1
|
Ma G, Gao A, Chen J, Liu P, Sarda R, Gulliver J, Wang Y, Joiner C, Hu M, Kim EJ, Yeger H, Le HD, Chen X, Li WJ, Xu W. Modeling high-risk Wilms tumors enables the discovery of therapeutic vulnerability. Cell Rep Med 2024; 5:101770. [PMID: 39368485 PMCID: PMC11513831 DOI: 10.1016/j.xcrm.2024.101770] [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] [Received: 04/28/2024] [Revised: 08/09/2024] [Accepted: 09/12/2024] [Indexed: 10/07/2024]
Abstract
Wilms tumor (WT) is the most common pediatric kidney cancer treated with standard chemotherapy. However, less-differentiated blastemal type of WT often relapses. To model the high-risk WT for therapeutic intervention, we introduce pluripotency factors into WiT49, a mixed-type WT cell line, to generate partially reprogrammed cells, namely WiT49-PRCs. When implanted into the kidney capsule in mice, WiT49-PRCs form kidney tumors and develop both liver and lung metastases, whereas WiT49 tumors do not metastasize. Histological characterization and gene expression signatures demonstrate that WiT49-PRCs recapitulate blastemal-predominant WTs. Moreover, drug screening in isogeneic WiT49 and WiT49-PRCs leads to the identification of epithelial- or blastemal-predominant WT-sensitive drugs, whose selectivity is validated in patient-derived xenografts (PDXs). Histone deacetylase (HDAC) inhibitors (e.g., panobinostat and romidepsin) are found universally effective across different WT and more potent than doxorubicin in PDXs. Taken together, WiT49-PRCs serve as a blastemal-predominant WT model for therapeutic intervention to treat patients with high-risk WT.
Collapse
Affiliation(s)
- Gui Ma
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI 53706, USA; Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI 53706, USA
| | - Ang Gao
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI 53706, USA; Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI 53706, USA
| | - Jiani Chen
- Department of Computational Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Peng Liu
- Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53706, USA; Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI 53706, USA
| | - Rakesh Sarda
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Jessica Gulliver
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Yidan Wang
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI 53706, USA; Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI 53706, USA
| | - Carstyn Joiner
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI 53706, USA; Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI 53706, USA
| | - Mingshan Hu
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI 53706, USA; Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI 53706, USA
| | - Eui-Jun Kim
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI 53706, USA; Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI 53706, USA
| | - Herman Yeger
- Program in Developmental and Stem Cell Biology, Peter Gilgan Centre for Research and Learning, SickKids, Toronto, ON M5G 0A4, Canada
| | - Hau D Le
- Department of Surgery, Division of Pediatric Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Xiang Chen
- Department of Computational Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Wan-Ju Li
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI, USA
| | - Wei Xu
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI 53706, USA; Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI 53706, USA.
| |
Collapse
|
2
|
Meng K, Zhao Z, Gao Y, Wu K, Liu W, Wang X, Zheng Y, Zhao W, Wang B. The synergistic effects of anoikis-related genes and EMT-related genes in the prognostic prediction of Wilms tumor. Front Mol Biosci 2024; 11:1469775. [PMID: 39351154 PMCID: PMC11439783 DOI: 10.3389/fmolb.2024.1469775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 09/03/2024] [Indexed: 10/04/2024] Open
Abstract
Wilms tumor (WT) is the most common type of malignant abdominal tumor in children; it exhibits a high degree of malignancy, grow rapidly, and is prone to metastasis. This study aimed to construct a prognosis model based on anoikis-related genes (ARGs) and epithelial-mesenchymal transition (EMT)-related genes (ERGs) for WT patients; we assessed the characteristics of the tumor microenvironment and treatment efficacy, as well as identifying potential therapeutic targets. To this end, we downloaded transcriptome sequencing data and clinical data for WT and normal renal cortices and used R to construct and validate the prognostic model based on ARGs and ERGs. Additionally, we performed clinical feature analysis, nomogram construction, mutation analysis, drug sensitivity analysis, Connectivity Map (cMAP) analysis, functional enrichment analysis, and immune infiltration analysis. Finally, we screened the hub gene using the STRING database and validated it via experiments. In this way, we constructed a model with good accuracy and robustness, which was composed of seven anoikis- and EMT-related genes. Paclitaxel and mesna were selected as potential chemotherapeutic drugs and adjuvant chemotherapeutic drugs for the WT high-risk group by using the Genomics of Drug Sensitivity in Cancer (GDSC) and cMAP compound libraries, respectively. We proved the existence of a strong correlation between invasive immune cells and prognostic genes and risk scores. Next, we selected NTRK2 as the hub gene, and in vitro experiments confirmed that its inhibition can significantly inhibit the proliferation and migration of tumor cells and promote late apoptosis. In summary, we screened out the potential biomarkers and chemotherapeutic drugs that can improve the prognosis of patients with WT.
Collapse
Affiliation(s)
- Kexin Meng
- Department of Medical Ultrasound, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Medical and Health Key Laboratory of Abdominal Medical Imaging, Jinan, China
| | - Zerui Zhao
- Department of Clinical Pharmacy, Clinical Trial Center, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Yaqing Gao
- Department of Medical Ultrasound, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Medical and Health Key Laboratory of Abdominal Medical Imaging, Jinan, China
| | - Keliang Wu
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Wei Liu
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xiaoqing Wang
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yi Zheng
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Wei Zhao
- Department of Clinical Pharmacy, Clinical Trial Center, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Bei Wang
- Department of Medical Ultrasound, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Medical and Health Key Laboratory of Abdominal Medical Imaging, Jinan, China
| |
Collapse
|
3
|
Whitworth J, Armstrong R, Maher ER. Wilms tumour resulting from paternal transmission of a TRIM28 pathogenic variant-A first report. Eur J Hum Genet 2024; 32:361-364. [PMID: 38282073 PMCID: PMC10923773 DOI: 10.1038/s41431-024-01545-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/03/2024] [Accepted: 01/16/2024] [Indexed: 01/30/2024] Open
Abstract
Wilms tumour (nephroblastoma) is a renal embryonal tumour that is frequently caused by constitutional variants in a small range of cancer predisposition genes. TRIM28 has recently been identified as one such gene. Previously, observational data strongly suggested a parent of origin effect, whereby Wilms tumour only occurred following maternal inheritance of a pathogenic genetic variant. However, here we report a child with bilateral Wilms tumour who had inherited a pathogenic TRIM28 variant from their father. This finding suggests that genetic counselling for paternally inherited pathogenic variants in TRIM28 should include discussion of a potential risk of Wilms tumour.
Collapse
Affiliation(s)
- James Whitworth
- University of Cambridge Department of Medical Genetics, Box 238 Level 6, Addenbrooke's Treatment Centre, Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge, CB2 0QQ, UK.
- Department of Clinical Genetics, Box 134 Level 6, Addenbrooke's Treatment Centre, Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge, CB2 0QQ, UK.
| | - Ruth Armstrong
- Department of Clinical Genetics, Box 134 Level 6, Addenbrooke's Treatment Centre, Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge, CB2 0QQ, UK
| | - Eamonn R Maher
- University of Cambridge Department of Medical Genetics, Box 238 Level 6, Addenbrooke's Treatment Centre, Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge, CB2 0QQ, UK
- Department of Clinical Genetics, Box 134 Level 6, Addenbrooke's Treatment Centre, Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge, CB2 0QQ, UK
- Aston Medical School, Aston University, Birmingham, B4 7ET, UK
| |
Collapse
|
4
|
Stevenson MJ, Phanor SK, Patel U, Gisselbrecht SS, Bulyk ML, O'Brien LL. Altered binding affinity of SIX1-Q177R correlates with enhanced WNT5A and WNT pathway effector expression in Wilms tumor. Dis Model Mech 2023; 16:dmm050208. [PMID: 37815464 PMCID: PMC10668032 DOI: 10.1242/dmm.050208] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 09/27/2023] [Indexed: 10/11/2023] Open
Abstract
Wilms tumors present as an amalgam of varying proportions of tissues located within the developing kidney, one being the nephrogenic blastema comprising multipotent nephron progenitor cells (NPCs). The recurring missense mutation Q177R in NPC transcription factors SIX1 and SIX2 is most correlated with tumors of blastemal histology and is significantly associated with relapse. Yet, the transcriptional regulatory consequences of SIX1/2-Q177R that might promote tumor progression and recurrence have not been investigated extensively. Utilizing multiple Wilms tumor transcriptomic datasets, we identified upregulation of the gene encoding non-canonical WNT ligand WNT5A in addition to other WNT pathway effectors in SIX1/2-Q177R mutant tumors. SIX1 ChIP-seq datasets from Wilms tumors revealed shared binding sites for SIX1/SIX1-Q177R within a promoter of WNT5A and at putative distal cis-regulatory elements (CREs). We demonstrate colocalization of SIX1 and WNT5A in Wilms tumor tissue and utilize in vitro assays that support SIX1 and SIX1-Q177R activation of expression from the WNT5A CREs, as well as enhanced binding affinity within the WNT5A promoter that may promote the differential expression of WNT5A and other WNT pathway effectors associated with SIX1-Q177R tumors.
Collapse
Affiliation(s)
- Matthew J. Stevenson
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Sabrina K. Phanor
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Urvi Patel
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Stephen S. Gisselbrecht
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Martha L. Bulyk
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Lori L. O'Brien
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
5
|
Faivre L, Crépin JC, Réda M, Nambot S, Carmignac V, Abadie C, Mirault T, Faure-Conter C, Mazereeuw-Hautier J, Maza A, Puzenat E, Collonge-Rame MA, Bursztejn AC, Philippe C, Thauvin-Robinet C, Chevarin M, Abasq-Thomas C, Amiel J, Arpin S, Barbarot S, Baujat G, Bessis D, Bourrat E, Boute O, Chassaing N, Coubes C, Demeer B, Edery P, El Chehadeh S, Goldenberg A, Hadj-Rabia S, Haye D, Isidor B, Jacquemont ML, Van Kien PK, Lacombe D, Lehalle D, Lambert L, Martin L, Maruani A, Morice-Picard F, Petit F, Phan A, Pinson L, Rossi M, Touraine R, Vanlerberghe C, Vincent M, Vincent-Delorme C, Whalen S, Willems M, Marle N, Verkarre V, Devalland C, Devouassoux-Shisheboran M, Abad M, Rioux-Leclercq N, Bonniaud B, Duffourd Y, Martel J, Binquet C, Kuentz P, Vabres P. Low risk of embryonic and other cancers in PIK3CA-related overgrowth spectrum: Impact on screening recommendations. Clin Genet 2023; 104:554-563. [PMID: 37580112 DOI: 10.1111/cge.14410] [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] [Received: 12/23/2022] [Revised: 07/07/2023] [Accepted: 07/19/2023] [Indexed: 08/16/2023]
Abstract
The PIK3CA-related overgrowth spectrum (PROS) encompasses various conditions caused by mosaic activating PIK3CA variants. PIK3CA somatic variants are also involved in various cancer types. Some generalized overgrowth syndromes are associated with an increased risk of Wilms tumor (WT). In PROS, abdominal ultrasound surveillance has been advocated to detect WT. We aimed to determine the risk of embryonic and other types of tumors in patients with PROS in order to evaluate surveillance relevance. We searched the clinical charts from 267 PROS patients for the diagnosis of cancer, and reviewed the medical literature for the risk of cancer. In our cohort, six patients developed a cancer (2.2%), and Kaplan Meier analyses estimated cumulative probabilities of cancer occurrence at 45 years of age was 5.6% (95% CI = 1.35%-21.8%). The presence of the PIK3CA variant was only confirmed in two out of four tumor samples. In the literature and our cohort, six cases of Wilms tumor/nephrogenic rests (0.12%) and four cases of other cancers have been reported out of 483 proven PIK3CA patients, in particular the p.(His1047Leu/Arg) variant. The risk of WT in PROS being lower than 5%, this is insufficient evidence to recommend routine abdominal imaging. Long-term follow-up studies are needed to evaluate the risk of other cancer types, as well as the relationship with the extent of tissue mosaicism and the presence or not of the variant in the tumor samples.
Collapse
Affiliation(s)
- Laurence Faivre
- Equipe INSERM UMR1231, Génétique des Anomalies du Développement, FHU TRANSLAD, Université Bourgogne Franche-Comté, Dijon, France
- Centre de Génétique, Centre de Référence Anomalies du Développement et Syndromes Malformatifs et FHU TRANSLAD, CHU Dijon Bourgogne, Dijon, France
| | - Jean-Charles Crépin
- Equipe INSERM UMR1231, Génétique des Anomalies du Développement, FHU TRANSLAD, Université Bourgogne Franche-Comté, Dijon, France
- Service de Dermatologie, CHU Dijon Bourgogne, Dijon, France
- Centre de référence Maladies Rares Génétiques à Expression Cutanée (MAGEC), CHU Dijon, Dijon, France
| | - Manon Réda
- Oncogénétique, Centre de lutte contre le cancer Georges François Leclerc, Dijon, France
| | - Sophie Nambot
- Equipe INSERM UMR1231, Génétique des Anomalies du Développement, FHU TRANSLAD, Université Bourgogne Franche-Comté, Dijon, France
- Centre de Génétique, Centre de Référence Anomalies du Développement et Syndromes Malformatifs et FHU TRANSLAD, CHU Dijon Bourgogne, Dijon, France
- Oncogénétique, Centre de lutte contre le cancer Georges François Leclerc, Dijon, France
| | - Virginie Carmignac
- Equipe INSERM UMR1231, Génétique des Anomalies du Développement, FHU TRANSLAD, Université Bourgogne Franche-Comté, Dijon, France
- Centre de référence Maladies Rares Génétiques à Expression Cutanée (MAGEC), CHU Dijon, Dijon, France
| | | | - Tristan Mirault
- Université Paris Cité, PARCC INSERM U970, Centre de référence des maladies vasculaires rares, Hôpital européen Georges-Pompidou, Assistance Publique Hôpitaux de Paris, Paris, France
| | | | | | - Aude Maza
- Service de Dermatologie, CHU Toulouse, Toulouse, France
| | - Eve Puzenat
- Service de Dermatologie, CHU Besançon, Besançon, France
| | | | | | - Christophe Philippe
- Equipe INSERM UMR1231, Génétique des Anomalies du Développement, FHU TRANSLAD, Université Bourgogne Franche-Comté, Dijon, France
- UF6254 Innovation en Diagnostic Génomique des Maladies Rares, Plate-forme de Biologie Hospitalo-Universitaire, CHU Dijon-Bourgogne, Dijon, France
| | - Christel Thauvin-Robinet
- Equipe INSERM UMR1231, Génétique des Anomalies du Développement, FHU TRANSLAD, Université Bourgogne Franche-Comté, Dijon, France
- Centre de Référence Déficiences Intellectuelles de Causes Rares, CHU Dijon Bourgogne, Dijon, France
| | - Martin Chevarin
- Equipe INSERM UMR1231, Génétique des Anomalies du Développement, FHU TRANSLAD, Université Bourgogne Franche-Comté, Dijon, France
- UF6254 Innovation en Diagnostic Génomique des Maladies Rares, Plate-forme de Biologie Hospitalo-Universitaire, CHU Dijon-Bourgogne, Dijon, France
| | - Claire Abasq-Thomas
- Département de Pédiatrie et Génétique Médicale, CHU Brest Morvan, Brest, France
| | - Jeanne Amiel
- Service de Médecine Génomique des Maladies Rares et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Stéphanie Arpin
- Service de Génétique Clinique et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, CHRU de Tours, Tours, France
| | | | - Geneviève Baujat
- Service de Médecine Génomique des Maladies Rares et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Didier Bessis
- Département de Dermatologie, CHRU de Montpellier, Montpellier, France
| | - Emmanuelle Bourrat
- Service de dermatologie, centre de référence maladies génétiques à expression cutanée MAGEC, CHU St-Louis, Service de pédiatrie générale, CHU Robert Debré, Paris, France
| | - Odile Boute
- Service de Génétique Clinique et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, CHU Lille, Lille, France
| | - Nicolas Chassaing
- Service de Génétique Médicale et Centre de Compétence Anomalies du Développement et Syndromes Malformatifs, CHU Toulouse, Toulouse, France
| | - Christine Coubes
- Département de Génétique Médicale, Maladies rares et Médecine Personnalisée, et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, CHRU de Montpellier, Montpellier, France
| | - Bénédicte Demeer
- Centre d'Activité de Génétique Clinique et Oncogénétique, CHU d'Amiens, Amiens, France
| | - Patrick Edery
- Service de génétique, Centre de Référence Anomalies du Développement, Hospices Civils de Lyon, Bron, France
- INSERM U1028, CNRS UMR5292, Centre de Recherche en Neurosciences de Lyon, GENDEV Team, Université Claude Bernard Lyon 1, Bron, France
| | - Salima El Chehadeh
- Service de Génétique Médicale, Centre de Référence Déficiences Intellectuelles de Causes Rares, Institut de Génétique Médicale d'Alsace (IGMA), CHRU de Strasbourg, Strasbourg, France
| | - Alice Goldenberg
- Service de Génétique et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, CHU de Rouen et Centre Normand de Génomique Médicale et Médecine Personnalisée, Rouen, France
| | - Smail Hadj-Rabia
- Service de Dermatologie et Centre de Référence des Maladies Rares Génétiques à Expression Cutanée (MAGEC), Université Paris Descartes-Sorbonne Paris Cité, Institut Imagine, Hôpital Universitaire Necker Enfants Malades, Paris, France
| | - Damien Haye
- Service de Génétique Clinique et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, CHRU de Tours, Tours, France
| | - Bertrand Isidor
- Service de Génétique Médicale et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, CHU de Nantes, Nantes, France
| | - Marie-Line Jacquemont
- Unité de Génétique Médicale et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, CHU de la Réunion, Saint-Pierre, France
| | - Philippe Khau Van Kien
- Unité de Génétique Médicale et Cytogénétique, Centre de Compétence Anomalies du Développement et Syndromes Malformatifs, CHU de Nîmes, Nîmes, France
| | - Didier Lacombe
- Service de Génétique Médicale et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, CHU de Bordeaux, Bordeaux, France
| | - Daphné Lehalle
- Equipe INSERM UMR1231, Génétique des Anomalies du Développement, FHU TRANSLAD, Université Bourgogne Franche-Comté, Dijon, France
| | - Laetitia Lambert
- Service de Génétique Clinique et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, CHU de Nancy, Nancy, France
| | | | | | - Fanny Morice-Picard
- Service de Génétique Clinique et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, CHU de Nancy, Nancy, France
- Service de Dermatologie, CHU de Bordeaux, Bordeaux, France
| | - Florence Petit
- Service de Génétique Clinique et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, CHU Lille, Lille, France
| | - Alice Phan
- Service de Dermatologie, CHU de Lyon, Lyon, France
| | - Lucile Pinson
- Département de Génétique Médicale, Maladies rares et Médecine Personnalisée, et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, CHRU de Montpellier, Montpellier, France
| | - Massimiliano Rossi
- Service de génétique, Centre de Référence Anomalies du Développement, Hospices Civils de Lyon, Bron, France
- INSERM U1028, CNRS UMR5292, Centre de Recherche en Neurosciences de Lyon, GENDEV Team, Université Claude Bernard Lyon 1, Bron, France
| | - Renaud Touraine
- Service de Génétique Clinique et Centre de Compétence Anomalies du Développement et Syndromes Malformatifs, CHU de Saint-Etienne, Saint-Etienne, France
| | - Clémence Vanlerberghe
- Service de Génétique Clinique et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, CHU Lille, Lille, France
| | - Marie Vincent
- Service de Génétique Médicale et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, CHU de Nantes, Nantes, France
| | - Catherine Vincent-Delorme
- Service de Génétique Clinique et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, CHU Lille, Lille, France
| | - Sandra Whalen
- Unité Fonctionnelle de Génétique Clinique, Hôpital Armand-Trousseau, Paris, France
| | - Marjolaine Willems
- Département de Génétique Médicale, Maladies rares et Médecine Personnalisée, et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, CHRU de Montpellier, Montpellier, France
| | - Nathalie Marle
- UF6254 Innovation en Diagnostic Génomique des Maladies Rares, Plate-forme de Biologie Hospitalo-Universitaire, CHU Dijon-Bourgogne, Dijon, France
| | - Virginie Verkarre
- Service d'Anatomie Pathologique, Hôpital Européen Georges Pompidou, Paris, France et INSERM UMR 970, Equipe 13, PARCC Université de Paris Cité, Paris, France
| | - Christine Devalland
- Service d'Anatomie Pathologique, Hôpital Nord Franche Comté, Trevenans, France
| | | | - Marine Abad
- Service d'Anatomie Pathologique, CHU Besançon, Besançon, France
| | | | | | - Yannis Duffourd
- Equipe INSERM UMR1231, Génétique des Anomalies du Développement, FHU TRANSLAD, Université Bourgogne Franche-Comté, Dijon, France
| | - Jehanne Martel
- Centre de référence Maladies Rares Génétiques à Expression Cutanée (MAGEC), CHU Dijon, Dijon, France
| | - Christine Binquet
- INSERM, Université de Bourgogne, CHU Dijon Bourgogne, CIC 1432, Module Épidémiologie Clinique, Dijon, France
| | - Paul Kuentz
- Equipe INSERM UMR1231, Génétique des Anomalies du Développement, FHU TRANSLAD, Université Bourgogne Franche-Comté, Dijon, France
- Oncobiologie Génétique Bioinformatique, PCBio, CHU Besançon, Besançon, France
| | - Pierre Vabres
- Equipe INSERM UMR1231, Génétique des Anomalies du Développement, FHU TRANSLAD, Université Bourgogne Franche-Comté, Dijon, France
- Service de Dermatologie, CHU Dijon Bourgogne, Dijon, France
- Centre de référence Maladies Rares Génétiques à Expression Cutanée (MAGEC), CHU Dijon, Dijon, France
| |
Collapse
|
6
|
Li X, An W, Pan H, Fan Y, Huang H, Wang Y, Shen W, Zu L, Meng F, Zhou X. Wilms' tumour gene 1 (WT1) enhances non-small cell lung cancer malignancy and is inhibited by microRNA-498-5p. BMC Cancer 2023; 23:824. [PMID: 37667197 PMCID: PMC10476375 DOI: 10.1186/s12885-023-11295-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 08/12/2023] [Indexed: 09/06/2023] Open
Abstract
BACKGROUND Wilms' tumour gene 1 (WT1) is clearly recognized as a tumour promoter in diversiform of human malignancies. Nevertheless, knowledge of its expression, functions and potential molecular mechanisms in non-small cell lung cancer (NSCLC) remains elusive. METHODS Differential expression of WT1 mRNA and protein between NSCLC and normal tissues were assessed by analyzing RNA-seq data from Oncomine and protein data from Human Protein Atlas, respectively. Subsequently, prognosis significance and immune cell infiltration were analyzed by Kaplan-Meier plotter and CIBERSORT. 60 pairs of local NSCLC tissues were involved to validate WT1 expression by quantitative PCR (qPCR) and Western blot. Moreover, Cell Counting Kit-8 (CCK-8), colony formation, transwell, dual luciferase reporter assays and in vivo xenograft tumour growth experiments were conducted to explore the function and mechanism of WT1 in NSCLC. RESULTS Our solid data indicated that WT1 was increased in NSCLC tissues and cell lines in comparison with their matched controls. In particular, its upregulation correlated with worse prognosis and immune infiltration of the patients. Functional assays demonstrated that knockdown of WT1 inhibited NSCLC malignancy, including inhibiting cell proliferation, survival and invasion. Further exploration discovered that microRNA-498-5p (miR-498-5p) was the upstream suppressor of WT1 by directly targeting the 3' untranslated region (UTR) of WT1 mRNA. Moreover, expression of miR-498-5p was notably decreased and inversely correlated with WT1 in NSCLC tissues. Finally, we proved that miR-498-5p was a potent tumour suppressor in NSCLC by suppressing cell proliferation, survival and invasion, while WT1 restoration could in turn disrupt this suppression both in vitro and in vivo. CONCLUSION The abnormal increase in WT1 contributes to the malignant properties of NSCLC cells, and miR-498-5p is a natural inhibitor of WT1. Our findings might facilitate the development of novel therapeutic strategies against NSCLC in the future.
Collapse
Grants
- No. 81302002, No. 82273019 to Xuebing Li; No. 81502166, No. 81972354, No. 82172901 to Xuexia Zhou National Natural Science Foundation of China
- No. 81302002, No. 82273019 to Xuebing Li; No. 81502166, No. 81972354, No. 82172901 to Xuexia Zhou National Natural Science Foundation of China
- No. 18JCYBJC92100, to Xuebing Li; No. 21JCQNJC01440, to Xuexia Zhou; No. 17JCYBJC25400, to Yaguang Fan; No. 17JCQNJC11700, to Hongli Pan Natural Science Foundation of Tianjin City
- No. 18JCYBJC92100, to Xuebing Li; No. 21JCQNJC01440, to Xuexia Zhou; No. 17JCYBJC25400, to Yaguang Fan; No. 17JCQNJC11700, to Hongli Pan Natural Science Foundation of Tianjin City
- No. 18JCYBJC92100, to Xuebing Li; No. 21JCQNJC01440, to Xuexia Zhou; No. 17JCYBJC25400, to Yaguang Fan; No. 17JCQNJC11700, to Hongli Pan Natural Science Foundation of Tianjin City
- No. 18JCYBJC92100, to Xuebing Li; No. 21JCQNJC01440, to Xuexia Zhou; No. 17JCYBJC25400, to Yaguang Fan; No. 17JCQNJC11700, to Hongli Pan Natural Science Foundation of Tianjin City
- No. CFC2020kyxm003, to Xuebing Li; No. CFC2020kyxm002, to Yaguang Fan Key Project of Cancer Foundation of China
- No. CFC2020kyxm003, to Xuebing Li; No. CFC2020kyxm002, to Yaguang Fan Key Project of Cancer Foundation of China
- No. ZYYFY2019022, to Fanrong Meng Foundation of Tianjin Medical University General Hospital
Collapse
Affiliation(s)
- Xuebing Li
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Wenzhe An
- Department of Neuropathology, Tianjin Key Laboratory of Injuries, Variations and Regeneration of the Nervous System, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System of Education Ministry, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Hongli Pan
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Yaguang Fan
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Hua Huang
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Yixuan Wang
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Wang Shen
- West China Hospital, Sichuan Lung Cancer Institute, Sichuan Lung Cancer Center, Sichuan University, Chengdu, China
| | - Lingling Zu
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Fanrong Meng
- Tianjin Prenatal Diagnostic Center, Obstetrics and Gynecology Department, Tianjin Medical University General Hospital, Tianjin, China
| | - Xuexia Zhou
- Department of Neuropathology, Tianjin Key Laboratory of Injuries, Variations and Regeneration of the Nervous System, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System of Education Ministry, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China.
| |
Collapse
|
7
|
Stoltze UK, Hildonen M, Hansen TVO, Foss-Skiftesvik J, Byrjalsen A, Lundsgaard M, Pignata L, Grønskov K, Tumer Z, Schmiegelow K, Brok JS, Wadt KAW. Germline (epi)genetics reveals high predisposition in females: a 5-year, nationwide, prospective Wilms tumour cohort. J Med Genet 2023; 60:842-849. [PMID: 37019617 PMCID: PMC10447365 DOI: 10.1136/jmg-2022-108982] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 03/10/2023] [Indexed: 04/07/2023]
Abstract
BACKGROUND Studies suggest that Wilms tumours (WT) are caused by underlying genetic (5%-10%) and epigenetic (2%-29%) mechanisms, yet studies covering both aspects are sparse. METHODS We performed prospective whole-genome sequencing of germline DNA in Danish children diagnosed with WT from 2016 to 2021, and linked genotypes to deep phenotypes. RESULTS Of 24 patients (58% female), 3 (13%, all female) harboured pathogenic germline variants in WT risk genes (FBXW7, WT1 and REST). Only one patient had a family history of WT (3 cases), segregating with the REST variant. Epigenetic testing revealed one (4%) additional patient (female) with uniparental disomy of chromosome 11 and Beckwith-Wiedemann syndrome (BWS). We observed a tendency of higher methylation of the BWS-related imprinting centre 1 in patients with WT than in healthy controls. Three patients (13%, all female) with bilateral tumours and/or features of BWS had higher birth weights (4780 g vs 3575 g; p=0.002). We observed more patients with macrosomia (>4250 g, n=5, all female) than expected (OR 9.98 (95% CI 2.56 to 34.66)). Genes involved in early kidney development were enriched in our constrained gene analysis, including both known (WT1, FBXW7) and candidate (CTNND1, FRMD4A) WT predisposition genes. WT predisposing variants, BWS and/or macrosomia (n=8, all female) were more common in female patients than male patients (p=0.01). CONCLUSION We find that most females (57%) and 33% of all patients with WT had either a genetic or another indicator of WT predisposition. This emphasises the need for scrutiny when diagnosing patients with WT, as early detection of underlying predisposition may impact treatment, follow-up and genetic counselling.
Collapse
Affiliation(s)
- Ulrik Kristoffer Stoltze
- Department of Clinical Genetics, Rigshospitalet, Copenhagen, Denmark
- Department of Pediatrics, Rigshospitalet, Copenhagen, Denmark
| | - Mathis Hildonen
- Department of Genetics, Kennedy Center-National Research Center on Rare Genetic Diseases, Glostrup, Denmark
| | | | | | - Anna Byrjalsen
- Department of Clinical Genetics, Rigshospitalet, Copenhagen, Denmark
| | - Malene Lundsgaard
- Department of Clinical Genetics, Aalborg University Hospital, Aalborg, North Denmark Region, Denmark
| | - Laura Pignata
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, Università Degli Studi Della Campania "Luigi Vanvitelli", Caserta, Italy
| | - Karen Grønskov
- Department of Clinical Genetics, Rigshospitalet, Copenhagen, Denmark
| | - Zeynep Tumer
- Applied Human Molecular Genetics, Kennedy Center, Copenhagen University Hospital, Rigshospitalet, Glostrup, Denmark
| | | | - Jesper Sune Brok
- Department of Pediatrics and Adolescent Medicine, Rigshospitalet, Copenhagen, Denmark
| | - Karin A W Wadt
- Department of Clinical Genetics, University Hospital of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
8
|
Sarkany B, Kuthi L, Kovacs G. Novel concept of Wilms' tumor development: involvement of pluripotential cells of ureteric bud. Hum Pathol 2023; 138:34-40. [PMID: 37209922 DOI: 10.1016/j.humpath.2023.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 05/15/2023] [Indexed: 05/22/2023]
Abstract
It is acknowledged that nephron develops after bilateral induction of the metanephric mesenchyma and branching ureteric bud (UB), and that nephrogenic rest and Wilms' tumor (nephroblastoma) arises from impaired differentiation of metanephric blastema. The aim of this study was to obtain more information on the involvement of UB derivatives in nephrogenic rest and Wilms' tumor. We applied immunohistochemistry to analyze nephrogenic rests and Wilms' tumors with mixed histology, including regressive and blastemal types. We used antibodies recognizing UB tip cells (ROBO1, SLIT2, RET), principal cells (AQP2), α- and β-intercalated cells (SLC26A4, SLC4A1, ATP6V1B1, ATP6V0D2), and their precursors (CA2). Tubules surrounded by tumorous blastemal cells resembling UB tip were positive for RET, ROBO1, and SLIT2 in Wilms' tumor. Moreover, CA2-positive tubular structures and ATP6V1B1- and ATP6V0D2-positive immature non-α- and non-β-intercalated cells were detected in nephrogenic rest and Wilms' tumor. We suggest that Wilms' tumor is more than nephroblastoma and propose a definition that Wilms tumor is a malignant embryonal neoplasm derived from pluripotential cells of nephrogenic blastema and of ureteric bud tip.
Collapse
Affiliation(s)
- Beatrix Sarkany
- Department of Urology, Medical School, University of Pecs, 7621, Pecs, Hungary
| | - Levente Kuthi
- Department of Pathology, University of Szeged, 6725, Szeged, Hungary
| | - Gyula Kovacs
- Department of Urology, Medical School, University of Pecs, 7621, Pecs, Hungary; Medical Faculty, Ruprecht-Karls-University, 69120, Heidelberg, Germany.
| |
Collapse
|
9
|
Zheng H, Liu J, Pan X, Cui X. Biomarkers for patients with Wilms tumor: a review. Front Oncol 2023; 13:1137346. [PMID: 37554168 PMCID: PMC10405734 DOI: 10.3389/fonc.2023.1137346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 06/27/2023] [Indexed: 08/10/2023] Open
Abstract
Wilms tumor, originating from aberrant fetal nephrogenesis, is the most common renal malignancy in childhood. The overall survival of children is approximately 90%. Although existing risk-stratification systems are helpful in identifying patients with poor prognosis, the recurrence rate of Wilms tumors remains as high as 15%. To resolve this clinical problem, diverse studies on the occurrence and progression of the disease have been conducted, and the results are encouraging. A series of molecular biomarkers have been identified with further studies on the mechanism of tumorigenesis. Some of these show prognostic value and have been introduced into clinical practice. Identification of these biomarkers can supplement the existing risk-stratification systems. In the future, more biomarkers will be discovered, and more studies are required to validate their roles in improving the detection rate of occurrence or recurrence of Wilms tumor and to enhance clinical outcomes.
Collapse
Affiliation(s)
| | | | - Xiuwu Pan
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xingang Cui
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
10
|
Pishbin F, Ziamajidi N, Abbasalipourkabir R, Najafi R, Farhadian M. Correlation of Wilms' Tumor 1 (WT1) with Oxidative Stress Markers and Expression of miR-361-5p; New Aspect of WT1 in Breast Cancer. Indian J Clin Biochem 2023; 38:338-350. [PMID: 37234179 PMCID: PMC10205927 DOI: 10.1007/s12291-022-01053-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 05/12/2022] [Indexed: 10/17/2022]
Abstract
Breast carcinoma is a heterogeneous disease that affects millions of women worldwide. Wilms' tumor 1 (WT1) is an oncogene that promotes proliferation, metastasis and reduces apoptosis. MicroRNAs (miR) are short noncoding RNAs with a major role in cancer metastasis. In present study, we investigated the association of serum level of WT1 with oxidative stress and expression of miR-361-5p in breast cancer. Serum samples of 45 patients and of 45 healthy women analyzed for protein level of WT1, malondialdehyde (MDA), total oxidant status (TOS), and total antioxidant capacity (TAC). Serum and tissue expression of miR-361-5p in 45 tumor tissues and 45 paired non-tumor adjacent tissues and 45 serum samples of patients and healthy women analyzed by qRT-PCR. Protein levels of WT1 not significantly difference in serum of patients compared to healthy controls. Serum levels of MDA and TOS in patients were higher, but TAC level was lower than healthy controls (p < 0.001). There was a positive correlation between WT1 with MDA and TOS, and a negative correlation between WT1 with TAC in patients. miR-361-5p expression in tumor tissues and serum of patients was lower than non-tumor adjacent tissues and serum of healthy controls, respectively (p < 0.001). Moreover, there was a negative correlation between miR-361-5p and WT1 in patients. The positive correlation between WT1 with MDA and TOS and negative correlation between TAC and miR-361-5p suggests that this gene can play an important role in worse prognoses in breast cancer. Additionally, miR-361-5p may serve as an invasive biomarker for early detection of breast cancer.
Collapse
Affiliation(s)
- Fariba Pishbin
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Nasrin Ziamajidi
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Molecular Medicine Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Roghayeh Abbasalipourkabir
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Rezvan Najafi
- Molecular Medicine Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Molecular Medicine and Genetics, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Maryam Farhadian
- Department of Biostatistics, School of Health, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
11
|
Jansson C, Mengelbier LH. Retinoic acid promotes differentiation of WiT49- but not of CCG99-11 Wilms tumour cells. Cancer Rep (Hoboken) 2023:e1819. [PMID: 37186071 DOI: 10.1002/cnr2.1819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/14/2023] [Accepted: 04/10/2023] [Indexed: 05/17/2023] Open
Abstract
BACKGROUND Most children with Wilms tumour are successfully treated with multidrug chemotherapy and surgery. These treatments cause severe side effects for the patients, an issue that needs to be addressed by exploring other treatment options with less or no side effects. One option is to complement current therapies with agents that could potentially induce tumour cell differentiation, for example retinoic acid (RA). AIMS To facilitate quick assessment of an agent's effect on Wilms tumour differentiation by a rapid in vitro model system. METHODS AND RESULTS Here WiT49 and CCG99-11 Wilms tumour cells were treated with 10 μM RA for 72 h or 9 days. Cultured cells were scraped off from Petri dishes, pelleted and embedded in paraffin in the same way as clinical tumour specimens are preserved. Cell morphology and differentiation were evaluated by analyses of haematoxylin eosin (H&E) and immunohistochemical stainings. Based on H&E, WT1 and CKAE1/3 stainings, RA treatment induced further epithelial differentiation of WiT49 cells, whereas there was no sign of induced maturation in CCG99-11 cells. Ki67 staining showed that RA inhibited cell proliferation in both cell lines. CONCLUSIONS Our study shows that in vitro culturing of WiT49 and CCG99-11 cells, followed by pelleting and paraffin embedding of cell pellets, could aid in a quick evaluation of potential differentiating agents against Wilms tumour. In addition, our results strengthen previous results that retinoic acid could be a potential complement to regular Wilms tumour treatment.
Collapse
Affiliation(s)
- Caroline Jansson
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Sweden
| | | |
Collapse
|
12
|
Dickinson K, Hammond L, Akpa M, Chu LL, Lalonde CT, Goumba A, Goodyer P. WT1 regulates expression of DNA repair gene Neil3 during nephrogenesis. Am J Physiol Renal Physiol 2023; 324:F245-F255. [PMID: 36546838 DOI: 10.1152/ajprenal.00207.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Mammalian nephrons arise from a population of nephron progenitor cells (NPCs) expressing the master transcription factor Wilms tumor-1 (WT1), which is crucial for NPC proliferation, migration, and differentiation. In humans, biallelic loss of WT1 precludes nephrogenesis and leads to the formation of Wilms tumor precursor lesions. We hypothesize that WT1 normally primes the NPC for nephrogenesis by inducing expression of NPC-specific DNA repair genes that protect the genome. We analyzed transcript levels for a panel of DNA repair genes in embryonic day 17.5 (E17.5) versus adult mouse kidneys and noted seven genes that were increased >20-fold. We then isolated Cited1+ NPCs from E17.5 kidneys and found that only one gene, nei-like DNA glycosylase 3 (Neil3), was enriched. RNAscope in situ hybridization of E17.5 mouse kidneys showed increased Neil3 expression in the nephrogenic zone versus mature nephron structures. To determine whether Neil3 expression is WT1 dependent, we knocked down Wt1 in Cited1+ NPCs (60% knockdown efficiency) and noted a 58% reduction in Neil3 transcript levels. We showed that WT1 interacts with the Neil3 promoter and that activity of a Neil3 promoter-reporter vector was increased twofold in WT1+ versus WT1- cells. We propose that Neil3 is a WT1-dependent DNA repair gene expressed at high levels in Cited1+ NPCs, where it repairs mutational injury to the genome during nephrogenesis. NEIL3 is likely just one of many such lineage-specific repair mechanisms that respond to genomic injury during kidney development.NEW & NOTEWORTHY We studied the molecular events leading to Wilms tumors as a model for the repair of genomic injury. Specifically, we showed that WT1 activates DNA repair gene Neil3 in nephron progenitor cells. However, our observations offer a much broader principle, demonstrating that the embryonic kidney invests in lineage-specific expression of DNA repair enzymes. Thus, it is conceivable that failure of these mechanisms could lead to a variety of "sporadic" congenital renal malformations and human disease.
Collapse
Affiliation(s)
- Kyle Dickinson
- Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada
| | - Leah Hammond
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Murielle Akpa
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Lee Lee Chu
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Caleb Tse Lalonde
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Alexandre Goumba
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Paul Goodyer
- Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada.,Department of Human Genetics, McGill University, Montreal, Quebec, Canada.,Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.,Department of Pediatrics, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
13
|
Ng JKM, Li JJX, Lai BSW, Tsang JY, Chan AWS, Cheung CMT, Ip ECC, Tse GM. WT1 as a myoepithelial marker: a comparative study of breast, cutaneous, and salivary gland lesions. Hum Pathol 2023; 135:76-83. [PMID: 36739952 DOI: 10.1016/j.humpath.2023.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 01/05/2023] [Accepted: 01/18/2023] [Indexed: 02/05/2023]
Abstract
WT1 immunostain is expressed in various benign and malignant neoplasms, as well as normal myoepithelial cells. WT1 shows differential expression in non-neoplastic, benign, and malignant neoplastic myoepithelial cells of the salivary gland. In this study, WT1 immunostain and other myoepithelial markers were compared to investigate the value of WT1 as a myoepithelial marker, and to delineate the expression profile of WT1 in nonsalivary gland myoepithelial cells. WT1, p63, and calponin immunostains were performed on normal and lesional tissues from the breast (adenosis, sclerosing adenosis, lactating adenoma, nipple adenoma, tubular adenoma, adenomyoepithelioma, and adenoid cystic carcinoma [ACC]), skin (cutaneous mixed tumor, hidradenoma, spiradenoma, and ACC), and salivary gland (pleomorphic adenoma and ACC). The stained slides were digitized and orientated with H&E images and assessed simultaneously using QuPath. A total of 129, 58, and 56 breast, cutaneous, and salivary gland lesions, respectively, were included. There was poor agreement between WT1-p63 and WT1-calponin (κ < 0.1) in all organs, with absence of WT1 expression in normal salivary gland myoepithelium and most ACCs. There were no significant differences in WT1 expression in myoepithelial cells in normal breast tissue and benign breast neoplasms. Compared to pleomorphic adenomas, cutaneous mixed tumors showed lower WT1 expression (P < .001). WT1 is a less sensitive myoepithelial marker than calponin and p63. However, its unique pattern of expression in salivary gland primary for pleomorphic adenomas/cutaneous mixed tumor can favor a diagnosis of benign salivary gland tumors, particularly in small biopsy specimens.
Collapse
Affiliation(s)
- Joanna K M Ng
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| | - Joshua J X Li
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| | - Billy S W Lai
- Department of Pathology, North District Hospital, Hong Kong
| | - Julia Y Tsang
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| | - Agnes W S Chan
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| | - Christina M T Cheung
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| | - Edric C C Ip
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| | - Gary M Tse
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong.
| |
Collapse
|
14
|
Julson JR, Beierle EA. Embryology and surgical anatomy of pediatric solid tumors. Semin Pediatr Surg 2022; 31:151233. [PMID: 36423516 PMCID: PMC10084944 DOI: 10.1016/j.sempedsurg.2022.151233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- J R Julson
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - E A Beierle
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA.
| |
Collapse
|
15
|
Shiraki T, Hayashi T, Ozue J, Watanabe M. Appropriate Amounts and Activity of the Wilms' Tumor Suppressor Gene, wt1, Are Required for Normal Pronephros Development of Xenopus Embryos. J Dev Biol 2022; 10:jdb10040046. [PMID: 36412640 PMCID: PMC9680428 DOI: 10.3390/jdb10040046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/23/2022] [Accepted: 10/27/2022] [Indexed: 12/14/2022] Open
Abstract
The Wilms' tumor suppressor gene, wt1, encodes a zinc finger-containing transcription factor that binds to a GC-rich motif and regulates the transcription of target genes. wt1 was first identified as a tumor suppressor gene in Wilms' tumor, a pediatric kidney tumor, and has been implicated in normal kidney development. The WT1 protein has transcriptional activation and repression domains and acts as a transcriptional activator or repressor, depending on the target gene and context. In Xenopus, an ortholog of wt1 has been isolated and shown to be expressed in the developing embryonic pronephros. To investigate the role of wt1 in pronephros development in Xenopus embryos, we mutated wt1 by CRISPR/Cas9 and found that the expression of pronephros marker genes was reduced. In reporter assays in which known WT1 binding sequences were placed upstream of the luciferase gene, WT1 activated transcription of the luciferase gene. The injection of wild-type or artificially altered transcriptional activity of wt1 mRNA disrupted the expression of pronephros marker genes in the embryos. These results suggest that the appropriate amounts and activity of WT1 protein are required for normal pronephros development in Xenopus embryos.
Collapse
Affiliation(s)
- Taisei Shiraki
- Graduate School of Sciences and Technology for Innovation, Tokushima University, 1-1 Minamijosanjima-Cho, Tokushima 770-8054, Japan
| | - Takuma Hayashi
- Graduate School of Sciences and Technology for Innovation, Tokushima University, 1-1 Minamijosanjima-Cho, Tokushima 770-8054, Japan
| | - Jotaro Ozue
- Graduate School of Sciences and Technology for Innovation, Tokushima University, 1-1 Minamijosanjima-Cho, Tokushima 770-8054, Japan
| | - Minoru Watanabe
- Graduate School of Sciences and Technology for Innovation, Tokushima University, 1-1 Minamijosanjima-Cho, Tokushima 770-8054, Japan
- Institute of Liberal Arts and Sciences, Tokushima University, 1-1 Minamijosanjima-Cho, Tokushima 770-8054, Japan
- Correspondence: ; Tel.: +81-088-656-7253
| |
Collapse
|
16
|
Nucleic acid therapy in pediatric cancer. Pharmacol Res 2022; 184:106441. [PMID: 36096420 DOI: 10.1016/j.phrs.2022.106441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/02/2022] [Accepted: 09/07/2022] [Indexed: 12/24/2022]
Abstract
The overall survival, progress free survival, and life quality of cancer patients have improved due to the advance in minimally invasive surgery, precision radiotherapy, and various combined chemotherapy in the last decade. Furthermore, the discovery of new types of therapeutics, such as immune checkpoint inhibitors and immune cell therapies have facilitated both patients and doctors to fight with cancers. Moreover, in the context of the development in biocompatible and cell type targeting nano-carriers as well as nucleic acid-based drugs for initiating and enhancing the anti-tumor response have come to the age. The treatment paradigms utilization of nucleic acids, including short interfering RNA (siRNA), antisense oligonucleotides (ASO), and messenger RNA (mRNA), can target specific protein expression to achieve the therapeutic effects. Over ten nucleic acid therapeutics have been approved by the FDA and EMA in rare diseases and genetic diseases as well as dozens of registered clinical trails for varies cancers. Though generally less dangerous of pediatric cancers than adult cancers was observed during the past decades, yet pediatric cancers accounted for a significant proportion of child deaths which hurt those family very deeply. Therefore, it is necessary to pay more attention for improving the treatment of pediatric cancer and discovering new nucleic acid therapeutics which may help to improve the therapeutic effect and prognoses in turns to ameliorate the survival period and quality of life for children patient. In this review, we focus on the nucleic acid therapy in pediatric cancers.
Collapse
|
17
|
MicroRNA-27a-5p Inhibits Proliferation, Migration, and Invasion and Promotes Apoptosis of Wilms' Tumor Cell by Targeting PBOV1. Mol Cell Biol 2022; 42:e0039721. [PMID: 35862727 PMCID: PMC9387294 DOI: 10.1128/mcb.00397-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Wilms' tumor is the most common type of renal tumor in children. MicroRNAs (miRNAs) are small noncoding RNAs that play crucial regulatory roles in tumorigenesis. We aimed to study the expression profile and function of miR-27a-5p in Wilms' tumor. miR-27a-5p expression was downregulated in human Wilms' tumor tissues. Functionally, overexpression of miR-27a-5p promoted cell apoptosis of Wilms' tumor cells. Furthermore, upregulated miR-27a-5p delayed xenograft Wilms' tumor tumorigenesis in vivo. Bioinformatics analysis predicted that miR-27a-5p directly targeted the 3'-untranslated region (3'-UTR) of PBOV1, and luciferase reporter assay confirmed the interaction between miR-27a-5p and PBOV1. The function of PBOV1 in Wilms' tumor was evaluated in vitro, and knockdown of PBOV1 dampened cell migration. In addition, overexpression of PBOV1 antagonized the tumor-suppressive effect of miR-27a-5p in Wilms' tumor cells. Collectively, our findings reveal the regulatory axis of miR-27a-5p/PBOV1 in Wilms' tumor, and miR-27a-5p might serve as a novel therapeutic target in Wilms' tumor.
Collapse
|
18
|
Yin X, Lin H, Lin L, Miao L, He J, Zhuo Z. LncRNAs and CircRNAs in cancer. MedComm (Beijing) 2022; 3:e141. [PMID: 35592755 PMCID: PMC9099016 DOI: 10.1002/mco2.141] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 04/14/2022] [Accepted: 04/15/2022] [Indexed: 12/24/2022] Open
Affiliation(s)
- Xin Yin
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center Guangzhou Medical University Guangzhou Guangdong China
- College of Pharmacy Jinan University Guangzhou Guangdong China
| | - Huiran Lin
- Faculty of Medicine Macau University of Science and Technology Macau China
| | - Lei Lin
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center Guangzhou Medical University Guangzhou Guangdong China
| | - Lei Miao
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center Guangzhou Medical University Guangzhou Guangdong China
| | - Jing He
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center Guangzhou Medical University Guangzhou Guangdong China
| | - Zhenjian Zhuo
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center Guangzhou Medical University Guangzhou Guangdong China
- Laboratory Animal Center, School of Chemical Biology and Biotechnology Peking University Shenzhen Graduate School Shenzhen China
| |
Collapse
|
19
|
Abstract
MicroRNAs (miRNAs) belong to a class of endogenous small noncoding RNAs that regulate gene expression at the posttranscriptional level, through both translational repression and mRNA destabilization. They are key regulators of kidney morphogenesis, modulating diverse biological processes in different renal cell lineages. Dysregulation of miRNA expression disrupts early kidney development and has been implicated in the pathogenesis of developmental kidney diseases. In this Review, we summarize current knowledge of miRNA biogenesis and function and discuss in detail the role of miRNAs in kidney morphogenesis and developmental kidney diseases, including congenital anomalies of the kidney and urinary tract and Wilms tumor. We conclude by discussing the utility of miRNAs as potentially novel biomarkers and therapeutic agents.
Collapse
Affiliation(s)
- Débora Malta Cerqueira
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- John G. Rangos Sr. Research Center, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Maliha Tayeb
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- John G. Rangos Sr. Research Center, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jacqueline Ho
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- John G. Rangos Sr. Research Center, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
20
|
Willis KR, Sathe AA, Xing C, Koduru P, Artunduaga M, Butler EB, Park JY, Kurmasheva RT, Houghton PJ, Chen KS, Rakheja D. Extrarenal Anaplastic Wilms Tumor: A Case Report With Genomic Analysis and Tumor Models. J Pediatr Hematol Oncol 2022; 44:147-154. [PMID: 35129140 PMCID: PMC9035038 DOI: 10.1097/mph.0000000000002413] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 12/29/2021] [Indexed: 11/25/2022]
Abstract
Primary extrarenal Wilms tumors are rare neoplasms that are presumed to arise from metanephric or mesonephric remnants outside of the kidney. Their pathogenesis is debated but has not been studied, and there are no reports of genomic descriptions of extrarenal Wilms tumors. We describe a diffusely anaplastic extrarenal Wilms tumor that occurred in the lower abdomen and upper pelvis of a 10-year-old boy. In addition to the clinical, histopathologic, and radiologic features, we describe the cytogenetic changes and exomic profile of the tumor. The tumor showed loss of the tumor suppressor AMER1, loss of chromosome regions 1p, 16q, and 22q, gain of chromosome 8, and loss of function TP53 mutation-findings known to occur in renal Wilms tumors. This is the first description of the exomic profile of a primary extrarenal Wilms tumor. Our data indicate that primary extrarenal Wilms tumors may follow the same pathogenetic pathways that are seen in renal Wilms tumors. Finally, we describe the establishment of first ever tumor models (primary cell line and patient-derived xenograft) from an extrarenal Wilms tumor.
Collapse
Affiliation(s)
| | - Adwait A Sathe
- Eugene McDermott Center for Human Growth and Development
| | - Chao Xing
- Eugene McDermott Center for Human Growth and Development
- Division of Pediatric Radiology, Department of Radiology
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, University of Texas Southwestern Medical Center
| | | | - Maddy Artunduaga
- Division of Pediatric Radiology, Department of Radiology
- Children's Health System of Texas, Dallas
| | - Erin B Butler
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, University of Texas Southwestern Medical Center
| | - Jason Y Park
- Departments of Pathology
- Children's Health System of Texas, Dallas
| | - Raushan T Kurmasheva
- Greehey Children's Cancer Research Institute
- Department of Molecular Medicine, University of Texas Health at San Antonio, San Antonio, TX
| | - Peter J Houghton
- Greehey Children's Cancer Research Institute
- Department of Molecular Medicine, University of Texas Health at San Antonio, San Antonio, TX
| | - Kenneth S Chen
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, University of Texas Southwestern Medical Center
- Children's Health System of Texas, Dallas
| | - Dinesh Rakheja
- Departments of Pathology
- Children's Health System of Texas, Dallas
| |
Collapse
|
21
|
Liu BH, Liu GB, Zhang BB, Shen J, Xie LL, Liu XQ, Yao W, Dong R, Bi YL, Dong KR. Tumor Suppressive Role of MUC6 in Wilms Tumor via Autophagy-Dependent β-Catenin Degradation. Front Oncol 2022; 12:756117. [PMID: 35574418 PMCID: PMC9097904 DOI: 10.3389/fonc.2022.756117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 03/28/2022] [Indexed: 11/13/2022] Open
Abstract
Wilms tumor is the most common renal malignancy in children. Known gene mutations account for about 40% of all wilms tumor cases, but the full map of genetic mutations in wilms tumor is far from clear. Whole genome sequencing and RNA sequencing were performed in 5 pairs of wilms tumor tissues and adjacent normal tissues to figure out important genetic mutations. Gene knock-down, CRISPR-induced mutations were used to investigate their potential effects in cell lines and in-vivo xenografted model. Mutations in seven novel genes (MUC6, GOLGA6L2, GPRIN2, MDN1, MUC4, OR4L1 and PDE4DIP) occurred in more than one patient. The most prevalent mutation was found in MUC6, which had 7 somatic exonic variants in 4 patients. In addition, TaqMan assay and immunoblot confirmed that MUC6 expression was reduced in WT tissues when compared with control tissues. Moreover, the results of MUC6 knock-down assay and CRISPR-induced MUC6 mutations showed that MUC6 inhibited tumor aggression via autophagy-dependent β-catenin degradation while its mutations attenuated tumor-suppressive effects of MUC6. Seven novel mutated genes (MUC6, GOLGA6L2, GPRIN2, MDN1, MUC4, OR4L1 and PDE4DIP) were found in WT, among which MUC6 was the most prevalent one. MUC6 acted as a tumor suppressive gene through autophagy dependent β-catenin pathway.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Yun-Li Bi
- Department of Pediatric Surgery, Shanghai Key Laboratory of Birth Defect, Children’s Hospital of Fudan University, Shanghai, China
| | - Kui-Ran Dong
- Department of Pediatric Surgery, Shanghai Key Laboratory of Birth Defect, Children’s Hospital of Fudan University, Shanghai, China
| |
Collapse
|
22
|
Hu M, Ma Q, Liu B, Wang Q, Zhang T, Huang T, Lv Z. Long Non-Coding RNAs in the Pathogenesis of Diabetic Kidney Disease. Front Cell Dev Biol 2022; 10:845371. [PMID: 35517509 PMCID: PMC9065414 DOI: 10.3389/fcell.2022.845371] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 03/08/2022] [Indexed: 01/09/2023] Open
Abstract
Diabetic kidney disease (DKD) is one of the major microvascular complications of diabetes mellitus, with relatively high morbidity and mortality globally but still in short therapeutic options. Over the decades, a large body of data has demonstrated that oxidative stress, inflammatory responses, and hemodynamic disorders might exert critical influence in the initiation and development of DKD, whereas the delicate pathogenesis of DKD remains profoundly elusive. Recently, long non-coding RNAs (lncRNAs), extensively studied in the field of cancer, are attracting increasing attentions on the development of diabetes mellitus and its complications including DKD, diabetic retinopathy, and diabetic cardiomyopathy. In this review, we chiefly focused on abnormal expression and function of lncRNAs in major resident cells (mesangial cell, endothelial cell, podocyte, and tubular epithelial cell) in the kidney, summarized the critical roles of lncRNAs in the pathogenesis of DKD, and elaborated their potential therapeutic significance, in order to advance our knowledge in this field, which might help in future research and clinical treatment for the disease.
Collapse
Affiliation(s)
- Mengsi Hu
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Nephrology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qiqi Ma
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Bing Liu
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Nephrology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qianhui Wang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Tingwei Zhang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Tongtong Huang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zhimei Lv
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Nephrology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- *Correspondence: Zhimei Lv,
| |
Collapse
|
23
|
Falcone MP, Pritchard-Jones K, Brok J, Mifsud W, Williams RD, Nakata K, Tugnait S, Al-Saadi R, Side L, Anderson J, Duncan C, Marks SD, Bockenhauer D, Chowdhury T. Long-term kidney function in children with Wilms tumour and constitutional WT1 pathogenic variant. Pediatr Nephrol 2022; 37:821-832. [PMID: 34608521 PMCID: PMC8960606 DOI: 10.1007/s00467-021-05125-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 02/25/2021] [Accepted: 05/05/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND Wilms tumour (WT) survivors, especially patients with associated syndromes or genitourinary anomalies due to constitutional WT1 pathogenic variant, have increased risk of kidney failure. We describe the long-term kidney function in children with WT and WT1 pathogenic variant to inform the surgical strategy and oncological management of such complex children. METHODS Retrospective analysis of patients with WT and constitutional WT1 pathogenic variant treated at a single centre between 1993 and 2016, reviewing genotype, phenotype, tumour histology, laterality, treatment, patient survival, and kidney outcome. RESULTS We identified 25 patients (60% male, median age at diagnosis 14 months, range 4-74 months) with WT1 deletion (4), missense (2), nonsense (8), frameshift (7), or splice site (4) pathogenic variant. Thirteen (52%) had bilateral disease, 3 (12%) had WT-aniridia, 1 had incomplete Denys-Drash syndrome, 11 (44%) had genitourinary malformation, and 10 (40%) had no phenotypic anomalies. Patient survival was 100% and 3 patients were in remission after relapse at median follow-up of 9 years. Seven patients (28%) commenced chronic dialysis of which 3 were after bilateral nephrectomies. The overall kidney survival for this cohort as mean time to start of dialysis was 13.38 years (95% CI: 10.3-16.4), where 7 patients experienced kidney failure at a median of 5.6 years. All of these 7 patients were subsequently transplanted. In addition, 2 patients have stage III and stage IV chronic kidney disease and 12 patients have albuminuria and/or treatment with ACE inhibitors. Four patients (3 frameshift; 1 WT1 deletion) had normal blood pressure and kidney function without proteinuria at follow-up from 1.5 to 12 years. CONCLUSIONS Despite the known high risk of kidney disease in patients with WT and constitutional WT1 pathogenic variant, nearly two-thirds of patients had sustained native kidney function, suggesting that nephron-sparing surgery (NSS) should be attempted when possible without compromising oncological risk. Larger international studies are needed for accurate assessment of WT1genotype-kidney function phenotype correlation.
Collapse
Affiliation(s)
- Maria Pia Falcone
- Department of Paediatric Oncology Great Ormond Street Hospital, UCL Institute of Child Health, NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK
- Paediatric Residency Program, University of Foggia, Foggia, Italy
| | - Kathryn Pritchard-Jones
- Department of Paediatric Oncology Great Ormond Street Hospital, UCL Institute of Child Health, NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK
| | - Jesper Brok
- Department of Paediatric Oncology Great Ormond Street Hospital, UCL Institute of Child Health, NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK
- Dept. of Paediatric Haematology and Oncology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - William Mifsud
- Department of Paediatric Oncology Great Ormond Street Hospital, UCL Institute of Child Health, NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK
| | - Richard D Williams
- Department of Paediatric Oncology Great Ormond Street Hospital, UCL Institute of Child Health, NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK
| | - Kayo Nakata
- Department of Paediatric Oncology Great Ormond Street Hospital, UCL Institute of Child Health, NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK
| | - Suzanne Tugnait
- Department of Paediatric Oncology Great Ormond Street Hospital, UCL Institute of Child Health, NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK
| | - Reem Al-Saadi
- Department of Paediatric Oncology Great Ormond Street Hospital, UCL Institute of Child Health, NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK
- Dept. of Histopathology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Lucy Side
- Dept. of Clinical Genetics, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - John Anderson
- Department of Paediatric Oncology Great Ormond Street Hospital, UCL Institute of Child Health, NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK
| | - Catriona Duncan
- Department of Paediatric Oncology Great Ormond Street Hospital, UCL Institute of Child Health, NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK
| | - Stephen D Marks
- Department of Paediatric Oncology Great Ormond Street Hospital, UCL Institute of Child Health, NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK
- Dept. of Paediatric Nephrology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Detlef Bockenhauer
- Dept. of Paediatric Nephrology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
- UCL Department of Renal Medicine, London, UK
| | - Tanzina Chowdhury
- Department of Paediatric Oncology Great Ormond Street Hospital, UCL Institute of Child Health, NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK.
| |
Collapse
|
24
|
Chen Y, Miao L, Lin H, Zhuo Z, He J. The role of m6A modification in pediatric cancer. Biochim Biophys Acta Rev Cancer 2022; 1877:188691. [PMID: 35122883 DOI: 10.1016/j.bbcan.2022.188691] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 01/25/2022] [Accepted: 01/28/2022] [Indexed: 02/07/2023]
Abstract
With the development of RNA modification research, the importance of N6-methyladenosine (m6A) in tumors cannot be ignored. m6A promotes the self-renewal of tumor stem cells and the proliferation of tumor cells. It affects post-transcriptional gene expression through epigenetic mechanisms, combining various factors to determine proteins' fate and altering the biological function. This modification process runs through the entire tumors, and genes affected by m6A modification may be the critical targets for cancers breakthroughs. Though generally less dangerous than adult cancer, pediatric cancer accounts for a significant proportion of child deaths. What is more alarming is that the occurrences of adult tumors are highly associated with the poor prognoses of pediatric tumors. Therefore, it is necessary to pay attention to the importance of pediatric cancer and discover new therapeutic targets, which will help improve the therapeutic effect and prognoses of the diseases. We collected and investigated m6A modification in pediatric cancers based on mRNA and non-coding RNA, finding that m6A factors were involved in glioma, hepatoblastoma, nephroblastoma, neuroblastoma, osteosarcoma, medulloblastoma, retinoblastoma, and acute lymphoblastic leukemia. Consequently, we summarized the relationships between the m6A factors and these pediatric cancers.
Collapse
Affiliation(s)
- Yongping Chen
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Lei Miao
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Huiran Lin
- Faculty of Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Zhenjian Zhuo
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China; Laboratory Animal Center, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China.
| | - Jing He
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China.
| |
Collapse
|
25
|
Su C, Huang R, Yu Z, Zheng J, Liu F, Liang H, Mo Z. Myelin and lymphocyte protein serves as a prognostic biomarker and is closely associated with the tumor microenvironment in the nephroblastoma. Cancer Med 2022; 11:1427-1438. [PMID: 35023304 PMCID: PMC8894696 DOI: 10.1002/cam4.4542] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/05/2021] [Accepted: 12/06/2021] [Indexed: 12/30/2022] Open
Abstract
Nephroblastoma, also known as Wilms' tumor (WT), is the most common renal tumor that occurs in children. Although the efficacy of treatment has been significantly improved by a series of comprehensive treatments, some patients still have poor prognosis. Myelin and lymphocyte (MAL) protein, a highly hydrophobic integrated membrane‐bound protein, has been implicated in many tumors and is also closely linked to kidney development. However, the relationship between MAL and WT has not yet been elucidated. Therefore, we attempted to evaluate the feasibility of MAL as a promising prognosis factor for WT. The differential expression of MAL was investigated using TARGET database and was verified using the Gene Expression Omnibus database and real‐time quantitative PCR. The prognostic ability of MAL was determined using Kaplan–Meier and Cox regression analyses. Pearson correlation analysis was applied to explore the relationship between MAL expression and methylation sites. The ESTIMATE and CIBERSORT algorithms showed that MAL expression was associated with the WT tumor microenvironment. Gene Set Enrichment Analysis (GSEA) indicated that multiple signaling pathways closely associated with tumorigenesis were differentially enriched between the high‐ and low‐MAL groups. In conclusion, our study comprehensively explored the potential of MAL as a prognosis factor for WT. Meanwhile, we also demonstrated that MAL, as a prognostic factor for WT, may be closely related to the tumor microenvironment.
Collapse
Affiliation(s)
- Cheng Su
- Department of Pediatric Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Guangxi Medical University, Nanning, China
| | | | - Zhenyuan Yu
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Key Laboratory of Colleges and Universities, Nanning, China
| | - Jie Zheng
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Key Laboratory of Colleges and Universities, Nanning, China
| | | | | | - Zengnan Mo
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Key Laboratory of Colleges and Universities, Nanning, China.,Institute of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
| |
Collapse
|
26
|
Patel K, Phipps WS, Luu HS, Rakheja D. Laboratory testing in pediatric cancer patients. Cancer Biomark 2022. [DOI: 10.1016/b978-0-12-824302-2.00010-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
27
|
González IA, Stewart DR, Schultz KAP, Field AP, Hill DA, Dehner LP. DICER1 tumor predisposition syndrome: an evolving story initiated with the pleuropulmonary blastoma. Mod Pathol 2022; 35:4-22. [PMID: 34599283 PMCID: PMC8695383 DOI: 10.1038/s41379-021-00905-8] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 08/09/2021] [Accepted: 08/09/2021] [Indexed: 02/06/2023]
Abstract
DICER1 syndrome (OMIM 606241, 601200) is a rare autosomal dominant familial tumor predisposition disorder with a heterozygous DICER1 germline mutation. The most common tumor seen clinically is the pleuropulmonary blastoma (PPB), a lung neoplasm of early childhood which is classified on its morphologic features into four types (IR, I, II and III) with tumor progression over time within the first 4-5 years of life from the prognostically favorable cystic type I to the unfavorable solid type III. Following the initial report of PPB, its association with other cystic neoplasms was demonstrated in family studies. The detection of the germline mutation in DICER1 provided the opportunity to identify and continue to recognize a number seemingly unrelated extrapulmonary neoplasms: Sertoli-Leydig cell tumor, gynandroblastoma, embryonal rhabdomyosarcomas of the cervix and other sites, multinodular goiter, differentiated and poorly differentiated thyroid carcinoma, cervical-thyroid teratoma, cystic nephroma-anaplastic sarcoma of kidney, nasal chondromesenchymal hamartoma, intestinal juvenile-like hamartomatous polyp, ciliary body medulloepithelioma, pituitary blastoma, pineoblastoma, primary central nervous system sarcoma, embryonal tumor with multilayered rosettes-like cerebellar tumor, PPB-like peritoneal sarcoma, DICER1-associated presacral malignant teratoid neoplasm and other non-neoplastic associations. Each of these neoplasms is characterized by a second somatic mutation in DICER1. In this review, we have summarized the salient clinicopathologic aspects of these tumors whose histopathologic features have several overlapping morphologic attributes particularly the primitive mesenchyme often with rhabdomyoblastic and chondroid differentiation and an uncommitted spindle cell pattern. Several of these tumors have an initial cystic stage from which there is progression to a high grade, complex patterned neoplasm. These pathologic findings in the appropriate clinical setting should serve to alert the pathologist to the possibility of a DICER1-associated neoplasm and initiate appropriate testing on the neoplasm and to alert the clinician about the concern for a DICER1 mutation.
Collapse
Affiliation(s)
- Iván A. González
- grid.239552.a0000 0001 0680 8770Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA USA
| | - Douglas R. Stewart
- grid.48336.3a0000 0004 1936 8075Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD USA
| | - Kris Ann P. Schultz
- International Pleuropulmonary Blastoma/DICER1 Registry, Children’s Minnesota, Minneapolis, MN USA ,Cancer and Blood Disorders, Children’s Minnesota, Minneapolis, MN USA
| | | | - D. Ashley Hill
- International Pleuropulmonary Blastoma/DICER1 Registry, Children’s Minnesota, Minneapolis, MN USA ,ResourcePath LLC, Sterling, VA USA ,grid.253615.60000 0004 1936 9510Division of Pathology, Children’s National Medical Center, George Washington University School of Medicine and Health Sciences, Washington, DC USA
| | - Louis P. Dehner
- International Pleuropulmonary Blastoma/DICER1 Registry, Children’s Minnesota, Minneapolis, MN USA ,grid.411019.cThe Lauren V. Ackerman Laboratory of Surgical Pathology, Barnes-Jewish and St. Louis Children’s Hospitals, Washington University Medical Center, St. Louis, MO USA
| |
Collapse
|
28
|
Geng G, Xiao Y, Shang Y, Zhang Y, Zhu F, Tang L, Peng F, Shen W, Jin Y, Yang Z, Li Q, Chen X. Naphthalenephenylalanine-phenylalanine-glycine-arginine-glycine-aspartic promotes self-assembly of nephron progenitor cells in decellularized scaffolds to construct bioengineered kidneys. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 134:112590. [DOI: 10.1016/j.msec.2021.112590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 11/09/2021] [Accepted: 11/29/2021] [Indexed: 10/19/2022]
|
29
|
Genetic Polymorphisms of the TGFB1 Signal Peptide and Promoter Region: Role in Wilms Tumor Susceptibility? J Kidney Cancer VHL 2021; 8:22-31. [PMID: 34722128 PMCID: PMC8532353 DOI: 10.15586/jkcvhl.v8i4.182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 08/19/2021] [Indexed: 11/18/2022] Open
Abstract
The aim of the present study was to investigate the rs1800468 (G-800A), rs1800469 (C-509T), rs1800470 (C29T), and rs1800471 (G74C) TGFB1 genetic polymorphisms and their haplotype structures in patients with Wilms Tumor (WT) and neoplasia-free controls. The genomic DNA was extracted from 35 WT patients and 160 neoplasia-free children, and the TGFB1 polymorphisms were genotyped by polymerase chain reaction, followed by restriction fragment length polymorphism. The haplotype structures were inferred, and permutation and logistic regression tests were performed to check for differences in haplotype distribution between the control and WT individuals. Positive associations were found in the recessive model for rs1800469 T allele (OR: 8.417; 95% CI: 3.177 to 22.297; P < 0.001) and for the rs1800470 C allele (OR: 3.000; 95% CI: 1.296 to 6.944; P = 0.01). Haplotype analysis revealed a significant negative association between GCTG and WT (OR: 0.236, 95% CI: 0.105 to 0.534; P = 0.0002); by contrast, the GTTG haplotype was associated with increased risk for WT (OR: 12.0; 95% CI: 4.202 to 34.270; P < 0.001). Furthermore, rs1800469 was negatively correlated with tumor size and a trend toward a positive correlation for capsular invasion was observed in the dominant model (Tau-b: −0.43, P = 0.02 and tau-b: 0.5, P = 0.06, respectively). This is the first study with rs1800468, rs1800469, rs1800470, and rs1800471 TGFB1 polymorphisms in WT, and our results suggest that the TGFB1 promoter and signal peptide region polymorphisms may be associated with WT susceptibility and clinical presentation.
Collapse
|
30
|
Grinberg S, Rous I, Cartry J. [An organoid biobank to help children with kidney cancer]. Med Sci (Paris) 2021; 37:811-813. [PMID: 34491194 DOI: 10.1051/medsci/2021126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Séverine Grinberg
- Master 2 Immunologie Translationnelle et Biothérapies (ITB), Parcours Immunologie, Mention BMC, Sorbonne Université, Paris, France
| | - Isaure Rous
- Master 2 Immunologie Translationnelle et Biothérapies (ITB), Parcours Immunologie, Mention BMC, Sorbonne Université, Paris, France
| | - Jérôme Cartry
- Inserm U1279, Collective Invasion (Fanny Jaulin), Institut Gustave Roussy, France
| |
Collapse
|
31
|
Maciaszek JL, Oak N, Nichols KE. Recent advances in Wilms' tumor predisposition. Hum Mol Genet 2021; 29:R138-R149. [PMID: 32412586 DOI: 10.1093/hmg/ddaa091] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/01/2020] [Accepted: 05/12/2020] [Indexed: 12/13/2022] Open
Abstract
Wilms' tumor (WT), the most common childhood kidney cancer, develops in association with an underlying germline predisposition in up to 15% of cases. Germline alterations affecting the WT1 gene and epigenetic alterations affecting the 11p15 locus are associated with a selective increase in WT risk. Nevertheless, WT also occurs in the context of more pleiotropic cancer predispositions, such as DICER1, Li-Fraumeni and Bloom syndrome, as well as Fanconi anemia. Recent germline genomic investigations have increased our understanding of the host genetic factors that influence WT risk, with sequencing of rare familial cases and large WT cohorts revealing an expanding array of predisposition genes and associated genetic conditions. Here, we describe evidence implicating WT1, the 11p15 locus, and the recently identified genes CTR9, REST and TRIM28 in WT predisposition. We discuss the clinical features, mode of inheritance and biological aspects of tumorigenesis, when known. Despite these described associations, many cases of familial WT remain unexplained. Continued investigations are needed to fully elucidate the landscape of germline genetic alterations in children with WT. Establishing a genetic diagnosis is imperative for WT families so that individuals harboring a predisposing germline variant can undergo surveillance, which should enable the early detection of tumors and use of less intensive treatments, thereby leading to improved overall outcomes.
Collapse
Affiliation(s)
- Jamie L Maciaszek
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Ninad Oak
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Kim E Nichols
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| |
Collapse
|
32
|
Wen-Jin C, Xiu-Wu P, Jian C, Da X, Jia-Xin C, Wei-Jie C, Lin-Hui W, Xin-Gang C. Study of cellular heterogeneity and differential dynamics of autophagy in human embryonic kidney development by single-cell RNA sequencing. Cancer Cell Int 2021; 21:460. [PMID: 34461918 PMCID: PMC8404318 DOI: 10.1186/s12935-021-02154-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 08/16/2021] [Indexed: 11/17/2022] Open
Abstract
Background Autophagy is believed to participate in embryonic development, but whether the expression of autophagy-associated genes undergoes changes during the development of human embryonic kidneys remains unknown. Methods In this work, we identified 36,151 human renal cells from embryonic kidneys of 9–18 gestational weeks in 16 major clusters by single-cell RNA sequencing (scRNA-seq), and detected 1350 autophagy-related genes in all fetal renal cells. The abundance of each cell cluster in Wilms tumor samples from scRNA-seq and GDC TARGET WT datasets was detected by CIBERSORTx. R package Monocle 3 was used to determine differentiation trajectories. Cyclone tool of R package scran was applied to calculate the cell cycle scores. R package SCENIC was used to investigate the transcriptional regulons. The FindMarkers tool from Seurat was used to calculate DEGs. GSVA was used to perform gene set enrichment analyses. CellphoneDB was utilized to analyze intercellular communication. Results It was found that cells in the 13th gestational week showed the lowest transcriptional level in each cluster in all stages. Nephron progenitors could be divided into four subgroups with diverse levels of autophagy corresponding to different SIX2 expressions. SSBpod (podocyte precursors) could differentiate into four types of podocytes (Pod), and autophagy-related regulation was involved in this process. Pseudotime analysis showed that interstitial progenitor cells (IPCs) potentially possessed two primitive directions of differentiation to interstitial cells with different expressions of autophagy. It was found that NPCs, pretubular aggregates and interstitial cell clusters had high abundance in Wilms tumor as compared with para-tumor samples with active intercellular communication. Conclusions All these findings suggest that autophagy may be involved in the development and cellular heterogeneity of early human fetal kidneys. In addition, part of Wilms tumor cancer cells possess the characteristics of some fetal renal cell clusters. Graphical abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-02154-w.
Collapse
Affiliation(s)
- Chen Wen-Jin
- Department of Urology, The Third Affiliated Hospital of Second Military Medical University, 700 North Moyu Road, Shanghai, 201805, China
| | - Pan Xiu-Wu
- Department of Urology, The Third Affiliated Hospital of Second Military Medical University, 700 North Moyu Road, Shanghai, 201805, China.,Department of Urology, Xinhua Hospital, Shanghai Jiaotong University, School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Chu Jian
- Department of Urology, The Third Affiliated Hospital of Second Military Medical University, 700 North Moyu Road, Shanghai, 201805, China.,Department of Urology, Gongli Hospital of Second Military Medical University, 219 Miaopu Road, Shanghai, 200135, China
| | - Xu Da
- Department of Urology, The Third Affiliated Hospital of Second Military Medical University, 700 North Moyu Road, Shanghai, 201805, China
| | - Chen Jia-Xin
- Department of Urology, The Third Affiliated Hospital of Second Military Medical University, 700 North Moyu Road, Shanghai, 201805, China
| | - Chen Wei-Jie
- Department of Urology, The Third Affiliated Hospital of Second Military Medical University, 700 North Moyu Road, Shanghai, 201805, China
| | - Wang Lin-Hui
- Department of Urology, Changzheng Hospital of Second Military Medical University, 415 Fengyang Road, Shanghai, 200003, China.
| | - Cui Xin-Gang
- Department of Urology, The Third Affiliated Hospital of Second Military Medical University, 700 North Moyu Road, Shanghai, 201805, China. .,Department of Urology, Xinhua Hospital, Shanghai Jiaotong University, School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China.
| |
Collapse
|
33
|
Attali-Padael Y, Armon L, Urbach A. Apoptosis induction by the stem cell factor LIN28A. Biol Cell 2021; 113:450-457. [PMID: 34437724 DOI: 10.1111/boc.202100011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 08/12/2021] [Accepted: 08/20/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND INFORMATION Lin28A and its paralog Lin28B are RNA binding proteins expressed in stem and progenitor cells, regulating the balance between their proliferation and differentiation. In-vivo and in-vitro experiments have shown that overexpression of these genes leads to abnormal cell proliferation, which results in many cases in cell transformation and tumor formation. RESULTS Here we show, for the first time, that Lin28A overexpression can also lead to the opposite effect, i.e. apoptosis induction. We further demonstrate that this effect is specific to Lin28A but not to Lin28B and that it is mediated via the Let-7 independent pathway in a complex mechanism that involves at least several proteins. CONCLUSIONS AND SIGNIFICANCE This unexpected observation suggests that cell fate regulation by Lin28 is dependent on a specific cellular/genetic context. Unraveling the cellular and molecular mechanisms underlying this Lin28A overexpression effect may pave the way for novel tumor therapeutic strategies, as Lin28 is commonly expressed in many types of tumors but not in most normal adult cells.
Collapse
Affiliation(s)
- Yael Attali-Padael
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Leah Armon
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Achia Urbach
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| |
Collapse
|
34
|
Yao Y, Chai X, Gong C, Zou L. WT1 inhibits AML cell proliferation in a p53-dependent manner. Cell Cycle 2021; 20:1552-1560. [PMID: 34288813 DOI: 10.1080/15384101.2021.1951938] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
WT1 has been reported to function as an oncogene and a tumor suppressor in acute myeloid leukemia (AML). The molecular mechanisms have not yet been fully elucidated. Here, we report that p53, served as a tumor suppressor, plays a critical role in regulating the function of WT1 in AML. For details, we performed a meta-analysis on 1131 AML cases, showing that WT1 gene mutation and TP53 gene exhibited a mutually exclusive predisposition in AML. p53 can be recruited to the promoter region of WT1's target genes to modulate their expression by physically interacting with WT1. The AML-derived p53 mutation (p53R248Q) can disrupt the interaction between WT1 and p53, resulting in the loss of modulation of WT1's target genes. Furthermore, wild-type p53 maintained the anti-proliferation activity of WT1 in AML cells. In contrast, WT1 promoted AML cell proliferation in the absence of p53 (or mutated p53). In conclusion, we demonstrated a novel explanation of the controversial function of WT1 in AML. These results provided a mechanism by which WT1 inhibited AML cell proliferation in a p53-dependent manner.
Collapse
Affiliation(s)
- Yiyun Yao
- Shanghai Ninth People's Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Xingxing Chai
- Department of Hematology, The Second People's Hospital of Lianyungang City, Jiangsu 222000, China
| | - Chen Gong
- Department of Geriatric Medicine, The Second People's Hospital of Lianyungang City, Jiangsu 222000, China
| | - Lifang Zou
- Shanghai Ninth People's Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| |
Collapse
|
35
|
Every Beat You Take-The Wilms' Tumor Suppressor WT1 and the Heart. Int J Mol Sci 2021; 22:ijms22147675. [PMID: 34299295 PMCID: PMC8306835 DOI: 10.3390/ijms22147675] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 07/06/2021] [Accepted: 07/16/2021] [Indexed: 12/23/2022] Open
Abstract
Nearly three decades ago, the Wilms’ tumor suppressor Wt1 was identified as a crucial regulator of heart development. Wt1 is a zinc finger transcription factor with multiple biological functions, implicated in the development of several organ systems, among them cardiovascular structures. This review summarizes the results from many research groups which allowed to establish a relevant function for Wt1 in cardiac development and disease. During development, Wt1 is involved in fundamental processes as the formation of the epicardium, epicardial epithelial-mesenchymal transition, coronary vessel development, valve formation, organization of the cardiac autonomous nervous system, and formation of the cardiac ventricles. Wt1 is further implicated in cardiac disease and repair in adult life. We summarize here the current knowledge about expression and function of Wt1 in heart development and disease and point out controversies to further stimulate additional research in the areas of cardiac development and pathophysiology. As re-activation of developmental programs is considered as paradigm for regeneration in response to injury, understanding of these processes and the molecules involved therein is essential for the development of therapeutic strategies, which we discuss on the example of WT1.
Collapse
|
36
|
Ceylan O, Caglar O. Importance of COX-2 expression in Wilms' tumour: A preliminary study. Int J Clin Pract 2021; 75:e14146. [PMID: 33713389 DOI: 10.1111/ijcp.14146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/10/2021] [Accepted: 03/07/2021] [Indexed: 11/29/2022] Open
Abstract
OBJECTIVES In Wilms' tumour (WT), secondary malignancies caused by the side effects of intensive treatments remain one of the important problems. Therefore, there is a need for new studies to identify low- and high-risk groups for WT and to improve the treatment regimens of children in the low-risk group. In our study, we aimed to determine the prognostic significance of the cyclooxygenase-2 (COX-2) biomarker in WT. MATERIALS AND METHODS Our study included 24 patients diagnosed with WT between January 2010 and December 2019. The correlation between COX-2 expression and significant prognostic parameters was investigated by studying the COX-2 antibody using the immunohistochemical method. RESULTS COX-2 expression was observed in 22 of the patients and it was more evident in the epithelial component. No significant correlation was observed between COX-2 positivity and prognostic parameters. There was also no statistically significant difference between the two groups regarding survival (P = .563). CONCLUSIONS In our study, no significant relationship was found between significant prognostic parameters and COX-2 expression. Since COX-2 expression was observed in almost all patients, we consider that the COX-2 pathway is effective during the development phase of WT.
Collapse
Affiliation(s)
- Onur Ceylan
- Department of Pathology, Ataturk University, Faculty of Medicine Erzurum, Erzurum, Turkey
| | - Ozgur Caglar
- Department of Pediatric Surgery, Ataturk University, Faculty of Medicine, Erzurum, Turkey
| |
Collapse
|
37
|
Chen L, Yin L, Qi Z, Li J, Wang X, Ma K, Liu X. Gene expression-based immune infiltration analyses of renal cancer and their associations with survival outcome. BMC Cancer 2021; 21:595. [PMID: 34030645 PMCID: PMC8146654 DOI: 10.1186/s12885-021-08244-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 04/23/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Renal cancer is a common malignant tumor with an increasing incidence rate. METHODS In this study, based on the gene expression profiles, we analyzed the compositions of tumor-infiltrating immune cells (TIICs) in renal cancer and paracancerous samples using CIBERSORT. The proportions of 22 TIICs subsets in 122 paired renal carcinoma and paracancerous samples, and 224 Wilms tumor (WT) samples varied between intragroup and intergroup. RESULTS After analyzed the difference of TIICs composition between renal cancer and paired paracancerous samples, we found that M0 macrophages and CD8 T cells were significantly elevated, while naive B cells were significantly decreased in renal cancer samples compared with paracancerous samples. Survival analysis showed that high overall TIICs proportion, the low proportion of resting mast cells and the high proportion of activated memory CD4 T cells were associated with poor prognosis of renal cancer patients. In addition, 3 clusters were identified by hierarchical clustering analysis, and they presented a distinct prognosis. Cluster 1 had superior survival outcomes, while cluster 2 had an inferior survival outcome. CONCLUSIONS Our study indicated that overall TIICs proportion, certain TIICs subset proportion, including resting mast cells and activated memory CD4 T cells, and distinct cluster patterns were associated with the prognosis of renal cancer, which was significant for the clinical surveillance and treatment of renal cancer.
Collapse
Affiliation(s)
- Lei Chen
- Department of Pediatric Surgery, Cangzhou Central Hospital, No.16 Xinhua West Road, Cangzhou, 061000, Hebei, China
| | - Liang Yin
- Department of Pediatric Surgery, Cangzhou Central Hospital, No.16 Xinhua West Road, Cangzhou, 061000, Hebei, China
| | - Zilong Qi
- Department of Pediatric Surgery, Cangzhou Central Hospital, No.16 Xinhua West Road, Cangzhou, 061000, Hebei, China
| | - Jinmin Li
- Department of Pediatric Surgery, Cangzhou Central Hospital, No.16 Xinhua West Road, Cangzhou, 061000, Hebei, China
| | - Xinning Wang
- Department of Pediatric Surgery, Cangzhou Central Hospital, No.16 Xinhua West Road, Cangzhou, 061000, Hebei, China
| | - Kun Ma
- Department of Pediatric Surgery, Cangzhou Central Hospital, No.16 Xinhua West Road, Cangzhou, 061000, Hebei, China
| | - Xiangyang Liu
- Department of Pediatric Surgery, Cangzhou Central Hospital, No.16 Xinhua West Road, Cangzhou, 061000, Hebei, China.
| |
Collapse
|
38
|
Prognostic Gene Expression, Stemness and Immune Microenvironment in Pediatric Tumors. Cancers (Basel) 2021; 13:cancers13040854. [PMID: 33670534 PMCID: PMC7922568 DOI: 10.3390/cancers13040854] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 02/15/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Tumors in children and young adults are rare and diagnostically distinct from those occurring in older patients. They frequently arise from developing cells, resembling stem cells, which may explain some of the clinical and biologic differences observed. The aim of this retrospective transcriptome study was to investigate the prognostic landscape, immune tumor microenvironment (TME) and stemness in a cohort of 4068 transcriptomes of such tumors. We find that patients’ prognosis correlates with distinct gene expression patterns similar to adult tumor types. Stemness defined by a computational stemness score (mRNAsi) correlates with clinical and molecular parameters that is distinct for each tumor type. In Wilms tumors that recapitulate normal kidney development microscopically, stemness correlates with distinct patterns of immune cell infiltration by transcriptome analysis and by cell localization in tumor tissue. Abstract Pediatric tumors frequently arise from embryonal cells, often displaying a stem cell-like (“small round blue”) morphology in tissue sections. Because recently “stemness” has been associated with a poor immune response in tumors, we investigated the association of prognostic gene expression, stemness and the immune microenvironment systematically using transcriptomes of 4068 tumors occurring mostly at the pediatric and young adult age. While the prognostic landscape of gene expression (PRECOG) and infiltrating immune cell types (CIBERSORT) is similar to that of tumor entities occurring mainly in adults, the patterns are distinct for each diagnostic entity. A high stemness score (mRNAsi) correlates with clinical and morphologic subtype in Wilms tumors, neuroblastomas, synovial sarcomas, atypical teratoid rhabdoid tumors and germ cell tumors. In neuroblastomas, a high mRNAsi is associated with shortened overall survival. In Wilms tumors a high mRNAsi correlates with blastemal morphology, whereas tumors with predominant epithelial or stromal differentiation have a low mRNAsi and a high percentage of M2 type macrophages. This could be validated in Wilms tumor tissue (n = 78). Here, blastemal areas are low in M2 macrophage infiltrates, while nearby stromal differentiated areas contain abundant M2 macrophages, suggesting local microanatomic regulation of the immune response.
Collapse
|
39
|
Li W, Hua R, Wang M, Zhang D, Zhu J, Zhang S, Yang Y, Cheng J, Zhou H, Zhang J, He J. H19 gene polymorphisms and Wilms tumor risk in Chinese children: a four-center case-control study. Mol Genet Genomic Med 2021; 9:e1584. [PMID: 33403826 PMCID: PMC8077085 DOI: 10.1002/mgg3.1584] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 11/05/2020] [Accepted: 12/15/2020] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Wilms tumor is the most common pediatric renal cancer. However, genetic bases behind Wilms tumor remain largely unknown. H19 is a critical maternally imprinted gene. Previous studies indicated that single nucleotide polymorphisms (SNPs) in the H19 can modify the risk of several human malignancies. Epigenetic errors at the H19 locus lead to biallelic silencing in Wilms tumors. Genetic variations in the H19 may be related to Wilms tumor susceptibility. METHODS We conducted a four-center study to investigate whether H19 SNP was a predisposing factor to Wilms tumor. Three polymorphisms in the H19 (rs2839698 G > A, rs3024270 C > G, rs217727 G > A) were genotyped in 355 cases and 1070 cancer-free controls, using Taqman method. Odds ratios (ORs) and 95% confidence intervals (CIs) were calculated to evaluate the strength of the associations. RESULTS We found that all of these three polymorphisms were significantly associated with Wilms tumor risk alterations. The rs2839698 G > A polymorphism (AG vs. GG: adjusted OR = 0.74, 95% CI = 0.57-0.96, p = 0.024; AA vs. GG: adjusted OR = 1.52, 95% CI = 1.05-2.22, p = 0.027), the rs3024270 C > G polymorphism (CG vs. CC: adjusted OR = 0.61, 95% CI = 0.46-0.81, p = 0.0007; and the rs217727 polymorphism (AG vs. GG: adjusted OR = 0.76, 95% CI = 0.58-0.99, p = 0.035). The Carriers of 1, 2, and 1-2 risk genotypes were inclined to develop Wilms tumor compared with those without risk genotype (adjusted OR = 1.36, 95% CI = 1.02-1.80, p = 0.037; adjusted OR = 1.84, 95% CI = 1.27-2.67, p = 0.001; adjusted OR = 1.50, 95% CI = 1.17-1.92, p = 0.002, respectively). The stratified analysis further revealed that rs2839698 AA, rs217727 AA, and 1-2 risk genotypes could strongly increase Wilms tumor risk among children above 18 months of age, males, and with clinical stage I+II disease. CONCLUSION Our findings indicate that genetic variations in the H19 may confer Wilms tumor risk.
Collapse
Affiliation(s)
- Wenya Li
- Department of Pediatric SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Rui‐Xi Hua
- Department of OncologyThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
- Department of Pediatric SurgeryGuangzhou Institute of PediatricsGuangdong Provincial Key Laboratory of Research in Structural Birth Defect DiseaseGuangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhouChina
| | - Mi Wang
- Department of Pediatric SurgeryGuangzhou Institute of PediatricsGuangdong Provincial Key Laboratory of Research in Structural Birth Defect DiseaseGuangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhouChina
| | - Da Zhang
- Department of Pediatric SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Jinhong Zhu
- Department of Pediatric SurgeryGuangzhou Institute of PediatricsGuangdong Provincial Key Laboratory of Research in Structural Birth Defect DiseaseGuangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhouChina
- Department of Clinical LaboratoryBiobankHarbin Medical University Cancer HospitalHarbinChina
| | - Songyang Zhang
- Department of Pediatric SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Yang Yang
- Department of Pediatric SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Jiwen Cheng
- Department of Pediatric Surgerythe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Haixia Zhou
- Department of HematologyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Jiao Zhang
- Department of Pediatric SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Jing He
- Department of Pediatric SurgeryGuangzhou Institute of PediatricsGuangdong Provincial Key Laboratory of Research in Structural Birth Defect DiseaseGuangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhouChina
| |
Collapse
|
40
|
Wang Z, Zhuo Z, Li L, Hua RX, Li L, Zhang J, Cheng J, Zhou H, Li S, He J, Yan S. The contribution of YTHDF2 gene rs3738067 A>G to the Wilms tumor susceptibility. J Cancer 2021; 12:6165-6169. [PMID: 34539889 PMCID: PMC8425210 DOI: 10.7150/jca.62154] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 08/09/2021] [Indexed: 02/07/2023] Open
Abstract
YTHDF2 is responsible for maintaining the dynamic N6-methyladenosine (m6A) modification balance and influences a variety of cancers. We tested whether YTHDF2 gene rs3738067 A>G polymorphism is related to Wilms tumor by genotyping samples of Chinese children (450 cases and 1317 controls). However, the rs3738067 A>G polymorphism showed no statistical significance with Wilms tumor susceptibility. Stratification analysis also revealed that there was no remarkable association of rs3738067 variant AG/GG genotype with Wilms tumor risk in every subgroup (age, gender, and clinical stages). In all, the results indicated YTHDF2 gene rs3738067 A>G polymorphism could not alter Wilms tumor risk significantly.
Collapse
Affiliation(s)
- Zhiyuan Wang
- Department of Pathology, The First Affiliated Hospital of Kunming Medical University, Kunming 650031, Yunnan, China
| | - Zhenjian Zhuo
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Linyan Li
- Department of Clinical Laboratory, Yunnan Key Laboratory of Laboratory Medicine, the First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China
| | - Rui-Xi Hua
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Li Li
- Kunming Key Laboratory of Children Infection and Immunity, Yunnan Key Laboratory of Children's Major Disease Research, Yunnan Institute of Pediatrics Research, Yunnan Medical Center for Pediatric Diseases, Kunming Children's Hospital, Kunming 650228, Yunnan, China
| | - Jiao Zhang
- Department of Pediatric Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Jiwen Cheng
- Department of Pediatric Surgery, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi, China
| | - Haixia Zhou
- Department of Hematology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang, China
| | - Suhong Li
- Department of Pathology, Children Hospital and Women Health Center of Shanxi, Taiyuan 030013, Shannxi, China
| | - Jing He
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
- ✉ Corresponding authors: Shan Yan, Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Biomedical Engineering Research Center, Kunming Medical University, No. 1168 Chunrongxi Road, Kunming 650500, Yunnan, China, E-mail: ; or Jing He, Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Guangzhou 510623, Guangdong, China, E-mail:
| | - Shan Yan
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Biomedical Engineering Research Center, Kunming Medical University, Kunming 650500, Yunnan, China
- ✉ Corresponding authors: Shan Yan, Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Biomedical Engineering Research Center, Kunming Medical University, No. 1168 Chunrongxi Road, Kunming 650500, Yunnan, China, E-mail: ; or Jing He, Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Guangzhou 510623, Guangdong, China, E-mail:
| |
Collapse
|
41
|
Lin A, Zhou M, Hua RX, Zhang J, Zhou H, Li S, Cheng J, Xia H, Fu W, He J. METTL3 polymorphisms and Wilms tumor susceptibility in Chinese children: A five-center case-control study. J Gene Med 2020; 22:e3255. [PMID: 32716082 DOI: 10.1002/jgm.3255] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/21/2020] [Accepted: 07/21/2020] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Wilms tumor is a common pediatric tumor worldwide. Methyltransferase like 3 (METTL3) is a core gene of the N6 -methyladenosine (m6 A) modification that widely affects the transcription of tumor-related genes in eukaryotes. METTL3 has been extensively investigated in various tumors but not Wilms tumor. METHODS We describe a five-center case-control study with 414 patients and 1199 controls aiming to explore the associations between METTL3 polymorphisms (rs1061026 T>G, rs1061027 C>A, rs1139130 A>G and rs1263801 G>C) and Wilms tumor susceptibility. A TaqMan real-time polymerase chain reaction was performed for genotyping. Odds ratios (ORs) and 95% confidence intervals (CIs) were reported as evaluation indicators to determine any associations. RESULTS Referring to the preliminary analysis results, protective genotypes were identified as rs1061026 TG/GG, rs1061027 CA/AA, rs1139130 GG and rs1263801 GC/CC. The children with three protective genotypes were less likely to develop Wilms tumor than children without protective genotypes (adjusted OR = 0.68, 95% CI = 0.46-0.999, p = 0.0496). Similarly, stratified analysis of the subgroup aged > 18 months, carrying 3 or 4 protective genotypes, was a protective factor for Wilms tumor compared to carrying 0-2 protective genotypes (adjusted OR = 0.59 95% CI = 0.39-0.91, p = 0.016). However, we did not observe any other significant results. CONCLUSIONS The combined effect of METTL3 polymorphisms reduce Wilms tumor susceptibility in Chinese children. This conclusion requires further verification.
Collapse
Affiliation(s)
- Ao Lin
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Mingming Zhou
- Department of Clinical Laboratory, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Zhejiang, Hangzhou, China
| | - Rui-Xi Hua
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jiao Zhang
- Department of Pediatric Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Haixia Zhou
- Department of Hematology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Suhong Li
- Department of Pathology, Children Hospital and Women Health Center of Shanxi, Taiyuan, Shannxi, China
| | - Jiwen Cheng
- Department of Pediatric Surgery, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Huimin Xia
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Wen Fu
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jing He
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
42
|
Nishikawa T, Wojciak JM, Dyson HJ, Wright PE. RNA Binding by the KTS Splice Variants of Wilms' Tumor Suppressor Protein WT1. Biochemistry 2020; 59:3889-3901. [PMID: 32955251 DOI: 10.1021/acs.biochem.0c00602] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Wilms' tumor suppressor protein WT1 regulates the expression of multiple genes through binding of the Cys2-His2 zinc finger domain to promoter sites. WT1 has also been proposed to be involved in post-transcriptional regulation, by binding to RNA using the same set of zinc fingers. WT1 has two major splice variants, where the Lys-Thr-Ser (KTS) tripeptide is inserted into the linker between the third and fourth zinc fingers. To obtain insights into the mechanism by which the different WT1 splice variants recognize both DNA and RNA, we have determined the solution structure of the WT1 (-KTS) zinc finger domain in complex with a 29mer stem-loop RNA. Zinc fingers 1-3 bind in a widened major groove favored by the presence of a bulge nucleotide in the double-stranded helical stem. Fingers 2 and 3 make specific contacts with the nucleobases in a conserved AUGG sequence in the helical stem. Nuclear magnetic resonance chemical shift mapping and relaxation analysis show that fingers 1-3 of the two splice variants (-KTS and +KTS) of WT1 form similar complexes with RNA. Finger 4 of the -KTS isoform interacts weakly with the RNA loop, an interaction that is abrogated in the +KTS isoform, and both isoforms bind with similar affinity to the RNA. In contrast, finger 4 is required for high-affinity binding to DNA and insertion of KTS into the linker of fingers 3 and 4 abrogates DNA binding. While finger 1 is required for RNA binding, it is dispensable for binding to consensus DNA sites.
Collapse
Affiliation(s)
- Tadateru Nishikawa
- Department of Integrative Structural and Computational Biology and Skaggs Institute of Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Jonathan M Wojciak
- Department of Integrative Structural and Computational Biology and Skaggs Institute of Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - H Jane Dyson
- Department of Integrative Structural and Computational Biology and Skaggs Institute of Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Peter E Wright
- Department of Integrative Structural and Computational Biology and Skaggs Institute of Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| |
Collapse
|
43
|
Li JC, Chang X, Chen Y, Li XZ, Zhang XL, Yang SM, Hu CJ, Zhang H. Loss of the Tumor Suppressor HACE1 Contributes to Cancer Progression. Curr Drug Targets 2020; 20:1018-1028. [PMID: 30827236 DOI: 10.2174/1389450120666190227184654] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 01/20/2019] [Accepted: 02/14/2019] [Indexed: 12/15/2022]
Abstract
HACE1 belongs to the family of HECT domain-containing E3 ligases, which plays an important role in the occurrence, invasion and metastatic process in many human malignancies. HACE1 is a tumor suppressor gene that is reduced in most cancer tissues compared to adjacent normal tissue. The loss or knocking out of HACE1 leads to enhanced tumor growth, invasion, and metastasis; in contrast, the overexpression of HACE1 can inhibit the development of tumors. Hypermethylation reduces the expression of HACE1, thereby promoting tumor development. HACE1 can inhibit the development of inflammation or tumors via the ubiquitination pathway. Therefore, HACE1 may be a potential therapeutic target, providing new strategies for disease prevention and treatment.
Collapse
Affiliation(s)
- Jun-Chen Li
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.,Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Xing Chang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Yang Chen
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Xin-Zhe Li
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Xiang-Lian Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Guangxi 530021, China
| | - Shi-Ming Yang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Chang-Jiang Hu
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Hao Zhang
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| |
Collapse
|
44
|
Drake KA, Chaney CP, Das A, Roy P, Kwartler CS, Rakheja D, Carroll TJ. Stromal β-catenin activation impacts nephron progenitor differentiation in the developing kidney and may contribute to Wilms tumor. Development 2020; 147:dev189597. [PMID: 32541007 PMCID: PMC7406317 DOI: 10.1242/dev.189597] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 06/02/2020] [Indexed: 02/03/2023]
Abstract
Wilms' tumor (WT) morphologically resembles the embryonic kidney, consisting of blastema, epithelial and stromal components, suggesting tumors arise from the dysregulation of normal development. β-Catenin activation is observed in a significant proportion of WTs; however, much remains to be understood about how it contributes to tumorigenesis. Although activating β-catenin mutations are observed in both blastema and stromal components of WT, current models assume that activation in the blastemal lineage is causal. Paradoxically, studies performed in mice suggest that activation of β-catenin in the nephrogenic lineage results in loss of nephron progenitor cell (NPC) renewal, a phenotype opposite to WT. Here, we show that activation of β-catenin in the stromal lineage non-autonomously prevents the differentiation of NPCs. Comparisons of the transcriptomes of kidneys expressing an activated allele of β-catenin in the stromal or nephron progenitor cells reveals that human WT more closely resembles the stromal-lineage mutants. These findings suggest that stromal β-catenin activation results in histological and molecular features of human WT, providing insights into how alterations in the stromal microenvironment may play an active role in tumorigenesis.
Collapse
Affiliation(s)
- Keri A Drake
- Division of Pediatric Nephrology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Christopher P Chaney
- Department of Molecular Biology and Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Amrita Das
- Amgen, Inc., San Francisco, CA 94080, USA
| | - Priti Roy
- Department of Ophthalmology and Visual Sciences, Chicago, IL 60612, USA
| | - Callie S Kwartler
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Dinesh Rakheja
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Thomas J Carroll
- Department of Molecular Biology and Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
45
|
Li Y, Gong H, Ding J, Zhao F, Du J, Wan J, Zhang J, Liu S, Li J, Wang L, Zhou B. Inhibition of GSK3 Represses the Expression of Retinoic Acid Synthetic Enzyme ALDH1A2 via Wnt/β-Catenin Signaling in WiT49 Cells. Front Cell Dev Biol 2020; 8:94. [PMID: 32258025 PMCID: PMC7092725 DOI: 10.3389/fcell.2020.00094] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 02/04/2020] [Indexed: 02/02/2023] Open
Abstract
Organogenesis, including renal development, requires an appropriate retinoic acid concentration, which is established by differential expression of aldehyde dehydrogenase 1 family member A2 (ALDH1A2) and cytochrome P450 family 26 subfamily A/B/C member 1 (CYP26A1/B1/C1). In the fetal kidney, ALDH1A2 expresses in the developing stroma and renal vesicle and its derivatives but does not present in the ureteric bud. It remains unclear what may contribute to this expression pattern. Here we show that the glycogen synthase kinase 3 alpha/beta (GSK3A/B) inhibitor CHIR99021 significantly represses ALDH1A2 expression in WiT49, which is a Wilms’ tumor cell line that exhibits “triphasic” differential potential and is used as a fetal kidney cell model. CHIR99021 fails to suppress ALDH1A2 as β-catenin is inhibited, suggesting that the downregulation of ALDH1A2 by CHIR99021 is through Wnt/β-catenin signaling. Ectopic expression of mouse Wnt1, Wnt3a, Wnt4, and Wnt9b represses ALDH1A2 expression in WiT49 cells. Using immunohistochemistry, we show an inverse correlation of Aldh1a2 expression with β-catenin in rat E18.5 kidney. ChIP demonstrated that β-catenin is recruited to the ALDH1A2 promoter, the conserved intron1G, and another site within intron 1 of ALDH1A2. Using a luciferase assay, we further show that the ALDH1A2 promoter and the intron1G element are involved in the repression of ALDH1A2 expression by CHIR99021. Our work demonstrates that ALDH1A2 expression can be directly repressed by the Wnt/β-catenin signaling in fetal kidney cells, suggesting that Wnt/β-catenin may play a role in maintaining the expression pattern of ALDH1A2 in the fetal kidney, thus controlling the availability and localization of retinoic acid and regulating aspects of kidney development.
Collapse
Affiliation(s)
- Yifan Li
- Central Laboratory, Huazhong University of Science and Technology Union Shenzhen Hospital and the Affiliated Shenzhen Sixth Hospital of Guangdong Medical University, Shenzhen, China.,Shenzhen Key Lab of Endogenous Infection, Huazhong University of Science and Technology Union Shenzhen Hospital and the Affiliated Shenzhen Sixth Hospital of Guangdong Medical University, Shenzhen, China
| | - Hui Gong
- Central Laboratory, Huazhong University of Science and Technology Union Shenzhen Hospital and the Affiliated Shenzhen Sixth Hospital of Guangdong Medical University, Shenzhen, China
| | - Jiangfeng Ding
- Department of Stomotology, Huazhong University of Science and Technology Union Shenzhen Hospital and the Affiliated Shenzhen Sixth Hospital of Guangdong Medical University, Shenzhen, China
| | - Fujuan Zhao
- Department of Pathology, Huazhong University of Science and Technology Union Shenzhen Hospital and the Affiliated Shenzhen Sixth Hospital of Guangdong Medical University, Shenzhen, China
| | - Jihui Du
- Central Laboratory, Huazhong University of Science and Technology Union Shenzhen Hospital and the Affiliated Shenzhen Sixth Hospital of Guangdong Medical University, Shenzhen, China
| | - Jun Wan
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China.,Division of Life Science, The Hong Kong University of Science and Technology, Kowloon, Hong Kong
| | - Juan Zhang
- Department of Pathology, Huazhong University of Science and Technology Union Shenzhen Hospital and the Affiliated Shenzhen Sixth Hospital of Guangdong Medical University, Shenzhen, China
| | - Shaoxiong Liu
- Department of Pathology, Huazhong University of Science and Technology Union Shenzhen Hospital and the Affiliated Shenzhen Sixth Hospital of Guangdong Medical University, Shenzhen, China
| | - Jing Li
- Department of Endocrinology, Huazhong University of Science and Technology Union Shenzhen Hospital and the Affiliated Shenzhen Sixth Hospital of Guangdong Medical University, Shenzhen, China
| | - Lei Wang
- Central Laboratory, Huazhong University of Science and Technology Union Shenzhen Hospital and the Affiliated Shenzhen Sixth Hospital of Guangdong Medical University, Shenzhen, China
| | - Bei Zhou
- Central Laboratory, Huazhong University of Science and Technology Union Shenzhen Hospital and the Affiliated Shenzhen Sixth Hospital of Guangdong Medical University, Shenzhen, China
| |
Collapse
|
46
|
Calandrini C, Schutgens F, Oka R, Margaritis T, Candelli T, Mathijsen L, Ammerlaan C, van Ineveld RL, Derakhshan S, de Haan S, Dolman E, Lijnzaad P, Custers L, Begthel H, Kerstens HHD, Visser LL, Rookmaaker M, Verhaar M, Tytgat GAM, Kemmeren P, de Krijger RR, Al-Saadi R, Pritchard-Jones K, Kool M, Rios AC, van den Heuvel-Eibrink MM, Molenaar JJ, van Boxtel R, Holstege FCP, Clevers H, Drost J. An organoid biobank for childhood kidney cancers that captures disease and tissue heterogeneity. Nat Commun 2020; 11:1310. [PMID: 32161258 PMCID: PMC7066173 DOI: 10.1038/s41467-020-15155-6] [Citation(s) in RCA: 176] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 02/21/2020] [Indexed: 01/02/2023] Open
Abstract
Kidney tumours are among the most common solid tumours in children, comprising distinct subtypes differing in many aspects, including cell-of-origin, genetics, and pathology. Pre-clinical cell models capturing the disease heterogeneity are currently lacking. Here, we describe the first paediatric cancer organoid biobank. It contains tumour and matching normal kidney organoids from over 50 children with different subtypes of kidney cancer, including Wilms tumours, malignant rhabdoid tumours, renal cell carcinomas, and congenital mesoblastic nephromas. Paediatric kidney tumour organoids retain key properties of native tumours, useful for revealing patient-specific drug sensitivities. Using single cell RNA-sequencing and high resolution 3D imaging, we further demonstrate that organoid cultures derived from Wilms tumours consist of multiple different cell types, including epithelial, stromal and blastemal-like cells. Our organoid biobank captures the heterogeneity of paediatric kidney tumours, providing a representative collection of well-characterised models for basic cancer research, drug-screening and personalised medicine. Pre-clinical cell culture models capturing the heterogeneity of childhood kidney tumours are limited. Here, the authors establish and characterise an organoid biobank of tumour and matched normal organoid cultures from over 50 children with different subtypes of kidney cancer.
Collapse
Affiliation(s)
- Camilla Calandrini
- Oncode Institute, Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands
| | - Frans Schutgens
- Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center, Uppsalalaan 8, 3584 CT, Utrecht, The Netherlands.,University Medical Center, Department of Nephrology and Hypertension, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Rurika Oka
- Oncode Institute, Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands
| | - Thanasis Margaritis
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands
| | - Tito Candelli
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands
| | - Luka Mathijsen
- Oncode Institute, Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands
| | - Carola Ammerlaan
- Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center, Uppsalalaan 8, 3584 CT, Utrecht, The Netherlands.,University Medical Center, Department of Nephrology and Hypertension, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Ravian L van Ineveld
- Oncode Institute, Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands
| | - Sepide Derakhshan
- Oncode Institute, Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands
| | - Sanne de Haan
- Oncode Institute, Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands
| | - Emmy Dolman
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands
| | - Philip Lijnzaad
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands
| | - Lars Custers
- Oncode Institute, Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands
| | - Harry Begthel
- Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center, Uppsalalaan 8, 3584 CT, Utrecht, The Netherlands
| | - Hindrik H D Kerstens
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands
| | - Lindy L Visser
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands
| | - Maarten Rookmaaker
- University Medical Center, Department of Nephrology and Hypertension, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Marianne Verhaar
- University Medical Center, Department of Nephrology and Hypertension, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Godelieve A M Tytgat
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands
| | - Patrick Kemmeren
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands
| | - Ronald R de Krijger
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands.,University Medical Center, Department of Pathology, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Reem Al-Saadi
- University College London, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
| | - Kathy Pritchard-Jones
- University College London, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
| | - Marcel Kool
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands.,Hopp Children's Cancer Center (KiTZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany.,Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Research Consortium (DKTK), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Anne C Rios
- Oncode Institute, Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands
| | | | - Jan J Molenaar
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands
| | - Ruben van Boxtel
- Oncode Institute, Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands
| | - Frank C P Holstege
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands
| | - Hans Clevers
- Oncode Institute, Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands.,Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center, Uppsalalaan 8, 3584 CT, Utrecht, The Netherlands
| | - Jarno Drost
- Oncode Institute, Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands.
| |
Collapse
|
47
|
Zhang X, Liu C, Li H, Guo L. Effects of miR-21 on proliferation and apoptosis of WT cells via PTEN/Akt pathway. Exp Ther Med 2020; 19:2155-2160. [PMID: 32104279 PMCID: PMC7027200 DOI: 10.3892/etm.2019.8376] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 12/13/2019] [Indexed: 12/29/2022] Open
Abstract
Micro ribonucleic acid (miR)-21 in the proliferation and apoptosis of Wilms' tumor (WT) cells was explored. SK-NEP-1 cells were transfected with miR-21 inhibitor to silence the expression of miR-21. Then, the effects of miR-21 silencing on the proliferation and apoptosis of WT SK-NEP-1 cells were detected through cell counting kit-8 (CCK-8), colony formation assay and flow cytometry. The targets of miR-21 were analyzed via TargetScan database. Fluorescence real-time quantitative polymerase chain reaction (RT-qPCR) assay and western blot analysis were conducted to detect the changes in messenger RNA (mRNA) and protein expression levels of gene of phosphate and tension homology deleted on chromosome ten (PTEN) after silencing miR-21. Whether miR-21 directly binds to PTEN was examined by activity detection via dual luciferase reporter gene assay. Western blotting was employed to detect the correlation of miR-21 with PTEN and protein kinase B (Akt). Compared with normal control (NC) group, miR-21 inhibitor group had significantly inhibited proliferation of SK-NEP-1 cells (P<0.05), notably reduced number of clones (P<0.05) and overtly raised proportion of apoptotic cells (P<0.05). The suppression of miR-21 expression upregulated the mRNA and protein expression levels of PTEN, and the results of activity detection via dual luciferase reporter gene assay indicated that miR-21 bound to PTEN 3'-untranslated region (UTR) to repress its expression (P<0.05). PTEN silencing increased phosphorylated Akt (p-Akt) level in SK-NEP-1 cells, but there was no changes in Akt protein level. After silencing both PTEN and miR-21, the decrease in p-Akt was reversed, thereby reversing the inhibitory effect of miR-21 on the proliferation of SK-NEP-1 cells (P<0.05). miR-21 affects the proliferation and apoptosis of WT SK-NEP-1 cells via the PTEN/Akt pathway.
Collapse
Affiliation(s)
- Xiuli Zhang
- Pediatrics Intensive Care Unit, Shanxian Central Hospital, Heze, Shandong 274300, P.R. China
| | - Chunyan Liu
- Pediatrics Intensive Care Unit, Shanxian Central Hospital, Heze, Shandong 274300, P.R. China
| | - Haiyan Li
- Pediatrics Intensive Care Unit, Shanxian Central Hospital, Heze, Shandong 274300, P.R. China
| | - Li Guo
- Pediatrics Intensive Care Unit, Shanxian Central Hospital, Heze, Shandong 274300, P.R. China
| |
Collapse
|
48
|
de Sá Pereira BM, Azevedo RMD, Aguirre Neto JCD, Menezes CF, Rodrigues KE, Faria PA, Camargo BD, Maschietto M. Intra-tumor genetic heterogeneity in Wilms tumor samples. ACTA ACUST UNITED AC 2020; 65:1496-1501. [PMID: 31994632 DOI: 10.1590/1806-9282.65.12.1496] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 06/18/2019] [Indexed: 12/20/2022]
Abstract
Childhood renal tumors account for ~7% of all childhood cancers, and most cases are embryonic Wilms' tumors (WT). Children with WT are usually treated by either COG or SIOP. The later treats the children using preoperative chemotherapy, but both have around 90% of overall survival in five years. WT is a genetically heterogeneous group with a low prevalence of known somatic alterations. Only around 30% of the cases present mutation in known genes, and there is a relatively high degree of intra-tumor genetic heterogeneity (ITGH). Besides potentially having an impact on the clinical outcome of patients, ITGH may interfere with the search for molecular markers that are prospectively being tested by COG and SIOP. In this review, we present the proposal of the current UMBRELLA SIOP Study 2017/Brazilian Renal Tumor Group that requires the multi-sampling collection of each tumor to better evaluate possible molecular markers, as well as to understand WT biology.
Collapse
Affiliation(s)
- Bruna M de Sá Pereira
- . Centro de Pesquisas (CPQ), Instituto Nacional de Câncer (Inca), Rio de Janeiro, RJ, Brasil
| | | | | | | | | | - Paulo A Faria
- . Divisão de Patologia (Dipat), Instituto Nacional de Câncer (Inca), Rio de Janeiro, RJ, Brasil
| | - Beatriz de Camargo
- . Centro de Pesquisas (CPQ), Instituto Nacional de Câncer (Inca), Rio de Janeiro, RJ, Brasil
| | - Mariana Maschietto
- . Laboratório Nacional de Biociências (LNBio), Centro Nacional de Pesquisa em Energia e Materiais (CNPEM), Campinas, SP, Brasil
| |
Collapse
|
49
|
Huang X, Zhao J, Fu W, Zhu J, Lou S, Tian X, Chen S, Ruan J, He J, Zhou H. The association of RAN and RANBP2 gene polymerphisms with Wilms tumor risk in Chinese children. J Cancer 2020; 11:804-809. [PMID: 31949483 PMCID: PMC6959007 DOI: 10.7150/jca.36651] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Accepted: 11/03/2019] [Indexed: 02/07/2023] Open
Abstract
Wilms tumor is considered to be the most common renal malignancy among children. RAN, a member of RAS superfamily, and its binding partner RANBP2 are related to the progression of multiple tumors. Nevertheless, the effects of the RAN and RANBP2 gene polymorphisms on the tumorigenesis of Wilms tumor remain unclarified. In this study, three potentially functional polymorphisms (rs56109543 C>T, rs7132224 A>G, and rs14035 C>T) in the RAN and one (rs2462788 C>T) in the RANBP2 were chosen to investigate their association with Wilms tumor susceptibility. Odds ratios (ORs) and 95% confidence intervals (CIs) were applied to assess the association of the selected polymorphisms with Wilms tumor susceptibility. Results shown that RAN rs7132224 AG/GG genotypes significantly increased Wilms tumor risk when compared to AA genotype (adjusted OR=1.40, 95% CI=1.01-1.95, P=0.047). Carriers of 1-3 risk genotypes have a significantly higher Wilms tumor risk than those without risk genotype (adjusted OR=1.49, 95% CI=1.07-2.07, P=0.020). Moreover, stratified analysis indicated that RAN rs56109543 CT/TT genotypes, RAN rs7132224 AG/GG genotypes and RANBP2 rs2462788 CT/TT genotypes remarkably increased Wilms tumor susceptibility among the subgroups. Our results indicated that RAN and RANBP2 polymorphisms were associated with Wilms tumor susceptibility in Chinese children. The role of RAN/RANBP2 in cancers deserves more attention.
Collapse
Affiliation(s)
- Xiaokai Huang
- Department of Hematology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang, China
| | - Jie Zhao
- Department of Hematology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang, China
| | - Wen Fu
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, China
| | - Jinhong Zhu
- Department of Clinical Laboratory, Biobank, Harbin Medical University Cancer Hospital, Harbin 150040, Heilongjiang, China
| | - Susu Lou
- Department of Hematology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang, China
| | - Xiaoqian Tian
- Department of Hematology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang, China
| | - Shanshan Chen
- Department of Hematology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang, China
| | - Jichen Ruan
- Department of Hematology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang, China
| | - Jing He
- Department of Hematology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang, China
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, China
- ✉ Corresponding authors: Haixia Zhou, Department of Hematology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109 West Xueyuan Road, Wenzhou 325027, Zhejiang, China, Tel./Fax: +86-13587898900, ; or Jing He, Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Guangzhou 510623, Guangdong, China, Tel./Fax: (+86-020)38076560,
| | - Haixia Zhou
- Department of Hematology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang, China
- ✉ Corresponding authors: Haixia Zhou, Department of Hematology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109 West Xueyuan Road, Wenzhou 325027, Zhejiang, China, Tel./Fax: +86-13587898900, ; or Jing He, Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Guangzhou 510623, Guangdong, China, Tel./Fax: (+86-020)38076560,
| |
Collapse
|
50
|
Zhong W, Zeng J, Xue J, Du A, Xu Y. Knockdown of lncRNA PVT1 alleviates high glucose-induced proliferation and fibrosis in human mesangial cells by miR-23b-3p/WT1 axis. Diabetol Metab Syndr 2020; 12:33. [PMID: 32322310 PMCID: PMC7161221 DOI: 10.1186/s13098-020-00539-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 04/01/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Diabetic nephropathy (DN) is a severe complication of diabetes with type 1 and 2. Long non-coding RNAs (lncRNAs) are being found to be involved in the DN pathogenesis. In this study, we aimed to further explore the effect and underlying mechanism of plasmacytoma variant translocation 1 (PVT1) in DN pathogenesis. METHODS The expression levels of PVT1, miR-23b-3p, and Wilms tumor protein 1 (WT1) mRNA were assessed by quantitative real-time polymerase chain reaction (qRT-PCR). Western blot analysis was performed to determine protein expression. Cell proliferation was detected using the 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetr-azolium (MTS) assay. The targeted correlation between miR-23b-3p and PVT1 or WT1 was verified by dual-luciferase reporter assay. RESULTS PVT1 and WT1 were highly expressed in the serum of DN patients and high glucose (HG)-induced mesangial cells (MCs). The knockdown of PVT1 or WT1 ameliorated HG-induced proliferation and fibrosis in MCs. Mechanistically, PVT1 modulated WT1 expression through acting as a molecular sponge of miR-23b-3p. The miR-23b-3p/WT1 axis mediated the protective effect of PVT1 knockdown on HG-induced proliferation and fibrosis in MCs. The NF-κB pathway was involved in the regulatory network of the PVT1/miR-23b-3p/WT1 axis in HG-induced MCs. CONCLUSION Our study suggested that PVT1 knockdown ameliorated HG-induced proliferation and fibrosis in MCs at least partially by regulating the miR-23b-3p/WT1/NF-κB pathway. Targeting PVT1 might be a potential therapeutic strategy for DN treatment.
Collapse
Affiliation(s)
- Wen Zhong
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, 169 East Lake Road, Wuchang District, Wuhan, 430071 Hubei China
| | - Jiaoe Zeng
- Department of Endocrine, Jingzhou Central Hospital and The Second Clinical Medical College, Yangtze University, Jingzhou, 434020 Hubei China
| | - Junli Xue
- Department of Endocrine, Jingzhou Central Hospital and The Second Clinical Medical College, Yangtze University, Jingzhou, 434020 Hubei China
| | - Aimin Du
- Department of Endocrine, Jingzhou Central Hospital and The Second Clinical Medical College, Yangtze University, Jingzhou, 434020 Hubei China
| | - Yancheng Xu
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, 169 East Lake Road, Wuchang District, Wuhan, 430071 Hubei China
| |
Collapse
|