Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 733
Filtrar
1.
BMC Pregnancy Childbirth ; 24(1): 662, 2024 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-39394069

RESUMO

BACKGROUND: Prenatal ultrasound findings of fetal bilateral echogenic kidneys accompanied by oligohydramnios can be highly stressful for both pregnant women and physicians. The diversity of underlying causes makes it challenging to confirm a prenatal diagnosis, predict postnatal outcomes, and counsel regarding recurrence risks in future pregnancies. CASE PRESENTATION: We report two cases of abnormal fetal echogenic kidneys with oligohydramnios detected in the early third trimester. Autosomal recessive polycystic kidney disease (ARPKD), a rare genetic syndrome, was initially suspected in both cases. However, postnatal diagnoses differed: the first case was confirmed as glomerulocystic kidney disease (GCKD) through renal pathology, while the second case was diagnosed as ARPKD with a compound heterozygous likely pathogenic PKHD1 mutation. CONCLUSION: Prenatal diagnosis of fetal echogenic kidneys with oligohydramnios should prioritize accurate diagnosis. Given the differences in the clinical spectrum, GCKD should be considered a differential diagnosis for this condition, particularly ARPKD. This study highlights the importance and benefits of molecular diagnosis and postnatal renal pathology for precise diagnosis and effective counseling.


Assuntos
Rim , Oligo-Hidrâmnio , Rim Policístico Autossômico Recessivo , Ultrassonografia Pré-Natal , Humanos , Feminino , Oligo-Hidrâmnio/diagnóstico por imagem , Gravidez , Adulto , Rim/anormalidades , Rim/diagnóstico por imagem , Rim Policístico Autossômico Recessivo/diagnóstico por imagem , Rim Policístico Autossômico Recessivo/complicações , Rim Policístico Autossômico Recessivo/genética , Diagnóstico Diferencial , Mutação , Receptores de Superfície Celular
2.
Zhongguo Dang Dai Er Ke Za Zhi ; 26(9): 954-960, 2024.
Artigo em Chinês | MEDLINE | ID: mdl-39267511

RESUMO

A 5-year-old girl was admitted due to one episode of melena and one episode of hematemesis. Upon admission, gastroscopy revealed esophageal and gastric varices. Abdominal CT scan, MRI, and color Doppler ultrasound suggested cirrhosis, intrahepatic bile duct dilation, and bilateral kidney enlargement. Genetic testing identified compound heterozygous mutations in the PKHD1 gene: c.2264C>T (p.Pro755Leu) and c.1886T>C (p.Val629Ala). The c.2264C>T (p.Pro755Leu) mutation is a known pathogenic variant with previous reports, while c.1886T>C (p.Val629Ala) is a novel mutation predicted to have pathogenic potential according to Mutation Taster and PolyPhen2. The child was diagnosed with autosomal recessive polycystic kidney disease. In children presenting with gastrointestinal bleeding without obvious causes, particularly those with liver or kidney disease, consideration should be given to the possibility of autosomal recessive polycystic kidney disease, and genetic testing should be conducted for definitive diagnosis when necessary.


Assuntos
Rim Policístico Autossômico Recessivo , Humanos , Feminino , Rim Policístico Autossômico Recessivo/genética , Rim Policístico Autossômico Recessivo/complicações , Pré-Escolar , Mutação , Receptores de Superfície Celular/genética
3.
Medicine (Baltimore) ; 103(31): e39216, 2024 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-39093746

RESUMO

BACKGROUND: This study aims to report the clinical characteristics of a child with autosomal recessive polycystic kidney disease (ARPKD) within a Chinese Zhuang ethnic family. METHODS: We used whole exome sequencing (WES) in the family to examine the genetic cause of the disease. Candidate pathogenic variants were validated by Sanger sequencing. RESULTS: We identified previously unreported mutations in the PKHD1 gene of the proband with ARPKD through WES: a splice site mutation c.6809-2A > T, a nonsense mutation c.4192C > T(p.Gln1398Ter), and a missense mutation c.2181T > G(p.Asn727Lys). Her mother is a heterozygous carrier of c.2181T > G(p.Asn727Lys) mutation. Her father is a carrier of c.6809-2A > T mutation and c.4192C > T(p.Gln1398Ter) mutation. CONCLUSIONS: The identification of novel mutations in the PKHD1 gene through WES not only expands the spectrum of known variants but also potentially enhances genetic counseling and prenatal diagnostic approaches for families affected by ARPKD.


Assuntos
Códon sem Sentido , Linhagem , Rim Policístico Autossômico Recessivo , Receptores de Superfície Celular , Humanos , Rim Policístico Autossômico Recessivo/genética , Receptores de Superfície Celular/genética , Feminino , China , Masculino , Sequenciamento do Exoma , Sítios de Splice de RNA/genética , Mutação de Sentido Incorreto , Adulto , População do Leste Asiático
4.
Biomedica ; 44(Sp. 1): 27-41, 2024 05 31.
Artigo em Inglês, Espanhol | MEDLINE | ID: mdl-39079142

RESUMO

Renal cystic diseases are common conditions whose etiology can be highly heterogeneous. They require a correct approach for adequate diagnosis and management. We aimed to illustrate part of the spectrum of renal cystic diseases through some clinical cases managed in our service. We describe 11 clinical cases including clinical entities such as renal multicystic dysplasia, and autosomal dominant and autosomal recessive polycystic renal disease, among other pathologies. Renal cystic diseases are heterogeneous in their clinical presentation, natural history, radiological findings, and genetic and pathophysiological basis. An integral clinical approach is needed to get a clear etiological diagnosis and offer adequate individualized care and follow-up for patients.


Las enfermedades quísticas renales son condiciones frecuentes cuya etiología puede ser muy heterogénea, por lo que se requiere un adecuado abordaje para su diagnóstico y manejo. El objetivo de este trabajo fue ilustrar parte del espectro de la enfermedad renal quística por medio de casos clínicos manejados en la Fundación Valle del Lili. Se describen 11 casos clínicos que incluyen enfermedades como displasia multiquística renal, enfermedad poliquística renal autosómica dominante y autosómica recesiva, entre otras. Las enfermedades quísticas renales varían en su presentación clínica, historia natural, hallazgos imagenológicos, bases genéticas y fisiopatológicas, por consiguiente, el enfoque diagnóstico y el manejo integral se debe realizar de forma individualizada y con un abordaje multidisciplinario.


Assuntos
Doenças Renais Císticas , Humanos , Criança , Masculino , Doenças Renais Císticas/genética , Doenças Renais Císticas/diagnóstico por imagem , Feminino , Pré-Escolar , Lactente , Adolescente , Rim Policístico Autossômico Dominante/genética , Rim Policístico Autossômico Dominante/diagnóstico por imagem , Rim Policístico Autossômico Recessivo/genética , Rim Policístico Autossômico Recessivo/diagnóstico por imagem , Rim Displásico Multicístico/genética , Rim Displásico Multicístico/diagnóstico por imagem
5.
Biochim Biophys Acta Mol Basis Dis ; 1870(7): 167309, 2024 10.
Artigo em Inglês | MEDLINE | ID: mdl-38885798

RESUMO

INTRODUCTION: Autosomal Recessive Polycystic Kidney Disease (ARPKD) is a rare paediatric disease primarily caused by sequence variants in PKHD1. ARPKD presents with considerable clinical variability relating to the type of PKHD1 sequence variant, but not its position. Animal models of Polycystic Kidney Disease (PKD) suggest a complex genetic landscape, with genetic modifiers as a potential cause of disease variability. METHODS: To investigate in an unbiased manner the molecular mechanisms of ARPKD and identify potential indicators of disease severity, Whole Exome Sequencing (WES) and RNA-Sequencing (RNA-Seq) were employed on human ARPKD kidneys and age-matched healthy controls. RESULTS: WES confirmed the clinical diagnosis of ARPKD in our patient cohort consisting of ten ARPKD kidneys. Sequence variant type, nor position of PKHD1 sequence variants, was linked to disease severity. Sequence variants in genes associated with other ciliopathies were detected in the ARPKD cohort, but only PKD1 could be linked to disease severity. Transcriptomic analysis on a subset of four ARPKD kidneys representing severe and moderate ARPKD, identified a significant number of genes relating to WNT signalling, cellular metabolism and development. Increased expression of WNT signalling-related genes was validated by RT-qPCR in severe and moderate ARPKD kidneys. Two individuals in our cohort with the same PKHD1 sequence variants but different rates of kidney disease progression, with displayed transcriptomic differences in the expression of WNT signalling genes. CONCLUSION: ARPKD kidney transcriptomics highlights changes in WNT signalling as potentially significant in ARPKD manifestation and severity, providing indicators for slowing down the progression of ARPKD.


Assuntos
Rim Policístico Autossômico Recessivo , Receptores de Superfície Celular , Via de Sinalização Wnt , Humanos , Rim Policístico Autossômico Recessivo/genética , Rim Policístico Autossômico Recessivo/patologia , Rim Policístico Autossômico Recessivo/metabolismo , Via de Sinalização Wnt/genética , Masculino , Feminino , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Sequenciamento do Exoma , Criança , Índice de Gravidade de Doença , Sequenciamento de Nucleotídeos em Larga Escala , Adolescente , Pré-Escolar , Rim/metabolismo , Rim/patologia
6.
BMC Nephrol ; 25(1): 209, 2024 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-38918687

RESUMO

BACKGROUND: Autosomal recessive polycystic kidney disease (ARPKD) is a rare inherited cystic disease characterized by bilateral renal cyst formation and congenital liver fibrosis. Cardiovascular disorders such as noncompaction of ventricular myocardium (NVM) have not been reported with ARPKD. CASE PRESENTATION: A 5-month-old girl was examined after presenting with a fever and turbid urine for one day and was diagnosed as urinary tract infection. Urinary ultrasound showed multiple round, small cysts varying in size in both kidneys. Genetic testing revealed two heterozygous mutations and one exon deletion in the polycystic kidney and hepatic disease 1 gene, indicating a diagnosis of ARPKD. During hospitalization, she was found to have chronic heart failure after respiratory tract infection, with an ejection fraction of 29% and fraction shortening of 13%. When the patient was 15 months old, it was found that she had prominent trabeculations and deep intertrabecular recesses with the appearance of blood flow from the ventricular cavity into the intertrabecular recesses by echocardiography. The noncompaction myocardium was 0.716 cm and compaction myocardium was 0.221 cm (N/C = 3.27), indicating a diagnosis of NVM. Liver and kidney function remained normal during four-year follow-up. CONCLUSIONS: This is the first report of NVM in a patient with ARPKD. It is unsure if the coexistence of NVM and ARPKD is a coincidence or they are different manifestations of ciliary dysfunction in the heart and kidneys.


Assuntos
Rim Policístico Autossômico Recessivo , Humanos , Feminino , Rim Policístico Autossômico Recessivo/complicações , Rim Policístico Autossômico Recessivo/genética , Rim Policístico Autossômico Recessivo/diagnóstico por imagem , Lactente , Miocárdio Ventricular não Compactado Isolado/complicações , Miocárdio Ventricular não Compactado Isolado/genética , Miocárdio Ventricular não Compactado Isolado/diagnóstico por imagem , Ciliopatias/genética , Ciliopatias/complicações
7.
Adv Sci (Weinh) ; 11(24): e2308820, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38634253

RESUMO

Serving as the cell's sensory antennae, primary cilia are linked to numerous human genetic diseases when they malfunction. DZIP1L, identified as one of the genetic causes of human autosomal recessive polycystic kidney disease (ARPKD), is an evolutionarily conserved ciliary basal body protein. Although it has been reported that DZIP1L is involved in the ciliary entry of PKD proteins, the underlying mechanism remains elusive. Here, an uncharacterized role of DZIP1L is reported in modulating the architecture and function of transition fibers (TFs), striking ciliary base structures essential for selective cilia gating. Using C. elegans as a model, C01G5.7 (hereafter termed DZIP-1) is identified as the sole homolog of DZIP1L, which specifically localizes to TFs. While DZIP-1 or ANKR-26 (the ortholog of ANKRD26) deficiency shows subtle impact on TFs, co-depletion of DZIP-1 and ANKR-26 disrupts TF assembly and cilia gating for soluble and membrane proteins, including the ortholog of ADPKD protein polycystin-2. Notably, the synergistic role for DZIP1L and ANKRD26 in the formation and function of TFs is highly conserved in mammalian cilia. Hence, the findings illuminate an evolutionarily conserved role of DZIP1L in TFs architecture and function, highlighting TFs as a vital part of the ciliary gate implicated in ciliopathies ARPKD.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Cílios , Animais , Humanos , Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Cílios/metabolismo , Cílios/genética , Rim Policístico Autossômico Recessivo/metabolismo , Rim Policístico Autossômico Recessivo/genética , Rim Policístico Autossômico Recessivo/patologia
8.
Pediatr Nephrol ; 39(9): 2569-2578, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38261064

RESUMO

The incidence of rare diseases is expected to be comparatively higher in the Middle East and North Africa (MENA) region than in other parts of the world, attributed to the high prevalence of consanguinity. Most MENA countries share social and economic statuses, cultural relativism, religious beliefs, and healthcare policies. Polycystic kidney diseases (PKDs) are the most common genetic causes of kidney failure, accounting for nearly 8.0% of dialysis cases. The development of PKDs is linked to variants in several genes, including PKD1, PKD2, PKHD1, DZIP1L, and CYS1. Autosomal recessive PKD (ARPKD) is the less common yet aggressive form of PKD. ARPKD has an estimated incidence between 1:10,000 and 1:40,000. Most patients with ARPKD require kidney replacement therapy earlier than patients with autosomal dominant polycystic kidney disease (ADPKD), often in their early years of life. This review gathered data from published research studies and reviews of ARPKD, highlighting the epidemiology, phenotypic presentation, investigations, genetic analysis, outcomes, and management. Although limited data are available, the published literature suggests that the incidence of ARPKD may be higher in the MENA region due to consanguineous marriages. Patients with ARPKD from the MENA region usually present at a later disease stage and have a relatively short time to progress to kidney failure. Limited data are available regarding the management practice in the region, which warrants further investigations.


Assuntos
Rim Policístico Autossômico Recessivo , Humanos , Rim Policístico Autossômico Recessivo/genética , Rim Policístico Autossômico Recessivo/epidemiologia , Rim Policístico Autossômico Recessivo/terapia , Rim Policístico Autossômico Recessivo/diagnóstico , Oriente Médio/epidemiologia , África do Norte/epidemiologia , Incidência , Criança , Consanguinidade , Prevalência
9.
Am J Kidney Dis ; 83(6): 829-833, 2024 06.
Artigo em Inglês | MEDLINE | ID: mdl-38211685

RESUMO

The etiologies of newborn deaths in neonatal intensive care units usually remain unknown, even after genetic testing. Whole-genome sequencing, combined with artificial intelligence-based methods for predicting the effects of non-coding variants, provide an avenue for resolving these deaths. Using one such method, SpliceAI, we identified a maternally inherited deep intronic PKHD1 splice variant (chr6:52030169T>C), in trans with a pathogenic missense variant (p.Thr36Met), in a newborn who died of autosomal recessive polycystic kidney disease at age 2 days. We validated the deep intronic variant's impact in maternal urine-derived cells expressing PKHD1. Reverse transcription polymerase chain reaction followed by Sanger sequencing showed that the variant causes inclusion of 147bp of the canonical intron between exons 29 and 30 of PKHD1 into the mRNA, including a premature stop codon. Allele-specific expression analysis at a heterozygous site in the mother showed that the mutant allele completely suppresses canonical splicing. In an unrelated healthy control, there was no evidence of transcripts including the novel splice junction. We returned a diagnostic report to the parents, who underwent in vitro embryo selection.


Assuntos
Íntrons , Rim Policístico Autossômico Recessivo , Receptores de Superfície Celular , Humanos , Recém-Nascido , Masculino , Íntrons/genética , Mutação de Sentido Incorreto , Rim Policístico Autossômico Recessivo/genética , Rim Policístico Autossômico Recessivo/diagnóstico , Receptores de Superfície Celular/genética
11.
Nephron ; 148(4): 264-272, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-36617405

RESUMO

The clinical features of cerebellar vermis hypoplasia, oligophrenia, ataxia, coloboma, and hepatic fibrosis (COACH) characterize the rare autosomal recessive multisystem disorder called COACH syndrome. COACH syndrome belongs to the spectrum of Joubert syndrome and related disorders (JSRDs) and liver involvement distinguishes COACH syndrome from the rest of the JSRD spectrum. Developmental delay and oculomotor apraxia occur early but with time, these can improve and may not be readily apparent or no longer need active medical management. Congenital hepatic fibrosis and renal disease, on the other hand, may develop late, and the temporal incongruity in organ system involvement may delay the recognition of COACH syndrome. We present a case of a young adult presenting late to a Renal Genetics Clinic for evaluation of renal cystic disease with congenital hepatic fibrosis, clinically suspected to have autosomal recessive polycystic kidney disease. Following genetic testing, a reevaluation of his medical records from infancy, together with reverse phenotyping and genetic phasing, led to a diagnosis of COACH syndrome.


Assuntos
Anormalidades Múltiplas , Encéfalo/anormalidades , Vermis Cerebelar , Cerebelo/anormalidades , Colestase , Coloboma , Doenças Genéticas Inatas , Deficiência Intelectual , Hepatopatias , Malformações do Sistema Nervoso , Rim Policístico Autossômico Recessivo , Adulto Jovem , Humanos , Coloboma/diagnóstico , Coloboma/genética , Rim Policístico Autossômico Recessivo/diagnóstico , Rim Policístico Autossômico Recessivo/genética , Diagnóstico Tardio , Genótipo , Cirrose Hepática/genética , Ataxia/diagnóstico , Ataxia/genética , Deficiência Intelectual/genética , Deficiências do Desenvolvimento
12.
Ann Hum Genet ; 88(1): 58-75, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37905714

RESUMO

Autosomal recessive polycystic kidney disease is an early onset inherited hepatorenal disorder affecting around 1 in 20,000 births with no approved specific therapies. The disease is almost always caused by variations in the polycystic kidney and hepatic disease 1 gene, which encodes fibrocystin (FC), a very large, single-pass transmembrane glycoprotein found in primary cilia, urine and urinary exosomes. By comparison to proteins involved in autosomal dominant PKD, our structural and molecular understanding of FC has lagged far behind such that there are no published experimentally determined structures of any part of the protein. Bioinformatics analyses predict that the ectodomain contains a long chain of immunoglobulin-like plexin-transcription factor domains, a protective antigen 14 domain, a tandem G8-TMEM2 homology region and a sperm protein, enterokinase and agrin domain. Here we review current knowledge on the molecular function of the protein from a structural perspective.


Assuntos
Rim Policístico Autossômico Recessivo , Receptores de Superfície Celular , Humanos , Rim Policístico Autossômico Recessivo/genética , Rim Policístico Autossômico Recessivo/metabolismo , Receptores de Superfície Celular/química , Receptores de Superfície Celular/genética , RNA , Fatores de Transcrição/química , Proteínas do Espermatozoide/química , Conformação Proteica
13.
Medicine (Baltimore) ; 102(50): e36573, 2023 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-38115240

RESUMO

RATIONAL: The disease of Caroli is a rare congenital disorder, characterized by the dilated intrahepatic bile ducts, resulting from mutations in the PKHD1 gene. Caroli syndrome, characterized by dilated intrahepatic bile ducts with congenital hepatic fibrosis, is linked to autosomal recessive polycystic kidney disease. The clinical manifestations of Caroli disease are not typical, and Caroli disease is easy to be missed and misdiagnosed. Therefore, we reported this case in the hope of raising awareness of the disease among clinicians. PATIENT CONCERNS: The clinical manifestation of a 10-year-old girl was subcutaneous hemorrhage. DIAGNOSES: Magnetic resonance imaging (MRI ) indicates that the person may have Caroli disease, cirrhosis, splenomegaly, portal hypertension, esophagogastric fundal varices, or sponge kidneys. INTERVENTION: The patient was advised for liver transplantation. OUTCOMES: The patient parents did not take our treatment advice, and they asked to go to a better hospital for further treatment, so we did not give the patient any treatment. LESSONS: This case serves as a reminder that if we encounter a patient with hemophilia in our clinic, we should not only consider hematologic diseases and cirrhosis, but also perform an epigastric MRI and magnetic resonance cholangiopancreatography to rule out Caroli disease.


Assuntos
Doença de Caroli , Hipertensão Portal , Rim Policístico Autossômico Recessivo , Feminino , Humanos , Criança , Doença de Caroli/complicações , Doença de Caroli/diagnóstico , Cirrose Hepática/patologia , Rim Policístico Autossômico Recessivo/genética , Hemorragia/etiologia
14.
Adv Kidney Dis Health ; 30(5): 397-406, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-38097330

RESUMO

Polycystic kidney diseases are a group of monogenically inherited disorders characterized by cyst development in the kidney with defects in primary cilia function central to pathogenesis. Autosomal dominant polycystic kidney disease (ADPKD) has progressive cystogenesis and accounts for 5-10% of kidney failure (KF) patients. There are two major ADPKD genes, PKD1 and PKD2, and seven minor loci. PKD1 accounts for ∼80% of patients and is associated with the most severe disease (KF is typically at 55-65 years); PKD2 accounts for ∼15% of families, with KF typically in the mid-70s. The minor genes are generally associated with milder kidney disease, but for DNAJB11 and ALG5, the age at KF is similar to PKD2. PKD1 and PKD2 have a high level of allelic heterogeneity, with no single pathogenic variant accounting for >2% of patients. Additional genetic complexity includes biallelic disease, sometimes causing very early-onset ADPKD, and mosaicism. Autosomal dominant polycystic liver disease is characterized by severe PLD but limited PKD. The two major genes are PRKCSH and SEC63, while GANAB, ALG8, and PKHD1 can present as ADPKD or autosomal dominant polycystic liver disease. Autosomal recessive polycystic kidney disease typically has an infantile onset, with PKHD1 being the major locus and DZIP1L and CYS1 being minor genes. In addition, there are a range of mainly recessive syndromic ciliopathies with PKD as part of the phenotype. Because of the phenotypic and genic overlap between the diseases, employing a next-generation sequencing panel containing all known PKD and ciliopathy genes is recommended for clinical testing.


Assuntos
Ciliopatias , Hepatopatias , Rim Policístico Autossômico Dominante , Rim Policístico Autossômico Recessivo , Humanos , Rim Policístico Autossômico Dominante/genética , Mutação , Hepatopatias/genética , Rim Policístico Autossômico Recessivo/genética , Fenótipo
15.
Adv Kidney Dis Health ; 30(5): 468-476, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-38097335

RESUMO

Autosomal recessive polycystic kidney disease (ARPKD) is the rare and usually early-onset form of polycystic kidney disease with a typical clinical presentation of enlarged cystic kidneys and liver involvement with congenital hepatic fibrosis or Caroli syndrome. ARPKD remains a clinical challenge in pediatrics, frequently requiring continuous and long-term multidisciplinary treatment. In this review, we aim to give an overview over clinical aspects of ARPKD and recent developments in our understanding of disease progression, risk patterns, and treatment of ARPKD.


Assuntos
Doença de Caroli , Rim Policístico Autossômico Recessivo , Criança , Humanos , Rim Policístico Autossômico Recessivo/diagnóstico , Receptores de Superfície Celular , Prognóstico , Cirrose Hepática/diagnóstico , Doença de Caroli/diagnóstico
16.
Semin Nephrol ; 43(4): 151434, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37996359

RESUMO

Cystic kidney diseases, when broadly defined, have a wide differential diagnosis extending from recessive diseases with a prenatal or pediatric diagnosis, to the most common autosomal-dominant polycystic kidney disease primarily affecting adults, and several other genetic or acquired etiologies that can manifest with kidney cysts. The most likely diagnoses to consider when assessing a patient with cystic kidney disease differ depending on family history, age stratum, radiologic characteristics, and extrarenal features. Accurate identification of the underlying condition is crucial to estimate the prognosis and initiate the appropriate management, identification of extrarenal manifestations, and counseling on recurrence risk in future pregnancies. There are significant differences in the clinical approach to investigating and managing kidney cysts in children compared with adults. Next-generation sequencing has revolutionized the diagnosis of inherited disorders of the kidney, despite limitations in access and challenges in interpreting the data. Disease-modifying treatments are lacking in the majority of kidney cystic diseases. For adults with rapid progressive autosomal-dominant polycystic kidney disease, tolvaptan (V2-receptor antagonist) has been approved to slow the rate of decline in kidney function. In this article, we examine the differences in the differential diagnosis and clinical management of cystic kidney disease in children versus adults, and we highlight the progress in molecular diagnostics and therapeutics, as well as some of the gaps meriting further attention.


Assuntos
Cistos , Neoplasias Renais , Rim Policístico Autossômico Dominante , Rim Policístico Autossômico Recessivo , Adulto , Gravidez , Feminino , Criança , Humanos , Rim Policístico Autossômico Recessivo/diagnóstico , Rim Policístico Autossômico Recessivo/genética , Rim Policístico Autossômico Recessivo/terapia , Rim , Rim Policístico Autossômico Dominante/complicações , Rim Policístico Autossômico Dominante/diagnóstico , Rim Policístico Autossômico Dominante/genética , Cistos/diagnóstico , Cistos/genética , Cistos/terapia
17.
Rev Assoc Med Bras (1992) ; 69(11): e20230334, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37909612

RESUMO

OBJECTIVE: Autosomal dominant polycystic kidney disease is an inherited kidney disorder with mutations in polycystin-1 or polycystin-2. Autosomal recessive polycystic kidney disease is a severe form of polycystic kidney disease that is characterized by enlarged kidneys and congenital hepatic fibrosis. Mutations at PKHD1 are responsible for all typical forms of autosomal recessive polycystic kidney disease. METHODS: We evaluated the children diagnosed with polycystic kidney disease between October 2020 and May 2022. The diagnosis was established by family history, ultrasound findings, and/or genetic analysis. The demographic, clinical, and laboratory findings were evaluated retrospectively. RESULTS: There were 28 children (male/female: 11:17) evaluated in this study. Genetic analysis was performed in all patients (polycystin-1 variants in 13, polycystin-2 variants in 7, and no variants in 8 patients). A total of 18 variants in polycystin-1 and polycystin-2 were identified and 9 (50%) of them were not reported before. A total of eight novel variants were identified as definite pathogenic or likely pathogenic mutations. There was no variant detected in the PKDH1 gene. CONCLUSION: Our results highlighted molecular features of Turkish children with polycystic kidney disease and demonstrated novel variations that can be utilized in clinical diagnosis and prognosis.


Assuntos
Rim Policístico Autossômico Recessivo , Humanos , Masculino , Criança , Feminino , Rim Policístico Autossômico Recessivo/genética , Rim Policístico Autossômico Recessivo/diagnóstico , Rim Policístico Autossômico Recessivo/patologia , Canais de Cátion TRPP/genética , Estudos Retrospectivos , Receptores de Superfície Celular/genética , Rim/patologia , Mutação
18.
Nat Commun ; 14(1): 6513, 2023 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-37845212

RESUMO

Fibrocystin/Polyductin (FPC), encoded by PKHD1, is associated with autosomal recessive polycystic kidney disease (ARPKD), yet its precise role in cystogenesis remains unclear. Here we show that FPC undergoes complex proteolytic processing in developing kidneys, generating three soluble C-terminal fragments (ICDs). Notably, ICD15, contains a novel mitochondrial targeting sequence at its N-terminus, facilitating its translocation into mitochondria. This enhances mitochondrial respiration in renal epithelial cells, partially restoring impaired mitochondrial function caused by FPC loss. FPC inactivation leads to abnormal ultrastructural morphology of mitochondria in kidney tubules without cyst formation. Moreover, FPC inactivation significantly exacerbates renal cystogenesis and triggers severe pancreatic cystogenesis in a Pkd1 mouse mutant Pkd1V/V in which cleavage of Pkd1-encoded Polycystin-1 at the GPCR Proteolysis Site is blocked. Deleting ICD15 enhances renal cystogenesis without inducing pancreatic cysts in Pkd1V/V mice. These findings reveal a direct link between FPC and a mitochondrial pathway through ICD15 cleavage, crucial for cystogenesis mechanisms.


Assuntos
Cisto Pancreático , Rim Policístico Autossômico Recessivo , Camundongos , Animais , Receptores de Superfície Celular/metabolismo , Rim/metabolismo , Rim Policístico Autossômico Recessivo/metabolismo , Canais de Cátion TRPP/genética , Canais de Cátion TRPP/metabolismo , Túbulos Renais/metabolismo
19.
J Mol Med (Berl) ; 101(9): 1141-1151, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37584738

RESUMO

Autosomal-recessive polycystic kidney disease (ARPKD; MIM #263200) is a severe, hereditary, hepato-renal fibrocystic disorder that causes early childhood morbidity and mortality. Mutations in the polycystic kidney and hepatic disease 1 (PKHD1) gene, which encodes the protein fibrocystin/polyductin complex (FPC), cause all typical forms of ARPKD. Several mouse lines carrying diverse, genetically engineered disruptions in the orthologous Pkhd1 gene have been generated, but none expresses the classic ARPKD renal phenotype. In the current study, we characterized a spontaneous mouse Pkhd1 mutation that is transmitted as a recessive trait and causes cysticliver (cyli), similar to the hepato-biliary disease in ARPKD, but which is exacerbated by age, sex, and parity. We mapped the mutation to Chromosome 1 and determined that an insertion/deletion mutation causes a frameshift within Pkhd1 exon 48, which is predicted to result in a premature termination codon (UGA). Pkhd1cyli/cyli (cyli) mice exhibit a severe liver pathology but lack renal disease. Further analysis revealed that several alternatively spliced Pkhd1 mRNA, all containing exon 48, were expressed in cyli kidneys, but in lower abundance than in wild-type kidneys, suggesting that these transcripts escaped from nonsense-mediated decay (NMD). We identified an AAAAAT motif in exon 48 upstream of the cyli mutation which could enable ribosomal frameshifting, thus potentially allowing production of sufficient amounts of FPC for renoprotection. This mechanism, expressed in a species-specific fashion, may help explain the disparities in the renal phenotype observed between Pkhd1 mutant mice and patients with PKHD1-related disease. KEY MESSAGES: The Pkhd1cyli/cyli mouse expresses cystic liver disease, but no kidney phenotype. Pkhd1 mRNA expression is decreased in cyli liver and kidneys compared to wild-type. Ribosomal frameshifting may be responsible for Pkhd1 mRNA escape from NMD. Pkhd1 mRNA escape from NMD could contribute to the absent kidney phenotype.


Assuntos
Hepatopatias , Rim Policístico Autossômico Recessivo , Pré-Escolar , Camundongos , Humanos , Animais , Rim Policístico Autossômico Recessivo/genética , Rim Policístico Autossômico Recessivo/patologia , Rim/metabolismo , Mutação , Fatores de Transcrição/genética , RNA Mensageiro/genética , Receptores de Superfície Celular/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA