Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 178
Filtrar
1.
PLoS Pathog ; 17(7): e1009381, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34197564

RESUMO

Clearance of viral infections, such as SARS-CoV-2 and influenza A virus (IAV), must be fine-tuned to eliminate the pathogen without causing immunopathology. As such, an aggressive initial innate immune response favors the host in contrast to a detrimental prolonged inflammation. The complement pathway bridges innate and adaptive immune system and contributes to the response by directly clearing pathogens or infected cells, as well as recruiting proinflammatory immune cells and regulating inflammation. However, the impact of modulating complement activation in viral infections is still unclear. In this work, we targeted the complement decay-accelerating factor (DAF/CD55), a surface protein that protects cells from non-specific complement attack, and analyzed its role in IAV infections. We found that DAF modulates IAV infection in vivo, via an interplay with the antigenic viral proteins hemagglutinin (HA) and neuraminidase (NA), in a strain specific manner. Our results reveal that, contrary to what could be expected, DAF potentiates complement activation, increasing the recruitment of neutrophils, monocytes and T cells. We also show that viral NA acts on the heavily sialylated DAF and propose that the NA-dependent DAF removal of sialic acids exacerbates complement activation, leading to lung immunopathology. Remarkably, this mechanism has no impact on viral loads, but rather on the host resilience to infection, and may have direct implications in zoonotic influenza transmissions.


Assuntos
Antígenos CD55/fisiologia , Vírus da Influenza A Subtipo H1N1/isolamento & purificação , Pulmão/imunologia , Viremia/imunologia , Animais , Líquido da Lavagem Broncoalveolar/imunologia , Antígenos CD55/química , Antígenos CD55/deficiência , Quimiotaxia de Leucócito , Ativação do Complemento , Glicoproteínas de Hemaglutininação de Vírus da Influenza/fisiologia , Adaptação ao Hospedeiro , Especificidade de Hospedeiro , Interações Hospedeiro-Patógeno , Vírus da Influenza A Subtipo H1N1/enzimologia , Vírus da Influenza A Subtipo H1N1/patogenicidade , Vírus da Influenza A Subtipo H1N1/fisiologia , Interferon gama/análise , Pulmão/patologia , Pulmão/virologia , Camundongos , Camundongos Endogâmicos C57BL , Ácido N-Acetilneuramínico , Neuraminidase/fisiologia , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/patologia , Carga Viral , Proteínas Virais/fisiologia , Virulência , Replicação Viral , Redução de Peso
2.
Front Immunol ; 10: 1074, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31164885

RESUMO

It has long been understood that the control and surveillance of tumors within the body involves an intricate dance between the adaptive and innate immune systems. At the center of the interplay between the adaptive and innate immune response sits the complement system-an evolutionarily ancient response that aids in the destruction of microorganisms and damaged cells, including cancer cells. Membrane-bound complement regulatory proteins (mCRPs), such as CD46, CD55, and CD59, are expressed throughout the body in order to prevent over-activation of the complement system. These mCRPs act as a double-edged sword however, as they can also over-regulate the complement system to the extent that it is no longer effective at eliminating cancerous cells. Recent studies are now indicating that mCRPs may function as a biomarker of a malignant transformation in numerous cancer types, and further, are being shown to interfere with anti-tumor treatments. This highlights the critical roles that therapeutic blockade of mCRPs can play in cancer treatment. Furthermore, with the complement system having the ability to both directly and indirectly control adaptive T-cell responses, the use of a combinatorial approach of complement-related therapy along with other T-cell activating therapies becomes a logical approach to treatment. This review will highlight the biomarker-related role that mCRP expression may have in the classification of tumor phenotype and predicted response to different anti-cancer treatments in the context of an emerging understanding that complement activation within the Tumor Microenvironment (TME) is actually harmful for tumor control. We will discuss what is known about complement activation and mCRPs relating to cancer and immunotherapy, and will examine the potential for combinatorial approaches of anti-mCRP therapy with other anti-tumor therapies, especially checkpoint inhibitors such as anti PD-1 and PD-L1 monoclonal antibodies (mAbs). Overall, mCRPs play an essential role in the immune response to tumors, and understanding their role in the immune response, particularly in modulating currently used cancer therapeutics may lead to better clinical outcomes in patients with diverse cancer types.


Assuntos
Antígenos CD55/fisiologia , Antígenos CD59/fisiologia , Imunoterapia/métodos , Proteína Cofatora de Membrana/fisiologia , Neoplasias/imunologia , Proteínas do Sistema Complemento/fisiologia , Humanos , Neoplasias/etiologia , Neoplasias/terapia , Receptores de Complemento 3b/fisiologia
3.
J Exp Med ; 214(9): 2715-2732, 2017 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-28838952

RESUMO

Effective targeting of cancer stem cells (CSCs) requires neutralization of self-renewal and chemoresistance, but these phenotypes are often regulated by distinct molecular mechanisms. Here we report the ability to target both of these phenotypes via CD55, an intrinsic cell surface complement inhibitor, which was identified in a comparative analysis between CSCs and non-CSCs in endometrioid cancer models. In this context, CD55 functions in a complement-independent manner and required lipid raft localization for CSC maintenance and cisplatin resistance. CD55 regulated self-renewal and core pluripotency genes via ROR2/JNK signaling and in parallel cisplatin resistance via lymphocyte-specific protein tyrosine kinase (LCK) signaling, which induced DNA repair genes. Targeting LCK signaling via saracatinib, an inhibitor currently undergoing clinical evaluation, sensitized chemoresistant cells to cisplatin. Collectively, our findings identify CD55 as a unique signaling node that drives self-renewal and therapeutic resistance through a bifurcating signaling axis and provides an opportunity to target both signaling pathways in endometrioid tumors.


Assuntos
Antineoplásicos/uso terapêutico , Antígenos CD55/fisiologia , Autorrenovação Celular/fisiologia , Cisplatino/uso terapêutico , Neoplasias do Endométrio/fisiopatologia , Animais , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Neoplasias do Endométrio/tratamento farmacológico , Feminino , Camundongos , Camundongos SCID , Transplante de Neoplasias , Células-Tronco Neoplásicas/fisiologia , Transdução de Sinais
4.
Exp Eye Res ; 138: 126-33, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26149093

RESUMO

CC chemokine ligand 2 (CCL2) recruits macrophages to reduce inflammatory responses. Decay-accelerating factor (DAF) is a membrane regulator of the classical and alternative pathways of complement activation. In view of the link between complement genes and retinal diseases, we evaluated the retinal phenotype of C57BL/6J mice and mice lacking Ccl2 and/or Daf1 at 12 months of age, using scanning laser ophthalmoscopic imaging, electroretinography (ERG), histology, immunohistochemistry, and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) analysis. In comparison to C57BL/6J mice, mutant mice had an increased number of autofluorescent foci, with the greatest number in the Ccl2(-/-)/Daf1(-/-) retina. ERG amplitudes in Ccl2(-/-)/Daf1(-/-), Ccl2(-/-) and Daf1(-/-) mice were reduced, with the greatest reduction in Ccl2(-/-)/Daf1(-/-) mice. TUNEL-positive cells were not seen in C57BL/6J retina, but were prevalent in the outer and inner nuclear layers of Ccl2(-/-)Daf1(-/-) mice and were present at reduced density in Ccl2(-/-) or Daf1(-/-) mice. Cell loss was most pronounced in the outer and inner nuclear layers of Ccl2(-/-)/Daf1(-/-) mice. The levels of the endoplasmic reticulum chaperone GPR78 and transcription factor ATF4 were significantly increased in the Ccl2(-/-)/Daf1(-/-) retina. In comparison to the C57BL/6J retina, the phosphorylation of NF-κB p65, p38, ERK and JNK was significantly upregulated while SIRT1 was significantly downregulated in the Ccl2(-/-)/Daf1(-/-) retina. Our results suggest that loss of Ccl2 and Daf1 causes retinal neuronal death and degeneration which is related to increased endoplasmic reticulum stress, oxidative stress and inflammation.


Assuntos
Antígenos CD55/fisiologia , Quimiocina CCL2/fisiologia , Degeneração Retiniana/etiologia , Degeneração Retiniana/fisiopatologia , Neurônios Retinianos/patologia , Fator 4 Ativador da Transcrição/metabolismo , Animais , Apoptose , Modelos Animais de Doenças , Eletrorretinografia , Chaperona BiP do Retículo Endoplasmático , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteínas de Choque Térmico/metabolismo , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Degeneração Retiniana/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
5.
J Mal Vasc ; 40(6): 384-90, 2015 Dec.
Artigo em Francês | MEDLINE | ID: mdl-26205796

RESUMO

Paroxysmal nocturnal hemoglobinuria (PNH) is a rare acquired disorder of hematopoietic stem cells. Somatic mutation in the phosphatidylinositol glycan class A (PIG-A), X-linked gene, is responsible for a deficiency in glycosphosphatidylinositol-anchored proteins (GPI-AP). The lack of one of the GPI-AP complement regulatory proteins (CD55, CD59) leads to hemolysis. The disease is diagnosed with hemolytic anemia, marrow failure and thrombosis. Thromboembolic complication occurs in 30% of patient after 10 years of follow-up and is the first event in one out of 10 patients. The two most common sites are hepatic and cerebral veins. These locations are correlated with high risk of death. Currently, these data are balanced with the use of a monoclonal antibody (Eculizumab), which has significantly improved the prognosis with a survival similar to general population after 36 months of follow-up. Anticoagulant treatment is recommended after a thromboembolic event but has no place in primary prophylaxis.


Assuntos
Hemoglobinúria Paroxística/sangue , Trombofilia/etiologia , Trombose/etiologia , Anticorpos Monoclonais Humanizados/uso terapêutico , Anticoagulantes/uso terapêutico , Transplante de Medula Óssea , Antígenos CD55/fisiologia , Antígenos CD59/fisiologia , Complexo de Ataque à Membrana do Sistema Complemento/antagonistas & inibidores , Complexo de Ataque à Membrana do Sistema Complemento/imunologia , GMP Cíclico/metabolismo , Gerenciamento Clínico , Endotélio Vascular/patologia , Feminino , Seguimentos , Glicosilfosfatidilinositóis/metabolismo , Hemoglobinúria Paroxística/complicações , Hemoglobinúria Paroxística/genética , Hemoglobinúria Paroxística/terapia , Humanos , Masculino , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Óxido Nítrico/metabolismo , Guias de Prática Clínica como Assunto , Trombofilia/tratamento farmacológico , Trombose/tratamento farmacológico , Trombose/prevenção & controle
6.
Biochem Biophys Res Commun ; 460(3): 518-24, 2015 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-25797618

RESUMO

CD55 is a glycosylphosphatidylinositol-anchored protein, which inhibits complement activation by acting on the complement C3 convertases. CD55 is widely localized in the cholesterol rich regions of the cell plasma membrane termed membrane rafts. CD55 is attached to these specialized regions via a GPI link on the outer leaflet of the plasma membrane. Membrane rafts anchor many important signaling proteins, which control several cellular functions within the cell. For example, we recently demonstrated that the membrane raft protein and complement inhibitor CD59 also controls insulin secretion by an intracellular mechanism. Therefore, we have in this study aimed at addressing the expression and function of CD55 in pancreatic beta cells. To this end, we observe that CD55 is highly expressed in INS1 832/13 beta cells as well as human pancreatic islets. Diabetic human islets show a tendency for increased expression of CD55 when compared to the healthy controls. Importantly, silencing of CD55 in INS1 832/13 cells does not affect their insulin secretory capacity. On the other hand, silencing of CD55 diminished the intensity of membrane rafts as determined by Atto-SM staining. We hence conclude that CD55 expression is affected by glycemic status in human islets and plays a critical role in maintaining the conserved structure of rafts in pancreatic islets, which is similar to that of the related complement inhibitor CD59. However CD55 does not interfere with insulin secretion in beta cells, which is in sharp contrast to the action of the complement inhibitor CD59.


Assuntos
Antígenos CD55/fisiologia , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Microdomínios da Membrana/metabolismo , Animais , Antígenos CD55/genética , Linhagem Celular , Perfilação da Expressão Gênica , Humanos , Secreção de Insulina , Ratos
7.
Eur J Cancer ; 50(12): 2152-61, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24915776

RESUMO

BACKGROUND: Membrane-bound complement restriction proteins (mCRPs) CD46, CD55 and CD59 enable tumour cells to evade complement dependent cytotoxicity and antibody-dependent killing mechanisms. But less is known about the role of these mCRPs in head and neck cancer. METHODS: In this study we determined the expression of the mCRPs on head and neck squamous cell carcinoma (HNSCC) cell lines, on tumour tissue and TDLNs (tumour-draining lymph nodes) as well as on lymphocytes from HNSCC patients. The influence of the HNSCC microenvironment on the mCRP regulation was analysed using Flow Cytometry, Western blotting and small interfering RNAs (siRNA) transfection studies. RESULTS: We examined the effects of the HNSCC tumour milieu on the expression levels of CD46, CD55 and CD59. We investigated the susceptibility of HNSCC cells to CDC (complement-dependent cytotoxicity) while silencing the mCRPs. Our results demonstrate a huge influence of the HNSCC tumour microenvironment on the regulation of mCRP expression and show a reciprocal regulation between the different mCRPs themselves. CONCLUSIONS: In summary, our data indicate that HNSCC has evolved different strategies to evade complement attacks and that the tumour microenvironment leads to the enhancement of complement resistance of the surrounding tissue.


Assuntos
Antígenos CD55/fisiologia , Antígenos CD59/fisiologia , Carcinoma de Células Escamosas/imunologia , Ativação do Complemento/fisiologia , Neoplasias de Cabeça e Pescoço/imunologia , Proteína Cofatora de Membrana/fisiologia , Neoplasias Bucais/imunologia , Microambiente Tumoral/fisiologia , Idoso , Idoso de 80 Anos ou mais , Linhagem Celular Tumoral , Feminino , Inativação Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Carcinoma de Células Escamosas de Cabeça e Pescoço , Microambiente Tumoral/imunologia
8.
Eur J Immunol ; 44(7): 2025-35, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24723363

RESUMO

A role for NKT cells has been implicated in sepsis, but the mechanism by which NKT cells contribute to sepsis remains unclear. Here, we examined WT and NKT-cell-deficient mice of C57BL/6 background during cecal ligation and puncture-induced sepsis. The levels of C5a, IFN-γ, and IL-10 were higher in the serum and peritoneal fluid of WT mice than in those of CD1d(-/-) mice, while the mortality rate was lower in CD1d(-/-) mice than in WT mice. C5a blockade decreased mortality of WT mice during sepsis, whereas it did not alter that of CD1d(-/-) mice. As assessed by intracellular staining, NKT cells expressed IFN-γ, while neutrophils expressed IL-10. Upon coculture, IL-10-deficient NKT cells enhanced IL-10 production by WT, but not IFN-γR-deficient, neutrophils. Meanwhile, CD1d(-/-) mice exhibited high CD55 expression on neutrophils during sepsis, whereas those cells from WT mice expressed minimal levels of CD55. Recombinant IL-10 administration into CD1d(-/-) mice reduced CD55 expression on neutrophils. Furthermore, adoptive transfer of sorted WT, but not IFN-γ-deficient, NKT cells into CD1d(-/-) mice suppressed CD55 expression on neutrophils, but increased IL-10 and C5a levels. Taken together, IFN-γ-producing NKT cells enhance C5a generation via IL-10-mediated inhibition of CD55 expression on neutrophils, thereby exacerbating sepsis.


Assuntos
Antígenos CD55/fisiologia , Complemento C5a/biossíntese , Interferon gama/fisiologia , Interleucina-10/fisiologia , Células T Matadoras Naturais/imunologia , Neutrófilos/imunologia , Sepse/imunologia , Animais , Antígenos CD1d/fisiologia , Complemento C5a/análise , Camundongos , Camundongos Endogâmicos C57BL
9.
Theriogenology ; 81(4): 613-24, 2014 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-24377861

RESUMO

The release of extracellular proteins is a part of the sperm capacitation process; this allows the sperm surface reorganization that enables the sperm to fertilize an oocyte. Some of the components released are 'decapacitation factors', an uncoordinated or early release of which may cause inappropriate surface destabilization and premature capacitation. We studied the involvement of glycosylphosphatidylinositol-anchored proteins (GPI-APs) in sperm capacitation, and reported that CD52 and CD55 exhibit bicarbonate-dependent release during in vitro sperm capacitation. Treating sperm with phosphatidylinositol-specific phospholipase C (PIPLC) resulted in the enzymatic cleavage of CD55, in both capacitating and noncapacitating conditions. Moreover, PIPLC treatment in noncapacitating conditions caused surface reorganization events that included exposure of the ganglioside GM1, aggregation of flotillin-1, and the swelling of the apical acrosome region; all of which have been reported to be associated with sperm capacitation. The acrosomal swelling was monitored using wet mount atomic force microscopy, a new imaging technique that allows nanometer-level sperm surface measurements in samples hydrated with physiological buffer rather than dried. Despite these surface changes, PIPLC treatment in identical incubation conditions did not stimulate hyperactive sperm motility or protein tyrosine phosphorylation (other hallmarks of sperm capacitation in vitro). In full capacitating conditions (i.e., the presence of bicarbonate and albumin), PIPLC treatment caused sperm deterioration. The possible role of GPI-APs removal from the sperm surface during sperm capacitation is discussed.


Assuntos
Antígenos CD/fisiologia , Antígenos de Neoplasias/fisiologia , Antígenos CD55/fisiologia , Gangliosídeos/fisiologia , Glicoproteínas/fisiologia , Capacitação Espermática/fisiologia , Espermatozoides/fisiologia , Suínos/fisiologia , Acrossomo/fisiologia , Animais , Antígeno CD52 , Feminino , Fertilização in vitro/veterinária , Immunoblotting/veterinária , Masculino , Microscopia de Força Atômica/veterinária , Motilidade dos Espermatozoides/fisiologia , Fosfolipases Tipo C/farmacologia
10.
Adv Exp Med Biol ; 790: 24-41, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23884584

RESUMO

The essential event in picornavirus entry is the delivery of the RNA genome to the cytoplasm of a target cell, where replication occurs. In the past several years progress has been made in understanding the structural changes in the virion important for uncoating and RNA release. In addition, for several viruses the endocytic mechanisms responsible for internalization have been identified, as have the cellular sites at which uncoating occurs. It has become clear that entry is not a passive process, and that viruses initiate specific signals required for entry. And we have begun to recognize that for a given virus, there may be multiple routes of entry, depending on the particular target cell and the receptors available on that cell.


Assuntos
Picornaviridae/fisiologia , Internalização do Vírus , Animais , Antígenos CD55/fisiologia , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus/fisiologia , Endocitose , Humanos , Picornaviridae/ultraestrutura , Receptores Virais/fisiologia , Ligação Viral
11.
Adv Exp Med Biol ; 735: 55-81, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23402019

RESUMO

For the last two decades, there had been remarkable advancement in understanding the role of complement regulatory proteins in autoimmune disorders and importance of complement inhibitors as therapeutics. Systemic lupus erythematosus is a prototype of systemic autoimmune disorders. The disease, though rare, is potentially fatal and afflicts women at their reproductive age. It is a complex disease with multiorgan involvement, and each patient presents with a different set of symptoms. The diagnosis is often difficult and is based on the diagnostic criteria set by the American Rheumatology Association. Presence of antinuclear antibodies and more specifically antidouble-stranded DNA indicates SLE. Since the disease is multifactorial and its phenotypes are highly heterogeneous, there is a need to identify multiple noninvasive biomarkers for SLE. Lack of validated biomarkers for SLE disease activity or response to treatment is a barrier to the efficient management of the disease, drug discovery, as well as development of new therapeutics. Recent studies with gene knockout mice have suggested that membrane-bound complement regulatory proteins (CRPs) may critically determine the sensitivity of host tissues to complement injury in autoimmune and inflammatory disorders. Case-controlled and followup studies carried out in our laboratory suggest an intimate relation between the level of DAF, MCP, CR1, and CD59 transcripts and the disease activity in SLE. Based on comparative evaluation of our data on these four membrane-bound complement regulatory proteins, we envisaged CR1 and MCP transcripts as putative noninvasive disease activity markers and the respective proteins as therapeutic targets for SLE. Following is a brief appraisal on membrane-bound complement regulatory proteins DAF, MCP, CR1, and CD59 as biomarkers and therapeutic targets for SLE.


Assuntos
Proteínas do Sistema Complemento/efeitos dos fármacos , Proteínas do Sistema Complemento/fisiologia , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Proteínas de Membrana/efeitos dos fármacos , Proteínas de Membrana/fisiologia , Animais , Biomarcadores , Antígenos CD55/fisiologia , Antígenos CD59/efeitos dos fármacos , Antígenos CD59/fisiologia , Humanos , Proteína Cofatora de Membrana/antagonistas & inibidores , Proteína Cofatora de Membrana/fisiologia , Receptores de Complemento/antagonistas & inibidores , Receptores de Complemento/fisiologia
12.
Adv Exp Med Biol ; 735: 83-96, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23402020

RESUMO

In this chapter, we present a concise historic prospective and a summary of accumulated knowledge on steroid hormones, DAF expression, and therapeutic implication of steroid hormone treatment on multiple pathologies, including infection and the host-pathogen interactions. DAF/CD55 plays multiple physiologic functions including tissue protection from the cytotoxic complement injury, an anti-inflammatory function due to its anti-adherence properties which enhance transmigration of monocytes and macrophages and reduce tissue injury. DAF physiologic functions are essential in many organ systems including pregnancy for protection of the semiallogeneic fetus or for preventing uncontrolled infiltration by white cells in their pro- and/or anti-inflammatory functions. DAF expression appears to have multiple regulatory tissue-specific and/or menstrual cycle-specific mechanisms, which involve complex signaling mechanisms. Regulation of DAF expression may involve a direct or an indirect effect of at least the estrogen, progesterone, and corticosteroid regulatory pathways. DAF is exploited in multiple pathologic conditions by pathogens and viruses in chronic tissue infection processes. The binding of Escherichia coli bearing Dr adhesins to the DAF/CD55 receptor is DAF density dependent and triggers internalization of E. coli via an endocytic pathway involving CD55, lipid rafts, and microtubules. Dr+ E. coli or Dr antigen may persist in vivo in the interstitium for several months. Further understanding of such processes should be instrumental in designing therapeutic strategies for multiple conditions involving DAF's protective or pathologic functions and tailoring host expression of DAF.


Assuntos
Antígenos CD55/fisiologia , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Esteroides/farmacologia , Esteroides/uso terapêutico , Adulto , Animais , Antígenos CD55/biossíntese , Antígenos CD55/efeitos dos fármacos , Proteínas do Sistema Complemento/imunologia , Feminino , Terapia de Reposição Hormonal , Humanos , Nefropatias/complicações , Óxido Nítrico/fisiologia , Trabalho de Parto Prematuro , Comunicação Parácrina/fisiologia , Gravidez , Progesterona/fisiologia , Esteroides/fisiologia
13.
J Virol ; 86(18): 9929-40, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22761385

RESUMO

Enveloped viruses can incorporate host cell membrane proteins during the budding process. Here we demonstrate that mumps virus (MuV) and vesicular stomatitis virus (VSV) assemble to include CD46 and CD55, two host cell regulators which inhibit propagation of complement pathways through distinct mechanisms. Using viruses which incorporated CD46 alone, CD55 alone, or both CD46 and CD55, we have tested the relative contribution of these regulators in resistance to complement-mediated neutralization. Virion-associated CD46 and CD55 were biologically active, with VSV showing higher levels of activity of both cofactors, which promoted factor I-mediated cleavage of C3b into iC3b as well as decay-accelerating factor (DAF) activity against the C3 convertase, than MuV. Time courses of in vitro neutralization with normal human serum (NHS) showed that both regulators could delay neutralization, but viruses containing CD46 alone were neutralized faster and more completely than viruses containing CD55 alone. A dominant inhibitory role for CD55 was most evident for VSV, where virus containing CD55 alone was not substantially different in neutralization kinetics from virus harboring both regulators. Electron microscopy showed that VSV neutralization proceeded through virion aggregation followed by lysis, with virion-associated CD55 providing a delay in both aggregation and lysis more substantial than that conferred by CD46. Our results demonstrate the functional significance of incorporation of host cell factors during virion envelope assembly. They also define pathways of virus complement-mediated neutralization and suggest the design of more effective viral vectors.


Assuntos
Antígenos CD55/fisiologia , Ativação do Complemento/fisiologia , Proteína Cofatora de Membrana/fisiologia , Vírus da Caxumba/imunologia , Vesiculovirus/imunologia , Animais , Antígenos CD55/genética , Células CHO , Ativação do Complemento/genética , Cricetinae , Cricetulus , Interações Hospedeiro-Patógeno/imunologia , Humanos , Proteína Cofatora de Membrana/genética , Microscopia Imunoeletrônica , Vírus da Caxumba/fisiologia , Vírus da Caxumba/ultraestrutura , Testes de Neutralização , Vesiculovirus/fisiologia , Vesiculovirus/ultraestrutura , Montagem de Vírus
14.
Exp Clin Transplant ; 10(1): 49-54, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22309420

RESUMO

OBJECTIVES: To analyze the protective effects against complement-mediated cytolysis of the MCP, DAF, and CD59 human complement regulatory proteins, alone and in combination, on NIH 3T3 mouse fibroblast cells. MATERIALS AND METHODS: We constructed 3 double and 3 single-human complement regulatory protein plasmids (pIRES-hMCP-hDAF, pIRES-hMCP-hCD59, pIRES-hDAF-hCD59, pIRES-A-hMCP, pIRES-B-hDAF, and pIRES-B-hCD59). The plasmids were transfected into NIH 3T3 cells, and stable transfectants were obtained by treatment with 200 kg/m3 G418 for 2 weeks. Normal human serum (50%) as a source of complement was added to the culture medium of stable transfectants. The 3-(4,5-dimethylthiazol-2-yl)- 2,5-diphenyltetrazolium bromide assay was used to analyze the protective ability of different human complement regulatory protein plasmids on complement-dependent cytolysis. RESULTS: The viability of double-human complement regulatory protein stable transfectants was significantly higher than that of single-human complement regulatory protein stable transfectants (P < .05). Among the double-transfectants, cells expressing pIRES-hMCP-hDAF and pIRES-hMCPhCD59 survived better than cells expressing pIREShDAF- hCD59 (91.75% ± 3.30% and 84.88% ± 2.36% vs 66.19% ± 6.52%; P < .05). Among the single transfectants, cells expressing pIRES-A-hMCP or pIRES-B-hDAF survived better than cells expressing pIRES-B-hCD59 or pIRES empty vector (53.76% ± 3.84% and 56.32% ± 2.83% vs 43.28% ± 0.96% and 40.27% ± 1.11%; P < .05). CONCLUSIONS: These results suggest that the MCP+DAF and MCP+CD59 combinations could be more effective than DAF+CD59 in protecting the NIH 3T3 cells from injury caused by complement-dependent cytolysis, whereas MCP or DAF alone is stronger than CD59 alone in inhibiting membrane attack complex formation.


Assuntos
Antígenos CD55/fisiologia , Antígenos CD59/fisiologia , Complexo de Ataque à Membrana do Sistema Complemento/fisiologia , Fibroblastos/fisiologia , Proteína Cofatora de Membrana/fisiologia , Animais , Antígenos CD55/genética , Antígenos CD59/genética , Sobrevivência Celular , Fibroblastos/citologia , Humanos , Proteína Cofatora de Membrana/genética , Camundongos , Modelos Animais , Células NIH 3T3 , Plasmídeos , Transfecção
15.
Transplantation ; 93(4): 390-7, 2012 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-22222784

RESUMO

BACKGROUND: Recent development of immunosuppressive therapy has provided a platform for clinical human leukocyte antigen (HLA)- and ABO-incompatible kidney transplantation. However, the prognosis seems to be different between the two. Accommodation, the condition of no injury even in the presence of antidonor antibody, is one of the key factors for successful transplantation with antidonor antibody. The purpose of this study was to compare signal transduction between anti-A/B and anti-HLA antibody reaction and to elucidate the mechanisms underlying accommodation. METHODS: Blood type A- or B-transferase gene was transfected into human EA.hy926 endothelial cells. After cell sorting, A- or B-expressing cells at high levels were obtained. The effects of anti-HLA and anti-A/B antibody binding on complement-mediated cytotoxicity and signal transduction were examined. RESULTS: Preincubation with anti-HLA antibodies only at low levels (<10% of saturation level) or anti-A/B antibodies at high levels (even at near saturation levels) for 24 hr resulted in resistance to complement-mediated cytotoxicity. Anti-A/B antibody ligation inactivated ERK1/2 pathway and increased complement regulatory proteins such as CD55 and CD59, whereas anti-HLA ligation activated PI3K/AKT pathway and increased cytoprotective genes such as hemeoxygenase-1 and ferritin H. CONCLUSION: Complement inhibition by upregulation of CD55 and CD59 through ERK1/2 inactivation might play a substantial role in accommodation after ABO-incompatible transplantation, which could also explain the intriguing finding of C4d deposition in the graft without rejection.


Assuntos
Sistema ABO de Grupos Sanguíneos/imunologia , Anticorpos Anti-Idiotípicos/imunologia , Células Endoteliais/fisiologia , Rejeição de Enxerto/prevenção & controle , Antígenos HLA/imunologia , Transplante de Órgãos/fisiologia , Transdução de Sinais/fisiologia , Anticorpos Anti-Idiotípicos/farmacologia , Apoferritinas/fisiologia , Antígenos CD55/fisiologia , Antígenos CD59/fisiologia , Células Cultivadas , Proteínas do Sistema Complemento/fisiologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/imunologia , Rejeição de Enxerto/imunologia , Heme Oxigenase-1/fisiologia , Humanos , Sistema de Sinalização das MAP Quinases/fisiologia , Proteína Oncogênica v-akt/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Transdução de Sinais/imunologia
17.
Am J Perinatol ; 28(7): 565-70, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21380985

RESUMO

Complement activation is thought to contribute to the pathogenesis of preterm labor (PTL). Decay-accelerating factor (DAF) is a natural complement pathway inhibitor. Our hypothesis was that DAF expression on maternal white blood cells (WBCs) in women with preterm labor is elevated compared with women with no preterm labor. Our secondary objective was to determine if differences in upregulation of DAF levels correlated with clinical outcomes. Serial blood samples were obtained from 30 patients with a clinical diagnosis of PTL and a control group of 30 pregnant individuals (same gestational age range) to determine DAF expression in peripheral WBCs in both groups. DAF expression was higher in women with PTL (less than 37 weeks) compared with the control group without PTL. Subjects with PTL who delivered before 34 weeks had less DAF expression and different kinetics of expression compared with those carrying pregnancies beyond 34 weeks. These data suggest that women with a clinical diagnosis of preterm labor have increased DAF expression on peripheral WBCs. Furthermore, it appears that failure to elevate DAF expression is associated with a risk of early premature delivery.


Assuntos
Antígenos CD55/fisiologia , Ativação do Complemento/fisiologia , Trabalho de Parto Prematuro/fisiopatologia , Adulto , Antígenos CD55/metabolismo , Feminino , Idade Gestacional , Humanos , Leucócitos/metabolismo , Trabalho de Parto Prematuro/metabolismo , Gravidez , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Regulação para Cima/fisiologia , Adulto Jovem
18.
J Cancer Res Clin Oncol ; 137(1): 81-7, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20221634

RESUMO

BACKGROUND: Decay-accelerating factor (DAF) is one of the key molecules involved in cell protection against autologous complement, which restricts the action of complement at critical stages of the cascade reaction. The effect of DAF on the survival of human cervical cancer cell (ME180) has not been demonstrated. METHODS: In this study we applied, for the first time, small interference RNA (siRNA) to knock down the expression of the DAF with the aim of exploiting complement more effectively for tumor cell damage. Meanwhile, we investigated the effects of DAF on the viability and migration, moreover the proliferation of ME180 cell. RESULTS: The results showed that the expression of DAF was significantly increased in human cervical cancer tissues. SiRNA inhibition of DAF expression enhanced complement-dependent cytolysis up to 32% in ME180 cells, which contributed to the control of C3 activation and increased the cells viability, migration and augment the number of ME180 cells. CONCLUSION: These data indicated that DAF siRNA described in this study may offer an additional alternative to improve the efficacy of antibody- and complement-based cancer immunotherapy.


Assuntos
Antígenos CD55/fisiologia , Neoplasias do Colo do Útero/metabolismo , Antígenos CD55/genética , Antígenos CD55/metabolismo , Movimento Celular , Proliferação de Células , Sobrevivência Celular/genética , Convertases de Complemento C3-C5/metabolismo , Regulação para Baixo , Feminino , Humanos , RNA Interferente Pequeno/metabolismo , Células Tumorais Cultivadas , Neoplasias do Colo do Útero/genética , Neoplasias do Colo do Útero/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...