Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
J Am Soc Nephrol ; 27(5): 1334-42, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26374608

RESUMEN

The complement-mediated renal diseases C3 glomerulopathy (C3G) and atypical hemolytic uremic syndrome (aHUS) strongly associate with inherited and acquired abnormalities in the regulation of the complement alternative pathway (AP). The major negative regulator of the AP is the plasma protein complement factor H (FH). Abnormalities in FH result in uncontrolled activation of C3 through the AP and associate with susceptibility to both C3G and aHUS. Although previously developed FH-deficient animal models have provided important insights into the mechanisms underlying susceptibility to these unique phenotypes, these models do not entirely reproduce the clinical observations. FH is predominantly synthesized in the liver. We generated mice with hepatocyte-specific FH deficiency and showed that these animals have reduced plasma FH levels with secondary reduction in plasma C3. Unlike mice with complete FH deficiency, hepatocyte-specific FH-deficient animals developed neither plasma C5 depletion nor accumulation of C3 along the glomerular basement membrane. In contrast, subtotal FH deficiency associated with mesangial C3 accumulation consistent with C3G. Although there was no evidence of spontaneous thrombotic microangiopathy, the hepatocyte-specific FH-deficient animals developed severe C5-dependent thrombotic microangiopathy after induction of complement activation within the kidney by accelerated serum nephrotoxic nephritis. Taken together, our data indicate that subtotal FH deficiency can give rise to either spontaneous C3G or aHUS after a complement-activating trigger within the kidney and that the latter is C5 dependent.


Asunto(s)
Síndrome Hemolítico Urémico Atípico/etiología , Complemento C3 , Factor H de Complemento/deficiencia , Enfermedades Renales/etiología , Glomérulos Renales , Microangiopatías Trombóticas/etiología , Animales , Activación de Complemento , Femenino , Enfermedades por Deficiencia de Complemento Hereditario , Riñón/irrigación sanguínea , Enfermedades Renales/complicaciones , Masculino , Ratones
2.
J Am Soc Nephrol ; 24(1): 43-52, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23184055

RESUMEN

Complement factor H (CFH) is a negative regulator of the alternative pathway of complement, and properdin is the sole positive regulator. CFH-deficient mice (CFH(-/-)) develop uncontrolled C3 activation and spontaneous renal disease characterized by accumulation of C3 along the glomerular basement membrane, but the role of properdin in the pathophysiology is unknown. Here, we studied mice deficient in both CFH and properdin (CFH(-/-).P(-/-)). Although CFH(-/-) mice had plasma depleted of both C3 and C5, CFH(-/-).P(-/-) animals exhibited depletion of C3 predominantly, recapitulating the plasma complement profile observed in humans with properdin-independent C3 nephritic factors. Glomerular inflammation, thickening of the capillary wall, and glomerular C3 staining were significantly increased in CFH(-/-).P(-/-) compared with CFH(-/-) mice. We previously reported that exogenous CFH ameliorates C3 staining of the glomerular basement membrane and triggers the appearance of mesangial C3 deposits in CFH(-/-) mice; here, we show that these effects require properdin. In summary, during uncontrolled activation of C3 driven by complete CFH deficiency, properdin influences the intraglomerular localization of C3, suggesting that therapeutic inhibition of properdin would be detrimental in this setting.


Asunto(s)
Glomerulonefritis/etiología , Enfermedades Renales/complicaciones , Properdina/deficiencia , Animales , Complemento C3/metabolismo , Factor H de Complemento/deficiencia , Glomerulonefritis/metabolismo , Enfermedades por Deficiencia de Complemento Hereditario , Humanos , Glomérulos Renales/metabolismo , Ratones , Ratones Endogámicos C57BL
3.
Cell Rep ; 43(6): 114253, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38781074

RESUMEN

Diabetic kidney disease (DKD), the most common cause of kidney failure, is a frequent complication of diabetes and obesity, and yet to date, treatments to halt its progression are lacking. We analyze kidney single-cell transcriptomic profiles from DKD patients and two DKD mouse models at multiple time points along disease progression-high-fat diet (HFD)-fed mice aged to 90-100 weeks and BTBR ob/ob mice (a genetic model)-and report an expanding population of macrophages with high expression of triggering receptor expressed on myeloid cells 2 (TREM2) in HFD-fed mice. TREM2high macrophages are enriched in obese and diabetic patients, in contrast to hypertensive patients or healthy controls in an independent validation cohort. Trem2 knockout mice on an HFD have worsening kidney filter damage and increased tubular epithelial cell injury, all signs of worsening DKD. Together, our studies suggest that strategies to enhance kidney TREM2high macrophages may provide therapeutic benefits for DKD.


Asunto(s)
Nefropatías Diabéticas , Dieta Alta en Grasa , Riñón , Macrófagos , Glicoproteínas de Membrana , Ratones Noqueados , Obesidad , Receptores Inmunológicos , Animales , Receptores Inmunológicos/metabolismo , Receptores Inmunológicos/genética , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/genética , Macrófagos/metabolismo , Obesidad/metabolismo , Obesidad/patología , Obesidad/complicaciones , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/patología , Ratones , Riñón/patología , Riñón/metabolismo , Humanos , Masculino , Ratones Endogámicos C57BL , Femenino
4.
J Am Soc Nephrol ; 23(7): 1155-60, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22626820

RESUMEN

Controlled activation of the complement system, a key component of innate immunity, enables destruction of pathogens with minimal damage to host tissue. Complement factor H (CFH), which inhibits complement activation, and five CFH-related proteins (CFHR1-5) compose a family of structurally related molecules. Combined deletion of CFHR3 and CFHR1 is common and confers a protective effect in IgA nephropathy. Here, we report an autosomal dominant complement-mediated GN associated with abnormal increases in copy number across the CFHR3 and CFHR1 loci. In addition to normal copies of these genes, affected individuals carry a unique hybrid CFHR3-1 gene. In addition to identifying an association between these genetic observations and complement-mediated kidney disease, these results provide insight into the protective role of the combined deletion of CFHR3 and CFHR1 in IgA nephropathy.


Asunto(s)
Proteínas Sanguíneas/genética , Quimera/genética , Complemento C3/metabolismo , Proteínas Inactivadoras del Complemento C3b/genética , Glomerulonefritis por IGA/genética , Glomerulonefritis por IGA/metabolismo , Adolescente , Adulto , Biopsia , Preescolar , Femenino , Predisposición Genética a la Enfermedad/genética , Genotipo , Humanos , Riñón/metabolismo , Riñón/patología , Masculino , Persona de Mediana Edad , Linaje
5.
Nat Commun ; 14(1): 1293, 2023 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-36894557

RESUMEN

Systemic immunity supports lifelong brain function. Obesity posits a chronic burden on systemic immunity. Independently, obesity was shown as a risk factor for Alzheimer's disease (AD). Here we show that high-fat obesogenic diet accelerated recognition-memory impairment in an AD mouse model (5xFAD). In obese 5xFAD mice, hippocampal cells displayed only minor diet-related transcriptional changes, whereas the splenic immune landscape exhibited aging-like CD4+ T-cell deregulation. Following plasma metabolite profiling, we identified free N-acetylneuraminic acid (NANA), the predominant sialic acid, as the metabolite linking recognition-memory impairment to increased splenic immune-suppressive cells in mice. Single-nucleus RNA-sequencing revealed mouse visceral adipose macrophages as a potential source of NANA. In vitro, NANA reduced CD4+ T-cell proliferation, tested in both mouse and human. In vivo, NANA administration to standard diet-fed mice recapitulated high-fat diet effects on CD4+ T cells and accelerated recognition-memory impairment in 5xFAD mice. We suggest that obesity accelerates disease manifestation in a mouse model of AD via systemic immune exhaustion.


Asunto(s)
Enfermedad de Alzheimer , Ratones , Humanos , Animales , Enfermedad de Alzheimer/metabolismo , Ácido N-Acetilneuramínico , Ratones Transgénicos , Trastornos de la Memoria/etiología , Obesidad/complicaciones , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad
6.
Front Immunol ; 14: 1139358, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37063857

RESUMEN

Background: Kidney transplant recipients are currently treated with nonspecific immunosuppressants that cause severe systemic side effects. Current immunosuppressants were developed based on their effect on T-cell activation rather than the underlying mechanisms driving alloimmune responses. Thus, understanding the role of the intragraft microenvironment will help us identify more directed therapies with lower side effects. Methods: To understand the role of the alloimmune response and the intragraft microenvironment in cellular rejection progression, we conducted a Single nucleus RNA sequencing (snRNA-seq) on one human non-rejecting kidney allograft sample, one borderline sample, and T-cell mediated rejection (TCMR) sample (Banff IIa). We studied the differential gene expression and enriched pathways in different conditions, in addition to ligand-receptor (L-R) interactions. Results: Pathway analysis of T-cells in borderline sample showed enrichment for allograft rejection pathway, suggesting that the borderline sample reflects an early rejection. Hence, this allows for studying the early stages of cellular rejection. Moreover, we showed that focal adhesion (FA), IFNg pathways, and endomucin (EMCN) were significantly upregulated in endothelial cell clusters (ECs) of borderline compared to ECs TCMR. Furthermore, we found that pericytes in TCMR seem to favor endothelial permeability compared to borderline. Similarly, T-cells interaction with ECs in borderline differs from TCMR by involving DAMPS-TLRs interactions. Conclusion: Our data revealed novel roles of T-cells, ECs, and pericytes in cellular rejection progression, providing new clues on the pathophysiology of allograft rejection.


Asunto(s)
Trasplante de Riñón , Humanos , Trasplante de Riñón/efectos adversos , Interferón gamma , Adhesiones Focales , Riñón , Aloinjertos , Inmunosupresores , Rechazo de Injerto
7.
Am J Kidney Dis ; 60(1): 121-5, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22503529

RESUMEN

Acute poststreptococcal glomerulonephritis is a common cause of acute nephritis in children. Transient hypocomplementemia and complete recovery are typical, with only a minority developing chronic disease. We describe a young girl who developed persistent kidney disease and hypocomplementemia after a streptococcal throat infection. Kidney biopsy 1 year after presentation showed isolated glomerular complement C3 deposition, membranoproliferative changes, and subendothelial, intramembranous and occasional subepithelial electron-dense deposits consistent with C3 glomerulopathy. Complement gene screening revealed a heterozygous single nucleotide insertion in exon 4 of the complement factor H-related protein 5 gene (CFHR5), resulting in a premature stop codon. This variant was not detected in 198 controls. Serum CFHR5 levels were reduced. The mother and sister of the index patient were heterozygous for the sequence variant, with no overt evidence of kidney disease. We speculate that this heterozygous CFHR5 sequence variant is a risk factor for the development of chronic kidney disease after streptococcal infection.


Asunto(s)
Proteínas del Sistema Complemento/deficiencia , Proteínas del Sistema Complemento/genética , Glomerulonefritis Membranoproliferativa/genética , Infecciones Estreptocócicas/complicaciones , Niño , Enfermedad Crónica , Complemento C3/análisis , Proteínas del Sistema Complemento/análisis , Femenino , Mutación del Sistema de Lectura , Glomerulonefritis Membranoproliferativa/epidemiología , Glomerulonefritis Membranoproliferativa/etiología , Glomerulonefritis Membranoproliferativa/patología , Heterocigoto , Humanos , Riñón/patología , Linaje , Polimorfismo de Nucleótido Simple , Factores de Riesgo , Análisis de Secuencia de Proteína , Infecciones Estreptocócicas/inmunología
8.
J Hum Genet ; 57(7): 459-64, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22622361

RESUMEN

Atypical hemolytic uremic syndrome (aHUS) is a severe renal disorder that is associated with mutations in genes encoding proteins of the alternative complement pathway. Previously, we identified pathogenic variations in genes encoding complement regulators (CFH, CFI and MCP) in our aHUS cohort. In this study, we screened for mutations in the alternative pathway regulator CFHR5 in 65 aHUS patients by means of PCR on genomic DNA and sequence analysis. Potential pathogenicity of genetic alterations was determined by published data on CFHR5 variants, evolutionary conservation and in silico mutation prediction programs. Detection of serum CFHR5 was performed by western blot analysis and enzyme-linked immunosorbent assay. A potentially pathogenic sequence variation was found in CFHR5 in three patients (4.6%). All variations were located in short consensus repeats that might be involved in binding to C3b, heparin or C-reactive protein. The identified CFHR5 mutations require functional studies to determine their relevance to aHUS, but they might be candidates for an altered genetic profile predisposing to the disease.


Asunto(s)
Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Proteínas del Sistema Complemento/genética , Síndrome Hemolítico-Urémico/genética , Adolescente , Secuencia de Aminoácidos , Síndrome Hemolítico Urémico Atípico , Estudios de Casos y Controles , Niño , Complejo de Ataque a Membrana del Sistema Complemento/genética , Vía Alternativa del Complemento , Proteínas del Sistema Complemento/metabolismo , Secuencia Conservada , Análisis Mutacional de ADN/métodos , Femenino , Pruebas Genéticas/métodos , Síndrome Hemolítico-Urémico/metabolismo , Heterocigoto , Homocigoto , Humanos , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Polimorfismo de Nucleótido Simple , Adulto Joven
9.
iScience ; 25(4): 104097, 2022 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-35372810

RESUMEN

High-resolution spatial transcriptomics enables mapping of RNA expression directly from intact tissue sections; however, its utility for the elucidation of disease processes and therapeutically actionable pathways remains unexplored. We applied Slide-seqV2 to mouse and human kidneys, in healthy and distinct disease paradigms. First, we established the feasibility of Slide-seqV2 in tissue from nine distinct human kidneys, which revealed a cell neighborhood centered around a population of LYVE1+ macrophages. Second, in a mouse model of diabetic kidney disease, we detected changes in the cellular organization of the spatially restricted kidney filter and blood-flow-regulating apparatus. Third, in a mouse model of a toxic proteinopathy, we identified previously unknown, disease-specific cell neighborhoods centered around macrophages. In a spatially restricted subpopulation of epithelial cells, we discovered perturbations in 77 genes associated with the unfolded protein response. Our studies illustrate and experimentally validate the utility of Slide-seqV2 for the discovery of disease-specific cell neighborhoods.

10.
J Clin Invest ; 131(5)2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33444290

RESUMEN

Mutations affecting mitochondrial coenzyme Q (CoQ) biosynthesis lead to kidney failure due to selective loss of podocytes, essential cells of the kidney filter. Curiously, neighboring tubular epithelial cells are spared early in disease despite higher mitochondrial content. We sought to illuminate noncanonical, cell-specific roles for CoQ, independently of the electron transport chain (ETC). Here, we demonstrate that CoQ depletion caused by Pdss2 enzyme deficiency in podocytes results in perturbations in polyunsaturated fatty acid (PUFA) metabolism and the Braf/Mapk pathway rather than ETC dysfunction. Single-nucleus RNA-Seq from kidneys of Pdss2kd/kd mice with nephrotic syndrome and global CoQ deficiency identified a podocyte-specific perturbation of the Braf/Mapk pathway. Treatment with GDC-0879, a Braf/Mapk-targeting compound, ameliorated kidney disease in Pdss2kd/kd mice. Mechanistic studies in Pdss2-depleted podocytes revealed a previously unknown perturbation in PUFA metabolism that was confirmed in vivo. Gpx4, an enzyme that protects against PUFA-mediated lipid peroxidation, was elevated in disease and restored after GDC-0879 treatment. We demonstrate broader human disease relevance by uncovering patterns of GPX4 and Braf/Mapk pathway gene expression in tissue from patients with kidney diseases. Our studies reveal ETC-independent roles for CoQ in podocytes and point to Braf/Mapk as a candidate pathway for the treatment of kidney diseases.


Asunto(s)
Ataxia/metabolismo , Indenos/farmacología , Enfermedades Renales/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Enfermedades Mitocondriales/metabolismo , Debilidad Muscular/metabolismo , Podocitos/metabolismo , Proteínas Proto-Oncogénicas B-raf/metabolismo , Pirazoles/farmacología , Ubiquinona/deficiencia , Transferasas Alquil y Aril/genética , Transferasas Alquil y Aril/metabolismo , Animales , Ataxia/tratamiento farmacológico , Ataxia/genética , Ataxia/patología , Sistemas de Liberación de Medicamentos , Células HEK293 , Humanos , Enfermedades Renales/tratamiento farmacológico , Enfermedades Renales/genética , Enfermedades Renales/patología , Peroxidación de Lípido/genética , Sistema de Señalización de MAP Quinasas/genética , Ratones , Enfermedades Mitocondriales/tratamiento farmacológico , Enfermedades Mitocondriales/genética , Enfermedades Mitocondriales/patología , Debilidad Muscular/tratamiento farmacológico , Debilidad Muscular/genética , Debilidad Muscular/patología , Podocitos/patología , Proteínas Proto-Oncogénicas B-raf/genética , RNA-Seq , Ubiquinona/genética , Ubiquinona/metabolismo
11.
bioRxiv ; 2020 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-32637960

RESUMEN

Drug repurposing is the only method capable of delivering treatments on the shortened time-scale required for patients afflicted with lung disease arising from SARS-CoV-2 infection. Mucin-1 (MUC1), a membrane-bound molecule expressed on the apical surfaces of most mucosal epithelial cells, is a biochemical marker whose elevated levels predict the development of acute lung injury (ALI) and respiratory distress syndrome (ARDS), and correlate with poor clinical outcomes. In response to the pandemic spread of SARS-CoV-2, we took advantage of a high content screen of 3,713 compounds at different stages of clinical development to identify FDA-approved compounds that reduce MUC1 protein abundance. Our screen identified Fostamatinib (R788), an inhibitor of spleen tyrosine kinase (SYK) approved for the treatment of chronic immune thrombocytopenia, as a repurposing candidate for the treatment of ALI. In vivo , Fostamatinib reduced MUC1 abundance in lung epithelial cells in a mouse model of ALI. In vitro , SYK inhibition by Fostamatinib promoted MUC1 removal from the cell surface. Our work reveals Fostamatinib as a repurposing drug candidate for ALI and provides the rationale for rapidly standing up clinical trials to test Fostamatinib efficacy in patients with COVID-19 lung injury.

12.
Cell Rep Med ; 1(8): 100137, 2020 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-33294858

RESUMEN

Drug repurposing has the advantage of identifying potential treatments on a shortened timescale. In response to the pandemic spread of SARS-CoV-2, we took advantage of a high-content screen of 3,713 compounds at different stages of clinical development to identify FDA-approved compounds that reduce mucin-1 (MUC1) protein abundance. Elevated MUC1 levels predict the development of acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) and correlate with poor clinical outcomes. Our screen identifies fostamatinib (R788), an inhibitor of spleen tyrosine kinase (SYK) approved for the treatment of chronic immune thrombocytopenia, as a repurposing candidate for the treatment of ALI. In vivo, fostamatinib reduces MUC1 abundance in lung epithelial cells in a mouse model of ALI. In vitro, SYK inhibition by the active metabolite R406 promotes MUC1 removal from the cell surface. Our work suggests fostamatinib as a repurposing drug candidate for ALI.

13.
Adv Chronic Kidney Dis ; 19(2): 84-92, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22449345

RESUMEN

The role of the complement system in renal disease has long been recognized, but there have been major advances in our understanding of its role over the past decade. Complement plays a critical role not only in host's defense against infection and preventing damage to "self" tissues but also mediates tissue injury, both in the glomerulus and tubulointerstitium. Although injury may originate in the glomerulus, resulting proteinuria and complement activation within the tubular lumen may lead to tubulointerstitial damage and progressive renal disease. Recent advances in our understanding of the mechanisms by which complement mediates renal injury have led to the development of promising strategies with which complement may be targeted to prevent renal injury and its associated complications.


Asunto(s)
Activación de Complemento/inmunología , Proteínas del Sistema Complemento/inmunología , Glomerulonefritis/inmunología , Animales , Anticuerpos Anticitoplasma de Neutrófilos/inmunología , Síndrome Hemolítico Urémico Atípico , Enfermedad Crónica , Diabetes Mellitus/inmunología , Progresión de la Enfermedad , Femenino , Síndrome Hemolítico-Urémico/inmunología , Humanos , Inmunoglobulina A/inmunología , Lupus Eritematoso Sistémico/inmunología , Masculino , Ratones , Nefritis Intersticial/inmunología , Ratas
14.
Contrib Nephrol ; 169: 198-210, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21252520

RESUMEN

Membranoproliferative glomerulonephritis (MPGN) is characterised by mesangial expansion and hypercellularity and capillary wall thickening with capillary wall and mesangial deposits of immunoglobulin and/or complement. Two main forms are described in humans: MPGN type I with subendothelial and mesangial electron-dense deposits on electron microscopy, and MPGN type II, or dense deposit disease, with electron dense transformation of the glomerular capillary wall. Spontaneous MPGN type I has been described in dogs and sheep in association with C3 deficiency. Induced models of MPGN type I have been described in mice with cryoglobulinaemia. Glomerulonephritis resembling MPGN type II has occurred spontaneously in pigs that have a genetic deficiency of the complement control protein factor H. The animals develop capillary wall deposits of C3 before birth. Mice have been genetically engineered with a deficiency of factor H and similarly develop glomerular capillary wall C3 with MPGN. This model has been used to study both pathogenesis and therapeutic interventions. In particular, MPGN associated with factor H deficiency is absolutely dependent on both the ability to activate C3 and on the ability of factor I to cleave C3b. There is an important role for C5 activation in the development of glomerular inflammation in this model. Factor H dysfunction is associated with an increased susceptibility to complement-activating nephrotoxic insults and in these scenarios C5 activation appears to play a major role in mediating glomerular injury.


Asunto(s)
Modelos Animales de Enfermedad , Glomerulonefritis Membranoproliferativa/fisiopatología , Animales , Complemento C3/deficiencia , Complemento C3/fisiología , Complemento C5/fisiología , Factor H de Complemento/deficiencia , Factor H de Complemento/fisiología , Perros , Humanos , Ratones , Porcinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA