Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Nat Commun ; 13(1): 7891, 2022 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-36550108

RESUMEN

Type 1 diabetes affects over nine million individuals globally, with approximately 40% developing diabetic kidney disease. Emerging evidence suggests that epigenetic alterations, such as DNA methylation, are involved in diabetic kidney disease. Here we assess differences in blood-derived genome-wide DNA methylation associated with diabetic kidney disease in 1304 carefully characterised individuals with type 1 diabetes and known renal status from two cohorts in the United Kingdom-Republic of Ireland and Finland. In the meta-analysis, we identify 32 differentially methylated CpGs in diabetic kidney disease in type 1 diabetes, 18 of which are located within genes differentially expressed in kidneys or correlated with pathological traits in diabetic kidney disease. We show that methylation at 21 of the 32 CpGs predict the development of kidney failure, extending the knowledge and potentially identifying individuals at greater risk for diabetic kidney disease in type 1 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1 , Nefropatías Diabéticas , Humanos , Metilación de ADN/genética , Epigenoma , Nefropatías Diabéticas/genética , Epigénesis Genética , Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 1/genética , Biomarcadores , ADN , Estudio de Asociación del Genoma Completo , Islas de CpG
2.
Front Med (Lausanne) ; 8: 642318, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34513853

RESUMEN

Objective: We aimed to use SARS-CoV-2 antibody tests to assess the asymptomatic seroprevalence of individuals in high-risk hospital cohorts who's previous COVID-19 exposure is unknown; staff, and patients requiring haemodialysis or chemotherapy after the first wave. Methods: In a single Center, study participants had five SARS-CoV-2 antibody tests done simultaneously; one rapid diagnostic test (RDT) (Superbio Colloidal Gold IgM/IgG), and four laboratory tests (Roche Elecsys® Anti-SARS-CoV-2 IgG [RE], Abbott Architect i2000SR IgG [AAr], Abbott Alinity IgG [AAl], and Abbott Architect IgM CMIA). To determine seroprevalence, only positive test results on laboratory assay were considered true positives. Results: There were 157 participants, of whom 103 (65.6%) were female with a median age of 50 years (range 19-90). The IgG component of the RDT showed a high number of false positives (n = 18), was inferior to the laboratory assays (p < 0.001 RDT vs. AAl/AAr, p < 0.001 RDT vs. RE), and had reduced specificity (85.5% vs. AAl/AAr, 87.2% vs. RE). Sero-concordance was 97.5% between IgG laboratory assays (RE vs. AAl/AAr). Specificity of the IgM component of the RDT compared to Abbott IgM CMIA was 95.4%. Ten participants had positivity in at least one laboratory assay, seven (9.9%) of which were seen in HCWs. Two (4.1%) hematology/oncology (H/O) patients and a single (2.7%) haemodialysis (HD) were asymptomatically seropositive. Asymptomatic seroprevalence of HCWs compared to patients was not significant (p = 0.105). Conclusion: HCWs (9.9%) had higher, although non-significant asymptomatic seroprevalence of SARS-CoV-2 antibodies compared to high-risk patients (H/O 4.1%, HD 2.7%). An IgM/IgG rapid diagnostic test was inferior to laboratory assays. Sero-concordance of 97.5% was found between IgG laboratory assays, RE vs. AAl/AAr.

3.
Artículo en Inglés | MEDLINE | ID: mdl-32723786

RESUMEN

High-throughput DNA testing is becoming established as a standard diagnostic test in the renal clinic. Previously published studies on cohorts of patients with unexplained chronic kidney disease of a suspected genetic aetiology have suggested a diagnostic yield for genomic sequencing of up to 18%. Here we determine the yield of targeted gene panel in a clinically unscreened cohort of patients referred for percutaneous native renal biopsy. Patients who underwent renal biopsy for investigation of chronic kidney disease were sequenced using a genomic sequencing panel covering 227 genes in which variation is known to be associated with monogenic chronic kidney disease (CKD). Candidate disease-causing variants were assessed for pathogenicity using guidelines from the American College for Medical Genetics and Genomics. Fifty CKD patients were recruited and sequenced. A molecular diagnosis was obtained for two patients (4%). A molecular diagnosis is possible using genomic testing in ∼4% of clinically unscreened patients undergoing renal biopsy. Genetic screening may be useful for diagnosis in a subset of CKD patients but is most valuable when applied to patients with suspected heritable forms of kidney disease.


Asunto(s)
Insuficiencia Renal Crónica/diagnóstico , Insuficiencia Renal Crónica/genética , Adulto , Anciano , Biopsia , Estudios de Cohortes , Pruebas Diagnósticas de Rutina/tendencias , Progresión de la Enfermedad , Femenino , Pruebas Genéticas , Genómica , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/tendencias , Humanos , Riñón/patología , Masculino , Persona de Mediana Edad
4.
Semin Immunol ; 40: 36-48, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30293857

RESUMEN

Inflammation is an essential response to injury and its timely and adequate resolution permits tissue repair and avoidance of chronic inflammation. Ageing is associated with increased inflammation, sub-optimal resolution and these act as drivers for a number of ageing-associated pathologies. We describe the role played by specialised proresolving lipid mediators (SPMs) in the resolution of inflammation and how insufficient levels of these mediators, or compromised responsiveness may play a role in the pathogenesis of many ageing-associated pathologies, e.g. Alzheimer's Disease, atherosclerosis, obesity, diabetes and kidney disease. Detailed examination of the resolution phase of inflammation highlights the potential to harness these lipid mediators and or mimetics of their bioactions, in particular, their synthetic analogues to promote effective resolution of inflammation, without compromising the host immune system.


Asunto(s)
Envejecimiento , Enfermedad de Alzheimer/inmunología , Aterosclerosis/inmunología , Inmunoterapia/métodos , Inflamación/terapia , Lípidos/inmunología , Obesidad/inmunología , Animales , Ácidos Docosahexaenoicos/metabolismo , Humanos , Mediadores de Inflamación/uso terapéutico , Lípidos/uso terapéutico , Lipoxinas/metabolismo
5.
J Thorac Cardiovasc Surg ; 150(6): 1622-8, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26409993

RESUMEN

OBJECTIVE: Postoperative acute kidney injury is a frequent and serious consequence of cardiac surgery. We undertook to investigate the association of obesity and the risk of acute kidney injury development after cardiac surgery. METHODS: A total of 432 patients who underwent cardiac surgery with cardiopulmonary bypass between October 2009 and August 2010 were included in the final retrospective analysis. Obesity was defined as body mass index 30 kg/m(2) or greater. Acute kidney injury was defined as a creatinine increase of 25% or more from baseline at 48 hours after surgery. RESULTS: The overall incidence of acute kidney injury was 29.9% (n = 129). There was an increased incidence of postoperative renal impairment in the obese versus nonobese cohort; however, this was not statistically significant (39% vs 25.9%, P = .007). Multivariate logistic regression revealed that body mass index 30 kg/m(2) or greater was independently associated with the development of postoperative acute kidney injury (odds ratio [OR], 2.12; 95% confidence interval [CI], 1.27-3.54; P = .004), as were age (OR, 0.98; 95% CI, 0.96-1.0; P = .04) and cardiopulmonary bypass time (OR, 0.99; 95% CI, 0.98-1.0; P = .048). CONCLUSIONS: Obesity with body mass index 30 kg/m(2) or greater is independently associated with an increased risk of acute kidney injury after cardiac surgery. Further understanding of the molecular basis of this association is critical to the design of preventative strategies.


Asunto(s)
Lesión Renal Aguda/etiología , Procedimientos Quirúrgicos Cardíacos/efectos adversos , Obesidad/complicaciones , Anciano , Índice de Masa Corporal , Puente Cardiopulmonar , Femenino , Humanos , Incidencia , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Factores de Riesgo
6.
Diabetes ; 64(12): 4238-46, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26307587

RESUMEN

Obesity has been posited as an independent risk factor for diabetic kidney disease (DKD), but establishing causality from observational data is problematic. We aimed to test whether obesity is causally related to DKD using Mendelian randomization, which exploits the random assortment of genes during meiosis. In 6,049 subjects with type 1 diabetes, we used a weighted genetic risk score (GRS) comprised of 32 validated BMI loci as an instrument to test the relationship of BMI with macroalbuminuria, end-stage renal disease (ESRD), or DKD defined as presence of macroalbuminuria or ESRD. We compared these results with cross-sectional and longitudinal observational associations. Longitudinal analysis demonstrated a U-shaped relationship of BMI with development of macroalbuminuria, ESRD, or DKD over time. Cross-sectional observational analysis showed no association with overall DKD, higher odds of macroalbuminuria (for every 1 kg/m(2) higher BMI, odds ratio [OR] 1.05, 95% CI 1.03-1.07, P < 0.001), and lower odds of ESRD (OR 0.95, 95% CI 0.93-0.97, P < 0.001). Mendelian randomization analysis showed a 1 kg/m(2) higher BMI conferring an increased risk in macroalbuminuria (OR 1.28, 95% CI 1.11-1.45, P = 0.001), ESRD (OR 1.43, 95% CI 1.20-1.72, P < 0.001), and DKD (OR 1.33, 95% CI 1.17-1.51, P < 0.001). Our results provide genetic evidence for a causal link between obesity and DKD in type 1 diabetes. As obesity prevalence rises, this finding predicts an increase in DKD prevalence unless intervention should occur.


Asunto(s)
Albuminuria/etiología , Diabetes Mellitus Tipo 1/complicaciones , Nefropatías Diabéticas/etiología , Fallo Renal Crónico/etiología , Obesidad/fisiopatología , Adulto , Albuminuria/epidemiología , Albuminuria/genética , Índice de Masa Corporal , Estudios de Casos y Controles , Estudios de Cohortes , Estudios Transversales , Nefropatías Diabéticas/epidemiología , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/fisiopatología , Femenino , Finlandia/epidemiología , Sitios Genéticos , Predisposición Genética a la Enfermedad , Humanos , Incidencia , Fallo Renal Crónico/epidemiología , Fallo Renal Crónico/genética , Estudios Longitudinales , Masculino , Análisis de la Aleatorización Mendeliana , Persona de Mediana Edad , Obesidad/complicaciones , Obesidad/epidemiología , Obesidad/genética , Prevalencia , Factores de Riesgo
7.
Nephrol Dial Transplant ; 28 Suppl 4: iv73-82, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24071659

RESUMEN

Renal dysfunction and disease, including hyperfiltration, proteinuria and hypofiltration, are commonly associated with obesity. Diabetic kidney disease is also common in obese cohorts. Weight loss interventions, including bariatric surgery, can effectively reduce weight and improve renal outcomes. Some of this effect may be due to the remission of Type 2 diabetes and hypertension. However, other mechanisms, including the resolution of inflammatory processes, may also contribute. The effect of bariatric surgery on renal function has only recently become a focus of particular investigation. In this study, we will review the effects of bariatric surgery on obesity-associated kidney disease. We will discuss the pitfalls in assessing renal function in obese cohorts and will examine the effect of bariatric surgery on renal function and urinary protein excretion using different mechanisms. We will give particular attention to the evidence for bariatric surgery in cohorts with established renal disease and suggest future directions. In particular, we will outline the evidence for inflammation as an important therapeutic target, and the emerging medical therapies being considered to exploit this target in obesity- and diabetes-related kidney disease.


Asunto(s)
Cirugía Bariátrica , Inflamación/prevención & control , Enfermedades Renales/fisiopatología , Riñón/fisiopatología , Obesidad/cirugía , Animales , Humanos , Enfermedades Renales/etiología , Enfermedades Renales/prevención & control , Obesidad/fisiopatología
8.
J Am Soc Nephrol ; 24(10): 1537-43, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24029427

RESUMEN

Sex and genetic variation influence the risk of developing diabetic nephropathy and ESRD in patients with type 1 diabetes. We performed a genome-wide association study in a cohort of 3652 patients from the Finnish Diabetic Nephropathy (FinnDiane) Study with type 1 diabetes to determine whether sex-specific genetic risk factors for ESRD exist. A common variant, rs4972593 on chromosome 2q31.1, was associated with ESRD in women (P<5×10(-8)) but not in men (P=0.77). This association was replicated in the meta-analysis of three independent type 1 diabetes cohorts (P=0.02) and remained significant for women (P<5×10(-8); odds ratio, 1.81 [95% confidence interval, 1.47 to 2.24]) upon combined meta-analysis of the discovery and replication cohorts. rs4972593 is located between the genes that code for the Sp3 transcription factor, which interacts directly with estrogen receptor α and regulates the expression of genes linked to glomerular function and the pathogenesis of nephropathy, and the CDCA7 transcription factor, which regulates cell proliferation. Further examination revealed potential transcription factor-binding sites within rs4972593 and predicted eight estrogen-responsive elements within 5 kb of this locus. Moreover, we found sex-specific differences in the glomerular expression levels of SP3 (P=0.004). Overall, these results suggest that rs4972593 is a sex-specific genetic variant associated with ESRD in patients with type 1 diabetes and may underlie the sex-specific protection against ESRD.


Asunto(s)
Cromosomas Humanos Par 2/genética , Diabetes Mellitus Tipo 1/complicaciones , Nefropatías Diabéticas/genética , Fallo Renal Crónico/genética , Adulto , Estudios de Casos y Controles , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Persona de Mediana Edad , Proteínas Nucleares/genética , Caracteres Sexuales , Factor de Transcripción Sp3/genética
9.
J Am Soc Nephrol ; 24(4): 627-37, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23520204

RESUMEN

Lipoxins, which are endogenously produced lipid mediators, promote the resolution of inflammation, and may inhibit fibrosis, suggesting a possible role in modulating renal disease. Here, lipoxin A4 (LXA4) attenuated TGF-ß1-induced expression of fibronectin, N-cadherin, thrombospondin, and the notch ligand jagged-1 in cultured human proximal tubular epithelial (HK-2) cells through a mechanism involving upregulation of the microRNA let-7c. Conversely, TGF-ß1 suppressed expression of let-7c. In cells pretreated with LXA4, upregulation of let-7c persisted despite subsequent stimulation with TGF-ß1. In the unilateral ureteral obstruction model of renal fibrosis, let-7c upregulation was induced by administering an LXA4 analog. Bioinformatic analysis suggested that targets of let-7c include several members of the TGF-ß1 signaling pathway, including the TGF-ß receptor type 1. Consistent with this, LXA4-induced upregulation of let-7c inhibited both the expression of TGF-ß receptor type 1 and the response to TGF-ß1. Overexpression of let-7c mimicked the antifibrotic effects of LXA4 in renal epithelia; conversely, anti-miR directed against let-7c attenuated the effects of LXA4. Finally, we observed that several let-7c target genes were upregulated in fibrotic human renal biopsies compared with controls. In conclusion, these results suggest that LXA4-mediated upregulation of let-7c suppresses TGF-ß1-induced fibrosis and that expression of let-7c targets is dysregulated in human renal fibrosis.


Asunto(s)
Riñón/efectos de los fármacos , Riñón/patología , Lipoxinas/farmacología , MicroARNs/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Cadherinas/efectos de los fármacos , Cadherinas/metabolismo , Células Cultivadas , Fibronectinas/efectos de los fármacos , Fibronectinas/metabolismo , Fibrosis , Humanos , Riñón/metabolismo , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , MicroARNs/efectos de los fármacos , Receptor Notch1/efectos de los fármacos , Receptor Notch1/metabolismo , Transducción de Señal , Trombospondinas/efectos de los fármacos , Trombospondinas/metabolismo , Factor de Crecimiento Transformador beta1/efectos de los fármacos
10.
Genes (Basel) ; 4(4): 596-619, 2013 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-24705265

RESUMEN

Up to 40% of patients with type 1 and type 2 diabetes will develop diabetic nephropathy (DN), resulting in chronic kidney disease and potential organ failure. There is evidence for a heritable genetic susceptibility to DN, but despite intensive research efforts the causative genes remain elusive. Recently, genome-wide association studies have discovered several novel genetic variants associated with DN. The identification of such variants may potentially allow for early identification of at risk patients. Here we review the current understanding of the key molecular mechanisms and genetic architecture of DN, and discuss the merits of employing an integrative approach to incorporate datasets from multiple sources (genetics, transcriptomics, epigenetic, proteomic) in order to fully elucidate the genetic elements contributing to this serious complication of diabetes.

11.
PLoS Genet ; 8(9): e1002921, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23028342

RESUMEN

Diabetic kidney disease, or diabetic nephropathy (DN), is a major complication of diabetes and the leading cause of end-stage renal disease (ESRD) that requires dialysis treatment or kidney transplantation. In addition to the decrease in the quality of life, DN accounts for a large proportion of the excess mortality associated with type 1 diabetes (T1D). Whereas the degree of glycemia plays a pivotal role in DN, a subset of individuals with poorly controlled T1D do not develop DN. Furthermore, strong familial aggregation supports genetic susceptibility to DN. However, the genes and the molecular mechanisms behind the disease remain poorly understood, and current therapeutic strategies rarely result in reversal of DN. In the GEnetics of Nephropathy: an International Effort (GENIE) consortium, we have undertaken a meta-analysis of genome-wide association studies (GWAS) of T1D DN comprising ~2.4 million single nucleotide polymorphisms (SNPs) imputed in 6,691 individuals. After additional genotyping of 41 top ranked SNPs representing 24 independent signals in 5,873 individuals, combined meta-analysis revealed association of two SNPs with ESRD: rs7583877 in the AFF3 gene (P = 1.2 × 10(-8)) and an intergenic SNP on chromosome 15q26 between the genes RGMA and MCTP2, rs12437854 (P = 2.0 × 10(-9)). Functional data suggest that AFF3 influences renal tubule fibrosis via the transforming growth factor-beta (TGF-ß1) pathway. The strongest association with DN as a primary phenotype was seen for an intronic SNP in the ERBB4 gene (rs7588550, P = 2.1 × 10(-7)), a gene with type 2 diabetes DN differential expression and in the same intron as a variant with cis-eQTL expression of ERBB4. All these detected associations represent new signals in the pathogenesis of DN.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Nefropatías Diabéticas/genética , Receptores ErbB/genética , Fallo Renal Crónico , Proteínas Nucleares/genética , Diabetes Mellitus Tipo 1/complicaciones , Nefropatías Diabéticas/etiología , Nefropatías Diabéticas/patología , Fibrosis/genética , Fibrosis/metabolismo , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Fallo Renal Crónico/etiología , Fallo Renal Crónico/genética , Fallo Renal Crónico/patología , Túbulos Renales/metabolismo , Túbulos Renales/patología , Polimorfismo de Nucleótido Simple , Sitios de Carácter Cuantitativo/genética , Receptor ErbB-4 , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo
12.
Diabetes ; 61(8): 2187-94, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22721967

RESUMEN

We formed the GEnetics of Nephropathy-an International Effort (GENIE) consortium to examine previously reported genetic associations with diabetic nephropathy (DN) in type 1 diabetes. GENIE consists of 6,366 similarly ascertained participants of European ancestry with type 1 diabetes, with and without DN, from the All Ireland-Warren 3-Genetics of Kidneys in Diabetes U.K. and Republic of Ireland (U.K.-R.O.I.) collection and the Finnish Diabetic Nephropathy Study (FinnDiane), combined with reanalyzed data from the Genetics of Kidneys in Diabetes U.S. Study (U.S. GoKinD). We found little evidence for the association of the EPO promoter polymorphism, rs161740, with the combined phenotype of proliferative retinopathy and end-stage renal disease in U.K.-R.O.I. (odds ratio [OR] 1.14, P = 0.19) or FinnDiane (OR 1.06, P = 0.60). However, a fixed-effects meta-analysis that included the previously reported cohorts retained a genome-wide significant association with that phenotype (OR 1.31, P = 2 × 10(-9)). An expanded investigation of the ELMO1 locus and genetic regions reported to be associated with DN in the U.S. GoKinD yielded only nominal statistical significance for these loci. Finally, top candidates identified in a recent meta-analysis failed to reach genome-wide significance. In conclusion, we were unable to replicate most of the previously reported genetic associations for DN, and significance for the EPO promoter association was attenuated.


Asunto(s)
Diabetes Mellitus Tipo 1/epidemiología , Nefropatías Diabéticas/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Adulto , Estudios de Casos y Controles , Diabetes Mellitus Tipo 1/genética , Nefropatías Diabéticas/epidemiología , Eritropoyetina/genética , Finlandia/epidemiología , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Irlanda/epidemiología , Fallo Renal Crónico/genética , Fenotipo , Regiones Promotoras Genéticas/genética , Estados Unidos/epidemiología , Población Blanca/genética
13.
Biochim Biophys Acta ; 1822(4): 589-99, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22266139

RESUMEN

Transforming growth factor-beta (TGF-ß1) is implicated in the onset and progression of renal fibrosis and diabetic nephropathy (DN), leading to a loss of epithelial characteristics of tubular cells. The transcriptional profile of renal tubular epithelial cells stimulated with TGF-ß1 was assessed using RNA-Seq, with 2027 differentially expressed genes identified. Promoter analysis of transcription factor binding sites in the TGF-ß1 responsive gene set predicted activation of multiple transcriptional networks, including NFκB. Comparison of RNA-Seq with microarray data from identical experimental conditions identified low abundance transcripts exclusive to RNA-Seq data. We compared these findings to human disease by analyzing transcriptomic data from renal biopsies of patients with DN versus control groups, identifying a shared subset of 179 regulated genes. ARK5, encoding an AMP-related kinase, and TGFBI - encoding transforming growth factor, beta-induced protein were induced by TGF-ß1 and also upregulated in human DN. Suppression of ARK5 attenuated fibrotic responses of renal epithelia to TGF-ß1 exposure; and silencing of TGFBI induced expression of the epithelial cell marker - E-cadherin. We identified low abundance transcripts in sequence data and validated expression levels of several transcripts (ANKRD56, ENTPD8) in tubular enriched kidney biopsies of DN patients versus living donors. In conclusion, we have defined a TGF-ß1-driven pro-fibrotic signal in renal epithelial cells that is also evident in the DN renal transcriptome.


Asunto(s)
Nefropatías Diabéticas/patología , Células Epiteliales/patología , Perfilación de la Expresión Génica , Riñón/metabolismo , Factor de Crecimiento Transformador beta1/fisiología , Western Blotting , Línea Celular , Nefropatías Diabéticas/genética , Humanos , Riñón/patología , Reacción en Cadena de la Polimerasa
14.
Blood ; 119(2): 591-601, 2012 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-22058112

RESUMEN

Genomic disorders affecting the genes encoding factor H (fH) and the 5 factor H related proteins have been described in association with atypical hemolytic uremic syndrome. These include deletions of CFHR3, CFHR1, and CFHR4 in association with fH autoantibodies and the formation of a hybrid CFH/CFHR1 gene. These occur through nonallelic homologous recombination secondary to the presence of large segmental duplications (macrohomology) in this region. Using multiplex ligation-dependent probe amplification to screen for such genomic disorders, we have identified a large atypical hemolytic uremic syndrome family where a deletion has occurred through microhomology-mediated end joining rather than nonallelic homologous recombination. In the 3 affected persons of this family, we have shown that the deletion results in formation of a CFH/CFHR3 gene. We have shown that the protein product of this is a 24 SCR protein that is secreted with normal fluid-phase activity but marked loss of complement regulation at cell surfaces despite increased heparin binding. In this study, we have therefore shown that microhomology in this area of chromosome 1 predisposes to disease associated genomic disorders and that the complement regulatory function of fH at the cell surface is critically dependent on the structural integrity of the whole molecule.


Asunto(s)
Apolipoproteínas/genética , Proteínas Sanguíneas/genética , Proteínas Inactivadoras del Complemento C3b/genética , Factor H de Complemento/genética , Eliminación de Gen , Predisposición Genética a la Enfermedad , Síndrome Hemolítico-Urémico/genética , Animales , Apolipoproteínas/metabolismo , Síndrome Hemolítico Urémico Atípico , Autoanticuerpos , Secuencia de Bases , Proteínas Sanguíneas/metabolismo , Western Blotting , Activación de Complemento , Proteínas Inactivadoras del Complemento C3b/metabolismo , Factor H de Complemento/metabolismo , Eritrocitos/metabolismo , Hemólisis , Síndrome Hemolítico-Urémico/metabolismo , Síndrome Hemolítico-Urémico/patología , Recombinación Homóloga , Humanos , Datos de Secuencia Molecular , Proteínas Mutantes Quiméricas/genética , Proteínas Mutantes Quiméricas/metabolismo , Mutación/genética , Linaje , Homología de Secuencia de Ácido Nucleico , Ovinos , Resonancia por Plasmón de Superficie
15.
BMC Cancer ; 11: 50, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21284875

RESUMEN

BACKGROUND: NET1, a RhoA guanine exchange factor, is up-regulated in gastric cancer (GC) tissue and drives the invasive phenotype of this disease. In this study, we aimed to determine the role of NET1 in GC by monitoring the proliferation, motility and invasion of GC cells in which NET1 has been stably knocked down. Additionally, we aimed to determine NET1-dependent transcriptomic events that occur in GC. METHODS: An in vitro model of stable knockdown of NET1 was achieved in AGS human gastric adenocarcinoma cells via lentiviral mediated transduction of short-hairpin (sh) RNA targeting NET1. Knockdown was assessed using quantitative PCR. Cell proliferation was assessed using an MTS assay and cell migration was assessed using a wound healing scratch assay. Cell invasion was assessed using a transwell matrigel invasion assay. Gene expression profiles were examined using affymetrix oligonucleotide U133A expression arrays. A student's t test was used to determine changes of statistical significance. RESULTS: GC cells were transduced with NET1 shRNA resulting in a 97% reduction in NET1 mRNA (p < 0.0001). NET1 knockdown significantly reduced the invasion and migration of GC cells by 94% (p < 0.05) and 24% (p < 0.001) respectively, while cell proliferation was not significantly altered following NET1 knockdown. Microarray analysis was performed on non-target and knockdown cell lines, treated with and without 10 µM lysophosphatidic acid (LPA) allowing us to identify NET1-dependent, LPA-dependent and NET1-mediated LPA-induced gene transcription. Differential gene expression was confirmed by quantitative PCR. Shortlisted NET1-dependent genes included STAT1, TSPAN1, TGFBi and CCL5 all of which were downregulatd upon NET1 downregulation. Shortlisted LPA-dependent genes included EGFR and PPARD where EGFR was upregulated and PPARD was downregulated upon LPA stimulation. Shortlisted NET1 and LPA dependent genes included IGFR1 and PIP5K3. These LPA induced genes were downregulated in NET1 knockdown cells. CONCLUSIONS: NET1 plays an important role in GC cell migration and invasion, key aspects of GC progression. Furthermore, the gene expression profile further elucidates the molecular mechanisms underpinning NET1-mediated aggressive GC cell behaviour.


Asunto(s)
Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/metabolismo , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Adenocarcinoma/patología , Procesos de Crecimiento Celular/fisiología , Movimiento Celular/fisiología , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Invasividad Neoplásica , Proteínas Oncogénicas/biosíntesis , Proteínas Oncogénicas/deficiencia , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Reproducibilidad de los Resultados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Neoplasias Gástricas/patología , Células Tumorales Cultivadas
16.
FEBS Lett ; 585(3): 531-8, 2011 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-21237163

RESUMEN

We describe the activation of Wnt signalling in mesangial cells by CCN2. CCN2 stimulates phosphorylation of LRP6 and GSK-3ß resulting in accumulation and nuclear localisation of ß-catenin, TCF/LEF activity and expression of Wnt targets. This is coincident with decreased phosphorylation of ß-catenin on Ser 33/37 and increased phosphorylation on Tyr142. DKK-1 and LRP6 siRNA reversed CCN2's effects. Microarray analyses of diabetic patients identified differentially expressed Wnt components. ß-Catenin is increased in type 1 diabetic and UUO mice and in in vitro models of hyperglycaemia and hypertension. These findings suggest that Wnt/CCN2 signalling plays a role in the pathogenesis of diabetic nephropathy.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Nefropatías Diabéticas/metabolismo , Proteínas Relacionadas con Receptor de LDL/metabolismo , Células Mesangiales/metabolismo , Transducción de Señal , Proteínas Wnt/metabolismo , Animales , Células Cultivadas , Factor de Crecimiento del Tejido Conjuntivo/genética , Diabetes Mellitus Tipo 1/inducido químicamente , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patología , Nefropatías Diabéticas/patología , Nefropatías Diabéticas/fisiopatología , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Hiperglucemia/metabolismo , Hipertensión/metabolismo , Corteza Renal/metabolismo , Corteza Renal/patología , Proteínas Relacionadas con Receptor de LDL/genética , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad , Masculino , Células Mesangiales/citología , Ratones , Ratones Endogámicos C57BL , Fosforilación , Transporte de Proteínas , ARN Interferente Pequeño , Obstrucción Ureteral/metabolismo , Obstrucción Ureteral/patología , beta Catenina/metabolismo
17.
Eur J Gastroenterol Hepatol ; 22(10): 1211-20, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20631625

RESUMEN

OBJECTIVES: This study explored gene expression differences in predicting response to pegylated interferon (IFN-PEG) and ribavirin (RBV) in hepatitis C infection. Current treatment for hepatitis C virus (HCV) with IFN-PEG alpha-2a/b and RBV is an expensive regimen with frequent significant side-effects where less than 60% of patients ultimately achieve a sustained virological response. Responders and nonresponders may not be identified for up to 6 months post-treatment. This dichotomy may be because of differences in the molecular genetic response. METHODS: Peripheral blood mononuclear cell samples were obtained from a cohort of 31 infected individuals within the first 24 h of treatment and the extracted RNA was hybridized to genome expression microarrays. Hepatitis C viral kinetics was also examined in these patients. The ability of differentially regulated genes to predict response to therapy was assessed with treatment outcome. RESULTS: Distinct patterns of gene expression distinguished responders from nonresponders to HCV treatment. The ultimate response to treatment with IFN-PEG and RBV was observed within the first 24 h of treatment by a greater drop in viral load (mean HCV RNA decline of 1.92+/-1.26 log10 IU/ml) in responders compared with nonresponders (P<0.007). Induced genes achieved maximal response within 12 h of therapy which coincided with a rapid decline in HCV RNA between 12 and 24 h. This study revealed that peripheral blood mononuclear cell metallothionein 2A, CCRL2, tumour necrosis factor-alpha-induced protein 6 (TNFAIP6) and IFN-induced protein with tetratricopeptide repeats 2 expression predicted viral treatment response to therapy verified by quantitative real time polymerase chain reaction. CONCLUSION: This study has identified a noninvasive gene microarray pattern and a set of verified genes to be predictive of hepatitis C patient response to IFN-PEG and RBV treatment within the first 24 h. The potential of this noninvasive diagnostic approach and identified genes as biomarkers of response to treatment warrants further investigation.


Asunto(s)
Hepatitis C Crónica , Interferón-alfa/uso terapéutico , Leucocitos Mononucleares , Ribavirina/uso terapéutico , Adulto , Antivirales/uso terapéutico , Farmacorresistencia Viral/genética , Femenino , Perfilación de la Expresión Génica , Hepatitis C Crónica/tratamiento farmacológico , Hepatitis C Crónica/genética , Hepatitis C Crónica/inmunología , Humanos , Interferón alfa-2 , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/fisiología , Leucocitos Mononucleares/virología , Masculino , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Valor Predictivo de las Pruebas , Proteínas Recombinantes , Factores de Tiempo
18.
Epigenetics ; 5(5): 396-401, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20458172

RESUMEN

We have previously identified differentially expressed genes in cell models of diabetic nephropathy and renal biopsies. Here we have performed quantitative DNA methylation profiling in cell models of diabetic nephropathy. Over 3,000 CpG units in the promoter regions of 192 candidate genes were assessed in unstimulated human mesangial cells (HMCs) and proximal tubular epithelial cells (PTCs) compared to HMCs or PTCs exposed to appropriate stimuli. A total of 301 CpG units across 38 genes (~20%) were identified as differentially methylated in unstimulated HMCs versus PTCs. Analysis of amplicon methylation values in unstimulated versus stimulated cell models failed to demonstrate a >20% difference between amplicons. In conclusion, our results demonstrate that (1) specific DNA methylation signatures are present in HMCs and PTCs, and (2) standard protocols for exposure of renal cells to stimuli that alter gene expression may be insufficient to replicate possible alterations in DNA methylation profiles in diabetic nephropathy.


Asunto(s)
Metilación de ADN , Nefropatías Diabéticas/genética , Perfilación de la Expresión Génica/métodos , Islas de CpG/genética , Epigénesis Genética , Células Epiteliales/metabolismo , Genoma Humano/genética , Humanos , Células Mesangiales/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas
19.
J Hum Genet ; 55(4): 248-51, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20203694

RESUMEN

Chemokine (C-C motif) ligand 5 (CCL5) and chemokine (C-C motif) receptor 5 are implicated in the pathogenesis of diabetic nephropathy (DN). We hypothesize that variants in these genes may be associated with DN. The CCL5 and chemokine receptor type 5 (CCR5) genes were resequenced, variants identified (n=58), allele frequencies determined in 46 individuals (92 chromosomes) and efficient haplotype tag single-nucleotide polymorphisms (htSNPs) selected to effectively evaluate the common variation in these genes. One reportedly functional gene variant and eight htSNPs were genotyped in a case-control association study involving Caucasian individuals with type 1 diabetes (267 cases with DN and 442 non-nephropathic diabetic controls). Genotyping was performed using MassARRAY iPLEX, TaqMan, gel electrophoresis and direct capillary sequencing. After correction for multiple testing, there were no statistically significant associations between variants in the CCL5 and CCR5 genes and DN.


Asunto(s)
Quimiocina CCL5/genética , Nefropatías Diabéticas/genética , Polimorfismo de Nucleótido Simple , Receptores CCR5/genética , Alelos , Frecuencia de los Genes , Estudios de Asociación Genética , Variación Genética , Genotipo , Humanos , Análisis de Secuencia de ADN
20.
J Am Soc Nephrol ; 21(5): 773-81, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20150533

RESUMEN

Gremlin, a cell growth and differentiation factor, promotes the development of diabetic nephropathy in animal models, but whether GREM1 gene variants associate with diabetic nephropathy is unknown. We comprehensively screened the 5' upstream region (including the predicted promoter), all exons, intron-exon boundaries, complete untranslated regions, and the 3' region downstream of the GREM1 gene. We identified 31 unique variants, including 24 with a minor allele frequency exceeding 5%, and 9 haplotype-tagging single nucleotide polymorphisms (htSNPs). We selected one additional variant that we predicted to alter transcription factor binding. We genotyped 709 individuals with type 1 diabetes of whom 267 had nephropathy (cases) and 442 had no evidence of kidney disease (controls). Three individual SNPs significantly associated with nephropathy at the 5% level, and two remained significant after adjustment for multiple testing. Subsequently, we genotyped a replicate population comprising 597 cases and 502 controls: this population supported an association with one of the SNPs (rs1129456; P = 0.0003). Combined analysis, adjusted for recruitment center (n = 8), suggested that the T allele conferred greater odds of nephropathy (OR 1.69; 95% CI 1.36 to 2.11). In summary, the GREM1 variant rs1129456 associates with diabetic nephropathy, perhaps explaining some of the genetic susceptibility to this condition.


Asunto(s)
Nefropatías Diabéticas/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Polimorfismo de Nucleótido Simple , Adulto , Secuencia de Aminoácidos , Secuencia de Bases , Estudios de Casos y Controles , Frecuencia de los Genes , Genotipo , Humanos , Desequilibrio de Ligamiento , Persona de Mediana Edad , Datos de Secuencia Molecular , Alineación de Secuencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA