Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 154
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Immunol Rev ; 313(1): 327-338, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36369971

RESUMEN

The glomerulus is often the prime target of dysregulated alternative pathway (AP) activation. In particular, AP activation is the key driver of two severe kidney diseases: atypical hemolytic uremic syndrome and C3 glomerulopathy. Both conditions are associated with a variety of predisposing molecular defects in AP regulation, such as genetic variants in complement regulators, autoantibodies targeting AP proteins, or autoantibodies that stabilize the AP convertases (C3- and C5-activating enzymes). It is noteworthy that these are systemic AP defects, yet in both diseases pathologic complement activation primarily affects the kidneys. In particular, AP activation is often limited to the glomerular capillaries. This tropism of AP-mediated inflammation for the glomerulus points to a unique interaction between AP proteins in plasma and this particular anatomic structure. In this review, we discuss the pre-clinical and clinical data linking the molecular causes of aberrant control of the AP with activation in the glomerulus, and the possible causes of this tropism. Based on these data, we propose a model for why the kidney is so uniquely and frequently targeted in patients with AP defects. Finally, we discuss possible strategies for preventing pathologic AP activation in the kidney.


Asunto(s)
Síndrome Hemolítico Urémico Atípico , Vía Alternativa del Complemento , Humanos , Vía Alternativa del Complemento/genética , Complemento C3/genética , Complemento C3/metabolismo , Riñón , Glomérulos Renales/metabolismo , Glomérulos Renales/patología , Síndrome Hemolítico Urémico Atípico/genética , Síndrome Hemolítico Urémico Atípico/patología , Autoanticuerpos
2.
Immunol Rev ; 313(1): 225-238, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36305168

RESUMEN

Uncontrolled alternative pathway activation is the primary driver of several diseases, and it contributes to the pathogenesis of many others. Consequently, diagnostic tests to monitor this arm of the complement system are increasingly important. Defects in alternative pathway regulation are strong risk factors for disease, and drugs that specifically block the alternative pathway are entering clinical use. A range of diagnostic tests have been developed to evaluate and monitor the alternative pathway, including assays to measure its function, expression of alternative pathway constituents, and activation fragments. Genetic studies have also revealed many disease-associated variants in alternative pathway genes that predict the risk of disease and prognosis. Newer imaging modalities offer the promise of non-invasively detecting and localizing pathologic complement activation. Together, these various tests help in the diagnosis of disease, provide important prognostic information, and can help guide therapy with complement inhibitory drugs.


Asunto(s)
Activación de Complemento , Proteínas del Sistema Complemento , Humanos , Proteínas del Sistema Complemento/genética , Pronóstico , Vía Alternativa del Complemento/genética
3.
J Immunol ; 211(8): 1240-1248, 2023 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-37682538

RESUMEN

B cell-targeted therapies are effective for treating multiple different kidney diseases in humans and also protect mice from Adriamycin nephropathy. Because glomerular IgM is frequently seen in both humans and mice with "nonimmune" forms of glomerular disease, we hypothesized that natural IgM binds to epitopes displayed in the injured glomerulus, exacerbating injury. To test this hypothesis, we induced Adriamycin nephropathy in BALB/C mice that cannot secrete soluble IgM (sIgM-/- mice) and compared them with BALB/C controls. Contrary to our prediction, we found that female sIgM-/- mice developed higher mortality and more severe kidney injury after injection of Adriamycin. The absence of soluble IgM did not reduce glomerular complement activation, and IgG was seen deposited within the injured glomeruli. Furthermore, we discovered that female sIgM-/- mice have higher levels of anti-cardiolipin IgG, and that IgG from these mice binds to epitopes in the injured kidney. These findings indicate that natural IgM may prevent generation of autoreactive IgG. Circulating levels of anti-cardiolipin IgG decreased after induction of kidney injury in female mice, consistent with deposition of the Abs in injured tissues. Better understanding of the mechanisms by which the immune system modulates and amplifies kidney injury may enable the development of targeted therapies to slow kidney disease progression.


Asunto(s)
Inmunoglobulina M , Enfermedades Renales , Animales , Femenino , Ratones , Doxorrubicina , Epítopos , Inmunoglobulina G , Ratones Endogámicos BALB C
4.
Mol Ther ; 32(4): 1061-1079, 2024 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-38382529

RESUMEN

Complement-mediated diseases can be treated using systemic inhibitors. However, complement components are abundant in circulation, affecting systemic inhibitors' exposure and efficacy. Furthermore, because of complement's essential role in immunity, systemic treatments raise infection risk in patients. To address these challenges, we developed antibody fusion proteins combining the alternative-pathway complement inhibitor factor H (fH1-5) with an anti-C3d monoclonal antibody (C3d-mAb-2fH). Because C3d is deposited at sites of complement activity, this molecule localizes to tissue complement while minimizing circulating complement engagement. These fusion proteins bind to deposited complement in diseased human skin sections and localize to activated complement in a primate skin injury model. We further explored the pharmacology of C3d-mAb-2fH proteins in rodent models with robust tissue complement activation. Doses of C3d-mAb-2fH >1 mg/kg achieved >75% tissue complement inhibition in mouse and rat injury models while avoiding circulating complement blockade. Glomerular-specific complement inhibition reduced proteinuria and preserved podocyte foot-process architecture in rat membranous nephropathy, indicating disease-modifying efficacy. These data indicate that targeting local tissue complement results in durable and efficacious complement blockade in skin and kidney while avoiding systemic inhibition, suggesting broad applicability of this approach in treating a range of complement-mediated diseases.


Asunto(s)
Factor H de Complemento , Enfermedades Renales , Humanos , Ratones , Ratas , Animales , Factor H de Complemento/genética , Complemento C3d/metabolismo , Enfermedades Renales/etiología , Anticuerpos , Activación de Complemento
5.
Kidney Int ; 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38844295

RESUMEN

Uncontrolled complement activation can cause or contribute to glomerular injury in multiple kidney diseases. Although complement activation plays a causal role in atypical hemolytic uremic syndrome and C3 glomerulopathy, over the past decade, a rapidly accumulating body of evidence has shown a role for complement activation in multiple other kidney diseases, including diabetic nephropathy and several glomerulonephritides. The number of available complement inhibitor therapies has also increased during the same period. In 2022, Kidney Diseases: Improving Global Outcomes (KDIGO) convened a Controversies Conference, "The Role of Complement in Kidney Disease," to address the expanding role of complement dysregulation in the pathophysiology, diagnosis, and management of various glomerular diseases, diabetic nephropathy, and other forms of hemolytic uremic syndrome. Conference participants reviewed the evidence for complement playing a primary causal or secondary role in progression for several disease states and considered how evidence of complement involvement might inform management. Participating patients with various complement-mediated diseases and caregivers described concerns related to life planning, implications surrounding genetic testing, and the need for inclusive implementation of effective novel therapies into clinical practice. The value of biomarkers in monitoring disease course and the role of the glomerular microenvironment in complement response were examined, and key gaps in knowledge and research priorities were identified.

6.
Pediatr Nephrol ; 39(5): 1387-1404, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-37733095

RESUMEN

Historically, the complement system (classical, lectin, alternative, and terminal pathways) is known to play a crucial role in the etiopathogenesis of many kidney diseases. Direct or indirect activation in these settings is revealed by consumption of complement proteins at the serum level and kidney tissue deposition seen by immunofluorescence and electron microscopy. The advent of eculizumab has shown that complement inhibitors may improve the natural history of certain kidney diseases. Since then, the number of available therapeutic molecules and experimental studies on complement inhibition has increased exponentially. In our narrative review, we give a summary of the main complement inhibitors that have completed phase II and phase III studies or are currently used in adult and pediatric nephrology. The relevant full-text works, abstracts, and ongoing trials (clinicaltrials.gov site) are discussed. Data and key clinical features are reported for eculizumab, ravulizumab, crovalimab, avacopan, danicopan, iptacopan, pegcetacoplan, and narsoplimab. Many of these molecules have been shown to be effective in reducing proteinuria and stabilizing kidney function in different complement-mediated kidney diseases. Thanks to their efficacy and target specificity, these novel drugs may radically improve the outcome of complement-mediated kidney diseases, contributing to an improvement in our understanding of their underlying pathophysiology.


Asunto(s)
Síndrome Hemolítico Urémico Atípico , Glomerulonefritis Membranoproliferativa , Enfermedades Renales , Adulto , Niño , Humanos , Inactivadores del Complemento/uso terapéutico , Inactivadores del Complemento/farmacología , Complemento C3/metabolismo , Glomerulonefritis Membranoproliferativa/tratamiento farmacológico , Síndrome Hemolítico Urémico Atípico/tratamiento farmacológico , Enfermedades Renales/tratamiento farmacológico , Activación de Complemento
7.
J Am Soc Nephrol ; 34(7): 1151-1154, 2023 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-36995143

RESUMEN

SIGNIFICANCE STATEMENT: Histologic quantification of complement C3 deposits in kidney biopsies provides prognostic information in patients with glomerulonephritis. Unfortunately, kidney biopsies are invasive procedures that cannot be performed regularly and only provide a snapshot of a small portion of one kidney at the time of sampling. We have developed a method to noninvasively detect specific C3 fragment deposition throughout both kidneys, using a monoclonal antibody targeting tissue-bound iC3b/C3d linked to a bioluminescent resonance energy transfer construct that emits near-infrared light. In a mouse model of glomerulonephritis, the probe detected iC3b/C3d in kidneys of live mice by bioluminescent imaging. This demonstrates that noninvasive imaging with an anti-iC3b/C3d probe can be used to monitor inflammation in the kidneys.


Asunto(s)
Complemento C3b , Glomerulonefritis , Animales , Ratones , Complemento C3d , Riñón/diagnóstico por imagen , Anticuerpos Monoclonales
8.
Pediatr Nephrol ; 38(5): 1411-1425, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36203104

RESUMEN

The complement cascade is an important part of the innate immune system. In addition to helping the body to eliminate pathogens, however, complement activation also contributes to the pathogenesis of a wide range of kidney diseases. Recent work has revealed that uncontrolled complement activation is the key driver of several rare kidney diseases in children, including atypical hemolytic uremic syndrome and C3 glomerulopathy. In addition, a growing body of literature has implicated complement in the pathogenesis of more common kidney diseases, including acute kidney injury (AKI). Complement-targeted therapeutics are in use for a variety of diseases, and an increasing number of therapeutic agents are under development. With the implication of complement in the pathogenesis of AKI, complement-targeted therapeutics could be trialed to prevent or treat this condition. In this review, we discuss the evidence that the complement system is activated in pediatric patients with AKI, and we review the role of complement proteins as biomarkers and therapeutic targets in patients with AKI.


Asunto(s)
Proteínas del Sistema Complemento , Enfermedades Renales , Riñón , Síndrome Hemolítico Urémico Atípico/terapia , Activación de Complemento , Riñón/patología , Humanos , Niño , Enfermedades Renales/terapia
9.
Kidney Int ; 102(6): 1331-1344, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36063874

RESUMEN

Complement activation at a particular location is determined by the balance of activating and inhibitory proteins. Factor H is a key regulator of the alternative pathway of complement, and genetic or acquired impairments in Factor H are associated with glomerular injury. The human Factor H-related proteins (FHRs) comprise a family of five proteins that are structurally related to Factor H. Variations in the genes or expression levels of the FHRs are also associated with glomerular disease, although the mechanisms of glomerular protection/injury are incompletely understood. To explore the role of the FHRs on complement regulation/dysregulation in the kidney, we expressed and purified recombinant murine FHRs (FHRs A, B, C and E). These four distinct FHRs contain binding regions with high amino acid sequence homology to binding regions within Factor H, but we observed different interactions of the FHRs with Factor H binding ligands, including heparin and C3d. There was differential binding of the FHRs to the resident kidney cell types (mesangial, glomerular endothelial, podocytes, and tubular epithelial). All four FHRs caused complement dysregulation on kidney cell surfaces in vitro, although the magnitude of the effect differed among the FHRs and also varied among the different kidney cells. However, only FHR E caused glomerular complement dysregulation when injected in vivo but did not exacerbate injury when injected into mice with ischemic acute kidney injury, an alternative pathway-mediated model. Thus, our experiments demonstrate that the FHRs have unique, and likely context-dependent, effects on the different cell types within the kidney.


Asunto(s)
Factor H de Complemento , Enfermedades Renales , Humanos , Ratones , Animales , Factor H de Complemento/genética , Factor H de Complemento/metabolismo , Activación de Complemento , Proteínas del Sistema Complemento/metabolismo , Riñón/metabolismo
10.
Am J Transplant ; 22(9): 2139-2157, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35583104

RESUMEN

Extracellular vesicles (EV) are emerging mediators in several diseases. However, their role in the pathophysiology of antibody-mediated allograft rejection (AMR) has been poorly investigated. Here, we investigated the role of EV isolated from AMR patients in inducing tubular senescence and endothelial to mesenchymal transition (EndMT) and analyzed their miRNA expression profile. By multiplex bead flow cytometry, we characterized the immunophenotype of plasma AMR-derived EV and found a prevalent platelet and endothelial cell origin. In vitro, AMR-derived EV induced tubular senescence by upregulating SA-ß Gal and CDKN1A mRNA. Furthermore, AMR-derived EV induced EndMT. The occurrence of tubular senescence and EndMT was confirmed by analysis of renal biopsies from the same AMR patients. Moreover, AMR-derived EV induced C3 gene upregulation and CFH downregulation in tubular epithelial cells, with C4d deposition on endothelial cells. Interestingly, RNase-mediated digestion of EV cargo completely abrogated tubular senescence and EndMT. By microarray analysis, miR-604, miR-515-3p, miR-let-7d-5p, and miR-590-3p were significantly upregulated in EV from AMR group compared with transplant controls, whereas miR-24-3p and miR-29a-3p were downregulated. Therefore, EV-associated miRNA could act as active player in AMR pathogenesis, unraveling potential mechanisms of accelerated graft senescence, complement activation and early fibrosis that might lead to new therapeutic intervention.


Asunto(s)
Vesículas Extracelulares , MicroARNs , Células Endoteliales/metabolismo , Células Epiteliales/metabolismo , Vesículas Extracelulares/metabolismo , Humanos , MicroARNs/genética , ARN Mensajero/metabolismo
11.
Am J Kidney Dis ; 79(4): 570-581, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34571062

RESUMEN

Blocking the complement system as a therapeutic strategy has been proposed for numerous glomerular diseases but presents myriad questions and challenges, not the least of which is demonstrating efficacy and safety. In light of these potential issues and because there are an increasing number of anticomplement therapy trials either planned or under way, the National Kidney Foundation facilitated an all-virtual scientific workshop entitled "Improving Clinical Trials for Anti-Complement Therapies in Complement-Mediated Glomerulopathies." Attended by patient representatives and experts in glomerular diseases, complement physiology, and clinical trial design, the aim of this workshop was to develop standards applicable for designing and conducting clinical trials for anticomplement therapies across a wide spectrum of complement-mediated glomerulopathies. Discussions focused on study design, participant risk assessment and mitigation, laboratory measurements and biomarkers to support these studies, and identification of optimal outcome measures to detect benefit, specifically for trials in complement-mediated diseases. This report summarizes the discussions from this workshop and outlines consensus recommendations.


Asunto(s)
Proteínas Inactivadoras de Complemento , Enfermedades Renales , Proteínas Inactivadoras de Complemento/uso terapéutico , Proteínas del Sistema Complemento , Humanos , Riñón
12.
Pediatr Nephrol ; 37(3): 521-535, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34002292

RESUMEN

C3 glomerulopathy is a relatively new clinical entity that represents a challenge both to diagnose and to treat. As new therapeutic agents that act as complement inhibitors become available, many with an oral formulation, a better understanding of this disease and of the underlying complement dysregulation driving it has become increasingly useful to optimize patient care. Moreover, recent advances in research have clarified the role of complement in other glomerular diseases in which its role was less established, namely in immune-complex membranoproliferative glomerulonephritis (IC-MPGN), ANCA-vasculitis, IgA nephropathy, and idiopathic membranous nephropathy. Complement inhibitors are being studied in adult and adolescent clinical trials for these indications. This review summarizes current knowledge and future perspectives on every aspect of the diagnosis and management of C3 glomerulopathy and elucidates current understanding of the role of complement in this condition and in other glomerular diseases in children. An overview of ongoing trials involving therapeutic agents targeting complement in glomerular diseases is also provided.


Asunto(s)
Glomerulonefritis Membranoproliferativa , Glomerulonefritis , Enfermedades Renales , Adolescente , Adulto , Niño , Inactivadores del Complemento/uso terapéutico , Femenino , Glomerulonefritis/diagnóstico , Glomerulonefritis/tratamiento farmacológico , Glomerulonefritis Membranoproliferativa/tratamiento farmacológico , Humanos , Enfermedades Renales/diagnóstico , Enfermedades Renales/tratamiento farmacológico , Masculino
14.
Am J Physiol Renal Physiol ; 321(4): F505-F516, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34459222

RESUMEN

Focal segmental glomerulosclerosis (FSGS) and minimal change disease (MCD) are common forms of idiopathic nephrotic syndrome. The causes of these diseases are incompletely understood, but the response of patients to immunosuppressive therapies suggests that their pathogenesis is at least in part immune mediated. Preclinical and clinical research indicates that activation of the classical pathway of complement contributes to glomerular injury in FSGS. Glomerular IgM deposits are also prominent in some patients, raising the possibility that IgM is a trigger of classical pathway activation. In the present study, we examined the pattern of complement activation in the glomeruli and plasma of patients with nephrotic syndrome. We also tested whether patients with FSGS and MCD have elevated levels of natural IgM reactive with epitopes on glomerular endothelial cells and cardiolipin. We found evidence of classical pathway activation in patients with idiopathic nephrotic syndrome compared with healthy control subjects. We also detected higher levels of self-reactive IgM to both targets. Based on these results, IgM and classical pathway activation may contribute to disease pathogenesis in some patients with FSGS and MCD.NEW & NOTEWORTHY IgM is detected in biopsies from some patients with nephrotic syndrome, although this has been attributed to passive trapping of the protein. We found, however, that IgM colocalizes with complement activation fragments in some glomeruli. We also found that affected patients had higher levels of IgM reactive to glomerular endothelial cell epitopes. Thus, IgM activates the complement system in the glomeruli of some patients with nephrotic syndrome and may contribute to injury.


Asunto(s)
Cardiolipinas/inmunología , Vía Clásica del Complemento , Proteínas del Sistema Complemento/análisis , Células Endoteliales/inmunología , Epítopos , Glomeruloesclerosis Focal y Segmentaria/inmunología , Inmunoglobulina M/análisis , Glomérulos Renales/inmunología , Nefrosis Lipoidea/inmunología , Síndrome Nefrótico/inmunología , Adulto , Anciano , Especificidad de Anticuerpos , Estudios de Casos y Controles , Vía Clásica del Complemento/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Femenino , Glomeruloesclerosis Focal y Segmentaria/sangre , Glomeruloesclerosis Focal y Segmentaria/tratamiento farmacológico , Glomeruloesclerosis Focal y Segmentaria/patología , Humanos , Inmunoglobulina M/sangre , Inmunosupresores/uso terapéutico , Glomérulos Renales/efectos de los fármacos , Glomérulos Renales/patología , Masculino , Persona de Mediana Edad , Nefrosis Lipoidea/tratamiento farmacológico , Nefrosis Lipoidea/patología , Síndrome Nefrótico/tratamiento farmacológico , Síndrome Nefrótico/patología , Resultado del Tratamiento , Adulto Joven
15.
Kidney Int ; 99(3): 537-539, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33637198

RESUMEN

Rhabdomyolysis is frequently associated with kidney injury. Unfortunately, there are no specific treatments for this condition, and patient care primarily consists of supportive measures. In this edition of Kidney International, Boudhabhay et al. demonstrate that myoglobin released from injured skeletal muscle cells triggers tubulointerstitial complement activation. In a mouse model of the disease, interventions that scavenged free heme or that prevented complement activation ameliorated kidney injury, raising the possibility that these strategies may be effective treatments for the condition.


Asunto(s)
Lesión Renal Aguda , Rabdomiólisis , Animales , Activación de Complemento , Células Epiteliales , Humanos , Ratones , Mioglobina , Rabdomiólisis/terapia
16.
J Immunol ; 203(12): 3136-3147, 2019 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-31732528

RESUMEN

Humoral autoimmunity is central to the development of systemic lupus erythematosus (SLE). Complement receptor type 2 (CR2)/CD21 plays a key role in the development of high-affinity Abs and long-lasting memory to foreign Ags. When CR2 is bound by its primary C3 activation fragment-derived ligand, designated C3d, it coassociates with CD19 on B cells to amplify BCR signaling. C3d and CR2 also mediate immune complex binding to follicular dendritic cells. As the development of SLE involves subversion of normal B cell tolerance checkpoints, one might expect that CR2 ligation by C3d-bound immune complexes would promote development of SLE. However, prior studies in murine models of SLE using gene-targeted Cr2-/- mice, which lack both CR2 and complement receptor 1 (CR1), have demonstrated contradictory results. As a new approach, we developed a highly specific mouse anti-mouse C3d mAb that blocks its interaction with CR2. With this novel tool, we show that disruption of the critical C3d-CR2 ligand-receptor binding step alone substantially ameliorates autoimmunity and renal disease in the MRL/lpr model of SLE.


Asunto(s)
Complejo Antígeno-Anticuerpo/inmunología , Complemento C3d/inmunología , Lupus Eritematoso Sistémico/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Complejo Antígeno-Anticuerpo/metabolismo , Autoanticuerpos/inmunología , Autoinmunidad , Biomarcadores , Complemento C3d/antagonistas & inhibidores , Complemento C3d/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Epítopos de Linfocito T/inmunología , Femenino , Humanos , Inmunoglobulina G/inmunología , Inmunoglobulina G/metabolismo , Mediadores de Inflamación , Ligandos , Lupus Eritematoso Sistémico/tratamiento farmacológico , Lupus Eritematoso Sistémico/metabolismo , Ratones , Ratones Endogámicos MRL lpr , Ratones Noqueados , Unión Proteica/efectos de los fármacos , Unión Proteica/inmunología
17.
Xenobiotica ; 51(6): 657-667, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33870862

RESUMEN

Effects of cholecalciferol (VitD3) and calcitriol (1,25-VitD3), on the expression and function of major vitamin D metabolizing enzymes (cytochrome P450 [CYP]2R1, CYP24A1) and select drug transport pathways (ABCB1/P-gp, SLCO4C1/OATP4C1) were evaluated in human kidney proximal tubule epithelial cells (hPTECs) under normal and uraemic serum conditions.hPTECs were incubated with 10% normal or uraemic serum for 24 h followed by treatment with 2% ethanol vehicle, or 100 and 240 nM doses of VitD3, or 1,25-VitD3 for 6 days. The effects of treatment on mRNA and protein expression and functional activity of select CYP enzymes and transporters were assessedUnder uraemic serum, treatment with 1,25-VitD3 resulted in increased mRNA but decreased protein expression of CYP2R1. Activity of CYP2R1 was not influenced by serum or VitD analogues. CYP24A1 expression was increased with 1,25-VitD3 under normal as well as uraemic serum, although to a lesser extent. ABCB1/P-gp mRNA expression increased under normal and uraemic serum, with exposure to 1,25-VitD3. SLCO4C1/OATP4C1 exhibited increased mRNA but decreased protein expression, under uraemic serum + 1,25-VitD3. Functional assessments of transport showed no changes regardless of exposure to serum or 1,25-VitD3.Key findings indicate that uraemic serum and VitD treatment led to differential effects on the functional expression of CYPs and transporters in hPTECs.


Asunto(s)
Transportadores de Anión Orgánico , Preparaciones Farmacéuticas , Uremia , Colecalciferol , Humanos , Riñón , Vitamina D
18.
Am J Respir Crit Care Med ; 201(2): 224-239, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31545648

RESUMEN

Rationale: Pulmonary hypertension (PH) is a life-threatening cardiopulmonary disorder in which inflammation and immunity have emerged as critical early pathogenic elements. Although proinflammatory processes in PH and pulmonary arterial hypertension (PAH) are the focus of extensive investigation, the initiating mechanisms remain elusive.Objectives: We tested whether activation of the complement cascade is critical in regulating proinflammatory and pro-proliferative processes in the initiation of experimental hypoxic PH and can serve as a prognostic biomarker of outcome in human PAH.Methods: We used immunostaining of lung tissues from experimental PH models and patients with PAH, analyses of genetic murine models lacking specific complement components or circulating immunoglobulins, cultured human pulmonary adventitial fibroblasts, and network medicine analysis of a biomarker risk panel from plasma of patients with PAH.Measurements and Main Results: Pulmonary perivascular-specific activation of the complement cascade was identified as a consistent critical determinant of PH and PAH in experimental animal models and humans. In experimental hypoxic PH, proinflammatory and pro-proliferative responses were dependent on complement (alternative pathway and component 5), and immunoglobulins, particularly IgG, were critical for activation of the complement cascade. We identified Csf2/GM-CSF as a primary complement-dependent inflammatory mediator. Furthermore, using network medicine analysis of a biomarker risk panel from plasma of patients with PAH, we demonstrated that complement signaling can serve as a prognostic factor for clinical outcome in PAH.Conclusions: This study establishes immunoglobulin-driven dysregulated complement activation as a critical pathobiological mechanism regulating proinflammatory and pro-proliferative processes in the initiation of experimental hypoxic PH and demonstrates complement signaling as a critical determinant of clinical outcome in PAH.


Asunto(s)
Activación de Complemento/inmunología , Fibroblastos/inmunología , Hipertensión Pulmonar/inmunología , Inmunoglobulina G/inmunología , Remodelación Vascular/inmunología , Animales , Complemento C3/inmunología , Complemento C5/inmunología , Factor B del Complemento/inmunología , Vía Alternativa del Complemento/inmunología , Modelos Animales de Enfermedad , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Humanos , Hipertensión Pulmonar/etiología , Hipoxia/complicaciones , Inmunoglobulinas/inmunología , Inflamación , Ratones , Ratones Noqueados , Pronóstico , Hipertensión Arterial Pulmonar/inmunología , Ratas
19.
J Immunol ; 200(1): 316-326, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29187587

RESUMEN

Factor H (FH) is a key alternative pathway regulator that controls complement activation both in the fluid phase and on specific cell surfaces, thus allowing the innate immune response to discriminate between self and foreign pathogens. However, the interrelationships between FH and a group of closely related molecules, designated the FH-related (FHR) proteins, are currently not well understood. Whereas some studies have suggested that human FHR proteins possess complement regulatory abilities, recent studies have shown that FHR proteins are potent deregulators. Furthermore, the roles of the FHR proteins have not been explored in any in vivo models of inflammatory disease. In this study, we report the cloning and expression of recombinant mouse FH and three FHR proteins (FHR proteins A-C). Results from functional assays show that FHR-A and FHR-B proteins antagonize the protective function of FH in sheep erythrocyte hemolytic assays and increase cell-surface C3b deposition on a mouse kidney proximal tubular cell line (TEC) and a human retinal pigment epithelial cell line (ARPE-19). We also report apparent KD values for the binding interaction of mouse C3d with mouse FH (3.85 µM), FHR-A (136 nM), FHR-B (546 nM), and FHR-C (1.04 µM), which directly correlate with results from functional assays. Collectively, our work suggests that similar to their human counterparts, a subset of mouse FHR proteins have an important modulatory role in complement activation. Further work is warranted to define the in vivo context-dependent roles of these proteins and determine whether FHR proteins are suitable therapeutic targets for the treatment of complement-driven diseases.


Asunto(s)
Proteínas Inactivadoras del Complemento C3b/genética , Factor H de Complemento/metabolismo , Vía Alternativa del Complemento , Riñón/fisiología , Epitelio Pigmentado de la Retina/fisiología , Animales , Línea Celular , Clonación Molecular , Proteínas Inactivadoras del Complemento C3b/metabolismo , Hemólisis , Humanos , Inmunidad Innata , Inmunomodulación , Ratones , Receptores de Complemento/metabolismo , Autotolerancia
20.
Semin Immunol ; 28(3): 260-7, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27282113

RESUMEN

Despite substantial opportunity and commercial interest in developing drugs that modulate the complement system in a broad range of non-orphan indications, several obstacles remain to be overcome. Among these issues is the biophysical nature of complement proteins, whose circulating levels are typically very high and whose turnover rates are relatively rapid, especially in the setting of chronic inflammatory conditions. This situation necessitates the use of very high levels of therapeutic compounds in order to achieve both multi-pathway and multiple effector mechanism inhibition. In addition, one must avoid infectious complications or the systemic impairment of the other important physiological functions of complement. Herein we focus on the development of a novel therapeutic strategy based on injured tissue-specific targeting of complement inhibitors using the antigen-combining domains of a small subset of natural IgM antibodies, which as endogenous antibodies specifically recognize sites of local damage across a broad range of tissues and locally activate complement C3, resulting in C3 fragment covalent fixation. Because the use of such recombinant tissue-targeting inhibitors precludes the utility of measuring systemic levels of complement biomarkers or function, since a goal of this targeting strategy is to leave those processes intact and unimpeded, we also briefly describe a new method designed to quantitatively measure using imaging modalities the inhibition of generation of fixed C3 fragments at sites of inflammation/injury. In addition to the ability to determine whether complement activation is locally constrained with the use of inhibitors, there is also a broader application of this imaging approach to inflammatory and autoimmune diseases characterized by local complement activation.


Asunto(s)
Inactivadores del Complemento/uso terapéutico , Proteínas del Sistema Complemento/metabolismo , Inflamación/diagnóstico , Inflamación/terapia , Animales , Autoinmunidad , Activación de Complemento , Diagnóstico por Imagen , Humanos , Terapia Molecular Dirigida , Especificidad de Órganos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA