Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 235
Filtrar
1.
Kidney Int ; 98(2): 314-322, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32461141

RESUMEN

The novel coronavirus disease COVID-19 originates in the lungs, but it may extend to other organs, causing, in severe cases, multiorgan damage, including cardiac injury and acute kidney injury. In severe cases, the presence of kidney injury is associated with increased risk of death, highlighting the relevance of this organ as a target of SARS-CoV-2 infection. COVID-19-associated tissue injury is not primarily mediated by viral infection, but rather is a result of the inflammatory host immune response, which drives hypercytokinemia and aggressive inflammation that affect lung parenchymal cells, diminishing oxygen uptake, but also endothelial cells, resulting in endotheliitis and thrombotic events and intravascular coagulation. The complement system represents the first response of the host immune system to SARS-CoV-2 infection, but there is growing evidence that unrestrained activation of complement induced by the virus in the lungs and other organs plays a major role in acute and chronic inflammation, endothelial cell dysfunction, thrombus formation, and intravascular coagulation, and ultimately contributes to multiple organ failure and death. In this review, we discuss the relative role of the different complement activation products in the pathogenesis of COVID-19-associated tissue inflammation and thrombosis and propose the hypothesis that blockade of the terminal complement pathway may represent a potential therapeutic option for the prevention and treatment of lung and multiorgan damage.


Asunto(s)
Betacoronavirus , Activación de Complemento , Infecciones por Coronavirus/inmunología , Neumonía Viral/inmunología , Animales , COVID-19 , Complemento C5a/fisiología , Infecciones por Coronavirus/complicaciones , Células Endoteliales/fisiología , Humanos , Inflamación/etiología , Lectina de Unión a Manosa/fisiología , Ratones , Pandemias , Neumonía Viral/complicaciones , SARS-CoV-2 , Trombosis/etiología , Enfermedades Vasculares/etiología
2.
Eur J Clin Invest ; 48(12): e13028, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30229880

RESUMEN

Critical illness is an aetiologically and clinically heterogeneous syndrome that is characterised by organ failure and immune dysfunction. Mortality in critically ill patients is driven by inflammation-associated organ damage and a profound vulnerability to nosocomial infection. Both factors are influenced by the activated complement protein C5a, released by unbridled activation of the complement system during critical illness. C5a exerts deleterious effects on organ systems directly and suppresses antimicrobial functions of key immune cells. Whilst several recent reports have added key knowledge of the cellular signalling pathways triggered by C5a, there remain a number of areas that are incompletely understood and therapeutic opportunities are still being evaluated. In this review, we summarise the cellular basis for C5a-induced vulnerability to nosocomial infection and organ dysfunction. We focus on cells of the innate immune system, highlighting the major areas in need of further research and potential avenues for targeted therapies.


Asunto(s)
Anafilatoxinas/fisiología , Complemento C5a/fisiología , Insuficiencia Multiorgánica/inmunología , Coagulación Sanguínea/inmunología , Plaquetas/inmunología , Sistema Cardiovascular/inmunología , Comunicación Celular/inmunología , Enfermedad Crítica , Endotelio Vascular/inmunología , Humanos , Enfermedades del Sistema Inmune/inmunología , Inmunidad Innata/inmunología , Receptor de Anafilatoxina C5a/fisiología
3.
Curr Hypertens Rep ; 19(11): 87, 2017 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-29046976

RESUMEN

PURPOSE OF REVIEW: Preeclampsia affects 3-4% of pregnancies with few treatment options to reduce maternal and fetal harm. Recent evidence that targeting the complement system may be an effective therapeutic strategy in prevention or treatment of preeclampsia will be reviewed. RECENT FINDINGS: Studies in humans confirm the safety and efficacy of C5 blockade in complement-mediated disorders of pregnancy, including preeclampsia. Animal models mimic the placental abnormalities and/or the maternal symptoms which characterize preeclampsia. These models in mouse and rat have defined a role for complement and its regulators in placental dysfunction, hypertension, proteinuria, endothelial dysfunction, fetal growth restriction, and angiogenic imbalance, thus informing future human studies. Targeting excessive complement activation, particularly the terminal complement complex (C5b-9) and C5a may be an effective strategy to prolong pregnancy in women with preeclampsia. Continued research is needed to identify the initiator(s) of activation, the pathways involved, and the key component(s) in the pathophysiology to allow development of safe and effective therapeutics to target complement without compromising its role in homeostasis and host defense.


Asunto(s)
Activación de Complemento/fisiología , Placenta/fisiopatología , Preeclampsia/fisiopatología , Animales , Complemento C5a/fisiología , Complejo de Ataque a Membrana del Sistema Complemento/fisiología , Modelos Animales de Enfermedad , Endotelio Vascular/fisiopatología , Femenino , Retardo del Crecimiento Fetal/fisiopatología , Homeostasis , Humanos , Hipertensión/fisiopatología , Ratones , Neovascularización Patológica/fisiopatología , Embarazo , Proteinuria/fisiopatología , Ratas
4.
Nephrol Dial Transplant ; 29(6): 1134-40, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24335382

RESUMEN

Ischaemia-reperfusion injury (IRI) is a frequent event in kidney transplantation, particularly when the kidney comes from a deceased donor. The brain death is usually associated with generalized ischaemia due to a hyperactivity of the sympathetic system. In spite of this, most donors have profound hypotension and require administration of vasoconstrictor agents. Warm ischaemia after kidney vessels clamping and the cold ischaemia after refrigeration also reduce oxygen and nutrients supply to tissues. The reperfusion further aggravates the state of oxidation and inflammation created by ischaemia. IRI first attacks endothelial cells and tubular epithelial cells. The lesions may be so severe that they lead to acute kidney injury (AKI) and delayed graft function (DGF), which can impair the graft survival. The unfavourable impact of DGF is worse when DGF is associated with acute rejection. Another consequence of IRI is the activation of the innate immunity. Danger signals released by dying cells alarm Toll-like receptors that, through adapter molecules and a chain of kinases, transmit the signal to transcription factors which encode the genes regulating inflammatory cells and mediators. In the inflammatory environment, dendritic cells (DCs) intercept the antigen, migrate to lymph nodes and present the antigen to immunocompetent cells, so activating the adaptive immunity and favouring rejection. Attempts to prevent IRI include optimal management of donor and recipient. Calcium-channel blockers, l-arginine and N-acetylcysteine could obtain a small reduction in the incidence of post-transplant DGF. Fenoldopam, Atrial Natriuretic Peptide, Brain Natriuretic Peptide and Dopamine proved to be helpful in reducing the risk of AKI in experimental models, but there is no controlled evidence that these agents may be of benefit in preventing DGF in kidney transplant recipients. Other antioxidants have been successfully used in experimental models of AKI but only a few studies of poor quality have been made in clinical transplantation with a few of these agents and we still lack of unambiguous demonstration that pre-treatment with these antioxidants can attenuate the impact of IRI in kidney transplantation. Interference with the signals leading to activation of innate immunity, inactivation of complement or manipulation of DCs is a promising therapeutic option for the near future.


Asunto(s)
Trasplante de Riñón , Daño por Reperfusión/patología , Acetilcisteína , Animales , Antioxidantes/uso terapéutico , Arginina , Isquemia Fría , Complemento C5a/fisiología , Funcionamiento Retardado del Injerto/inmunología , Funcionamiento Retardado del Injerto/fisiopatología , Células Dendríticas/fisiología , Supervivencia de Injerto/fisiología , Humanos , Inmunidad Innata/fisiología , Inflamación/patología , Riñón/inmunología , Riñón/fisiopatología , Trasplante de Riñón/efectos adversos , Masculino , Daño por Reperfusión/etiología , Daño por Reperfusión/inmunología , Daño por Reperfusión/fisiopatología , Donantes de Tejidos , Receptores Toll-Like/inmunología , Isquemia Tibia
5.
J Immunol ; 188(10): 5086-93, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22491257

RESUMEN

There is accumulating evidence that the complement activation product, C5a, can orchestrate cellular immune functions. IL-27(p28/EBI3) is an emerging key player essential for regulating inflammatory responses and T cells. In this article, we report that C5a robustly suppressed IL-27(p28) gene expression and release in peritoneal macrophages. These cells from C57BL/6J mice abundantly produced IL-27(p28) after engagement of either the TLR3 (polyinosinic-polycytidylic acid) or TLR4 (LPS) receptor. Genetic deficiency of either TLR4 or LBP completely incapacitated the ability of macrophages to secrete IL-27(p28) in response to LPS. IL-27(p28)-producing macrophages also expressed the C5aR receptor, thus displaying an IL-27(p28)(+)F4/80(+)C5aR(+) phenotype. C5a suppressed IL-27(p28) in LPS-stimulated macrophages via interactions with the C5aR receptor rather than the C5L2 receptor. After endotoxemia, C5aR(-/-) mice displayed higher plasma levels of IL-27(p28) compared with C57BL/6J mice. C5a did not affect the release of IL-27(p28) or the frequency of IL-27(p28)(+)F4/80(+) macrophages after engagement of TLR3. Mechanistically, LPS activated both the NF-κB and the PI3K/Akt pathways, whereas C5a activated only the PI3K/Akt pathway. Engagement of PI3K/Akt was inhibitory for IL-27(p28) production, because PI3K/Akt pharmacologic blockade resulted in increased amounts of IL-27(p28) and reversed the suppressive effects of C5a. Blockade of PI3K/Akt in endotoxemic C57BL/6J mice resulted in higher generation of IL-27(p28). In contrast, the PI3K/Akt pathway was not involved in TLR3-mediated release of IL-27(p28). These data provide new evidence about how complement activation may selectively interfere with production of T cell regulatory cytokines by APCs in the varying contexts of either bacterial (TLR4 pathway) or viral (TLR3 pathway) infection.


Asunto(s)
Activación de Complemento/inmunología , Complemento C5a/fisiología , Regulación hacia Abajo/inmunología , Interleucinas/antagonistas & inhibidores , Interleucinas/biosíntesis , Macrófagos Peritoneales/inmunología , Receptor Toll-Like 3/fisiología , Receptor Toll-Like 4/fisiología , Animales , Células Cultivadas , Interleucinas/metabolismo , Lipopolisacáridos/metabolismo , Macrófagos Peritoneales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Anafilatoxina C5a , Receptores de Quimiocina/antagonistas & inhibidores , Receptores de Quimiocina/fisiología , Receptor Toll-Like 3/biosíntesis , Receptor Toll-Like 4/antagonistas & inhibidores , Receptor Toll-Like 4/deficiencia
6.
J Immunol ; 189(6): 2985-94, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22914051

RESUMEN

Promoting complement (C) activation may enhance immunological mechanisms of anti-tumor Abs for tumor destruction. However, C activation components, such as C5a, trigger inflammation, which can promote tumor growth. We addressed the role of C5a on tumor growth by transfecting both human carcinoma and murine lymphoma with mouse C5a. In vitro growth kinetics of C5a, control vector, or parental cells revealed no significant differences. Tumor-bearing mice with C5a-transfected xenografted tumor cells had significantly less tumor burden as compared with control vector tumors. NK cells and macrophages infiltrated C5a-expressing tumors with significantly greater frequency, whereas vascular endothelial growth factor, arginase, and TNF-α production were significantly less. Tumor-bearing mice with high C5a-producing syngeneic lymphoma cells had significantly accelerated tumor progression with more Gr-1+CD11b+ myeloid cells in the spleen and overall decreased CD4+ and CD8+ T cells in the tumor, tumor-draining lymph nodes, and the spleen. In contrast, tumor-bearing mice with low C5a-producing lymphoma cells had a significantly reduced tumor burden with increased IFN-γ-producing CD4+ and CD8+ T cells in the spleen and tumor-draining lymph nodes. These studies suggest concentration of local C5a within the tumor microenvironment is critical in determining its role in tumor progression.


Asunto(s)
Complemento C5a/fisiología , Linfoma/inmunología , Linfoma/patología , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/patología , Microambiente Tumoral/inmunología , Animales , Carcinoma/genética , Carcinoma/inmunología , Carcinoma/patología , Línea Celular Tumoral , Movimiento Celular/genética , Movimiento Celular/inmunología , Técnicas de Cocultivo , Complemento C5a/genética , Complemento C5a/metabolismo , Progresión de la Enfermedad , Femenino , Humanos , Inmunidad Innata/genética , Leucocitos/inmunología , Leucocitos/metabolismo , Leucocitos/patología , Linfoma/genética , Ratones , Ratones SCID , Ratones Transgénicos , Neoplasias Ováricas/genética , Microambiente Tumoral/genética
7.
J Immunol ; 189(9): 4674-83, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-23028051

RESUMEN

The complement system contributes to various immune and inflammatory diseases, including cancer. In this study, we investigated the capacity of lung cancer cells to activate complement and characterized the consequences of complement activation on tumor progression. We focused our study on the production and role of the anaphylatoxin C5a, a potent immune mediator generated after complement activation. We first measured the capacity of lung cancer cell lines to deposit C5 and release C5a. C5 deposition, after incubation with normal human serum, was higher in lung cancer cell lines than in nonmalignant bronchial epithelial cells. Notably, lung malignant cells produced complement C5a even in the absence of serum. We also found a significant increase of C5a in plasma from patients with non-small cell lung cancer, suggesting that the local production of C5a is followed by its systemic diffusion. The contribution of C5a to lung cancer growth in vivo was evaluated in the Lewis lung cancer model. Syngeneic tumors of 3LL cells grew slower in mice treated with an antagonist of the C5a receptor. C5a did not modify 3LL cell proliferation in vitro but induced endothelial cell chemotaxis and blood-vessels formation. C5a also contributed to the immunosuppressive microenvironment required for tumor growth. In particular, blockade of C5a receptor significantly reduced myeloid-derived suppressor cells and immunomodulators ARG1, CTLA-4, IL-6, IL-10, LAG3, and PDL1 (B7H1). In conclusion, lung cancer cells have the capacity to generate C5a, a molecule that creates a favorable tumor microenvironment for lung cancer progression.


Asunto(s)
Carcinoma Pulmonar de Lewis/inmunología , Carcinoma Pulmonar de Lewis/patología , Complemento C5a/fisiología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Microambiente Tumoral/inmunología , Adenocarcinoma/sangre , Adenocarcinoma/inmunología , Adenocarcinoma/patología , Animales , Carcinoma Pulmonar de Lewis/prevención & control , Carcinoma de Pulmón de Células no Pequeñas/sangre , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Células Escamosas/sangre , Carcinoma de Células Escamosas/inmunología , Carcinoma de Células Escamosas/patología , Línea Celular Transformada , Línea Celular Tumoral , Activación de Complemento/genética , Activación de Complemento/inmunología , Complemento C5a/biosíntesis , Complemento C5a/genética , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Tolerancia Inmunológica/genética , Neoplasias Pulmonares/prevención & control , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica/inmunología , Neovascularización Patológica/patología , Neovascularización Patológica/terapia , Receptor de Anafilatoxina C5a/antagonistas & inhibidores , Receptor de Anafilatoxina C5a/fisiología , Mucosa Respiratoria/citología , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/metabolismo , Microambiente Tumoral/genética
8.
J Immunol ; 186(12): 6657-60, 2011 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-21572031

RESUMEN

Cerebral malaria is the most severe complication of Plasmodium falciparum infection and accounts for a large number of malaria fatalities worldwide. Recent studies demonstrated that C5(-/-) mice are resistant to experimental cerebral malaria (ECM) and suggested that protection was due to loss of C5a-induced inflammation. Surprisingly, we observed that C5aR(-/-) mice were fully susceptible to disease, indicating that C5a is not required for ECM. C3aR(-/-) and C3aR(-/-) × C5aR(-/-) mice were equally susceptible to ECM as were wild-type mice, indicating that neither complement anaphylatoxin receptor is critical for ECM development. In contrast, C9 deposition in the brains of mice with ECM suggested an important role for the terminal complement pathway. Treatment with anti-C9 Ab significantly increased survival time and reduced mortality in ECM. Our data indicate that protection from ECM in C5(-/-) mice is mediated through inhibition of membrane attack complex formation and not through C5a-induced inflammation.


Asunto(s)
Complejo de Ataque a Membrana del Sistema Complemento/fisiología , Malaria Cerebral/etiología , Animales , Encéfalo/inmunología , Complemento C5a/fisiología , Complemento C9/antagonistas & inhibidores , Complejo de Ataque a Membrana del Sistema Complemento/antagonistas & inhibidores , Susceptibilidad a Enfermedades , Inflamación , Malaria Cerebral/inmunología , Ratones , Ratones Noqueados , Receptor de Anafilatoxina C5a , Receptores de Complemento , Tasa de Supervivencia
9.
Br J Anaesth ; 111(5): 778-87, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23756248

RESUMEN

BACKGROUND: Nosocomial infection occurs commonly in intensive care units (ICUs). Although critical illness is associated with immune activation, the prevalence of nosocomial infections suggests concomitant immune suppression. This study examined the temporal occurrence of immune dysfunction across three immune cell types, and their relationship with the development of nosocomial infection. METHODS: A prospective observational cohort study was undertaken in a teaching hospital general ICU. Critically ill patients were recruited and underwent serial examination of immune status, namely percentage regulatory T-cells (Tregs), monocyte deactivation (by expression) and neutrophil dysfunction (by CD88 expression). The occurrence of nosocomial infection was determined using pre-defined, objective criteria. RESULTS: Ninety-six patients were recruited, of whom 95 had data available for analysis. Relative to healthy controls, percentage Tregs were elevated 6-10 days after admission, while monocyte HLA-DR and neutrophil CD88 showed broader depression across time points measured. Thirty-three patients (35%) developed nosocomial infection, and patients developing nosocomial infection showed significantly greater immune dysfunction by the measures used. Tregs and neutrophil dysfunction remained significantly predictive of infection in a Cox hazards model correcting for time effects and clinical confounders {hazard ratio (HR) 2.4 [95% confidence interval (CI) 1.1-5.4] and 6.9 (95% CI 1.6-30), respectively, P=0.001}. Cumulative immune dysfunction resulted in a progressive risk of infection, rising from no cases in patients with no dysfunction to 75% of patients with dysfunction of all three cell types (P=0.0004). CONCLUSIONS: Dysfunctions of T-cells, monocytes, and neutrophils predict acquisition of nosocomial infection, and combine additively to stratify risk of nosocomial infection in the critically ill.


Asunto(s)
Enfermedad Crítica/epidemiología , Infección Hospitalaria/epidemiología , Inmunidad Celular/fisiología , Adolescente , Adulto , Anciano , Recuento de Linfocito CD4 , Estudios de Cohortes , Complemento C5a/fisiología , Infección Hospitalaria/microbiología , Femenino , Antígenos HLA-DR/inmunología , Humanos , Masculino , Persona de Mediana Edad , Monocitos/inmunología , Neutrófilos/inmunología , Pronóstico , Estudios Prospectivos , Receptor de Anafilatoxina C5a/biosíntesis , Linfocitos T Reguladores/inmunología , Adulto Joven
10.
Adv Exp Med Biol ; 735: 111-21, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23402022

RESUMEN

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.


Asunto(s)
Complemento C5a/fisiología , Vía Alternativa del Complemento/fisiología , Secuencia de Aminoácidos , Animales , Complemento C5a/deficiencia , Complemento C5a/genética , Vía Clásica del Complemento/fisiología , Humanos , Ratones , Ratones Noqueados , Datos de Secuencia Molecular
11.
Adv Exp Med Biol ; 735: 197-206, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23402028

RESUMEN

Periodontitis is a prevalent oral chronic inflammatory disease which, in severe forms, may exert a major impact on systemic health. Clinical and histological observations, as well as experimental animal studies, suggest involvement of the complement system in periodontitis. However, the precise roles of the various complement components and pathways in periodontitis have only recently started to be elucidated. In this chapter, we review recent progress in the field and discuss the potential of complement-targeted therapeutics in the treatment of periodontitis.


Asunto(s)
Proteínas del Sistema Complemento/fisiología , Periodontitis/tratamiento farmacológico , Animales , Complemento C5a/fisiología , Humanos , Periodontitis/microbiología , Periodontitis/fisiopatología , Receptores de Complemento/fisiología
12.
Blood ; 116(4): 631-9, 2010 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-20424189

RESUMEN

Thrombosis is a common complication of end-stage renal disease, particularly in patients on hemodialysis. Although substantial progress has been made in preventing thrombotic complications in various other groups of patients, the mechanisms of thrombosis during hemodialysis require clarification. In this report, we demonstrate that complement activation triggered by hemodialysis biomaterials, and the subsequent generation of the complement anaphylatoxin C5a, results in the expression of functionally active tissue factor (TF) in peripheral blood neutrophils. Because TF is a key initiator of coagulation in vivo, we postulate that the recurring complement activation that occurs during long-term hemodialysis contributes to thrombosis in dialyzed end-stage renal disease patients. Furthermore, we found that complement contributed to the induction of granulocyte colony-stimulating factor, which has been implicated in the pathogenesis of thrombosis in patients treated with the recombinant form of this molecule. Importantly, the inhibition of complement activation attenuated the TF expression and granulocyte colony-stimulating factor induction in blood passing through a hemodialysis circuit, suggesting that the complement system could become a new therapeutic target for preventing thrombosis in patients with chronic renal failure who are maintained on hemodialysis.


Asunto(s)
Complemento C5a/fisiología , Diálisis Renal/efectos adversos , Trombosis/etiología , Anciano , Anafilatoxinas/farmacología , Anafilatoxinas/fisiología , Coagulación Sanguínea/efectos de los fármacos , Células Cultivadas , Activación de Complemento/efectos de los fármacos , Complemento C5a/metabolismo , Complemento C5a/farmacología , Femenino , Factor Estimulante de Colonias de Granulocitos/metabolismo , Humanos , Fallo Renal Crónico/sangre , Fallo Renal Crónico/complicaciones , Fallo Renal Crónico/metabolismo , Fallo Renal Crónico/terapia , Leucocitos/efectos de los fármacos , Leucocitos/metabolismo , Leucocitos/patología , Masculino , Persona de Mediana Edad , Receptor de Anafilatoxina C5a/metabolismo , Tromboplastina/metabolismo , Factores de Tiempo
13.
J Cell Biochem ; 112(9): 2594-605, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21598302

RESUMEN

There is a tight interaction of the bone and the immune system. However, little is known about the relevance of the complement system, an important part of innate immunity and a crucial trigger for inflammation. The aim of this study was, therefore, to investigate the presence and function of complement in bone cells including osteoblasts, mesenchymal stem cells (MSC), and osteoclasts. qRT-PCR and immunostaining revealed that the central complement receptors C3aR and C5aR, complement C3 and C5, and membrane-bound regulatory proteins CD46, CD55, and CD59 were expressed in human MSC, osteoblasts, and osteoclasts. Furthermore, osteoblasts and particularly osteoclasts were able to activate complement by cleaving C5 to its active form C5a as measured by ELISA. Both C3a and C5a alone were unable to trigger the release of inflammatory cytokines interleukin (IL)-6 and IL-8 from osteoblasts. However, co-stimulation with the pro-inflammatory cytokine IL-1ß significantly induced IL-6 and IL-8 expression as well as the expression of receptor activator of nuclear factor-kappaB ligand (RANKL) and osteoprotegerin (OPG) indicating that complement may modulate the inflammatory response of osteoblastic cells in a pro-inflammatory environment as well as osteoblast-osteoclast interaction. While C3a and C5a did not affect osteogenic differentiation, osteoclastogenesis was significantly induced even in the absence of RANKL and macrophage-colony stimulating factor (M-CSF) suggesting that complement could directly regulate osteoclast formation. It can therefore be proposed that complement may enhance the inflammatory response of osteoblasts and increase osteoclast formation, particularly in a pro-inflammatory environment, for example, during bone healing or in inflammatory bone disorders.


Asunto(s)
Complemento C3a/farmacología , Complemento C5a/farmacología , Interleucina-1beta/farmacología , Osteoblastos/fisiología , Osteoclastos/fisiología , Adulto , Antígenos CD/genética , Antígenos CD/metabolismo , Diferenciación Celular , Células Cultivadas , Complemento C3a/metabolismo , Complemento C3a/fisiología , Complemento C5a/metabolismo , Complemento C5a/fisiología , Expresión Génica , Humanos , Inflamación , Interleucina-1beta/fisiología , Masculino , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/fisiología , Microscopía Fluorescente , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteogénesis , Osteoprotegerina/metabolismo , Proteolisis , Ligando RANK/metabolismo , Receptores de Complemento/genética , Receptores de Complemento/metabolismo , Adulto Joven
14.
J Neurochem ; 119(5): 1041-51, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21929539

RESUMEN

Inflammation is a key factor in a number of neurodegenerative diseases including systemic lupus erythematosus. The complement system is an important mechanism in initiating and amplifying inflammation. Our recent studies demonstrate that C5a, a protein fragment generated during complement activation could alter the blood-brain barrier integrity, and thereby disturb the brain microenvironment. To understand the mechanism by which this occurs, we examined the effects of C5a on apoptosis, translocation of nuclear factor-κB (NF-κb) and the expression of Iκbα, MAPK, CREB and TJ protein, zona occludens (ZO-1) in mouse brain endothelial cells. Apoptosis was examined by DNA laddering and caspase 3 activity and the distribution of the ZO-1 and the p65 subunit of NF-κB were determined by immunofluorescence. Inhibition of CD88 reduced translocation of NF-κb into the nucleus, altered ZO-1 at the interfaces of neighboring cells, decreased caspase 3 activity and prevented apoptosis in these cells. Our results indicate that signaling through CD88 regulates the blood-brain barrier in a NF-κb-dependent manner. These studies suggest that the C5a receptor, CD88 is a promising therapeutic target that will reduce NF-κb-signaling cascades in inflammatory settings.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Complemento C5a/fisiología , Lupus Eritematoso Sistémico/metabolismo , Lupus Eritematoso Sistémico/patología , FN-kappa B/fisiología , Receptor de Anafilatoxina C5a/fisiología , Transducción de Señal/fisiología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Línea Celular Transformada , Células Endoteliales/metabolismo , Células Endoteliales/patología , Ratones , Ratones Endogámicos C57BL , FN-kappa B/antagonistas & inhibidores , Cultivo Primario de Células
15.
Eur J Immunol ; 40(5): 1355-68, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20213734

RESUMEN

In visceral leishmaniasis, the draining LN (DLN) is the initial site for colonization and establishment of infection after intradermal transmission by the sand fly vector; however, little is known about the developing immune response within this site. Using an intradermal infection model, which allows for parasite visceralization, we have examined the ongoing immune responses in the DLN of BALB/c mice infected with Leishmania infantum. Although not unexpected, at early times post-infection there is a marked B-cell expansion in the DLN, which persists throughout infection. However, the characteristics of this response were of interest; as early as day 7 post-infection, polyclonal antibodies (TNP, OVA, chromatin) were observed and the levels appeared comparable to the specific anti-leishmania response. Although B-cell-deficient JhD BALB/c mice are relatively resistant to infection, neither B-cell-derived IL-10 nor B-cell antigen presentation appear to be primarily responsible for the elevated parasitemia. However, passive transfer and reconstitution of JhD BALB/c with secretory immunoglobulins, (IgM or IgG; specific or non-specific immune complexes) results in increased susceptibility to L. infantum infection. Further, JhD BALB/c mice transgenetically reconstituted to secrete IgM demonstrated exacerbated disease in comparison to WT BALB/c mice as early as 2 days post-infection. Evidence suggests that complement activation (generation of C5a) and signaling via the C5a receptor (CD88) is related to the disease exacerbation caused by IgM rather than cytokine levels (IL-10 or IFN-gamma). Overall these studies indicate that polyclonal B-cell activation, which is known to be associated with human visceral leishmaniasis, is an early and intrinsic characteristic of disease and may represent a target for therapeutic intervention.


Asunto(s)
Anticuerpos Antiprotozoarios/inmunología , Linfocitos B/inmunología , Inmunoglobulina M/inmunología , Leishmaniasis Visceral/inmunología , Parasitemia/inmunología , Animales , Presentación de Antígeno , Complemento C5a/antagonistas & inhibidores , Complemento C5a/fisiología , Progresión de la Enfermedad , Femenino , Hipergammaglobulinemia/etiología , Hipergammaglobulinemia/inmunología , Inmunidad Innata , Inmunización Pasiva , Inmunoglobulina G/inmunología , Interleucina-10/fisiología , Leishmania infantum/crecimiento & desarrollo , Leishmania infantum/inmunología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/parasitología , Ganglios Linfáticos/patología , Activación de Linfocitos , Depleción Linfocítica , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos
16.
Eur J Immunol ; 40(4): 1079-88, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20140904

RESUMEN

Complement 5a (C5a) and Interleukin-17 (IL-17) are two important inflammatory mediators in sepsis. Here we studied the mechanisms underlying regulation of IL-17 by anaphylatoxin C5a. We found that C5a blockade increased the survival rate of mice following cecal ligation and puncture (CLP)-induced sepsis and decreased IL-17 expression in vivo. IL-17 was secreted mainly by gammadelta T cells in this model. Importantly, our data suggest that C5a participates in the regulation of IL-17 secretion by gammadelta T cells. Dendritic cells (DC) were found to act as a "bridge" between C5a and gammadelta T cells in a mechanism involving IL-6 and transforming growth factor beta (TGF-beta). These results imply that C5a affects the crosstalk between DC and gammadelta T cells during sepsis development, and this may result in a large production of inflammatory mediators such as IL-17.


Asunto(s)
Comunicación Celular/fisiología , Complemento C5a/fisiología , Células Dendríticas/inmunología , Interleucina-17/biosíntesis , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Sepsis/inmunología , Subgrupos de Linfocitos T/inmunología , Traslado Adoptivo , Animales , Ciego/lesiones , Técnicas de Cocultivo , Complemento C5a/farmacología , Células Dendríticas/efectos de los fármacos , Interleucina-17/genética , Interleucina-6/biosíntesis , Perforación Intestinal/complicaciones , Linfotoxina-alfa/biosíntesis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Recombinantes/farmacología , Sepsis/etiología , Organismos Libres de Patógenos Específicos , Subgrupos de Linfocitos T/trasplante
17.
Blood ; 114(5): 1005-15, 2009 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-19491392

RESUMEN

Toll-like receptors (TLRs) and complement are 2 major components of innate immunity that provide a first-line host defense and shape the adaptive immune responses. We show here that coincidental activation of complement and several TLRs in mice led to the synergistic production of serum factors that promoted T-helper cell 17 (Th17) differentiation from anti-CD3/CD28 or antigen-stimulated T cells. Although multiple TLR-triggered cytokines were regulated by complement, Th17 cell-promoting activity in the serum was correlated with interleukin (IL)-6 induction, and antibody neutralization of IL-6 abrogated the complement effect. By using both in vitro and in vivo approaches, we examined in more detail the mechanism and physiologic implication of complement/TLR4 interaction on Th17-cell differentiation. We found that the complement effect required C5a receptor, was evident at physiologically relevant levels of C5a, and could be demonstrated in cultured peritoneal macrophages as well as in the setting of antigen immunization. Importantly, despite an inhibitory effect of complement on IL-23 production, complement-promoted Th17 cells were functionally competent in causing autoimmunity in an adoptive transfer model of experimental autoimmune encephalomyelitis. Collectively, these data establish a link between complement/TLR interaction and Th17-cell differentiation and provide new insight into the mechanism of action of complement in autoimmunity.


Asunto(s)
Activación de Complemento , Inflamación/inmunología , Interleucina-6/fisiología , Linfocitos T Colaboradores-Inductores/citología , Receptores Toll-Like/fisiología , Traslado Adoptivo , Animales , Antígenos CD55/genética , Células Cultivadas/citología , Células Cultivadas/metabolismo , Complemento C5a/fisiología , Complemento C5a/uso terapéutico , Citocinas/sangre , Citocinas/fisiología , Encefalomielitis Autoinmune Experimental/etiología , Encefalomielitis Autoinmune Experimental/inmunología , Inmunidad Innata , Inmunización , Inflamación/inducido químicamente , Interleucina-6/biosíntesis , Interleucina-6/sangre , Interleucina-6/deficiencia , Interleucina-6/genética , Linfopoyesis , Macrófagos Peritoneales/citología , Macrófagos Peritoneales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Anafilatoxina C5a/deficiencia , Receptor de Anafilatoxina C5a/fisiología , Proteínas Recombinantes/uso terapéutico , Organismos Libres de Patógenos Específicos , Linfocitos T Colaboradores-Inductores/clasificación , Linfocitos T Colaboradores-Inductores/inmunología , Receptor Toll-Like 4/fisiología
18.
J Immunol ; 182(6): 3827-36, 2009 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-19265162

RESUMEN

Mesenchymal stem cells (MSCs) have a great potential for tissue repair, especially if they can be delivered efficiently to sites of tissue injury. Since complement activation occurs whenever there is tissue damage, the effects of the complement activation products C3a and C5a on MSCs were examined. Both C3a and C5a were chemoattractants for human bone marrow-derived MSCs, which expressed both the C3a receptor (C3aR) and the C5a receptor (C5aR; CD88) on the cell surface. Specific C3aR and C5aR inhibitors blocked the chemotactic response, as did pertussis toxin, indicating that the response was mediated by the known anaphylatoxin receptors in a G(i) activation-dependent fashion. While C5a causes strong and prolonged activation of various signaling pathways in many different cell types, the response observed with C3a is generally transient and weak. However, we show herein that in MSCs both C3a and C5a caused prolonged and robust ERK1/2 and Akt phosphorylation. Phospho-ERK1/2 was translocated to the nucleus in both C3a and C5a-stimulated MSCs, which was associated with subsequent phosphorylation of the transcription factor Elk, which could not be detected in other cell types stimulated with C3a. More surprisingly, the C3aR itself was translocated to the nucleus in C3a-stimulated MSCs, especially at low cell densities. Since nuclear activation/translocation of G protein-coupled receptors has been shown to induce long-term effects, this novel observation implies that C3a exerts far-reaching consequences on MSC biology. These results suggest that the anaphylatoxins C3a and C5a present in injured tissues contribute to the recruitment of MSCs and regulation of their behavior.


Asunto(s)
Factores Quimiotácticos/fisiología , Complemento C3a/fisiología , Complemento C5a/fisiología , Células Madre Mesenquimatosas/enzimología , Células Madre Mesenquimatosas/inmunología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Línea Celular , Células Cultivadas , Quimiotaxis de Leucocito/inmunología , Humanos , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor de Anafilatoxina C5a , Receptores de Complemento/biosíntesis , Receptores de Complemento/genética , Factores de Tiempo
19.
J Immunol ; 182(9): 5412-8, 2009 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-19380788

RESUMEN

Complement effectors are known to contribute to host cell injury in several inflammatory diseases. Contrary to this paradigm, in this study utilizing surgical liver resection (partial hepatectomy) in various complement-deficient mice as a model, we have demonstrated that complement anaphylatoxins C3a and C5a are required for the survival of liver cells during regeneration. The mechanisms of these cytoprotective functions of complement were related to the regulation of IL-6 and TNF production or release after liver resection. Disturbances in the cytokine milieu, induced by a loss of complement activity, were found to alter prosurvival signaling, including the IL-6/STAT3 and PI3K/Akt/mammalian target of rapamycin pathways. In conclusion, this study documents functions of complement proteins as prosurvival factors that, through their interactions with cytokines, inhibit apoptotic signaling in proliferating cells of epithelial origin.


Asunto(s)
Proteínas del Sistema Complemento/fisiología , Hígado/citología , Hígado/inmunología , Animales , Muerte Celular/genética , Muerte Celular/inmunología , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Activación de Complemento/genética , Activación de Complemento/inmunología , Complemento C3/deficiencia , Complemento C3/genética , Complemento C3/fisiología , Complemento C3a/deficiencia , Complemento C3a/genética , Complemento C3a/fisiología , Complemento C5a/deficiencia , Complemento C5a/genética , Complemento C5a/fisiología , Proteínas del Sistema Complemento/deficiencia , Proteínas del Sistema Complemento/genética , Hepatectomía , Regeneración Hepática/genética , Regeneración Hepática/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
20.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 42(1): 74-7, 2011 Jan.
Artículo en Zh | MEDLINE | ID: mdl-21355306

RESUMEN

OBJECTIVE: To investigate the effect of C3a and C5a and their receptor antagonists (C3aRA and C5aRA) on the expression of BETAcatenin in renal tubular epithelial cell line HK-2. METHODS: Cells were divided in into C3a group and C5a group which was further divided into four subgroups: C3a group (control, 1 micromol/L TGF-beta1, 50 nmol/L C3a, 1 micromol/L C3aRA); C5a group (control, 1 micromol/L TGF-beta1, 50 nmol/L C5a, 2.5 micromol/L C5aRA). Real time PCR and Western blot were used to detect mRNA and protein expression of beta-catenin. RESULTS: Real-time PCR and Western blot demonstrated that 1 micromol/L TGF-beta1 could increase the expression of beta-catenin; C3a and C5a presented the similar inducible effect as TGF-beta1, which could be blocked by C3aR antagonist and C5aR antagonist (C3aRA and C5aRA). CONCLUSION: Anaphylatoxin C3a and C5a can induce mRNA and protein expression of beta-catenin in renal tubular epithelial cells, which could be blocked by C3aRA and C5aRA. C3a and C5a may participate in tubular epithelial-mesenchymal transition in vitro.


Asunto(s)
Complemento C3a/fisiología , Complemento C5a/fisiología , Túbulos Renales/metabolismo , Receptores de Complemento/antagonistas & inhibidores , beta Catenina/metabolismo , Línea Celular , Humanos , Túbulos Renales/citología , Receptor de Anafilatoxina C5a , Receptores de Complemento/fisiología , beta Catenina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA