Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
2.
Can J Kidney Health Dis ; 10: 20543581231154185, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36798634

RESUMO

Purpose of Review: To understand the impact of kidney disease in Canada and the priority areas of kidney research that can benefit from patient-oriented, precision medicine research using novel technologies. Sources of Information: Information was collected through discussions between health care professionals, researchers, and patient partners. Literature was compiled using search engines (PubMed, PubMed central, Medline, and Google) and data from the Canadian Organ Replacement Register. Methods: We reviewed the impact, prevalence, economic burden, causes of kidney disease, and priority research areas in Canada. After reviewing the priority areas for kidney research, potential avenues for future research that can integrate precision medicine initiatives for patient-oriented research were outlined. Key Findings: Chronic kidney disease (CKD) remains among the top causes of morbidity and mortality in the world and exerts a large financial strain on the health care system. Despite the increasing number of people with CKD, funding for basic kidney research continues to trail behind other diseases. Current funding strategies favor existing clinical treatment and patient educational strategies. The identification of genetic factors for various forms of kidney disease in the adult and pediatric populations provides mechanistic insight into disease pathogenesis. Allocation of resources and funding toward existing high-yield personalized research initiatives have the potential to significantly affect patient-oriented research outcomes but will be difficult due to a constant decline of funding for kidney research. Limitations: This is an overview primarily focused on Canadian-specific literature rather than a comprehensive systematic review of the literature. The scope of our findings and conclusions may not be applicable to health care systems in other countries.


Justification: Cette étude visait à mieux comprendre l'impact de l'insuffisance rénale au Canada et à déterminer les domaines de recherche prioritaires en santé rénale qui pourraient tirer profit des recherches axées sur le patient qui sont menées en médecine de précision avec de nouvelles technologies. Sources: L'information provient de discussions entre les professionnels de la santé, les chercheurs et les patients partenaires. La littérature a été compilée à l'aide de moteurs de recherche (PubMed, PubMed central, Medline et Google) et de données provenant du Registre canadien des insuffisances et des transplantations d'organes. Méthodologie: Nous avons examiné les répercussions, la prévalence, le fardeau économique et les causes des maladies rénales, ainsi que les domaines prioritaires de la recherche au Canada. Après avoir examiné les domaines de recherche prioritaires en santé rénale, nous avons décrit les possibles orientations de recherche pouvant intégrer des initiatives de la recherche axée sur le patient menée en médecine de précision. Principaux résultats: L'insuffisance rénale chronique (IRC) demeure l'une des principales causes de morbidité et de mortalité dans le monde et elle exerce une forte pression financière sur le système de santé. Malgré le nombre croissant de personnes atteintes d'IRC, le financement de la recherche fondamentale en santé rénale reste à la traîne des autres maladies; et le financement actuel favorise les stratégies existantes pour le traitement clinique et l'éducation des patients. L'identification de facteurs génétiques liés aux diverses formes de néphropathies dans les populations adultes et pédiatriques fournit une compréhension mécanistique de la pathogenèse de la maladie. L'attribution des ressources et du financement à des initiatives de recherche personnalisées existantes et connues pour leur rendement élevé pourrait avoir une incidence considérable sur les résultats de la recherche axée sur le patient, mais elle sera difficile compte tenu de la diminution constante du financement de la recherche en santé rénale. Limites: Il s'agit d'un portrait axé principalement sur la littérature canadienne et non d'un examen systématique complet de la littérature. La portée des résultats et conclusions pourrait ne pas s'appliquer aux systèmes de santé d'autres pays.

3.
Front Nephrol ; 3: 1331847, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38292052

RESUMO

Ciliopathies are a group of rare genetic disorders caused by defects to the structure or function of the primary cilium. They often affect multiple organs, leading to brain malformations, congenital heart defects, and anomalies of the retina or skeletal system. Kidney abnormalities are among the most frequent ciliopathic phenotypes manifesting as smaller, dysplastic, and cystic kidneys that are often accompanied by renal fibrosis. Many renal ciliopathies cause chronic kidney disease and often progress to end-stage renal disease, necessitating replacing therapies. There are more than 35 known ciliopathies; each is a rare hereditary condition, yet collectively they account for a significant proportion of chronic kidney disease worldwide. The primary cilium is a tiny microtubule-based organelle at the apex of almost all vertebrate cells. It serves as a "cellular antenna" surveying environment outside the cell and transducing this information inside the cell to trigger multiple signaling responses crucial for tissue morphogenesis and homeostasis. Hundreds of proteins and unique cellular mechanisms are involved in cilia formation. Recent evidence suggests that actin remodeling and regulation at the base of the primary cilium strongly impacts ciliogenesis. In this review, we provide an overview of the structure and function of the primary cilium, focusing on the role of actin cytoskeleton and its regulators in ciliogenesis. We then describe the key clinical, genetic, and molecular aspects of renal ciliopathies. We highlight what is known about actin regulation in the pathogenesis of these diseases with the aim to consider these recent molecular findings as potential therapeutic targets for renal ciliopathies.

4.
Neuromuscul Disord ; 32(6): 512-515, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35584999

RESUMO

Chylothorax has been reported in rare cases of X-linked myotubular myopathy, but the pathophysiology of this association is not fully understood. We report a case of a neonate presenting prenatally with hydrops and chylothorax. The patient died at 17 days of life due to respiratory failure secondary to severe pulmonary hypertension. Comprehensive genetic testing identified a de novo hemizygous frameshift mutation in the MTM1 gene (c.142-143del, p.Glu48Serfs*12) with subsequent autopsy confirming the diagnosis of X-linked myotubular myopathy. Lung microscopy demonstrated primary pulmonary lymphangiectasia as the cause for the massive chylothorax. To the best of our knowledge, this is the first reported case of molecularly confirmed X-linked myotubular myopathy with pulmonary lymphangiectasia with prenatal findings of hydrops, chylothorax and postnatal severe pulmonary hypertension.


Assuntos
Quilotórax , Hipertensão Pulmonar , Miopatias Congênitas Estruturais , Quilotórax/complicações , Quilotórax/genética , Edema/complicações , Edema/genética , Feminino , Testes Genéticos , Humanos , Hipertensão Pulmonar/etiologia , Hipertensão Pulmonar/genética , Recém-Nascido , Miopatias Congênitas Estruturais/complicações , Miopatias Congênitas Estruturais/diagnóstico , Miopatias Congênitas Estruturais/genética , Gravidez , Proteínas Tirosina Fosfatases não Receptoras/genética
5.
Am J Med Genet A ; 188(5): 1355-1367, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35040250

RESUMO

Spina bifida (SB) is the second most common nonlethal congenital malformation. The existence of monogenic SB mouse models and human monogenic syndromes with SB features indicate that human SB may be caused by monogenic genes. We hypothesized that whole exome sequencing (WES) allows identification of potential candidate genes by (i) generating a list of 136 candidate genes for SB, and (ii) by unbiased exome-wide analysis. We generated a list of 136 potential candidate genes from three categories and evaluated WES data of 50 unrelated SB cases for likely deleterious variants in 136 potential candidate genes, and for potential SB candidate genes exome-wide. We identified 6 likely deleterious variants in 6 of the 136 potential SB candidate genes in 6 of the 50 SB cases, whereof 4 genes were derived from mouse models, 1 gene was derived from human nonsyndromic SB, and 1 gene was derived from candidate genes known to cause human syndromic SB. In addition, by unbiased exome-wide analysis, we identified 12 genes as potential candidates for SB. Identification of these 18 potential candidate genes in larger SB cohorts will help decide which ones can be considered as novel monogenic causes of human SB.


Assuntos
Exoma , Disrafismo Espinal , Animais , Modelos Animais de Doenças , Exoma/genética , Humanos , Camundongos , Disrafismo Espinal/genética , Sequenciamento do Exoma
6.
Eur J Hum Genet ; 30(1): 111-116, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34707297

RESUMO

ITSN1 plays an important role in brain development. Recent studies in large cohorts of subjects with neurodevelopmental disorders have identified de novo variants in ITSN1 gene thereby suggesting that this gene is involved in the development of such disorders. The aim of this study is to provide further proof of such a link. We performed trio exome sequencing in a patient presenting autism, intellectual disability, and severe behavioral difficulties. Additional affected patients with a neurodevelopmental disorder harboring a heterozygous variant in ITSN1 (NM_003024.2) were collected through a worldwide collaboration. All patients underwent detailed phenotypic and genetic assessment and data was collected and shared by healthcare givers. We identified ten novel patients from eight families with heterozygous truncating or missense variants in ITSN1 gene. In addition, four previously published patients from large meta-analysis studies were included. In total, 7/14 patients presented a de novo variant in ITSN1. All patients showed neurodevelopmental disorders from autism spectrum disorders (90%), intellectual disability (86%), and epilepsy (30%). We demonstrated that truncating variants are in the first half of ITSN1 whereas missense variants are clustered in C-terminal region. We suggest ITSN1 gene is involved in development of an autism spectrum disorder with variable additional neurodevelopmental deficiency, thus confirming the hypothesis that ITSN1 is important for brain development.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/genética , Deficiências do Desenvolvimento/genética , Epilepsia/genética , Deficiência Intelectual/genética , Proteínas Adaptadoras de Transporte Vesicular/química , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Adolescente , Adulto , Criança , Pré-Escolar , Deficiências do Desenvolvimento/patologia , Epilepsia/diagnóstico , Genes Dominantes , Humanos , Deficiência Intelectual/patologia , Mutação com Perda de Função , Masculino , Mutação de Sentido Incorreto , Fenótipo
7.
Am J Med Genet A ; 185(12): 3784-3792, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34338422

RESUMO

The acronym VATER/VACTERL refers to the rare nonrandom association of the following component features (CFs): vertebral defects (V), anorectal malformations (ARM) (A), cardiac anomalies (C), tracheoesophageal fistula with or without esophageal atresia (TE), renal malformations (R), and limb anomalies (L). For the clinical diagnosis, the presence of at least three CFs is required, individuals presenting with only two CFs have been categorized as VATER/VACTERL-like. The majority of VATER/VACTERL individuals displays a renal phenotype. Hitherto, variants in FGF8, FOXF1, HOXD13, LPP, TRAP1, PTEN, and ZIC3 have been associated with the VATER/VACTERL association; however, large-scale re-sequencing could only confirm TRAP1 and ZIC3 as VATER/VACTERL disease genes, both associated with a renal phenotype. In this study, we performed exome sequencing in 21 individuals and their families with a renal VATER/VACTERL or VATER/VACTERL-like phenotype to identify potentially novel genetic causes. Exome analysis identified biallelic and X-chromosomal hemizygous potentially pathogenic variants in six individuals (29%) in B9D1, FREM1, ZNF157, SP8, ACOT9, and TTLL11, respectively. The online tool GeneMatcher revealed another individual with a variant in ZNF157. Our study suggests six biallelic and X-chromosomal hemizygous VATER/VACTERL disease genes implicating all six genes in the expression of human renal malformations.


Assuntos
Malformações Anorretais/genética , Atresia Esofágica/genética , Predisposição Genética para Doença , Cardiopatias/genética , Fístula Traqueoesofágica/genética , Malformações Anorretais/complicações , Malformações Anorretais/patologia , Proteínas do Citoesqueleto/genética , Proteínas de Ligação a DNA/genética , Atresia Esofágica/complicações , Atresia Esofágica/patologia , Feminino , Genes Ligados ao Cromossomo X/genética , Estudos de Associação Genética , Proteínas de Choque Térmico HSP90/genética , Cardiopatias/complicações , Cardiopatias/patologia , Hemizigoto , Proteínas de Homeodomínio/genética , Humanos , Rim/anormalidades , Masculino , Receptores de Interleucina/genética , Fístula Traqueoesofágica/complicações , Fístula Traqueoesofágica/patologia , Fatores de Transcrição/genética , Sequenciamento do Exoma
8.
BMJ Case Rep ; 14(4)2021 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-33849881

RESUMO

A 39-year-old woman was referred to the cancer genetics outpatient clinic for a clinical diagnosis of Carney complex (CNC) in her deceased brother. The patient had some characteristic clinical features such as periorbital lentigines and coarse facial features, suggestive of CNC; however, she did not meet major diagnostic criteria for CNC. Previous extensive investigations revealed a mild insulin-like growth factor 1 elevation, a stable left adrenal gland adenoma and a slightly enlarged pituitary gland. Single gene sequencing confirmed a novel pathogenic mutation in the PRKAR1A gene. This case, to our knowledge, is the first report of this mutation identified in a family of French-Canadian origin. This report broadens our understanding of the genotypic and phenotypic spectrum of this rare disease, while it highlights the value of a multidisciplinary approach in rare diseases, for genetic testing facilitated a timely diagnosis and enabled the initiation of early surveillance of CNC-related manifestations in our patient.


Assuntos
Complexo de Carney , Adulto , Canadá , Complexo de Carney/diagnóstico , Complexo de Carney/genética , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/genética , Feminino , Testes Genéticos , Humanos , Masculino , Mutação
9.
Sci Adv ; 7(1)2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33523862

RESUMO

Nephrotic syndrome (NS) is a leading cause of chronic kidney disease. We found recessive NOS1AP variants in two families with early-onset NS by exome sequencing. Overexpression of wild-type (WT) NOS1AP, but not cDNA constructs bearing patient variants, increased active CDC42 and promoted filopodia and podosome formation. Pharmacologic inhibition of CDC42 or its effectors, formin proteins, reduced NOS1AP-induced filopodia formation. NOS1AP knockdown reduced podocyte migration rate (PMR), which was rescued by overexpression of WT Nos1ap but not by constructs bearing patient variants. PMR in NOS1AP knockdown podocytes was also rescued by constitutively active CDC42Q61L or the formin DIAPH3 Modeling a NOS1AP patient variant in knock-in human kidney organoids revealed malformed glomeruli with increased apoptosis. Nos1apEx3-/Ex3- mice recapitulated the human phenotype, exhibiting proteinuria, foot process effacement, and glomerulosclerosis. These findings demonstrate that recessive NOS1AP variants impair CDC42/DIAPH-dependent actin remodeling, cause aberrant organoid glomerulogenesis, and lead to a glomerulopathy in humans and mice.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Nefropatias , Síndrome Nefrótica , Podócitos , Actinas/genética , Actinas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Forminas/genética , Humanos , Nefropatias/metabolismo , Camundongos , Síndrome Nefrótica/genética , Síndrome Nefrótica/metabolismo , Podócitos/metabolismo
10.
Kidney Int Rep ; 6(2): 460-471, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33615071

RESUMO

INTRODUCTION: Steroid-resistant nephrotic syndrome (SRNS) is the second most common cause of chronic kidney disease during childhood. Identification of 63 monogenic human genes has delineated 12 distinct pathogenic pathways. METHODS: Here, we generated 2 independent sets of nephrotic syndrome (NS) candidate genes to augment the discovery of additional monogenic causes based on whole-exome sequencing (WES) data from 1382 families with NS. RESULTS: We first identified 63 known monogenic causes of NS in mice from public databases and scientific publications, and 12 of these genes overlapped with the 63 known human monogenic SRNS genes. Second, we used a set of 64 genes that are regulated by the transcription factor Wilms tumor 1 (WT1), which causes SRNS if mutated. Thirteen of these WT1-regulated genes overlapped with human or murine NS genes. Finally, we overlapped these lists of murine and WT1 candidate genes with our list of 120 candidate genes generated from WES in 1382 NS families, to identify novel candidate genes for monogenic human SRNS. Using this approach, we identified 7 overlapping genes, of which 3 genes were shared by all datasets, including SYNPO. We show that loss-of-function of SYNPO leads to decreased CDC42 activity and reduced podocyte migration rate, both of which are rescued by overexpression of wild-type complementary DNA (cDNA), but not by cDNA representing the patient mutation. CONCLUSION: Thus, we identified 3 novel candidate genes for human SRNS using 3 independent, nonoverlapping hypotheses, and generated functional evidence for SYNPO as a novel potential monogenic cause of NS.

11.
Kidney Int Rep ; 6(2): 472-483, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33615072

RESUMO

INTRODUCTION: Most of the approximately 60 genes that if mutated cause steroid-resistant nephrotic syndrome (SRNS) are highly expressed in the glomerular podocyte, rendering SRNS a "podocytopathy." METHODS: We performed whole-exome sequencing (WES) in 1200 nephrotic syndrome (NS) patients. RESULTS: We discovered homozygous truncating and homozygous missense mutation in SYNPO2 (synaptopodin-2) (p.Lys1124∗ and p.Ala1134Thr) in 2 patients with childhood-onset NS. We found SYNPO2 expression in both podocytes and mesangial cells; however, notably, immunofluorescence staining of adult human and rat kidney cryosections indicated that SYNPO2 is localized mainly in mesangial cells. Subcellular localization studies reveal that in these cells SYNPO2 partially co-localizes with α-actinin and filamin A-containing F-actin filaments. Upon transfection in mesangial cells or podocytes, EGFP-SYNPO2 co-localized with α-actinin-4, which gene is mutated in autosomal dominant SRNS in humans. SYNPO2 overexpression increases mesangial cell migration rate (MMR), whereas shRNA knockdown reduces MMR. Decreased MMR was rescued by transfection of wild-type mouse Synpo2 cDNA but only partially by cDNA representing mutations from the NS patients. The increased mesangial cell migration rate (MMR) by SYNPO2 overexpression was inhibited by ARP complex inhibitor CK666. SYNPO2 shRNA knockdown in podocytes decreased active Rac1, which was rescued by transfection of wild-type SYNPO2 cDNA but not by cDNA representing any of the 2 mutant variants. CONCLUSION: We show that SYNPO2 variants may lead to Rac1-ARP3 dysregulation, and may play a role in the pathogenesis of nephrotic syndrome.

12.
J Am Soc Nephrol ; 32(3): 580-596, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33593823

RESUMO

BACKGROUND: Galloway-Mowat syndrome (GAMOS) is characterized by neurodevelopmental defects and a progressive nephropathy, which typically manifests as steroid-resistant nephrotic syndrome. The prognosis of GAMOS is poor, and the majority of children progress to renal failure. The discovery of monogenic causes of GAMOS has uncovered molecular pathways involved in the pathogenesis of disease. METHODS: Homozygosity mapping, whole-exome sequencing, and linkage analysis were used to identify mutations in four families with a GAMOS-like phenotype, and high-throughput PCR technology was applied to 91 individuals with GAMOS and 816 individuals with isolated nephrotic syndrome. In vitro and in vivo studies determined the functional significance of the mutations identified. RESULTS: Three biallelic variants of the transcriptional regulator PRDM15 were detected in six families with proteinuric kidney disease. Four families with a variant in the protein's zinc-finger (ZNF) domain have additional GAMOS-like features, including brain anomalies, cardiac defects, and skeletal defects. All variants destabilize the PRDM15 protein, and the ZNF variant additionally interferes with transcriptional activation. Morpholino oligonucleotide-mediated knockdown of Prdm15 in Xenopus embryos disrupted pronephric development. Human wild-type PRDM15 RNA rescued the disruption, but the three PRDM15 variants did not. Finally, CRISPR-mediated knockout of PRDM15 in human podocytes led to dysregulation of several renal developmental genes. CONCLUSIONS: Variants in PRDM15 can cause either isolated nephrotic syndrome or a GAMOS-type syndrome on an allelic basis. PRDM15 regulates multiple developmental kidney genes, and is likely to play an essential role in renal development in humans.


Assuntos
Proteínas de Ligação a DNA/genética , Hérnia Hiatal/genética , Microcefalia/genética , Mutação de Sentido Incorreto , Nefrose/genética , Fatores de Transcrição/genética , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Linhagem Celular , Pré-Escolar , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/deficiência , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Técnicas de Inativação de Genes , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Lactente , Recém-Nascido , Masculino , Modelos Moleculares , Síndrome Nefrótica/genética , Podócitos/metabolismo , Polimorfismo de Nucleotídeo Único , Pronefro/embriologia , Pronefro/metabolismo , Estabilidade Proteica , Fatores de Transcrição/química , Fatores de Transcrição/deficiência , Xenopus laevis/embriologia , Xenopus laevis/genética , Dedos de Zinco/genética
13.
Am J Hum Genet ; 108(2): 357-367, 2021 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-33508234

RESUMO

Focal segmental glomerulosclerosis (FSGS) is the main pathology underlying steroid-resistant nephrotic syndrome (SRNS) and a leading cause of chronic kidney disease. Monogenic forms of pediatric SRNS are predominantly caused by recessive mutations, while the contribution of de novo variants (DNVs) to this trait is poorly understood. Using exome sequencing (ES) in a proband with FSGS/SRNS, developmental delay, and epilepsy, we discovered a nonsense DNV in TRIM8, which encodes the E3 ubiquitin ligase tripartite motif containing 8. To establish whether TRIM8 variants represent a cause of FSGS, we aggregated exome/genome-sequencing data for 2,501 pediatric FSGS/SRNS-affected individuals and 48,556 control subjects, detecting eight heterozygous TRIM8 truncating variants in affected subjects but none in control subjects (p = 3.28 × 10-11). In all six cases with available parental DNA, we demonstrated de novo inheritance (p = 2.21 × 10-15). Reverse phenotyping revealed neurodevelopmental disease in all eight families. We next analyzed ES from 9,067 individuals with epilepsy, yielding three additional families with truncating TRIM8 variants. Clinical review revealed FSGS in all. All TRIM8 variants cause protein truncation clustering within the last exon between residues 390 and 487 of the 551 amino acid protein, indicating a correlation between this syndrome and loss of the TRIM8 C-terminal region. Wild-type TRIM8 overexpressed in immortalized human podocytes and neuronal cells localized to nuclear bodies, while constructs harboring patient-specific variants mislocalized diffusely to the nucleoplasm. Co-localization studies demonstrated that Gemini and Cajal bodies frequently abut a TRIM8 nuclear body. Truncating TRIM8 DNVs cause a neuro-renal syndrome via aberrant TRIM8 localization, implicating nuclear bodies in FSGS and developmental brain disease.


Assuntos
Proteínas de Transporte/genética , Deficiências do Desenvolvimento/genética , Epilepsia/genética , Glomerulosclerose Segmentar e Focal/genética , Espaço Intranuclear/metabolismo , Síndrome Nefrótica/genética , Síndrome Nefrótica/metabolismo , Proteínas do Tecido Nervoso/genética , Adulto , Animais , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Linhagem Celular , Criança , Pré-Escolar , Códon sem Sentido , Deficiências do Desenvolvimento/metabolismo , Epilepsia/metabolismo , Feminino , Glomerulosclerose Segmentar e Focal/metabolismo , Humanos , Rim/metabolismo , Masculino , Camundongos , Mutação , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/metabolismo , Fenótipo , Podócitos/metabolismo , Sequenciamento do Exoma
14.
Kidney Int ; 99(2): 336-345, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33122025

RESUMO

The Rho family of small GTPases (Rho GTPases) are the master regulators of the actin cytoskeleton and consist of 22 members. Previous studies implicated dysregulation of Rho GTPases in podocytes in the pathogenesis of proteinuric glomerular diseases. Rho GTPases are primarily regulated by the three families of proteins; guanine nucleotide exchange factors (GEFs; 82 members), GTPase-activating proteins (GAPs; 69 members), and GDP dissociation inhibitors (GDIs; 3 members). Since the regulatory proteins far outnumber their substrate Rho GTPases and act in concert in a cell/context-dependent manner, the upstream regulatory mechanism directing Rho GTPases in podocytes is largely unknown. In this review, we summarize recent advances in the understanding of the role of Rho GTPase regulatory proteins in podocytes, including the known mutations of these proteins that cause proteinuria in humans. We also provide critical appraisal of the in vivo and in vitro studies and identify the knowledge gap in the field that will require further studies.


Assuntos
Proteínas Monoméricas de Ligação ao GTP , Podócitos , Proteínas Ativadoras de GTPase/genética , Proteínas Ativadoras de GTPase/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Humanos , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Podócitos/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo
15.
Nephrol Dial Transplant ; 36(2): 237-246, 2021 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-33097957

RESUMO

BACKGROUND: An underlying monogenic cause of early-onset chronic kidney disease (CKD) can be detected in ∼20% of individuals. For many etiologies of CKD manifesting before 25 years of age, >200 monogenic causative genes have been identified to date, leading to the elucidation of mechanisms of renal pathogenesis. METHODS: In 51 families with echogenic kidneys and CKD, we performed whole-exome sequencing to identify novel monogenic causes of CKD. RESULTS: We discovered a homozygous truncating mutation in the transcription factor gene transcription factor CP2-like 1 (TFCP2L1) in an Arabic patient of consanguineous descent. The patient developed CKD by the age of 2 months and had episodes of severe hypochloremic, hyponatremic and hypokalemic alkalosis, seizures, developmental delay and hypotonia together with cataracts. We found that TFCP2L1 was localized throughout kidney development particularly in the distal nephron. Interestingly, TFCP2L1 induced the growth and development of renal tubules from rat mesenchymal cells. Conversely, the deletion of TFCP2L1 in mice was previously shown to lead to reduced expression of renal cell markers including ion transporters and cell identity proteins expressed in different segments of the distal nephron. TFCP2L1 localized to the nucleus in HEK293T cells only upon coexpression with its paralog upstream-binding protein 1 (UBP1). A TFCP2L1 mutant complementary DNA (cDNA) clone that represented the patient's mutation failed to form homo- and heterodimers with UBP1, an essential step for its transcriptional activity. CONCLUSION: Here, we identified a loss-of-function TFCP2L1 mutation as a potential novel cause of CKD in childhood accompanied by a salt-losing tubulopathy.


Assuntos
Transição Epitelial-Mesenquimal , Nefropatias/etiologia , Mutação , Proteínas Repressoras/genética , Animais , Criança , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Feminino , Células HEK293 , Humanos , Nefropatias/metabolismo , Nefropatias/patologia , Camundongos , Camundongos Knockout , Ratos , Proteínas Repressoras/metabolismo , Análise de Célula Única , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Sequenciamento do Exoma
16.
Am J Hum Genet ; 107(6): 1113-1128, 2020 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-33232676

RESUMO

The discovery of >60 monogenic causes of nephrotic syndrome (NS) has revealed a central role for the actin regulators RhoA/Rac1/Cdc42 and their effectors, including the formin INF2. By whole-exome sequencing (WES), we here discovered bi-allelic variants in the formin DAAM2 in four unrelated families with steroid-resistant NS. We show that DAAM2 localizes to the cytoplasm in podocytes and in kidney sections. Further, the variants impair DAAM2-dependent actin remodeling processes: wild-type DAAM2 cDNA, but not cDNA representing missense variants found in individuals with NS, rescued reduced podocyte migration rate (PMR) and restored reduced filopodia formation in shRNA-induced DAAM2-knockdown podocytes. Filopodia restoration was also induced by the formin-activating molecule IMM-01. DAAM2 also co-localizes and co-immunoprecipitates with INF2, which is intriguing since variants in both formins cause NS. Using in vitro bulk and TIRF microscopy assays, we find that DAAM2 variants alter actin assembly activities of the formin. In a Xenopus daam2-CRISPR knockout model, we demonstrate actin dysregulation in vivo and glomerular maldevelopment that is rescued by WT-DAAM2 mRNA. We conclude that DAAM2 variants are a likely cause of monogenic human SRNS due to actin dysregulation in podocytes. Further, we provide evidence that DAAM2-associated SRNS may be amenable to treatment using actin regulating compounds.


Assuntos
Actinas/metabolismo , Variação Genética , Proteínas dos Microfilamentos/genética , Síndrome Nefrótica/genética , Proteínas rho de Ligação ao GTP/genética , Alelos , Animais , Animais Geneticamente Modificados , Movimento Celular/genética , Citoplasma/metabolismo , Forminas/metabolismo , Humanos , Rim/metabolismo , Glomérulos Renais/metabolismo , Mutação de Sentido Incorreto , Podócitos/metabolismo , Pseudópodes/metabolismo , RNA Interferente Pequeno/metabolismo , Sequenciamento do Exoma , Xenopus
17.
Am J Hum Genet ; 107(4): 727-742, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32891193

RESUMO

Congenital anomalies of the kidney and urinary tract (CAKUT) constitute one of the most frequent birth defects and represent the most common cause of chronic kidney disease in the first three decades of life. Despite the discovery of dozens of monogenic causes of CAKUT, most pathogenic pathways remain elusive. We performed whole-exome sequencing (WES) in 551 individuals with CAKUT and identified a heterozygous de novo stop-gain variant in ZMYM2 in two different families with CAKUT. Through collaboration, we identified in total 14 different heterozygous loss-of-function mutations in ZMYM2 in 15 unrelated families. Most mutations occurred de novo, indicating possible interference with reproductive function. Human disease features are replicated in X. tropicalis larvae with morpholino knockdowns, in which expression of truncated ZMYM2 proteins, based on individual mutations, failed to rescue renal and craniofacial defects. Moreover, heterozygous Zmym2-deficient mice recapitulated features of CAKUT with high penetrance. The ZMYM2 protein is a component of a transcriptional corepressor complex recently linked to the silencing of developmentally regulated endogenous retrovirus elements. Using protein-protein interaction assays, we show that ZMYM2 interacts with additional epigenetic silencing complexes, as well as confirming that it binds to FOXP1, a transcription factor that has also been linked to CAKUT. In summary, our findings establish that loss-of-function mutations of ZMYM2, and potentially that of other proteins in its interactome, as causes of human CAKUT, offering new routes for studying the pathogenesis of the disorder.


Assuntos
Proteínas de Ligação a DNA/genética , Epigênese Genética , Fatores de Transcrição Forkhead/genética , Mutação , Proteínas Repressoras/genética , Fatores de Transcrição/genética , Sistema Urinário/metabolismo , Anormalidades Urogenitais/genética , Proteínas de Anfíbios/antagonistas & inibidores , Proteínas de Anfíbios/genética , Proteínas de Anfíbios/metabolismo , Animais , Estudos de Casos e Controles , Criança , Pré-Escolar , Proteínas de Ligação a DNA/metabolismo , Família , Feminino , Fatores de Transcrição Forkhead/metabolismo , Heterozigoto , Humanos , Lactente , Larva/genética , Larva/crescimento & desenvolvimento , Larva/metabolismo , Masculino , Camundongos , Camundongos Knockout , Morfolinos/genética , Morfolinos/metabolismo , Linhagem , Ligação Proteica , Proteínas Repressoras/metabolismo , Fatores de Transcrição/metabolismo , Sistema Urinário/anormalidades , Anormalidades Urogenitais/metabolismo , Anormalidades Urogenitais/patologia , Sequenciamento do Exoma , Xenopus
18.
Hum Genet ; 138(10): 1105-1115, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31230195

RESUMO

Congenital anomalies of the kidney and urinary tract (CAKUT) are the most common cause of chronic kidney disease (~ 45%) that manifests before 30 years of age. The genetic locus containing COL4A1 (13q33-34) has been implicated in vesicoureteral reflux (VUR), but mutations in COL4A1 have not been reported in CAKUT. We hypothesized that COL4A1 mutations cause CAKUT in humans. We performed whole exome sequencing (WES) in 550 families with CAKUT. As negative control cohorts we used WES sequencing data from patients with nephronophthisis (NPHP) with no genetic cause identified (n = 257) and with nephrotic syndrome (NS) due to monogenic causes (n = 100). We identified a not previously reported heterozygous missense variant in COL4A1 in three siblings with isolated VUR. When examining 549 families with CAKUT, we identified nine additional different heterozygous missense mutations in COL4A1 in 11 individuals from 11 unrelated families with CAKUT, while no COL4A1 mutations were identified in a control cohort with NPHP and only one in the cohort with NS. Most individuals (12/14) had isolated CAKUT with no extrarenal features. The predominant phenotype was VUR (9/14). There were no clinical features of the COL4A1-related disorders (e.g., HANAC syndrome, porencephaly, tortuosity of retinal arteries). Whereas COL4A1-related disorders are typically caused by glycine substitutions in the collagenous domain (84.4% of variants), only one variant in our cohort is a glycine substitution within the collagenous domain (1/10). We identified heterozygous COL4A1 mutations as a potential novel autosomal dominant cause of CAKUT that is allelic to the established COL4A1-related disorders and predominantly caused by non-glycine substitutions.


Assuntos
Colágeno Tipo IV/genética , Anormalidades Congênitas/diagnóstico , Anormalidades Congênitas/genética , Rim/anormalidades , Mutação , Fenótipo , Sistema Urinário/anormalidades , Alelos , Substituição de Aminoácidos , Biologia Computacional/métodos , Análise Mutacional de DNA , Bases de Dados Genéticas , Evolução Molecular , Feminino , Estudos de Associação Genética , Loci Gênicos , Genômica/métodos , Heterozigoto , Humanos , Doenças Renais Císticas/diagnóstico , Doenças Renais Císticas/genética , Masculino , Síndrome Nefrótica/diagnóstico , Síndrome Nefrótica/genética , Navegador , Sequenciamento do Exoma
19.
Can J Kidney Health Dis ; 6: 2054358119839080, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31065378

RESUMO

PURPOSE OF REVIEW: Infections are a major contributor to morbidity and mortality in end-stage renal disease (ESRD) patients. A better understanding of the interplay between infectious processes and ESRD may eventually lead to the development of targeted treatment strategies aimed at lowering overall disease morbidity and mortality. Monogenic causes are a major contributor to the development of adult chronic kidney disease (CKD). Recent studies identified a genetic cause in 10% to 20% of adults with CKD. With the introduction of whole-exome sequencing (WES) into clinical mainstay, this proportion is expected to increase in the future. Once patients develop CKD/ESRD due to a genetic cause, secondary changes, such as a compromised immune status, affect overall disease progression and clinical outcomes. Stratification according to genotype may enable us to study its effects on secondary disease outcomes, such as infectious risk. Moreover, this knowledge will enable us to better understand the molecular interplay between primary disease and secondary disease outcomes. SOURCES OF INFORMATION: We conducted a literature review using search engines such as PubMed, PubMed central, and Medline, as well as cumulative knowledge from our respective areas of expertise. METHODS: This is a transdisciplinary perspective on infectious complications in ESRD due to monogenic causes, such as autosomal dominant polycystic kidney disease (ADPKD), combining expertise in genomics and immunology. KEY FINDINGS: In ADPKD, infection is a frequent complication manifesting primarily as lower urinary tract infection and less frequently as renal infection. Infectious episodes may be a direct consequence of a specific underlying structural abnormality, for example the characteristic cysts, among others. However, evidence suggests that infectious disease risk is also increased in ESRD due to secondary not-well-understood disease mechanisms. These disease mechanisms may vary depending on the underlying nature of the primary disease. While the infectious disease risk is well documented in ADPKD, there are currently insufficient data on the risk in other monogenic causes of ESRD. WES in combination with novel technologies, such as RNA sequencing and single-cell RNA sequencing, can provide insight into the molecular mechanisms of disease progression in different monogenic causes of CKD/ESRD and may lead to the development of novel risk-stratification profiles in the future. LIMITATIONS: This is not a systematic review of the literature and the proposed perspective is tainted by the authors' point of view on the topic. IMPLICATIONS: WES in combination with novel technologies such as RNA sequencing may enable us to fully unravel underlying disease mechanisms and secondary disease outcomes in monogenic causes of CKD and better characterize individual risk profiles. This understanding will hopefully facilitate the development of novel targeted therapies.


CONTEXTE MOTIVANT LA REVUE: Les infections contribuent largement à la morbidité et à la mortalité observées chez les patients atteints d'insuffisance rénale terminale (IRT). Une meilleure compréhension des interactions entre le processus infectieux et l'IRT pourrait éventuellement mener au développement de traitements ciblés visant la réduction de la morbidité et de la mortalité liées à la maladie. Les causes monogéniques sont en bonne partie responsables du développement de l'insuffisance rénale chronique (IRC) chez l'adulte. Des études récentes pointent vers une cause génétique dans 10 à 20 % des cas d'IRC, une proportion qui devrait s'accroître avec l'introduction du séquençage de l'exome entier (WES) comme soutien clinique principal. Lorsque les patients évoluent vers l'IRC/IRT de cause génétique, des changements secondaires, notamment un état immunologique fragilisé, affectent la progression globale de la maladie et les résultats cliniques. La stratification selon le génotype pourrait permettre d'étudier ses effets sur l'issue de pathologies secondaires comme le risque infectieux. En outre, cette information nous permettrait de mieux comprendre l'interaction moléculaire entre les résultats des pathologies primaires et secondaires. SOURCES: Nous avons procédé à une revue de la littérature à l'aide des moteurs de recherche PubMed, PubMed central et Medline, de même qu'avec nos connaissances cumulatives dans nos domaines d'expertise respectifs. MÉTHODOLOGIE: Il s'agit d'une perspective interdisciplinaire sur les complications infectieuses en contexte d'IRT dues à des causes monogéniques, notamment une la polykystose rénale autosomique dominante (ADPKD), qui combine l'expertise en génomique et en immunologie. PRINCIPAUX RÉSULTATS: Les infections constituent une complication fréquente en contexte d'ADPKD et se manifestent principalement sous la forme d'une infection urinaire basse et moins souvent comme une infection rénale. Les épisodes infectieux pourraient être une conséquence directe d'une anomalie structurelle sous-jacente, notamment des kystes caractéristiques, entre autres. Toutefois, des données indiquent que le risque de maladie infectieuse en contexte d'IRT augmente aussi en raison de mécanismes secondaires mal connus; ceux-ci peuvent varier selon la nature sous-jacente de la pathologie primaire. Bien que le risque de maladie infectieuse soit bien documenté en contexte d'ADPKD, on dispose actuellement de données insuffisantes sur ce risque pour les autres causes monogéniques de l'IRT. Le WES, combiné aux nouvelles technologies telles que le séquençage d'ARN et le séquençage d'ARN unicellulaire, peut éclairer sur les mécanismes moléculaires régissant la progression de la maladie pour les différentes causes monogéniques de l'IRC/IRT et pourrait jouer un rôle dans l'élaboration de nouveaux profils de stratification des risques dans le futur. LIMITES: L'étude ne constitue pas une revue systématique de la littérature. De plus, la perspective proposée est teintée du point de vue des auteurs sur le sujet. IMPLICATIONS: Le WES, combiné aux nouvelles technologies telles que le séquençage d'ARN, pourrait nous permettre d'abord de mieux comprendre les mécanismes sous-jacents de la maladie et l'issue des pathologies secondaires des causes monogéniques de l'IRT, puis de mieux caractériser les profils de risque individuels. Ces informations, nous l'espérons, contribueront à faciliter le développement de nouveaux traitements ciblés.

20.
PLoS One ; 14(5): e0216426, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31095586

RESUMO

Focal segmental glomerulosclerosis (FSGS) is frequently found in biopsies of patients with steroid resistant nephrotic syndrome (SRNS). The pathogenesis of SRNS/FSGS is often unknown and the disease will recur in up to 50% of patients post-transplant, indicating the presence of circulating podocyte-toxic factor(s). Several studies have reported clinical improvement after anti-TNFα therapy. However, prediction of the clinical outcome in SRNS/FSGS is difficult, and novel predictive biomarkers are needed. An image-based assay, which measures disassembly of focal adhesion complexes in cultured podocytes, was used to ascertain the presence of podocyte toxic activity in SRNS/FSGS sera. Expression of TNFα pathway genes was analysed in the Nephroseq FSGS cohort and in cultured podocytes treated with SRNS/FSGS sera. Podocyte toxic activity was detected in 48/96 SRNS/FSGS patients. It did not correlate with serum TNFα levels, age, sex, ethnicity or glomerular filtration rate. In ~25% of the toxic samples, the toxicity was strongly inhibited by blockade of TNFα signaling. Transcriptional profiling of human FSGS biopsies and podocytes treated with FSGS sera revealed significant increases in expression of TNFα pathway genes. We identified patients with serum podocyte toxic activity who may be at risk for FSGS recurrence, and those patients in whom serum podocyte toxicity may be reversed by TNFα blockade. Activation of TNFα pathway genes occurs in podocytes of FSGS patients suggesting a causative effect of this pathway in response to circulating factor(s). In vitro analyses of patient sera may stratify patients according to prognostic outcomes and potential responses to specific clinical interventions.


Assuntos
Glomerulosclerose Segmentar e Focal/metabolismo , Síndrome Nefrótica/metabolismo , Podócitos/metabolismo , Soro/metabolismo , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo , Adolescente , Adulto , Biópsia , Linhagem Celular Transformada , Criança , Feminino , Glomerulosclerose Segmentar e Focal/patologia , Humanos , Masculino , Síndrome Nefrótica/patologia , Podócitos/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...