1
|
Sánchez-Cazorla E, Carrera N, García-González MÁ. HNF1B Transcription Factor: Key Regulator in Renal Physiology and Pathogenesis. Int J Mol Sci 2024; 25:10609. [PMID: 39408938 PMCID: PMC11476927 DOI: 10.3390/ijms251910609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/19/2024] [Accepted: 09/30/2024] [Indexed: 10/20/2024] Open
Abstract
The HNF1B gene, located on chromosome 17q12, encodes a transcription factor essential for the development of several organs. It regulates the expression of multiple genes in renal, pancreatic, hepatic, neurological, and genitourinary tissues during prenatal and postnatal development, influencing processes such as nephrogenesis, cellular polarity, tight junction formation, cilia development, ion transport in the renal tubule, and renal metabolism. Mutations that alter the function of Hnf1b deregulate those processes, leading to various pathologies characterized by both renal and extrarenal manifestations. The main renal diseases that develop are polycystic kidney disease, hypoplastic or dysplastic kidneys, structural abnormalities, Congenital Anomalies of the Kidney and Urinary Tract (CAKUT), and electrolyte imbalances such as hyperuricemia and hypomagnesemia. Extrarenal manifestations include Maturity-Onset Diabetes of the Young (MODY), hypertransaminasemia, genital and urinary tract malformations, Autism Spectrum Disorder (ASD), and other neurodevelopmental disorders. Patients with HNF1B alterations typically carry either punctual mutations or a monoallelic microdeletion in the 17q12 region. Future research on the molecular mechanisms and genotype-phenotype correlations in HNF1B-related conditions will enhance our understanding, leading to improved clinical management, genetic counseling, monitoring, and patient care.
Collapse
Affiliation(s)
- Eloísa Sánchez-Cazorla
- Group of Genetics and Developmental Biology of Renal Disease, Laboratory of Nephrology, No. 11, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital (CHUS), 15706 Santiago de Compostela, Spain;
- Genomic Medicine Group, Clinical University Hospital (CHUS), 15706 Santiago de Compostela, Spain
| | - Noa Carrera
- Group of Genetics and Developmental Biology of Renal Disease, Laboratory of Nephrology, No. 11, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital (CHUS), 15706 Santiago de Compostela, Spain;
- Genomic Medicine Group, Clinical University Hospital (CHUS), 15706 Santiago de Compostela, Spain
- RICORS 2040 (Kidney Disease), ISCIII, 15706 Santiago de Compostela, Spain
| | - Miguel Ángel García-González
- Group of Genetics and Developmental Biology of Renal Disease, Laboratory of Nephrology, No. 11, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital (CHUS), 15706 Santiago de Compostela, Spain;
- Genomic Medicine Group, Clinical University Hospital (CHUS), 15706 Santiago de Compostela, Spain
- RICORS 2040 (Kidney Disease), ISCIII, 15706 Santiago de Compostela, Spain
| |
Collapse
|
2
|
Torban E, Goodyer P. Wilms' tumor gene 1: lessons from the interface between kidney development and cancer. Am J Physiol Renal Physiol 2024; 326:F3-F19. [PMID: 37916284 DOI: 10.1152/ajprenal.00248.2023] [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: 08/17/2023] [Revised: 10/27/2023] [Accepted: 10/27/2023] [Indexed: 11/03/2023] Open
Abstract
In 1990, mutations of the Wilms' tumor-1 gene (WT1), encoding a transcription factor in the embryonic kidney, were found in 10-15% of Wilms' tumors; germline WT1 mutations were associated with hereditary syndromes involving glomerular and reproductive tract dysplasia. For more than three decades, these discoveries prompted investigators to explore the embryonic role of WT1 and the mechanisms by which loss of WT1 leads to malignant transformation. Here, we discuss how alternative splicing of WT1 generates isoforms that act in a context-specific manner to activate or repress target gene transcription. WT1 also regulates posttranscriptional regulation, alters the epigenetic landscape, and activates miRNA expression. WT1 functions at multiple stages of kidney development, including the transition from resting stem cells to committed nephron progenitor, which it primes to respond to WNT9b signals from the ureteric bud. WT1 then drives nephrogenesis by activating WNT4 expression and directing the development of glomerular podocytes. We review the WT1 mutations that account for Denys-Drash syndrome, Frasier syndrome, and WAGR syndrome. Although the WT1 story began with Wilms' tumors, an understanding of the pathways that link aberrant kidney development to malignant transformation still has some important gaps. Loss of WT1 in nephrogenic rests may leave these premalignant clones with inadequate DNA repair enzymes and may disturb the epigenetic landscape. Yet none of these observations provide a complete picture of Wilms' tumor pathogenesis. It appears that the WT1 odyssey is unfinished and still holds a great deal of untilled ground to be explored.
Collapse
Affiliation(s)
- Elena Torban
- Department of Medicine, McGill University and Research Institute of McGill University Health Center, Montreal, Quebec, Canada
| | - Paul Goodyer
- Department of Human Genetics, Montreal Children's Hospital and McGill University, Montreal, Quebec, Canada
- Department of Pediatrics, Montreal Children's Hospital and McGill University, Montreal, Quebec, Canada
| |
Collapse
|
3
|
Shi M, Fu P, Bonventre JV, McCracken KW. Directed differentiation of ureteric bud and collecting duct organoids from human pluripotent stem cells. Nat Protoc 2023; 18:2485-2508. [PMID: 37460630 PMCID: PMC11154671 DOI: 10.1038/s41596-023-00847-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 04/24/2023] [Indexed: 08/09/2023]
Abstract
Developing models of human kidney tissue in vitro is an important challenge in regenerative nephrology research, given the paucity of novel and effective therapies in kidney disease. However, the de novo generation of kidney tissues from human pluripotent stem cells (hPSCs) is challenging owing to the structural and functional complexity of the organ, as well its developmental origin from two distinct embryologic populations: the metanephric mesenchyme and the ureteric bud (UB). Directed differentiation strategies have been developed to generate kidney organoids containing nephron-like structures; we recently reported an efficient and practical method to generate UB tissues. Here, we describe a detailed step-by-step protocol for differentiation of hPSCs into three-dimensonal UB organoids that exhibit complex morphological development and the capacity to differentiate into functional collecting duct tissues. Over 3 d, hPSCs are induced into PAX2+GATA3+ pronephric (anterior) intermediate mesoderm fates in monolayer cultures at high efficiency. The cells are aggregated into three-dimensional spheroids, which then assemble and organize into nephric duct-like tissue over 4 d. When embedded into an extracellular matrix, the spheroids grow into UB organoids that recapitulate fetal branching morphogenesis for 1 week of culture. When switched to permissive conditions, the UB organoids spontaneously differentiate to form collecting duct principal cells. This approach provides robust and reproducible methods that can be readily adopted by users with basic experience in hPSC and organoid differentiation to generate UB tissues, which may be used to investigate human kidney development, model disease processes and catalyze further efforts in engineering functional kidney tissue.
Collapse
Affiliation(s)
- Min Shi
- Division of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, China
- Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ping Fu
- Division of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, China
| | - Joseph V Bonventre
- Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Harvard Stem Cell Institute, Cambridge and Boston, MA, USA.
- Division of Engineering in Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Kyle W McCracken
- Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Division of Nephrology, Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
4
|
Ahram DF, Lim TY, Ke J, Jin G, Verbitsky M, Bodria M, Kil BH, Chatterjee D, Piva SE, Marasa M, Zhang JY, Cocchi E, Caridi G, Gucev Z, Lozanovski VJ, Pisani I, Izzi C, Savoldi G, Gnutti B, Capone VP, Morello W, Guarino S, Esposito P, Lambert S, Radhakrishnan J, Appel GB, Uy NS, Rao MK, Canetta PA, Bomback AS, Nestor JG, Hays T, Cohen DJ, Finale C, van Wijk JA, La Scola C, Baraldi O, Tondolo F, Di Renzo D, Jamry-Dziurla A, Pezzutto A, Manca V, Mitrotti A, Santoro D, Conti G, Martino M, Giordano M, Gesualdo L, Zibar L, Masnata G, Bonomini M, Alberti D, La Manna G, Caliskan Y, Ranghino A, Marzuillo P, Kiryluk K, Krzemień G, Miklaszewska M, Lin F, Montini G, Scolari F, Fiaccadori E, Arapović A, Saraga M, McKiernan J, Alam S, Zaniew M, Szczepańska M, Szmigielska A, Sikora P, Drożdż D, Mizerska-Wasiak M, Mane S, Lifton RP, Tasic V, Latos-Bielenska A, Gharavi AG, Ghiggeri GM, Materna-Kiryluk A, Westland R, Sanna-Cherchi S. Rare Single Nucleotide and Copy Number Variants and the Etiology of Congenital Obstructive Uropathy: Implications for Genetic Diagnosis. J Am Soc Nephrol 2023; 34:1105-1119. [PMID: 36995132 PMCID: PMC10278788 DOI: 10.1681/asn.0000000000000132] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 01/18/2023] [Indexed: 03/31/2023] Open
Abstract
SIGNIFICANCE STATEMENT Congenital obstructive uropathy (COU) is a prevalent human developmental defect with highly heterogeneous clinical presentations and outcomes. Genetics may refine diagnosis, prognosis, and treatment, but the genomic architecture of COU is largely unknown. Comprehensive genomic screening study of 733 cases with three distinct COU subphenotypes revealed disease etiology in 10.0% of them. We detected no significant differences in the overall diagnostic yield among COU subphenotypes, with characteristic variable expressivity of several mutant genes. Our findings therefore may legitimize a genetic first diagnostic approach for COU, especially when burdening clinical and imaging characterization is not complete or available. BACKGROUND Congenital obstructive uropathy (COU) is a common cause of developmental defects of the urinary tract, with heterogeneous clinical presentation and outcome. Genetic analysis has the potential to elucidate the underlying diagnosis and help risk stratification. METHODS We performed a comprehensive genomic screen of 733 independent COU cases, which consisted of individuals with ureteropelvic junction obstruction ( n =321), ureterovesical junction obstruction/congenital megaureter ( n =178), and COU not otherwise specified (COU-NOS; n =234). RESULTS We identified pathogenic single nucleotide variants (SNVs) in 53 (7.2%) cases and genomic disorders (GDs) in 23 (3.1%) cases. We detected no significant differences in the overall diagnostic yield between COU sub-phenotypes, and pathogenic SNVs in several genes were associated to any of the three categories. Hence, although COU may appear phenotypically heterogeneous, COU phenotypes are likely to share common molecular bases. On the other hand, mutations in TNXB were more often identified in COU-NOS cases, demonstrating the diagnostic challenge in discriminating COU from hydronephrosis secondary to vesicoureteral reflux, particularly when diagnostic imaging is incomplete. Pathogenic SNVs in only six genes were found in more than one individual, supporting high genetic heterogeneity. Finally, convergence between data on SNVs and GDs suggest MYH11 as a dosage-sensitive gene possibly correlating with severity of COU. CONCLUSIONS We established a genomic diagnosis in 10.0% of COU individuals. The findings underscore the urgent need to identify novel genetic susceptibility factors to COU to better define the natural history of the remaining 90% of cases without a molecular diagnosis.
Collapse
Affiliation(s)
- Dina F. Ahram
- Department of Medicine, Division of Nephrology, Columbia University, New York, New York
| | - Tze Y. Lim
- Department of Medicine, Division of Nephrology, Columbia University, New York, New York
| | - Juntao Ke
- Department of Medicine, Division of Nephrology, Columbia University, New York, New York
| | - Gina Jin
- Department of Medicine, Division of Nephrology, Columbia University, New York, New York
| | - Miguel Verbitsky
- Department of Medicine, Division of Nephrology, Columbia University, New York, New York
| | - Monica Bodria
- Division of Nephrology and Renal Transplantation, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Byum Hee Kil
- Department of Medicine, Division of Nephrology, Columbia University, New York, New York
| | - Debanjana Chatterjee
- Department of Medicine, Division of Nephrology, Columbia University, New York, New York
| | - Stacy E. Piva
- Department of Medicine, Division of Nephrology, Columbia University, New York, New York
| | - Maddalena Marasa
- Department of Medicine, Division of Nephrology, Columbia University, New York, New York
| | - Jun Y. Zhang
- Department of Medicine, Division of Nephrology, Columbia University, New York, New York
| | - Enrico Cocchi
- Department of Medicine, Division of Nephrology, Columbia University, New York, New York
| | - Gianluca Caridi
- Division of Nephrology and Renal Transplantation, IRCCS Istituto Giannina Gaslini, Genoa, Italy
- Laboratory on Molecular Nephrology, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Zoran Gucev
- Medical Faculty of Skopje, University Children's Hospital, Skopje, Macedonia
| | - Vladimir J. Lozanovski
- Medical Faculty of Skopje, University Children's Hospital, Skopje, Macedonia
- Department of General, Visceral and Transplant Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Isabella Pisani
- Unità Operativa Nefrologia, Azienda Ospedaliero-Universitaria di Parma, Dipartimento di Medicina e Chirurgia, Università di Parma, Parma, Italy
| | - Claudia Izzi
- Division of Nephrology, Department of Obstetrics and Gynecology, ASST Spedali Civili of Brescia, Brescia, Italy
| | | | - Barbara Gnutti
- Medical Genetics Laboratory, ASST-Spedali Civili, Brescia, Italy
| | - Valentina P. Capone
- Department of Medicine, Division of Nephrology, Columbia University, New York, New York
- Pediatric Nephrology, Dialysis and Transplant Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - William Morello
- Pediatric Nephrology, Dialysis and Transplant Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefano Guarino
- Department of Woman and Child and of General and Specialized Surgery, Università degli Studi della Campania “Luigi Vanvitelli,” Naples, Italy
| | - Pasquale Esposito
- Department of Internal Medicine, University of Genoa, Genova, Italy
- Unit of Nephrology, IRCCS San Martino Polyclinic Hospital, Genoa, Italy
| | - Sarah Lambert
- Yale School of Medicine/Yale New Haven Health System, New Haven, Connecticut
| | - Jai Radhakrishnan
- Department of Medicine, Division of Nephrology, Columbia University, New York, New York
| | - Gerald B. Appel
- Department of Medicine, Division of Nephrology, Columbia University, New York, New York
| | - Natalie S. Uy
- Division of Pediatric Nephrology, Department of Pediatric, NewYork-Presbyterian Morgan Stanley Children's Hospital, Columbia University Irving Medical Center, New York, New York
| | - Maya K. Rao
- Department of Medicine, Division of Nephrology, Columbia University, New York, New York
| | - Pietro A. Canetta
- Department of Medicine, Division of Nephrology, Columbia University, New York, New York
| | - Andrew S. Bomback
- Department of Medicine, Division of Nephrology, Columbia University, New York, New York
| | - Jordan G. Nestor
- Department of Medicine, Division of Nephrology, Columbia University, New York, New York
| | - Thomas Hays
- Department of Pediatrics, Division of Neonatology, Columbia University, New York, New York
| | - David J. Cohen
- Department of Medicine, Division of Nephrology, Columbia University, New York, New York
| | - Carolina Finale
- Nephrology, Dialysis and Renal Transplantation Unit, Azienda Ospedaliera Universitaria Ospedali Riuniti Umberto I, Lancisi, Salesi of Ancona, Ancona, Italy
| | - Joanna A.E. van Wijk
- Department of Pediatric Nephrology, Emma Children's Hospital, University of Amsterdam, Amsterdam, The Netherlands
| | - Claudio La Scola
- Nephrology and Dialysis Unit, Department of Pediatrics, Azienda Ospedaliero Universitaria Sant'Orsola-Malpighi, Bologna, Italy
| | - Olga Baraldi
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Francesco Tondolo
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Dacia Di Renzo
- “Spirito Santo” Hospital of Pescara, Pediatric Surgery of “G. d'Annunzio” University of Chieti-Pescara, Chieti, Italy
| | - Anna Jamry-Dziurla
- Polish Registry of Congenital Malformations, Chair and Department of Medical Genetics, University of Medical Sciences, Poznan, Poland
| | - Alessandro Pezzutto
- Nephrology and Dialysis Unit, Department of Medicine, SS Annunziata Hospital, “G. d'Annunzio” University, Chieti, Italy
| | - Valeria Manca
- Department of Pediatric Urology, Azienda Ospedaliera Brotzu, Cagliari, Italy
| | - Adele Mitrotti
- Department of Medicine, Division of Nephrology, Columbia University, New York, New York
- Section of Nephrology, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Domenico Santoro
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Giovanni Conti
- Department of Pediatric Nephrology, Azienda Ospedaliera Universitaria “G. Martino,” Messina, Italy
| | - Marida Martino
- Pediatric Nephrology and Dialysis Unit, Pediatric Hospital “Giovanni XXIII,” Bari, Italy
| | - Mario Giordano
- Pediatric Nephrology and Dialysis Unit, Pediatric Hospital “Giovanni XXIII,” Bari, Italy
| | - Loreto Gesualdo
- Section of Nephrology, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Lada Zibar
- Department of Nephrology, University Hospital Merkur, Zagreb, Croatia
- Faculty of Medicine, University Josip Juraj Strossmayer in Osijek, Osijek, Croatia
| | - Giuseppe Masnata
- Department of Pediatric Urology, Azienda Ospedaliera Brotzu, Cagliari, Italy
| | - Mario Bonomini
- Nephrology and Dialysis Unit, Department of Medicine, SS Annunziata Hospital, “G. d'Annunzio” University, Chieti, Italy
| | | | - Gaetano La Manna
- IRCCS Azienda Ospedaliera di Bologna, Nephrology, Dialysis and Kidney Transplant Unit, St. Orsola University Hospital, Bologna, Italy
| | - Yasar Caliskan
- Division of Nephrology, Saint Louis University School of Medicine, Saint Louis, Missouri
| | - Andrea Ranghino
- Nephrology, Dialysis and Renal Transplantation Unit, Azienda Ospedaliera Universitaria Ospedali Riuniti Umberto I, Lancisi, Salesi of Ancona, Ancona, Italy
| | - Pierluigi Marzuillo
- Department of Woman and Child and of General and Specialized Surgery, Università degli Studi della Campania “Luigi Vanvitelli,” Naples, Italy
| | - Krzysztof Kiryluk
- Department of Medicine, Division of Nephrology, Columbia University, New York, New York
| | - Grażyna Krzemień
- Department of Pediatrics and Nephrology, Medical University of Warsaw, Warsaw, Poland
| | - Monika Miklaszewska
- Department of Pediatric Nephrology and Hypertension, Jagiellonian University Medical College, Krakow, Poland
| | - Fangming Lin
- Division of Pediatric Nephrology, Department of Pediatric, NewYork-Presbyterian Morgan Stanley Children's Hospital, Columbia University Irving Medical Center, New York, New York
| | - Giovanni Montini
- Pediatric Nephrology, Dialysis and Transplant Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, Giuliana and Bernardo Caprotti Chair of Pediatrics, University of Milano, Milano, Italy
| | - Francesco Scolari
- Division of Nephrology and Dialysis, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia and ASST Spedali Civili of Brescia, Brescia, Italy
| | - Enrico Fiaccadori
- Unità Operativa Nefrologia, Azienda Ospedaliero-Universitaria di Parma, Dipartimento di Medicina e Chirurgia, Università di Parma, Parma, Italy
| | - Adela Arapović
- Department of Pediatrics, University Hospital of Split, Split, Croatia
- School of Medicine, University of Split, Split, Croatia
| | - Marijan Saraga
- Department of Pediatrics, University Hospital of Split, Split, Croatia
- School of Medicine, University of Split, Split, Croatia
| | - James McKiernan
- Department of Urology, Columbia University Irving Medical Center, New York, New York
| | - Shumyle Alam
- Department of Urology, Columbia University Irving Medical Center, New York, New York
- Division of Pediatric Urology, MUSC Health-University Medical Center, Charleston, South Carolina
| | - Marcin Zaniew
- Department of Pediatrics, University of Zielona Góra, Zielona Góra, Poland
| | - Maria Szczepańska
- Department of Pediatrics, FMS in Zabrze, Medical University of Silesia, Katowice, Poland
| | - Agnieszka Szmigielska
- Department of Pediatrics and Nephrology, Medical University of Warsaw, Warsaw, Poland
| | - Przemysław Sikora
- Department of Pediatric Nephrology, Medical University of Lublin, Lublin, Poland
| | - Dorota Drożdż
- Department of Pediatric Nephrology and Hypertension, Jagiellonian University Medical College, Krakow, Poland
| | | | - Shrikant Mane
- Yale Center for Mendelian Genomics (YCMG), New Haven, Connecticut
| | | | - Velibor Tasic
- Medical Faculty of Skopje, University Children's Hospital, Skopje, Macedonia
| | - Anna Latos-Bielenska
- Polish Registry of Congenital Malformations, Chair and Department of Medical Genetics, University of Medical Sciences, Poznan, Poland
| | - Ali G. Gharavi
- Department of Medicine, Division of Nephrology, Columbia University, New York, New York
| | - Gian Marco Ghiggeri
- Division of Nephrology and Renal Transplantation, IRCCS Istituto Giannina Gaslini, Genoa, Italy
- Laboratory on Molecular Nephrology, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Anna Materna-Kiryluk
- Polish Registry of Congenital Malformations, Chair and Department of Medical Genetics, University of Medical Sciences, Poznan, Poland
| | - Rik Westland
- Department of Pediatric Nephrology, Emma Children's Hospital, University of Amsterdam, Amsterdam, The Netherlands
| | - Simone Sanna-Cherchi
- Department of Medicine, Division of Nephrology, Columbia University, New York, New York
| |
Collapse
|
5
|
Wille S, Peukert N, Haak R, Riedel J, Mayer S, Kluth D, Lacher M, Gosemann JH, Markel M. Development of the Urinary Tract in Fetal Rats: A Micro-CT Study. Eur J Pediatr Surg 2023; 33:53-60. [PMID: 36395789 DOI: 10.1055/s-0042-1758681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Micro-computed tomography (micro-CT) is an established tool to study fetal development in rodents. This study aimed to use micro-CT imaging to visualize the development of the urinary tract in fetal rats. MATERIALS AND METHODS Fetal rats from embryonic day (ED) 15, ED17, ED19, ED21, and N0 (newborn) (n = 6 per group; 3 males) were fixed and desiccated using the "critical point" technique. We utilized the micro-CT system (SkyScan) and analyzed the resulting scans with CTAn, DataViewer, and ImageJ to visualize the morphology and quantify the volumes of kidney, bladder, adrenal gland, as well as length of the ureter. RESULTS High-resolution micro-CT showed continuous growth of both kidneys from ED15 to N0, with the highest increase between ED19 and ED21. The length of the ureter increased from ED15 to ED21 and remained stable until birth. The volume of the bladder steadily increased from ED15 to N0.In females, a statistically higher volume of the adrenal gland on ED21 was observed, whereas no sex-specific differences were seen for kidney, ureter, and bladder development. CONCLUSION Micro-CT depicts an excellent tool to study urinary tract development in the fetal and neonatal rat. It enables the metric quantification of longitudinal anatomic changes in high definition without previous destructive tissue preparation. The present study revealed sex-specific differences of the adrenal gland development and provides comprehensive data for the understanding of fetal urinary tract development, inspiring future research on congenital urological malformations.
Collapse
Affiliation(s)
- Stephanie Wille
- Department of Pediatric Surgery, University of Leipzig, Leipzig, Germany.,Department of Interdisciplinary Medical Intensive Care, University of Leipzig, Leipzig, Germany
| | - Nicole Peukert
- Department of Pediatric Surgery, University of Leipzig, Leipzig, Germany
| | - Rainer Haak
- Department of Cariology, Endodontology, and Periodontology, University of Leipzig, Leipzig, Germany
| | - Jan Riedel
- Department of Pediatric Surgery, University of Leipzig, Leipzig, Germany
| | - Steffi Mayer
- Department of Pediatric Surgery, University of Leipzig, Leipzig, Germany
| | - Dietrich Kluth
- Department of Pediatric Surgery, University of Leipzig, Leipzig, Germany
| | - Martin Lacher
- Department of Pediatric Surgery, University of Leipzig, Leipzig, Germany
| | | | - Moritz Markel
- Department of Pediatric Surgery, University of Leipzig, Leipzig, Germany
| |
Collapse
|
6
|
Nishiyama K, Sanefuji M, Kurokawa M, Iwaya Y, Hamada N, Sonoda Y, Ogawa M, Shimono M, Suga R, Kusuhara K, Ohga S. Maternal Chronic Disease and Congenital Anomalies of the Kidney and Urinary Tract in Offspring: A Japanese Cohort Study. Am J Kidney Dis 2022; 80:619-628.e1. [PMID: 35439592 DOI: 10.1053/j.ajkd.2022.03.003] [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] [Received: 09/08/2021] [Accepted: 03/01/2022] [Indexed: 02/02/2023]
Abstract
RATIONALE & OBJECTIVE Several maternal chronic diseases have been reported as risk factors for congenital anomalies of the kidney and urinary tract (CAKUT) in offspring. However, these investigations used case-control designs, and cases with isolated genitourinary CAKUT were not distinguished from cases in which CAKUT were present with extrarenal congenital anomalies (complicated CAKUT). We examined the association of maternal diseases with isolated and complicated CAKUT in offspring using data from a prospective cohort study. STUDY DESIGN A nationwide prospective birth cohort study. SETTING & PARTICIPANTS 100,239 children enrolled in the Japan Environment and Children's Study between January 2011 and March 2014 at 15 research centers. Physicians' diagnoses in mothers and children were collected from medical record transcripts and questionnaires. EXPOSURES Medical histories of maternal noncommunicable diseases, including obesity, hypertension, diabetes mellitus, kidney disease, hyperthyroidism, hypothyroidism, psychiatric disease, epilepsy, cancer, and autoimmune disease. OUTCOMES CAKUT diagnosed during the first 3 years of life, classified as isolated or complicated. ANALYTICAL APPROACH Multivariable Poisson regression with generalized estimating equations accounting for clustering by clinical center. RESULTS Among the 100,239 children, 560 (0.6%) had CAKUT, comprising 454 (81%) isolated and 106 (19%) complicated forms. The risk of isolated CAKUT was increased in children of mothers who experienced kidney disease (adjusted risk ratio [RR], 1.80 [95% CI, 1.12-2.91]) or cancer (RR, 2.11 [95% CI, 1.15-3.86]). Furthermore, the risk of complicated CAKUT was increased in children of mothers with diabetes mellitus (RR, 3.04 [95% CI, 1.64-5.61]). LIMITATIONS Lack of standardization or prespecification of clinical definitions, diagnostic criteria, measurements, and testing. Genetic testing was not performed. CONCLUSIONS Isolated CAKUTs and complicated CAKUTs were associated with different maternal diseases. The results may inform clinical management of pregnancy and highlight potential differences in the genesis of isolated and complicated forms of CAKUT.
Collapse
Affiliation(s)
- Kei Nishiyama
- Departments of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masafumi Sanefuji
- Departments of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Research Center for Environment and Developmental Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Mari Kurokawa
- Department of Pediatrics, Fukuoka Higashi Medical Center, Koga, Fukuoka, Japan
| | - Yuka Iwaya
- Departments of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Norio Hamada
- Department of Obstetrics and Gynecology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Research Center for Environment and Developmental Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuri Sonoda
- Departments of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Research Center for Environment and Developmental Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masanobu Ogawa
- Department of Obstetrics and Gynecology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masayuki Shimono
- Department of Pediatrics, University of Occupational and Environmental Health, Kitakyushu, Japan; Regional Center for Japan Environment and Children's Study, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Reiko Suga
- Regional Center for Japan Environment and Children's Study, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Koichi Kusuhara
- Department of Pediatrics, University of Occupational and Environmental Health, Kitakyushu, Japan; Regional Center for Japan Environment and Children's Study, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Shouichi Ohga
- Departments of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Research Center for Environment and Developmental Medical Sciences, Kyushu University, Fukuoka, Japan
| | | |
Collapse
|
7
|
Wu CHW, Lim TY, Wang C, Seltzsam S, Zheng B, Schierbaum L, Schneider S, Mann N, Connaughton DM, Nakayama M, van der Ven AT, Dai R, Kolvenbach CM, Kause F, Ottlewski I, Stajic N, Soliman NA, Kari JA, El Desoky S, Fathy HM, Milosevic D, Turudic D, Al Saffar M, Awad HS, Eid LA, Ramanathan A, Senguttuvan P, Mane SM, Lee RS, Bauer SB, Lu W, Hilger AC, Tasic V, Shril S, Sanna-Cherchi S, Hildebrandt F. Copy Number Variation Analysis Facilitates Identification of Genetic Causation in Patients with Congenital Anomalies of the Kidney and Urinary Tract. EUR UROL SUPPL 2022; 44:106-112. [PMID: 36185583 PMCID: PMC9520493 DOI: 10.1016/j.euros.2022.08.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/10/2022] [Indexed: 11/27/2022] Open
Abstract
Background Congenital anomalies of the kidneys and urinary tract (CAKUT) are the most common cause of chronic kidney disease among children and adults younger than 30 yr. In our previous study, whole-exome sequencing (WES) identified a known monogenic cause of isolated or syndromic CAKUT in 13% of families with CAKUT. However, WES has limitations and detection of copy number variations (CNV) is technically challenging, and CNVs causative of CAKUT have previously been detected in up to 16% of cases. Objective To detect CNVs causing CAKUT in this WES cohort and increase the diagnostic yield. Design setting and participants We performed a genome-wide single nucleotide polymorphism (SNP)-based CNV analysis on the same CAKUT cohort for whom WES was previously conducted. Outcome measurements and statistical analysis We evaluated and classified the CNVs using previously published predefined criteria. Results and limitations In a cohort of 170 CAKUT families, we detected a pathogenic CNV known to cause CAKUT in nine families (5.29%, 9/170). There were no competing variants on genome-wide CNV analysis or WES analysis. In addition, we identified novel likely pathogenic CNVs that may cause a CAKUT phenotype in three of the 170 families (1.76%). Conclusions CNV analysis in this cohort of 170 CAKUT families previously examined via WES increased the rate of diagnosis of genetic causes of CAKUT from 13% on WES to 18% on WES + CNV analysis combined. We also identified three candidate loci that may potentially cause CAKUT. Patient summary We conducted a genetics study on families with congenital anomalies of the kidney and urinary tract (CAKUT). We identified gene mutations that can explain CAKUT symptoms in 5.29% of the families, which increased the percentage of genetic causes of CAKUT to 18% from a previous study, so roughly one in five of our patients with CAKUT had a genetic cause. These analyses can help patients with CAKUT and their families in identifying a possible genetic cause.
Collapse
Affiliation(s)
- Chen-Han Wilfred Wu
- Department of Pediatrics, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Urology, Case Western Reserve University and University Hospitals, Cleveland, OH, USA
- Department of Genetics and Genome Sciences, Case Western Reserve University and University Hospitals, Cleveland, OH, USA
| | - Tze Y. Lim
- Division of Nephrology, Columbia University Irving Medical Center, New York, NY, USA
| | - Chunyan Wang
- Department of Pediatrics, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Steve Seltzsam
- Department of Pediatrics, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Bixia Zheng
- Department of Pediatrics, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Luca Schierbaum
- Department of Pediatrics, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Sophia Schneider
- Department of Pediatrics, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Nina Mann
- Department of Pediatrics, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Dervla M. Connaughton
- Department of Pediatrics, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Makiko Nakayama
- Department of Pediatrics, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Amelie T. van der Ven
- Department of Pediatrics, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Rufeng Dai
- Department of Pediatrics, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Caroline M. Kolvenbach
- Department of Pediatrics, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Franziska Kause
- Department of Pediatrics, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Isabel Ottlewski
- Department of Pediatrics, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Natasa Stajic
- Department of Pediatric Nephrology, Institute for Mother and Child Health Care, Belgrade, Serbia
| | - Neveen A. Soliman
- Department of Pediatrics, Center of Pediatric Nephrology & Transplantation, Cairo University, Egyptian Group for Orphan Renal Diseases, Cairo, Egypt
| | - Jameela A. Kari
- Department of Pediatrics, King AbdulAziz University, Jeddah, Saudi Arabia
| | - Sherif El Desoky
- Department of Pediatrics, King AbdulAziz University, Jeddah, Saudi Arabia
| | - Hanan M. Fathy
- Pediatric Nephrology Unit, University of Alexandria, Alexandria, Egypt
| | - Danko Milosevic
- Department of Pediatric Nephrology, University Hospital Center Zagreb, Zagreb, Croatia
| | - Daniel Turudic
- Department of Pediatric Nephrology, University Hospital Center Zagreb, Zagreb, Croatia
| | - Muna Al Saffar
- Department of Pediatrics, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Paediatrics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Hazem S. Awad
- Pediatric Nephrology Department, Dubai Hospital, Dubai, United Arab Emirates
| | - Loai A. Eid
- Pediatric Nephrology Department, Dubai Hospital, Dubai, United Arab Emirates
- Department of Pediatrics, Dubai Medical College and Kidney Centre of Excellence, Al Jalila Children’s Specialty Hospital, Dubai, United Arab Emirates
| | - Aravind Ramanathan
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Prabha Senguttuvan
- Department of Pediatric Nephrology, Dr. Mehta’s Multi-Specialty Hospital, Chennai, India
| | - Shrikant M. Mane
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Richard S. Lee
- Department of Urology, Boston Children’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Stuart B. Bauer
- Department of Urology, Boston Children’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Weining Lu
- Renal Section, Department of Medicine, Boston University Medical Center, Boston, MA, USA
| | - Alina C. Hilger
- Department of Pediatric and Adolescent Medicine, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Velibor Tasic
- Medical Faculty Skopje, University Children’s Hospital, Skopje, Macedonia
| | - Shirlee Shril
- Department of Pediatrics, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Simone Sanna-Cherchi
- Division of Nephrology, Columbia University Irving Medical Center, New York, NY, USA
| | - Friedhelm Hildebrandt
- Department of Pediatrics, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
- Corresponding author. Division of Nephrology, Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA 02115, USA. Tel. +1 617 3556129; Fax: +1 617 8300365.
| |
Collapse
|
8
|
Chen H, Zhang W, Maskey N, Yang F, Zheng Z, Li C, Wang R, Wu P, Mao S, Zhang J, Yan Y, Li W, Yao X. Urological cancer organoids, patients' avatars for precision medicine: past, present and future. Cell Biosci 2022; 12:132. [PMID: 35986387 PMCID: PMC9389738 DOI: 10.1186/s13578-022-00866-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 07/31/2022] [Indexed: 11/29/2022] Open
Abstract
Urological cancers are common malignant cancers worldwide, with annually increasing morbidity and mortality rates. For decades, two-dimensional cell cultures and animal models have been widely used to study the development and underlying molecular mechanisms of urological cancers. However, they either fail to reflect cancer heterogeneity or are time-consuming and labour-intensive. The recent emergence of a three-dimensional culture model called organoid has the potential to overcome the shortcomings of traditional models. For example, organoids can recapitulate the histopathological and molecular diversity of original cancer and reflect the interaction between cancer and surrounding cells or stroma by simulating tumour microenvironments. Emerging evidence suggests that urine-derived organoids can be generated, which could be a novel non-invasive liquid biopsy method that provides new ideas for clinical precision therapy. However, the current research on organoids has encountered some bottlenecks, such as the lack of a standard culture process, the need to optimize the culture medium and the inability to completely simulate the immune system in vivo. Nonetheless, cell co-culture and organoid-on-a-chip have significant potential to solve these problems. In this review, the latest applications of organoids in drug screening, cancer origin investigation and combined single-cell sequencing are illustrated. Furthermore, the development and application of organoids in urological cancers and their challenges are summarised.
Collapse
|
9
|
Wu H, Liu F, Shangguan Y, Yang Y, Shi W, Hu W, Zeng Z, Hu N, Zhang X, Hocher B, Tang D, Yin L, Dai Y. Integrating spatial transcriptomics with single-cell transcriptomics reveals a spatiotemporal gene landscape of the human developing kidney. Cell Biosci 2022; 12:80. [PMID: 35659756 PMCID: PMC9164720 DOI: 10.1186/s13578-022-00801-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 04/29/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Research on spatiotemporal gene landscape can provide insights into the spatial characteristics of human kidney development and facilitate kidney organoid cultivation. Here, we profiled the spatiotemporal gene programs of the human embryonic kidneys at 9 and 18 post-conception weeks (PCW) by integrating the application of microarray-based spatial transcriptomics and single-cell transcriptomics. RESULTS We mapped transcriptomic signatures of scRNA-seq cell types upon the 9 and 18 PCW kidney sections based on cell-type deconvolution and multimodal intersection analyses, depicting a spatial landscape of developing cell subpopulations. We established the gene characteristics in the medullary regions and revealed a strong mitochondrial oxidative phosphorylation and glycolysis activity in the deeper medullary region. We also built a regulatory network centered on GDNF-ETV4 for nephrogenic niche development based on the weighted gene co-expression network analysis and highlighted the key roles of Wnt, FGF, and JAG1-Notch2 signaling in maintaining renal branching morphogenesis. CONCLUSIONS Our findings obtained by this spatiotemporal gene program are expected to improve the current understanding of kidney development.
Collapse
Affiliation(s)
- Hongwei Wu
- Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, 518020, Guangdong, China.,Institute of Nephrology and Blood Purification, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, 510632, China
| | - Fanna Liu
- Institute of Nephrology and Blood Purification, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, 510632, China
| | - Yu Shangguan
- Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, 518020, Guangdong, China
| | - Yane Yang
- Shenzhen Far East Women & Children Hospital, Shenzhen, 518000, Guangdong, China
| | - Wei Shi
- Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, 518020, Guangdong, China
| | - Wenlong Hu
- Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, 518020, Guangdong, China
| | - Zhipeng Zeng
- Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, 518020, Guangdong, China
| | - Nan Hu
- Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, 518020, Guangdong, China
| | - Xinzhou Zhang
- Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, 518020, Guangdong, China
| | - Berthold Hocher
- Department of Medicine Nephrology, Medical Faculty, Mannheim Heidelberg University, 68167, Mannheim, Germany
| | - Donge Tang
- Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, 518020, Guangdong, China.
| | - Lianghong Yin
- Institute of Nephrology and Blood Purification, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, 510632, China.
| | - Yong Dai
- Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, 518020, Guangdong, China. .,Institute of Nephrology and Blood Purification, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, 510632, China. .,Guangxi Key Laboratory of Metabolic Disease Research, Central Laboratory of Guilin NO. 924 Hospital, Guilin, 541002, China.
| |
Collapse
|
10
|
Comparative whole-genome transcriptome analysis in renal cell populations reveals high tissue specificity of MAPK/ERK targets in embryonic kidney. BMC Biol 2022; 20:112. [PMID: 35550069 PMCID: PMC9102746 DOI: 10.1186/s12915-022-01309-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 04/25/2022] [Indexed: 12/19/2022] Open
Abstract
Background MAPK/ERK signaling is a well-known mediator of extracellular stimuli controlling intracellular responses to growth factors and mechanical cues. The critical requirement of MAPK/ERK signaling for embryonic stem cell maintenance is demonstrated, but specific functions in progenitor regulation during embryonic development, and in particular kidney development remain largely unexplored. We previously demonstrated MAPK/ERK signaling as a key regulator of kidney growth through branching morphogenesis and normal nephrogenesis where it also regulates progenitor expansion. Here, we performed RNA sequencing-based whole-genome expression analysis to identify transcriptional MAPK/ERK targets in two distinct renal populations: the ureteric bud epithelium and the nephron progenitors. Results Our analysis revealed a large number (5053) of differentially expressed genes (DEGs) in nephron progenitors and significantly less (1004) in ureteric bud epithelium, reflecting likely heterogenicity of cell types. The data analysis identified high tissue-specificity, as only a fraction (362) of MAPK/ERK targets are shared between the two tissues. Tissue-specific MAPK/ERK targets participate in the regulation of mitochondrial energy metabolism in nephron progenitors, which fail to maintain normal mitochondria numbers in the MAPK/ERK-deficient tissue. In the ureteric bud epithelium, a dramatic decline in progenitor-specific gene expression was detected with a simultaneous increase in differentiation-associated genes, which was not observed in nephron progenitors. Our experiments in the genetic model of MAPK/ERK deficiency provide evidence that MAPK/ERK signaling in the ureteric bud maintains epithelial cells in an undifferentiated state. Interestingly, the transcriptional targets shared between the two tissues studied are over-represented by histone genes, suggesting that MAPK/ERK signaling regulates cell cycle progression and stem cell maintenance through chromosome condensation and nucleosome assembly. Conclusions Using tissue-specific MAPK/ERK inactivation and RNA sequencing in combination with experimentation in embryonic kidneys, we demonstrate here that MAPK/ERK signaling maintains ureteric bud tip cells, suggesting a regulatory role in collecting duct progenitors. We additionally deliver new mechanistic information on how MAPK/ERK signaling regulates progenitor maintenance through its effects on chromatin accessibility and energy metabolism. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-022-01309-z.
Collapse
|
11
|
Seltzsam S, Wang C, Zheng B, Mann N, Connaughton DM, Wu CHW, Schneider S, Schierbaum L, Kause F, Kolvenbach CM, Nakayama M, Dai R, Ottlewski I, Schneider R, Deutsch K, Buerger F, Klämbt V, Mao Y, Onuchic-Whitford AC, Nicolas-Frank C, Yousef K, Pantel D, Lai EW, Salmanullah D, Majmundar AJ, Bauer SB, Rodig NM, Somers MJG, Traum AZ, Stein DR, Daga A, Baum MA, Daouk GH, Tasic V, Awad HS, Eid LA, El Desoky S, Shalaby M, Kari JA, Fathy HM, Soliman NA, Mane SM, Shril S, Ferguson MA, Hildebrandt F. Reverse phenotyping facilitates disease allele calling in exome sequencing of patients with CAKUT. Genet Med 2022; 24:307-318. [PMID: 34906515 PMCID: PMC8876311 DOI: 10.1016/j.gim.2021.09.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 06/08/2021] [Accepted: 09/14/2021] [Indexed: 12/31/2022] Open
Abstract
PURPOSE Congenital anomalies of the kidneys and urinary tract (CAKUT) constitute the leading cause of chronic kidney disease in children. In total, 174 monogenic causes of isolated or syndromic CAKUT are known. However, syndromic features may be overlooked when the initial clinical diagnosis of CAKUT is made. We hypothesized that the yield of a molecular genetic diagnosis by exome sequencing (ES) can be increased by applying reverse phenotyping, by re-examining the case for signs/symptoms of the suspected clinical syndrome that results from the genetic variant detected by ES. METHODS We conducted ES in an international cohort of 731 unrelated families with CAKUT. We evaluated ES data for variants in 174 genes, in which variants are known to cause isolated or syndromic CAKUT. In cases in which ES suggested a previously unreported syndromic phenotype, we conducted reverse phenotyping. RESULTS In 83 of 731 (11.4%) families, we detected a likely CAKUT-causing genetic variant consistent with an isolated or syndromic CAKUT phenotype. In 19 of these 83 families (22.9%), reverse phenotyping yielded syndromic clinical findings, thereby strengthening the genotype-phenotype correlation. CONCLUSION We conclude that employing reverse phenotyping in the evaluation of syndromic CAKUT genes by ES provides an important tool to facilitate molecular genetic diagnostics in CAKUT.
Collapse
Affiliation(s)
- Steve Seltzsam
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Chunyan Wang
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA; Department of Nephrology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Bixia Zheng
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Nina Mann
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Dervla M Connaughton
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Chen-Han Wilfred Wu
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA; Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Sophia Schneider
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Luca Schierbaum
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Franziska Kause
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Caroline M Kolvenbach
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Makiko Nakayama
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Rufeng Dai
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Isabel Ottlewski
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Ronen Schneider
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Konstantin Deutsch
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Florian Buerger
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Verena Klämbt
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Youying Mao
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Ana C Onuchic-Whitford
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA; Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Camille Nicolas-Frank
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Kirollos Yousef
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Dalia Pantel
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA; Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Ethan W Lai
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Daanya Salmanullah
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Amar J Majmundar
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Stuart B Bauer
- Department of Urology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Nancy M Rodig
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Michael J G Somers
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Avram Z Traum
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Deborah R Stein
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Ankana Daga
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Michelle A Baum
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Ghaleb H Daouk
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Velibor Tasic
- Medical Faculty Skopje, University Children's Hospital, Skopje, North Macedonia
| | - Hazem S Awad
- Pediatric Nephrology Department, Dubai Hospital, Dubai, United Arab Emirates
| | - Loai A Eid
- Pediatric Nephrology Department, Dubai Hospital, Dubai, United Arab Emirates
| | - Sherif El Desoky
- Department of Pediatrics, King Abdul Aziz University, Jeddah, Saudi Arabia; Pediatric Nephrology Center of Excellence, Department of Pediatrics, King Abdul Aziz University, Jeddah, Saudi Arabia
| | - Mohammed Shalaby
- Department of Pediatrics, King Abdul Aziz University, Jeddah, Saudi Arabia; Pediatric Nephrology Center of Excellence, Department of Pediatrics, King Abdul Aziz University, Jeddah, Saudi Arabia
| | - Jameela A Kari
- Department of Pediatrics, King Abdul Aziz University, Jeddah, Saudi Arabia; Pediatric Nephrology Center of Excellence, Department of Pediatrics, King Abdul Aziz University, Jeddah, Saudi Arabia
| | - Hanan M Fathy
- Pediatric Nephrology Unit, University of Alexandria, Alexandria, Egypt
| | - Neveen A Soliman
- Department of Pediatrics, Center of Pediatric Nephrology and Transplantation, Kasr Al Ainy School of Medicine, Cairo University, Cairo, Egypt
| | - Shrikant M Mane
- Department of Genetics, Yale University School of Medicine, New Haven, CT
| | - Shirlee Shril
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Michael A Ferguson
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Friedhelm Hildebrandt
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA.
| |
Collapse
|
12
|
Wang IY, Chung CF, Babayeva S, Sogomonian T, Torban E. Loss of Planar Cell Polarity Effector Fuzzy Causes Renal Hypoplasia by Disrupting Several Signaling Pathways. J Dev Biol 2021; 10:jdb10010001. [PMID: 35076510 PMCID: PMC8788523 DOI: 10.3390/jdb10010001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/09/2021] [Accepted: 12/19/2021] [Indexed: 12/20/2022] Open
Abstract
In vertebrates, the planar cell polarity (PCP) pathway regulates tissue morphogenesis during organogenesis, including the kidney. Mutations in human PCP effector proteins have been associated with severe syndromic ciliopathies. Importantly, renal hypoplasia has been reported in some patients. However, the developmental disturbance that causes renal hypoplasia is unknown. Here, we describe the early onset of profound renal hypoplasia in mice homozygous for null mutation of the PCP effector gene, Fuzzy. We found that this phenotype is caused by defective branching morphogenesis of the ureteric bud (UB) in the absence of defects in nephron progenitor specification or in early steps of nephrogenesis. By using various experimental approaches, we show that the loss of Fuzzy affects multiple signaling pathways. Specifically, we found mild involvement of GDNF/c-Ret pathway that drives UB branching. We noted the deficient expression of molecules belonging to the Bmp, Fgf and Shh pathways. Analysis of the primary cilia in the UB structures revealed a significant decrease in ciliary length. We conclude that renal hypoplasia in the mouse Fuzzy mutants is caused by defective UB branching associated with dysregulation of ciliary and non-ciliary signaling pathways. Our work suggests a PCP effector-dependent pathogenetic mechanism that contributes to renal hypoplasia in mice and humans.
Collapse
Affiliation(s)
- Irene-Yanran Wang
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada; (I.-Y.W.); (C.-F.C.); (S.B.); (T.S.)
- McGill University Health Center Research Institute, Montreal, QC H4A 3J1, Canada
| | - Chen-Fang Chung
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada; (I.-Y.W.); (C.-F.C.); (S.B.); (T.S.)
- McGill University Health Center Research Institute, Montreal, QC H4A 3J1, Canada
| | - Sima Babayeva
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada; (I.-Y.W.); (C.-F.C.); (S.B.); (T.S.)
- McGill University Health Center Research Institute, Montreal, QC H4A 3J1, Canada
| | - Tamara Sogomonian
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada; (I.-Y.W.); (C.-F.C.); (S.B.); (T.S.)
- McGill University Health Center Research Institute, Montreal, QC H4A 3J1, Canada
| | - Elena Torban
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada; (I.-Y.W.); (C.-F.C.); (S.B.); (T.S.)
- McGill University Health Center Research Institute, Montreal, QC H4A 3J1, Canada
- Correspondence:
| |
Collapse
|
13
|
Makayes Y, Resnick E, Hinden L, Aizenshtein E, Shlomi T, Kopan R, Nechama M, Volovelsky O. Increasing mTORC1 Pathway Activity or Methionine Supplementation during Pregnancy Reverses the Negative Effect of Maternal Malnutrition on the Developing Kidney. J Am Soc Nephrol 2021; 32:1898-1912. [PMID: 33958489 PMCID: PMC8455268 DOI: 10.1681/asn.2020091321] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 03/01/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Low nephron number at birth is associated with a high risk of CKD in adulthood because nephrogenesis is completed in utero. Poor intrauterine environment impairs nephron endowment via an undefined molecular mechanism. A calorie-restricted diet (CRD) mouse model examined the effect of malnutrition during pregnancy on nephron progenitor cells (NPCs). METHODS Daily caloric intake was reduced by 30% during pregnancy. mRNA expression, the cell cycle, and metabolic activity were evaluated in sorted Six2 NPCs. The results were validated using transgenic mice, oral nutrient supplementation, and organ cultures. RESULTS Maternal CRD is associated with low nephron number in offspring, compromising kidney function at an older age. RNA-seq identified cell cycle regulators and the mTORC1 pathway, among other pathways, that maternal malnutrition in NPCs modifies. Metabolomics analysis of NPCs singled out the methionine pathway as crucial for NPC proliferation and maintenance. Methionine deprivation reduced NPC proliferation and lowered NPC number per tip in embryonic kidney cultures, with rescue from methionine metabolite supplementation. Importantly, in vivo, the negative effect of caloric restriction on nephrogenesis was prevented by adding methionine to the otherwise restricted diet during pregnancy or by removing one Tsc1 allele in NPCs. CONCLUSIONS These findings show that mTORC1 signaling and methionine metabolism are central to the cellular and metabolic effects of malnutrition during pregnancy on NPCs, contributing to nephrogenesis and later, to kidney health in adulthood.
Collapse
Affiliation(s)
- Yaniv Makayes
- Pediatric Nephrology Unit and Research Lab, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Elad Resnick
- Pediatric Nephrology Unit and Research Lab, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Liad Hinden
- Faculty of Medicine, School of Pharmacy, Institute for Drug Research, The Hebrew University, Jerusalem, Israel
| | | | | | - Raphael Kopan
- Division of Developmental Biology, Department of Pediatrics, University of Cincinnati College of Medicine and Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Morris Nechama
- Pediatric Nephrology Unit and Research Lab, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel
- Wohl’s Translation Research Institute at Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Oded Volovelsky
- Pediatric Nephrology Unit and Research Lab, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Israel
- Wohl’s Translation Research Institute at Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
14
|
Generation of patterned kidney organoids that recapitulate the adult kidney collecting duct system from expandable ureteric bud progenitors. Nat Commun 2021; 12:3641. [PMID: 34131121 PMCID: PMC8206157 DOI: 10.1038/s41467-021-23911-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Accepted: 05/21/2021] [Indexed: 01/19/2023] Open
Abstract
Current kidney organoids model development and diseases of the nephron but not the contiguous epithelial network of the kidney’s collecting duct (CD) system. Here, we report the generation of an expandable, 3D branching ureteric bud (UB) organoid culture model that can be derived from primary UB progenitors from mouse and human fetal kidneys, or generated de novo from human pluripotent stem cells. In chemically-defined culture conditions, UB organoids generate CD organoids, with differentiated principal and intercalated cells adopting spatial assemblies reflective of the adult kidney’s collecting system. Aggregating 3D-cultured nephron progenitor cells with UB organoids in vitro results in a reiterative process of branching morphogenesis and nephron induction, similar to kidney development. Applying an efficient gene editing strategy to remove RET activity, we demonstrate genetically modified UB organoids can model congenital anomalies of kidney and urinary tract. Taken together, these platforms will facilitate an enhanced understanding of development, regeneration and diseases of the mammalian collecting duct system. Here, the authors model the collecting duct system in kidneys by taking ureteric bud (UB) progenitor cells from both mouse and human primary tissues, as well as from hESC and hiPSC to generate organoids, which can model congenital anomalies of the kidney and urinary tract.
Collapse
|
15
|
Torban E, Sokol SY. Planar cell polarity pathway in kidney development, function and disease. Nat Rev Nephrol 2021; 17:369-385. [PMID: 33547419 PMCID: PMC8967065 DOI: 10.1038/s41581-021-00395-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2021] [Indexed: 02/08/2023]
Abstract
Planar cell polarity (PCP) refers to the coordinated orientation of cells in the tissue plane. Originally discovered and studied in Drosophila melanogaster, PCP is now widely recognized in vertebrates, where it is implicated in organogenesis. Specific sets of PCP genes have been identified. The proteins encoded by these genes become asymmetrically distributed to opposite sides of cells within a tissue plane and guide many processes that include changes in cell shape and polarity, collective cell movements or the uniform distribution of cell appendages. A unifying characteristic of these processes is that they often involve rearrangement of actomyosin. Mutations in PCP genes can cause malformations in organs of many animals, including humans. In the past decade, strong evidence has accumulated for a role of the PCP pathway in kidney development including outgrowth and branching morphogenesis of ureteric bud and podocyte development. Defective PCP signalling has been implicated in the pathogenesis of developmental kidney disorders of the congenital anomalies of the kidney and urinary tract spectrum. Understanding the origins, molecular constituents and cellular targets of PCP provides insights into the involvement of PCP molecules in normal kidney development and how dysfunction of PCP components may lead to kidney disease.
Collapse
Affiliation(s)
- Elena Torban
- McGill University and McGill University Health Center Research Institute, 1001 Boulevard Decarie, Block E, Montreal, Quebec, Canada, H4A3J1.,Corresponding authors: Elena Torban (); Sergei Sokol ()
| | - Sergei Y. Sokol
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, 10029, USA,Corresponding authors: Elena Torban (); Sergei Sokol ()
| |
Collapse
|
16
|
Li H, Kurtzeborn K, Kupari J, Gui Y, Siefker E, Lu B, Mätlik K, Olfat S, Montaño-Rodríguez AR, Huh SH, Costantini F, Andressoo JO, Kuure S. Postnatal prolongation of mammalian nephrogenesis by excess fetal GDNF. Development 2021; 148:268366. [PMID: 34032268 PMCID: PMC8180252 DOI: 10.1242/dev.197475] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 04/26/2021] [Indexed: 01/21/2023]
Abstract
Nephron endowment, defined during the fetal period, dictates renal and related cardiovascular health throughout life. We show here that, despite its negative effects on kidney growth, genetic increase of GDNF prolongs the nephrogenic program beyond its normal cessation. Multi-stage mechanistic analysis revealed that excess GDNF maintains nephron progenitors and nephrogenesis through increased expression of its secreted targets and augmented WNT signaling, leading to a two-part effect on nephron progenitor maintenance. Abnormally high GDNF in embryonic kidneys upregulates its known targets but also Wnt9b and Axin2, with concomitant deceleration of nephron progenitor proliferation. Decline of GDNF levels in postnatal kidneys normalizes the ureteric bud and creates a permissive environment for continuation of the nephrogenic program, as demonstrated by morphologically and molecularly normal postnatal nephron progenitor self-renewal and differentiation. These results establish that excess GDNF has a bi-phasic effect on nephron progenitors in mice, which can faithfully respond to GDNF dosage manipulation during the fetal and postnatal period. Our results suggest that sensing the signaling activity level is an important mechanism through which GDNF and other molecules contribute to nephron progenitor lifespan specification. Summary: Dosage of neurotropic factor GDNF regulates nephron progenitors and in utero growth factor augmentation can extend postnatal lifespan and differentiation of nephron progenitors.
Collapse
Affiliation(s)
- Hao Li
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland.,Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland
| | - Kristen Kurtzeborn
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland.,Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland
| | - Jussi Kupari
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Yujuan Gui
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Edward Siefker
- Department of Developmental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68105, USA
| | - Benson Lu
- Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032, USA
| | - Kärt Mätlik
- Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland.,Department of Pharmacology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Soophie Olfat
- Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland.,Department of Pharmacology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Ana R Montaño-Rodríguez
- Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland.,Department of Pharmacology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Sung-Ho Huh
- Department of Developmental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68105, USA
| | - Franklin Costantini
- Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032, USA
| | - Jaan-Olle Andressoo
- Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland.,Department of Pharmacology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland.,Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Satu Kuure
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland.,GM-unit, Laboratory Animal Centre, Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland
| |
Collapse
|
17
|
Li H, Hohenstein P, Kuure S. Embryonic Kidney Development, Stem Cells and the Origin of Wilms Tumor. Genes (Basel) 2021; 12:genes12020318. [PMID: 33672414 PMCID: PMC7926385 DOI: 10.3390/genes12020318] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/17/2021] [Accepted: 02/18/2021] [Indexed: 12/23/2022] Open
Abstract
The adult mammalian kidney is a poorly regenerating organ that lacks the stem cells that could replenish functional homeostasis similarly to, e.g., skin or the hematopoietic system. Unlike a mature kidney, the embryonic kidney hosts at least three types of lineage-specific stem cells that give rise to (a) a ureter and collecting duct system, (b) nephrons, and (c) mesangial cells together with connective tissue of the stroma. Extensive interest has been raised towards these embryonic progenitor cells, which are normally lost before birth in humans but remain part of the undifferentiated nephrogenic rests in the pediatric renal cancer Wilms tumor. Here, we discuss the current understanding of kidney-specific embryonic progenitor regulation in the innate environment of the developing kidney and the types of disruptions in their balanced regulation that lead to the formation of Wilms tumor.
Collapse
Affiliation(s)
- Hao Li
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, FIN-00014 Helsinki, Finland;
| | - Peter Hohenstein
- Department of Human Genetics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
| | - Satu Kuure
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, FIN-00014 Helsinki, Finland;
- GM-Unit, Laboratory Animal Center, Helsinki Institute of Life Science, University of Helsinki, FIN-00014 Helsinki, Finland
- Correspondence: ; Tel.: +358-2941-59395
| |
Collapse
|
18
|
Rooney KM, Woolf AS, Kimber SJ. Towards Modelling Genetic Kidney Diseases with Human Pluripotent Stem Cells. Nephron Clin Pract 2021; 145:285-296. [PMID: 33774632 DOI: 10.1159/000514018] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 12/19/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Kidney disease causes major suffering and premature mortality worldwide. With no cure for kidney failure currently available, and with limited options for treatment, there is an urgent need to develop effective pharmaceutical interventions to slow or prevent kidney disease progression. SUMMARY In this review, we consider the feasibility of using human pluripotent stem cell-derived kidney tissues, or organoids, to model genetic kidney disease. Notable successes have been made in modelling genetic tubular diseases (e.g., cystinosis), polycystic kidney disease, and medullary cystic kidney disease. Organoid models have also been used to test novel therapies that ameliorate aberrant cell biology. Some progress has been made in modelling congenital glomerular disease, even though glomeruli within organoids are developmentally immature. Less progress has been made in modelling structural kidney malformations, perhaps because sufficiently mature metanephric mesenchyme-derived nephrons, ureteric bud-derived branching collecting ducts, and a prominent stromal cell population are not generated together within a single protocol. Key Messages: We predict that the field will advance significantly if organoids can be generated with a full complement of cell lineages and with kidney components displaying key physiological functions, such as glomerular filtration. The future economic upscaling of reproducible organoid generation will facilitate more widespread research applications, including the potential therapeutic application of these stem cell-based technologies.
Collapse
Affiliation(s)
- Kirsty M Rooney
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Adrian S Woolf
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
- Royal Manchester Children's Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Susan J Kimber
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
19
|
Zhou X, Wang Y, Shao B, Wang C, Hu P, Qiao F, Xu Z. Molecular diagnostic in fetuses with isolated congenital anomalies of the kidney and urinary tract by whole-exome sequencing. J Clin Lab Anal 2020; 34:e23480. [PMID: 32779812 PMCID: PMC7676188 DOI: 10.1002/jcla.23480] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/11/2020] [Accepted: 06/18/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND In prenatal care, accumulating evidences has demonstrated that whole-exome sequencing (WES) expedites the genetic diagnosis of fetal structural anomalies. However, the clinical value of WES in the diagnosis of prenatal isolated congenital anomalies of the kidney and urinary tract (CAKUT) is unknown. METHODS Forty-one fetuses with unexplained isolated CAKUT, normal karyotype and negative chromosomal microarray analysis (CMA) results, underwent WES and were accordingly grouped as (a) Group 1: complex cases with bilateral renal abnormalities (N = 19); and (b) Group 2: cases with isolated unilateral fetal renal abnormalities (N = 22). RESULTS The detection rate of WES for pathogenic variants and incidental variants was 7.32% (3/41) and 2.4% (1/41), respectively. The three pathogenic variants were identified in the genes ACTA2 (multisystem smooth muscle dysfunction syndrome), PKHD1 (autosomal recessive form of polycystic kidney disease), and PKD1 (autosomal dominant polycystic kidney disease type 1). The incidental variants were detected in genes PPM1D (syndromic neurodevelopmental disorders). Furthermore, all above fetuses carrying pathogenic variants came from bilateral kidney anomalies. Thus, the detection rate was 0 for fetuses with unilateral fetal renal abnormalities and 15.7% (3/19) for bilateral renal abnormalities. CONCLUSION This cohort shows that prenatal WES is a supplementary approach for the etiologic diagnosis of unexplained isolated CAKUT with negative CMA, especially for fetuses with bilateral renal abnormality.
Collapse
Affiliation(s)
- Xiaoyan Zhou
- State Key Laboratory of Reproductive MedicineDepartment of Prenatal DiagnosisWomen's Hospital of Nanjing Medical UniversityNanjingChina
- Department of ObstetricsThe Affiliated Huai an No. 1 People's Hospital of NanjingMedical UniversityHuai anChina
| | - Yan Wang
- State Key Laboratory of Reproductive MedicineDepartment of Prenatal DiagnosisWomen's Hospital of Nanjing Medical UniversityNanjingChina
| | - Binbin Shao
- State Key Laboratory of Reproductive MedicineDepartment of Prenatal DiagnosisWomen's Hospital of Nanjing Medical UniversityNanjingChina
| | - Chen Wang
- State Key Laboratory of Reproductive MedicineDepartment of Prenatal DiagnosisWomen's Hospital of Nanjing Medical UniversityNanjingChina
| | - Ping Hu
- State Key Laboratory of Reproductive MedicineDepartment of Prenatal DiagnosisWomen's Hospital of Nanjing Medical UniversityNanjingChina
| | - Fengchang Qiao
- State Key Laboratory of Reproductive MedicineDepartment of Prenatal DiagnosisWomen's Hospital of Nanjing Medical UniversityNanjingChina
| | - Zhengfeng Xu
- State Key Laboratory of Reproductive MedicineDepartment of Prenatal DiagnosisWomen's Hospital of Nanjing Medical UniversityNanjingChina
| |
Collapse
|
20
|
El Andalousi J, Khairallah H, Zhuang Y, Ryan AK, Gupta IR. Role of Claudins in Renal Branching Morphogenesis. Physiol Rep 2020; 8:e14492. [PMID: 32975899 PMCID: PMC7518295 DOI: 10.14814/phy2.14492] [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: 05/18/2020] [Revised: 05/25/2020] [Accepted: 05/26/2020] [Indexed: 11/26/2022] Open
Abstract
Claudins are a family of tight junction proteins that are expressed during mouse kidney development. They regulate paracellular transport of solutes along the nephron and contribute to the final composition of the urinary filtrate. To understand their roles during development, we used a protein reagent, a truncated version of the Clostridium perfringens enterotoxin (C-CPE), to specifically remove a subset of claudin family members from mouse embryonic kidney explants at embryonic day 12. We observed that treatment with C-CPE decreased the number and the complexity of ureteric bud tips that formed: there were more single and less bifid ureteric bud tips when compared to control-treated explants. In addition, C-CPE-treated explants exhibited ureteric bud tips with larger lumens when compared to control explants (p < .05). Immunofluorescent analysis revealed decreased expression and localization of Claudin-3, -4, -6, and -8 to tight junctions of ureteric bud tips following treatment with C-CPE. Interestingly, Claudin-7 showed higher expression in the basolateral membrane of the ureteric bud lineage and poor localization to the tight junctions of the ureteric bud lineage both in controls and in C-CPE-treated explants. Taken together, it appears that claudin proteins may play a role in ureteric bud branching morphogenesis through changes in lumen formation that may affect the efficiency by which ureteric buds emerge and branch.
Collapse
Affiliation(s)
- Jasmine El Andalousi
- Research Institute of McGill University Health CentreMontreal Children's HospitalMontréalQuébecCanada
| | - Halim Khairallah
- Department of Human GeneticsMcGill UniversityMontréalQuébecCanada
| | - Yuan Zhuang
- Department of Human GeneticsMcGill UniversityMontréalQuébecCanada
| | - Aimee K. Ryan
- Department of Human GeneticsMcGill UniversityMontréalQuébecCanada
- Department of PediatricsMontreal Children's HospitalMcGill UniversityMontréalQuébecCanada
| | - Indra R. Gupta
- Department of Human GeneticsMcGill UniversityMontréalQuébecCanada
- Department of PediatricsMontreal Children's HospitalMcGill UniversityMontréalQuébecCanada
| |
Collapse
|
21
|
Harer MW, Charlton JR, Tipple TE, Reidy KJ. Preterm birth and neonatal acute kidney injury: implications on adolescent and adult outcomes. J Perinatol 2020; 40:1286-1295. [PMID: 32277164 DOI: 10.1038/s41372-020-0656-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 03/02/2020] [Accepted: 03/11/2020] [Indexed: 02/06/2023]
Abstract
As a result of preterm birth, immature kidneys are exposed to interventions in the NICU that promote survival, but are nephrotoxic. Furthermore, the duration of renal development may be truncated in these vulnerable neonates. Immaturity and nephrotoxic exposures predispose preterm newborns to acute kidney injury (AKI), particularly in the low birth weight and extremely preterm gestational age groups. Several studies have associated preterm birth as a risk factor for future chronic kidney disease (CKD). However, only a few publications have investigated the impact of neonatal AKI on CKD development. Here, we will review the evidence linking preterm birth and AKI in the NICU to CKD and highlight the knowledge gaps and opportunities for future research. For neonatal intensive care studies, we propose the inclusion of AKI as an important short-term morbidity outcome and CKD findings such as a reduced glomerular filtration rate in the assessment of long-term outcomes.
Collapse
Affiliation(s)
- Matthew W Harer
- Department of Pediatrics, Division of Neonatology, University of Wisconsin-Madison, Madison, WI, USA
| | - Jennifer R Charlton
- Department of Pediatrics, Division of Nephrology, University of Virginia Children's Hospital, Box 800386, Charlottesville, VA, USA.
| | - Trent E Tipple
- Department of Pediatrics, Section of Neonatal-Perinatal Medicine, University of Oklahoma College of Medicine, Oklahoma City, OK, USA
| | - Kimberly J Reidy
- Department of Pediatrics, Division of Pediatric Nephrology, Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
22
|
Hilliard SA, Li Y, Dixon A, El-Dahr SS. Mdm4 controls ureteric bud branching via regulation of p53 activity. Mech Dev 2020; 163:103616. [PMID: 32464196 DOI: 10.1016/j.mod.2020.103616] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 05/19/2020] [Accepted: 05/21/2020] [Indexed: 02/07/2023]
Abstract
The antagonism between Mdm2 and its close homolog Mdm4 (also known as MdmX) and p53 is vital for embryogenesis and organogenesis. Previously, we demonstrated that targeted disruption of Mdm2 in the Hoxb7+ ureteric bud (Ub) lineage, which gives rise to the renal collecting system, causes renal hypodysplasia culminating in perinatal lethality. In this study, we examine the unique role of Mdm4 in establishing the collecting duct system of the murine kidney. Hoxb7Cre driven loss of Mdm4 in the Ub lineage (UbMdm4-/-) disrupts branching morphogenesis and triggers UB cell apoptosis. UbMdm4-/- kidneys exhibit abnormally dilated Ub tips while the medulla is hypoplastic. These structural alterations result in secondary depletion of nephron progenitors and nascent nephrons. As a result, newborn UbMdm4-/- mice have hypo-dysplastic kidneys. Transcriptional profiling revealed downregulation of the Ret-tyrosine kinase pathway components, Gdnf, Wnt11, Sox8, Etv4 and Cxcr4 in the UbMdm4-/- mice relative to controls. Moreover, the expression levels of the canonical Wnt signaling members Axin2 and Wnt9b are downregulated. Mdm4 deletion upregulated p53 activity and p53-target gene expression including Cdkn1a (p21), Gdf15, Ccng1, PERP, and Fas. Germline loss of p53 in UbMdm4-/- mice largely rescues kidney development and terminal differentiation of the collecting duct. We conclude that Mdm4 plays a unique and vital role in Ub branching morphogenesis and collecting system development.
Collapse
Affiliation(s)
- Sylvia A Hilliard
- Tulane University School of Medicine, Department of Pediatrics, Section of Pediatric Nephrology, New Orleans, LA 70112, United States of America
| | - Yuwen Li
- Tulane University School of Medicine, Department of Pediatrics, Section of Pediatric Nephrology, New Orleans, LA 70112, United States of America
| | - Angelina Dixon
- Tulane University School of Medicine, Department of Pediatrics, Section of Pediatric Nephrology, New Orleans, LA 70112, United States of America
| | - Samir S El-Dahr
- Tulane University School of Medicine, Department of Pediatrics, Section of Pediatric Nephrology, New Orleans, LA 70112, United States of America.
| |
Collapse
|
23
|
Huang B, Liu Z, Vonk A, Zeng Z, Li Z. Epigenetic regulation of kidney progenitor cells. Stem Cells Transl Med 2020; 9:655-660. [PMID: 32163228 PMCID: PMC7214665 DOI: 10.1002/sctm.19-0289] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 01/26/2020] [Accepted: 02/17/2020] [Indexed: 12/15/2022] Open
Abstract
The reciprocal interactions among the different embryonic kidney progenitor populations lay the basis for proper kidney organogenesis. During kidney development, three types of progenitor cells, including nephron progenitor cells, ureteric bud progenitor cells, and interstitial progenitor cells, generate the three major kidney structures—the nephrons, the collecting duct network, and the stroma, respectively. Epigenetic mechanisms are well recognized for playing important roles in organism development, in fine‐tuned control of physiological activities, and in responses to environment stimuli. Recently, evidence supporting the importance of epigenetic mechanisms underlying kidney organogenesis has emerged. In this perspective, we summarize the research progress and discuss the potential contribution of novel stem cell, organoid, and next‐generation sequencing tools in advancing this field in the future.
Collapse
Affiliation(s)
- Biao Huang
- Division of Nephrology and Hypertension, Department of Medicine and USC/UKRO Kidney Research Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Zhenqing Liu
- Division of Oral Biology and Medicine, School of Dentistry, UCLA, Los Angeles, California, USA
| | - Ariel Vonk
- Division of Nephrology and Hypertension, Department of Medicine and USC/UKRO Kidney Research Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Zipeng Zeng
- Division of Nephrology and Hypertension, Department of Medicine and USC/UKRO Kidney Research Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Zhongwei Li
- Division of Nephrology and Hypertension, Department of Medicine and USC/UKRO Kidney Research Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
24
|
Rutledge EA, Lindström NO, Michos O, McMahon AP. Genetic manipulation of ureteric bud tip progenitors in the mammalian kidney through an Adamts18 enhancer driven tet-on inducible system. Dev Biol 2020; 458:164-176. [PMID: 31734175 PMCID: PMC6995766 DOI: 10.1016/j.ydbio.2019.11.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 11/13/2019] [Accepted: 11/13/2019] [Indexed: 11/22/2022]
Abstract
The ureteric epithelial progenitor (UEP) population within the embryonic kidney generates the arborized epithelial network of the kidney's collecting system and plays a critical role in the expansion and induction of the surrounding nephron progenitor pool. Adamts18 shows UEP- restricted expression in the kidney and progenitor tip-restricted expression in several other organs undergoing branching epithelial growth. Adamts18 is encoded by 23 exons. Genetic removal of genomic sequence spanning exons 1 to 3 led to a specific loss of Adamts18 expression in UEPs, suggesting this region may encode a UEP-specific enhancer. Intron 2 (3 kb) was shown to have enhancer activity driving expression of the doxycycline inducible tet-on transcriptional regulator (rtTA) in an Adamts18en-rtTA transgenic mouse strain. Crossing Adamts18en-rtTA mice to a doxycycline dependent GFP reporter mouse enabled the live imaging of embryonic kidney explants. This facilitated the analysis of ureteric epithelial branching events at the cellular level. Ablation of UEPs at the initiation of ureteric bud outgrowth through the doxycycline-mediated induction of Diphtheria Toxin A (DTA) generated a range of phenotypes from complete kidneys agenesis, to duplex kidneys with double ureters. The latter outcome points to the potential of regulative processes to restore UEPs. In contrast, overexpression of YAP prior to ureteric bud outgrowth led to a complete failure of kidney development. Elevating YAP levels at later stages retarded branching growth. A similar phenotype was observed with the overexpression of MYC within the branch-tip localized UEP population. These experiments showcase the utility of the Adamts18en-rtTA transgenic model to the investigation of cellular and molecular events specific to branch tip progenitors within the mammalian kidney complementing existing CRE-dependent genetic tools. Further, the illustrative examples point to areas where new insight may be gained into the regulation of UEP programs.
Collapse
Affiliation(s)
- Elisabeth A Rutledge
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad-CIRM Center for Regenerative Medicine and Stem Cell Research, W.M. Keck School of Medicine of the University of Southern California, CA, 90089, USA
| | - Nils O Lindström
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad-CIRM Center for Regenerative Medicine and Stem Cell Research, W.M. Keck School of Medicine of the University of Southern California, CA, 90089, USA
| | - Odysse Michos
- Department of Biosystems, Science and Engineering (D-BSSE), ETH Zurich, Basel, 4058, Switzerland
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad-CIRM Center for Regenerative Medicine and Stem Cell Research, W.M. Keck School of Medicine of the University of Southern California, CA, 90089, USA.
| |
Collapse
|
25
|
Rutledge EA, McMahon AP. Mutational analysis of genes with ureteric progenitor cell-specific expression in branching morphogenesis of the mouse kidney. Dev Dyn 2020; 249:765-774. [PMID: 32017326 DOI: 10.1002/dvdy.157] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 01/09/2020] [Accepted: 01/27/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Ureteric progenitor cells (UPCs) within the branch tips of the arborizing ureteric epithelium of the kidney's developing collecting system establish the shape and cellular organization of the collecting network, and drive the nephrogenic program through their interactions with nephron progenitor cells. In a previous study, expression screening identified a cohort of genes showing UPC-enriched expression including D17H6S56E-5, Hs3st3a1, Hs3st3b1, and Tmem59l. Each of these is also enriched in branch tips of assembling airways of the developing lungs. Here, we used Crispr-CAS9 directed gene editing to mutate each of these targets to address their potential role(s) in UPC programs. RESULTS Single (D17H6S56E-5 and Tmem59l) and double (Hs3st3a1 and Hs3st3b1) mutants were viable, fertile, and displayed varying frequencies of ureter duplications and no overt lung phenotype. Ureter duplications arise spontaneously through multiple outgrowths of the ureteric bud at the onset of kidney development. Tmem59l mutants and Hs3st3a1/Hs3st3b1 compound mutants showed a weakly penetrant, but statistically significant increase in duplicated ureters compared to C57BL6/J and SW wild-type mouse strains. CONCLUSIONS Tmem59l and Hs3st3a1/Hs3st3b1 activities contribute to the regulatory programs restricting ureteric outgrowth in the developing mouse kidney. However, the low penetrance of the observed phenotype precludes a detailed analysis of their specific actions.
Collapse
Affiliation(s)
- Elisabeth A Rutledge
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad-CIRM Center for Regenerative Medicine and Stem Cell Research, W.M. Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad-CIRM Center for Regenerative Medicine and Stem Cell Research, W.M. Keck School of Medicine of the University of Southern California, Los Angeles, California
| |
Collapse
|
26
|
Kuure S, Sariola H. Mouse Models of Congenital Kidney Anomalies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1236:109-136. [PMID: 32304071 DOI: 10.1007/978-981-15-2389-2_5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Congenital anomalies of the kidney and urinary tract (CAKUT) are common birth defects, which cause the majority of chronic kidney diseases in children. CAKUT covers a wide range of malformations that derive from deficiencies in embryonic kidney and lower urinary tract development, including renal aplasia, hypodysplasia, hypoplasia, ectopia, and different forms of ureter abnormalities. The majority of the genetic causes of CAKUT remain unknown. Research on mutant mice has identified multiple genes that critically regulate renal differentiation. The data generated from this research have served as an excellent resource to identify the genetic bases of human kidney defects and have led to significantly improved diagnostics. Furthermore, genetic data from human CAKUT studies have also revealed novel genes regulating kidney differentiation.
Collapse
Affiliation(s)
- Satu Kuure
- GM-Unit, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland. .,Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland. .,Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - Hannu Sariola
- Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Paediatric Pathology, HUSLAB, Helsinki University Central Hospital, Helsinki, Finland
| |
Collapse
|
27
|
Kanda S, Ohmuraya M, Akagawa H, Horita S, Yoshida Y, Kaneko N, Sugawara N, Ishizuka K, Miura K, Harita Y, Yamamoto T, Oka A, Araki K, Furukawa T, Hattori M. Deletion in the Cobalamin Synthetase W Domain-Containing Protein 1 Gene Is associated with Congenital Anomalies of the Kidney and Urinary Tract. J Am Soc Nephrol 2020; 31:139-147. [PMID: 31862704 PMCID: PMC6934996 DOI: 10.1681/asn.2019040398] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Accepted: 10/02/2019] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND Researchers have identified about 40 genes with mutations that result in the most common cause of CKD in children, congenital anomalies of the kidney and urinary tract (CAKUT), but approximately 85% of patients with CAKUT lack mutations in these genes. The anomalies that comprise CAKUT are clinically heterogenous, and thought to be caused by disturbances at different points in kidney development. However, identification of novel CAKUT-causing genes remains difficult because of their variable expressivity, incomplete penetrance, and heterogeneity. METHODS We investigated two generations of a family that included two siblings with CAKUT. Although the parents and another child were healthy, the two affected siblings presented the same manifestations, unilateral renal agenesis and contralateral renal hypoplasia. To search for a novel causative gene of CAKUT, we performed whole-exome and whole-genome sequencing of DNA from the family members. We also generated two lines of genetically modified mice with a gene deletion present only in the affected siblings, and performed immunohistochemical and phenotypic analyses of these mice. RESULTS We found that the affected siblings, but not healthy family members, had a homozygous deletion in the Cobalamin Synthetase W Domain-Containing Protein 1 (CBWD1) gene. Whole-genome sequencing uncovered genomic breakpoints, which involved exon 1 of CBWD1, harboring the initiating codon. Immunohistochemical analysis revealed high expression of Cbwd1 in the nuclei of the ureteric bud cells in the developing kidneys. Cbwd1-deficient mice showed CAKUT phenotypes, including hydronephrosis, hydroureters, and duplicated ureters. CONCLUSIONS The identification of a deletion in CBWD1 gene in two siblings with CAKUT implies a role for CBWD1 in the etiology of some cases of CAKUT.
Collapse
Affiliation(s)
- Shoichiro Kanda
- Department of Pediatrics, The University of Tokyo, Tokyo, Japan;
- Department of Pediatric Nephrology
| | - Masaki Ohmuraya
- Department of Genetics, Hyogo College of Medicine, Hyogo, Japan
| | - Hiroyuki Akagawa
- Tokyo Women's Medical University Institute for Integrated Medical Sciences, Tokyo, Japan
| | - Shigeru Horita
- Department of Pathology, Kidney Center, School of Medicine, and
| | | | | | | | | | | | - Yutaka Harita
- Department of Pediatrics, The University of Tokyo, Tokyo, Japan
| | - Toshiyuki Yamamoto
- Tokyo Women's Medical University Institute for Integrated Medical Sciences, Tokyo, Japan
- Institute of Medical Genetics, Tokyo Women's Medical University, Tokyo, Japan
| | - Akira Oka
- Department of Pediatrics, The University of Tokyo, Tokyo, Japan
| | - Kimi Araki
- Division of Developmental Genetics, Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, Japan; and
| | - Toru Furukawa
- Tokyo Women's Medical University Institute for Integrated Medical Sciences, Tokyo, Japan
- Department of Investigative Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | |
Collapse
|
28
|
Dapkunas A, Rantanen V, Gui Y, Lalowski M, Sainio K, Kuure S, Sariola H. Simple 3D culture of dissociated kidney mesenchyme mimics nephron progenitor niche and facilitates nephrogenesis Wnt-independently. Sci Rep 2019; 9:13433. [PMID: 31530822 PMCID: PMC6748995 DOI: 10.1038/s41598-019-49526-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 08/21/2019] [Indexed: 02/06/2023] Open
Abstract
Kidney mesenchyme (KM) and nephron progenitors (NPs) depend on WNT activity, and their culture in vitro requires extensive repertoire of recombinant proteins and chemicals. Here we established a robust, simple culture of mouse KM using a combination of 3D Matrigel and growth media supplemented with Fibroblast Growth Factor 2 (FGF2) and Src inhibitor PP2. This allows dissociated KM to spontaneously self-organize into spheres. To reassess the requirement of WNT activity in KM self-organization and NPs maintenance, cells were cultured with short pulse of high-dose GSK3β inhibitor BIO, on a constant low-dose or without BIO. Robust proliferation at 48 hours and differentiation at 1 week were observed in cultures with high BIO pulse. Importantly, dissociated KM cultured without BIO, similarly to that exposed to constant low dose of BIO, maintained NPs up to one week and spontaneously differentiated into nephron tubules at 3 weeks of culture. Our results show that KM is maintained and induced to differentiate in a simple culture system. They also imply that GSK3β/WNT-independent pathways contribute to the maintenance and induction of mouse KM. The robust and easy 3D culture enables further characterization of NPs, and may facilitate disease modeling when applied to human cells.
Collapse
Affiliation(s)
- Arvydas Dapkunas
- Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, FI-00014, Helsinki, Finland. .,Meilahti Clinical Proteomics Core Facility, Helsinki Institute of Life Science, University of Helsinki, FI-00014, Helsinki, Finland.
| | - Ville Rantanen
- Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, FI-00014, Helsinki, Finland.,Genome-Scale Biology Research Program, Faculty of Medicine, University of Helsinki, FI-00014, Helsinki, Finland
| | - Yujuan Gui
- Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, FI-00014, Helsinki, Finland
| | - Maciej Lalowski
- Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, FI-00014, Helsinki, Finland.,Meilahti Clinical Proteomics Core Facility, Helsinki Institute of Life Science, University of Helsinki, FI-00014, Helsinki, Finland
| | - Kirsi Sainio
- Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, FI-00014, Helsinki, Finland
| | - Satu Kuure
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, FI-00014, Helsinki, Finland.,GM-unit, Laboratory Animal Centre, Helsinki Institute of Life Science, University of Helsinki, FI-00014, Helsinki, Finland
| | - Hannu Sariola
- Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, FI-00014, Helsinki, Finland
| |
Collapse
|
29
|
Development of the urogenital system is regulated via the 3'UTR of GDNF. Sci Rep 2019; 9:5302. [PMID: 30923332 PMCID: PMC6438985 DOI: 10.1038/s41598-019-40457-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 02/13/2019] [Indexed: 12/30/2022] Open
Abstract
Mechanisms controlling ureter lenght and the position of the kidney are poorly understood. Glial cell-line derived neurotrophic factor (GDNF) induced RET signaling is critical for ureteric bud outgrowth, but the function of endogenous GDNF in further renal differentiation and urogenital system development remains discursive. Here we analyzed mice where 3′ untranslated region (UTR) of GDNF is replaced with sequence less responsive to microRNA-mediated regulation, leading to increased GDNF expression specifically in cells naturally transcribing Gdnf. We demonstrate that increased Gdnf leads to short ureters in kidneys located in an abnormally caudal position thus resembling human pelvic kidneys. High GDNF levels expand collecting ductal progenitors at the expense of ureteric trunk elongation and result in expanded tip and short trunk phenotype due to changes in cell cycle length and progenitor motility. MEK-inhibition rescues these defects suggesting that MAPK-activity mediates GDNF’s effects on progenitors. Moreover, Gdnf hyper mice are infertile likely due to effects of excess GDNF on distal ureter remodeling. Our findings suggest that dysregulation of GDNF levels, for example via alterations in 3′UTR, may account for a subset of congenital anomalies of the kidney and urinary tract (CAKUT) and/or congenital infertility cases in humans and pave way to future studies.
Collapse
|
30
|
Hochane M, van den Berg PR, Fan X, Bérenger-Currias N, Adegeest E, Bialecka M, Nieveen M, Menschaart M, Chuva de Sousa Lopes SM, Semrau S. Single-cell transcriptomics reveals gene expression dynamics of human fetal kidney development. PLoS Biol 2019; 17:e3000152. [PMID: 30789893 PMCID: PMC6400406 DOI: 10.1371/journal.pbio.3000152] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 03/05/2019] [Indexed: 01/30/2023] Open
Abstract
The current understanding of mammalian kidney development is largely based on mouse models. Recent landmark studies revealed pervasive differences in renal embryogenesis between mouse and human. The scarcity of detailed gene expression data in humans therefore hampers a thorough understanding of human kidney development and the possible developmental origin of kidney diseases. In this paper, we present a single-cell transcriptomics study of the human fetal kidney. We identified 22 cell types and a host of marker genes. Comparison of samples from different developmental ages revealed continuous gene expression changes in podocytes. To demonstrate the usefulness of our data set, we explored the heterogeneity of the nephrogenic niche, localized podocyte precursors, and confirmed disease-associated marker genes. With close to 18,000 renal cells from five different developmental ages, this study provides a rich resource for the elucidation of human kidney development, easily accessible through an interactive web application.
Collapse
Affiliation(s)
- Mazène Hochane
- Leiden Institute of Physics, Leiden University, Leiden, The Netherlands
| | | | - Xueying Fan
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Esmée Adegeest
- Leiden Institute of Physics, Leiden University, Leiden, The Netherlands
| | - Monika Bialecka
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Maaike Nieveen
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Susana M. Chuva de Sousa Lopes
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Reproductive Medicine, Ghent University Hospital, Ghent, Belgium
| | - Stefan Semrau
- Leiden Institute of Physics, Leiden University, Leiden, The Netherlands
| |
Collapse
|
31
|
Abstract
Kidney organogenesis has been a widely used classical model system to study inductive tissue interactions that guide differentiation of many organs. The basis for this is in the pioneering work done during the early 1950s when the conditions of how to support ex vivo growth and differentiation of developing kidneys were revealed. Importantly, culturing developing kidneys remains as an essential instrument to advance our understanding of molecular and cellular regulation of morphogenesis even today. Despite the fact that embryonic kidneys have been cultured for decades, it is not a trivial method and requires specific anatomical and developmental biology knowledge. This chapter outlines the general steps in organ culture and details the requirements for successful kidney explant differentiation.
Collapse
|
32
|
O'Brien LL, Combes AN, Short KM, Lindström NO, Whitney PH, Cullen-McEwen LA, Ju A, Abdelhalim A, Michos O, Bertram JF, Smyth IM, Little MH, McMahon AP. Wnt11 directs nephron progenitor polarity and motile behavior ultimately determining nephron endowment. eLife 2018; 7:e40392. [PMID: 30516471 PMCID: PMC6281319 DOI: 10.7554/elife.40392] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 11/16/2018] [Indexed: 01/09/2023] Open
Abstract
A normal endowment of nephrons in the mammalian kidney requires a balance of nephron progenitor self-renewal and differentiation throughout development. Here, we provide evidence for a novel action of ureteric branch tip-derived Wnt11 in progenitor cell organization and interactions within the nephrogenic niche, ultimately determining nephron endowment. In Wnt11 mutants, nephron progenitors dispersed from their restricted niche, intermixing with interstitial progenitors. Nephron progenitor differentiation was accelerated, kidneys were significantly smaller, and the nephron progenitor pool was prematurely exhausted, halving the final nephron count. Interestingly, RNA-seq revealed no significant differences in gene expression. Live imaging of nephron progenitors showed that in the absence of Wnt11 they lose stable attachments to the ureteric branch tips, continuously detaching and reattaching. Further, the polarized distribution of several markers within nephron progenitors is disrupted. Together these data highlight the importance of Wnt11 signaling in directing nephron progenitor behavior which determines a normal nephrogenic program.
Collapse
Affiliation(s)
- Lori L O'Brien
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell ResearchKeck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Alexander N Combes
- Institute for Molecular BioscienceThe University of QueenslandBrisbaneAustralia
- Department of Anatomy and NeuroscienceThe University of MelbourneMelbourneAustralia
- Murdoch Children’s Research InstituteRoyal Children's HospitalMelbourneAustralia
| | - Kieran M Short
- Department of Anatomy and Neuroscience, Monash Biomedicine Discovery InstituteMonash UniversityMelbourneAustralia
- Development and Stem Cells Program, Monash Biomedicine Discovery InstituteMonash UniversityMelbourneAustralia
| | - Nils O Lindström
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell ResearchKeck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Peter H Whitney
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell ResearchKeck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Luise A Cullen-McEwen
- Department of Anatomy and Neuroscience, Monash Biomedicine Discovery InstituteMonash UniversityMelbourneAustralia
| | - Adler Ju
- Institute for Molecular BioscienceThe University of QueenslandBrisbaneAustralia
| | - Ahmed Abdelhalim
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell ResearchKeck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Odyssé Michos
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell ResearchKeck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - John F Bertram
- Department of Anatomy and Neuroscience, Monash Biomedicine Discovery InstituteMonash UniversityMelbourneAustralia
| | - Ian M Smyth
- Department of Anatomy and Neuroscience, Monash Biomedicine Discovery InstituteMonash UniversityMelbourneAustralia
- Development and Stem Cells Program, Monash Biomedicine Discovery InstituteMonash UniversityMelbourneAustralia
| | - Melissa H Little
- Institute for Molecular BioscienceThe University of QueenslandBrisbaneAustralia
- Department of Anatomy and NeuroscienceThe University of MelbourneMelbourneAustralia
- Murdoch Children’s Research InstituteRoyal Children's HospitalMelbourneAustralia
- Department of PediatricsUniversity of MelbourneParkvilleAustralia
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell ResearchKeck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| |
Collapse
|
33
|
Ye M, Xu L, Fu M, Chen D, Mattina T, Zufardi O, Rossi E, Bush KT, Nigam SK, Grossfeld P. Gene-targeted deletion in mice of the Ets-1 transcription factor, a candidate gene in the Jacobsen syndrome kidney "critical region," causes abnormal kidney development. Am J Med Genet A 2018; 179:71-77. [PMID: 30422383 DOI: 10.1002/ajmg.a.40481] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 04/13/2018] [Accepted: 06/29/2018] [Indexed: 11/12/2022]
Abstract
Ets-1 is a member of the Ets family of transcription factors and has critical roles in multiple biological functions. Structural kidney defects occur at an increased frequency in Jacobsen syndrome (OMIM #147791), a rare chromosomal disorder caused by deletions in distal 11q, implicating at least one causal gene in distal 11q. In this study, we define an 8.1 Mb "critical region" for kidney defects in Jacobsen syndrome, which spans ~50 genes. We demonstrate that gene-targeted deletion of Ets-1 in mice results in some of the most common congenital kidney defects occurring in Jacobsen syndrome, including: duplicated kidney, hypoplastic kidney, and dilated renal pelvis and calyces. Taken together, our results implicate Ets-1 in normal mammalian kidney development and, potentially, in the pathogenesis of some of the most common types of human structural kidney defects.
Collapse
Affiliation(s)
- Maoqing Ye
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Lian Xu
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Mengxia Fu
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Dongrui Chen
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Teresa Mattina
- Division of Medical Genetics, Department of Biomedical and Biotechnological Sciences (BIOMETEC), Catania, Italy
| | - Orsetta Zufardi
- Department of Medical Genetics, University of Pavia, Pavia, Italy
| | - Elena Rossi
- Department of Medical Genetics, University of Pavia, Pavia, Italy
| | - Kevin T Bush
- Department of Pediatrics, University of California San Diego School of Medicine, San Diego, California
| | - Sanjay K Nigam
- Department of Pediatrics, University of California San Diego School of Medicine, San Diego, California.,Department of Medicine, University of California San Diego School of Medicine, San Diego, California
| | - Paul Grossfeld
- Division of Pediatric Cardiology, University of California San Diego School of Medicine, San Diego, California
| |
Collapse
|
34
|
Kurtzeborn K, Cebrian C, Kuure S. Regulation of Renal Differentiation by Trophic Factors. Front Physiol 2018; 9:1588. [PMID: 30483151 PMCID: PMC6240607 DOI: 10.3389/fphys.2018.01588] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 10/23/2018] [Indexed: 12/11/2022] Open
Abstract
Classically, trophic factors are considered as proteins which support neurons in their growth, survival, and differentiation. However, most neurotrophic factors also have important functions outside of the nervous system. Especially essential renal growth and differentiation regulators are glial cell line-derived neurotrophic factor (GDNF), bone morphogenetic proteins (BMPs), and fibroblast growth factors (FGFs). Here we discuss how trophic factor-induced signaling contributes to the control of ureteric bud (UB) branching morphogenesis and to maintenance and differentiation of nephrogenic mesenchyme in embryonic kidney. The review includes recent advances in trophic factor functions during the guidance of branching morphogenesis and self-renewal versus differentiation decisions, both of which dictate the control of kidney size and nephron number. Creative utilization of current information may help better recapitulate renal differentiation in vitro, but it is obvious that significantly more basic knowledge is needed for development of regeneration-based renal therapies.
Collapse
Affiliation(s)
- Kristen Kurtzeborn
- Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Medicum, University of Helsinki, Helsinki, Finland
| | - Cristina Cebrian
- Developmental Biology Division, Cincinnati Children’s Hospital, Cincinnati, OH, United States
| | - Satu Kuure
- Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Medicum, University of Helsinki, Helsinki, Finland
- GM-Unit, Laboratory Animal Centre, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| |
Collapse
|
35
|
van der Ven AT, Connaughton DM, Ityel H, Mann N, Nakayama M, Chen J, Vivante A, Hwang DY, Schulz J, Braun DA, Schmidt JM, Schapiro D, Schneider R, Warejko JK, Daga A, Majmundar AJ, Tan W, Jobst-Schwan T, Hermle T, Widmeier E, Ashraf S, Amar A, Hoogstraaten CA, Hugo H, Kitzler TM, Kause F, Kolvenbach CM, Dai R, Spaneas L, Amann K, Stein DR, Baum MA, Somers MJG, Rodig NM, Ferguson MA, Traum AZ, Daouk GH, Bogdanović R, Stajić N, Soliman NA, Kari JA, El Desoky S, Fathy HM, Milosevic D, Al-Saffar M, Awad HS, Eid LA, Selvin A, Senguttuvan P, Sanna-Cherchi S, Rehm HL, MacArthur DG, Lek M, Laricchia KM, Wilson MW, Mane SM, Lifton RP, Lee RS, Bauer SB, Lu W, Reutter HM, Tasic V, Shril S, Hildebrandt F. Whole-Exome Sequencing Identifies Causative Mutations in Families with Congenital Anomalies of the Kidney and Urinary Tract. J Am Soc Nephrol 2018; 29:2348-2361. [PMID: 30143558 PMCID: PMC6115658 DOI: 10.1681/asn.2017121265] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 06/11/2018] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Congenital anomalies of the kidney and urinary tract (CAKUT) are the most prevalent cause of kidney disease in the first three decades of life. Previous gene panel studies showed monogenic causation in up to 12% of patients with CAKUT. METHODS We applied whole-exome sequencing to analyze the genotypes of individuals from 232 families with CAKUT, evaluating for mutations in single genes known to cause human CAKUT and genes known to cause CAKUT in mice. In consanguineous or multiplex families, we additionally performed a search for novel monogenic causes of CAKUT. RESULTS In 29 families (13%), we detected a causative mutation in a known gene for isolated or syndromic CAKUT that sufficiently explained the patient's CAKUT phenotype. In three families (1%), we detected a mutation in a gene reported to cause a phenocopy of CAKUT. In 15 of 155 families with isolated CAKUT, we detected deleterious mutations in syndromic CAKUT genes. Our additional search for novel monogenic causes of CAKUT in consanguineous and multiplex families revealed a potential single, novel monogenic CAKUT gene in 19 of 232 families (8%). CONCLUSIONS We identified monogenic mutations in a known human CAKUT gene or CAKUT phenocopy gene as the cause of disease in 14% of the CAKUT families in this study. Whole-exome sequencing provides an etiologic diagnosis in a high fraction of patients with CAKUT and will provide a new basis for the mechanistic understanding of CAKUT.
Collapse
Affiliation(s)
- Amelie T van der Ven
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Dervla M Connaughton
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Hadas Ityel
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Nina Mann
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Makiko Nakayama
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jing Chen
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Asaf Vivante
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Daw-Yang Hwang
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Julian Schulz
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Daniela A Braun
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - David Schapiro
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ronen Schneider
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jillian K Warejko
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ankana Daga
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Amar J Majmundar
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Weizhen Tan
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Tilman Jobst-Schwan
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Tobias Hermle
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Eugen Widmeier
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Shazia Ashraf
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ali Amar
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Charlotte A Hoogstraaten
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Hannah Hugo
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Thomas M Kitzler
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Franziska Kause
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Caroline M Kolvenbach
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Rufeng Dai
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Leslie Spaneas
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Kassaundra Amann
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Deborah R Stein
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Michelle A Baum
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Michael J G Somers
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Nancy M Rodig
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Michael A Ferguson
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Avram Z Traum
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ghaleb H Daouk
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Radovan Bogdanović
- Department of Pediatric Nephrology, Institute for Mother and Child Health Care, Belgrade, Serbia
| | - Natasa Stajić
- Department of Pediatric Nephrology, Institute for Mother and Child Health Care, Belgrade, Serbia
| | - Neveen A Soliman
- Department of Pediatrics, Center of Pediatric Nephrology and Transplantation, Cairo University, Egypt
- Egyptian Group for Orphan Renal Diseases, Cairo, Egypt
| | - Jameela A Kari
- Department of Pediatrics and
- Pediatric Nephrology Center of Excellence, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Sherif El Desoky
- Department of Pediatrics and
- Pediatric Nephrology Center of Excellence, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Hanan M Fathy
- Pediatric Nephrology Unit, University of Alexandria, Alexandria, Egypt
| | - Danko Milosevic
- University of Zagreb School of Medicine, University Hospital Center Zagreb, Zagreb, Croatia
| | - Muna Al-Saffar
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
- United Arab Emirates University, Abu Dhabi, United Arab Emirates
| | - Hazem S Awad
- Pediatric Nephrology Department, Dubai Kidney Center Of Excellence, Dubai Hospital, Dubai, United Arab Emirates
| | - Loai A Eid
- Pediatric Nephrology Department, Dubai Kidney Center Of Excellence, Dubai Hospital, Dubai, United Arab Emirates
| | - Aravind Selvin
- Department of Pediatric Nephrology, Institute of Child Health and Hospital for Children, The Tamil Nadu Dr. M.G.R. Medical University, Chennai, Tamil Nadu, India
| | - Prabha Senguttuvan
- Department of Pediatric Nephrology, Dr. Mehta's Multi-Specialty Hospital, Chennai, Tamil Nadu, India
| | | | - Heidi L Rehm
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, Massachusetts
| | - Daniel G MacArthur
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, Massachusetts
- Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | - Monkol Lek
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, Massachusetts
- Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | - Kristen M Laricchia
- Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | - Michael W Wilson
- Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | - Shrikant M Mane
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut
| | - Richard P Lifton
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut
- Rockefeller University, New York, New York
| | - Richard S Lee
- Department of Urology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Stuart B Bauer
- Department of Urology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Weining Lu
- Renal Section, Department of Medicine and Pathology, Boston University Medical Center, Boston, Massachusetts
| | - Heiko M Reutter
- Institute of Human Genetics and
- Department of Neonatology and Pediatric Intensive Care, Children's Hospital, University of Bonn, Bonn, Germany; and
| | - Velibor Tasic
- Medical Faculty Skopje, University Children's Hospital, Skopje, Macedonia
| | - Shirlee Shril
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Friedhelm Hildebrandt
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts;
| |
Collapse
|
36
|
Kimura H, Nishikawa M, Yanagawa N, Nakamura H, Miyamoto S, Hamon M, Hauser P, Zhao L, Jo OD, Komeya M, Ogawa T, Yanagawa N. Effect of fluid shear stress on in vitro cultured ureteric bud cells. BIOMICROFLUIDICS 2018; 12:044107. [PMID: 30034570 PMCID: PMC6039298 DOI: 10.1063/1.5035328] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Accepted: 06/26/2018] [Indexed: 05/08/2023]
Abstract
Most kidney cells are continuously exposed to fluid shear stress (FSS) from either blood flow or urine flow. Recent studies suggest that changes in FSS could contribute to the function and injury of these kidney cells. However, it is unclear whether FSS influences kidney development when urinary flow starts in the embryonic kidneys. In this study, we evaluated the influence of FSS on in vitro cultured ureteric bud (UB) cells by using a pumpless microfluidic device, which offers the convenience of conducting parallel cell culture experiments while also eliminating the need for cumbersome electronic driven equipment and intricate techniques. We first validated the function of the device by both mathematical model and experimental measurements. UB cells dissected from E15.5 mouse embryonic kidneys were cultured in the pumpless microfluidic device and subjected to FSS in the range of 0.4-0.6 dyn mm-2 for 48 h (dynamic). Control UB cells were similarly cultured in the device and maintained under a no-flow condition (static). We found from our present study that the exposure to FSS for up to 48 h led to an increase in mRNA expression levels of UB tip cell marker genes (Wnt11, Ret, Etv4) with a decrease in stalk cell marker genes (Wnt7b, Tacstd2). In further support of the enrichment of UB tip cell population in response to FSS, we also found that exposure to FSS led to a remarkable reduction in the binding of lectin Dolichos Biflorus Agglutinin. In conclusion, results of our present study show that exposure to FSS led to an enrichment in UB tip cell populations, which could contribute to the development and function of the embryonic kidney when urine flow starts at around embryonic age E15.5 in mouse. Since UB tip cells are known to be the proliferative progenitor cells that contribute to the branching morphogenesis of the collecting system in the kidney, our finding could imply an important link between the FSS from the initiation of urine flow and the development and function of the kidney.
Collapse
Affiliation(s)
| | | | | | - Hiroko Nakamura
- Department of Mechanical Engineering, School of Engineering, Tokai University, Hiratsuka, Kanagawa 259-1292, Japan
| | - Shunsuke Miyamoto
- Department of Mechanical Engineering, School of Engineering, Tokai University, Hiratsuka, Kanagawa 259-1292, Japan
| | | | | | - Lifu Zhao
- Medical and Research Services, Greater Los Angeles Veterans Affairs Healthcare System at Sepulveda, North Hills, California 91343, USA
| | - Oak D. Jo
- Medical and Research Services, Greater Los Angeles Veterans Affairs Healthcare System at Sepulveda, North Hills, California 91343, USA
| | - Mitsuru Komeya
- Department of Urology, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa 236-0004, Japan
| | | | | |
Collapse
|
37
|
Butterworth MB. The tale of two (distal nephron) cell types. Am J Physiol Renal Physiol 2018; 314:F930-F931. [PMID: 29363323 DOI: 10.1152/ajprenal.00044.2018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- Michael B Butterworth
- Department of Cell Biology, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| |
Collapse
|
38
|
Tumelty KE, Higginson-Scott N, Fan X, Bajaj P, Knowlton KM, Shamashkin M, Coyle AJ, Lu W, Berasi SP. Identification of direct negative cross-talk between the SLIT2 and bone morphogenetic protein-Gremlin signaling pathways. J Biol Chem 2018; 293:3039-3055. [PMID: 29317497 DOI: 10.1074/jbc.m117.804021] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 12/06/2017] [Indexed: 12/28/2022] Open
Abstract
Slit guidance ligand 2 (SLIT2) is a large, secreted protein that binds roundabout (ROBO) receptors on multiple cell types, including neurons and kidney podocytes. SLIT2-ROBO-mediated signaling regulates neuronal migration and ureteric bud (UB) outgrowth during kidney development as well as glomerular filtration in adult kidneys. Additionally, SLIT2 binds Gremlin, an antagonist of bone morphogenetic proteins (BMPs), and BMP-Gremlin signaling also regulates UB formation. However, direct cross-talk between the ROBO2-SLIT2 and BMP-Gremlin signaling pathways has not been established. Here, we report the discovery of negative feedback between the SLIT2 and BMP-Gremlin signaling pathways. We found that the SLIT2-Gremlin interaction inhibited both SLIT2-ROBO2 signaling in neurons and Gremlin antagonism of BMP activity in myoblasts and fibroblasts. Furthermore, BMP2 down-regulated SLIT2 expression and promoter activity through canonical BMP signaling. Gremlin treatment, BMP receptor inhibition, and SMAD family member 4 (SMAD4) knockdown rescued BMP-mediated repression of SLIT2. BMP2 treatment of nephron progenitor cells derived from human embryonic stem cells decreased SLIT2 expression, further suggesting an interaction between the BMP2-Gremlin and SLIT2 pathways in human kidney cells. In conclusion, our study has revealed direct negative cross-talk between two pathways, previously thought to be unassociated, that may regulate both kidney development and adult tissue maintenance.
Collapse
Affiliation(s)
- Kathleen E Tumelty
- From the Centers for Therapeutic Innovation, Pfizer Inc., Cambridge, Massachusetts 02139
| | - Nathan Higginson-Scott
- From the Centers for Therapeutic Innovation, Pfizer Inc., Cambridge, Massachusetts 02139
| | - Xueping Fan
- the Renal Section, Department of Medicine, Boston University Medical Center, Boston, Massachusetts 02118, and
| | - Piyush Bajaj
- the Drug Safety Research and Development, Pfizer Inc., Groton, Connecticut 06340
| | - Kelly M Knowlton
- From the Centers for Therapeutic Innovation, Pfizer Inc., Cambridge, Massachusetts 02139
| | - Michael Shamashkin
- From the Centers for Therapeutic Innovation, Pfizer Inc., Cambridge, Massachusetts 02139
| | - Anthony J Coyle
- From the Centers for Therapeutic Innovation, Pfizer Inc., Cambridge, Massachusetts 02139
| | - Weining Lu
- the Renal Section, Department of Medicine, Boston University Medical Center, Boston, Massachusetts 02118, and
| | - Stephen P Berasi
- From the Centers for Therapeutic Innovation, Pfizer Inc., Cambridge, Massachusetts 02139,
| |
Collapse
|
39
|
Wang H, Zhang C, Wang X, Lian Y, Guo B, Han M, Zhang X, Zhu X, Xu S, Guo Z, Bi Y, Shen Q, Wang X, Liu J, Zhuang Y, Ni T, Xu H, Wu X. Disruption of Gen1 Causes Congenital Anomalies of the Kidney and Urinary Tract in Mice. Int J Biol Sci 2018; 14:10-20. [PMID: 29483821 PMCID: PMC5821045 DOI: 10.7150/ijbs.22768] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Accepted: 10/02/2017] [Indexed: 01/20/2023] Open
Abstract
Congenital anomalies of the kidney and urinary tract (CAKUT) are among the most common developmental defects in humans. Despite of several known CAKUT-related loci (HNF1B, PAX2, EYA1, etc.), the genetic etiology of CAKUT remains to be elucidated for most patients. In this study, we report that disruption of the Holliday Junction resolvase gene Gen1 leads to renal agenesis, duplex kidney, hydronephrosis, and vesicoureteral reflux (VUR) in mice. GEN1 interacts with SIX1 and enhances the transcriptional activity of SIX1/EYA1, a key regulatory complex of the GDNF morphogen. Gen1 mutation impairs Grem1 and Gdnf expression, resulting in excessive ureteric bud formation and defective ureteric bud branching during early kidney development. These results revealed an unidentified role of GEN1 in kidney development and suggested its contribution to CAKUT.
Collapse
Affiliation(s)
- Herui Wang
- State Key Laboratory of Genetic Engineering and National Center for International Research of Development and Disease, Institute of Developmental Biology and Molecular Medicine, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai 200433, China.,Shanghai Kidney Development and Pediatric Kidney Disease Research Center, Children's Hospital of Fudan University, Shanghai 201102, China.,Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Chi Zhang
- State Key Laboratory of Genetic Engineering and National Center for International Research of Development and Disease, Institute of Developmental Biology and Molecular Medicine, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai 200433, China.,Shanghai Kidney Development and Pediatric Kidney Disease Research Center, Children's Hospital of Fudan University, Shanghai 201102, China.,Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, CO 80309, USA
| | - Xiaowen Wang
- Shanghai Kidney Development and Pediatric Kidney Disease Research Center, Children's Hospital of Fudan University, Shanghai 201102, China.,Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430000, China
| | - Yaru Lian
- State Key Laboratory of Genetic Engineering and National Center for International Research of Development and Disease, Institute of Developmental Biology and Molecular Medicine, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai 200433, China.,MOE Key Laboratory of Contemporary Anthropology, Fudan University, Shanghai 200433, China
| | - Bin Guo
- State Key Laboratory of Genetic Engineering and National Center for International Research of Development and Disease, Institute of Developmental Biology and Molecular Medicine, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Miao Han
- State Key Laboratory of Genetic Engineering and National Center for International Research of Development and Disease, Institute of Developmental Biology and Molecular Medicine, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai 200433, China.,MOE Key Laboratory of Contemporary Anthropology, Fudan University, Shanghai 200433, China
| | - Xiaoe Zhang
- Shanghai Kidney Development and Pediatric Kidney Disease Research Center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Xiaoting Zhu
- State Key Laboratory of Genetic Engineering and National Center for International Research of Development and Disease, Institute of Developmental Biology and Molecular Medicine, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Sixian Xu
- State Key Laboratory of Genetic Engineering and National Center for International Research of Development and Disease, Institute of Developmental Biology and Molecular Medicine, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Zengli Guo
- State Key Laboratory of Genetic Engineering and National Center for International Research of Development and Disease, Institute of Developmental Biology and Molecular Medicine, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Yunli Bi
- Shanghai Kidney Development and Pediatric Kidney Disease Research Center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Qian Shen
- Shanghai Kidney Development and Pediatric Kidney Disease Research Center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Xiang Wang
- Shanghai Kidney Development and Pediatric Kidney Disease Research Center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Jiaojiao Liu
- Shanghai Kidney Development and Pediatric Kidney Disease Research Center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Yuan Zhuang
- State Key Laboratory of Genetic Engineering and National Center for International Research of Development and Disease, Institute of Developmental Biology and Molecular Medicine, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai 200433, China.,Department of Immunology, Duke University Medical Center, Durham, NC 27710, USA
| | - Ting Ni
- State Key Laboratory of Genetic Engineering and National Center for International Research of Development and Disease, Institute of Developmental Biology and Molecular Medicine, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai 200433, China.,MOE Key Laboratory of Contemporary Anthropology, Fudan University, Shanghai 200433, China
| | - Hong Xu
- Shanghai Kidney Development and Pediatric Kidney Disease Research Center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Xiaohui Wu
- State Key Laboratory of Genetic Engineering and National Center for International Research of Development and Disease, Institute of Developmental Biology and Molecular Medicine, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai 200433, China.,Shanghai Kidney Development and Pediatric Kidney Disease Research Center, Children's Hospital of Fudan University, Shanghai 201102, China
| |
Collapse
|
40
|
Boghossian NS, Sicko RJ, Giannakou A, Dimopoulos A, Caggana M, Tsai MY, Yeung EH, Pankratz N, Cole BR, Romitti PA, Browne ML, Fan R, Liu A, Kay DM, Mills JL. Rare copy number variants identified in prune belly syndrome. Eur J Med Genet 2017; 61:145-151. [PMID: 29174092 DOI: 10.1016/j.ejmg.2017.11.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 10/31/2017] [Accepted: 11/21/2017] [Indexed: 11/26/2022]
Abstract
Prune belly syndrome (PBS), also known as Eagle-Barrett syndrome, is a rare congenital disorder characterized by absence or hypoplasia of the abdominal wall musculature, urinary tract anomalies, and cryptorchidism in males. The etiology of PBS is largely unresolved, but genetic factors are implicated given its recurrence in families. We examined cases of PBS to identify novel pathogenic copy number variants (CNVs). A total of 34 cases (30 males and 4 females) with PBS identified from all live births in New York State (1998-2005) were genotyped using Illumina HumanOmni2.5 microarrays. CNVs were prioritized if they were absent from in-house controls, encompassed ≥10 consecutive probes, were ≥20 Kb in size, had ≤20% overlap with common variants in population reference controls, and had ≤20% overlap with any variant previously detected in other birth defect phenotypes screened in our laboratory. We identified 17 candidate autosomal CNVs; 10 cases each had one CNV and four cases each had two CNVs. The CNVs included a 158 Kb duplication at 4q22 that overlaps the BMPR1B gene; duplications of different sizes carried by two cases in the intron of STIM1 gene; a 67 Kb duplication 202 Kb downstream of the NOG gene, and a 1.34 Mb deletion including the MYOCD gene. The identified rare CNVs spanned genes involved in mesodermal, muscle, and urinary tract development and differentiation, which might help in elucidating the genetic contribution to PBS. We did not have parental DNA and cannot identify whether these CNVs were de novo or inherited. Further research on these CNVs, particularly BMP signaling is warranted to elucidate the pathogenesis of PBS.
Collapse
Affiliation(s)
- Nansi S Boghossian
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC, United States; Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States.
| | - Robert J Sicko
- Division of Genetics, Wadsworth Center, Department of Health, Albany, NY, United States
| | - Andreas Giannakou
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Aggeliki Dimopoulos
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Michele Caggana
- Division of Genetics, Wadsworth Center, Department of Health, Albany, NY, United States
| | - Michael Y Tsai
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Edwina H Yeung
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Nathan Pankratz
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Benjamin R Cole
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Paul A Romitti
- Department of Epidemiology, College of Public Health, The University of Iowa, Iowa City, IA, United States
| | - Marilyn L Browne
- New York State Department of Health, Congenital Malformations Registry, Albany, NY, United States; University at Albany School of Public Health, Rensselaer, NY, United States
| | - Ruzong Fan
- Department of Biostatistics, Bioinformatics, and Biomathematics, Georgetown University Medical Center (GUMC), Washington, DC, United States
| | - Aiyi Liu
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Denise M Kay
- Division of Genetics, Wadsworth Center, Department of Health, Albany, NY, United States
| | - James L Mills
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
41
|
Desgrange A, Heliot C, Skovorodkin I, Akram SU, Heikkilä J, Ronkainen VP, Miinalainen I, Vainio SJ, Cereghini S. HNF1B controls epithelial organization and cell polarity during ureteric bud branching and collecting duct morphogenesis. Development 2017; 144:4704-4719. [PMID: 29158444 DOI: 10.1242/dev.154336] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 11/03/2017] [Indexed: 12/16/2022]
Abstract
Kidney development depends crucially on proper ureteric bud branching giving rise to the entire collecting duct system. The transcription factor HNF1B is required for the early steps of ureteric bud branching, yet the molecular and cellular events regulated by HNF1B are poorly understood. We report that specific removal of Hnf1b from the ureteric bud leads to defective cell-cell contacts and apicobasal polarity during the early branching events. High-resolution ex vivo imaging combined with a membranous fluorescent reporter strategy show decreased mutant cell rearrangements during mitosis-associated cell dispersal and severe epithelial disorganization. Molecular analysis reveals downregulation of Gdnf-Ret pathway components and suggests that HNF1B acts both upstream and downstream of Ret signaling by directly regulating Gfra1 and Etv5 Subsequently, Hnf1b deletion leads to massively mispatterned ureteric tree network, defective collecting duct differentiation and disrupted tissue architecture, which leads to cystogenesis. Consistently, mRNA-seq analysis shows that the most impacted genes encode intrinsic cell-membrane components with transporter activity. Our study uncovers a fundamental and recurring role of HNF1B in epithelial organization during early ureteric bud branching and in further patterning and differentiation of the collecting duct system in mouse.
Collapse
Affiliation(s)
- Audrey Desgrange
- Sorbonne Universités, UPMC Université Paris 06, IBPS - UMR7622, F-75005 Paris, France .,CNRS, UMR7622, Institut de Biologie Paris-Seine (IBPS) - Developmental Biology Laboratory, F-75005 Paris, France
| | - Claire Heliot
- Sorbonne Universités, UPMC Université Paris 06, IBPS - UMR7622, F-75005 Paris, France.,CNRS, UMR7622, Institut de Biologie Paris-Seine (IBPS) - Developmental Biology Laboratory, F-75005 Paris, France
| | - Ilya Skovorodkin
- Faculty of Biochemistry and Molecular Medicine, Biocenter, University of Oulu; Laboratory of Developmental Biology, Biocenter Oulu and InfoTech, Department of Medical Biochemistry and Molecular Medicine, Oulu Center for Cell Matrix Research, 90220 Oulu, Finland
| | - Saad U Akram
- Center for Machine Vision Research and Signal Analysis (CMVS), University of Oulu, FIN-90014, Oulu, Finland
| | - Janne Heikkilä
- Center for Machine Vision Research and Signal Analysis (CMVS), University of Oulu, FIN-90014, Oulu, Finland
| | | | | | - Seppo J Vainio
- Faculty of Biochemistry and Molecular Medicine, Biocenter, University of Oulu; Laboratory of Developmental Biology, Biocenter Oulu and InfoTech, Department of Medical Biochemistry and Molecular Medicine, Oulu Center for Cell Matrix Research, 90220 Oulu, Finland
| | - Silvia Cereghini
- Sorbonne Universités, UPMC Université Paris 06, IBPS - UMR7622, F-75005 Paris, France .,CNRS, UMR7622, Institut de Biologie Paris-Seine (IBPS) - Developmental Biology Laboratory, F-75005 Paris, France
| |
Collapse
|
42
|
van der Ven AT, Vivante A, Hildebrandt F. Novel Insights into the Pathogenesis of Monogenic Congenital Anomalies of the Kidney and Urinary Tract. J Am Soc Nephrol 2017; 29:36-50. [PMID: 29079659 DOI: 10.1681/asn.2017050561] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Congenital anomalies of the kidneys and urinary tract (CAKUT) comprise a large spectrum of congenital malformations ranging from severe manifestations, such as renal agenesis, to potentially milder conditions, such as vesicoureteral reflux. CAKUT causes approximately 40% of ESRD that manifests within the first three decades of life. Several lines of evidence indicate that CAKUT is often caused by recessive or dominant mutations in single (monogenic) genes. To date, approximately 40 monogenic genes are known to cause CAKUT if mutated, explaining 5%-20% of patients. However, hundreds of different monogenic CAKUT genes probably exist. The discovery of novel CAKUT-causing genes remains challenging because of this pronounced heterogeneity, variable expressivity, and incomplete penetrance. We here give an overview of known genetic causes for human CAKUT and shed light on distinct renal morphogenetic pathways that were identified as relevant for CAKUT in mice and humans.
Collapse
Affiliation(s)
- Amelie T van der Ven
- Divison of Nephrology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Asaf Vivante
- Divison of Nephrology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Friedhelm Hildebrandt
- Divison of Nephrology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
43
|
Minuth W. Reading First Coordinates from the Nephrogenic Zone in Human Fetal Kidney. Nephron Clin Pract 2017; 138:137-146. [DOI: 10.1159/000481441] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 09/11/2017] [Indexed: 01/15/2023] Open
|
44
|
Naiman N, Fujioka K, Fujino M, Valerius MT, Potter SS, McMahon AP, Kobayashi A. Repression of Interstitial Identity in Nephron Progenitor Cells by Pax2 Establishes the Nephron-Interstitium Boundary during Kidney Development. Dev Cell 2017; 41:349-365.e3. [PMID: 28535371 DOI: 10.1016/j.devcel.2017.04.022] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 03/10/2017] [Accepted: 04/24/2017] [Indexed: 12/31/2022]
Abstract
The kidney contains the functional units, the nephrons, surrounded by the renal interstitium. Previously we discovered that, once Six2-expressing nephron progenitor cells and Foxd1-expressing renal interstitial progenitor cells form at the onset of kidney development, descendant cells from these populations contribute exclusively to the main body of nephrons and renal interstitial tissues, respectively, indicating a lineage boundary between the nephron and renal interstitial compartments. Currently it is unclear how lineages are regulated during kidney organogenesis. We demonstrate that nephron progenitor cells lacking Pax2 fail to differentiate into nephron cells but can switch fates into renal interstitium-like cell types. These data suggest that Pax2 function maintains nephron progenitor cells by repressing a renal interstitial cell program. Thus, the lineage boundary between the nephron and renal interstitial compartments is maintained by the Pax2 activity in nephron progenitor cells during kidney organogenesis.
Collapse
Affiliation(s)
- Natalie Naiman
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Kaoru Fujioka
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Mari Fujino
- Department of Medicine, Institute for Stem Cell and Regenerative Medicine, University of Washington, 750 Republican Street, Seattle, WA 98109, USA
| | - M Todd Valerius
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - S Steven Potter
- Division of Developmental Biology, Cincinnati Children's Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad-CIRM Center for Regenerative Medicine and Stem Cell Research, W.M. Keck School of Medicine of the University of Southern California, 1425 San Pablo Street, Los Angeles, CA 90033, USA
| | - Akio Kobayashi
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA; Department of Medicine, Institute for Stem Cell and Regenerative Medicine, University of Washington, 750 Republican Street, Seattle, WA 98109, USA.
| |
Collapse
|
45
|
Rutledge EA, Benazet JD, McMahon AP. Cellular heterogeneity in the ureteric progenitor niche and distinct profiles of branching morphogenesis in organ development. Development 2017; 144:3177-3188. [PMID: 28705898 DOI: 10.1242/dev.149112] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 07/10/2017] [Indexed: 12/26/2022]
Abstract
Branching morphogenesis creates arborized epithelial networks. In the mammalian kidney, an epithelial progenitor pool at ureteric branch tips (UBTs) creates the urine-transporting collecting system. Using region-specific mouse reporter strains, we performed an RNA-seq screen, identifying tip- and stalk-enriched gene sets in the developing collecting duct system. Detailed in situ hybridization studies of tip-enriched predictions identified UBT-enriched gene sets conserved between the mouse and human kidney. Comparative spatial analysis of their UBT niche expression highlighted distinct patterns of gene expression revealing novel molecular heterogeneity within the UBT progenitor population. To identify kidney-specific and shared programs of branching morphogenesis, comparative expression studies on the developing mouse lung were combined with in silico analysis of the developing mouse salivary gland. These studies highlight a shared gene set with multi-organ tip enrichment and a gene set specific to UBTs. This comprehensive analysis extends our current understanding of the ureteric branch tip niche.
Collapse
Affiliation(s)
- Elisabeth A Rutledge
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad-CIRM Center for Regenerative Medicine and Stem Cell Research, W.M. Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
| | - Jean-Denis Benazet
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad-CIRM Center for Regenerative Medicine and Stem Cell Research, W.M. Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA.,Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, CA 94143, USA
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad-CIRM Center for Regenerative Medicine and Stem Cell Research, W.M. Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|
46
|
El-Dahr SS, Li Y, Liu J, Gutierrez E, Hering-Smith KS, Signoretti S, Pignon JC, Sinha S, Saifudeen Z. p63+ ureteric bud tip cells are progenitors of intercalated cells. JCI Insight 2017; 2:89996. [PMID: 28469077 DOI: 10.1172/jci.insight.89996] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 03/23/2017] [Indexed: 01/13/2023] Open
Abstract
During renal branching morphogenesis, ureteric bud tip cells (UBTC) serve as the progenitor epithelium for all cell types of the collecting duct. While the transcriptional circuitry of ureteric bud (UB) branching has been intensively studied, the transcriptional control of UBTC differentiation has been difficult to ascertain. This is partly due to limited knowledge of UBTC-specific transcription factors that mark the progenitor state. Here, we identify the transcription factor p63 (also known as TP63), a master regulator of basal stem cells in stratified epithelia, as a specific marker of mouse and human UBTC. Nuclear p63 marks Ret+ UBTC transiently and is silenced by the end of nephrogenesis. Lineage tracing revealed that a subset of UBTC expressing the ΔNp63 isoform (N-terminus truncated p63) is dedicated to generating cortical intercalated cells. Germline targeting of ΔNp63 in mice caused a marked reduction in intercalated cells near the time of birth, indicating that p63 not only marks UBTC, but also is essential for their differentiation. We conclude that the choice of UBTC progenitors to differentiate is determined earlier than previously recognized and that UBTC progenitors are prepatterned and fate restricted. These findings prompt the rethinking of current paradigms of collecting duct differentiation and may have implications for regenerative renal medicine.
Collapse
Affiliation(s)
| | | | | | | | | | - Sabina Signoretti
- Department of Pathology, Brigham and Women's Hospital, Harvard School of Medicine, Boston, Massachusetts, USA
| | - Jean-Christophe Pignon
- Department of Pathology, Brigham and Women's Hospital, Harvard School of Medicine, Boston, Massachusetts, USA
| | - Satrajit Sinha
- Department of Biochemistry, Jacobs School of Medicine, University of Buffalo, New York, USA
| | | |
Collapse
|
47
|
Abstract
Congenital abnormalities of the kidney and urinary tract (CAKUT) are one of the leading congenital defects to be identified on prenatal ultrasound. CAKUT represent a broad spectrum of abnormalities, from transient hydronephrosis to severe bilateral renal agenesis. CAKUT are a major contributor to chronic and end stage kidney disease (CKD/ESKD) in children. Prenatal imaging is useful to identify CAKUT, but will not detect all defects. Both genetic abnormalities and the fetal environment contribute to CAKUT. Monogenic gene mutations identified in human CAKUT have advanced our understanding of molecular mechanisms of renal development. Low nephron number and solitary kidneys are associated with increased risk of adult onset CKD and ESKD. Premature and low birth weight infants represent a high risk population for low nephron number. Additional research is needed to identify biomarkers and appropriate follow-up of premature and low birth weight infants into adulthood.
Collapse
Affiliation(s)
- Stacy Rosenblum
- Department of Pediatrics/Neonatology, Children's Hospital of Montefiore/Einstein, Bronx, NY, USA
| | - Abhijeet Pal
- Department of Pediatrics/Nephrology, Children's Hospital of Montefiore/Einstein, Bronx, NY, USA
| | - Kimberly Reidy
- Department of Pediatrics/Nephrology, Children's Hospital of Montefiore/Einstein, Bronx, NY, USA.
| |
Collapse
|
48
|
Abstract
PURPOSE OF REVIEW Allogenic kidney transplantation use is limited because of a shortage of kidney organ donors and the risks associated with a long-term immunosuppression. An emerging treatment prospect is autologous transplants of ex vivo produced human kidneys. Here we will review the research advances in this area. RECENT FINDINGS The creation of human induced pluripotent cells (iPSCs) from somatic cells and the emergence of several differentiation protocols that are able to convert iPSCs cells into self-organizing kidney organoids are two large steps toward assembling a human kidney in vitro. Several groups have successfully generated urine-producing kidney organoids upon transplantation in a mouse host. Additional advances in culturing nephron progenitors in vitro may provide another source for kidney engineering, and the emergence of genome editing technology will facilitate correction of congenital mutations. SUMMARY Basic research into the development of metanephric kidneys and iPSC differentiation protocols, the therapeutic use of iPSCs, along with emergence of new technologies such as CRISPR/Cas9 genome editing have accelerated a trend that may prove transformative in the treatment of ESRD and congenital kidney disorders.
Collapse
Affiliation(s)
- Oded Volovelsky
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Division of Nephrology and Hypertension, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Raphael Kopan
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
49
|
Abstract
Over a decade ago, it was proposed that the regulation of tubular repair in the kidney might involve the recapitulation of developmental pathways. Although the kidney cannot generate new nephrons after birth, suggesting a low level of regenerative competence, the tubular epithelial cells of the nephrons can proliferate to repair the damage after AKI. However, the debate continues over whether this repair involves a persistent progenitor population or any mature epithelial cell remaining after injury. Recent reports have highlighted the expression of Sox9, a transcription factor critical for normal kidney development, during postnatal epithelial repair in the kidney. Indeed, the proliferative response of the epithelium involves expression of several pathways previously described as being involved in kidney development. In some instances, these pathways are also apparently involved in the maladaptive responses observed after repeated injury. Whether development and repair in the kidney are the same processes or we are misinterpreting the similar expression of genes under different circumstances remains unknown. Here, we review the evidence for this link, concluding that such parallels in expression may more correctly represent the use of the same pathways in a distinct context, likely triggered by similar stressors.
Collapse
Affiliation(s)
- Melissa Helen Little
- Murdoch Children's Research Institute, Melbourne, Australia; and .,Department of Pediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Australia
| | - Pamela Kairath
- Murdoch Children's Research Institute, Melbourne, Australia; and.,Department of Pediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Australia
| |
Collapse
|
50
|
Abstract
The treatment of renal failure has seen little change in the past 70 years. Patients with end-stage renal disease (ESRD) are treated with renal replacement therapy, including dialysis or organ transplantation. The growing imbalance between the availability of donor organs and prevalence of ESRD is pushing an increasing number of patients to undergo dialysis. Although the prospect of new treatment options for patients through regenerative medicine has long been suggested, advances in the generation of human kidney cell types through the directed differentiation of human pluripotent stem cells over the past 2 years have brought this prospect closer to delivery. These advances are the result of careful research into mammalian embryogenesis. By understanding the decision points made within the embryo to pattern the kidney, it is now possible to recreate self-organizing kidney tissues in vitro. In this Review, we describe the key decision points in kidney development and how these decisions have been mimicked experimentally. Recreation of human nephrons from human pluripotent stem cells opens the door to patient-derived disease models and personalized drug and toxicity screening. In the long term, we hope that these efforts will also result in the generation of bioengineered organs for the treatment of kidney disease.
Collapse
|