Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
1.
Cutan Ocul Toxicol ; 42(3): 137-143, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37335830

RESUMEN

PURPOSE: The complement system is considered to play an important role in the progression of myopia, whereas the influence of complement activation on the human scleral fibroblasts (HSFs) remains unknown. Hence, the effect of complement 3a (C3a) on HSFs was investigated in this study. METHODS: HSFs were cultured with exogenous C3a at 0.1 µM for various periods following different measurement protocols, and cells without C3a treatment served as negative control (NC). Cell viability was investigated using the MTS assay after 3 days of C3a treatment. Cell proliferation was evaluated by the 5-Ethynyl-20-Deoxyuridine (EdU) assay following C3a stimulation for 24 hours. Apoptosis was assessed by Annexin V-fluorescein isothiocyanate (FITC)/propidium iodide (PI) double staining following C3a stimulation for 48 hours and the stained cells were analysed using flow cytometry. The levels of type I collagen and matrix metalloproteinase-2 (MMP-2) were analysed using ELISA following C3a stimulation for 36 and 60 hours. The level of CD59 were analysed using western blot following C3a stimulation for 60 hours. RESULTS: The MTS assay revealed that cell viability was attenuated by 13% and 8% after C3a for 2 and 3 days, respectively (P < 0.05). The EdU assay demonstrated a 9% decrease in proliferation rate for the C3a-treated cells after 24 hours (P < 0.05). The apoptosis analysis revealed an increased percentage of cells in early apoptosis (P = 0.02) and total apoptosis (P = 0.02) in the C3a-treated group. Compared with NC group, the level of MMP-2 was increased by 17.6% (P = 0.002), whereas the levels of type I collagen and CD59 were respectively decreased by 12.5% (P = 0.024) and 21.6% (P = 0.044) with C3a treatment for 60 hours. CONCLUSIONS: These results indicated that C3a-induced complement activation is potentially involved in inducing myopic-associated scleral extracellular matrix remodelling via mediating the proliferation and function of HSFs.


Asunto(s)
Complemento C3a , Metaloproteinasa 2 de la Matriz , Humanos , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 2 de la Matriz/farmacología , Complemento C3a/metabolismo , Complemento C3a/farmacología , Colágeno Tipo I/metabolismo , Colágeno Tipo I/farmacología , Fibroblastos , Apoptosis
2.
Cytotherapy ; 20(12): 1427-1436, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30377040

RESUMEN

BACKGROUND AIMS: Mesenchymal stromal cells (MSCs) enhance islet function both in vitro and in vivo, at least in part by secreting ligands that activate islet G-protein coupled receptors (GPCRs). We assessed whether pre-treatment with a defined "cocktail" of MSC-secreted GPCR ligands enhances islet functional survival in vitro and improves the outcomes of islet transplantation in an experimental model of diabetes. METHODS: Isolated islets were cultured for 48 h with ANXA1, SDF-1 or C3a, alone or in combination. Glucose-stimulated insulin secretion (GSIS) and cytokine-induced apoptosis were measured immediately after the 48 h culture period and at 24 h or 72 h following removal of the ligands from the culture media. Islets were syngeneically transplanted underneath the kidney capsule of streptozotocin-induced diabetic C57BL/6 mice and blood glucose levels monitored for 28 days. RESULTS: Pre-culturing islets with a cocktail of ANXA1/SDF-1/C3a potentiated GSIS and protected islet cells from cytokine-induced apoptosis in vitro. These effects were maintained for up to 72 h after the removal of the factors from the culture medium, suggesting a sustained protection of islet graft functional survival during the immediate post-transplantation period. Islets pre-treated with the cocktail of MSC secretory factors were more effective in reducing blood glucose in diabetic mice, consistent with their improved functional survival in vivo. DISCUSSION: Pre-culturing islets with a cocktail of MSC secretory products offers a well-defined, cell-free approach to improve clinical islet transplantation outcomes while avoiding many of the safety, regulatory and logistical hurdles of incorporating MSCs into transplantation protocols.


Asunto(s)
Quimiocina CXCL12/farmacología , Complemento C3a/farmacología , Trasplante de Islotes Pancreáticos/métodos , Células Madre Mesenquimatosas/metabolismo , Animales , Anexina A1/genética , Anexina A1/metabolismo , Anexina A1/farmacología , Apoptosis/fisiología , Supervivencia Celular/fisiología , Células Cultivadas , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Complemento C3a/genética , Complemento C3a/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/terapia , Glucosa/metabolismo , Secreción de Insulina , Islotes Pancreáticos/citología , Islotes Pancreáticos/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos , Receptores Acoplados a Proteínas G/metabolismo
3.
Mol Vis ; 21: 264-72, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25814824

RESUMEN

PURPOSE: Age-related macular degeneration (AMD) is the leading cause of irreversible blindness in people 50 years of age or older in developed countries. The homozygous CC genotype in the complement factor H (CFH) Y402H single nucleotide polymorphism (SNP; rs1061170) is widely recognized as a risk factor for the development of AMD. In this study, we examined vitreal levels of granulocyte macrophage colony-stimulating factor (GM-CSF), a hematopoietic cytokine, and macrophages in the choroid of postmortem human eyes genotyped for the CFH Y402H SNP. METHODS: Twenty-two pairs of postmortem, non-diseased, human donor eyes were obtained. The vitreous and retinal tissues of the left eyes were collected for GM-CSF level measurement and CFH Y402H genotyping, respectively. The right eyes were paraffin-embedded and sectioned for immunohistochemistry using a macrophage and microglia marker, CD68. Cell cultures of RPE cells were stimulated with complement C3a, C5a, 4-hydroxynonenal (HNE), or tumor necrosis factor alpha (TNF-α), and GM-CSF expression was measured with a suspension assay or quantitative PCR. RESULTS: Eyes genotyped with the CC or the CT risk variant of the CFH Y402H SNP showed significantly increased levels of GM-CSF in the vitreous compared to eyes with the protective TT variant (mean ± standard error of mean, 607.54±85.83 pg/ml or 656.32±15.20 pg/ml versus 286.69±81.96 pg/ml, p<0.05). The choroid of eye tissues genotyped with the CC variant showed higher levels of CD68 immunoreactivity than the tissues genotyped with the TT variant (p<0.05). The GM-CSF levels detected in the supernatant of RPE cells in culture treated with HNE or TNF-α were significantly higher compared to the non-treated control (145.88±5.06 pg/ml and 149.32±3.76 pg/ml versus 123.27±4.05 pg/ml, p<0.05). Furthermore, the gene expression of GM-CSF detected in the lysate of RPE cells stimulated with complement C3a or C5a showed significantly increased fold changes compared to the non-treated control (C3a: 2.38±0.31 fold, p<0.05; C5a: 2.84±0.54 fold, p<0.01). CONCLUSIONS: Our data showed a relationship between the CFH Y402H polymorphism and GM-CSF levels in the vitreous and accumulation of choroidal macrophages in the postmortem eye. These data suggest that the at-risk variant of the CFH gene may contribute to the dysregulation of proinflammatory cytokines locally in the eye.


Asunto(s)
Coroides/metabolismo , Factor H de Complemento/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Macrófagos/citología , Polimorfismo de Nucleótido Simple , Cuerpo Vítreo/metabolismo , Aldehídos/farmacología , Sustitución de Aminoácidos , Autopsia , Células Cultivadas , Coroides/química , Coroides/citología , Complemento C3a/farmacología , Complemento C5a/farmacología , Factor H de Complemento/metabolismo , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Femenino , Expresión Génica , Genotipo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Macrófagos/metabolismo , Masculino , Persona de Mediana Edad , Epitelio Pigmentado de la Retina/citología , Epitelio Pigmentado de la Retina/efectos de los fármacos , Epitelio Pigmentado de la Retina/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Cuerpo Vítreo/química , Cuerpo Vítreo/citología
4.
Mediators Inflamm ; 2013: 713284, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23737652

RESUMEN

Acylation stimulating protein (ASP) is an adipokine derived from the immune complement system, which stimulates fat storage and is typically increased in obesity, type 2 diabetes, and cardiovascular disease. Using a diet-induced obesity (DIO) mouse model, the acute effects of ASP on energy metabolism and inflammatory processes in vivo were evaluated. We hypothesized that ASP would specifically exert proinflammatory effects. C57Bl/6 wild-type mice were put on a high-fat-high-sucrose diet for 12 weeks. Mice were then subjected to both glucose and insulin tolerance tests, each manipulation being preceded by recombinant ASP or vehicle (control) bolus injection. ASP supplementation increased whole-body glucose excursion, and this was accomplished with reduced concomitant insulin levels. However, ASP did not directly alter insulin sensitivity. ASP supplementation induced a proinflammatory phenotype, with higher levels of cytokines including IL-6 and TNF-α in plasma and in adipose tissue, liver, and skeletal muscle mRNA. Additionally, ASP increased M1 macrophage content of these tissues. ASP exerted a direct concentration-dependent role in the migration and M1 activation of cultured macrophages. Altogether, the in vivo and in vitro experiments demonstrate that ASP plays a role in both energy metabolism and inflammation, with paradoxical whole-body glucose-sensitizing yet proinflammatory effects.


Asunto(s)
Complemento C3a/farmacología , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Animales , Movimiento Celular/efectos de los fármacos , Grasas de la Dieta/efectos adversos , Humanos , Insulina/farmacología , Resistencia a la Insulina/fisiología , Interleucina-6/sangre , Macrófagos/citología , Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/sangre , Obesidad/etiología , Obesidad/metabolismo , Factor de Necrosis Tumoral alfa/sangre
5.
Cancer Res Commun ; 2(7): 725-738, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35937458

RESUMEN

Pancreatic cancer is one of the deadliest cancers, against which current immunotherapy strategies are not effective. Herein, we analyzed the immune cell composition of the tumor microenvironment of pancreatic cancer samples in The Cancer Genome Atlas and found that the presence of intratumoral NK cells correlates with survival. Subsequent analysis also indicated that NK cell exclusion from the microenvironment is found in a high percentage of clinical pancreatic cancers and in preclinical models of pancreatic cancer. Mechanistically, NK cell exclusion is regulated in part by complement C3a and its receptor signaling. Inhibition of the C3a receptor enhances NK cell infiltration in syngeneic mouse models of pancreatic cancer resulting in tumor growth delay. However, tumor growth inhibition mediated by NK cells is not sufficient alone for complete tumor regression, but is potentiated when combined with radiation therapy. Our findings indicate that although C3a inhibition is a promising approach to enhance NK cell-based immunotherapy against pancreatic cancer, its combination with radiation therapy hold greater therapeutic benefit.


Asunto(s)
Complemento C3a , Neoplasias Pancreáticas , Animales , Ratones , Complemento C3a/farmacología , Neoplasias Pancreáticas/radioterapia , Células Asesinas Naturales , Inmunoterapia/métodos , Microambiente Tumoral , Neoplasias Pancreáticas
6.
Neuropharmacology ; 205: 108927, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-34921829

RESUMEN

Activation of microglia and astrocytes following germinal matrix hemorrhage and intraventricular hemorrhage (GMH-IVH) plays a detrimental role in posthemorrhagic hydrocephalus (PHH). It is still unclear whether or how an interaction occurs between microglia and astrocytes in PHH. Here, we investigated the role of the C3/C3aR pathway in microglia and astrocyte interactions and whether C3/C3aR-targeted inhibition could alleviate PHH following GMH-IVH. A total of 152 Sprague-Dawley rats at postnatal day seven (P7) were enrolled in the study, and collagenase VII was used to induce GMH-IVH. Minocycline (45 mg/kg) was administered to inhibit microglial activation. Complement C3a peptide and C3aR antagonist (SB 290157, 10 mg/kg) were used to regulate the C3/C3aR pathway. As a result, the data demonstrated that periventricular C3aR+/Iba-1+ microglia and C3+/GFAP+ astrocytes were significantly increased in GMH-IVH pups at 28 days after surgery. Intranasal C3a peptide upregulated C3aR expression in microglia. Inhibition of microglia by minocycline decreased both C3+/GFAP+ astrocytes and the colocalization volume of Iba-1 and GFAP. In addition, intraperitoneally injected C3aRA alleviated the periventricular colocalization volume of microglia and astrocytes. Compared with vehicle-treated pups, the protein level of IL-1ß, IL-6 and TNF-α in cerebral spinal fluid and brain tissue at 28 days following GMH-IVH were reduced in C3aRA-treated pups. Moreover, hydrocephalus was alleviated, and long-term cognitive ability were improved in the C3aRA-treated group. Our data presented simultaneous periventricular astrogliosis and microgliosis of pups following GMH-IVH and proved their potential interaction through the C3/C3aR pathway, indicating C3aRA as a potential pharmacological treatment of PHH in neonates.


Asunto(s)
Arginina/análogos & derivados , Astrocitos/efectos de los fármacos , Compuestos de Bencidrilo/farmacología , Hemorragia Cerebral/tratamiento farmacológico , Hemorragia Cerebral/metabolismo , Complemento C3a/farmacología , Hidrocefalia/tratamiento farmacológico , Microglía/efectos de los fármacos , Receptores Acoplados a Proteínas G/efectos de los fármacos , Animales , Animales Recién Nacidos , Arginina/administración & dosificación , Arginina/farmacología , Compuestos de Bencidrilo/administración & dosificación , Hemorragia Cerebral/complicaciones , Hemorragia Cerebral Intraventricular/complicaciones , Hemorragia Cerebral Intraventricular/tratamiento farmacológico , Hemorragia Cerebral Intraventricular/metabolismo , Complemento C3a/administración & dosificación , Modelos Animales de Enfermedad , Hidrocefalia/etiología , Hidrocefalia/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/antagonistas & inhibidores
7.
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
8.
J Exp Med ; 186(2): 199-207, 1997 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-9221749

RESUMEN

The pathophysiological relevance of the complement split product C3a as a proinflammatory mediator is still ill defined. The expression pattern of the human C3a receptor (C3aR) can provide important clues for the role of this anaphylatoxin in inflammation. There is strong evidence for C3aR expression on basophils, and eosinophils, but additionally, only on tumor cell lines of leukemic or hepatic origin. It is unclear whether neutrophils also express the C3aR, but need a costimulus provided by eosinophils for certain biological responses, or whether neutrophils lack the C3aR and respond to C3a via a secondary stimulus generated by eosinophils, i.e., by an indirect mode. In the present study, polyclonal antiserum raised against the second extracellular loop of the C3aR was used to characterize C3aR expression on peripheral blood leukocytes. For high degree purification of neutrophils, a negative selection method was established that decreased the contamination with CD9(bright+) eosinophils down to <0.2%. Flow cytometric analyses, functional assays, and binding assays on highly purified neutrophils confirmed C3aR expression and coupling. Monocytes were identified as an additional C3aR-positive cell population of the peripheral blood. The expression of the C3aR on eosinophils could be confirmed. In contrast, the receptor could not be detected on unchallenged B or T lymphocytes (or lymphocyte-derived Raji cells).


Asunto(s)
Linfocitos B/química , Complemento C3a/metabolismo , Glicoproteínas de Membrana , Monocitos/química , Neutrófilos/química , Receptores de Complemento/análisis , Linfocitos T/química , Animales , Antígenos CD/análisis , Northern Blotting , Calcio/metabolismo , Complemento C3a/farmacología , Citometría de Flujo , Humanos , ARN Mensajero/análisis , Conejos , Ratas , Tetraspanina 29
9.
J Exp Med ; 181(6): 2119-27, 1995 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-7760001

RESUMEN

Inflammatory action of the potent chemotaxin C5a has been well characterized on a variety of human cell types, including neutrophils, monocytes, basophils, and eosinophils. The cellular effects of C3a are less well defined. Contradictory reports have been published for C3a activation of neutrophils. Recent reports that C3a activates both basophils and eosinophils prompted us to reinvestigate the effects of C3a stimulation on eosinophils. We hypothesized that C3a activation of eosinophils, cells that are present in most neutrophil preparations, might lead to neutrophil activation. Using neutrophils of 98% purity, we observed no evidence of cellular activation after stimulation with either C3a, recombinant human C3a (rhC3a), or the synthetic C3a analogue C3a 57-77, Y57. Eosinophils purified to > 98% purity displayed concentration-dependent polarization, chemotaxis, and enzyme release by stimulation with C3a, rhC3a, and the synthetic C3a analogue. An inactive form of C3a, C3adesArg, failed to stimulate either eosinophils or neutrophils. Using neutrophil preparations containing 5-9% eosinophils, up to 20% of neutrophils became polarized after exposure to C3a. Likewise, we demonstrated that supernatant from C3a-stimulated eosinophils promotes neutrophil chemotaxis. Eosinophil polarization experiments were repeated in the presence of antibody to the C5a receptor (C5aR) to show that C3a and C5a interact with different receptors. C3a activates eosinophils in the presence of anti-C5aR antibody at concentrations that fully block C5a activation. We conclude that eosinophils are directly activated by either C3a or C5a, whereas C3a failed to activate neutrophils. C3a acts on eosinophils via a receptor that is distinct from C5aR. Since neutrophils are indirectly stimulated by C3a, eosinophils contaminating neutrophil preparations may explain earlier reports that C3a activates human neutrophils.


Asunto(s)
Quimiotaxis de Leucocito/fisiología , Complemento C3a/farmacología , Eosinófilos/fisiología , Activación Neutrófila , Neutrófilos/fisiología , Secuencia de Aminoácidos , Quimiotaxis de Leucocito/efectos de los fármacos , Complemento C5a/farmacología , Relación Dosis-Respuesta a Droga , Eosinófilos/efectos de los fármacos , Glucuronidasa/análisis , Glucuronidasa/sangre , Humanos , Cinética , Datos de Secuencia Molecular , Neutrófilos/efectos de los fármacos , Fragmentos de Péptidos/síntesis química , Fragmentos de Péptidos/farmacología , Proteínas Recombinantes/farmacología , Factores de Tiempo
10.
Stem Cells ; 27(11): 2824-32, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19785034

RESUMEN

Anaphylatoxin C3a is a third complement component (C3)-derived peptide, the multiple functions of which range from stimulation of inflammation to neuroprotection. In a previous study, we have shown that signaling through C3a receptor positively regulates in vivo neurogenesis in adult mouse brain. Here, we studied the direct effects of C3a on adult mouse whole brain-derived neural progenitor cells (NPCs) in vitro. Our results demonstrate that NPCs bind C3a in a specific and reversible manner and that C3a stimulates neuronal differentiation of NPCs. Furthermore, C3a stimulated the migration of NPCs induced by low concentrations of stromal cell-derived factor (SDF)-1alpha, whereas it inhibited NPC migration at high concentration of SDF-1alpha. In the same manner, C3a modulated SDF-1alpha-induced extracellular-signal-regulated kinases 1 and 2 (ERK1/2) phosphorylation in these cells. In addition, C3a had inhibitory effect on SDF-1alpha-induced neuronal differentiation of NPCs. These data show that C3a modulates SDF-1alpha-induced differentiation and migration of these cells, conceivably through the regulation of ERK1/2 phosphorylation. Our results provide the first evidence that C3a regulates neurogenesis by directly affecting the fate and properties of NPCs.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Complemento C3a/farmacología , Factores Inmunológicos/farmacología , Neuronas/citología , Neuronas/efectos de los fármacos , Células Madre/citología , Animales , Western Blotting , Células Cultivadas , Quimiocina CXCL12/farmacología , Humanos , Inmunoprecipitación , Ratones , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación/efectos de los fármacos , ARN Mensajero , Células Madre/metabolismo
11.
Front Immunol ; 11: 615236, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33597949

RESUMEN

Both, aberrant mast cell responses and complement activation contribute to allergic diseases. Since mast cells are highly responsive to C3a and C5a, while Interleukin-33 (IL-33) is a potent mast cell activator, we hypothesized that IL-33 critically regulates mast cell responses to complement anaphylatoxins. We sought to understand whether C3a and C5a differentially activate primary human mast cells, and probe whether IL-33 regulates C3a/C5a-induced mast cell activities. Primary human mast cells were generated from peripheral blood precursors or isolated from healthy human lung tissue, and mast cell complement receptor expression, degranulation, mediator release, phosphorylation patterns, and calcium flux were assessed. Human mast cells of distinct origin express constitutively higher levels of C3aR1 than C5aR1, and both receptors are downregulated by anaphylatoxins. While C3a is a potent mast cell degranulation inducer, C5a is a weaker secretagogue with more delayed effects. Importantly, IL-33 potently enhances the human mast cell reactivity to C3a and C5a (degranulation, cytokine and chemokine release), independent of changes in C3a or C5a receptor expression or the level of Ca2+ influx. Instead, this reflects differential dynamics of intracellular signaling such as ERK1/2 phosphorylation. Since primary human mast cells respond differentially to anaphylatoxin stimulation, and that IL-33 is a key regulator of mast cell responses to complement anaphylatoxins, this is likely to aggravate Th2 immune responses. This newly identified cross-regulation may be important for controlling exacerbated complement- and mast cell-dependent Th2 responses and thus provides an additional rationale for targeting anti-IL33 therapeutically in allergic diseases.


Asunto(s)
Complemento C3a/farmacología , Complemento C5a/farmacología , Interleucina-33/farmacología , Mastocitos/efectos de los fármacos , Antígenos CD/biosíntesis , Antígenos CD/genética , Células Sanguíneas , Señalización del Calcio/efectos de los fármacos , Degranulación de la Célula/efectos de los fármacos , Células Cultivadas , Complemento C3a/inmunología , Complemento C5a/inmunología , Sinergismo Farmacológico , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Interferón gamma/farmacología , Interleucina-4/farmacología , Ligandos , Pulmón/citología , Mastocitos/inmunología , Mastocitos/metabolismo , Proteínas de la Membrana/metabolismo , Especificidad de Órganos , Fosforilación , Procesamiento Proteico-Postraduccional , Receptores de Complemento/biosíntesis , Receptores de Complemento/genética
12.
Sheng Li Xue Bao ; 61(1): 56-64, 2009 Feb 25.
Artículo en Zh | MEDLINE | ID: mdl-19224055

RESUMEN

Perilipin and adipophilin, two significant lipid droplet (LD)-specific proteins, participate in storing fat or ectopic lipid deposition and fat mobilization in many types of mammalian cells. Acylation stimulating protein (ASP) is a novel adipocyte-derived hormone known for a major determinant for triglyceride synthesis (TGS) and lipid metabolism. The present study was aimed to investigate: (1) whether ASP, rather than insulin, is a powerful potentiator which could physiologically and directly influence TGS during 3T3-L1 preadipocyte differentiation; (2) whether ASP exposure at indicated time points during 3T3-L1 preadipocyte differentiation could influence the gene/protein expression of adipophilin and perilipin. 3T3-L1 preadipocytes were differentiated by traditional hormone cocktail and divided into control, ASP and insulin groups according to the treatment of ASP (1 mmol/L) or insulin (100 nmol/L). ASP-stimulated and insulin-stimulated TGS rate at indicated time points (0 d, 3 d, 6 d, 9 d) were assayed by measuring the incorporation of [(3)H]-oleic acid into TG, and the corresponding glucose transport was assayed by [(3)H]-2-DG uptake. The effects of ASP or insulin on gene/protein expression of adipophilin and perilipin at indicated time points were evaluated by semi-quantitative reverse transcription-polymerase chain reaction (RT-PCR) and Western blot. The results obtained were as follows: (1) on the 3rd and 6th day of differentiation, ASP dramatically enhanced TGS rate compared with control group (P<0.05, P<0.01); There was no significant difference in TGS rate between insulin group and control group; (2) on the 6th and 9th day of differentiation, both ASP and insulin promoted glucose uptake (P<0.05, P<0.01), and the promoting effect in ASP group was greater than that in insulin group; (3) ASP elevated adipophilin gene and protein expression at the very early stage of differentiation (P<0.05, P<0.001) and had no significant effect from the 4th day of differentiation. Perilipin gene and protein expression increased throughout preadipocyte differentiation and its expression was up-regulated following ASP stimulation from the 3rd day of differentiation (P<0.05, P<0.001) to the end of differentiation (P<0.05); (4) Insulin did not affect gene and protein variation pattern of adipophilin and perilipin. Taken together, this study provides evidence that ASP-evoked changes in gene and protein expression of adipophilin and perilipin correlate with ASP-stimulated TGS acceleration, and adipophilin and perilipin are involved in the molecular mechanism of ASP-induced adipogenesis and LD formation.


Asunto(s)
Adipocitos/citología , Proteínas Portadoras/metabolismo , Diferenciación Celular , Complemento C3a/farmacología , Proteínas de la Membrana/metabolismo , Fosfoproteínas/metabolismo , Células 3T3-L1 , Animales , Expresión Génica , Insulina/farmacología , Ratones , Perilipina-1 , Perilipina-2
13.
Biochim Biophys Acta ; 1768(2): 346-53, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17169328

RESUMEN

Antimicrobial peptides are generated during activation of the complement system [Nordahl et al. Proc. Natl. Acad. Sci. U. S. A. 2004, 101:16879-16884]. Here we show that the anaphylatoxin C3a exerts antimicrobial effects against the yeast Candida. Fluorescence microscopy and electron microscopy analysis demonstrated that C3a-derived peptides bound to the cell surface of Candida, and induced membrane perturbations and release of extracellular material. Various Candida isolates were found to induce complement degradation, leading to generation of C3a. Arginine residues were found to be critical for the antifungal and membrane breaking activity of a C3a-derived antimicrobial peptide, CNY21 (C3a; Cys57-Arg77). A CNY21 variant with increased positive net charge displayed enhanced antifungal activity. Thus, C3a-derived peptides can be utilized as templates in the development of peptide-based antifungal therapies.


Asunto(s)
Antifúngicos/farmacología , Péptidos Catiónicos Antimicrobianos/farmacología , Candida albicans/efectos de los fármacos , Complemento C3a/farmacología , Secuencia de Aminoácidos , Humanos , Liposomas , Pruebas de Sensibilidad Microbiana , Datos de Secuencia Molecular
14.
Mol Immunol ; 103: 125-132, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30261438

RESUMEN

Multiple studies have identified that complement becomes activated during inflammation of the intestines yet it is unclear what roles the split complement molecules play. The epithelium, in particular, may be impacted and accordingly, we first discovered that colonic cell lines indeed possess the C5aR. Here we examined whether these cells also possess the C3aR. We determined that T84, HT-29 and Caco2 all possess C3aR mRNA and protein; T84 and HT29 were used to further explore the consequence of C3a binding the C3aR. C3a led to increased mRNA for CXCL2, CXCL8 and CXCL11. Polarized T84 monolayers responded to apically applied C3a with increased CXCL8 mRNA more rapidly than if the C3a was applied basolaterally. Polarized monolayers also increased permeability when treated with C3a. ERK1/2 was activated by C3a and the increase in CXCL8 mRNA was ERK-dependent in both T84 and HT-29. C3a resulted in activation of Gαi, determined by the ERK1/2 signal showing sensitivity to pertussis toxin. The transmembrane signal was further mapped to include Ras and c-Raf. Finally, we show that the C3aR is expressed by primary cells in mouse enteroids. We conclude that complement activation will contribute to the epithelial response during inflammation through C3a binding to the C3aR including by priming the cells to upregulate mRNA for selected chemokines.


Asunto(s)
Quimiocinas/inmunología , Complemento C3a/farmacología , Células Epiteliales/efectos de los fármacos , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/inmunología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Mediadores de Inflamación/inmunología , Animales , Células CACO-2 , Línea Celular Tumoral , Células Cultivadas , Quimiocinas/genética , Quimiocinas/metabolismo , Colon/patología , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Células HT29 , Humanos , Mediadores de Inflamación/metabolismo , Ratones Endogámicos C57BL , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/inmunología , Receptores Acoplados a Proteínas G/metabolismo
15.
Mol Immunol ; 93: 266-277, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28860090

RESUMEN

Candida albicans the most frequently isolated clinical fungal pathogen can cause local as well as systemic and life-threatening infections particularly in immune-compromised individuals. A better and more detailed understanding how C. albicans evades human immune attack is therefore needed for identifying fungal immune-evasive proteins and develop new therapies. Here, we identified Pra1, the pH-regulated C. albicans antigen as a hierarchical complement inhibitor that targets C3, the central human complement component. Pra1 cleaved C3 at a unique site and further inhibited effector function of the activation fragments. The newly formed C3a-like peptide lacked the C-terminal arginine residue needed for C3a-receptor binding and activation. Moreover, Pra1 also blocked C3a-like antifungal activity as shown in survival assays, and the C3b-like molecule formed by Pra1 was degraded by the host protease Factor I. Pra1 also bound to C3a and C3b generated by human convertases and blocked their effector functions, like C3a antifungal activity shown by fungal survival, blocked C3a binding to human C3a receptor-expressing HEK cells, activation of Fura2-AM loaded cells, intracellular Ca2+ signaling, IL-8 release, C3b deposition, as well as opsonophagocytosis and killing by human neutrophils. Thus, upon infection C. albicans uses Pra1 to destroy C3 and to disrupt host complement attack. In conclusion, candida Pra1 represents the first fungal C3-cleaving protease identified and functions as a fungal master regulator of innate immunity and as a central fungal immune-escape protein.


Asunto(s)
Candida albicans/enzimología , Complemento C3/antagonistas & inhibidores , Proteínas Fúngicas/fisiología , Secuencia de Aminoácidos , Unión Competitiva , Señalización del Calcio/efectos de los fármacos , Candida albicans/efectos de los fármacos , Candida albicans/inmunología , Línea Celular , Complemento C3/inmunología , Complemento C3/metabolismo , Complemento C3/farmacología , Complemento C3a/antagonistas & inhibidores , Complemento C3a/farmacología , Complemento C3b/antagonistas & inhibidores , Complemento C3b/farmacología , Proteínas Fúngicas/antagonistas & inhibidores , Proteínas Fúngicas/farmacología , Células HEK293 , Humanos , Interleucina-8/metabolismo , Neutrófilos/efectos de los fármacos , Neutrófilos/fisiología , Proteínas Opsoninas/inmunología , Fragmentos de Péptidos/metabolismo , Fagocitosis/efectos de los fármacos , Inhibidores de Proteasas/farmacología , Proteolisis , Receptores de Complemento/antagonistas & inhibidores , Receptores de Complemento/metabolismo , Virulencia/inmunología
16.
Transplantation ; 101(7): 1559-1572, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28640789

RESUMEN

BACKGROUND: Antibody-mediated rejection (AMR) of most solid organs is characterized by evidence of complement activation and/or intragraft macrophages (C4d + and CD68+ biopsies). We previously demonstrated that crosslinking of HLA I by antibodies triggered endothelial activation and monocyte adhesion. We hypothesized that activation of the classical complement pathway at the endothelial cell surface by HLA antibodies would enhance monocyte adhesion through soluble split product generation, in parallel with direct endothelial activation downstream of HLA signaling. METHODS: Primary human aortic endothelial cells (HAEC) were stimulated with HLA class I antibodies in the presence of intact human serum complement. C3a and C5a generation, endothelial P-selectin expression, and adhesion of human primary and immortalized monocytes (Mono Mac 6) were measured. Alternatively, HAEC or monocytes were directly stimulated with purified C3a or C5a. Classical complement activation was inhibited by pretreatment of complement with an anti-C1s antibody (TNT003). RESULTS: Treatment of HAEC with HLA antibody and human complement increased the formation of C3a and C5a. Monocyte recruitment by human HLA antibodies was enhanced in the presence of intact human serum complement or purified C3a or C5a. Specific inhibition of the classical complement pathway using TNT003 or C1q-depleted serum significantly reduced adhesion of monocytes in the presence of human complement. CONCLUSIONS: Despite persistent endothelial viability in the presence of HLA antibodies and complement, upstream complement anaphylatoxin production exacerbates endothelial exocytosis and leukocyte recruitment. Upstream inhibition of classical complement may be therapeutic to dampen mononuclear cell recruitment and endothelial activation characteristic of microvascular inflammation during AMR.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Adhesión Celular/efectos de los fármacos , Inactivadores del Complemento/farmacología , Vía Clásica del Complemento/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Antígenos HLA-A/inmunología , Inmunosupresores/farmacología , Monocitos/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Complemento C3a/farmacología , Complemento C5a/farmacología , Relación Dosis-Respuesta a Droga , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Exocitosis/efectos de los fármacos , Antígenos HLA-A/metabolismo , Humanos , Antígeno de Macrófago-1/inmunología , Antígeno de Macrófago-1/metabolismo , Monocitos/inmunología , Monocitos/metabolismo , Selectina-P/inmunología , Selectina-P/metabolismo
17.
FASEB J ; 19(7): 798-800, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15758041

RESUMEN

Growing evidence suggests that anaphylatoxins, C3a and C5a, play important roles in innate immunity and may also participate in the pathogenesis of asthma. Previous studies with animal models and immunohistochemistry analysis of lung tissue indicated that anaphylatoxins may regulate airway hyperresponsiveness (AHR) in asthma via the activation of their cell surface G protein-coupled receptors (C3aR and C5aR) in airway smooth muscle (ASM) cells. Using RT-PCR, flow cytometry, and confocal microscopy, we made the surprising observation that while C3aR and C5aR were expressed in human mast cells, they were not present in cultured primary human or murine ASM cells. Furthermore, we could not detect C3aR in smooth muscle-positive cells of human trachea or bronchus. Interestingly, incubation of human mast cells with ASM cells, but not its culture supernatant, caused a significant enhancement of C3a-induced mast cell degranulation. Although stem cell factor (SCF) and its receptor c-kit are constitutively expressed on ASM cells and mast cells, respectively, neutralizing antibodies to SCF and c-kit failed to inhibit ASM cell-mediated enhancement of mast cell degranulation. However, dexamethasone-treated ASM cells were normal for cell surface SCF expression but were significantly less effective in enhancing C3a-induced mast cell degranulation when compared with untreated cells. These findings suggest that cell-cell interaction between ASM cells and mast cells, via a SCF-c-kit-independent but dexamethasone-sensitive mechanism, enhances C3a-induced mast cell degranulation, which likely regulates ASM function, thus contributing to the pathogenesis of asthma.


Asunto(s)
Degranulación de la Célula/fisiología , Complemento C3a/fisiología , Mastocitos/fisiología , Músculo Liso/fisiología , Sistema Respiratorio/citología , Animales , Línea Celular , Células Cultivadas , Técnicas de Cocultivo , Complemento C3a/análisis , Complemento C3a/farmacología , Complemento C5a/farmacología , Medios de Cultivo Condicionados , Dexametasona/farmacología , Humanos , Inmunohistoquímica , Antígeno de Macrófago-1/análisis , Antígeno de Macrófago-1/genética , Antígeno de Macrófago-1/fisiología , Macrófagos/química , Macrófagos/fisiología , Macrófagos Alveolares/química , Mastocitos/química , Ratones , Músculo Liso/química , ARN Mensajero/análisis , Receptor de Anafilatoxina C5a/análisis , Receptor de Anafilatoxina C5a/genética , Receptor de Anafilatoxina C5a/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Tráquea/química , Tráquea/citología , Transfección
18.
Int Immunopharmacol ; 6(7): 1109-18, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16714214

RESUMEN

Matrix metalloproteinase 9 (MMP-9) is a crucial proteinase, utilized by both eosinophils and neutrophils, that mediates transmigration through extracellular basement membranes. We have found that neutralization of MMP-9 by a monoclonal antibody or a chemical inhibitor blocked C5a dependent chemotaxis of these granulocytes in vitro. The levels of MMP-9 secreted by the action of C5a from eosinophils were about 50-fold lower than those from neutrophils, consistent with results from confocal microscopy, where the density of MMP-9 containing granules was fewer within eosinophils than in neutrophils. Zymography indicated gelatin degrading activity of the molecular size of pro MMP-9 in supernatants from eosinophils and neutrophils stimulated by C5a, with no evidence of proteolytic activation. Instead MMP-9 activation appeared oxidative, since inhibition of NADPH oxidase and nitric oxide synthase by DPI or L-NIL abrogated C5a-mediated chemotaxis through basement membranes. In keeping with this mode of activation, C5a, known as an agent of superoxide generation, was also found to induce secretion of nitric oxide from human eosinophils and rat granulocytes and monocytes. In conclusion C5a is an important mediator that brings about secretion and oxidative activation of MMP-9, a requisite protease for transmigration, from both eosinophils and neutrophils.


Asunto(s)
Complemento C5a/farmacología , Eosinófilos/inmunología , Metaloproteinasa 9 de la Matriz/inmunología , Neutrófilos/inmunología , Animales , Anticuerpos Monoclonales/farmacología , Quimiotaxis de Leucocito/efectos de los fármacos , Complemento C3a/farmacología , Inhibidores Enzimáticos/farmacología , Eosinófilos/efectos de los fármacos , Humanos , Interleucina-5/farmacología , Inhibidores de la Metaloproteinasa de la Matriz , Neutrófilos/efectos de los fármacos , Óxido Nítrico/biosíntesis , Piranos/farmacología , Ratas
19.
Mol Immunol ; 42(5): 581-7, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15607817

RESUMEN

Complement component C3a causes a robust degranulation in human mast cells. Whether C3a also stimulates chemokine production in human mast cells and what signaling pathway it activates is not known. In the present study, we demonstrate that CD34+ cell-derived primary mast cells and a human mast cell line LAD 2 express surface C3a receptors at similar levels. Furthermore, C3a caused approximately 50% internalization of cell surface C3a receptors in both cell types. We therefore used LAD 2 cells as a model to study C3a-induced biological responses and signaling in human mast cells. We found that C3a stimulated substantial degranulation and induced chemokine monocyte chemoattractant protein 1 (MCP-1/CCL2) and regulated upon activation, normal T-cell expressed and secreted (RANTES/CCL5) production in LAD 2 mast cells. C3a caused a rapid and sustained extracellular-signal-regulated kinase (ERK) phosphorylation and Akt phosphorylation in LAD 2 mast cells. Furthermore, U0126 and LY294002, which respectively inhibit MEK-induced ERK phosphorylation and PI3 kinase-mediated Akt phosphorylation had distinct effects on C3a-induced responses. Thus, U0126, which blocked C3a-induced RANTES/CCL5 production by 50.6+/-2.3%, inhibited MCP-1/CCL2 generation by 85.2+/-0.6%. In contrast, LY294002 had no effect on C3a-induced RANTES/CCL5 production but blocked MCP-1/CCL2 generation by 83.7+/-1.5%. These data demonstrate that C3a activates divergent signaling pathways to induce chemokine production in human mast cells.


Asunto(s)
Quimiocina CCL2/biosíntesis , Quimiocina CCL5/biosíntesis , Quimiocinas CC/biosíntesis , Complemento C3a/farmacología , Mastocitos/efectos de los fármacos , Mastocitos/inmunología , Células Cultivadas , Complemento C3a/metabolismo , Complemento C5a/metabolismo , Complemento C5a/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Antígeno de Macrófago-1/metabolismo , Mastocitos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal
20.
Biomed Res Int ; 2016: 6958752, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27747237

RESUMEN

Complement activation, specifically complement 3 (C3) activation and C3a generation, contributes to an imbalance between angiogenic stimulation by vascular endothelial growth factor (VEGF) and angiogenic inhibition by pigment epithelial derived factor (PEDF), leading to pathological angiogenesis. This study aimed to investigate the effects of C3a and small interfering RNA (siRNA) targeting C3 on the levels of VEGF and PEDF mRNAs in human retinal pigment epithelial (RPE) cells. ARPE-19 cells were cultured in the presence of exogenous C3a at 0.1 µM and 0.3 µM C3a for 24, 48, and 72 hours. 0.1 pmol/µL duplexes of siRNA targeting C3 were applied for C3a inhibition by transfecting ARPE-19 cells for 48 hours. RT-PCR was performed to examine the level of VEGF and PEDF mRNA. A random siRNA duplex was set for control siRNA. Results demonstrated that exogenous C3a significantly upregulated VEGF and downregulated PEDF mRNA levels in cultured ARPE-19 cells, and siRNA targeting C3 transfection reversed the above changes, significantly reducing VEGF and enhancing PEDF mRNAs level in ARPE-19 cells compared to the control. The present data provided evidence that reducing C3 activation can decreases VEGF and increase PEDF mRNA level in RPE and may serve as a potential therapy in pathological angiogenesis.


Asunto(s)
Complemento C3a/farmacología , Células Epiteliales/metabolismo , Proteínas del Ojo/genética , Factores de Crecimiento Nervioso/genética , Epitelio Pigmentado de la Retina/citología , Serpinas/genética , Factor A de Crecimiento Endotelial Vascular/genética , Línea Celular , Complemento C3a/genética , Complemento C3a/metabolismo , Células Epiteliales/efectos de los fármacos , Proteínas del Ojo/metabolismo , Humanos , Factores de Crecimiento Nervioso/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Serpinas/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA