Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Immunology ; 170(2): 180-192, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37222083

RESUMEN

The complement system, an arm of the innate immune system plays a critical role in both health and disease. The complement system is highly complex with dual possibilities, helping or hurting the host, depending on the location and local microenvironment. The traditionally known functions of complement include surveillance, pathogen recognition, immune complex trafficking, processing and pathogen elimination. The noncanonical functions of the complement system include their roles in development, differentiation, local homeostasis and other cellular functions. Complement proteins are present in both, the plasma and on the membranes. Complement activation occurs both extra- and intracellularly, which leads to considerable pleiotropy in their activity. In order to design more desirable and effective therapies, it is important to understand the different functions of complement, and its location-based and tissue-specific responses. This manuscript will provide a brief overview into the complex nature of the complement cascade, outlining some of their complement-independent functions, their effects at different locale, and their implication in disease settings.


Asunto(s)
Activación de Complemento , Proteínas del Sistema Complemento
2.
J Immunol ; 210(2): 119-125, 2023 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-36596217

RESUMEN

The complement field has recently experienced a strong resurgence of interest because of the unexpected discovery of new complement functions extending complement's role beyond immunity and pathogen clearance, a growing list of diseases in which complement plays a role, and the proliferation of complement therapeutics. Importantly, although the majority of complement components in the circulation are generated by the liver and activated extracellularly, complement activation unexpectedly also occurs intracellularly across a broad range of cells. Such cell-autonomous complement activation can engage intracellular complement receptors, which then drive noncanonical cell-specific effector functions. Thus, much remains to be discovered about complement biology. In this brief review, we focus on novel noncanonical activities of complement in its "classic areas of operation" (kidney and brain biology, infection, and autoimmunity), with an outlook on the next generation of complement-targeted therapeutics.


Asunto(s)
Activación de Complemento , Proteínas del Sistema Complemento
4.
J Immunol Res ; 2022: 1737419, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35097132

RESUMEN

Complement factor H (FH) is a key regulator of the alternative pathway of complement, in man and mouse. Earlier, our studies revealed that the absence of FH causes the C57BL6 mouse to become susceptible to chronic serum sickness (CSS) along with an increase in the renal infiltration of macrophages compared to controls. To understand if the increased recruitment of macrophages (Mϕs) to the kidney was driving inflammation and propagating injury, we examined the effect of Mϕ depletion with clodronate in FH knockout mice with CSS. Eight-week-old FHKO mice were treated with apoferritin (4 mg/mouse) for 5 wks and with either vehicle (PBS) or clodronate (50 mg/kg ip, 3 times/wk for the last 3 weeks). The administration of clodronate decreased monocytes and Mϕs in the kidneys by >80%. Kidney function assessed by BUN and albumin remained closer to normal on depletion of Mϕs. Clodronate treatment prevented the alteration in cytokines, TNFα and IL-6, and increase in gene expression of connective tissue growth factor (CTGF), TGFß-1, matrix metalloproteinase-9 (MMP9), fibronectin, laminin, and collagen in FHKO mice with CSS (P < 0.05). Clodronate treatment led to relative protection from immune complex- (IC-) mediated disease pathology during CSS as assessed by the significantly reduced glomerular pathology (GN) and extracellular matrix. Our results suggest that complement activation is one of the mechanism that regulates the macrophage landscape and thereby fibrosis. The exact mechanism remains to be deciphered. In brief, our data shows that Mϕs play a critical role in FH-dependent ICGN and Mϕ depletion reduces disease progression.


Asunto(s)
Glomerulonefritis/inmunología , Enfermedades del Complejo Inmune/inmunología , Riñón/metabolismo , Macrófagos/inmunología , Animales , Apoferritinas/administración & dosificación , Movimiento Celular , Ácido Clodrónico/administración & dosificación , Factor H de Complemento/metabolismo , Progresión de la Enfermedad , Fibrosis , Riñón/inmunología , Riñón/patología , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
5.
Kidney Int ; 100(4): 824-836, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34139209

RESUMEN

Factor H (FH) is a critical regulator of the alternative complement pathway and its deficiency or mutation underlie kidney diseases such as dense deposit disease. Since vascular dysfunction is an important facet of kidney disease, maintaining optimal function of the lining endothelial cells is important for vascular health. To investigate the molecular mechanisms that are regulated by FH in endothelial cells, FH deficient and sufficient mouse kidney endothelial cell cultures were established. Endothelial FH deficiency resulted in cytoskeletal remodeling, increased angiogenic potential, loss of cellular layer integrity and increased cell proliferation. FH reconstitution prevented these FH-dependent proliferative changes. Respiratory flux analysis showed reduced basal mitochondrial respiration, ATP production and maximal respiratory capacity in FH deficient endothelial cells, while proton leak remained unaltered. Similar changes were observed in FH deficient human glomerular endothelial cells indicating the translational potential of these studies. Gene expression analysis revealed that the FH-dependent gene changes in mouse kidney endothelial cells include significant upregulation of genes involved in inflammation and the complement system. The transcription factor nuclear factor-kB, that regulates many biological processes, was translocated from the cytoplasm to the nucleus in the absence of FH. Thus, our studies show the functional relevance of intrinsic FH in kidney endothelial cells in man and mouse.


Asunto(s)
Factor H de Complemento , Enfermedades Renales , Animales , Factor H de Complemento/genética , Vía Alternativa del Complemento , Células Endoteliales , Humanos , Riñón , Ratones
6.
Immunobiology ; 225(5): 152003, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32962822

RESUMEN

Complement (C) system is a double edge sword acting as the first line of defense on the one hand and causing aggravation of disease on the other. C activation when unregulated affects different organs including muscle regeneration. However, the effect of factor H (FH), a critical regulator of the alternative C pathway in muscle remains to be studied. FH deficiency results in excessive C activation and generates proinflammatory fragments C5a and C3a as byproducts. C3a and C5a signal through their respective receptors, C5aR and C3aR. In this study, we investigated the role of FH and downstream C5a/C5aR signaling in muscle architecture and function. Using the FH knockout (fh-/-) and fh-/-/C5aR-/double knockout mice we explored the role of C, specifically the alternative C pathway in muscle dysfunction. Substantial C3 and C9 deposits occur along the walls of the fh-/- muscle fibers indicative of unrestricted C activation. Physical performance assessments of the fh-/- mice show reduced grip endurance (76 %), grip strength (14 %) and rotarod balance (36 %) compared to controls. Histological analysis revealed a shift in muscle fiber populations indicated by an increase in glycolytic MHC IIB fibers and reduction in oxidative MHC IIA fibers. Consistent with this finding, mitochondrial DNA (mtDNA) and citrate synthase (CS) expression were both reduced indicating possible reduction in mitochondrial biomass. In addition, our results showed a significant increase in TGFß expression and altered TGFß localization in this setting. The architecture of cytoskeletal proteins actin and vimentin in the fh-/- muscle was changed that could lead to contractile weakness and loss of skeletal muscle elasticity. The muscle pathology in fh-/- mice was reduced in fh-/-/C5aR-/- double knockout (DKO) mice, highlighting partial C5aR dependence. Our results for the first time demonstrate an important role of FH in physical performance and skeletal muscle health.


Asunto(s)
Complemento C5a/metabolismo , Factor H de Complemento/genética , Músculo Esquelético/metabolismo , Resistencia Física/genética , Receptor de Anafilatoxina C5a/metabolismo , Actinas/metabolismo , Animales , Complemento C3/análisis , Complemento C3/genética , Complemento C5a/análisis , Factor H de Complemento/metabolismo , ADN Mitocondrial , Expresión Génica , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Fatiga Muscular/genética , Fuerza Muscular/genética , Receptor de Anafilatoxina C5a/genética , Prueba de Desempeño de Rotación con Aceleración Constante , Vimentina/metabolismo
7.
Precis Clin Med ; 3(1): 34-43, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32257532

RESUMEN

Systemic lupus erythematosus (SLE) is an autoimmune disease that is a challenge to diagnose and treat. There is an urgent need for biomarkers to help define organ involvement, and more effective therapies. A unique population of T cells, the CD3+CD4-CD8- (DNeg) cells, is significantly increased in lupus patients. Twenty-seven cases (53%) of pediatric SLE patients had elevated DNeg cells in their peripheral blood, which correlated with kidney function (R2  = 0.54). Significant infiltration of DNeg cells was observed in both adult and pediatric lupus kidneys by immunofluorescence. For the first time, this study provides direct evidence that DNeg cells facilitate kidney injury in preclinical 8-week-old MRL/lpr lupus mice. In lupus mice, the increase in DNeg cells tracked with worsening disease and correlated with kidney function (R2  = 0.85). Our results show that DNeg cells per se can cause kidney dysfunction, increase in number with increase in disease pathology, and could serve as a potential biomarker.

8.
J Cell Immunol ; 1(2): 33-42, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31984378

RESUMEN

Systemic lupus erythematosus is an autoimmune disease affecting multiple organs with devastating pathological consequences. Current treatment regimens largely rely on immunosuppressants and corticosteroids to attenuate autoimmune activity. However, such treatments have toxic side effects, often lacks efficacy, and inherently leaves the patient prone to infections, making the discovery of novel biomarkers and therapeutic targets an urgent need. Neutrophil extracellular traps (NETs) that participate in host defense are generated by neutrophils by a process called NETosis. NETs play an important role in the pathogenesis of SLE. In this review, we discuss the current literature regarding the role of NETs in SLE while entertaining the possibility that NETosis could serve as therapeutic targets thereby rendering the treatment more specific and effective in comparison to the current lupus therapy.

9.
Immunobiology ; 223(12): 761-771, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30107932

RESUMEN

Complement system is an important arm of the immune system that promotes inflammation. Complement Factor H (FH) is a critical regulator of the alternative complement pathway. Its absence causes pathology in different organs resulting in diseases such as age related macular degeneration and dense deposit disease. Recent studies suggest that the complement system plays a role in bone development and homeostasis. To determine the role of FH in bone architecture, we studied the FH knockout (fh-/-) mice. 3D reconstructions of femur from 16 week old fh-/- mice reveal significant changes, such as decreased BV/TV (4.5%, p < 0.02), trabecular number (22%, p < 0.01), tissue mineral density (16%, p < 0.04), and increased marrow area (16% p < 0.01), compared to their wild type (WT) counterparts. Kidney function and histology remained normal indicating that bone changes occurred prior to kidney dysfunction. Next we examined cultured osteoblasts and osteoclasts isolated from bone marrow. FH is expressed ubiquitously in the osteoblasts and in the cytoplasm of osteoclasts. The changes caused by absence of FH include: increase in number of osteoblasts (362%) and osteoclasts (342%), increase in RNA (180%) and protein expression of cathepsin K and increased osteoclast function (pit formation, 233%). Actin rearrangement in both osteoblasts and osteoclasts was altered, with a loss of integrity of the F-actin ring at the periphery of the osteoclasts. For the first time our studies demonstrate a direct role of FH in the maintenance of bone structure and function and is highlighted as a promising therapeutic target in bone diseases.


Asunto(s)
Huesos/inmunología , Huesos/metabolismo , Factor H de Complemento/inmunología , Actinas/metabolismo , Animales , Biomarcadores , Resorción Ósea/genética , Resorción Ósea/metabolismo , Resorción Ósea/patología , Huesos/diagnóstico por imagen , Huesos/patología , Factor H de Complemento/genética , Factor H de Complemento/metabolismo , Técnica del Anticuerpo Fluorescente , Inmunohistoquímica , Ratones , Ratones Noqueados , Osteoblastos/inmunología , Osteoblastos/metabolismo , Osteoclastos/inmunología , Osteoclastos/metabolismo , Fenotipo , Microtomografía por Rayos X
10.
Mol Immunol ; 102: 26-31, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30007547

RESUMEN

The brain is an immune privileged organ, uniquely placed in the body. Two systems involved in maintaining brain homeostasis and in protecting the brain are the blood-brain barrier (BBB) and the complement system. The BBB is present in the vasculature of the brain and is the dynamic interface between brain and body that regulates what enters and leaves the brain, thereby maintaining the brain microenvironment optimal for brain function. The complement system is ubiquitous, being present systemically and in the brain, both membrane bound and in circulation. It is an important arm of the body's defense that helps maintain homeostasis by eliminating debris and damaged cells, participating in destroying pathogens, promoting inflammation and conveying 'danger signals'. Recent studies reveal that the complement system plays an important role in normal brain development. However, when the complement system is overwhelmed, complement activation could contribute to loss of BBB integrity resulting in brain pathology. Studies support an association between complement proteins and BBB dysfunction, with the mechanisms being slowly unraveled. This review will provide an overview of both these systems, how they intersect and interact with each other.


Asunto(s)
Barrera Hematoencefálica , Encéfalo , Proteínas del Sistema Complemento , Animales , Activación de Complemento/fisiología , Humanos
12.
Immunobiology ; 223(6-7): 460-465, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29478734

RESUMEN

Microbiota consists of more than 1014 microorganisms that inhabit different areas of the body including the gastrointestinal tract, mainly the mouth and gut. It includes viruses, fungi, protozoa, archaea and bacteria. The microbiota interacts closely with host leading to a dynamic relationship that results in the biological effects observed. Its diverse genetic material (microbiome) interacts closely with the host immune system and cells, and therefore is closely associated with inflammation, immune tolerance, adaptive immunity and autoimmune diseases. Bacterial microbiota, which is the mostly studied lives in harmony with the host and maintains a symbiotic relationship. Therefore it plays an important role in immunological, metabolic, and neurological aspects and thereby the well-being of the host. Alteration of the homeostatic environment or the dynamic balance of microorganisms can result in dysbiosis or disease. However, does dysbiosis cause disease, aggravate disease or is the result of the disease remains to be defined, it could be a bit of all three factors. More recently, a number of studies demonstrate that these microorganisms could contribute to disease. Alteration of the tightly balanced composition of bacterial microbiota (dysbiosis) leads to exacerbation, rapid progression and worsening of disease states. It is important to identify the 'healthy' microbes that maintain a healthy environment, the 'sensitive' microbes that go awry with disease, the 'bad' microbes that cause disease and the 'therapeutic' microbes that can help rectify the changes. Increased relative abundance of certain bacterial species has been linked to triggering autoimmune diseases. Despite the burgeoning literature in the field, the molecular mechanisms by which the microbiota impacts the body in health and disease remain largely unknown. In this review, we will discuss recent advancements in our understanding of the gut bacterial microbiota associated with inflammatory and immunological processes and the role they play in the autoimmune disease, systemic lupus erythematosus.


Asunto(s)
Bacterias , Disbiosis/inmunología , Mucosa Intestinal/microbiología , Lupus Eritematoso Sistémico/microbiología , Microbiota/inmunología , Animales , Autoinmunidad , Homeostasis , Humanos , Tolerancia Inmunológica , Mucosa Intestinal/inmunología , Lupus Eritematoso Sistémico/inmunología
13.
Immunol Invest ; 46(8): 816-832, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29058550

RESUMEN

The complement system which is a critical mediator of innate immunity plays diverse roles in the neuropathogenesis of HIV-1 infection such as clearing HIV-1 and promoting productive HIV-1 replication. In the development of HIV-1 associated neurological disorders (HAND), there may be an imbalance between complement activation and regulation, which may contribute to the neuronal damage as a consequence of HIV-1 infection. It is well recognized that opiate abuse exacerbates HIV-1 neuropathology, however, little is known about the role of complement proteins in opiate induced neuromodulation, specifically in the presence of co-morbidity such as HIV-1 infection. Complement levels are significantly increased in the HIV-1-infected brain, thus HIV-induced complement synthesis may represent an important mechanism for the pathogenesis of AIDS in the brain, but remains underexplored. Anti-HIV-1 antibodies are able to initiate complement activation in HIV-1 infected CNS cells such as microglia and astrocytes during the course of disease progression; however, this complement activation fails to clear and eradicate HIV-1 from infected cells. In addition, the antiretroviral agents used for HIV therapy cause dysregulation of lipid metabolism, endothelial, and adipocyte cell function, and activation of pro-inflammatory cytokines. We speculate that both HIV-1 and opiates trigger a cytokine-mediated pro-inflammatory stimulus that modulates the complement cascade to exacerbate the virus-induced neurological damage. We examined the expression levels of C1q, SC5b-9, C5L2, C5aR, C3aR, and C9 key members of the complement cascade both in vivo in post mortem brain frontal cortex tissue from patients with HAND who used/did not use heroin, and in vitro using human microglial cultures treated with HIV tat and/or heroin. We observed significant expression of C1q and SC5b-9 by immunofluorescence staining in both the brain cortical and hippocampal region in HAND patients who abused heroin. Additionally, we observed increased gene expression of C5aR, C3aR, and C9 in the brain tissue of both HIV-1 infected patients with HAND who abused and did not abuse heroin, as compared to HIV negative controls. Our results show a significant increase in the expression of complement proteins C9, C5L2, C5aR, and C3aR in HIV transfected microglia and an additional increase in the levels of these complement proteins in heroin-treated HIV transfected microglia. This study highlights the a) potential roles of complement proteins in the pathogenesis of HIV-1-related neurodegenerative disorders; b) the combined effect of an opiate, like heroin, and HIV viral protein like HIV tat on complement proteins in normal human microglial cells and HIV transfected microglial cells. In the context of HAND, targeting selective steps in the complement cascade could help ameliorating the HIV burden in the CNS, thus investigations of complement-related therapeutic approaches for the treatment of HAND are warranted.


Asunto(s)
Nefropatía Asociada a SIDA/inmunología , Proteínas del Sistema Complemento/metabolismo , Lóbulo Frontal/metabolismo , Infecciones por VIH/inmunología , VIH-1/fisiología , Dependencia de Heroína/inmunología , Mediadores de Inflamación/metabolismo , Microglía/metabolismo , Nefropatía Asociada a SIDA/epidemiología , Cadáver , Células Cultivadas , Comorbilidad , Activación de Complemento , Citocinas/metabolismo , Infecciones por VIH/epidemiología , Dependencia de Heroína/epidemiología , Humanos , Inmunomodulación , Microglía/patología , Microglía/virología , Regulación hacia Arriba , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo
14.
Immunology ; 148(4): 407-19, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27213693

RESUMEN

Blood-brain barrier (BBB) dysfunction complicates central nervous system lupus, an important aspect of systemic lupus erythematosus. To gain insight into the underlying mechanism, vascular corrosion casts of brain were generated from the lupus mouse model, MRL/lpr mice and the MRL/MpJ congenic controls. Scanning electron microscopy of the casts showed loss of vascular endothelial cells in lupus mice compared with controls. Immunostaining revealed a significant increase in caspase 3 expression in the brain vascular endothelial cells, which suggests that apoptosis could be an important mechanism causing cell loss, and thereby loss of BBB integrity. Complement activation occurs in lupus resulting in increased generation of circulating C5a, which caused the endothelial layer to become 'leaky'. In this study, we show that C5a and lupus serum induced apoptosis in cultured human brain microvascular endothelial cells (HBMVECs), whereas selective C5a receptor 1 (C5aR1) antagonist reduced apoptosis in these cells, demonstrating C5a/C5aR1-dependence. Gene expression of initiator caspases, caspase 1 and caspase 8, and pro-apoptotic proteins death-associated protein kinase 1, Fas-associated protein (FADD), cell death-inducing DNA fragmentation factor 45 000 MW subunit A-like effector B (CIDEB) and BCL2-associated X protein were increased in HBMVECs treated with lupus serum or C5a, indicating that both the intrinsic and extrinsic apoptotic pathways could be critical mediators of brain endothelial cell apoptosis in this setting. Overall, our findings suggest that C5a/C5aR1 signalling induces apoptosis through activation of FADD, caspase 8/3 and CIDEB in brain endothelial cells in lupus. Further elucidation of the underlying apoptotic mechanisms mediating the reduced endothelial cell number is important in establishing the potential therapeutic effectiveness of C5aR1 inhibition that could prevent and/or reduce BBB alterations and preserve the physiological function of BBB in central nervous system lupus.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis , Encéfalo/patología , Complemento C5a/metabolismo , Endotelio Vascular/inmunología , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Vasculitis por Lupus del Sistema Nervioso Central/inmunología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/inmunología , Encéfalo/metabolismo , Caspasa 3/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/patología , Humanos , Ratones , Ratones Endogámicos MRL lpr , Péptidos Cíclicos/farmacología , Receptor de Anafilatoxina C5a/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos
15.
Cell Mol Immunol ; 13(5): 651-7, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-26166765

RESUMEN

Activation of complement generates C5a which leads to signaling through C5aR1. This is tightly controlled, including by the plasma proteins factor H (FH) and carboxypeptidase N. Here we studied a chronic serum sickness (CSS) model of glomerulonephritis (GN) in which there is an active humoral immune response, formation of glomerular immune complexes (ICs), and resulting glomerular inflammation. The antibody response, glomerular IC deposition, the degree of GN, and consequent renal functional insufficiency in CSS were all worse in FH-/- mice compared to wild-type FH+/+ animals. This was ameliorated in the former by giving a C5aR1 antagonist for the final 3 weeks of the 5-week protocol. In contrast, blocking CP-mediated inactivation of C5a increased these disease measures. Thus, complement regulation by both plasma FH and CP to limit the quantity of active C5a is important in conditions where the humoral immune response is directed to a continuously present foreign antigen. Signaling through C5aR1 enhances the humoral immune response as well as the inflammatory response to ICs that have formed in glomeruli. Both effects are relevant even after disease has begun. Thus, pharmacological targeting of C5a in IC-mediated GN has potential clinical relevance.


Asunto(s)
Complejo Antígeno-Anticuerpo/metabolismo , Carboxipeptidasas/metabolismo , Glomerulonefritis/inmunología , Receptor de Anafilatoxina C5a/antagonistas & inhibidores , Animales , Apoferritinas/metabolismo , Complemento C5a/metabolismo , Factor H de Complemento/deficiencia , Factor H de Complemento/metabolismo , Glomerulonefritis/patología , Inmunidad Humoral , Inmunoglobulina G/metabolismo , Riñón/patología , Riñón/fisiopatología , Pruebas de Función Renal , Masculino , Ratones Endogámicos C57BL , Receptor de Anafilatoxina C5a/metabolismo
16.
Immunology ; 146(1): 130-43, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26059553

RESUMEN

The blood-brain barrier (BBB) plays a crucial role in brain homeostasis, thereby maintaining the brain environment precise for optimal neuronal function. Its dysfunction is an intriguing complication of systemic lupus erythematosus (SLE). SLE is a systemic autoimmune disorder where neurological complications occur in 5-50% of cases and is associated with impaired BBB integrity. Complement activation occurs in SLE and is an important part of the clinical profile. Our earlier studies demonstrated that C5a generated by complement activation caused the loss of brain endothelial layer integrity in rodents. The goal of the current study was to determine the translational potential of these studies to a human system. To assess this, we used a two dimensional in vitro BBB model constructed using primary human brain microvascular endothelial cells and astroglial cells, which closely emulates the in vivo BBB allowing the assessment of BBB integrity. Increased permeability monitored by changes in transendothelial electrical resistance and cytoskeletal remodelling caused by actin fiber rearrangement were observed when the cells were exposed to lupus serum and C5a, similar to the observations in mice. In addition, our data show that C5a/C5aR1 signalling alters nuclear factor-κB translocation into nucleus and regulates the expression of the tight junction proteins, claudin-5 and zonula occludens 1 in this setting. Our results demonstrate for the first time that C5a regulates BBB integrity in a neuroinflammatory setting where it affects both endothelial and astroglial cells. In addition, we also demonstrate that our previous findings in a mouse model, were emulated in human cells in vitro, bringing the studies one step closer to understanding the translational potential of C5a/C5aR1 blockade as a promising therapeutic strategy in SLE and other neurodegenerative diseases.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Complemento C5a/metabolismo , Lupus Eritematoso Sistémico/patología , Receptor de Anafilatoxina C5a/metabolismo , Citoesqueleto de Actina/metabolismo , Transporte Activo de Núcleo Celular , Adolescente , Astrocitos/inmunología , Encéfalo/irrigación sanguínea , Células Cultivadas , Niño , Claudina-5/biosíntesis , Activación de Complemento/inmunología , Complemento C5a/inmunología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Impedancia Eléctrica , Células Endoteliales/inmunología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Humanos , Moléculas de Adhesión de Unión/biosíntesis , Lupus Eritematoso Sistémico/inmunología , Masculino , FN-kappa B/metabolismo , Transporte de Proteínas , Receptor de Anafilatoxina C5a/antagonistas & inhibidores , Receptor de Anafilatoxina C5a/inmunología , Uniones Estrechas/metabolismo , Proteína de la Zonula Occludens-1/biosíntesis
17.
Kidney Int ; 87(5): 930-9, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25565310

RESUMEN

In chronic serum sickness, glomerular immune complexes form, yet C57BL/6 mice do not develop glomerulonephritis unless complement factor H (CfH) is absent, indicating the relevance of complement regulation. Complement receptor 3 (CD11b) and Fcγ receptors on leukocytes, and CfH on platelets, can bind immune complexes. Here we induced immune complex-mediated glomerulonephritis in CfH(-/-) mice chimeric for wild-type, CfH(-/-), CD11b(-/-), or FcRγ(-/-) bone marrow stem cells. Glomerulonephritis was worse in CD11b(-/-) chimeras compared with all others, whereas disease in FcRγ(-/-) and wild-type chimeras was comparable. Disease tracked strongly with humoral immune responses, but not glomerular immune complex deposits. Interstitial inflammation with M1 macrophages strongly correlated with glomerulonephritis scores. CD11b(-/-) chimeras had significantly more M1 macrophages and CD4(+) T cells. The renal dendritic cell populations originating from bone marrow-derived CD11c(+) cells were similar in all experimental groups. CD11b(+) cells bearing colony-stimulating factor 1 receptor were present in kidneys, including CD11b(-/-) chimeras; these cells correlated negatively with glomerulonephritis scores. Thus, experimental immune complex-mediated glomerulonephritis is associated with accumulation of M1 macrophages and CD4(+) T cells in kidneys and functional renal insufficiency. Hence, CD11b on mononuclear cells is instrumental in generating an anti-inflammatory response in the inflamed kidney.


Asunto(s)
Antígeno CD11b/metabolismo , Factor H de Complemento/metabolismo , Glomerulonefritis/inmunología , Leucocitos/metabolismo , Receptores de IgG/metabolismo , Animales , Complejo Antígeno-Anticuerpo/metabolismo , Apoferritinas/inmunología , Médula Ósea/metabolismo , Antígeno CD11b/genética , Antígenos CD18/metabolismo , Factor H de Complemento/genética , Glomerulonefritis/metabolismo , Inmunoglobulina G/metabolismo , Ratones Endogámicos C57BL , Receptor de Anafilatoxina C5a/metabolismo , Receptores de IgG/genética , Enfermedad del Suero/complicaciones
18.
Immunology ; 143(3): 363-73, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24801999

RESUMEN

Blood-brain barrier (BBB) disturbance is a crucial occurrence in many neurological diseases, including systemic lupus erythematosus (SLE). Our previous studies showed that experimental lupus serum altered the integrity of the mouse brain endothelial layer, an important constituent of the BBB. Complement activation occurs in lupus with increased circulating complement components. Using a genomics approach, we identified the microRNA (miRNA) altered in mouse brain endothelial cells (bEnd3) by lupus serum and the complement protein, C5a. Of the 318 miRNA evaluated, 23 miRNAs were altered by lupus serum and 32 were altered by C5a alone compared with controls. Seven miRNAs (P < 0 · 05) were differentially expressed by both treatments: mmu-miR-133a*, mmu-miR-193*, mmu-miR-26b, mmu-miR-28*, mmu-miR-320a, mmu-miR-423-3p and mmu-miR-509-5p. The microarray results were validated by quantitative RT-PCR. In line with the in vitro results, expression of miR-26b and miR-28* were also significantly up-regulated in lupus mouse brain which was reduced by C5a receptor inhibition. Target prediction analysis revealed miR gene targets encoding components involved in inflammation, matrix arrangement, and apoptosis, pathways known to play important roles in central nervous system lupus. Our findings suggest that the miRNAs reported in this study may represent novel therapeutic targets in central nervous system lupus and other similar neuroinflammatory settings.


Asunto(s)
Encéfalo/inmunología , Encéfalo/metabolismo , Complemento C5a/metabolismo , Células Endoteliales/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , MicroARNs/genética , Animales , Barrera Hematoencefálica/inmunología , Barrera Hematoencefálica/metabolismo , Línea Celular , Células Cultivadas , Complemento C5a/inmunología , Modelos Animales de Enfermedad , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inmunología , Ratones , Permeabilidad
19.
Physiol Genomics ; 45(19): 877-88, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23922129

RESUMEN

Colistin (polymixin E) is an antibiotic prescribed with resurging frequency for multidrug resistant gram negative bacterial infections. It is associated with nephrotoxicity in humans in up to 55% of cases. Little is known regarding genes involved in colistin nephrotoxicity. A murine model of colistin-mediated kidney injury was developed. C57/BL6 mice were administered saline or colistin at a dose of 16 mg/kg/day in 2 divided intraperitoneal doses and killed after either 3 or 15 days of colistin. After 15 days, mice exposed to colistin had elevated blood urea nitrogen (BUN), creatinine, and pathologic evidence of acute tubular necrosis and apoptosis. After 3 days, mice had neither BUN elevation nor substantial pathologic injury; however, urinary neutrophil gelatinase-associated lipocalin was elevated (P = 0.017). An Illumina gene expression array was performed on kidney RNA harvested 72 h after first colistin dose to identify differentially expressed genes early in drug treatment. Array data revealed 21 differentially expressed genes (false discovery rate < 0.1) between control and colistin-exposed mice, including LGALS3 and CCNB1. The gene signature was significantly enriched for genes involved in cell cycle proliferation. RT-PCR, immunoblot, and immunostaining validated the relevance of key genes and proteins. This murine model offers insights into the potential mechanism of colistin-mediated nephrotoxicity. Further studies will determine whether the identified genes play a causative or protective role in colistin-induced nephrotoxicity.


Asunto(s)
Puntos de Control del Ciclo Celular/efectos de los fármacos , Colistina/toxicidad , Riñón/efectos de los fármacos , Riñón/patología , Animales , Peso Corporal/efectos de los fármacos , Análisis por Conglomerados , Colistina/administración & dosificación , Modelos Animales de Enfermedad , Galectina 3/metabolismo , Perfilación de la Expresión Génica , Etiquetado Corte-Fin in Situ , Riñón/metabolismo , Túbulos Renales/efectos de los fármacos , Túbulos Renales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Necrosis , Antígeno Nuclear de Célula en Proliferación/metabolismo , Reproducibilidad de los Resultados
20.
Immunology ; 139(3): 328-37, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23347386

RESUMEN

Complement factor H (Cfh) is a key regulator of the complement cascade and protects C57BL/6 mice from immune complex-mediated complement-dependent glomerulonephritis. In chronic serum sickness (CSS) there are increased deposits of immune complexes in the glomeruli with inflammation and a scarring phenotype. As cucurmin is an effective anti-inflammatory agent and reduces complement activation, we hypothesized that it should alleviate renal disease in this setting. To determine the effectiveness of curcumin, an apoferritin-induced CSS model in Cfh-deficient (Cfh(-/-)) mice was used. Curcumin treatment (30 mg/kg) given every day in parallel with apoferritin reduced glomerulonephritis and enhanced kidney function (blood urea nitrogen, 45·4 ± 7·5 versus 35·6 ± 5·1; albuminuria, 50·1 ± 7·1 versus 15·7 ± 7·1; glomerulonephritis, 2·62 + 0·25 versus 2 + 0·3, P < 0·05). In line with reduced IgG deposits in mice with CSS given curcumin, C9 deposits were reduced indicating reduced complement activation. Mice treated with curcumin had a significant reduction in the number of splenic CD19(+) B cells and the ratio of CD19 : CD3 cells (P < 0·05) with no change in the T-cell population. Myeloperoxidase assay showed reduced macrophages in the kidney. However, a significant reduction in the M2 subset of splenic macrophages by apoferritin was prevented by curcumin, suggesting a protective function. Curcumin treatment reduced mRNA expression of inflammatory proteins monocyte chemoattractant protein-1 and transforming growth factor-ß and matrix proteins, fibronectin, laminin and collagen. Our results clearly illustrate that curcumin reduces glomerulosclerosis, improves kidney function and could serve as a therapeutic agent during serum sickness.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Curcumina/uso terapéutico , Glomerulonefritis/tratamiento farmacológico , Enfermedades del Complejo Inmune/tratamiento farmacológico , Enfermedad del Suero/tratamiento farmacológico , Animales , Antiinflamatorios no Esteroideos/administración & dosificación , Antiinflamatorios no Esteroideos/farmacología , Apoferritinas/administración & dosificación , Enfermedad Crónica , Activación de Complemento/efectos de los fármacos , Factor H de Complemento/deficiencia , Curcumina/administración & dosificación , Curcumina/farmacología , Glomerulonefritis/etiología , Humanos , Pruebas de Función Renal , Masculino , Ratones , Ratones Endogámicos C57BL , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...