Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 436
Filtrar
1.
J Clin Immunol ; 44(8): 180, 2024 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-39153074

RESUMEN

Schimke immuno-osseous dysplasia is a rare multisystemic disorder caused by biallelic loss of function of the SMARCAL1 gene that plays a pivotal role in replication fork stabilization and thus DNA repair. Individuals affected from this disease suffer from disproportionate growth failure, steroid resistant nephrotic syndrome leading to renal failure and primary immunodeficiency mediated by T cell lymphopenia. With infectious complications being the leading cause of death in this disease, researching the nature of the immunodeficiency is crucial, particularly as the state is exacerbated by loss of antibodies due to nephrotic syndrome or immunosuppressive treatment. Building on previous findings that identified the loss of IL-7 receptor expression as a possible cause of the immunodeficiency and increased sensitivity to radiation-induced damage, we have employed spectral cytometry and multiplex RNA-sequencing to assess the phenotype and function of T cells ex-vivo and to study changes induced by in-vitro UV irradiation and reaction of cells to the presence of IL-7. Our findings highlight the mature phenotype of T cells with proinflammatory Th1 skew and signs of exhaustion and lack of response to IL-7. UV light irradiation caused a severe increase in the apoptosis of T cells, however the expression of the genes related to immune response and regulation remained surprisingly similar to healthy cells. Due to the disease's rarity, more studies will be necessary for complete understanding of this unique immunodeficiency.


Asunto(s)
Reparación del ADN , Osteocondrodisplasias , Enfermedades de Inmunodeficiencia Primaria , Humanos , Enfermedades de Inmunodeficiencia Primaria/genética , Enfermedades de Inmunodeficiencia Primaria/diagnóstico , Enfermedades de Inmunodeficiencia Primaria/inmunología , Osteocondrodisplasias/genética , Osteocondrodisplasias/inmunología , Reparación del ADN/genética , ADN Helicasas/genética , Síndrome Nefrótico/etiología , Síndrome Nefrótico/genética , Linfocitos T/inmunología , Arteriosclerosis/genética , Arteriosclerosis/etiología , Arteriosclerosis/inmunología , Masculino , Femenino , Embolia Pulmonar/genética , Embolia Pulmonar/etiología , Enfermedades Óseas Metabólicas/etiología , Enfermedades Óseas Metabólicas/genética , Trastornos del Crecimiento/genética , Trastornos del Crecimiento/etiología , Rayos Ultravioleta/efectos adversos , Niño , Apoptosis/genética , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/inmunología
2.
Nephrology (Carlton) ; 29(9): 612-616, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38838054

RESUMEN

Primary coenzyme Q10 deficiency-1, caused by COQ2 disease-causing variants, is an autosomal recessive disorder, and genetic testing is the gold standard for diagnosing this condition. A Chinese boy with steroid-resistant nephrotic syndrome, focal segmental glomerulosclerosis, and progressive kidney insufficiency was included in the study. Electron microscopy revealed the glomerular basement membrane with irregular thickness and lamellation with diffuse effacement of foot processes in the podocytes, and swollen mitochondria with abnormal cristae in the podocytes. Coenzyme Q10 supplementation started about 3 weeks after the onset of mild kidney dysfunction did not improve the proband's kidney outcome. Proband-only whole-exome sequencing and Sanger sequencing revealed two heteroallelic COQ2 variants: a maternally inherited novel variant c.1013G > A[p.(Gly338Glu)] in exon 6 and a variant of unknown origin c.1159C > T[p.(Arg387*)] in exon 7. Subsequent long-read sequencing demonstrated these two variants were located on different alleles. Our report extends the phenotypic and genotypic spectrum of COQ2 glomerulopathy.


Asunto(s)
Membrana Basal Glomerular , Glomeruloesclerosis Focal y Segmentaria , Síndrome Nefrótico , Ubiquinona , Humanos , Masculino , Síndrome Nefrótico/genética , Glomeruloesclerosis Focal y Segmentaria/genética , Glomeruloesclerosis Focal y Segmentaria/patología , Membrana Basal Glomerular/ultraestructura , Membrana Basal Glomerular/patología , Ubiquinona/análogos & derivados , Ubiquinona/deficiencia , Fenotipo , Predisposición Genética a la Enfermedad , Ataxia/genética , Secuenciación del Exoma , Debilidad Muscular/genética , Biopsia , Mutación , Enfermedades Mitocondriales/genética , Enfermedades Mitocondriales/patología , Transferasas Alquil y Aril
3.
Int J Mol Sci ; 25(11)2024 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-38891998

RESUMEN

Approximately 30% of steroid-resistant nephrotic syndromes are attributed to monogenic disorders that involve 27 genes. Mutations in KANK family members have also been linked to nephrotic syndrome; however, the precise mechanism remains elusive. To investigate this, podocyte-specific Kank1 knockout mice were generated to examine phenotypic changes. In the initial assessment under normal conditions, Kank1 knockout mice showed no significant differences in the urinary albumin-creatinine ratio, blood urea nitrogen, serum creatinine levels, or histological features compared to controls. However, following kidney injury with adriamycin, podocyte-specific Kank1 knockout mice exhibited a significantly higher albumin-creatinine ratio and a significantly greater sclerotic index than control mice. Electron microscopy revealed more extensive foot process effacement in the knockout mice than in control mice. In addition, KANK1-deficient human podocytes showed increased detachment and apoptosis following adriamycin exposure. These findings suggest that KANK1 may play a protective role in mitigating podocyte damage under pathological conditions.


Asunto(s)
Proteínas del Citoesqueleto , Doxorrubicina , Ratones Noqueados , Podocitos , Animales , Humanos , Masculino , Ratones , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Apoptosis , Proteínas del Citoesqueleto/metabolismo , Proteínas del Citoesqueleto/genética , Síndrome Nefrótico/metabolismo , Síndrome Nefrótico/genética , Síndrome Nefrótico/patología , Podocitos/metabolismo , Podocitos/patología , Proteínas Supresoras de Tumor/metabolismo , Proteínas Supresoras de Tumor/genética
4.
Immun Inflamm Dis ; 12(6): e1278, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38860750

RESUMEN

BACKGROUND: The current study aims to explore the relationship between tumor necrosis factor-α (TNF-α) polymorphism and the risk of primary nephrotic syndrome (PNS). METHODS: A total of 250 PNS patients were selected for this study, as well as 300 volunteers serving as the control group. TNF-α polymorphism were assessed using the polymerase chain reaction-restriction fragment length polymorphism method. In addition, a meta-analysis was conducted to analyze previously published literature on this topic. RESULTS: No significant differences were observed in the genotypes frequency or alleles frequency among the study populations. Meta-analysis results revealed a positive association between TNF-α rs1800629 polymorphism and allele contrast in African populations (p = 0), homozygote comparison (p = .007), heterozygote comparison (p = .026), recessive genetic model (p = .011), and dominant genetic model (p = .000). CONCLUSIONS: TNF-α rs1800629 polymorphism does not appear to confer any increased risk for PNS.


Asunto(s)
Predisposición Genética a la Enfermedad , Síndrome Nefrótico , Polimorfismo de Nucleótido Simple , Factor de Necrosis Tumoral alfa , Síndrome Nefrótico/genética , Humanos , Factor de Necrosis Tumoral alfa/genética , Estudios de Casos y Controles , Masculino , Femenino , Frecuencia de los Genes , Adulto , Genotipo , Alelos , Persona de Mediana Edad , Factores de Riesgo
5.
Pediatr Nephrol ; 39(11): 3205-3208, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-38780768

RESUMEN

BACKGROUND: Alport syndrome is a genetically heterogenous disorder resulting from variants in genes coding for alpha-3/4/5 chains of Collagen IV, which results in defective basement membranes in the kidney, cochlea and eye. The syndrome has different inheritance patterns and historically, was thought of as a disease affecting solely males. CASE: A 15-year-old female presented with pedal oedema, hypertension and proteinuria. She underwent a kidney biopsy which showed findings in keeping with focal segmental glomerulosclerosis. Her condition was refractory to steroids. Steroid-resistant nephrotic syndrome genetics were sent, revealing a rare pathogenic variant in the COL4A5 gene. CONCLUSION: Heterozygous females with X-linked Alport syndrome can develop chronic kidney disease and hearing loss. Clinicians should be mindful when reviewing kidney histology to include Alport syndrome as a differential for female patients. COL4A3-5 genes should be included in all steroid-resistant nephrotic syndrome genetic panels.


Asunto(s)
Colágeno Tipo IV , Nefritis Hereditaria , Síndrome Nefrótico , Humanos , Femenino , Adolescente , Síndrome Nefrótico/genética , Síndrome Nefrótico/diagnóstico , Síndrome Nefrótico/complicaciones , Síndrome Nefrótico/tratamiento farmacológico , Colágeno Tipo IV/genética , Nefritis Hereditaria/genética , Nefritis Hereditaria/complicaciones , Nefritis Hereditaria/diagnóstico , Nefritis Hereditaria/patología , Biopsia , Riñón/patología , Glomeruloesclerosis Focal y Segmentaria/genética , Glomeruloesclerosis Focal y Segmentaria/diagnóstico , Glomeruloesclerosis Focal y Segmentaria/patología , Glomeruloesclerosis Focal y Segmentaria/complicaciones , Glomeruloesclerosis Focal y Segmentaria/tratamiento farmacológico , Mutación
6.
Pediatr Nephrol ; 39(9): 2637-2640, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38587560

RESUMEN

Nuclear factor kappa B (NF-κB) family plays a central role in the human immune system. Heterozygous variants in NFKB2 typically cause immunodeficiency with various degrees of central adrenal insufficiency, autoimmunity, and ectodermal dysplasia. No reported case has presented kidney failure as an initial symptom. Moreover, documentation of kidney involvement of this disease is limited. CASE DIAGNOSIS: A 2-year-old female who presented with dyspnea and hypertensive emergency in the setting of new-onset nephrotic syndrome with acute-on chronic kidney injury with resultant chronic kidney disease (CKD) was found to have a novel heterozygous N-terminal variant in NFKB2 (c.880del: p. Tyr294Ilefs*4) with mild hypogammaglobulinemia, but no adrenal insufficiency or ectodermal dysplasia. She became dialysis-dependent during her initial hospitalization and developed CKD stage 5D, requiring continued peritoneal dialysis. She is currently awaiting kidney transplantation. CONCLUSIONS: Whether nephrotic syndrome or kidney injury or failure is the primary symptom of this variant or secondary to some event remains unknown. Further case accumulation is warranted.


Asunto(s)
Subunidad p52 de NF-kappa B , Síndrome Nefrótico , Insuficiencia Renal Crónica , Humanos , Femenino , Síndrome Nefrótico/genética , Síndrome Nefrótico/diagnóstico , Síndrome Nefrótico/complicaciones , Preescolar , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/diagnóstico , Insuficiencia Renal Crónica/complicaciones , Subunidad p52 de NF-kappa B/genética , Hipertensión/genética , Hipertensión/etiología , Hipertensión/diagnóstico , Diálisis Renal , Lesión Renal Aguda/genética , Lesión Renal Aguda/etiología , Lesión Renal Aguda/diagnóstico , Diálisis Peritoneal , Crisis Hipertensiva
7.
Am J Physiol Renal Physiol ; 326(5): F780-F791, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38482553

RESUMEN

Steroid-resistant nephrotic syndrome (SRNS) is the second most frequent cause of chronic kidney disease before the age of 25 yr. Nephrin, encoded by NPHS1, localizes to the slit diaphragm of glomerular podocytes and is the predominant structural component of the glomerular filtration barrier. Biallelic variants in NPHS1 can cause congenital nephrotic syndrome of the Finnish type, for which, to date, no causative therapy is available. Recently, adeno-associated virus (AAV) vectors targeting the glomerular podocyte have been assessed as a means for gene replacement therapy. Here, we established quantitative and reproducible phenotyping of a published, conditional Nphs1 knockout mouse model (Nphs1tm1.1Pgarg/J and Nphs2-Cre+) in preparation for a gene replacement study using AAV vectors. Nphs1 knockout mice (Nphs1fl/fl Nphs2-Cre+) exhibited 1) a median survival rate of 18 days (range: from 9 to 43 days; males: 16.5 days and females: 20 days); 2) an average foot process (FP) density of 1.0 FP/µm compared with 2.0 FP/µm in controls and a mean filtration slit density of 2.64 µm/µm2 compared with 4.36 µm/µm2 in controls; 3) a high number of proximal tubular microcysts; 4) the development of proteinuria within the first week of life as evidenced by urine albumin-to-creatinine ratios; and 5) significantly reduced levels of serum albumin and elevated blood urea nitrogen and creatinine levels. For none of these phenotypes, significant differences between sexes in Nphs1 knockout mice were observed. We quantitatively characterized five different phenotypic features of congenital nephrotic syndrome in Nphs1fl/fl Nphs2-Cre+ mice. Our results will facilitate future gene replacement therapy projects by allowing for sensitive detection of even subtle molecular effects.NEW & NOTEWORTHY To evaluate potential, even subtle molecular, therapeutic effects of gene replacement therapy (GRT) in a mouse model, prior rigorous quantifiable and reproducible disease phenotyping is necessary. Here, we, therefore, describe such a phenotyping effort in nephrin (Nphs1) knockout mice to establish the basis for GRT for congenital nephrotic syndrome. We believe that our findings set an important basis for upcoming/ongoing gene therapy approaches in the field of nephrology, especially for monogenic nephrotic syndrome.


Asunto(s)
Proteínas de la Membrana , Síndrome Nefrótico , Podocitos , Animales , Femenino , Masculino , Ratones , Modelos Animales de Enfermedad , Terapia Genética/métodos , Vectores Genéticos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones Noqueados , Síndrome Nefrótico/genética , Síndrome Nefrótico/terapia , Fenotipo , Podocitos/metabolismo
8.
Kidney Int ; 105(4): 663-665, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38519231

RESUMEN

In the current issue, Kuzmuk et al. offer a therapeutic option for patients with NPHS2 R138Q-associated nephrotic syndrome. For the first time in hereditary podocytopathies, this is offered by restoring the membrane targeting of a pathogenic protein. The idea that it is enough to liberate podocin from the trap of keratin 8, a key member of endoplasmic-reticulum-associated protein degradation complex, was brilliantly recognized based on former results obtained in cystic fibrosis.


Asunto(s)
Queratinas , Síndrome Nefrótico , Humanos , Queratinas/uso terapéutico , Síndrome Nefrótico/tratamiento farmacológico , Síndrome Nefrótico/genética , Mutación
9.
BMC Nephrol ; 25(1): 87, 2024 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-38448817

RESUMEN

BACKGROUND: This article reports an extremely rare case of lipoprotein glomerulopathy (LPG) with apolipoprotein E gene (APOE) Chicago mutation in a young Chinese male. Only five cases or families with APOE Chicago mutations have been reported in the literature. CASE PRESENTATION: The young male patient is manifested with nephrotic syndrome, accompanied by hyperlipidemia with a preferable increase in triglycerides and elevated ApoE level. Renal biopsy of the patient showed highly dilated glomerular capillaries filled with vacuolar lipids, segmentally fused podocyte foot processes, vacuolar degeneration of renal tubular epithelial cells and absence of electron-dense material, which indicates the diagnosis of LPG. Whole-exome gene sequencing identified the heterozygous mutation of NM_000041.4:c.494G > C (p.Arg165Pro), which is in the exon 4 of the APOE gene and also known as APOE Chicago mutation, a rare mutation of LPG. Further family pedigree gene analysis clarified that the mutation was inherited from the patient's mother, who does not have high ApoE levels or renal manifestations. This is also consistent with the incomplete penetrance of APOE gene mutations in LPG. Under lipid-lowering treatments, including a low-fat diet and fenofibrate, the patient's urinary protein was partially controlled, and the albumin level was recovered. CONCLUSION: Patients with nephrotic syndrome and elevated ApoE levels should be prompted into renal biopsy to avoid delay of appropriate treatment and unnecessary use of glucocorticoids. This case of LPG was diagnosed by renal biopsy and further verified with genetic sequencing. The timely diagnosis and treatment improved the patient's symptoms. This case is one of only six reported LPG cases or families with APOE Chicago mutation in the world.


Asunto(s)
Enfermedades Renales , Síndrome Nefrótico , Humanos , Masculino , Apolipoproteínas E/genética , Chicago , Síndrome Nefrótico/diagnóstico , Síndrome Nefrótico/genética
10.
Pediatr Nephrol ; 39(9): 2601-2609, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38326647

RESUMEN

Historically, specific mutations in WT1 gene have been associated with distinct syndromes based on phenotypic characteristics, including Denys-Drash syndrome (DDS), Frasier syndrome (FS), Meacham syndrome, and WAGR syndrome. DDS is classically defined by the triad of steroid-resistant nephrotic syndrome (SRNS) onset in the first year of life, disorders of sex development (DSD), and a predisposition to Wilms tumor (WT). Currently, a paradigm shift acknowledges a diverse spectrum of presentations beyond traditional syndromic definitions. Consequently, the concept of WT1-related disorders becomes more precise. A genotype-phenotype correlation has been established, emphasizing that the location and type of WT1 mutations significantly influence the clinical presentation, the condition severity, and the chronology of patient manifestations. Individuals presenting with persistent proteinuria, with or without nephrotic syndrome, and varying degrees of kidney dysfunction accompanied by genital malformations should prompt suspicion of WT1 mutations. Recent genetic advances enable a more accurate estimation of malignancy risk in these patients, facilitating a conservative nephron-sparing surgery (NSS) approach in select cases, with a focus on preserving residual kidney function and delaying nephrectomies. Other key management strategies include kidney transplantation and addressing DSD and gonadoblastoma. In summary, recent genetic insights underscore the imperative to implement individualized, integrated, and multidisciplinary management strategies for WT1-related disorders. This approach is pivotal in optimizing patient outcomes and addressing the complexities associated with these diverse clinical manifestations.


Asunto(s)
Síndrome de Denys-Drash , Mutación , Proteínas WT1 , Humanos , Síndrome de Denys-Drash/genética , Síndrome de Denys-Drash/diagnóstico , Síndrome de Denys-Drash/terapia , Proteínas WT1/genética , Fenotipo , Síndrome Nefrótico/genética , Síndrome Nefrótico/diagnóstico , Síndrome Nefrótico/terapia , Tumor de Wilms/genética , Tumor de Wilms/terapia , Tumor de Wilms/diagnóstico , Síndrome de Frasier/genética , Síndrome de Frasier/terapia , Síndrome de Frasier/diagnóstico
11.
Nat Commun ; 15(1): 1241, 2024 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-38336808

RESUMEN

Paraneoplastic syndromes occur in cancer patients and originate from dysfunction of organs at a distance from the tumor or its metastasis. A wide range of organs can be affected in paraneoplastic syndromes; however, the pathological mechanisms by which tumors influence host organs are poorly understood. Recent studies in the fly uncovered that tumor secreted factors target host organs, leading to pathological effects. In this study, using a Drosophila gut tumor model, we characterize a mechanism of tumor-induced kidney dysfunction. Specifically, we find that Pvf1, a PDGF/VEGF signaling ligand, secreted by gut tumors activates the PvR/JNK/Jra signaling pathway in the principal cells of the kidney, leading to mis-expression of renal genes and paraneoplastic renal syndrome-like phenotypes. Our study describes an important mechanism by which gut tumors perturb the function of the kidney, which might be of clinical relevance for the treatment of paraneoplastic syndromes.


Asunto(s)
Proteínas de Drosophila , Síndrome Nefrótico , Síndromes Paraneoplásicos , Animales , Humanos , Drosophila/metabolismo , Síndrome Nefrótico/genética , Síndromes Paraneoplásicos/terapia , Riñón/metabolismo , Transducción de Señal , Proteínas del Huevo/metabolismo , Proteínas de Drosophila/metabolismo
13.
Kidney Int ; 105(2): 218-230, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38245210

RESUMEN

Glomerular diseases are classified using a descriptive taxonomy that is not reflective of the heterogeneous underlying molecular drivers. This limits not only diagnostic and therapeutic patient management, but also impacts clinical trials evaluating targeted interventions. The Nephrotic Syndrome Study Network (NEPTUNE) is poised to address these challenges. The study has enrolled >850 pediatric and adult patients with proteinuric glomerular diseases who have contributed to deep clinical, histologic, genetic, and molecular profiles linked to long-term outcomes. The NEPTUNE Knowledge Network, comprising combined, multiscalar data sets, captures each participant's molecular disease processes at the time of kidney biopsy. In this editorial, we describe the design and implementation of NEPTUNE Match, which bridges a basic science discovery pipeline with targeted clinical trials. Noninvasive biomarkers have been developed for real-time pathway analyses. A Molecular Nephrology Board reviews the pathway maps together with clinical, laboratory, and histopathologic data assembled for each patient to compile a Match report that estimates the fit between the specific molecular disease pathway(s) identified in an individual patient and proposed clinical trials. The NEPTUNE Match report is communicated using established protocols to the patient and the attending nephrologist for use in their selection of available clinical trials. NEPTUNE Match represents the first application of precision medicine in nephrology with the aim of developing targeted therapies and providing the right medication for each patient with primary glomerular disease.


Asunto(s)
Enfermedades Renales , Síndrome Nefrótico , Adulto , Niño , Humanos , Biomarcadores , Ensayos Clínicos como Asunto , Glomérulos Renales/patología , Síndrome Nefrótico/diagnóstico , Síndrome Nefrótico/genética , Síndrome Nefrótico/terapia
14.
Pediatr Dev Pathol ; 27(4): 359-363, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38291869

RESUMEN

Steroid resistant nephrotic syndrome (SRNS) accounts for 30% of all cases of nephrotic syndrome (NS) in children and frequently leads to end stage kidney disease (ESKD). About 30% of children with SRNS demonstrate causative mutations in podocyte- associated genes. Early identification of genetic forms of SRNS is critical to avoid potentially harmful immunosuppressive therapy. A 2-year-old male patient with NS and no family history of renal disease did not respond to 4-week steroid treatment. Kidney biopsy demonstrated mesangial proliferative glomerulopathy with basement membrane dysmorphism. Tacrolimus and Lisinopril were added to therapy pending results of genetic testing. Kidney Gene panel showed a NPHS2 c.413G>A (p.Arg138Gln) homozygous pathogenic variant. This missense variant is considered a common pathogenic founder mutation in European populations. A diagnosis of autosomal-recessive form of nonsyndromic SRNS due to NPHS2 causative variant was made. Immunosuppresive therapy was stopped, Lizinopril dose was increased and weekly infusions of Albumin/furosemide were initiated to manage edema. This case demonstrates that early genetic testing in children with SRNS avoids prolonged potentially harmful immunosuppressive therapy, allows for timely genetic family counseling, and allows earlier consideration for future living related donor kidney transplantation.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular , Proteínas de la Membrana , Síndrome Nefrótico , Humanos , Masculino , Síndrome Nefrótico/genética , Síndrome Nefrótico/patología , Síndrome Nefrótico/diagnóstico , Preescolar , Proteínas de la Membrana/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Mutación Missense
15.
Pathol Res Pract ; 254: 155092, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38218042

RESUMEN

Schimke immuno-osseous dysplasia (SIOD) is a rare multi-system condition caused by biallelic loss-of-function mutations in the SMARCAL1 gene. This disorder is characterized by disproportionate growth failure, T-cell deficiency, and renal dysfunction. Pathogenic variants in the SMARCAL1 gene have been reported in only approximately half of SIOD-affected individuals. Among these alterations, nonsense and frameshift mutations generally lead to a severe phenotype with early onset. In this study, we identified novel mutations in an Iranian patient with SIOD. A 4-year-old girl with developmental delay and facial dysmorphism was referred to our center for molecular diagnosis. We applied whole-exome and Sanger sequencing for co-segregation analysis. Subsequently, bioinformatic analysis was performed to assess the pathogenic effects of the variants and their post-transcriptional effects. We discovered two novel mutations (c.2281delT and c.2283delA) in exon 15 of the SMARCAL1 gene, resulting in a truncated protein with a loss of 193 amino acids (p.S761Rfs*1). Variant effect predictors indicated that these variants are pathogenic, and multi-sequence alignments revealed high conservation of this region among different species. Given that our patient exhibited severe a phenotype and passed away soon after receiving a definitive molecular diagnosis, we propose that the loss of the helicase C-terminal domain in the deleted part of SMARCAL1 may lead to the severe form of SIOD. Besides, the combination of growth retardation and bone abnormalities also plays a crucial role in the early diagnosis of the disease.


Asunto(s)
Arteriosclerosis , Síndromes de Inmunodeficiencia , Síndrome Nefrótico , Osteocondrodisplasias , Enfermedades de Inmunodeficiencia Primaria , Embolia Pulmonar , Femenino , Humanos , Preescolar , Irán , Enfermedades de Inmunodeficiencia Primaria/genética , Enfermedades de Inmunodeficiencia Primaria/complicaciones , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/complicaciones , Síndromes de Inmunodeficiencia/metabolismo , Síndrome Nefrótico/diagnóstico , Síndrome Nefrótico/genética , Síndrome Nefrótico/complicaciones , ADN Helicasas/genética
16.
Exp Cell Res ; 435(2): 113931, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38253280

RESUMEN

The mortality rate linked with nephrotic syndrome (NS) is quite high. The renal tubular injury influences the response of NS patients to steroid treatment. KN motif and ankyrin repeat domains 2 (KANK2) regulates actin polymerization, which is required for renal tubular cells to maintain their function. In this study, we found that the levels of KANK2 in patients with NS were considerably lower than those in healthy controls, especially in NS patients with acute kidney injury (AKI). To get a deeper understanding of the KANK2 transcriptional control mechanism, the core promoter region of the KANK2 gene was identified. KANK2 was further found to be positively regulated by E2F Transcription Factor 1 (E2F1), Transcription Factor AP-2 Gamma (TFAP2C), and Nuclear Respiratory Factor 1 (NRF1), both at mRNA and protein levels. Knocking down E2F1, TFAP2C, or NRF1 deformed the cytoskeleton of renal tubular cells and reduced F-actin content. EMSA and ChIP assays confirmed that all three transcription factors could bind to the upstream promoter transcription site of KANK2 to transactivate KANK2 in renal tubular epithelial cells. Our study suggests that E2F1, TFAP2C, and NRF1 play essential roles in regulating the KANK2 transcription, therefore shedding fresh light on the development of putative therapeutic options for the treatment of NS patients.


Asunto(s)
Síndrome Nefrótico , Factor Nuclear 1 de Respiración , Humanos , Factor Nuclear 1 de Respiración/metabolismo , Síndrome Nefrótico/genética , Factores de Transcripción/metabolismo , Regulación de la Expresión Génica , Regiones Promotoras Genéticas/genética , Factor de Transcripción E2F1/genética , Factor de Transcripción AP-2/genética
17.
Pediatr Nephrol ; 39(5): 1421-1425, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-37985485

RESUMEN

A two-and-a-half-month-old female infant presented with generalized edema for 10 days. At presentation, she had periorbital puffiness, moderate ascites, and pedal edema. Laboratory investigations revealed serum albumin 1.3 g/dL, spot urine protein to creatinine ratio (Up:Uc) 20.87 mg/mg, total cholesterol 380 mg/dL, and serum creatinine 0.31 mg/dL. Exome sequencing revealed compound heterozygous variants in LAMA5 gene (NM_005560.6). There was a heterozygous likely pathogenic missense variant in exon 2: LAMA5: c.385C > A (depth 195 ×) and another heterozygous pathogenic variant in exon 31: LAMA5: c.3932_3936dup; parental segregation by Sanger sequencing proved that the variants were in trans. Kidney biopsy showed diffuse mesangial sclerosis (DMS). Our case adds LAMA5 gene to the constellation of genes causing DMS, in addition to the classically described WT1, LAMB2, and PLCE1 genes and to the list of genes causing congenital nephrotic syndrome (CNS).


Asunto(s)
Síndrome Nefrótico , Esclerosis , Femenino , Humanos , Lactante , Edema , Mutación , Síndrome Nefrótico/diagnóstico , Síndrome Nefrótico/genética , Síndrome Nefrótico/congénito
18.
Kidney Int ; 105(4): 744-758, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37995908

RESUMEN

Podocin is a key membrane scaffolding protein of the kidney podocyte essential for intact glomerular filtration. Mutations in NPHS2, the podocin-encoding gene, represent the commonest form of inherited nephrotic syndrome (NS), with early, intractable kidney failure. The most frequent podocin gene mutation in European children is R138Q, causing retention of the misfolded protein in the endoplasmic reticulum. Here, we provide evidence that podocin R138Q (but not wild-type podocin) complexes with the intermediate filament protein keratin 8 (K8) thereby preventing its correct trafficking to the plasma membrane. We have also identified a small molecule (c407), a compound that corrects the Cystic Fibrosis Transmembrane Conductance Regulator protein defect, that interrupts this complex and rescues mutant protein mistrafficking. This results in both the correct localization of podocin at the plasma membrane and functional rescue in both human patient R138Q mutant podocyte cell lines, and in a mouse inducible knock-in model of the R138Q mutation. Importantly, complete rescue of proteinuria and histological changes was seen when c407 was administered both via osmotic minipumps or delivered orally prior to induction of disease or crucially via osmotic minipump two weeks after disease induction. Thus, our data constitute a therapeutic option for patients with NS bearing a podocin mutation, with implications for other misfolding protein disorders. Further studies are necessary to confirm our findings.


Asunto(s)
Síndrome Nefrótico , Animales , Niño , Humanos , Ratones , Péptidos y Proteínas de Señalización Intracelular/genética , Queratina-8/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Chaperonas Moleculares/genética , Mutación , Síndrome Nefrótico/tratamiento farmacológico , Síndrome Nefrótico/genética , Síndrome Nefrótico/patología
19.
Pediatr Nephrol ; 39(6): 1685-1707, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37728640

RESUMEN

Focal segmental glomerulosclerosis (FSGS) is a complex disease which describes different kinds of kidney defects, not exclusively linked with podocyte defects. Since nephrin mutation was first described in association with early-onset nephrotic syndrome (NS), many advancements have been made in understanding genetic patterns associated with FSGS. New genetic causes of FSGS have been discovered, displaying unexpected genotypes, and recognizing possible site of damage. Many recent large-scale sequencing analyses on patients affected by idiopathic chronic kidney disease (CKD), kidney failure (KF) of unknown origin, or classified as FSGS, have revealed collagen alpha IV genes, as one of the most frequent sites of pathogenic mutations. Also, recent interest in complex and systemic lysosomal storage diseases, such as Fabry disease, has highlighted GLA mutations as possible causes of FSGS. Tubulointerstitial disease, recently classified by KDIGO based on genetic subtypes, when associated with UMOD variants, may phenotypically gain FSGS features, as well as ciliopathy genes or others, otherwise leading to completely different phenotypes, but found carrying pathogenic variants with associated FSGS phenotype. Thus, glomerulosclerosis may conceal different heterogeneous conditions. When a kidney biopsy is performed, the principal objective is to provide an accurate diagnosis. The broad spectrum of phenotypic expression and genetic complexity is demonstrating that a combined path of management needs to be applied. Genetic investigation should not be reserved only to selected cases, but rather part of medical management, integrating with clinical and renal pathology records. FSGS heterogeneity should be interpreted as an interesting opportunity to discover new pathways of CKD, requiring prompt genotype-phenotype correlation. In this review, we aim to highlight how FSGS represents a peculiar kidney condition, demanding multidisciplinary management, and in which genetic analysis may solve some otherwise unrevealed idiopathic cases. Unfortunately there is not a uniform correlation between specific mutations and FSGS morphological classes, as the same variants may be identified in familial cases or sporadic FSGS/NS or manifest a variable spectrum of the same disease. These non-specific features make diagnosis challenging. The complexity of FSGS genotypes requires new directions. Old morphological classification does not provide much information about the responsible cause of disease and misdiagnoses may expose patients to immunosuppressive therapy side effects, mistaken genetic counseling, and misguided kidney transplant programs.


Asunto(s)
Glomeruloesclerosis Focal y Segmentaria , Síndrome Nefrótico , Insuficiencia Renal Crónica , Humanos , Glomeruloesclerosis Focal y Segmentaria/diagnóstico , Cicatriz/patología , Glomérulos Renales/patología , Riñón/patología , Síndrome Nefrótico/genética , Colágeno Tipo IV/genética , Insuficiencia Renal Crónica/patología
20.
Genes (Basel) ; 14(12)2023 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-38136965

RESUMEN

Pathogenic gene variants encoding nuclear pore complex (NPC) proteins were previously implicated in the pathogenesis of steroid-resistant nephrotic syndrome (SRNS). The NUP85 gene, encoding nucleoporin, is related to a very rare form of SRNS with limited genotype-phenotype information. We identified an Italian boy affected with an SRNS associated with severe neurodevelopmental impairment characterized by microcephaly, axial hypotonia, lack of achievement of motor milestones, and refractory seizures with an associated hypsarrhythmic pattern on electroencephalography. Brain magnetic resonance imaging (MRI) showed hypoplasia of the corpus callosum and a simplified gyration of the cerebral cortex. Since the age of 3 years, the boy was followed up at our Pediatric Nephrology Department for an SRNS, with a focal segmental glomerulosclerosis at renal biopsy. The boy died 32 months after SRNS onset, and a Whole-Exome Sequencing analysis revealed a novel compound heterozygous variant in NUP85 (NM_024844.5): 611T>A (p.Val204Glu), c.1904T>G (p.Leu635Arg), inherited from the father and mother, respectively. We delineated the clinical phenotypes of NUP85-related disorders, reviewed the affected individuals so far reported in the literature, and overall expanded both the phenotypic and the molecular spectrum associated with this ultra-rare genetic condition. Our study suggests a potential occurrence of severe neurological phenotypes as part of the NUP85-related clinical spectrum and highlights an important involvement of nucleoporin in brain developmental processes and neurological function.


Asunto(s)
Neuronas , Podocitos , Niño , Preescolar , Humanos , Masculino , Mutación , Síndrome Nefrótico/genética , Síndrome Nefrótico/patología , Neuronas/metabolismo , Neuronas/patología , Proteínas de Complejo Poro Nuclear/genética , Podocitos/metabolismo , Podocitos/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA