Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 104
Filtrar
1.
FASEB J ; 36(11): e22599, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36250902

RESUMEN

Emerging evidence suggest that C3aR plays important roles in homeostasis, host defense and disease. Although it is known that C3aR is protective in several models of acute bacterial infections, the role for C3aR in chronic infection is largely unknown. Here we show that C3aR is protective in experimental chronic pyelonephritis. Global C3aR deficient (C3ar-/- ) mice had higher renal bacterial load, more pronounced renal histological lesions, increased renal apoptotic cell accumulation, tissue inflammation and extracellular matrix deposition following renal infection with uropathogenic E. coli (UPEC) strain IH11128, compared to WT control mice. Myeloid C3aR deficient (Lyz2-C3ar-/- ) mice exhibited a similar disease phenotype to global C3ar-/- mice. Pharmacological treatment with a C3aR agonist reduced disease severity in experimental chronic pyelonephritis. Furthermore, macrophages of C3ar-/- mice exhibited impaired ability to phagocytose UPEC. Our data clearly demonstrate a protective role for C3aR against experimental chronic pyelonephritis, macrophage C3aR plays a major role in the protection, and C3aR is necessary for phagocytosis of UPEC by macrophages. Our observation that C3aR agonist curtailed the pathology suggests a therapeutic potential for activation of C3aR in chronic infection.


Asunto(s)
Infecciones por Escherichia coli , Pielonefritis , Receptores de Complemento , Animales , Ratones , Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/patología , Inflamación/inmunología , Inflamación/microbiología , Inflamación/patología , Riñón/microbiología , Riñón/patología , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/patología , Pielonefritis/inmunología , Pielonefritis/microbiología , Pielonefritis/patología , Pielonefritis/prevención & control , Escherichia coli Uropatógena/patogenicidad , Receptores de Complemento/agonistas , Receptores de Complemento/deficiencia , Receptores de Complemento/genética , Receptores de Complemento/inmunología , Matriz Extracelular/metabolismo
2.
Nat Commun ; 12(1): 7172, 2021 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-34887405

RESUMEN

Complement receptor of immunoglobulin superfamily (CRIg) is expressed on liver macrophages and directly binds complement component C3b or Gram-positive bacteria to mediate phagocytosis. CRIg plays important roles in several immune-mediated diseases, but it is not clear how its pathogen recognition and phagocytic functions maintain homeostasis and prevent disease. We previously associated cytolysin-positive Enterococcus faecalis with severity of alcohol-related liver disease. Here, we demonstrate that CRIg is reduced in liver tissues from patients with alcohol-related liver disease. CRIg-deficient mice developed more severe ethanol-induced liver disease than wild-type mice; disease severity was reduced with loss of toll-like receptor 2. CRIg-deficient mice were less efficient than wild-type mice at clearing Gram-positive bacteria such as Enterococcus faecalis that had translocated from gut to liver. Administration of the soluble extracellular domain CRIg-Ig protein protected mice from ethanol-induced steatohepatitis. Our findings indicate that ethanol impairs hepatic clearance of translocated pathobionts, via decreased hepatic CRIg, which facilitates progression of liver disease.


Asunto(s)
Enterococcus faecalis/inmunología , Infecciones por Bacterias Grampositivas/inmunología , Hepatopatías Alcohólicas/inmunología , Macrófagos/inmunología , Receptores de Complemento 3b/inmunología , Receptores de Complemento/inmunología , Animales , Traslocación Bacteriana , Complemento C3b/inmunología , Enterococcus faecalis/fisiología , Etanol/efectos adversos , Femenino , Tracto Gastrointestinal/microbiología , Infecciones por Bacterias Grampositivas/genética , Infecciones por Bacterias Grampositivas/microbiología , Humanos , Hígado/efectos de los fármacos , Hígado/inmunología , Hígado/microbiología , Hepatopatías Alcohólicas/etiología , Hepatopatías Alcohólicas/genética , Hepatopatías Alcohólicas/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Complemento/deficiencia , Receptores de Complemento/genética , Receptores de Complemento 3b/genética
3.
Front Immunol ; 12: 604812, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33692783

RESUMEN

The C3a receptor (C3aR) is a seven trans-membrane domain G-protein coupled receptor with a range of immune modulatory functions. C3aR is activated by the third complement component (C3) activation derived peptide C3a and a neuropeptide TLQP-21. In the central nervous system (CNS), C3aR is expressed by neural progenitors, neurons as well as glial cells. The non-immune functions of C3aR in the adult CNS include regulation of basal neurogenesis, injury-induced neural plasticity, and modulation of glial cell activation. In the developing brain, C3aR and C3 have been shown to play a role in neural progenitor cell proliferation and neuronal migration with potential implications for autism spectrum disorder, and adult C3aR deficient (C3aR-/-) mice were reported to exhibit subtle deficit in recall memory. Here, we subjected 3 months old male C3aR-/- mice to a battery of behavioral tests and examined their brain morphology. We found that the C3aR-/- mice exhibit a short-term memory deficit and increased locomotor activity, but do not show any signs of autistic behavior as assessed by self-grooming behavior. We also found regional differences between the C3aR-/- and wild-type (WT) mice in the morphology of motor and somatosensory cortex, as well as amygdala and hippocampus. In summary, constitutive absence of C3aR signaling in mice leads to neurodevelopmental abnormalities that persist into adulthood and are associated with locomotive hyperactivity and altered cognitive functions.


Asunto(s)
Trastorno por Déficit de Atención con Hiperactividad/etiología , Trastorno por Déficit de Atención con Hiperactividad/metabolismo , Encéfalo/metabolismo , Encéfalo/fisiopatología , Receptores de Complemento/deficiencia , Animales , Biomarcadores , Encéfalo/patología , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Predisposición Genética a la Enfermedad , Hipocampo/metabolismo , Hipocampo/fisiopatología , Inmunohistoquímica , Ratones , Ratones Noqueados , Neurogénesis/genética
4.
J Neuroinflammation ; 17(1): 336, 2020 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-33176797

RESUMEN

BACKGROUND: The risk of glaucoma increases significantly with age and exposure to elevated intraocular pressure, two factors linked with neuroinflammation. The complement cascade is a complex immune process with many bioactive end-products, including mediators of inflammation. Complement cascade activation has been shown in glaucoma patients and models of glaucoma. However, the function of complement-mediated inflammation in glaucoma is largely untested. Here, the complement peptide C3a receptor 1 was genetically disrupted in DBA/2J mice, an ocular hypertensive model of glaucoma, to test its contribution to neurodegeneration. METHODS: A null allele of C3ar1 was backcrossed into DBA/2J mice. Development of iris disease, ocular hypertension, optic nerve degeneration, retinal ganglion cell activity, loss of RGCs, and myeloid cell infiltration in C3ar1-deficient and sufficient DBA/2J mice were compared across multiple ages. RNA sequencing was performed on microglia from primary culture to determine global effects of C3ar1 on microglia gene expression. RESULTS: Deficiency in C3ar1 lowered the risk of degeneration in ocular hypertensive mice without affecting intraocular pressure elevation at 10.5 months of age. Differences were found in the percentage of mice affected, but not in individual characteristics of disease progression. The protective effect of C3ar1 deficiency was then overcome by additional aging and ocular hypertensive injury. Microglia and other myeloid-derived cells were the primary cells identified that express C3ar1. In the absence of C3ar1, microglial expression of genes associated with neuroinflammation and other immune functions were differentially expressed compared to WT. A network analysis of these data suggested that the IL10 signaling pathway is a major interaction partner of C3AR1 signaling in microglia. CONCLUSIONS: C3AR1 was identified as a damaging neuroinflammatory factor. These data help suggest complement activation causes glaucomatous neurodegeneration through multiple mechanisms, including inflammation. Microglia and infiltrating myeloid cells expressed high levels of C3ar1 and are the primary candidates to mediate its effects. C3AR1 appeared to be a major regulator of microglia reactivity and neuroinflammatory function due to its interaction with IL10 signaling and other immune related pathways. Targeting myeloid-derived cells and C3AR1 signaling with therapies is expected to add to or improve neuroprotective therapeutic strategies.


Asunto(s)
Degeneración Nerviosa/metabolismo , Nervio Óptico/metabolismo , Receptores de Complemento/biosíntesis , Receptores de Complemento/deficiencia , Animales , Animales Recién Nacidos , Células Cultivadas , Femenino , Redes Reguladoras de Genes/fisiología , Masculino , Ratones , Ratones Endogámicos DBA , Ratones Noqueados , Ratones Transgénicos , Degeneración Nerviosa/genética , Degeneración Nerviosa/patología , Nervio Óptico/patología , Receptores de Complemento/genética
5.
BMJ Open Diabetes Res Care ; 7(1): e000817, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31798904

RESUMEN

Objective: Diabetic nephropathy (DN) is the leading cause of chronic kidney disease and end-stage renal disease. Emerging evidence suggests that complement activation is involved in the pathogenesis of DN. The aim of this study was to investigate the pathogenic role of C3a and C3a receptor (C3aR) in DN. Research design and methods: The expression of C3aR was examined in the renal specimen of patients with DN. Using a C3aR gene knockout mice (C3aR-/-), we evaluated kidney injury in diabetic mice. The mouse gene expression microarray was performed to further explore the pathogenic role of C3aR. Then the underlying mechanism was investigated in vitro with macrophage treated with C3a. Results: Compared with normal controls, the renal expression of C3aR was significantly increased in patients with DN. C3aR-/- diabetic mice developed less severe diabetic renal damage compared with wild-type (WT) diabetic mice, exhibiting significantly lower level of albuminuria and milder renal pathological injury. Microarray profiling uncovered significantly suppressed inflammatory responses and T-cell adaptive immunity in C3aR-/- diabetic mice compared with WT diabetic mice, and this result was further verified by immunohistochemical staining of renal CD4+, CD8+ T cells and macrophage infiltration. In vitro study demonstrated C3a can enhance macrophage-secreted cytokines which could induce inflammatory responses and differentiation of T-cell lineage. Conclusions: C3aR deficiency could attenuate diabetic renal damage through suppressing inflammatory responses and T-cell adaptive immunity, possibly by influencing macrophage-secreted cytokines. Thus, C3aR may be a promising therapeutic target for DN.


Asunto(s)
Inmunidad Adaptativa/inmunología , Activación de Complemento/inmunología , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Tipo 2/complicaciones , Nefropatías Diabéticas/prevención & control , Receptores de Complemento/deficiencia , Animales , Biomarcadores/análisis , Citocinas/metabolismo , Nefropatías Diabéticas/etiología , Nefropatías Diabéticas/patología , Humanos , Riñón/inmunología , Riñón/metabolismo , Riñón/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Pronóstico , Receptores de Complemento/fisiología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/patología
6.
Biochem Biophys Res Commun ; 516(3): 858-865, 2019 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-31266632

RESUMEN

The innate immune response contributes to hepatic steatosis and nonalcoholic fatty liver disease (NAFLD). However, the pathogenic mechanism of NAFLD is still poorly understood. The costimulatory molecule V-set and immunoglobulin domain-containing protein-4 (Vsig4), which is exclusively expressed on macrophages, shows significant role in regulating macrophage-mediated inflammation. Here, we attempted to explore if Vsig4 expression was involved in high fat diet (HFD)-induced NAFLD. The results indicated that Vsig4 expression was markedly down-regulated in fatty livers of NAFLD patients and obese mice. Vsig4 knockout accelerated HFD-induced metabolic dysfunction. In addition, the loss of Vsig4 significantly promoted insulin resistance and lipid deposition in liver samples of HFD-challenged mice. Furthermore, HFD-induced inflammation was apparently accelerated in Vsig4 knockout mice by further activating nuclear factor-κB (NF-κB) signaling pathway. Also, Vsig4 deficient mice exhibited greater collagen accumulation in hepatic samples in HFD-challenged mice compared to the WT mice, which was through promoting transforming growth factor-ß1 (TGFß1) signaling. Importantly, we found that lipopolysaccharide (LPS)- or TGFß1-stimulated inflammation and fibrosis in primary hepatocytes and hepatic stellate cells, respectively, were markedly exacerbated by co-culture with condition medium from bone marrow-derived macrophages (BMDMs) with Vsig4 deficiency. Finally, transplantation of bone marrow cells from control mice to Vsig4-knockout mice restored the severity of steatosis, inflammation and fibrosis after HFD feeding. Therefore, loss of Vsig4 accelerated the severity of lipid deposition, fibrosis and the inflammatory response. Vsig4 could be a therapeutic target for NAFLD treatment.


Asunto(s)
Cirrosis Hepática/genética , Macrófagos/inmunología , Enfermedad del Hígado Graso no Alcohólico/genética , Obesidad/genética , Receptores de Complemento/genética , Animales , Trasplante de Médula Ósea , Colágeno/genética , Colágeno/inmunología , Dieta Alta en Grasa/efectos adversos , Regulación de la Expresión Génica , Células Estrelladas Hepáticas/inmunología , Células Estrelladas Hepáticas/patología , Hepatocitos/inmunología , Hepatocitos/patología , Humanos , Inmunidad Innata , Inflamación , Resistencia a la Insulina , Cirrosis Hepática/etiología , Cirrosis Hepática/inmunología , Cirrosis Hepática/terapia , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/patología , Enfermedad del Hígado Graso no Alcohólico/terapia , Obesidad/etiología , Obesidad/patología , Obesidad/terapia , Cultivo Primario de Células , Receptores de Complemento/deficiencia , Receptores de Complemento/inmunología , Transducción de Señal , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/inmunología
7.
Neuromolecular Med ; 21(4): 467-473, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31102134

RESUMEN

The complement system is a key regulator of the innate immune response against diseased tissue that functions across multiple organ systems. Dysregulation of complement contributes to the pathogenesis of a number of neurological diseases including stroke. The C3a anaphylatoxin, via its cognate C3a receptor (C3aR), mediates inflammation by promoting breakdown of the blood-brain barrier and the massive infiltration of leukocytes into ischemic brain in experimental stroke models. Studies utilizing complement deficient mice as well as pharmacologic C3aR antagonists have shown a reduction in tissue injury and mortality in murine stroke models. The development of tissue-specific C3aR knockout mice and more specific C3aR antagonists is warranted to facilitate our understanding of the role of the C3aR in brain ischemia with the ultimate goal of clinical translation of therapies targeting C3aR in stroke patients.


Asunto(s)
Complemento C3a/fisiología , Neuroinmunomodulación , Receptores de Complemento/fisiología , Accidente Cerebrovascular/inmunología , Animales , Arginina/análogos & derivados , Arginina/uso terapéutico , Compuestos de Bencidrilo/uso terapéutico , Barrera Hematoencefálica , Activación de Complemento , Inactivadores del Complemento/uso terapéutico , Modelos Animales de Enfermedad , Humanos , Inmunidad Innata , Ratones , Ratones Noqueados , Fármacos Neuroprotectores/uso terapéutico , Receptores de Complemento/antagonistas & inhibidores , Receptores de Complemento/deficiencia , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/epidemiología , Accidente Cerebrovascular/fisiopatología , Investigación Biomédica Traslacional
8.
Am J Transplant ; 19(6): 1628-1640, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30565852

RESUMEN

Activation, differentiation, and expansion of alloreactive CD8+ T cells, the dominant effectors that mediate murine heart allograft rejection, requires allorecognition, costimulation, and cytokine-initiated signals. While previous work showed that alloreactive CD4+ T cell immunity entails immune cell-produced and locally activated complement, whether and how C3a receptor 1 (C3aR1) signaling impacts transplant outcomes and the mechanisms linking C3aR1 to alloreactive CD8+ T cell activation/expansion remain unclear. Herein we show that recipient C3aR1 deficiency or pharmacological C3aR1 blockade synergizes with tacrolimus to significantly prolong allograft survival versus tacrolimus-treated controls (median survival time 21 vs. 14 days, P < .05). Recipient C3aR1-deficiency reduced the frequencies of posttransplant, donor-reactive CD8+ T cells twofold. Reciprocal adoptive transfers of naive WT or C3ar1-/- CD8+ T cells into syngeneic WT or C3ar1-/- allograft recipients showed that T cell-expressed C3aR1 induces CD8+ T proliferation, mTOR activation and transcription factor T-bet expression. Host C3aR1 indirectly facilitates alloreactive CD8+ T cell proliferation/expansion by amplifying antigen presenting cell costimulatory molecule expression and innate cytokine production. In addition to expanding mechanistic insight, our findings identify C3aR1 as a testable therapeutic target for future studies aimed at improving human transplant outcomes.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Trasplante de Corazón , Receptores de Complemento/deficiencia , Traslado Adoptivo , Aloinjertos , Animales , Linfocitos T CD8-positivos/citología , Diferenciación Celular/inmunología , Citocinas/biosíntesis , Femenino , Supervivencia de Injerto/inmunología , Humanos , Inmunosupresores/administración & dosificación , Isoantígenos , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Receptores de Complemento/genética , Receptores de Complemento/inmunología , Transducción de Señal/inmunología , Tacrolimus/administración & dosificación
9.
Mol Immunol ; 101: 176-181, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30449309

RESUMEN

The complement system of innate immunity is emerging as a novel player in neurodevelopmental processes. The receptor for C3a, C3aR, shares a close evolutionary and functional relationship with C5a receptors. Whilst the C5a receptor, C5aR1, has been demonstrated to promote embryonic neural stem cell proliferation, little is known about the role of C3aR in this process. Here we show that C3aR is expressed in a similar manner to C5aR1 in mice, at the apical pole of the embryonic ventricular zone, though it has an opposing function. Using in utero delivery of C3aR agonist and antagonist compounds to the embryonic ventricle, we demonstrate that C3aR functions to decrease proliferation of apical neural progenitor cells (NPC). Intriguingly, C3aR-/- animals also have altered NPC proliferation, but demonstrate an opposing phenotype to animals subjected to pharmacological blockade of C3aR. Finally, despite a grossly normal development of C3aR-/- animals, cognitive behavioural testing of adult mice showed subtle deficits in recall memory. These data demonstrate that in addition to C5a, C3a also has a critical role in the normal development of the mammalian brain.


Asunto(s)
Cognición , Embrión de Mamíferos/citología , Células-Madre Neurales/metabolismo , Receptores de Complemento/metabolismo , Animales , Conducta Animal , Proliferación Celular , Células Cultivadas , Memoria Inmunológica , Masculino , Ratones Endogámicos C57BL , Células-Madre Neurales/citología , Receptores de Complemento/deficiencia , Transducción de Señal
10.
Circulation ; 138(16): 1720-1735, 2018 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-29802205

RESUMEN

BACKGROUND: Platelets have distinct roles in the vascular system in that they are the major mediator of thrombosis, critical for restoration of tissue integrity, and players in vascular inflammatory conditions. In close spatiotemporal proximity, the complement system acts as the first line of defense against invading microorganisms and is a key mediator of inflammation. Whereas the fluid phase cross-talk between the complement and coagulation systems is well appreciated, the understanding of the pathophysiological implications of such interactions is still scant. METHODS: We analyzed coexpression of the anaphylatoxin receptor C3aR with activated glycoprotein IIb/IIIa on platelets of 501 patients with coronary artery disease using flow cytometry; detected C3aR expression in human or murine specimen by polymerase chain reaction, immunofluorescence, Western blotting, or flow cytometry; and examined the importance of platelet C3aR by various in vitro platelet function tests, in vivo bleeding time, and intravital microscopy. The pathophysiological relevance of C3aR was scrutinized with the use of disease models of myocardial infarction and stroke. To approach underlying molecular mechanisms, we identified the platelet small GTPase Rap1b using nanoscale liquid chromatography coupled to tandem mass spectrometry. RESULTS: We found a strong positive correlation of platelet complement C3aR expression with activated glycoprotein IIb/IIIa in patients with coronary artery disease and coexpression of C3aR with glycoprotein IIb/IIIa in thrombi obtained from patients with myocardial infarction. Our results demonstrate that the C3a/C3aR axis on platelets regulates distinct steps of thrombus formation such as platelet adhesion, spreading, and Ca2+ influx. Using C3aR-/- mice or C3-/- mice with reinjection of C3a, we uncovered that the complement activation fragment C3a regulates bleeding time after tail injury and thrombosis. Notably, C3aR-/- mice were less prone to experimental stroke and myocardial infarction. Furthermore, reconstitution of C3aR-/- mice with C3aR+/+ platelets and platelet depletion experiments demonstrated that the observed effects on thrombosis, myocardial infarction, and stroke were specifically caused by platelet C3aR. Mechanistically, C3aR-mediated signaling regulates the activation of Rap1b and thereby bleeding arrest after injury and in vivo thrombus formation. CONCLUSIONS: Overall, our findings uncover a novel function of the anaphylatoxin C3a for platelet function and thrombus formation, highlighting a detrimental role of imbalanced complement activation in cardiovascular diseases.


Asunto(s)
Coagulación Sanguínea , Plaquetas/metabolismo , Inmunidad Innata , Infarto del Miocardio/sangre , Receptores de Complemento/sangre , Accidente Cerebrovascular/sangre , Trombosis/sangre , Animales , Plaquetas/inmunología , Señalización del Calcio , Activación de Complemento , Complemento C3/genética , Complemento C3/inmunología , Complemento C3/metabolismo , Modelos Animales de Enfermedad , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Infarto del Miocardio/inmunología , Activación Plaquetaria , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Complejo GPIb-IX de Glicoproteína Plaquetaria/genética , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Receptores de Complemento/deficiencia , Receptores de Complemento/genética , Receptores de Complemento/inmunología , Accidente Cerebrovascular/inmunología , Trombosis/inmunología
11.
Rheumatology (Oxford) ; 57(8): 1400-1407, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29718374

RESUMEN

Objectives: The aim was to describe the clinical characteristics and epidemiology of hypocomplementaemic urticarial vasculitis (HUV; anti-C1q vasculitis) in two geographically defined areas of Sweden. Methods: In the health-care districts surrounding Skåne University Hospital (mean population 950 560) and Linköping University Hospital (mean population 428 503), all incident cases of HUV residing within the study areas at the onset of disease were identified during the years 2000-15. The diagnosis of HUV was confirmed by review of medical records. Only patients meeting the proposed diagnostic HUV criteria and/or the 2012 Chapel Hill consensus definitions in combination with an ever-positive anti-C1q antibody test were included. Results: Sixteen patients (14 females) were identified during the study period. The median (interquartile range) age at diagnosis was 51 (40.7-56.7) years. Median (interquartile range) time of follow-up from diagnosis to 31 December 2015, or death, was 94 (46.5-136.2) months. The most frequent manifestations at diagnosis were urticaria (100%), arthritis (88%), followed by biopsy-proven glomerulonephritis (19%), episcleritis/scleritis (19%) and recurrent abdominal pain (13%). The annual incidence rate per million inhabitants was estimated as 0.7 (95% CI: 0.4, 1.1). Sixty-three per cent suffered from pulmonary disease at the last follow-up. Two patients died during the follow-up period. One patient underwent lung transplantation, and two patients proceeded to end-stage renal disease. The point prevalence on 31 December 2015 was 9.5/million (95% CI: 4.5, 14.5). Conclusion: Hypocomplementaemic urticarial vasculitis constitutes a rare, but not always benign condition. Renal and lung manifestations were severe in some cases, highlighting the need for careful screening and monitoring of this potentially serious condition.


Asunto(s)
Glicoproteínas de Membrana/deficiencia , Vigilancia de la Población , Receptores de Complemento/deficiencia , Urticaria/epidemiología , Vasculitis/epidemiología , Adulto , Anciano , Femenino , Estudios de Seguimiento , Humanos , Incidencia , Masculino , Glicoproteínas de Membrana/sangre , Persona de Mediana Edad , Receptores de Complemento/sangre , Estudios Retrospectivos , Suecia/epidemiología , Urticaria/sangre , Urticaria/complicaciones , Vasculitis/sangre , Vasculitis/etiología
12.
PLoS One ; 13(12): e0208559, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30596651

RESUMEN

Neuroinflammation and neurodegeneration are common during prion infection, but the mechanisms that underlie these pathological features are not well understood. Several components of innate immunity, such as Toll-like receptor (TLR) 4 and Complement C1q, have been shown to influence prion disease. To identify additional components of innate immunity that might impact prion disease within the central nervous system (CNS), we screened RNA from brains of pre-clinical and clinical 22L-infected mice for alterations in genes associated with innate immunity. Transcription of several genes encoding damage-associated molecular pattern (DAMP) proteins and receptors were increased in the brains of prion-infected mice. To investigate the role of some of these proteins in prion disease of the CNS, we infected mice deficient in DAMP receptor genes Tlr2, C3ar1, and C5ar1 with 22L scrapie. Elimination of TLR2 accelerated disease by a median of 10 days, while lack of C3aR1 or C5aR1 had no effect on disease tempo. Histopathologically, all knockout mouse strains tested were similar to infected control mice in gliosis, vacuolation, and PrPSc deposition. Analysis of proinflammatory markers in the brains of infected knockout mice indicated only a few alterations in gene expression suggesting that C5aR1 and TLR2 signaling did not act synergistically in the brains of prion-infected mice. These results indicate that signaling through TLR2 confers partial neuroprotection during prion infection.


Asunto(s)
Neuroprotección , Enfermedades por Prión/patología , Receptor Toll-Like 2/metabolismo , Anafilatoxinas/análisis , Animales , Encéfalo/metabolismo , Encéfalo/patología , Quimiocinas/metabolismo , Proteínas del Sistema Complemento/metabolismo , Citocinas/metabolismo , Susceptibilidad a Enfermedades , Expresión Génica , Inmunidad Innata/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedades por Prión/metabolismo , Enfermedades por Prión/veterinaria , ARN/genética , ARN/metabolismo , Receptor de Anafilatoxina C5a/deficiencia , Receptor de Anafilatoxina C5a/genética , Receptor de Anafilatoxina C5a/metabolismo , Receptores de Complemento/deficiencia , Receptores de Complemento/genética , Receptores de Complemento/metabolismo , Índice de Severidad de la Enfermedad , Transducción de Señal , Receptor Toll-Like 2/deficiencia , Receptor Toll-Like 2/genética
13.
Nat Commun ; 8(1): 2078, 2017 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-29233958

RESUMEN

Regeneration of skeletal muscle following injury is accompanied by transient inflammation. Here we show that complement is activated in skeletal muscle injury and plays a key role during regeneration. Genetic ablation of complement C3 or its inactivation with Cobra Venom Factor (CVF) result in impaired muscle regeneration following cardiotoxin-induced injury in mice. The effect of complement in muscle regeneration is mediated by the alternative pathway and C3a receptor (C3aR) signaling, as deletion of Cfb, a key alternative pathway component, or C3aR leads to impaired regeneration and reduced monocyte/macrophage infiltration. Monocytes from C3aR-deficient mice express a reduced level of adhesion molecules, cytokines and genes associated with antigen processing and presentation. Exogenous administration of recombinant CCL5 to C3aR-deficient mice rescues the defects in inflammatory cell recruitment and regeneration. These findings reveal an important role of complement C3a in skeletal muscle regeneration, and suggest that manipulating complement system may produce therapeutic benefit in muscle injury and regeneration.


Asunto(s)
Complemento C3a/fisiología , Inflamación/inmunología , Monocitos/fisiología , Músculo Esquelético/fisiología , Receptores de Complemento/fisiología , Regeneración/inmunología , Animales , Trasplante de Médula Ósea , Cardiotoxinas/toxicidad , Movimiento Celular/fisiología , Células Cultivadas , Quimiocina CCL5/metabolismo , Quimera/fisiología , Complemento C3a/antagonistas & inhibidores , Vía Alternativa del Complemento/fisiología , Modelos Animales de Enfermedad , Venenos Elapídicos/farmacología , Humanos , Macrófagos/fisiología , Masculino , Ratones , Ratones Noqueados , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/lesiones , Receptores de Complemento/deficiencia , Regeneración/efectos de los fármacos , Transducción de Señal/fisiología , Cicatrización de Heridas/fisiología
14.
Nat Commun ; 8(1): 1322, 2017 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-29109438

RESUMEN

Exacerbation of macrophage-mediated inflammation contributes to pathogenesis of various inflammatory diseases, but the immunometabolic programs underlying regulation of macrophage activation are unclear. Here we show that V-set immunoglobulin-domain-containing 4 (VSIG4), a B7 family-related protein that is expressed by resting macrophages, inhibits macrophage activation in response to lipopolysaccharide. Vsig4 -/- mice are susceptible to high-fat diet-caused obesity and murine hepatitis virus strain-3 (MHV-3)-induced fulminant hepatitis due to excessive macrophage-dependent inflammation. VSIG4 activates the PI3K/Akt-STAT3 pathway, leading to pyruvate dehydrogenase kinase-2 (PDK2) upregulation and subsequent phosphorylation of pyruvate dehydrogenase, which results in reduction in pyruvate/acetyl-CoA conversion, mitochondrial reactive oxygen species secretion, and macrophage inhibition. Conversely, interruption of Vsig4 or Pdk2 promotes inflammation. Forced expression of Vsig4 in mice ameliorates MHV-3-induced viral fulminant hepatitis. These data show that VSIG4 negatively regulates macrophage activation by reprogramming mitochondrial pyruvate metabolism.


Asunto(s)
Activación de Macrófagos/fisiología , Ácido Pirúvico/metabolismo , Receptores de Complemento/metabolismo , Animales , Infecciones por Coronavirus/etiología , Dieta Alta en Grasa/efectos adversos , Células HEK293 , Hepatitis Viral Animal/etiología , Humanos , Inflamación/etiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Virus de la Hepatitis Murina , Obesidad/etiología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , Células RAW 264.7 , Especies Reactivas de Oxígeno/metabolismo , Receptores de Complemento/deficiencia , Receptores de Complemento/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Células THP-1
15.
Diabetes ; 65(10): 2888-99, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27659228

RESUMEN

Type 2 diabetes and cardiovascular disease are complex disorders involving metabolic and inflammatory mechanisms. Here we investigated whether sCD93, a group XIV c-type lectin of the endosialin family, plays a role in metabolic dysregulation or carotid intima-media thickness (IMT). Although no association was observed between sCD93 and IMT, sCD93 levels were significantly lower in subjects with type 2 diabetes (n = 901, mean ± SD 156.6 ± 40.0 ng/mL) compared with subjects without diabetes (n = 2,470, 164.1 ± 44.8 ng/mL, P < 0.0001). Genetic variants associated with diabetes risk (DIAGRAM Consortium) did not influence sCD93 levels (individually or combined in a single nucleotide polymorphism score). In a prospective cohort, lower sCD93 levels preceded the development of diabetes. Consistent with this, a cd93-deficient mouse model (in addition to apoe deficiency) demonstrated no difference in atherosclerotic lesion development compared with apoe(-/-) cd93-sufficient littermates. However, cd93-deficient mice showed impaired glucose clearance and insulin sensitivity (compared with littermate controls) after eating a high-fat diet. The expression of cd93 was observed in pancreatic islets, and leaky vessels were apparent in cd93-deficient pancreases. We further demonstrated that stress-induced release of sCD93 is impaired by hyperglycemia. Therefore, we propose CD93 as an important component in glucometabolic regulation.


Asunto(s)
Grosor Intima-Media Carotídeo , Glicoproteínas de Membrana/metabolismo , Receptores de Complemento/metabolismo , Animales , Apolipoproteínas E , Aterosclerosis/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Células Endoteliales/metabolismo , Femenino , Genotipo , Humanos , Masculino , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Páncreas/metabolismo , Receptores de Complemento/deficiencia , Receptores de Complemento/genética
16.
J Immunol ; 196(11): 4783-92, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27183625

RESUMEN

The complement peptide C3a is a key component of the innate immune system and a major fragment produced following complement activation. We used a murine model of melanoma (B16-F0) to identify a hitherto unknown role for C3a-C3aR signaling in promoting tumor growth. The results show that the development and growth of B16-F0 melanomas is retarded in mice lacking C3aR, whereas growth of established melanomas can be arrested by C3aR antagonism. Flow cytometric analysis showed alterations in tumor-infiltrating leukocytes in the absence of C3aR. Specifically, neutrophils and CD4(+) T lymphocyte subpopulations were increased, whereas macrophages were reduced. The central role of neutrophils was confirmed by depletion experiments that reversed the tumor inhibitory effects observed in C3aR-deficient mice and returned tumor-infiltrating CD4(+) T cells to control levels. Analysis of the tumor microenvironment showed upregulation of inflammatory genes that may contribute to the enhanced antitumor response observed in C3aR-deficient mice. C3aR deficiency/inhibition was also protective in murine models of BRAF(V600E) mutant melanoma and colon and breast cancer, suggesting a tumor-promoting role for C3aR signaling in a range of tumor types. We propose that C3aR activation alters the tumor inflammatory milieu, thereby promoting tumor growth. Therapeutic inhibition of C3aR may therefore be an effective means to trigger an antitumor response in melanoma and other cancers.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Carcinogénesis/inmunología , Melanoma/inmunología , Melanoma/patología , Neutrófilos/inmunología , Receptores de Complemento/inmunología , Animales , Linfocitos T CD4-Positivos/patología , Células Cultivadas , Femenino , Melanoma/genética , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Neutrófilos/patología , Receptores de Complemento/deficiencia
17.
J Neuroinflammation ; 13: 23, 2016 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-26822321

RESUMEN

BACKGROUND: The complement system is becoming increasingly recognized as a key participant in many neurodegenerative diseases of the brain. Complement-deficient animals exhibit reduced neuroinflammation. METHODS: In the present study, we administered intracerebroventricularly lipopolysaccharide (LPS) to mimic local infection of the brain and investigated the role of key complement component C3 in brain vasculature endothelial activation and leukocyte recruitment. The degree of neutrophil infiltration was determined by esterase staining. Leukocyte-endothelial interactions were measured using intravital microscopy. Cerebral endothelial activation was evaluated using real-time PCR and Western blotting. RESULTS: Neutrophil infiltration into the brain cortex and hippocampus was significantly reduced in C3(-/-) mice and C3aR(-/-) mice but not in C6(-/-) mice. We detected markedly attenuated leukocyte-endothelial interactions in the brain microvasculature of C3(-/-) mice. Accordingly, in response to LPS administration, the brain microvasculature in these mice had decreased expression of P-selectin, E-selectin, intercellular cell adhesion molecule 1 (ICAM-1), and vascular cell adhesion molecule 1 (VCAM-1). Depletion of C3 from the circulation also caused reduction in VCAM-1 and E-selectin expression and leukocyte recruitment, suggesting that C3 in the circulation contributed to brain endothelial activation. Furthermore, C3(-/-) mice exhibited decreased leukocyte recruitment into the brain upon tumor necrosis factor-α (TNF-α) stimulation. C3a activated the phosphorylation of p38 mitogen-activated protein kinase (MAPK) and nuclear factor-κB (NF-κB) and induced the upregulation of VCAM-1 and ICAM-1 expression in murine primary cerebral endothelial cells in vitro. CONCLUSIONS: Our study provides the first evidence that C3a plays a critical role in cerebral endothelial activation and leukocyte recruitment during inflammation in the brain.


Asunto(s)
Encéfalo/citología , Complemento C3a/metabolismo , Células Endoteliales/fisiología , Leucocitos/fisiología , Infiltración Neutrófila/fisiología , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/fisiología , Encéfalo/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , Adhesión Celular/genética , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Células Cultivadas , Complemento C3a/genética , Células Endoteliales/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Inyecciones Intraventriculares , Leucocitos/efectos de los fármacos , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Microvasos/efectos de los fármacos , Microvasos/fisiología , FN-kappa B/metabolismo , Infiltración Neutrófila/efectos de los fármacos , Receptores de Complemento/deficiencia , Receptores de Complemento/genética , Factores de Tiempo
18.
PLoS One ; 10(11): e0143841, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26619292

RESUMEN

Antigen-specific IgG antibodies, passively administered to mice or humans together with large particulate antigens like erythrocytes, can completely suppress the antibody response against the antigen. This is used clinically in Rhesus prophylaxis, where administration of IgG anti-RhD prevents RhD-negative women from becoming immunized against RhD-positive fetal erythrocytes aquired transplacentally. The mechanisms by which IgG suppresses antibody responses are poorly understood. We have here addressed whether complement or Fc-receptors for IgG (FcγRs) are required for IgG-mediated suppression. IgG, specific for sheep red blood cells (SRBC), was administered to mice together with SRBC and the antibody responses analyzed. IgG was able to suppress early IgM- as well as longterm IgG-responses in wildtype mice equally well as in mice lacking FcγRIIB (FcγRIIB knockout mice) or FcγRI, III, and IV (FcRγ knockout mice). Moreover, IgG was able to suppress early IgM responses equally well in mice lacking C1q (C1qA knockout mice), C3 (C3 knockout mice), or complement receptors 1 and 2 (Cr2 knockout mice) as in wildtype mice. Owing to the previously described severely impaired IgG responses in the complement deficient mice, it was difficult to assess whether passively administered IgG further decreased their IgG response. In conclusion, Fc-receptor binding or complement-activation by IgG does not seem to be required for its ability to suppress antibody responses to xenogeneic erythrocytes.


Asunto(s)
Complemento C1q/deficiencia , Complemento C3/deficiencia , Inmunoglobulina G/administración & dosificación , Receptores de Complemento/deficiencia , Receptores de IgG/metabolismo , Ovinos/sangre , Animales , Formación de Anticuerpos , Transfusión de Eritrocitos , Técnicas de Inactivación de Genes , Inmunización/métodos , Inmunoglobulina M/metabolismo , Ratones , Ovinos/inmunología
19.
Mucosal Immunol ; 8(4): 874-85, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25465103

RESUMEN

Exposure to cigarette smoke can initiate sterile inflammatory responses in the lung and activate myeloid dendritic cells (mDCs) that induce differentiation of T helper type 1 (Th1) and Th17 cells in the emphysematous lungs. Consumption of complement proteins increases in acute inflammation, but the contribution of complement protein 3 (C3) to chronic cigarette smoke-induced immune responses in the lung is not clear. Here, we show that following chronic exposure to cigarette smoke, C3-deficient (C3(-/-)) mice develop less emphysema and have fewer CD11b(+)CD11c(+) mDCs infiltrating the lungs as compared with wild-type mice. Proteolytic cleavage of C3 by neutrophil elastase releases C3a, which in turn increases the expression of its receptor (C3aR) on lung mDCs. Mice deficient in the C3aR (C3ar(-/-)) partially phenocopy the attenuated responses to chronic smoke observed in C3(-/-) mice. Consistent with a role for C3 in emphysema, C3 and its active fragments are deposited on the lung tissue of smokers with emphysema, and smoke-exposed mice. Together, these findings suggest a critical role for C3a through autocrine/paracrine induction of C3aR in the pathogenesis of cigarette smoke-induced sterile inflammation and provide new therapeutic targets for the treatment of emphysema.


Asunto(s)
Enfisema/etiología , Enfisema/metabolismo , Receptores de Complemento/metabolismo , Fumar/efectos adversos , Animales , Comunicación Autocrina , Quimiotaxis de Leucocito/genética , Quimiotaxis de Leucocito/inmunología , Activación de Complemento , Complemento C3/genética , Complemento C3/inmunología , Complemento C3/metabolismo , Complemento C3a/inmunología , Complemento C3a/metabolismo , Modelos Animales de Enfermedad , Enfisema/diagnóstico , Regulación de la Expresión Génica , Humanos , Elastasa de Leucocito/metabolismo , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/patología , Metaloproteinasa 12 de la Matriz/metabolismo , Ratones Noqueados , Comunicación Paracrina , Proteolisis , Receptores de Complemento/deficiencia , Receptores de Complemento/genética , Transducción de Señal
20.
J Immunol ; 193(3): 1278-89, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24981453

RESUMEN

Listeria monocytogenes is a Gram-positive intracellular bacterium that is acquired through tainted food and may lead to systemic infection and possible death. Despite the importance of the innate immune system in fighting L. monocytogenes infection, little is known about the role of complement and its activation products, including the potent C3a anaphylatoxin. In a model of systemic L. monocytogenes infection, we show that mice lacking the receptor for C3a (C3aR(-/-)) are significantly more sensitive to infection compared with wild-type mice, as demonstrated by decreased survival, increased bacterial burden, and increased damage to their livers and spleens. The inability of the C3aR(-/-) mice to clear the bacterial infection was not caused by defective macrophages or by a reduction in cytokines/chemokines known to be critical in the host response to L. monocytogenes, including IFN-γ and TNF-α. Instead, TUNEL staining, together with Fas, active caspase-3, and Bcl-2 expression data, indicates that the increased susceptibility of C3aR(-/-) mice to L. monocytogenes infection was largely caused by increased L. monocytogenes-induced apoptosis of myeloid and lymphoid cells in the spleen that are required for ultimate clearance of L. monocytogenes, including neutrophils, macrophages, dendritic cells, and T cells. These findings reveal an unexpected function of C3a/C3aR signaling during the host immune response that suppresses Fas expression and caspase-3 activity while increasing Bcl-2 expression, thereby providing protection to both myeloid and lymphoid cells against L. monocytogenes-induced apoptosis.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/fisiología , Apoptosis/inmunología , Complemento C3a/metabolismo , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Listeriosis/patología , Receptores de Complemento/fisiología , Animales , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/deficiencia , Proteínas Reguladoras de la Apoptosis/genética , Caspasa 3/metabolismo , Inhibidores de Caspasas/farmacología , Complemento C3a/inmunología , Modelos Animales de Enfermedad , Regulación hacia Abajo/genética , Regulación hacia Abajo/inmunología , Inmunosupresores/farmacología , Inmunosupresores/uso terapéutico , Listeria monocytogenes/patogenicidad , Listeriosis/genética , Linfocitos/inmunología , Linfocitos/metabolismo , Linfocitos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/inmunología , Células Mieloides/metabolismo , Células Mieloides/patología , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Receptores de Complemento/deficiencia , Receptores de Complemento/genética , Transducción de Señal/genética , Transducción de Señal/inmunología , Regulación hacia Arriba/genética , Regulación hacia Arriba/inmunología , Receptor fas/antagonistas & inhibidores , Receptor fas/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA