Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 102
Filtrar
Mais filtros

País/Região como assunto
Intervalo de ano de publicação
1.
BMC Nephrol ; 25(1): 55, 2024 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-38355500

RESUMO

BACKGROUND: The International Classification of Diseases (ICD) coding system is the industry standard tool for billing, disease classification, and epidemiology purposes. Prior research has demonstrated ICD codes to have poor accuracy, particularly in relation to rapidly progressing chronic kidney disease (CKD) patients. In 2016, the ICD system moved to revision 10. This study examines subjects in a large insurer database to determine the accuracy of ICD-10 CKD-staging codes to diagnose patients rapidly progressing towards end-stage kidney disease (ESKD). PATIENTS AND METHODS: Serial observations of outpatient serum creatinine measurements from 2016 to 2021 of 315,903 patients were transformed to estimated glomerular filtration rate (eGFR) to identify CKD stage-3 and advanced patients diagnosed clinically (eGFR-CKD). CKD-staging codes from the same time period of 59,386 patients and used to identify stage-3 and advanced patients diagnosed by ICD-code (ICD-CKD). eGFR-CKD and ICD-CKD diagnostic accuracy was compared between a total of 334,610 patients. RESULTS: 5,618 patients qualified for the progression analysis; 72 were identified as eGFR rapid progressors; 718 had multiple codes to qualify as ICD rapid progressors. Sensitivity was 5.56%, with positive predictive value (PPV) 5.6%. 34,858 patients were diagnosed as eGFR-CKD stage-3 patients; 17,549 were also diagnosed as ICD-CKD stage-3 patients, for a sensitivity of 50.34%, with PPV of 58.71%. 4,069 patients reached eGFR-CKD stage-4 with 2,750 ICD-CKD stage-4 patients, giving a sensitivity of 67.58%, PPV of 42.43%. 959 patients reached eGFR-CKD stage-5 with 566 ICD-CKD stage-5 patients, giving a sensitivity of 59.02%, PPV of 35.85%. CONCLUSION: This research shows that recent ICD revisions have not improved identification of rapid progressors in diagnostic accuracy, although marked increases in sensitivity for stage-3 (50.34% vs. 24.68%), and PPV in stage-3 (58.71% vs. 40.08%), stage-4 (42.43% vs. 18.52%), and stage-5 (35.85% vs. 4.51%) were observed. However, sensitivity in stage-5 compares poorly (59.02% vs. 91.05%).


Assuntos
Falência Renal Crônica , Insuficiência Renal Crônica , Humanos , Classificação Internacional de Doenças , Insuficiência Renal Crônica/diagnóstico , Insuficiência Renal Crônica/epidemiologia , Falência Renal Crônica/diagnóstico , Falência Renal Crônica/epidemiologia , Taxa de Filtração Glomerular , Testes Diagnósticos de Rotina
2.
Immunology ; 170(2): 180-192, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37222083

RESUMO

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.


Assuntos
Ativação do Complemento , Proteínas do Sistema Complemento
3.
Kidney Int ; 100(4): 824-836, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34139209

RESUMO

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.


Assuntos
Fator H do Complemento , Nefropatias , Animais , Fator H do Complemento/genética , Via Alternativa do Complemento , Células Endoteliais , Humanos , Rim , Camundongos
4.
Clin Immunol ; 198: 102-105, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30472267

RESUMO

Owing to an increasing shortage of donor organs, the majority of patients with end-stage kidney disease remains reliant on extracorporeal hemodialysis (HD) in order to counter the lifelong complications of a failing kidney. While HD remains a life-saving option for these patients, mounting evidence suggests that it also fuels a vicious cycle of thromboinflammation that can increase the risk of cardiovascular disease. During HD, blood-borne innate immune systems become inappropriately activated on the biomaterial surface, instigating proinflammatory reactions that can alter endothelial and vascular homeostasis. Complement activation, early during the HD process, has been shown to fuel a multitude of detrimental thromboinflammatory reactions that collectively contribute to patient morbidity. Here we discuss emerging aspects of complement's involvement in HD-induced inflammation and put forth the concept that targeted intervention at the level of C3 might constitute a promising therapeutic approach in HD patients.


Assuntos
Complemento C3/antagonistas & inibidores , Inativadores do Complemento/uso terapêutico , Inflamação/tratamento farmacológico , Diálise Renal/efeitos adversos , Humanos
5.
Am J Physiol Renal Physiol ; 315(3): F487-F502, 2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-29693447

RESUMO

Several lines of evidence suggest that gut bacterial microbiota is altered in patients with chronic kidney disease (CKD), though the mechanism of which this dysbiosis takes place is not well understood. Recent studies delineated changes in gut microbiota in both CKD patients and experimental animal models using microarray chips. We present 16S ribosomal RNA gene sequencing of both stool pellets and small bowel contents of C57BL/6J mice that underwent a remnant kidney model and establish that changes in microbiota take place in the early gastrointestinal tract. Increased intestinal urea concentration has been hypothesized as a leading contributor to dysbiotic changes in CKD. We show that urea transporters (UT)-A and UT-B mRNA are both expressed throughout the whole gastrointestinal tract. The noted increase in intestinal urea concentration appears to be independent of UTs' expression. Urea supplementation in drinking water resulted in alteration in bacterial gut microbiota that is quite different than that seen in CKD. This indicates that increased intestinal urea concentration might not fully explain the CKD- associated dysbiosis.


Assuntos
Bactérias/metabolismo , Disbiose , Microbioma Gastrointestinal , Intestino Delgado/microbiologia , Insuficiência Renal Crônica/microbiologia , Ureia/metabolismo , Uremia/microbiologia , Administração Oral , Animais , Bactérias/classificação , Bactérias/genética , Modelos Animais de Doenças , Fezes/microbiologia , Interações Hospedeiro-Patógeno , Hidrólise , Intestino Delgado/metabolismo , Masculino , Proteínas de Membrana Transportadoras/metabolismo , Camundongos Endogâmicos C57BL , Insuficiência Renal Crônica/metabolismo , Ribotipagem , Ureia/administração & dosagem , Urease/metabolismo , Uremia/metabolismo
6.
Immunol Invest ; 46(8): 816-832, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29058550

RESUMO

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.


Assuntos
Nefropatia Associada a AIDS/imunologia , Proteínas do Sistema Complemento/metabolismo , Lobo Frontal/metabolismo , Infecções por HIV/imunologia , HIV-1/fisiologia , Dependência de Heroína/imunologia , Mediadores da Inflamação/metabolismo , Microglia/metabolismo , Nefropatia Associada a AIDS/epidemiologia , Cadáver , Células Cultivadas , Comorbidade , Ativação do Complemento , Citocinas/metabolismo , Infecções por HIV/epidemiologia , Dependência de Heroína/epidemiologia , Humanos , Imunomodulação , Microglia/patologia , Microglia/virologia , Regulação para Cima , Produtos do Gene tat do Vírus da Imunodeficiência Humana/metabolismo
7.
Immunology ; 148(4): 407-19, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27213693

RESUMO

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.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Apoptose , Encéfalo/patologia , Complemento C5a/metabolismo , Endotélio Vascular/imunologia , Proteína de Domínio de Morte Associada a Fas/metabolismo , Vasculite Associada ao Lúpus do Sistema Nervoso Central/imunologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/imunologia , Encéfalo/metabolismo , Caspase 3/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/patologia , Humanos , Camundongos , Camundongos Endogâmicos MRL lpr , Peptídeos Cíclicos/farmacologia , Receptor da Anafilatoxina C5a/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos
8.
Am J Physiol Renal Physiol ; 310(9): F895-908, 2016 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-26887830

RESUMO

Thrombotic microangiopathy (TMA) is a disorder characterized by microvascular occlusion that can lead to thrombocytopenia, hemolytic anemia, and glomerular damage. Complement activation is the central event in most cases of TMA. Primary forms of TMA are caused by mutations in genes encoding components of the complement or regulators of the complement cascade. Recently, we and others have described a genetic form of TMA caused by mutations in the gene diacylglycerol kinase-ε (DGKE) that encodes the lipid kinase DGKε (Lemaire M, Fremeaux-Bacchi V, Schaefer F, Choi MR, Tang WH, Le Quintrec M, Fakhouri F, Taque S, Nobili F, Martinez F, Ji WZ, Overton JD, Mane SM, Nurnberg G, Altmuller J, Thiele H, Morin D, Deschenes G, Baudouin V, Llanas B, Collard L, Majid MA, Simkova E, Nurnberg P, Rioux-Leclerc N, Moeckel GW, Gubler MC, Hwa J, Loirat C, Lifton RP. Nat Genet 45: 531-536, 2013; Ozaltin F, Li BH, Rauhauser A, An SW, Soylemezoglu O, Gonul II, Taskiran EZ, Ibsirlioglu T, Korkmaz E, Bilginer Y, Duzova A, Ozen S, Topaloglu R, Besbas N, Ashraf S, Du Y, Liang CY, Chen P, Lu DM, Vadnagara K, Arbuckle S, Lewis D, Wakeland B, Quigg RJ, Ransom RF, Wakeland EK, Topham MK, Bazan NG, Mohan C, Hildebrandt F, Bakkaloglu A, Huang CL, Attanasio M. J Am Soc Nephrol 24: 377-384, 2013). DGKε is unrelated to the complement pathway, which suggests that unidentified pathogenic mechanisms independent of complement dysregulation may result in TMA. Studying Dgke knockout mice may help to understand the pathogenesis of this disease, but no glomerular phenotype has been described in these animals so far. Here we report that Dgke null mice present subclinical microscopic anomalies of the glomerular endothelium and basal membrane that worsen with age and develop glomerular capillary occlusion when exposed to nephrotoxic serum. We found that induction of cyclooxygenase-2 and of the proangiogenic prostaglandin E2 are impaired in Dgke null kidneys and are associated with reduced expression of the antithrombotic cell adhesion molecule platelet endothelial cell adhesion molecule-1/CD31 in the glomerular endothelium. Notably, prostaglandin E2 supplementation was able to rescue motility defects of Dgke knockdown cells in vitro and to restore angiogenesis in a test in vivo. Our results unveil an unexpected role of Dgke in the induction of cyclooxygenase-2 and in the regulation of glomerular prostanoids synthesis under stress.


Assuntos
Ciclo-Oxigenase 2/biossíntese , Diacilglicerol Quinase/genética , Dinoprostona/biossíntese , Endotélio/patologia , Glomerulonefrite/patologia , Glomérulos Renais/metabolismo , Glomérulos Renais/patologia , Envelhecimento/patologia , Animais , Movimento Celular , Glomerulonefrite/enzimologia , Glomerulonefrite/metabolismo , Testes de Função Renal , Glomérulos Renais/enzimologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neovascularização Fisiológica , Cicatrização
9.
Immunology ; 146(1): 130-43, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26059553

RESUMO

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.


Assuntos
Barreira Hematoencefálica/metabolismo , Complemento C5a/metabolismo , Lúpus Eritematoso Sistêmico/patologia , Receptor da Anafilatoxina C5a/metabolismo , Citoesqueleto de Actina/metabolismo , Transporte Ativo do Núcleo Celular , Adolescente , Astrócitos/imunologia , Encéfalo/irrigação sanguínea , Células Cultivadas , Criança , Claudina-5/biossíntese , Ativação do Complemento/imunologia , Complemento C5a/imunologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Impedância Elétrica , Células Endoteliais/imunologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Humanos , Moléculas de Adesão Juncional/biossíntese , Lúpus Eritematoso Sistêmico/imunologia , Masculino , NF-kappa B/metabolismo , Transporte Proteico , Receptor da Anafilatoxina C5a/antagonistas & inibidores , Receptor da Anafilatoxina C5a/imunologia , Junções Íntimas/metabolismo , Proteína da Zônula de Oclusão-1/biossíntese
10.
Kidney Int ; 87(5): 930-9, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25565310

RESUMO

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.


Assuntos
Antígeno CD11b/metabolismo , Fator H do Complemento/metabolismo , Glomerulonefrite/imunologia , Leucócitos/metabolismo , Receptores de IgG/metabolismo , Animais , Complexo Antígeno-Anticorpo/metabolismo , Apoferritinas/imunologia , Medula Óssea/metabolismo , Antígeno CD11b/genética , Antígenos CD18/metabolismo , Fator H do Complemento/genética , Glomerulonefrite/metabolismo , Imunoglobulina G/metabolismo , Camundongos Endogâmicos C57BL , Receptor da Anafilatoxina C5a/metabolismo , Receptores de IgG/genética , Doença do Soro/complicações
11.
Immunology ; 143(3): 363-73, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24801999

RESUMO

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.


Assuntos
Encéfalo/imunologia , Encéfalo/metabolismo , Complemento C5a/metabolismo , Células Endoteliais/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , MicroRNAs/genética , Animais , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/metabolismo , Linhagem Celular , Células Cultivadas , Complemento C5a/imunologia , Modelos Animais de Doenças , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/imunologia , Camundongos , Permeabilidade
12.
Kidney Int ; 86(4): 663-6, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25265946

RESUMO

Complement activation is actively regulated to prevent injudicious activation, such as on peritubular endothelia and basolateral aspects of tubules. Miao et al. studied mice in which the key complement regulator, Crry, was deleted from tubular cells. This lacked functional consequence in unmanipulated animals. Yet, following ischemia-reperfusion, there was greater injury due to alternative pathway activation of C5. When the balance between complement activation and regulation is tipped towards the former, pathologic complement activation can ensue.


Assuntos
Predisposição Genética para Doença , Receptores de Complemento/genética , Receptores de Complemento/metabolismo , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/patologia , Animais
13.
J Am Soc Nephrol ; 24(3): 377-84, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23274426

RESUMO

Renal microangiopathies and membranoproliferative GN (MPGN) can manifest similar clinical presentations and histology, suggesting the possibility of a common underlying mechanism in some cases. Here, we performed homozygosity mapping and whole exome sequencing in a Turkish consanguineous family and identified DGKE gene variants as the cause of a membranoproliferative-like glomerular microangiopathy. Furthermore, we identified two additional DGKE variants in a cohort of 142 unrelated patients diagnosed with membranoproliferative GN. This gene encodes the diacylglycerol kinase DGKε, which is an intracellular lipid kinase that phosphorylates diacylglycerol to phosphatidic acid. Immunofluorescence confocal microscopy demonstrated that mouse and rat Dgkε colocalizes with the podocyte marker WT1 but not with the endothelial marker CD31. Patch-clamp experiments in human embryonic kidney (HEK293) cells showed that DGKε variants affect the intracellular concentration of diacylglycerol. Taken together, these results not only identify a genetic cause of a glomerular microangiopathy but also suggest that the phosphatidylinositol cycle, which requires DGKE, is critical to the normal function of podocytes.


Assuntos
Diacilglicerol Quinase/genética , Glomerulonefrite Membranoproliferativa/enzimologia , Glomerulonefrite Membranoproliferativa/genética , Nefropatias/enzimologia , Nefropatias/genética , Mutação , Sequência de Aminoácidos , Animais , Sequência de Bases , Estudos de Coortes , Consanguinidade , DNA/genética , Diacilglicerol Quinase/metabolismo , Diagnóstico Diferencial , Diglicerídeos/metabolismo , Feminino , Variação Genética , Glomerulonefrite Membranoproliferativa/patologia , Células HEK293 , Humanos , Nefropatias/patologia , Glomérulos Renais/enzimologia , Masculino , Camundongos , Dados de Sequência Molecular , Linhagem , Podócitos/metabolismo , Polimorfismo de Nucleotídeo Único , Ratos , Homologia de Sequência de Aminoácidos , Turquia
14.
Immunology ; 139(3): 328-37, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23347386

RESUMO

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.


Assuntos
Anti-Inflamatórios não Esteroides/uso terapêutico , Curcumina/uso terapêutico , Glomerulonefrite/tratamento farmacológico , Doenças do Complexo Imune/tratamento farmacológico , Doença do Soro/tratamento farmacológico , Animais , Anti-Inflamatórios não Esteroides/administração & dosagem , Anti-Inflamatórios não Esteroides/farmacologia , Apoferritinas/administração & dosagem , Doença Crônica , Ativação do Complemento/efeitos dos fármacos , Fator H do Complemento/deficiência , Curcumina/administração & dosagem , Curcumina/farmacologia , Glomerulonefrite/etiologia , Humanos , Testes de Função Renal , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Resultado do Tratamento
15.
J Immunol ; 186(3): 1849-60, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21187439

RESUMO

The most prevalent severe manifestation of systemic lupus erythematosus is nephritis, which is characterized by immune complex deposition, inflammation, and scarring in glomeruli and the tubulointerstitium. Numerous studies indicated that glomerulonephritis results from a systemic break in B cell tolerance, resulting in the local deposition of immune complexes containing Abs reactive with ubiquitous self-Ags. However, the pathogenesis of systemic lupus erythematosus tubulointerstitial disease is not known. In this article, we demonstrate that in more than half of a cohort of 68 lupus nephritis biopsies, the tubulointerstitial infiltrate was organized into well-circumscribed T:B cell aggregates or germinal centers (GCs) containing follicular dendritic cells. Sampling of the in situ-expressed Ig repertoire revealed that both histological patterns were associated with intrarenal B cell clonal expansion and ongoing somatic hypermutation. However, in the GC histology, the proliferating cells were CD138(-)CD20(+) centroblasts, whereas they were CD138(+)CD20(low/-) plasmablasts in T:B aggregates. The presence of GCs or T:B aggregates was strongly associated with tubular basement membrane immune complexes. These data implicate tertiary lymphoid neogenesis in the pathogenesis of lupus tubulointerstitial inflammation.


Assuntos
Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/patologia , Túbulos Renais/imunologia , Túbulos Renais/patologia , Nefrite Lúpica/imunologia , Nefrite Lúpica/patologia , Imunidade Adaptativa , Adolescente , Adulto , Sequência de Aminoácidos , Sequência de Bases , Movimento Celular/genética , Movimento Celular/imunologia , Criança , Pré-Escolar , Células Clonais , Células Dendríticas Foliculares/imunologia , Células Dendríticas Foliculares/patologia , Feminino , Centro Germinativo/imunologia , Centro Germinativo/patologia , Humanos , Lactente , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Nefrite Lúpica/genética , Masculino , Dados de Sequência Molecular , Plasmócitos/imunologia , Plasmócitos/patologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/patologia , Adulto Jovem
16.
Clin Dev Immunol ; 2013: 836989, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24489579

RESUMO

Using a reversible UUO model (rUUO), we have demonstrated that C57BL/6 mice are susceptible to development of CKD after obstruction-mediated kidney injury while BALB/c mice are resistant. We hypothesized that selective systemic depletion of subpopulations of inflammatory cells during injury or repair might alter the development of CKD. To investigate the impact of modification of Th-lymphocytes or macrophage responses on development of CKD after rUUO, we used an anti-CD4 antibody (GK1.5) or liposomal clodronate to systemically deplete CD4(+) T cells or monocyte/macrophages, respectively, prior to and throughout the rUUO protocol. Flow cytometry and immunohistochemistry confirmed depletion of target cell populations. C57BL/6 mice treated with the GK1.5 antibody to deplete CD4(+) T cells had higher BUN levels and delayed recovery from rUUO. Treatment of C57BL/6 mice with liposomal clodronate to deplete monocyte/macrophages led to a relative protection from CKD as assessed by BUN values. Our results demonstrate that modulation of the inflammatory response during injury and repair altered the susceptibility of C57BL/6 mice to development of CKD in our rUUO model.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Depleção Linfocítica , Macrófagos/imunologia , Monócitos/imunologia , Insuficiência Renal Crônica/imunologia , Insuficiência Renal Crônica/patologia , Animais , Atrofia , Linfócitos T CD4-Positivos/metabolismo , Modelos Animais de Doenças , Fibrose , Imunofenotipagem , Terapia de Imunossupressão/métodos , Macrófagos/metabolismo , Masculino , Camundongos , Monócitos/metabolismo , Fenótipo , Obstrução Ureteral
17.
Am J Physiol Renal Physiol ; 302(12): F1529-36, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22492944

RESUMO

The complement cascade is an important part of the innate immune system, but pathological activation of this system causes tissue injury in several autoimmune and inflammatory diseases, including immune complex glomerulonephritis. We examined whether mice with targeted deletion of the gene for factor B (fB(-/-) mice) and selective deficiency in the alternative pathway of complement are protected from injury in the nephrotoxic serum (NTS) nephritis model of antibody-mediated glomerulonephritis. When the acute affects of the anti-glomerular basement membrane antibody were assessed, fB(-/-) mice developed a degree of injury similar to wild-type controls. If the mice were presensitized with sheep IgG or if the mice were followed for 5 mo postinjection, however, the fB(-/-) mice developed milder injury than wild-type mice. The immune response of fB(-/-) mice exposed to sheep IgG was similar to that of wild-type mice, but the fB(-/-) mice had less glomerular C3 deposition and lower levels of albuminuria. These results demonstrate that fB(-/-) mice are not significantly protected from acute heterologous injury in NTS nephritis but are protected from autologous injury in response to a planted glomerular antigen. Thus, although the glomerulus is resistant to antibody-initiated, alternative pathway-mediated injury, inhibition of this complement pathway may be beneficial in chronic immune complex-mediated diseases.


Assuntos
Via Alternativa do Complemento/fisiologia , Glomérulos Renais/imunologia , Nefrite/imunologia , Animais , Complemento C3/imunologia , Fator B do Complemento/genética , Modelos Animais de Doenças , Glomérulos Renais/patologia , Camundongos , Camundongos Knockout , Nefrite/patologia , Proteinúria/imunologia , Proteinúria/patologia
18.
Kidney Int ; 82(9): 961-8, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22832515

RESUMO

Chronic serum sickness leads to the formation of glomerular immune complexes; however, C57BL/6 mice do not develop glomerulonephritis unless complement factor H (CFH) is absent from the plasma. Here we studied the role for C5a receptor (R) in this setting. The exaggerated humoral immune response in CFH(-/-) mice was normalized in CFH(-/-)C5aR(-/-) double knockout mice, highlighting the C5aR dependence. The CFH knockout mice developed proliferative glomerulonephritis with endocapillary F4/80+ macrophage infiltration, a process reduced in the double knockout mice. There was no interstitial inflammation by histologic criteria or flow cytometry for F4/80+ Ly6C(hi)CCR2(hi) inflammatory macrophages. There were, however, more interstitial CD3+ CD4+ T lymphocytes in CFH knockout mice with chronic serum sickness, while double knockout mice had greater than 5-fold more Ly6C(lo)CCR2(lo) anti-inflammatory macrophages compared to the CFH knockout mice. Mice lacking C5aR were significantly protected from functional renal disease as assessed by blood urea nitrogen levels. Thus, IgG- and iC3b-containing immune complexes are not inflammatory in C57BL/6 mice. Yet when these mice lack CFH, sufficient C3b persists in glomeruli to generate C5a and activate C5aR.


Assuntos
Glomerulonefrite/imunologia , Doenças do Complexo Imune/imunologia , Nefropatias/imunologia , Receptor da Anafilatoxina C5a/genética , Receptor da Anafilatoxina C5a/imunologia , Animais , Fator H do Complemento/deficiência , Fator H do Complemento/genética , Fator H do Complemento/imunologia , Modelos Animais de Doenças , Glomerulonefrite/genética , Glomerulonefrite/patologia , Doenças da Deficiência Hereditária de Complemento , Doenças do Complexo Imune/genética , Doenças do Complexo Imune/patologia , Rim/imunologia , Rim/patologia , Nefropatias/genética , Nefropatias/patologia , Macrófagos/imunologia , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Doença do Soro/genética , Doença do Soro/imunologia , Doença do Soro/patologia , Linfócitos T/imunologia , Linfócitos T/patologia
19.
J Immunol ; 185(6): 3759-67, 2010 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-20702729

RESUMO

Complement receptor 1 (CR1) on human erythrocytes (Es) and complement factor H (CFH) on rodent platelets perform immune adherence, which is a function that allows the processing of immune complexes (ICs) bearing C3 by the mononuclear phagocyte system. Similar immune adherence occurs in the glomerular podocyte by CR1 in humans and CFH in rodents. As a model for human IC processing, we studied transgenic mice lacking CFH systemically but with human CR1 on Es. These CR1(hu)Tg/CFH(-/-) mice spontaneously developed proliferative glomerulonephritis, which was accelerated in a chronic serum sickness model by active immunization with heterologous apoferritin. ICs containing Ag, IgG and C3 bound to Es in CR1(hu)Tg/CFH(-/-) mice. In this setting, there was increased IC deposition in glomeruli, attributable to the presence of CR1 on Es, together with the absence of CFH on platelets and podocytes. In the absence of plasma CFH, the accumulated ICs activated complement, which led to spontaneous and chronic serum sickness-induced proliferative glomerulonephritis. These findings illustrate the complexities of complement-dependent IC processing by blood cells and in the glomerulus, and the importance of CFH as a plasma complement regulator.


Assuntos
Fator H do Complemento/deficiência , Eritrócitos/imunologia , Glomerulonefrite Membranoproliferativa/genética , Glomerulonefrite Membranoproliferativa/imunologia , Doenças do Complexo Imune/genética , Doenças do Complexo Imune/imunologia , Processamento de Proteína Pós-Traducional/imunologia , Receptores de Complemento 3b/sangue , Animais , Plaquetas/imunologia , Plaquetas/metabolismo , Plaquetas/patologia , Ativação do Complemento/genética , Ativação do Complemento/imunologia , Fator H do Complemento/genética , Modelos Animais de Doenças , Eritrócitos/metabolismo , Eritrócitos/patologia , Glomerulonefrite Membranoproliferativa/sangue , Glomerulonefrite Membranoproliferativa/patologia , Humanos , Doenças do Complexo Imune/sangue , Doenças do Complexo Imune/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Podócitos/imunologia , Podócitos/metabolismo , Podócitos/patologia , Processamento de Proteína Pós-Traducional/genética , Receptores de Complemento 3b/genética , Doença do Soro/sangue , Doença do Soro/genética , Doença do Soro/imunologia , Índice de Gravidade de Doença
20.
J Am Soc Nephrol ; 22(2): 285-95, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21148254

RESUMO

Complement factor H (CfH) is a key regulator of the alternative pathway, and its presence on mouse platelets and podocytes allows the processing of immune complexes. Because of the role of immune complexes in the pathophysiology of lupus nephritis, we studied the role of CfH in the development of nephritis in MRL-lpr mice, an animal model of lupus. At 12 weeks, CfH-deficient MRL-lpr mice had significantly more albuminuria and higher BUN levels than MRL-lpr controls. Cfh-deficient MRL-lpr mice also experienced earlier mortality: at 14 weeks, 6 of 9 CfH-deficient MRL-lpr mice had died of renal failure, whereas all 11 littermate CfH-sufficient MRL-lpr mice were alive (P ≤ 0.001). Histologically, CfH-deficient MRL-lpr mice developed severe diffuse lupus nephritis by 12 weeks (glomerulonephritis scores of 2.6 ± 0.4 versus 0.4 ± 0.2 in littermate controls, P = 0.001). Similar to other CfH-deficient mouse models on nonautoimmune backgrounds, immunofluorescence staining showed extensive linear C3 staining along glomerular capillary walls. IgG was present in the mesangium and peripheral capillary walls along with excessive infiltration of macrophages and neutrophils. Ultrastructurally, there were subendothelial and subepithelial immune deposits and extensive podocyte foot process effacement. In summary, the loss of CfH accelerates the development of lupus nephritis and recapitulates the functional and structural features of the human disease. This illustrates the critical role of complement regulation and metabolism of immune complexes in the pathogenesis of lupus nephritis.


Assuntos
Fator H do Complemento/deficiência , Nefrite Lúpica/etiologia , Animais , Anticorpos Antinucleares/sangue , Nitrogênio da Ureia Sanguínea , Antígeno CD11b/análise , Fator H do Complemento/fisiologia , Glomérulos Renais/ultraestrutura , Nefrite Lúpica/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos MRL lpr , Insuficiência Renal/etiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA