51
|
Rowe JH. 50 Years Ago in TheJournalofPediatrics: Denys-Drash Syndrome links developmental biology to oncogenesis. J Pediatr 2020; 219:145. [PMID: 32204799 DOI: 10.1016/j.jpeds.2019.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Jared H Rowe
- Division of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
52
|
Cheng C, Chen L, Wen S, Lin Z, Jiang X. Case Report: Denys-Drash Syndrome With WT1 Causative Variant Presenting as Atypical Hemolytic Uremic Syndrome. Front Pediatr 2020; 8:605889. [PMID: 33392118 PMCID: PMC7775732 DOI: 10.3389/fped.2020.605889] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 11/03/2020] [Indexed: 01/04/2023] Open
Abstract
The WT1 variant is confirmed to be pathogenic for Denys-Drash syndrome (DDS), a rare disorder characterized by early-onset nephrotic syndrome and renal failure, pseudo-hermaphroditism, and a high risk of Wilms' tumor. Several cases of DDS presenting with atypical hemolytic uremic syndrome (aHUS) have been reported. Here we report the case of a 2-year-old child who was diagnosed with WT1 missense variant, associated with DDS and initial presentation of aHUS. Complement factor H autoantibodies were negative. Complement regulatory system-related gene variants were not found, but a de novo heterozygous c.754G>A missense variant in exon 9 of WT1 gene was detected, resulting in a p. Asp252Asn substitution, by next-generation sequencing. The patient was a female morphologically but proved to be a genetic male because of karyotype 46, XY with normally developed female external genitalia. Bilateral nephrectomy and renal transplantation were performed 1 year later, and there was no recurrence of aHUS at 10 months after transplantation.
Collapse
Affiliation(s)
- Cheng Cheng
- Department of Pediatrics, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Lizhi Chen
- Department of Pediatrics, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Sijia Wen
- Department of Pediatrics, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhilang Lin
- Department of Pediatrics, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaoyun Jiang
- Department of Pediatrics, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
53
|
Wang D, Horton JR, Zheng Y, Blumenthal RM, Zhang X, Cheng X. Role for first zinc finger of WT1 in DNA sequence specificity: Denys-Drash syndrome-associated WT1 mutant in ZF1 enhances affinity for a subset of WT1 binding sites. Nucleic Acids Res 2019; 46:3864-3877. [PMID: 29294058 PMCID: PMC5934627 DOI: 10.1093/nar/gkx1274] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 12/22/2017] [Indexed: 12/14/2022] Open
Abstract
Wilms tumor protein (WT1) is a Cys2-His2 zinc-finger transcription factor vital for embryonic development of the genitourinary system. The protein contains a C-terminal DNA binding domain with four tandem zinc-fingers (ZF1-4). An alternative splicing of Wt1 can add three additional amino acids-lysine (K), threonine (T) and serine (S)-between ZF3 and ZF4. In the -KTS isoform, ZF2-4 determine the sequence-specificity of DNA binding, whereas the function of ZF1 remains elusive. Three X-ray structures are described here for wild-type -KTS isoform ZF1-4 in complex with its cognate DNA sequence. We observed four unique ZF1 conformations. First, like ZF2-4, ZF1 can be positioned continuously in the DNA major groove forming a 'near-cognate' complex. Second, while ZF2-4 make base-specific interactions with one DNA molecule, ZF1 can interact with a second DNA molecule (or, presumably, two regions of the same DNA molecule). Third, ZF1 can intercalate at the joint of two tail-to-head DNA molecules. If such intercalation occurs on a continuous DNA molecule, it would kink the DNA at the ZF1 binding site. Fourth, two ZF1 units can dimerize. Furthermore, we examined a Denys-Drash syndrome-associated ZF1 mutation (methionine at position 342 is replaced by arginine). This mutation enhances WT1 affinity for a guanine base. X-ray crystallography of the mutant in complex with its preferred sequence revealed the interactions responsible for this affinity change. These results provide insight into the mechanisms of action of WT1, and clarify the fact that ZF1 plays a role in determining sequence specificity of this critical transcription factor.
Collapse
Affiliation(s)
- Dongxue Wang
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - John R Horton
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yu Zheng
- RGENE, Inc., 953 Indiana Street, San Francisco, CA 94107, USA
| | - Robert M Blumenthal
- Department of Medical Microbiology and Immunology, and Program in Bioinformatics, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Xing Zhang
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xiaodong Cheng
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA.,Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
54
|
Rahimi N, Zhang Y, Mina A, Altman JK, Sukhanova M, Frankfurt O, Jennings L, Lu X, Behdad A, Chen Q, Chen YH, Gao J. An integrative approach reveals genetic complexity and epigenetic perturbation in acute promyelocytic leukemia: a single institution experience. Hum Pathol 2019; 91:1-10. [PMID: 31125631 DOI: 10.1016/j.humpath.2019.05.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 05/08/2019] [Accepted: 05/08/2019] [Indexed: 12/14/2022]
Abstract
Acute promyelocytic leukemia (APL) is a distinct type of acute myeloid leukemia that is defined by the presence of the translocations that mostly involve the RARA gene. The most frequent translocation is the t(15;17), which fuses the RARA gene with the PML gene. Previous studies have shown that other cooperative mutations are required for the development of APL after the initiating event of the t(15;17). In this study, we combined cytogenetics with next-generation sequencing and single-nucleotide polymorphism array to study the genetic complexity in 20 APL cases diagnosed in our institution. All but 3 cases had additional genetic aberrations. Our study demonstrated that somatic mutations are frequent events in APL. In addition to the previously reported recurrent cooperative mutations in the FLT3, WT1, and RAS genes, we identified mutations in several epigenetic modifiers, including TET2, EZH2, and DNMT3A, co-occurring with either FLT3 or WT1 mutations. Mutations of the WT1 gene and chromosome 11p copy neutral loss of heterozygosity affecting WT1 are present in a third of the cases in our series. Two-thirds of APL cases in our study demonstrated a global reduction but focal accumulation of H3K27 methylase (H3K27me) expression, indicating a disorganized chromatin methylation pattern with generally more accessible chromatin status. Our study confirmed genetic complexity of APL and revealed that epigenetic aberrations are more common than previously expected. Although epigenetic modulation is not a common treatment strategy in APL, targeting this pathway may have some clinical utility in refractory or relapsed APL cases.
Collapse
Affiliation(s)
- Nina Rahimi
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Yanming Zhang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Alain Mina
- Department of Hematology and Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Jessica K Altman
- Department of Hematology and Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Madina Sukhanova
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Olga Frankfurt
- Department of Hematology and Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Lawrence Jennings
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Xinyan Lu
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Amir Behdad
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Qing Chen
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Yi-Hua Chen
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Juehua Gao
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
55
|
Hamatani H, Sakairi T, Ikeuchi H, Kaneko Y, Maeshima A, Nojima Y, Hiromura K. TGF‐β1 alters DNA methylation levels in promoter and enhancer regions of the
WT1
gene in human podocytes. Nephrology (Carlton) 2019; 24:575-584. [DOI: 10.1111/nep.13411] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/23/2018] [Indexed: 01/09/2023]
Affiliation(s)
- Hiroko Hamatani
- Department of Nephrology and RheumatologyGunma University Graduate School of Medicine Maebashi Japan
| | - Toru Sakairi
- Department of Nephrology and RheumatologyGunma University Graduate School of Medicine Maebashi Japan
| | - Hidekazu Ikeuchi
- Department of Nephrology and RheumatologyGunma University Graduate School of Medicine Maebashi Japan
| | - Yoriaki Kaneko
- Department of Nephrology and RheumatologyGunma University Graduate School of Medicine Maebashi Japan
| | - Akito Maeshima
- Department of Nephrology and RheumatologyGunma University Graduate School of Medicine Maebashi Japan
| | - Yoshihisa Nojima
- Department of Rheumatology and NephrologyJapan Red Cross Maebashi Hospital Maebashi Japan
| | - Keiju Hiromura
- Department of Nephrology and RheumatologyGunma University Graduate School of Medicine Maebashi Japan
| |
Collapse
|
56
|
Voigt EA, Haralambieva IH, Larrabee BL, Kennedy RB, Ovsyannikova IG, Schaid DJ, Poland GA. Polymorphisms in the Wilms Tumor Gene Are Associated With Interindividual Variations in Rubella Virus-Specific Cellular Immunity After Measles-Mumps-Rubella II Vaccination. J Infect Dis 2019; 217:560-566. [PMID: 29253144 DOI: 10.1093/infdis/jix538] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 10/03/2017] [Indexed: 02/07/2023] Open
Abstract
Rubella vaccination induces widely variable immune responses in vaccine recipients. While rubella vaccination is effective at inducing immunity to rubella infection in most subjects, up to 5% of individuals do not achieve or maintain long-term protective immunity. To expand upon our previous work identifying genetic polymorphisms that are associated with these interindividual differences in humoral immunity to rubella virus, we performed a genome-wide association study in a large cohort of 1843 subjects to discover single-nucleotide polymorphisms (SNPs) associated with rubella virus-specific cellular immune responses. We identified SNPs in the Wilms tumor protein gene (WT1) that were significantly associated (P < 5 × 10-8) with interindividual variations in rubella-specific interleukin 6 secretion from subjects' peripheral blood mononuclear cells postvaccination. No SNPs were found to be significantly associated with variations in rubella-specific interferon-γ secretion. Our findings demonstrate that genetic polymorphisms in the WT1 gene in subjects of European ancestry are associated with interindividual differences in rubella virus-specific cellular immunity after measles-mumps-rubella II vaccination.
Collapse
Affiliation(s)
- Emily A Voigt
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester
| | | | - Beth L Larrabee
- Mayo Clinic Division of Biostatistics, Mayo Clinic, Rochester, Minnesota
| | | | | | - Daniel J Schaid
- Mayo Clinic Division of Biostatistics, Mayo Clinic, Rochester, Minnesota
| | | |
Collapse
|
57
|
Abstract
Wilms tumour is the most common renal malignancy of childhood. The disease is curable in the majority of cases, albeit at considerable cost in terms of late treatment-related effects in some children. However, one in ten children with Wilms tumour will die of their disease despite modern treatment approaches. The genetic changes that underpin Wilms tumour have been defined by studies of familial cases and by unbiased DNA sequencing of tumour genomes. Together, these approaches have defined the landscape of cancer genes that are operative in Wilms tumour, many of which are intricately linked to the control of fetal nephrogenesis. Advances in our understanding of the germline and somatic genetic changes that underlie Wilms tumour may translate into better patient outcomes. Improvements in risk stratification have already been seen through the introduction of molecular biomarkers into clinical practice. A host of additional biomarkers are due to undergo clinical validation. Identifying actionable mutations has led to potential new targets, with some novel compounds undergoing testing in early phase trials. Avenues that warrant further exploration include targeting Wilms tumour cancer genes with a non-redundant role in nephrogenesis and targeting the fetal renal transcriptome.
Collapse
Affiliation(s)
- Taryn Dora Treger
- Wellcome Sanger Institute, Cambridge, UK
- Department of Paediatrics, University of Cambridge, Cambridge, UK
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Tanzina Chowdhury
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Kathy Pritchard-Jones
- UCL Great Ormond Street Institute of Child Health, London, UK.
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK.
| | - Sam Behjati
- Wellcome Sanger Institute, Cambridge, UK
- Department of Paediatrics, University of Cambridge, Cambridge, UK
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| |
Collapse
|
58
|
Genetic testing in steroid-resistant nephrotic syndrome: why, who, when and how? Pediatr Nephrol 2019; 34:195-210. [PMID: 29181713 PMCID: PMC6311200 DOI: 10.1007/s00467-017-3838-6] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 10/24/2017] [Accepted: 10/25/2017] [Indexed: 12/23/2022]
Abstract
Steroid-resistant nephrotic syndrome (SRNS) is a common cause of chronic kidney disease in childhood and has a significant risk of rapid progression to end-stage renal disease. The identification of over 50 monogenic causes of SRNS has revealed dysfunction in podocyte-associated proteins in the pathogenesis of proteinuria, highlighting their essential role in glomerular function. Recent technological advances in high-throughput sequencing have enabled indication-driven genetic panel testing for patients with SRNS. The availability of genetic testing, combined with the significant phenotypic variability of monogenic SRNS, poses unique challenges for clinicians when directing genetic testing. This highlights the need for clear clinical guidelines that provide a systematic approach for mutational screening in SRNS. The likelihood of identifying a causative mutation is inversely related to age at disease onset and is increased with a positive family history or the presence of extra-renal manifestations. An unequivocal molecular diagnosis could allow for a personalised treatment approach with weaning of immunosuppressive therapy, avoidance of renal biopsy and provision of accurate, well-informed genetic counselling. Identification of novel causative mutations will continue to unravel the pathogenic mechanisms of glomerular disease and provide new insights into podocyte biology and glomerular function.
Collapse
|
59
|
Abstract
Zusammenfassung
Das steroid-resistente nephrotische Syndrom (SRNS) mit dem histomorphologischen Korrelat der fokal-segmentalen Glomerulosklerose (FSGS) stellt eine bedeutende Ursache für eine terminale Niereninsuffizienz im Kindesalter, aber auch bei erwachsenen Patienten dar. Das Erkrankungsspektrum zeichnet sich durch eine große genetische Heterogenität aus, wobei auch nicht genetische Ursachen bei der FSGS beobachtet werden. Die genetische Grundlage des SRNS/FSGS-Komplexes ist v. a. für ältere Kinder/Jugendliche und Erwachsene bisher noch unzureichend verstanden. Die eindeutige Abgrenzung genetischer SRNS/FSGS-Ursachen ist unerlässlich, da sich bereits heute hieraus eine Vielzahl an klinischen Implikationen ergeben. Die Identifikation unbekannter Erkrankungsallele oder Erkrankungsgene kann zudem Erkenntnisse bringen, die ein gänzlich neues Verständnis der Pathomechanismen ermöglichen. Durch umfassende genetische Untersuchungen besteht die Möglichkeit, die ungelöste genetische Basis der Rekurrenz der FSGS-Erkrankung bei bislang Varianten-negativen Patienten zu finden.
Collapse
Affiliation(s)
- Julia Hoefele
- Aff1 Institut für Humangenetik Klinikum rechts der Isar, Technische Universität München Trogerstr. 32 81675 München Deutschland
| | - Bodo B. Beck
- Aff2 0000 0000 8852 305X grid.411097.a Institut für Humangenetik Uniklinik Köln Kerpener Str. 34 50937 Köln Deutschland
| | - Lutz T. Weber
- Aff3 0000 0000 8852 305X grid.411097.a Klinik und Poliklinik für Kinder- und Jugendmedizin Uniklinik Köln Kerpener Str. 62 50937 Köln Deutschland
| | - Paul Brinkkötter
- Aff4 0000 0000 8852 305X grid.411097.a Klinik II für Innere Medizin Uniklinik Köln Kerpener Str. 62 50937 Köln Deutschland
| |
Collapse
|
60
|
Management of Denys-Drash syndrome: A case series based on an international survey. Clin Nephrol Case Stud 2018; 6:36-44. [PMID: 30450273 PMCID: PMC6236398 DOI: 10.5414/cncs109515] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 09/12/2018] [Indexed: 01/16/2023] Open
Abstract
Denys-Drash syndrome (DDS), a condition caused by mutations in the tumor-suppressor gene WT-1, is associated with a triad of disorders: ambiguous genitalia, nephrotic syndrome leading to end-stage renal disease (ESRD), and Wilms’ tumor. Given the variable disease course, management is challenging. We aimed to describe the evolution of DDS and the range of management strategies by summarizing the clinical courses of cases collected from a questionnaire sent to the international pediatric nephrology community. 15 respondents provided information on 23 patients; 21 DDS cases were confirmed and analyzed. At DDS diagnosis, 6 patients had a Wilms’ tumor (group A) and 15 had no Wilms’ tumor (group B). Three group A patients had unilateral nephrectomy. Two of these still had renal function, with no second tumor, at 36 months and 16 years of age, and 1 progressed to ESRD. Three had bilateral nephrectomy before ESRD. Eight group B patients progressed to ESRD, all of whom later had all renal tissue removed. Two group B patients subsequently developed a unilateral Wilms’ tumor and had bilateral nephrectomy pre-ESRD. Three had bilateral nephrectomy prior to reaching ESRD without ever having a Wilms’ tumor. Two group B patients remained tumor-free with renal function at last follow-up. Two main management approaches were taken: pre-emptive nephrectomy prior to ESRD and conservative surveillance. Based on the known risks associated with ESRD in infants and young children, the variable course of DDS, and the relatively good prognosis associated with Wilms’ tumor, a guiding principle of preservation of renal function is most logical. Most would advocate bilateral prophylactic nephrectomy after ESRD is reached due to the high tumor risk, which is likely heightened after transplant.
Collapse
|
61
|
Shimizu N, Matsuda M. Identification of a Novel Zebrafish Mutant Line that Develops Testicular Germ Cell Tumors. Zebrafish 2018; 16:15-28. [PMID: 30300574 DOI: 10.1089/zeb.2018.1604] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Testicular tumors are the most common solid malignant tumors in men 20-35 years of age. Although most of testicular tumors are curable, current treatments still fail in 15%-20% of patients. However, insufficient understanding of the molecular basis and lack of animal models limit development of more effective treatments. This study reports the identification of a novel zebrafish mutant line, ns1402, which develops testicular germ cell tumors (TGCTs). While both male and female ns1402 mutants were fertile at young age, male ns1402 mutants became infertile as early as 9 months of age. This infertility was associated with progressive loss of mature sperm. Failure of spermatogenesis was, at least in part, explained by progressive loss of mature Leydig cells, a source of testosterone that is essential for spermatogenesis. Interestingly, TGCTs in ns1402 mutants contained a large number of Sertoli cells and gene expression profiles of Sertoli cells were altered before loss of mature Leydig cells. This suggests that changes in Sertoli cell properties happened first, followed by loss of mature Leydig cells and failure of spermatogenesis. Taken together, this study emphasizes the importance of cell-cell interactions and cell signaling in the testis for spermatogenesis and tissue homeostasis.
Collapse
Affiliation(s)
- Nobuyuki Shimizu
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey
| | - Miho Matsuda
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey
| |
Collapse
|
62
|
Murphy AJ, Davidoff AM. Bilateral Wilms Tumor: A Surgical Perspective. CHILDREN-BASEL 2018; 5:children5100134. [PMID: 30250006 PMCID: PMC6210093 DOI: 10.3390/children5100134] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 09/13/2018] [Accepted: 09/21/2018] [Indexed: 12/27/2022]
Abstract
Historically, the management of bilateral Wilms tumor (BWT) was non-standardized and suffered from instances of prolonged chemotherapy and inconsistent surgical management which resulted in suboptimal renal and oncologic outcomes. Because of the risk of end-stage renal disease associated with the management of BWT, neoadjuvant chemotherapy and nephron-sparing surgery have been adopted as the guiding management principles. This management strategy balances acceptable oncologic outcomes against the risk of end-stage renal disease. A recent multi-institutional Children’s Oncology Group study (AREN0534) has confirmed the benefits of standardized 3-drug neoadjuvant chemotherapy and the utilization of nephron-sparing surgery in BWT patients; however, less than 50% of patients underwent bilateral nephron-sparing surgery. The coordination of neoadjuvant chemotherapy and the timing and implementation of bilateral nephron-sparing surgery are features of BWT management that require collaboration between oncologists and surgeons. This review discusses the surgical management strategy in the context of BWT disease biology, with an emphasis on timepoints during therapy at which surgical decision making can greatly impact this disease and minimize long-term toxicities.
Collapse
Affiliation(s)
- Andrew J Murphy
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
- Division of Pediatric Surgery, Department of Surgery, University of Tennessee Health Science Center, Memphis, TN 38105, USA.
| | - Andrew M Davidoff
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
- Division of Pediatric Surgery, Department of Surgery, University of Tennessee Health Science Center, Memphis, TN 38105, USA.
| |
Collapse
|
63
|
The genetic architecture of aniridia and Gillespie syndrome. Hum Genet 2018; 138:881-898. [PMID: 30242502 PMCID: PMC6710220 DOI: 10.1007/s00439-018-1934-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Accepted: 09/06/2018] [Indexed: 12/13/2022]
Abstract
Absence of part or all of the iris, aniridia, is a feature of several genetically distinct conditions. This review focuses on iris development and then the clinical features and molecular genetics of these iris malformations. Classical aniridia, a panocular eye malformation including foveal hypoplasia, is the archetypal phenotype associated with heterozygous PAX6 loss-of-function mutations. Since this was identified in 1991, many genetic mechanisms of PAX6 inactivation have been elucidated, the commonest alleles being intragenic mutations causing premature stop codons, followed by those causing C-terminal extensions. Rarely, aniridia cases are associated with FOXC1, PITX2 and/or their regulatory regions. Aniridia can also occur as a component of many severe global eye malformations. Gillespie syndrome—a triad of partial aniridia, non-progressive cerebellar ataxia and intellectual disability—is phenotypically and genotypically distinct from classical aniridia. The causative gene has recently been identified as ITPR1. The same characteristic Gillespie syndrome-like iris, with aplasia of the pupillary sphincter and a scalloped margin, is seen in ACTA2-related multisystemic smooth muscle dysfunction syndrome. WAGR syndrome (Wilms tumour, aniridia, genitourinary anomalies and mental retardation/intellectual disability), is caused by contiguous deletion of PAX6 and WT1 on chromosome 11p. Deletions encompassing BDNF have been causally implicated in the obesity and intellectual disability associated with the condition. Lastly, we outline a genetic investigation strategy for aniridia in light of recent developments, suggesting an approach based principally on chromosomal array and gene panel testing. This strategy aims to test all known aniridia loci—including the rarer, life-limiting causes—whilst remaining simple and practical.
Collapse
|
64
|
Abstract
PURPOSE OF REVIEW The current review focuses on the neonatal presentation of disorders of sex development, summarize the current approach to the evaluation of newborns and describes recent advances in understanding of underlying genetic aetiology of these conditions. RECENT FINDINGS Several possible candidate genes as well as other adverse environmental factors have been described as contributing to several clinical subgroups of 46,XY DSDs. Moreover, registry-based studies showed that infants with suspected DSD may have extragenital anomalies and in 46,XY cases, being small for gestational age (SGA), cardiac and neurological malformations are the commonest concomitant conditions. SUMMARY Considering that children and adults with DSD may be at risk of several comorbidities a clear aetiological diagnosis will guide further management. To date, a firm diagnosis is not reached in over half of the cases of 46,XY DSD. Whilst it is likely that improved diagnostic resources will bridge this gap in the future, the next challenge to the clinical community will be to show that such advances will result in an improvement in clinical care.
Collapse
|
65
|
Hodhod A, El-Sherbiny M. 46-XY Denys-Drash Syndrome. Is There a Role for Nephron-sparing Modalities in Management of Renal Masses? A Report of 2 Cases. Urology 2018; 117:153-155. [PMID: 29723589 DOI: 10.1016/j.urology.2018.04.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 04/14/2018] [Accepted: 04/24/2018] [Indexed: 11/18/2022]
Abstract
Denys-Drash syndrome (DDS) is a combination of genital and urinary anomalies that are mostly associated with renal and gonadal malignancies. We report 2 patients who presented with genital ambiguity and were diagnosed as 46-XY DDS. Both patients had renal masses during follow-up and underwent partial nephrectomy aiming to have transplant at older age.
Collapse
Affiliation(s)
- Amr Hodhod
- Department of Pediatric Surgery, McGill University, The Montreal Children's Hospital, Montréal, Quebec, Canada.
| | - Mohamed El-Sherbiny
- Department of Pediatric Surgery, McGill University, The Montreal Children's Hospital, Montréal, Quebec, Canada
| |
Collapse
|
66
|
Kalish JM, Doros L, Helman LJ, Hennekam RC, Kuiper RP, Maas SM, Maher ER, Nichols KE, Plon SE, Porter CC, Rednam S, Schultz KAP, States LJ, Tomlinson GE, Zelley K, Druley TE. Surveillance Recommendations for Children with Overgrowth Syndromes and Predisposition to Wilms Tumors and Hepatoblastoma. Clin Cancer Res 2018; 23:e115-e122. [PMID: 28674120 DOI: 10.1158/1078-0432.ccr-17-0710] [Citation(s) in RCA: 129] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Revised: 04/23/2017] [Accepted: 05/09/2017] [Indexed: 12/13/2022]
Abstract
A number of genetic syndromes have been linked to increased risk for Wilms tumor (WT), hepatoblastoma (HB), and other embryonal tumors. Here, we outline these rare syndromes with at least a 1% risk to develop these tumors and recommend uniform tumor screening recommendations for North America. Specifically, for syndromes with increased risk for WT, we recommend renal ultrasounds every 3 months from birth (or the time of diagnosis) through the seventh birthday. For HB, we recommend screening with full abdominal ultrasound and alpha-fetoprotein serum measurements every 3 months from birth (or the time of diagnosis) through the fourth birthday. We recommend that when possible, these patients be evaluated and monitored by cancer predisposition specialists. At this time, these recommendations are not based on the differential risk between different genetic or epigenetic causes for each syndrome, which some European centers have implemented. This differentiated approach largely represents distinct practice environments between the United States and Europe, and these guidelines are designed to be a broad framework within which physicians and families can work together to implement specific screening. Further study is expected to lead to modifications of these recommendations. Clin Cancer Res; 23(13); e115-e22. ©2017 AACRSee all articles in the online-only CCR Pediatric Oncology Series.
Collapse
Affiliation(s)
- Jennifer M Kalish
- Division of Human Genetics, Children's Hospital of Philadelphia and the Department of Pediatrics at the Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| | - Leslie Doros
- Cancer Genetics Clinic, Children's National Medical Center, Washington, DC
| | - Lee J Helman
- Center for Cancer Research and Pediatric Oncology Branch, National Cancer Institute, Rockville, Maryland
| | - Raoul C Hennekam
- Department of Pediatrics, University of Amsterdam, Amsterdam, the Netherlands
| | - Roland P Kuiper
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Saskia M Maas
- Department of Clinical Genetics, Academic Medical Center, Amsterdam, the Netherlands
| | - Eamonn R Maher
- Department of Medical Genetics, University of Cambridge, and Cambridge NIHR Biomedical Research Centre, Cambridge, United Kingdom
| | - Kim E Nichols
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Sharon E Plon
- Department of Pediatrics/Hematology-Oncology, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas
| | | | - Surya Rednam
- Department of Pediatrics/Hematology-Oncology, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas
| | - Kris Ann P Schultz
- Division of Cancer and Blood Disorders, Children's Hospitals and Clinics of Minnesota, Minneapolis, Minnesota
| | - Lisa J States
- Division of Radiology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Gail E Tomlinson
- Division of Pediatric Hematology-Oncology and Greehey Children's Cancer Research Institute, The University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Kristin Zelley
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Todd E Druley
- Division of Pediatric Hematology and Oncology, Washington University, St. Louis, Missouri
| |
Collapse
|
67
|
Heeley JM, Hollander AS, Austin PF, Merritt DF, Wesevich VG, Amarillo IE. Risk association of congenital anomalies in patients with ambiguous genitalia: A 22-year single-center experience. J Pediatr Urol 2018; 14:153.e1-153.e7. [PMID: 29157626 DOI: 10.1016/j.jpurol.2017.09.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 09/22/2017] [Indexed: 11/19/2022]
Abstract
BACKGROUND Ambiguous genitalia refers to a form of differences of sex development (DSD) wherein the appearance of the external genitalia is atypical. This rare condition presents challenges in decision-making and clinical management. Review of historical data may reveal areas for clinical research to improve care for patients with ambiguous genitalia. OBJECTIVE This chart review was performed to identify patients with ambiguous genitalia, and to classify them as having 46,XX DSD, 46,XY DSD, or sex chromosome DSD. Within these categories, we looked at establishment of specific diagnoses, type and frequency of other congenital anomalies and neoplasms, and gender assignment, as well as incidence of gender reassignment and transition. METHODS We performed a retrospective chart review of patients diagnosed with DSD conditions from 1995 to 2016 using ICD9 codes. For the purpose of this study, review was limited to individuals assessed to have neonatal "ambiguous genitalia" or "indeterminate sex." RESULTS Review identified 128 patients evaluated for ambiguous genitalia from 22 years of experience (Figure). Approximately half of these (53%) had 46,XY karyotype, 35% had 46,XX, and the remaining 12% had sex chromosome aberrations. Diagnostic rate for 46,XX DSD was higher at 64%, all of which were congenital adrenal hyperplasia, while diagnostic rate for 46,XY DSD was 11.7% for a molecularly confirmed diagnosis and 24% if clinical diagnoses were included. The most common anomalies included cardiac anomalies in 28/128 (22%), skeletal anomalies in 19/128 (15%), and failure to thrive or growth problems in 19/128 (15%). Additional congenital anomalies were found in 53 out of 128 patients (41%). There were three reported neoplasms in this group: gonadoblastoma, hepatoblastoma, and myelodysplastic syndrome with monosomy 7. Gender assignment was consistent with chromosomes in approximately 90% of XX and XY patients. There were three recorded gender reassignments or transitions. DISCUSSION Diagnostic rate for ambiguous genitalia is low, especially in 46,XY DSD. Most neonates were assigned gender consistent with their chromosomes. Given the high rate of associated anomalies, screening for cardiac or other anomalies in patients with ambiguous genitalia may be beneficial. CONCLUSION Patients with ambiguous genitalia often have additional congenital anomalies. Establishment of a specific diagnosis is uncommon in 46,XY patients. A few patients have gender reassignment outside of the newborn period. Ongoing collection of clinical data on this population may reveal new information regarding long-term health, quality of life, and establishment of more diagnoses with improved molecular techniques.
Collapse
Affiliation(s)
- Jennifer M Heeley
- Division of Genetics and Genomics, Department of Pediatrics, Washington University School of Medicine in St. Louis, St Louis, MO, USA
| | - Abby S Hollander
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, Washington University School of Medicine in St. Louis, St Louis, MO, USA
| | - Paul F Austin
- Division of Urology, Department of Surgery, Washington University School of Medicine in St. Louis, St Louis, MO, USA
| | - Diane F Merritt
- Division of Pediatric and Adolescent Gynecology, Department of Obstetrics and Gynecology, Washington University School of Medicine in St. Louis, St Louis, MO, USA
| | | | - Ina E Amarillo
- Division of Laboratory and Genomic Medicine, Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, St Louis, MO, USA.
| |
Collapse
|
68
|
Asfahani RI, Tahoun MM, Miller-Hodges EV, Bellerby J, Virasami AK, Sampson RD, Moulding D, Sebire NJ, Hohenstein P, Scambler PJ, Waters AM. Activation of podocyte Notch mediates early Wt1 glomerulopathy. Kidney Int 2018; 93:903-920. [PMID: 29398135 PMCID: PMC6169130 DOI: 10.1016/j.kint.2017.11.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Revised: 11/07/2017] [Accepted: 11/09/2017] [Indexed: 01/26/2023]
Abstract
The Wilms' tumor suppressor gene, WT1, encodes a zinc finger protein that regulates podocyte development and is highly expressed in mature podocytes. Mutations in the WT1 gene are associated with the development of renal failure due to the formation of scar tissue within glomeruli, the mechanisms of which are poorly understood. Here, we used a tamoxifen-based CRE-LoxP system to induce deletion of Wt1 in adult mice to investigate the mechanisms underlying evolution of glomerulosclerosis. Podocyte apoptosis was evident as early as the fourth day post-induction and increased during disease progression, supporting a role for Wt1 in mature podocyte survival. Podocyte Notch activation was evident at disease onset with upregulation of Notch1 and its transcriptional targets, including Nrarp. There was repression of podocyte FoxC2 and upregulation of Hey2 supporting a role for a Wt1/FoxC2/Notch transcriptional network in mature podocyte injury. The expression of cleaved Notch1 and HES1 proteins in podocytes of mutant mice was confirmed in early disease. Furthermore, induction of podocyte HES1 expression was associated with upregulation of genes implicated in epithelial mesenchymal transition, thereby suggesting that HES1 mediates podocyte EMT. Lastly, early pharmacological inhibition of Notch signaling ameliorated glomerular scarring and albuminuria. Thus, loss of Wt1 in mature podocytes modulates podocyte Notch activation, which could mediate early events in WT1-related glomerulosclerosis.
Collapse
Affiliation(s)
- Rowan I Asfahani
- Programme of Developmental Biology of Birth Defects, Great Ormond Street Institute of Child Health, University College of London, London, UK
| | - Mona M Tahoun
- Programme of Developmental Biology of Birth Defects, Great Ormond Street Institute of Child Health, University College of London, London, UK; Clinical and Chemical Pathology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Eve V Miller-Hodges
- MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland
| | - Jack Bellerby
- Programme of Developmental Biology of Birth Defects, Great Ormond Street Institute of Child Health, University College of London, London, UK
| | - Alex K Virasami
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Robert D Sampson
- Institute of Ophthalmology, University College of London, London, UK
| | - Dale Moulding
- Programme of Developmental Biology of Birth Defects, Great Ormond Street Institute of Child Health, University College of London, London, UK
| | - Neil J Sebire
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | | | - Peter J Scambler
- Programme of Developmental Biology of Birth Defects, Great Ormond Street Institute of Child Health, University College of London, London, UK
| | - Aoife M Waters
- Programme of Developmental Biology of Birth Defects, Great Ormond Street Institute of Child Health, University College of London, London, UK; Great Ormond Street Hospital NHS Foundation Trust, London, UK.
| |
Collapse
|
69
|
Ramsawhook A, Ruzov A, Coyle B. Wilms' Tumor Protein 1 and Enzymatic Oxidation of 5-Methylcytosine in Brain Tumors: Potential Perspectives. Front Cell Dev Biol 2018; 6:26. [PMID: 29623275 PMCID: PMC5874295 DOI: 10.3389/fcell.2018.00026] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 03/05/2018] [Indexed: 12/24/2022] Open
Abstract
The patterns of 5-methylcytosine (5mC) and its oxidized derivatives, 5-hydroxymethylcytosine, 5-formylcytosine, and 5-carboxylcytosine (5caC) are reportedly altered in a range of cancers. Likewise, Wilms' Tumor protein 1 (WT1), a transcription factor essential for urogenital, epicardium, and kidney development exhibits aberrant expression in multiple tumors. Interestingly, WT1 directly interacts with TET proteins that catalyze the enzymatic oxidation of 5mC and exhibits high affinity for 5caC-containing DNA substrates in vitro. Here we review recent developments in the fields of Tet-dependent 5mC oxidation and WT1 biology and explore potential perspectives for studying the interplay between TETs and WT1 in brain tumors.
Collapse
Affiliation(s)
- Ashley Ramsawhook
- Wolfson Centre for Stem Cells, Tissue Engineering and Modelling (STEM), Centre for Biomolecular Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Alexey Ruzov
- Wolfson Centre for Stem Cells, Tissue Engineering and Modelling (STEM), Centre for Biomolecular Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Beth Coyle
- Children's Brain Tumour Research Centre, Medical School, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
70
|
Goudie C, Witkowski L, Vairy S, McCluggage WG, Foulkes WD. Paediatric ovarian tumours and their associated cancer susceptibility syndromes. J Med Genet 2017; 55:1-10. [DOI: 10.1136/jmedgenet-2017-104926] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 09/26/2017] [Accepted: 10/28/2017] [Indexed: 01/08/2023]
Abstract
Non-epithelial ovarian tumours are rare neoplasms that occasionally arise in childhood and adolescence. They can be associated with various cancer susceptibility syndromes. The morphological overlap seen across these tumours and their rarity can make the diagnosis challenging. In the case of an incorrect diagnosis, the underlying genetic susceptibility may be missed. In this review, we outline the genetic background of ovarian non-epithelial tumours arising in children, emphasizing the genes harbouring pathogenic germline variants associated with each tumour type. Specifically, juvenile granulosa cell tumours, Sertoli-Leydig cell tumours, sex cord tumours with annular tubules, Sertoli cell tumours, germ cell tumours and small cell carcinoma of the ovary of hypercalcaemic type are discussed in this review. For each tumour type, we detail the personal and family history features and the presenting characteristics of the ovarian tumour as well as the pathological features and molecular markers that point towards a cancer predisposition syndrome. Throughout, we stress the need for specialised pathological review in difficult cases.
Collapse
|
71
|
Kirschner KM, Sciesielski LK, Krueger K, Scholz H. Wilms tumor protein-dependent transcription of VEGF receptor 2 and hypoxia regulate expression of the testis-promoting gene Sox9 in murine embryonic gonads. J Biol Chem 2017; 292:20281-20291. [PMID: 29042436 DOI: 10.1074/jbc.m117.816751] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 10/11/2017] [Indexed: 01/24/2023] Open
Abstract
Wilms tumor protein 1 (WT1) has been implicated in the control of several genes in sexual development, but its function in gonad formation is still unclear. Here, we report that WT1 stimulates expression of Kdr, the gene encoding VEGF receptor 2, in murine embryonic gonads. We found that WT1 and KDR are co-expressed in Sertoli cells of the testes and somatic cells of embryonic ovaries. Vivo-morpholino-mediated WT1 knockdown decreased Kdr transcripts in cultured embryonic gonads at multiple developmental stages. Furthermore, WT1 bound to the Kdr promoter in the chromatin of embryonic testes and ovaries. Forced expression of the WT1(-KTS) isoform, which functions as a transcription factor, increased KDR mRNA levels, whereas the WT1(+KTS) isoform, which acts presumably on the post-transcriptional level, did not. ChIP indicated that WT1(-KTS), but not WT1(+KTS), binds to the KDR promoter. Treatment with the KDR tyrosine kinase inhibitor SU1498 or the KDR ligand VEGFA revealed that KDR signaling represses the testis-promoting gene Sox9 in embryonic XX gonads. WT1 knockdown abrogated the stimulatory effect of SU1498-mediated KDR inhibition on Sox9 expression. Exposure to 1% O2 to mimic the low-oxygen conditions in the embryo increased Vegfa expression but did not affect Sox9 mRNA levels in gonadal explants. However, incubation in 1% O2 in the presence of SU1498 significantly reduced Sox9 transcripts in cultured testes and increased Sox9 levels in ovaries. These findings demonstrate that both the local oxygen environment and WT1, which enhances KDR expression, contribute to sex-specific Sox9 expression in developing murine gonads.
Collapse
Affiliation(s)
| | - Lina K Sciesielski
- Klinik für Neonatologie, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | | | - Holger Scholz
- Institut für Vegetative Physiologie, 10117 Berlin, Germany.
| |
Collapse
|
72
|
Abstract
The study of genes mutated in human disease often leads to new insights into biology as well as disease mechanisms. One such gene is Wilms' tumour 1 (WT1), which plays multiple roles in development, tissue homeostasis and disease. In this Primer, I summarise how this multifaceted gene functions in various mammalian tissues and organs, including the kidney, gonads, heart and nervous system. This is followed by a discussion of our current understanding of the molecular mechanisms by which WT1 and its two major isoforms regulate these processes at the transcriptional and post-transcriptional levels.
Collapse
Affiliation(s)
- Nicholas D Hastie
- MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road (S), Edinburgh, EH4 2XU, UK
| |
Collapse
|
73
|
Kaverina NV, Eng DG, Largent AD, Daehn I, Chang A, Gross KW, Pippin JW, Hohenstein P, Shankland SJ. WT1 Is Necessary for the Proliferation and Migration of Cells of Renin Lineage Following Kidney Podocyte Depletion. Stem Cell Reports 2017; 9:1152-1166. [PMID: 28966119 PMCID: PMC5639431 DOI: 10.1016/j.stemcr.2017.08.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 08/25/2017] [Accepted: 08/28/2017] [Indexed: 12/17/2022] Open
Abstract
Wilms' tumor suppressor 1 (WT1) plays an important role in cell proliferation and mesenchymal-epithelial balance in normal development and disease. Here, we show that following podocyte depletion in three experimental models, and in patients with focal segmental glomerulosclerosis (FSGS) and membranous nephropathy, WT1 increased significantly in cells of renin lineage (CoRL). In an animal model of FSGS in RenWt1fl/fl reporter mice with inducible deletion of WT1 in CoRL, CoRL proliferation and migration to the glomerulus was reduced, and glomerular disease was worse compared with wild-type mice. To become podocytes, CoRL undergo mesenchymal-to-epithelial transformation (MET), typified by reduced staining for mesenchymal markers (MYH11, SM22, αSMA) and de novo expression of epithelial markers (E-cadherin and cytokeratin18). Evidence for changes in MET markers was barely detected in RenWt1fl/fl mice. Our results show that following podocyte depletion, WT1 plays essential roles in CoRL proliferation and migration toward an adult podocyte fate.
Collapse
Affiliation(s)
- Natalya V Kaverina
- Division of Nephrology, University of Washington School of Medicine, 750 Republican Street, Seattle, WA 98109, USA
| | - Diana G Eng
- Division of Nephrology, University of Washington School of Medicine, 750 Republican Street, Seattle, WA 98109, USA
| | - Andrea D Largent
- Division of Nephrology, University of Washington School of Medicine, 750 Republican Street, Seattle, WA 98109, USA
| | - Ilse Daehn
- Department of Medicine, Division of Nephrology, The Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, New York, NY 10029, USA
| | - Anthony Chang
- Department of Pathology, University of Chicago, 5841 S Maryland Ave, Chicago, IL 60637, USA
| | - Kenneth W Gross
- Department of Molecular and Cellular Biology, Roswell Park Cancer Institute, Elm & Carlton Streets, Buffalo, NY 14263, USA
| | - Jeffrey W Pippin
- Division of Nephrology, University of Washington School of Medicine, 750 Republican Street, Seattle, WA 98109, USA
| | - Peter Hohenstein
- The Roslin Institute, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK
| | - Stuart J Shankland
- Division of Nephrology, University of Washington School of Medicine, 750 Republican Street, Seattle, WA 98109, USA.
| |
Collapse
|
74
|
Eid W, Biason-Lauber A. Why boys will be boys and girls will be girls: Human sex development and its defects. ACTA ACUST UNITED AC 2017; 108:365-379. [PMID: 28033664 DOI: 10.1002/bdrc.21143] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Among the most defining events of an individual's life, is the development of a human embryo into male or a female. The phenotypic sex of an individual depends on the type of gonad that develops in the embryo, a process which itself is determined by the genetic setting of the individual. The development of the gonads is different from any other organ, as they possess the potential to differentiate into two functionally distinct organs, testes, or ovaries. Sex development can be divided into two distinctive processes, "sex determination," which is the commitment of the undifferentiated gonad into either a testis or an ovary, a process that is genetically programmed in a critically timed manner and "sex differentiation," which takes place through hormones produced by the gonads, once the developmental sex determination decision has been made. Disruption of any of the genes involved in either the testicular or ovarian development pathway could lead to disorders of sex development. In this review, we provide an insight into the factors important for sex determination, their antagonistic actions and whenever possible, references on the "prismatic" clinical cases are given. Birth Defects Research (Part C) 108:365-379, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Wassim Eid
- Division of Endocrinology, Department of Medicine, University of Fribourg, Fribourg, Switzerland.,Department of Biochemistry, Medical Research Institute, University of Alexandria, Alexandria, Egypt
| | - Anna Biason-Lauber
- Division of Endocrinology, Department of Medicine, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
75
|
Wen L, Yuan Q, Sun M, Niu M, Wang H, Fu H, Zhou F, Yao C, Wang X, Li Z, He Z. Generation and characteristics of human Sertoli cell line immortalized by overexpression of human telomerase. Oncotarget 2017; 8:16553-16570. [PMID: 28152522 PMCID: PMC5369984 DOI: 10.18632/oncotarget.14985] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 01/24/2017] [Indexed: 12/25/2022] Open
Abstract
Sertoli cells are required for normal spermatogenesis and they can be reprogrammed to other types of functional cells. However, the number of primary Sertoli cells is rare and human Sertoli cell line is unavailable. In this study, we have for the first time reported a stable human Sertoli cell line, namely hS1 cells, by overexpression of human telomerase. The hS1 cells expressed a number of hallmarks for human Sertoli cells, including SOX9, WT1, GDNF, SCF, BMP4, BMP6, GATA4, and VIM, and they were negative for 3β-HSD, SMA, and VASA. Higher levels of AR and FSHR were observed in hS1 cells compared to primary human Sertoli cells. Microarray analysis showed that 70.4% of global gene profiles of hS1 cells were similar to primary human Sertoli cells. Proliferation assay demonstrated that hS1 cells proliferated rapidly and they could be passaged for more than 30 times in 6 months. Neither Y chromosome microdeletion nor tumorgenesis was detected in this cell line and 90% normal karyotypes existed in hS1 cells. Collectively, we have established the first human Sertoli cell line with phenotype of primary human Sertoli cells, an unlimited proliferation potential and high safety, which could offer sufficient human Sertoli cells for basic research as well as reproductive and regenerative medicine.
Collapse
Affiliation(s)
- Liping Wen
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Qingqing Yuan
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Min Sun
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Minghui Niu
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Hong Wang
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Hongyong Fu
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Fan Zhou
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Chencheng Yao
- Department of Andrology, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai 200080, China
| | - Xiaobo Wang
- Department of Andrology, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai 200080, China
| | - Zheng Li
- Department of Andrology, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai 200080, China
| | - Zuping He
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.,Shanghai Institute of Andrology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200001, China.,Shanghai Key Laboratory of Assisted Reproduction and Reproductive Genetics, Shanghai 200127, China.,Shanghai Key Laboratory of Reproductive Medicine, Shanghai 200025, China
| |
Collapse
|
76
|
Alge JL, Wenderfer SE, Hicks J, Bekheirnia MR, Schady DA, Kain JS, Braun MC. Hemolytic uremic syndrome as the presenting manifestation of WT1 mutation and Denys-Drash syndrome: a case report. BMC Nephrol 2017; 18:243. [PMID: 28720077 PMCID: PMC5516385 DOI: 10.1186/s12882-017-0643-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Accepted: 06/27/2017] [Indexed: 11/30/2022] Open
Abstract
Background Hemolytic uremic syndrome (HUS) can occur as a primary process due to mutations in complement genes or secondary to another underlying disease. HUS sometimes occurs in the setting of glomerular diseases, and it has been described in association with Denys-Drash syndrome (DDS), which is characterized by the triad of abnormal genitourinary development; a pathognomonic glomerulopathy, diffuse mesangial sclerosis; and the development of Wilms tumor. Case presentation We report the case of a 46, XX female infant who presented with HUS and biopsy-proven thrombotic microangiopathy. Next generation sequencing of genes with known mutations causative of atypical HUS found that she was homozygous for the Complement Factor H H3 haplotype and heterozygous for a variant of unknown significance in the DGKE gene. Whole exome sequencing identified a de novo heterozygous WT1 c.1384C > T; p.R394W mutation, which is classically associated with Denys-Drash syndrome (DDS). At the time of bilateral nephrectomy five months after her initial biopsy, she had diffuse mesangial sclerosis, typical of Denys-Drash syndrome, without evidence of thrombotic microangiopathy. Conclusion This unique case highlights HUS as a rare but important manifestation of WT1 mutation and provides new insight into the genetics underlying this association.
Collapse
Affiliation(s)
- Joseph L Alge
- Baylor College of Medicine, Department of Pediatrics, Pediatrician-Scientist Training and Development Program, Houston, TX, USA.
| | - Scott E Wenderfer
- Baylor College of Medicine, Department of Pediatrics, Renal Section, Houston, TX, USA.,Texas Children's Hospital, Houston, TX, USA
| | - John Hicks
- Baylor College of Medicine, Department of Pathology and Immunology, Houston, TX, USA.,Texas Children's Hospital, Houston, TX, USA
| | - Mir Reza Bekheirnia
- Baylor College of Medicine, Department of Molecular and Human Genetics, Houston, 77030, USA
| | - Deborah A Schady
- Baylor College of Medicine, Department of Pathology and Immunology, Houston, TX, USA.,Texas Children's Hospital, Houston, TX, USA
| | | | - Michael C Braun
- Baylor College of Medicine, Department of Pediatrics, Renal Section, Houston, TX, USA.,Texas Children's Hospital, Houston, TX, USA
| |
Collapse
|
77
|
Park E, Ahn YH, Kang HG, Yoo KH, Won NH, Lee KB, Moon KC, Seong MW, Gwon TR, Park SS, Cheong HI. COQ6 Mutations in Children With Steroid-Resistant Focal Segmental Glomerulosclerosis and Sensorineural Hearing Loss. Am J Kidney Dis 2017; 70:139-144. [DOI: 10.1053/j.ajkd.2016.10.040] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 10/15/2016] [Indexed: 01/15/2023]
|
78
|
Wang F, Zhang Y, Mao J, Yu Z, Yi Z, Yu L, Sun J, Wei X, Ding F, Zhang H, Xiao H, Yao Y, Tan W, Lovric S, Ding J, Hildebrandt F. Spectrum of mutations in Chinese children with steroid-resistant nephrotic syndrome. Pediatr Nephrol 2017; 32:1181-1192. [PMID: 28204945 PMCID: PMC5478193 DOI: 10.1007/s00467-017-3590-y] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 12/06/2016] [Accepted: 12/23/2016] [Indexed: 01/21/2023]
Abstract
BACKGROUND The aim of this study was to elucidate whether genetic screening test results of pediatric patients with steroid-resistant nephrotic syndrome (SRNS) vary with ethnicity. METHODS Using high-throughput DNA sequencing, 28 nephrotic syndrome-related genes were analyzed in 110 chil-dren affected by SRNS and 10 children with isolated proteinuria enrolled by 5 centers in China (67 boys, 53 girls). Their age at disease onset ranged from 1 day to 208 months (median, 48.8 months). Patients were excluded if their age at onset of disease was over 18 years or if they were diagnosed as having Alport syndrome. RESULTS A genetic etiology was identified in 28.3% of our cohort and the likelihood of establishing a genetic diagnosis decreased as the age at onset of nephrotic syndrome increased. The most common mutated genes were ADCK4 (6.67%), NPHS1 (5.83%), WT1 (5.83%), and NPHS2 (3.33%), and the difference in the frequencies of ADCK4 and NPHS2 mutations between this study and a study on monogenic causes of SRNS in the largest international cohort of 1,783 different families was significant. A case of congenital nephrotic syndrome was attributed to a homozygous missense mutation in ADCK4, and a de novo missense mutation in TRPC6 was detected in a case of infantile nephrotic syndrome. CONCLUSIONS Our results showed that, in the first and the largest multicenter cohort of Chinese pediatric SRNS reported to date, ADCK4 is the most common causative gene, whereas there is a low prevalence of NPHS2 mutations. Our data indicated that the genetic testing results for pediatric SRNS patients vary with different ethnicities, and this information will help to improve management of the disease in clinical practice.
Collapse
Affiliation(s)
- Fang Wang
- Department of Pediatrics, Peking University First Hospital, Beijing, P. R. China
| | - Yanqin Zhang
- Department of Pediatrics, Peking University First Hospital, Beijing, P. R. China
| | - Jianhua Mao
- Department of Nephrology, The Children Hospital of Zhejiang University School of Medicine, Hangzhou, P. R. China
| | - Zihua Yu
- Department of Pediatrics, Fuzhou Dongfang Hospital, Fuzhou, P. R. China
| | - Zhuwen Yi
- Department of Pediatrics, The second Xiangya Hospital, Central South University, Changsha, P. R. China
| | - Li Yu
- Department of Pediatrics, Guangzhou First People’s Hospital, Guangzhou, P. R. China
| | - Jun Sun
- Binhai Genomics Institute, Tianjin Translational Genomics Center, BGI-Tianjin, BGI-shenzhen, Tianjin, P. R. China,BGI-Shenzhen, Shenzhen, P. R. China
| | - Xiuxiu Wei
- Binhai Genomics Institute, Tianjin Translational Genomics Center, BGI-Tianjin, BGI-shenzhen, Tianjin, P. R. China,BGI-Shenzhen, Shenzhen, P. R. China
| | - Fangrui Ding
- Department of Pediatrics, Peking University First Hospital, Beijing, P. R. China
| | - Hongwen Zhang
- Department of Pediatrics, Peking University First Hospital, Beijing, P. R. China
| | - Huijie Xiao
- Department of Pediatrics, Peking University First Hospital, Beijing, P. R. China
| | - Yong Yao
- Department of Pediatrics, Peking University First Hospital, Beijing, P. R. China
| | - Weizhen Tan
- Division of Nephrology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Svjetlana Lovric
- Division of Nephrology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jie Ding
- Department of Pediatrics, Peking University First Hospital, No. 1, Xi An Men Da Jie, Beijing, 100034, People's Republic of China.
| | - Friedhelm Hildebrandt
- Division of Nephrology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Enders 561, Boston, MA, 02115, USA.
| |
Collapse
|
79
|
Dogra S, Kaskel F. Steroid-resistant nephrotic syndrome: a persistent challenge for pediatric nephrology. Pediatr Nephrol 2017; 32:965-974. [PMID: 27783158 DOI: 10.1007/s00467-016-3459-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 06/23/2016] [Accepted: 06/24/2016] [Indexed: 12/19/2022]
Abstract
Steroid-resistant nephrotic syndrome remains a challenge to treat, but various efforts are underway to better understand the pathogenesis and improve patient outcomes. This review provides an update on the newer advances in understanding the molecular etiologies for a variety of podocyte abnormalities, potential circulating factors that may initiate and sustain the steroid-resistant state, genetic mutations, and precision medicine treatment modalities in this continuously perplexing disorder.
Collapse
Affiliation(s)
- Samriti Dogra
- Division of Pediatric Nephrology, Department of Pediatrics, Connecticut Children's Medical Center, 282 Washington Street, Hartford, CT, 06095, USA.
| | - Frederick Kaskel
- Division of Pediatric Nephrology, Department of Pediatrics, Children's Hospital at Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
80
|
WT1 Alternative Splicing: Role of Its Isoforms in Neuroblastoma. J Mol Neurosci 2017; 62:131-141. [DOI: 10.1007/s12031-017-0930-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 05/08/2017] [Indexed: 02/07/2023]
|
81
|
Reina-Alcaina L, Izquierdo-Morejón E, Romero-Hoyuela A, Miñana-López B. Tumor paratesticular atípico. Mesotelioma papilar bien diferenciado. Rev Int Androl 2017. [DOI: 10.1016/j.androl.2016.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
82
|
Ross OE, Kujubu DA, Sim JJ. Focal Segmental Glomerulosclerosis in a Patient with Ambiguous Genitalia: A Diagnostic Dilemma. Perm J 2017; 21:16-092. [PMID: 28333603 DOI: 10.7812/tpp/16-092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
INTRODUCTION The renal condition referred to as focal segmental glomerulosclerosis (FSGS) presents a diagnostic dilemma for the clinician. It encompasses and displays a nonspecific histologic appearance on a kidney biopsy specimen, rather than a unique disease entity. This characteristic of FSGS often makes treatment decisions and prognostication difficult. A 34-year-old man, who was born with ambiguous genitalia, had received a diagnosis of FSGS in young adulthood and now had advanced kidney disease. He underwent genetic testing to determine whether a genetic disorder was underlying his kidney disease and to ascertain his risk of FSGS recurrence if he were to receive a kidney transplant. The literature pertaining to genetic causes of FSGS is reviewed. We present here a diagnostic dilemma that clinicians face when confronted by a case of FSGS for which the underlying cause is unclear.
Collapse
Affiliation(s)
- Oliver E Ross
- Nephrologist at the Los Angeles Medical Center in CA.
| | - Dean A Kujubu
- Nephrologist at the Los Angeles Medical Center in CA.
| | - John J Sim
- Nephrologist at the Los Angeles Medical Center in CA.
| |
Collapse
|
83
|
Schenk H, Müller-Deile J, Kinast M, Schiffer M. Disease modeling in genetic kidney diseases: zebrafish. Cell Tissue Res 2017; 369:127-141. [PMID: 28331970 DOI: 10.1007/s00441-017-2593-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 02/22/2017] [Indexed: 01/07/2023]
Abstract
Growing numbers of translational genomics studies are based on the highly efficient and versatile zebrafish (Danio rerio) vertebrate model. The increasing types of zebrafish models have improved our understanding of inherited kidney diseases, since they not only display pathophysiological changes but also give us the opportunity to develop and test novel treatment options in a high-throughput manner. New paradigms in inherited kidney diseases have been developed on the basis of the distinct genome conservation of approximately 70 % between zebrafish and humans in terms of existing gene orthologs. Several options are available to determine the functional role of a specific gene or gene sets. Permanent genome editing can be induced via complete gene knockout by using the CRISPR/Cas-system, among others, or via transient modification by using various morpholino techniques. Cross-species rescues succeeding knockdown techniques are employed to determine the functional significance of a target gene or a specific mutation. This article summarizes the current techniques and discusses their perspectives.
Collapse
Affiliation(s)
- Heiko Schenk
- Department of Medicine/Nephrology, Hannover Medical School, Hannover, Germany
- Mount Desert Island Biological Laboratory, Salisbury Cove, Bar Harbor, Me., USA
| | - Janina Müller-Deile
- Department of Medicine/Nephrology, Hannover Medical School, Hannover, Germany
- Mount Desert Island Biological Laboratory, Salisbury Cove, Bar Harbor, Me., USA
| | - Mark Kinast
- Department of Medicine/Nephrology, Hannover Medical School, Hannover, Germany
- Mount Desert Island Biological Laboratory, Salisbury Cove, Bar Harbor, Me., USA
| | - Mario Schiffer
- Department of Medicine/Nephrology, Hannover Medical School, Hannover, Germany.
- Mount Desert Island Biological Laboratory, Salisbury Cove, Bar Harbor, Me., USA.
| |
Collapse
|
84
|
Ripperger T, Bielack SS, Borkhardt A, Brecht IB, Burkhardt B, Calaminus G, Debatin KM, Deubzer H, Dirksen U, Eckert C, Eggert A, Erlacher M, Fleischhack G, Frühwald MC, Gnekow A, Goehring G, Graf N, Hanenberg H, Hauer J, Hero B, Hettmer S, von Hoff K, Horstmann M, Hoyer J, Illig T, Kaatsch P, Kappler R, Kerl K, Klingebiel T, Kontny U, Kordes U, Körholz D, Koscielniak E, Kramm CM, Kuhlen M, Kulozik AE, Lamottke B, Leuschner I, Lohmann DR, Meinhardt A, Metzler M, Meyer LH, Moser O, Nathrath M, Niemeyer CM, Nustede R, Pajtler KW, Paret C, Rasche M, Reinhardt D, Rieß O, Russo A, Rutkowski S, Schlegelberger B, Schneider D, Schneppenheim R, Schrappe M, Schroeder C, von Schweinitz D, Simon T, Sparber-Sauer M, Spix C, Stanulla M, Steinemann D, Strahm B, Temming P, Thomay K, von Bueren AO, Vorwerk P, Witt O, Wlodarski M, Wössmann W, Zenker M, Zimmermann S, Pfister SM, Kratz CP. Childhood cancer predisposition syndromes-A concise review and recommendations by the Cancer Predisposition Working Group of the Society for Pediatric Oncology and Hematology. Am J Med Genet A 2017; 173:1017-1037. [PMID: 28168833 DOI: 10.1002/ajmg.a.38142] [Citation(s) in RCA: 152] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 12/19/2016] [Accepted: 12/30/2016] [Indexed: 12/12/2022]
Abstract
Heritable predisposition is an important cause of cancer in children and adolescents. Although a large number of cancer predisposition genes and their associated syndromes and malignancies have already been described, it appears likely that there are more pediatric cancer patients in whom heritable cancer predisposition syndromes have yet to be recognized. In a consensus meeting in the beginning of 2016, we convened experts in Human Genetics and Pediatric Hematology/Oncology to review the available data, to categorize the large amount of information, and to develop recommendations regarding when a cancer predisposition syndrome should be suspected in a young oncology patient. This review summarizes the current knowledge of cancer predisposition syndromes in pediatric oncology and provides essential information on clinical situations in which a childhood cancer predisposition syndrome should be suspected.
Collapse
Affiliation(s)
- Tim Ripperger
- Department of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Stefan S Bielack
- Pediatrics 5 (Oncology, Hematology, Immunology), Klinikum Stuttgart-Olgahospital, Stuttgart, Germany
| | - Arndt Borkhardt
- Medical Faculty, Department of Pediatric Oncology, Hematology and Clinical Immunology, University Children's Hospital, Heinrich Heine University, Düsseldorf, Germany
| | - Ines B Brecht
- General Pediatrics, Hematology/Oncology, University Children's Hospital Tuebingen, Tuebingen, Germany.,Department of Pediatrics and Adolescent Medicine, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Birgit Burkhardt
- Pediatric Hematology and Oncology, University Children's Hospital Muenster, Muenster, Germany
| | - Gabriele Calaminus
- Pediatric Hematology and Oncology, University Children's Hospital Muenster, Muenster, Germany
| | - Klaus-Michael Debatin
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Hedwig Deubzer
- Department of Pediatric Oncology and Hematology, Charité University Medicine, Berlin, Germany
| | - Uta Dirksen
- Pediatric Hematology and Oncology, University Children's Hospital Muenster, Muenster, Germany
| | - Cornelia Eckert
- Department of Pediatric Oncology and Hematology, Charité University Medicine, Berlin, Germany
| | - Angelika Eggert
- Department of Pediatric Oncology and Hematology, Charité University Medicine, Berlin, Germany
| | - Miriam Erlacher
- Faculty of Medicine, Division of Pediatric Hematology and Oncology Medical Center, Department of Pediatrics and Adolescent Medicine, University of Freiburg, Freiburg, Germany
| | - Gudrun Fleischhack
- Pediatric Oncology and Hematology, Pediatrics III, University Hospital of Essen, Essen, Germany
| | - Michael C Frühwald
- Children's Hospital Augsburg, Swabian Children's Cancer Center, Augsburg, Germany
| | - Astrid Gnekow
- Children's Hospital Augsburg, Swabian Children's Cancer Center, Augsburg, Germany
| | - Gudrun Goehring
- Department of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Norbert Graf
- Department of Pediatric Hematology and Oncology, University of Saarland, Homburg, Germany
| | - Helmut Hanenberg
- Medical Faculty, Department of Pediatric Oncology, Hematology and Clinical Immunology, University Children's Hospital, Heinrich Heine University, Düsseldorf, Germany.,Department of Otorhinolaryngology and Head/Neck Surgery, Heinrich Heine University, Düsseldorf, Germany
| | - Julia Hauer
- Medical Faculty, Department of Pediatric Oncology, Hematology and Clinical Immunology, University Children's Hospital, Heinrich Heine University, Düsseldorf, Germany
| | - Barbara Hero
- Department of Pediatric Hematology and Oncology, University of Cologne, Cologne, Germany
| | - Simone Hettmer
- Faculty of Medicine, Division of Pediatric Hematology and Oncology Medical Center, Department of Pediatrics and Adolescent Medicine, University of Freiburg, Freiburg, Germany
| | - Katja von Hoff
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Martin Horstmann
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Juliane Hoyer
- Institute of Human Genetics, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Thomas Illig
- Department of Human Genetics, Hannover Medical School, Hannover, Germany.,Hannover Unified Biobank, Hannover Medical School, Hannover, Germany
| | - Peter Kaatsch
- German Childhood Cancer Registry (GCCR), Institute for Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Roland Kappler
- Department of Pediatric Surgery, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Kornelius Kerl
- Pediatric Hematology and Oncology, University Children's Hospital Muenster, Muenster, Germany
| | - Thomas Klingebiel
- Hospital for Children and Adolescents, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Udo Kontny
- Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Medical Center, Aachen, Germany
| | - Uwe Kordes
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dieter Körholz
- Department of Pediatric Hematology and Oncology, Justus Liebig University, Giessen, Germany
| | - Ewa Koscielniak
- Pediatrics 5 (Oncology, Hematology, Immunology), Klinikum Stuttgart-Olgahospital, Stuttgart, Germany
| | - Christof M Kramm
- Division of Pediatric Hematology and Oncology, University Medical Center Goettingen, Goettingen, Germany
| | - Michaela Kuhlen
- Medical Faculty, Department of Pediatric Oncology, Hematology and Clinical Immunology, University Children's Hospital, Heinrich Heine University, Düsseldorf, Germany
| | - Andreas E Kulozik
- Department of Pediatric Oncology, Hematology and Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Britta Lamottke
- Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | - Ivo Leuschner
- Kiel Paediatric Tumor Registry, Department of Paediatric Pathology, University of Kiel, Kiel, Germany
| | - Dietmar R Lohmann
- Institute of Human Genetics, University Hospital Essen, Essen, Germany.,Eye Oncogenetics Research Group, University Hospital Essen, Essen, Germany
| | - Andrea Meinhardt
- Department of Pediatric Hematology and Oncology, Justus Liebig University, Giessen, Germany
| | - Markus Metzler
- Department of Pediatrics and Adolescent Medicine, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Lüder H Meyer
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Olga Moser
- Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Medical Center, Aachen, Germany
| | - Michaela Nathrath
- Department of Pediatric Oncology, Klinikum Kassel, Kassel, Germany.,Clinical Cooperation Group Osteosarcoma, Helmholtz Zentrum Munich, Neuherberg, Germany.,Pediatric Oncology Center, Technical University Munich, Munich, Germany
| | - Charlotte M Niemeyer
- Faculty of Medicine, Division of Pediatric Hematology and Oncology Medical Center, Department of Pediatrics and Adolescent Medicine, University of Freiburg, Freiburg, Germany
| | - Rainer Nustede
- Department of Surgery, Children's Hospital, Hannover Medical School, Hannover, Germany
| | - Kristian W Pajtler
- Department of Pediatric Oncology, Hematology and Immunology, Heidelberg University Hospital, Heidelberg, Germany.,Division of Pediatric Neuro-Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Claudia Paret
- Department of Pediatric Hematology/Oncology, University Medical Center Mainz, Mainz, Germany
| | - Mareike Rasche
- Pediatric Oncology and Hematology, Pediatrics III, University Hospital of Essen, Essen, Germany
| | - Dirk Reinhardt
- Pediatric Oncology and Hematology, Pediatrics III, University Hospital of Essen, Essen, Germany
| | - Olaf Rieß
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tuebingen, Germany
| | - Alexandra Russo
- Department of Pediatric Hematology/Oncology, University Medical Center Mainz, Mainz, Germany
| | - Stefan Rutkowski
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | | | - Reinhard Schneppenheim
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Martin Schrappe
- Department of Pediatrics, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Christopher Schroeder
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tuebingen, Germany
| | - Dietrich von Schweinitz
- Department of Pediatric Surgery, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Thorsten Simon
- Department of Pediatric Hematology and Oncology, University of Cologne, Cologne, Germany
| | - Monika Sparber-Sauer
- Pediatrics 5 (Oncology, Hematology, Immunology), Klinikum Stuttgart-Olgahospital, Stuttgart, Germany
| | - Claudia Spix
- German Childhood Cancer Registry (GCCR), Institute for Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Martin Stanulla
- Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | - Doris Steinemann
- Department of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Brigitte Strahm
- Faculty of Medicine, Division of Pediatric Hematology and Oncology Medical Center, Department of Pediatrics and Adolescent Medicine, University of Freiburg, Freiburg, Germany
| | - Petra Temming
- Pediatric Oncology and Hematology, Pediatrics III, University Hospital of Essen, Essen, Germany.,Eye Oncogenetics Research Group, University Hospital Essen, Essen, Germany
| | - Kathrin Thomay
- Department of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Andre O von Bueren
- Department of Pediatrics, Division of Pediatric Hematology and Oncology, University Medical Center Goettingen, Goettingen, Germany.,Division of Pediatric Hematology and Oncology, University Hospital of Geneva, Geneva, Switzerland
| | - Peter Vorwerk
- Pediatric Oncology, Otto von Guericke University Children's Hospital, Magdeburg, Germany
| | - Olaf Witt
- Department of Pediatric Oncology, Hematology and Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Marcin Wlodarski
- Faculty of Medicine, Division of Pediatric Hematology and Oncology Medical Center, Department of Pediatrics and Adolescent Medicine, University of Freiburg, Freiburg, Germany
| | - Willy Wössmann
- Department of Pediatric Hematology and Oncology, Justus Liebig University, Giessen, Germany
| | - Martin Zenker
- Institute of Human Genetics, University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg, Germany
| | - Stefanie Zimmermann
- Hospital for Children and Adolescents, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Stefan M Pfister
- Department of Pediatric Oncology, Hematology and Immunology, Heidelberg University Hospital, Heidelberg, Germany.,Division of Pediatric Neuro-Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christian P Kratz
- Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
85
|
Abstract
The process of sexual differentiation is central for reproduction of almost all metazoan and therefore for maintenance of practically all multicellular organisms. In sex development we can distinguish two different processes: First, sex determination is the developmental decision that directs the undifferentiated embryo into a sexually dimorphic individual. In mammals, sex determination equals gonadal development. The second process known as sex differentiation takes place once the sex determination decision has been made through factors produced by the gonads that determine the development of the phenotypic sex. Most of the knowledge on the factors involved in sexual development came from animal models and from studies of cases in whom the genetic or the gonadal sex does not match the phenotypical sex, i.e., patients affected by disorders of sex development (DSD). Generally speaking, factors influencing sex determination are transcriptional regulators, whereas factors important for sex differentiation are secreted hormones and their receptors. This review focuses on the factors involved in gonadal determination, and whenever possible, references on the "prismatic" clinical cases are given.
Collapse
Affiliation(s)
- Anna Biason-Lauber
- Department of Medicine, University of Fribourg, Chemin du Musée 5, 1700, Fribourg, Switzerland.
| |
Collapse
|
86
|
Carter S, Dixit A, Lunn A, Deorukhkar A, Christian M. Renal failure from birth-AKI or CKD? Answers. Pediatr Nephrol 2016; 31:2259-2262. [PMID: 26891727 DOI: 10.1007/s00467-016-3332-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Revised: 01/13/2016] [Accepted: 01/13/2016] [Indexed: 10/22/2022]
Affiliation(s)
- Sean Carter
- School of Medicine, University of Nottingham, Nottingham, UK
| | - Abhijit Dixit
- Nottingham Children's Hospital, Nottingham University Hospitals NHS Trust, Derby Road, Nottingham, NG7 2UH, UK
| | - Andrew Lunn
- Nottingham Children's Hospital, Nottingham University Hospitals NHS Trust, Derby Road, Nottingham, NG7 2UH, UK
| | - Anjum Deorukhkar
- Nottingham Children's Hospital, Nottingham University Hospitals NHS Trust, Derby Road, Nottingham, NG7 2UH, UK
| | - Martin Christian
- Nottingham Children's Hospital, Nottingham University Hospitals NHS Trust, Derby Road, Nottingham, NG7 2UH, UK.
| |
Collapse
|
87
|
Hersmus R, van Bever Y, Wolffenbuttel KP, Biermann K, Cools M, Looijenga LHJ. The biology of germ cell tumors in disorders of sex development. Clin Genet 2016; 91:292-301. [PMID: 27716895 DOI: 10.1111/cge.12882] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 09/29/2016] [Accepted: 09/30/2016] [Indexed: 01/01/2023]
Abstract
Development of a malignant germ cell tumor, i.e., germ cell cancer (GCC) in individuals with disorders of sex development (DSD) depends on a number of (epi-)genetic factors related to early gonadal- and germ cell development, possibly related to genetic susceptibility. Fetal development of germ cells is orchestrated by strict processes involving specification, migration and the development of a proper gonadal niche. In this review we will discuss the early (epi-)genetic events in normal and aberrant germ cell and gonadal development. Focus will be on the formation of the precursor lesions of GCC in individuals who have DSD. In our view, expression of the different embryonic markers in, and epigenetic profile of the precursor lesions reflects the developmental stage in which these cells are blocked in their maturation. Therefore, these are not a primary pathogenetic driving force. Progression later in life towards a full blown cancer likely depends on additional factors such as a changed endocrine environment in a susceptible individual. Genetic susceptibility is, as evidenced by the presence of specific risk genetic variants (SNPs) in patients with a testicular GCC, related to genes involved in early germ cell and gonadal development.
Collapse
Affiliation(s)
- Remko Hersmus
- Department of Pathology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Yolande van Bever
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Katja P Wolffenbuttel
- Department of Pediatric Urology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Katharina Biermann
- Department of Pathology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Martine Cools
- Department of Pediatric Endocrinology, Ghent University Hospital and Ghent University, Ghent, Belgium
| | | |
Collapse
|
88
|
Brok J, Treger TD, Gooskens SL, van den Heuvel-Eibrink MM, Pritchard-Jones K. Biology and treatment of renal tumours in childhood. Eur J Cancer 2016; 68:179-195. [PMID: 27969569 DOI: 10.1016/j.ejca.2016.09.005] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 08/25/2016] [Accepted: 09/01/2016] [Indexed: 02/08/2023]
Abstract
In Europe, almost 1000 children are diagnosed with a malignant renal tumour each year. The vast majority of cases are nephroblastoma, also known as Wilms' tumour (WT). Most children are treated according to Société Internationale d'Oncologie Pédiatrique Renal Tumour Study Group (SIOP-RTSG) protocols with pre-operative chemotherapy, surgery, and post-operative treatment dependent on stage and histology. Overall survival approaches 90%, but a subgroup of WT, with high-risk histology and/or relapsed disease, still have a much poorer prognosis. Outcome is similarly poor for the rare non-WT, particularly for malignant rhabdoid tumour of the kidney, metastatic clear cell sarcoma of the kidney (CCSK), and metastatic renal cell carcinoma (RCC). Improving outcome and long-term quality of life requires more accurate risk stratification through biological insights. Biomarkers are also needed to signpost potential targeted therapies for high-risk subgroups. Our understanding of Wilms' tumourigenesis is evolving and several signalling pathways, microRNA processing and epigenetics are now known to play pivotal roles. Most rhabdoid tumours display somatic and/or germline mutations in the SMARCB1 gene, whereas CCSK and paediatric RCC reveal a more varied genetic basis, including characteristic translocations. Conducting early-phase trials of targeted therapies is challenging due to the scarcity of patients with refractory or relapsed disease, the rapid progression of relapse and the genetic heterogeneity of the tumours with a low prevalence of individual somatic mutations. A further consideration in improving population survival rates is the geographical variation in outcomes across Europe. This review provides a comprehensive overview of the current biological knowledge of childhood renal tumours alongside the progress achieved through international collaboration. Ongoing collaboration is needed to ensure consistency of outcomes through standardised diagnostics and treatment and incorporation of biomarker research. Together, these objectives constitute the rationale for the forthcoming SIOP-RTSG 'UMBRELLA' study.
Collapse
Affiliation(s)
- Jesper Brok
- Cancer Section, University College London, Institute of Child Health, UK; Department of Paediatric Haematology and Oncology, Rigshospitalet, Copenhagen University Hospital, Denmark.
| | - Taryn D Treger
- Cancer Section, University College London, Institute of Child Health, UK
| | - Saskia L Gooskens
- Department of Paediatric Oncology, Princess Máxima Center for Pediatric Oncology and University of Utrecht, The Netherlands; Department of Paediatric Haematology and Oncology, Erasmus MC - Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Marry M van den Heuvel-Eibrink
- Department of Paediatric Oncology, Princess Máxima Center for Pediatric Oncology and University of Utrecht, The Netherlands
| | | |
Collapse
|
89
|
Goyal S, Mishra K, Sarkar U, Sharma S, Kumari A. Diagnostic utility of Wilms' tumour-1 protein (WT-1) immunostaining in paediatric renal tumours. Indian J Med Res 2016; 143:S59-S67. [PMID: 27748279 PMCID: PMC5080930 DOI: 10.4103/0971-5916.191776] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Background & objectives: Renal tumours constitute about 7 per cent of all neoplasms in children. It is important to differentiate Wilms’ tumour (commonest tumour) from non-Wilms’ tumours. The aim of this study was to evaluate the immunoexpression and diagnostic role of Wilms’ tumour-1 protein (WT1) in paediatric renal tumours. Methods: A total of 53 cases of renal tumours in children (below 18 yr) who underwent total nephrectomy were included in this retrospective study. WT1 immunostaining was done using mouse monoclonal WT1 antibody (clone: 6F-H2). Results: Of the 53 cases, 38 (72%) were of Wilms’ tumour. Non-Wilms’ group (15) included six cases of mesoblastic nephroma (MN), two each of clear cell sarcoma (CCSK), renal cell carcinoma (RCC) and peripheral neuroectodermal tumour (PNET) and one each of angiomyolipoma (AML), rhabdomyosarcoma (RMS) and malignant rhabdoid tumour (MRT). Proportion of WT1 positivity in Wilms’ tumour was 100 per cent in contrast to 26.7 per cent in non-Wilms’ tumours (P<0.001). Epithelial and blastemal components of Wilms’ tumour showed moderate (2+) nuclear and cytoplasmic staining in 80 (24/30) and 75 per cent (24/32) cases, respectively. MN, PNET, CCSK and AML were negative for WT1. RMS, RCC and MRT showed cytoplasmic staining, strongest in RMS. No significant association was seen between WT1 expression and NWTSG (National Wilms’ Tumor Study Group) stage. Interpretation & conclusions: WT1 helps to differentiate Wilms’ tumour from other paediatric renal tumours. It may help in differentiating the two subgroups of Wilms’ tumour which have distinct molecular pathogenesis and biological behaviour, however, further prospective studies are required for validation of this hypothesis.
Collapse
Affiliation(s)
- Surbhi Goyal
- Department of Pathology, University College of Medical Sciences, Delhi, India
| | - Kiran Mishra
- Department of Pathology, University College of Medical Sciences, Delhi, India
| | - Urvee Sarkar
- Department of Pathology, University College of Medical Sciences, Delhi, India
| | - Satendra Sharma
- Department of Pathology, University College of Medical Sciences, Delhi, India
| | - Anita Kumari
- Department of Pathology, University College of Medical Sciences, Delhi, India
| |
Collapse
|
90
|
Repression of CMIP transcription by WT1 is relevant to podocyte health. Kidney Int 2016; 90:1298-1311. [PMID: 27650733 DOI: 10.1016/j.kint.2016.07.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Revised: 07/11/2016] [Accepted: 07/14/2016] [Indexed: 12/23/2022]
Abstract
The WT1 (Wilm's tumor suppressor) gene is expressed throughout life in podocytes and is essential for the functional integrity of the glomerular filtration barrier. We have previously shown that CMIP (C-Maf inducing protein) is overproduced in podocyte diseases and alters intracellular signaling. Here we isolated the proximal region of the human CMIP promoter and showed by chromatin immunoprecipitation assays and electrophoretic-mobility shift that Wilm's tumor protein (WT1) bound to 2 WT1 response elements, located at positions -290/-274 and -57/-41 relative to transcription start site. Unlike the human CMIP gene, only one Wt1 response element was identified in the mouse Cmip proximal promoter located at position -217/-206. Luciferase reporter assays indicated that WT1 dose-dependently inhibited the transcriptional induction of the CMIP promoter. Transfection of decoy oligonucleotides mimicking the WT1 response elements prevented the inhibition of WT1 on CMIP promoter activity. Furthermore, WT1 silencing promoted Cmip expression. In line with these findings, the abundance of Cmip was early and significantly increased at the transcript and protein level in podocytes displaying a primary defect in Wt1, including Denys-Drash syndrome and Frasier syndrome. Thus, WT1 is a major repressor of the CMIP gene in physiological situations, while conditional deletion of CMIP in the developing kidney did not affect the development of mature glomeruli.
Collapse
|
91
|
Hashimoto H, Zhang X, Zheng Y, Wilson GG, Cheng X. Denys-Drash syndrome associated WT1 glutamine 369 mutants have altered sequence-preferences and altered responses to epigenetic modifications. Nucleic Acids Res 2016; 44:10165-10176. [PMID: 27596598 PMCID: PMC5137435 DOI: 10.1093/nar/gkw766] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 08/19/2016] [Accepted: 08/23/2016] [Indexed: 01/10/2023] Open
Abstract
Mutations in human zinc-finger transcription factor WT1 result in abnormal development of the kidneys and genitalia and an array of pediatric problems including nephropathy, blastoma, gonadal dysgenesis and genital discordance. Several overlapping phenotypes are associated with WT1 mutations, including Wilms tumors, Denys-Drash syndrome (DDS), Frasier syndrome (FS) and WAGR syndrome (Wilms tumor, aniridia, genitourinary malformations, and mental retardation). These conditions vary in severity from individual to individual; they can be fatal in early childhood, or relatively benign into adulthood. DDS mutations cluster predominantly in zinc fingers (ZF) 2 and 3 at the C-terminus of WT1, which together with ZF4 determine the sequence-specificity of DNA binding. We examined three DDS associated mutations in ZF2 of human WT1 where the normal glutamine at position 369 is replaced by arginine (Q369R), lysine (Q369K) or histidine (Q369H). These mutations alter the sequence-specificity of ZF2, we find, changing its affinity for certain bases and certain epigenetic forms of cytosine. X-ray crystallography of the DNA binding domains of normal WT1, Q369R and Q369H in complex with preferred sequences revealed the molecular interactions responsible for these affinity changes. DDS is inherited in an autosomal dominant fashion, implying a gain of function by mutant WT1 proteins. This gain, we speculate, might derive from the ability of the mutant proteins to sequester WT1 into unproductive oligomers, or to erroneously bind to variant target sequences.
Collapse
Affiliation(s)
- Hideharu Hashimoto
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Xing Zhang
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Yu Zheng
- RGENE, Inc., 953 Indiana Street, San Francisco, CA 94107, USA
| | | | - Xiaodong Cheng
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
92
|
New insight into the pathogenesis of minimal change nephrotic syndrome: Role of the persistence of respiratory tract virus in immune disorders. Autoimmun Rev 2016; 15:632-7. [DOI: 10.1016/j.autrev.2016.02.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 02/03/2016] [Indexed: 12/31/2022]
|
93
|
Sampson MG, Gillies CE, Robertson CC, Crawford B, Vega-Warner V, Otto EA, Kretzler M, Kang HM. Using Population Genetics to Interrogate the Monogenic Nephrotic Syndrome Diagnosis in a Case Cohort. J Am Soc Nephrol 2016; 27:1970-83. [PMID: 26534921 PMCID: PMC4926977 DOI: 10.1681/asn.2015050504] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 09/06/2015] [Indexed: 01/02/2023] Open
Abstract
To maximize clinical benefits of genetic screening of patients with nephrotic syndrome (NS) to diagnose monogenic causes, reliably distinguishing NS-causing variants from the background of rare, noncausal variants prevalent in all genomes is vital. To determine the prevalence of monogenic NS in a North American case cohort while accounting for background prevalence of genetic variation, we sequenced 21 implicated monogenic NS genes in 312 participants from the Nephrotic Syndrome Study Network and 61 putative controls from the 1000 Genomes Project (1000G). These analyses were extended to available sequence data from approximately 2500 subjects from the 1000G. A typical pathogenicity filter identified causal variants for NS in 4.2% of patients and 5.8% of subjects from the 1000G. We devised a more stringent pathogenicity filtering strategy, reducing background prevalence of causal variants to 1.5%. When applying this stringent filter to patients, prevalence of monogenic NS was 2.9%; of these patients, 67% were pediatric, and 44% had FSGS on biopsy. The rate of complete remission did not associate with monogenic classification. Thus, we identified factors contributing to inaccurate monogenic classification of NS and developed a more accurate variant filtering strategy. The prevalence and clinical correlates of monogenic NS in this sporadically affected cohort differ substantially from those reported for patients referred for genetic analysis. Particularly in unselected, population-based cases, considering putative causal variants in known NS genes from a probabilistic rather than a deterministic perspective may be more precise. We also introduce GeneVetter, a web tool for monogenic assessment of rare disease.
Collapse
Affiliation(s)
| | | | | | | | | | - Edgar A Otto
- Departments of Pediatrics and Communicable Diseases, and
| | - Matthias Kretzler
- Internal Medicine, Division of Nephrology and Department of Computational Medicine and Bioinformatics, University of Michigan School of Medicine, Ann Arbor, Michigan; and
| | - Hyun Min Kang
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, Michigan
| |
Collapse
|
94
|
Sampson MG, Pollak MR. Opportunities and Challenges of Genotyping Patients With Nephrotic Syndrome in the Genomic Era. Semin Nephrol 2016. [PMID: 26215859 DOI: 10.1016/j.semnephrol.2015.04.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Both targeted and genome-wide linkage and association studies have identified a number of genes and genetic variants associated with nephrotic syndrome (NS). Genotype-phenotype studies of patients with these variants have identified correlations of clear clinical significance. Combined with improved genomic technologies, this has resulted in increasing, and justifiable, enthusiasm for incorporating our patients' genomic information into our clinical management decisions. Here, we summarize our understanding of NS-associated genetic factors, namely rare causal mutations or common risk alleles in apolipoprotein L1. We discuss the complexities inherent in trying to ascribe risk or causality to these variants, particularly as we seek to extend genetic testing to a broader group of patients, including many with sporadic disease. Overall, the thoughtful application and interpretation of these genetic tests will maximize the benefits to our patients with NS in the form of more precise clinical care.
Collapse
Affiliation(s)
- Matthew G Sampson
- Department of Pediatrics, Division of Nephrology, University of Michigan School of Medicine, Ann Arbor, MI.
| | - Martin R Pollak
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| |
Collapse
|
95
|
Deng C, Dai R, Li X, Liu F. Genetic variation frequencies in Wilms' tumor: A meta-analysis and systematic review. Cancer Sci 2016; 107:690-9. [PMID: 26892980 PMCID: PMC4970837 DOI: 10.1111/cas.12910] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 02/11/2016] [Accepted: 02/12/2016] [Indexed: 12/11/2022] Open
Abstract
Over the last few decades, numerous biomarkers in Wilms' tumor have been confirmed and shown variations in prevalence. Most of these studies were based on small sample sizes. We carried out a meta-analysis of the research published from 1992 to 2015 to obtain more precise and comprehensive outcomes for genetic tests. In the present study, 70 out of 5175 published reports were eligible for the meta-analysis, which was carried out using Stata 12.0 software. Pooled prevalence for gene mutations WT1, WTX, CTNNB1, TP53, MYCN, DROSHA, and DGCR8 was 0.141 (0.104, 0.178), 0.147 (0.110, 0.184), 0.140 (0.100, 0.190), 0.410 (0.214, 0.605), 0.071 (0.041, 0.100), 0.082 (0.048, 0.116), and 0.036 (0.026, 0.046), respectively. Pooled prevalence of loss of heterozygosity at 1p, 11p, 11q, 16q, and 22q was 0.109 (0.084, 0.133), 0.334 (0.295, 0.373), 0.199 (0.146, 0.252), 0.151 (0.129, 0.172), and 0.148 (0.108, 0.189), respectively. Pooled prevalence of 1q and chromosome 12 gain was 0.218 (0.161, 0.275) and 0.273 (0.195, 0.350), respectively. The limited prevalence of currently known genetic alterations in Wilms' tumors indicates that significant drivers of initiation and progression remain to be discovered. Subgroup analyses indicated that ethnicity may be one of the sources of heterogeneity. However, in meta-regression analyses, no study-level characteristics of indicators were found to be significant. In addition, the findings of our sensitivity analysis and possible publication bias remind us to interpret results with caution.
Collapse
Affiliation(s)
- Changkai Deng
- Department of Urology Surgery, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorder, Key Laboratory of Pediatrics in Chongqing (CSTC2009CA5002), Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China.,Chengdu Women and Children's Central Hospital, Chengdu, China
| | - Rong Dai
- Chengdu Center for Disease Control and Prevention, Chengdu, China
| | - Xuliang Li
- Department of Urology Surgery, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorder, Key Laboratory of Pediatrics in Chongqing (CSTC2009CA5002), Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
| | - Feng Liu
- Department of Urology Surgery, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorder, Key Laboratory of Pediatrics in Chongqing (CSTC2009CA5002), Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
| |
Collapse
|
96
|
Finken MJJ, Hendriks YMC, van der Voorn JP, Veening MA, Lombardi MP, Rotteveel J. WT1 deletion leading to severe 46,XY gonadal dysgenesis, Wilms tumor and gonadoblastoma: case report. Horm Res Paediatr 2016; 83:211-6. [PMID: 25613702 DOI: 10.1159/000368964] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 10/06/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Heterozygous missense mutations in the WT1 gene that affect the function of the wild-type allele have been identified in Denys-Drash syndrome, which is characterized by severe gonadal dysgenesis, early-onset nephropathy and a predisposition to renal and gonadal cancer. Intron 9 splice-site mutations that influence the balance between WT1 isoforms cause a nearly similar phenotype, known as Frasier syndrome. Nonsense mutations and deletions only lead to WT1 haploinsufficiency and, hence, to less severe gonadal dysgenesis and late-onset nephropathy. WT1 analysis is mandatory in 46,XY gonadal dysgenesis with renal abnormality. PATIENT We describe a newborn with 46,XY severe partial gonadal dysgenesis, in whom structural renal anomalies and proteinuria were excluded. Gonadectomy was performed at the age of 1 month and the microscopy was thought to be suggestive for a gonadoblastoma. At the age of 9 months, the patient presented with a bilateral Wilms tumor. RESULTS We found a heterozygous WT1 whole-gene deletion but no other gene defects. CONCLUSIONS This case description illustrates that a WT1 deletion might be associated with a more severe phenotype than previously thought. It also illustrates that, even in the absence of renal abnormality, it is recommended to test promptly for WT1 defects in 46,XY gonadal dysgenesis.
Collapse
Affiliation(s)
- Martijn J J Finken
- Department of Pediatric Endocrinology, VU University Medical Center, Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
97
|
Hillen LM, Kamsteeg EJ, Schoots J, Tiebosch AT, Speel EJ, Roemen GM, Peutz-Koostra CJ, Stumpel CTRM. Refining the Diagnosis of Congenital Nephrotic Syndrome on Long-term Stored Tissue: c.1097G>A (p.(Arg366His)) WT1 Mutation Causing Denys Drash Syndrome. Fetal Pediatr Pathol 2016; 35:112-9. [PMID: 26882358 DOI: 10.3109/15513815.2016.1139018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Congenital nephrotic syndrome (CNS) caused by a mutation in the Wilms tumor 1 suppressor gene (WT1) is part of Denys Drash Syndrome or Frasier syndrome. In the framework of genetic counseling, the diagnosis of CNS can be refined with gene mutation studies on long-term stored formalin-fixed paraffin-embedded tissue from postmortem examination. We report a case of diffuse mesangial sclerosis with perinatal death caused by a de novo mutation in the WT1 gene in a girl with an XY-genotype. This is the first case of Denys Drash Syndrome with the uncommon missense c.1097G>A [p.(Arg366His)] mutation in the WT1 gene which has been diagnosed on long-term stored formalin-fixed paraffin-embedded tissue in 1993. This emphasizes the importance of retained and adequately stored tissue as a resource in the ongoing medical care and counseling.
Collapse
Affiliation(s)
- Lisa Maria Hillen
- a Department of Pathology , Maastricht University Medical Center , Maastricht , The Netherlands
| | - Erik Jan Kamsteeg
- b Department of Clinical Genetics , University Medical Center , Nijmegen , The Netherlands
| | - Jeroen Schoots
- b Department of Clinical Genetics , University Medical Center , Nijmegen , The Netherlands
| | - Anton Tom Tiebosch
- c Department of Pathology , Martini Ziekenhuis Groningen , Groningen , The Netherlands
| | - Ernst Jan Speel
- a Department of Pathology , Maastricht University Medical Center , Maastricht , The Netherlands
| | - Guido M Roemen
- a Department of Pathology , Maastricht University Medical Center , Maastricht , The Netherlands
| | - Carine J Peutz-Koostra
- a Department of Pathology , Maastricht University Medical Center , Maastricht , The Netherlands
| | - Constance T R M Stumpel
- d Department of Clinical Genetics and School for Oncology & Developmental Biology (GROW) , Maastricht University Medical Center , Maastricht , The Netherlands
| |
Collapse
|
98
|
Romaniuk PJ. Measuring Equilibrium Binding Constants for the WT1-DNA Interaction Using a Filter Binding Assay. Methods Mol Biol 2016; 1467:155-176. [PMID: 27417968 DOI: 10.1007/978-1-4939-4023-3_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Equilibrium binding of WT1 to specific sites in DNA and potentially RNA molecules is central in mediating the regulatory roles of this protein. In order to understand the functional effects of mutations in the nucleic acid-binding domain of WT1 proteins and/or mutations in the DNA- or RNA-binding sites, it is necessary to measure the equilibrium constant for formation of the protein-nucleic acid complex. This chapter describes the use of a filter binding assay to make accurate measurements of the binding of the WT1 zinc finger domain to the consensus WT1-binding site in DNA. The method described is readily adapted to the measurement of the effects of mutations in either the WT1 zinc finger domain or the putative binding sites within a promoter element or cellular RNA.
Collapse
Affiliation(s)
- Paul J Romaniuk
- Department of Biochemistry and Microbiology, University of Victoria, 3800 Finnerty Road, Victoria, BC, Canada, V8P 5C2.
| |
Collapse
|
99
|
Abstract
In this chapter, the role of WT1 in childhood cancer is discussed, using the key examples Wilms' tumor, desmoplastic small round cell of childhood, and leukemia. The role of WT1 in each disease is described and mirrored to the role of WT1 in normal development.
Collapse
Affiliation(s)
- Jocelyn Charlton
- UCL Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
| | - Kathy Pritchard-Jones
- UCL Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK.
- Hugh and Catherine Stevenson Professor of Paediatric Oncology, UCL Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK.
| |
Collapse
|
100
|
Weber S, Büscher AK, Hagmann H, Liebau MC, Heberle C, Ludwig M, Rath S, Alberer M, Beissert A, Zenker M, Hoyer PF, Konrad M, Klein HG, Hoefele J. Dealing with the incidental finding of secondary variants by the example of SRNS patients undergoing targeted next-generation sequencing. Pediatr Nephrol 2016; 31:73-81. [PMID: 26248470 DOI: 10.1007/s00467-015-3167-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 06/29/2015] [Accepted: 06/30/2015] [Indexed: 01/15/2023]
Abstract
BACKGROUND Steroid-resistant nephrotic syndrome (SRNS) is a severe cause of progressive renal disease. Genetic forms of SRNS can present with autosomal recessive or autosomal dominant inheritance. Recent studies have identified mutations in multiple podocyte genes responsible for SRNS. Improved sequencing methods (next-generation sequencing, NGS) now promise rapid mutational testing of SRNS genes. METHODS In the present study, a simultaneous screening of ten SRNS genes in 37 SRNS patients was performed by NGS. RESULTS In 38 % of the patients, causative mutations in one SRNS gene were found. In 22 % of the patients, in addition to these mutations, a secondary variant in a different gene was identified. CONCLUSIONS This high incidence of accumulating sequence variants was unexpected but, although they might have modifier effects, the pathogenic potential of these additional sequence variants seems unclear so far. The example of molecular diagnostics by NGS in SRNS patients shows that these new sequencing technologies might provide further insight into molecular pathogenicity in genetic disorders but will also generate results, which will be difficult to interpret and complicate genetic counseling. Although NGS promises more frequent identification of disease-causing mutations, the identification of causative mutations, the interpretation of incidental findings and possible pitfalls might pose problems, which hopefully will decrease by further experience and elucidation of molecular interactions.
Collapse
Affiliation(s)
- Stefanie Weber
- Pediatric Nephrology, Pediatrics II, University of Duisburg-Essen, Essen, Germany
| | - Anja K Büscher
- Pediatric Nephrology, Pediatrics II, University of Duisburg-Essen, Essen, Germany
| | - Henning Hagmann
- Renal Division, Department II of Internal Medicine and Center for Molecular Medicine, University of Cologne, Cologne, Germany
| | - Max C Liebau
- Renal Division, Department II of Internal Medicine and Center for Molecular Medicine, University of Cologne, Cologne, Germany
- Department of Pediatrics, University Hospital of Cologne, Cologne, Germany
| | - Christian Heberle
- Center for Human Genetics and Laboratory Diagnostics Dr. Klein, Dr. Rost and Colleagues, Martinsried, Germany
| | - Michael Ludwig
- Department of Clinical Chemistry and Clinical Pharmacology, University of Bonn, Bonn, Germany
| | - Sabine Rath
- Center for Human Genetics and Laboratory Diagnostics Dr. Klein, Dr. Rost and Colleagues, Martinsried, Germany
| | - Martin Alberer
- Department of Infectious Diseases and Tropical Medicine, Ludwig-Maximilians University, Munich, Germany
| | - Antje Beissert
- Pediatric Nephrology, University Children's Hospital, Würzburg, Germany
| | - Martin Zenker
- Institute of Human Genetics, University Hospital, Magdeburg, Germany
| | - Peter F Hoyer
- Pediatric Nephrology, Pediatrics II, University of Duisburg-Essen, Essen, Germany
| | | | - Hanns-Georg Klein
- Center for Human Genetics and Laboratory Diagnostics Dr. Klein, Dr. Rost and Colleagues, Martinsried, Germany
| | - Julia Hoefele
- Center for Human Genetics and Laboratory Diagnostics Dr. Klein, Dr. Rost and Colleagues, Martinsried, Germany.
| |
Collapse
|