a first defense against non-self cells. Neoplastic transformation is accompanied by an increased capacity of the malignant cells to activate complement. In fact, clinical data demonstrate complement activation in cancer patients. On the basis of the use of protective mechanisms by malignant cells, complement activation has traditionally been considered part of the body's immunosurveillance against cancer. Inhibitory mechanisms of complement activation allow cancer cells to escape from complement-mediated elimination and hamper the clinical efficacy of monoclonal antibody-based cancer immunotherapies. To overcome this limitation, many strategies have been developed with the goal of improving complement-mediated effector mechanisms. However, significant work in recent years has identified new and surprising roles for complement activation within the tumor microenvironment. Recent reports suggest that complement elements can promote tumor growth in the context of chronic inflammation. This chapter reviews the data describing the role of complement activation in cancer immunity, which offers insights that may aid the development of more effective therapeutic approaches to control cancer.


Assuntos
Proliferação de Células , Proteínas do Sistema Complemento/fisiologia , Neoplasias/patologia , Animais , Ativação do Complemento , Via Alternativa do Complemento/fisiologia , Via Clássica do Complemento/fisiologia , Humanos , Imunidade/fisiologia , Imunoterapia , Neoplasias/imunologia , Neoplasias/terapia , Microambiente Tumoral/imunologia
13.
Behav Brain Res ; 250: 114-22, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23680162

RESUMO

The cerebellum plays an essential role in motor learning. Recently, orexins, the newfound lateral hypothalamic neuropeptides, have been found to excite Purkinje cells in the cerebellar cortex and neurons in the deep cerebellar nuclei (DCN). However, little is known about their roles in cerebellum-dependent motor learning. Therefore, the present study was designed to investigate the functional significance of hypothalamic orexinergic system during trace eyeblink conditioning, a tractable behavioral model system of cerebellum-dependent motor learning. It was revealed that the orexin 1 receptors (OXR1) were specifically localized on the soma of Purkinje cells and large DCN neurons. Furthermore, interfering with the endogenous orexins' effects on the cerebellum via the selective OXR1 antagonist SB-334867 disrupted the timing rather than the acquisition of trace conditioned eyeblink responses. In addition to the behavioral effects, the SB-334867 prevented the increase in peak amplitude of cerebellar theta oscillations with learning. These results suggest that the endogenous orexins may modulate motor learning via the activation of cerebellar OXR1.


Assuntos
Piscadela/fisiologia , Cerebelo/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neuropeptídeos/metabolismo , Ritmo Teta/fisiologia , Análise de Variância , Animais , Benzoxazóis/farmacologia , Piscadela/efeitos dos fármacos , Cerebelo/citologia , Cerebelo/efeitos dos fármacos , Via Clássica do Complemento/efeitos dos fármacos , Via Clássica do Complemento/fisiologia , Cobaias , Masculino , Microinjeções , Naftiridinas , Receptores de Orexina , Células de Purkinje/efeitos dos fármacos , Células de Purkinje/fisiologia , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores de Neuropeptídeos/antagonistas & inibidores , Ritmo Teta/efeitos dos fármacos , Fatores de Tempo , Ureia/análogos & derivados , Ureia/farmacologia
14.
Adv Exp Med Biol ; 735: 111-21, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23402022

RESUMO

When acute inflammatory states are induced by treatment with chemical mediators in C5-deficient mice, neutrophil influxes are commonly decreased. Therefore, the neutrophil C5a receptor (C5aR) is believed to be a member of the pro-inflammatory receptors. However, C5aR deficiency endows mouse neutrophils with increased sensitivity to Pseudomonas aeruginosa. We have demonstrated that C5aR accepts not only C5a but also ribosomal protein S19 (RP S19) oligomers. RP S19 oligomers released from apoptotic cells promote apoptosis or induce dual agonistic and antagonistic effects on the chemotaxis of macrophages and neutrophils in an autocrine or paracrine manner, respectively. We assumed that the function of C5aR in apoptotic cells is almost the same as that in neutrophils infiltrating acute inflammatory lesions. Therefore, we believe that RP S19 oligomers can explain the opposite response of neutrophils in C5aR-deficient mice. In the present study, we found that antihuman RP S19 rabbit IgG cross-reacted with mouse RP S19 monomers and oligomers in plasma and serum, respectively, whereas anti-human C5a rabbit IgG only cross-reacted with mouse RP S19 oligomers in serum. To examine a role of RP S19 oligomers in vivo, we injected carrageenan (50 microg/100 microL) into the thoracic cavities of mice in the simultaneous presence of rabbit IgG and antihuman C5a rabbit IgG (100 microg/100 microL). Before 4 h and after 24 h, we did not observe any inflammatory cues in pleural exudates and lung substances from control mice. However, infiltrating neutrophils were detected in pleural exudates and lung tissues at 4 h after the addition of anti-human RP S19 rabbit IgG. Moreover, anti-human C5a rabbit IgG retards the initiation phase of carrageenan-induced mouse plurality. Many of the neutrophils infiltrating the thoracic cavities of the mice remained annexin V-negative. Neutrophil infiltration into pneumonic lesions became more severe, as alveolar septal destruction and haemorrhage concomitant with increased numbers of neutrophils in the pleural exudates were observed. These in vivo data demonstrate that the neutrophil C5aR acts as a dual pro-inflammatory and pro-apoptosis receptor during the initiation and the resolution phases of acute inflammation, respectively.


Assuntos
Complemento C5a/fisiologia , Via Alternativa do Complemento/fisiologia , Sequência de Aminoácidos , Animais , Complemento C5a/deficiência , Complemento C5a/genética , Via Clássica do Complemento/fisiologia , Humanos , Camundongos , Camundongos Knockout , Dados de Sequência Molecular
15.
Infect Immun ; 80(10): 3669-78, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22802341

RESUMO

Neonatal meningitis Escherichia coli (NMEC) is the most common Gram-negative organism that is associated with neonatal meningitis, which usually develops as a result of hematogenous spread of the bacteria. There are two key pathogenesis processes for NMEC to penetrate into the brain, the essential step for the development of E. coli meningitis: a high-level bacteremia and traversal of the blood-brain barrier (BBB). Our previous study has shown that the bacterial outer membrane protein NlpI contributes to NMEC binding to and invasion of brain microvascular endothelial cells, the major component cells of the BBB, suggesting a role for NlpI in NMEC crossing of the BBB. In this study, we showed that NlpI is involved in inducing a high level of bacteremia. In addition, NlpI contributed to the recruitment of the complement regulator C4bp to the surface of NMEC to evade serum killing, which is mediated by the classical complement pathway. NlpI may be involved in the interaction between C4bp and OmpA, which is an outer membrane protein that directly interacts with C4bp on the bacterial surface. The involvement of NlpI in two key pathogenesis processes of NMEC meningitis may make this bacterial factor a potential target for prevention and therapy of E. coli meningitis.


Assuntos
Bacteriemia/microbiologia , Via Clássica do Complemento/fisiologia , Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Antígenos de Histocompatibilidade/metabolismo , Lipoproteínas/metabolismo , Animais , Animais Recém-Nascidos , Bacteriemia/sangue , Bacteriemia/imunologia , Complemento C3b , Via Clássica do Complemento/imunologia , Modelos Animais de Doenças , Escherichia coli/genética , Infecções por Escherichia coli/imunologia , Regulação Bacteriana da Expressão Gênica/fisiologia , Humanos , Camundongos , Mutação , Neutrófilos/imunologia , Distribuição Aleatória , Soro , Fatores de Tempo
16.
Infect Immun ; 80(10): 3399-409, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22825444

RESUMO

Escherichia coli is a common Gram-negative organism that causes bacteremia. Prc, a bacterial periplasmic protease, and its homologues are known to be involved in the pathogenesis of Gram-negative bacterial infections. The present study examined the role of Prc in E. coli bacteremia and characterized the ability of the prc mutant of the pathogenic E. coli strain RS218 to cause bacteremia and survive in human serum. The prc mutant of RS218 exhibited a decreased ability to cause a high level of bacteremia and was more sensitive to serum killing than strain RS218. This sensitivity was due to the mutant's decreased ability to avoid the activation of the antibody-dependent and -independent classical complement cascades as well as its decreased resistance to killing mediated by the membrane attack complex, the end product of complement system activation. The demonstration of Prc in the evasion of classical complement-mediated serum killing of pathogenic E. coli makes this factor a potential target for the development of therapeutic and preventive measures against E. coli bacteremia.


Assuntos
Bacteriemia/microbiologia , Proteínas do Sistema Complemento/metabolismo , Endopeptidases/metabolismo , Infecções por Escherichia coli/microbiologia , Escherichia coli/enzimologia , Animais , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/metabolismo , Via Clássica do Complemento/fisiologia , Endopeptidases/genética , Escherichia coli/genética , Regulação Bacteriana da Expressão Gênica/fisiologia , Regulação Enzimológica da Expressão Gênica/fisiologia , Humanos , Imunoglobulina G/sangue , Camundongos , Camundongos Endogâmicos BALB C , Mutação , Coelhos
17.
Innate Immun ; 18(2): 350-63, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21450789

RESUMO

C1q is a versatile innate immune molecule that serves as the initiation subcomponent of the classical complement pathway. In addition, it is also a potent pattern recognition molecule, the versatility of which has fuelled its functional flexibility. C1q recognises an array of self, non-self and altered-self ligands. The broad-spectrum ligand-binding potential of C1q is facilitated by the modular organisation of the heterotrimeric globular head region, its ability to change its conformation in a very subtle way, and the manner in which this ancient molecule appears to have evolved to deal with the different types of ligands. Over recent years, molecules that resemble C1q have been put together to form the C1q family. In this review, we briefly summarise complement-dependent and complement-independent functions of C1q, its cognate receptors and key members of the rapidly growing C1q family.


Assuntos
Ativação do Complemento/fisiologia , Complemento C1q/fisiologia , Imunidade Inata/fisiologia , Animais , Via Clássica do Complemento/fisiologia , Humanos , Glicoproteínas de Membrana/fisiologia , Receptores de Complemento/fisiologia
18.
Clin Exp Rheumatol ; 29(6): 1049-56, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22153664

RESUMO

OBJECTIVES: Immune complexes (ICs) from sera of juvenile idiopathic arthritis (JIA) patients show increased complement opsonisation; however, a definitive role for involvement of the classical or alternative pathway is not entirely clear. To delineate the role of these pathways, we measured activated complement products bound to circulating IC (CICs) in the sera of JIA patients. METHODS: Sera from 100 JIA patients and 22 healthy children were collected. C1q, C4, C3, C3d, and membrane attack complex (MAC) bound to CICs were measured by enzyme-linked immunosorbent assay. Data was compared to IgM rheumatoid factor (RF), IgG anti-cyclic citrullinated peptide (CCP) antibodies, C-reactive protein (CRP), and erythrocyte sedimentation rate (ESR) levels. RESULTS: Mean levels of C1q, C4, and MAC bound to CICs were significantly elevated in JIA patients compared to healthy children. C1q correlated significantly with C4 and MAC bound to CICs and C4 and MAC also demonstrated significant correlation. No significant differences were noted in complement components bound to CICs when evaluating IgM RF, anti-CCP antibody, and CRP positivity. A significant correlation was noted between MAC bound to CICs and ESR. C1q and MAC bound to CICs mean levels were significantly higher in patients with an elevated ESR compared to those with a normal ESR level. CONCLUSIONS: JIA patients have elevated levels of complement components bound to CICs, particularly from the classical pathway. Moreover, classical pathway components were associated with ESR, a marker of disease activity. MAC bound to CICs also correlated significantly with ESR, further supporting the notion of complement-mediated tissue injury that is triggered by IC-mediated classical pathway activation.


Assuntos
Complexo Antígeno-Anticorpo/imunologia , Artrite Juvenil/imunologia , Ativação do Complemento , Complemento C1q/imunologia , Complemento C4/imunologia , Via Clássica do Complemento/fisiologia , Complexo Antígeno-Anticorpo/sangue , Artrite/sangue , Artrite/imunologia , Artrite/patologia , Artrite Juvenil/sangue , Artrite Juvenil/patologia , Criança , Complemento C1q/análise , Complemento C4/análise , Feminino , Humanos , Masculino , Ligação Proteica
19.
FASEB J ; 25(12): 4198-210, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21856781

RESUMO

The acute-phase protein C-reactive protein (CRP) recruits C1q to the surface of damaged cells and thereby initiates complement activation. However, CRP also recruits complement inhibitors, such as C4b-binding protein (C4bp) and factor H, which both block complement progression at the level of C3 and inhibits inflammation. To define how CRP modulates the classic complement pathway, we studied the interaction of CRP with the classic pathway inhibitor C4bp. Monomeric CRP (mCRP), but not pentameric CRP (pCRP), binds C4bp and enhances degradation of C4b and C3b. Both C1q, the initiator, and C4bp, the inhibitor of the classic pathway, compete for mCRP binding, and this competition adjusts the local balance of activation and inhibition. After attachment of pCRP to the surface of necrotic rat myocytes, generation of mCRP was demonstrated over a period of 18 h. Similarly, a biological role for mCRP, C1q, and C4bp in the disease setting of acute myocardial infarction was revealed. In this inflamed tissue, mCRP, pCRP, C4bp, C1q, and C4d were detected in acetone-fixed and in unfixed tissue. Protein levels were enhanced 6 h to 5 d after infarction. Thus, mCRP bound to damaged cardiomyocytes recruits C1q to activate and also C4bp to control the classic complement pathway.


Assuntos
Proteína C-Reativa/metabolismo , Via Clássica do Complemento/fisiologia , Animais , Proteína C-Reativa/química , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Células Cultivadas , Complemento C1q/metabolismo , Proteína de Ligação ao Complemento C4b , Antígenos de Histocompatibilidade/metabolismo , Humanos , Células Jurkat , Infarto do Miocárdio/imunologia , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Miócitos Cardíacos/imunologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Necrose , Ligação Proteica , Conformação Proteica , Estrutura Quaternária de Proteína , Ratos , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Ressonância de Plasmônio de Superfície
20.
J Biol Chem ; 286(18): 16459-69, 2011 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-21454703

RESUMO

C1q is the recognition subunit of the first component of the classical complement pathway. It participates in clearance of immune complexes and apoptotic cells as well as in defense against pathogens. Inappropriate activation of the complement contributes to cellular and tissue damage in different pathologies, urging the need for the development of therapeutic agents that are able to inhibit the complement system. In this study, we report heme as an inhibitor of C1q. Exposure of C1q to heme significantly reduced the activation of the classical complement pathway, mediated by C-reactive protein (CRP) and IgG. Interaction analyses revealed that heme reduces the binding of C1q to CRP and IgG. Furthermore, we demonstrated that the inhibition of C1q interactions results from a direct binding of heme to C1q. Formation of complex of heme with C1q caused changes in the mechanism of recognition of IgG and CRP. Taken together, our data suggest that heme is a natural negative regulator of the classical complement pathway at the level of C1q. Heme may play a role at sites of excessive tissue damage and hemolysis where large amounts of free heme are released.


Assuntos
Complemento C1q/metabolismo , Via Clássica do Complemento/fisiologia , Heme/metabolismo , Proteína C-Reativa/química , Proteína C-Reativa/metabolismo , Complemento C1q/antagonistas & inibidores , Complemento C1q/química , Heme/química , Hemólise/fisiologia , Humanos , Imunoglobulina G/química , Imunoglobulina G/metabolismo , Ligação Proteica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...