47 results on '"Hildebrandt, Friedhelm"'
Search Results
2. List of contributors
- Author
-
Benson, Merrill D., primary, Connaughton, Dervla M., additional, Friedman, David J., additional, Ginsburg, David, additional, Hildebrandt, Friedhelm, additional, Jalanko, Hannu, additional, Kääriäinen, Helena, additional, Luger, Selina M., additional, Morrissette, Jennifer J.D., additional, Mrug, Michal, additional, Peslak, Scott, additional, Pollak, Martin R., additional, Pyeritz, Reed E., additional, Rahbari Oskoui, Frederic, additional, Rizk, Dana V., additional, Roth, Jacquelyn J., additional, Saigusa, Takamitsu, additional, Sayani, Farzana, additional, Shavit, Jordan A., additional, Stadtmauer, Edward A., additional, Sullivan, Kathleen E., additional, and Weyand, Angela C., additional
- Published
- 2023
- Full Text
- View/download PDF
3. Nephronophthisis and Medullary Cystic Kidney Disease
- Author
-
Braun, Daniela A., primary and Hildebrandt, Friedhelm, additional
- Published
- 2018
- Full Text
- View/download PDF
4. Contributors
- Author
-
Abudayyeh, Ala, primary, Adrogué, Horacio J., additional, Allon, Michael, additional, Arif-Tiwari, Hina, additional, Barratt, Jonathan, additional, Berns, Jeffrey S., additional, Bjornstad, Petter, additional, Bomback, Andrew S., additional, Bowling, C. Barrett, additional, Braun, Daniela A., additional, Brewster, Ursula C., additional, Carson, John M., additional, Cattran, Daniel C., additional, Chandran, Sindhu, additional, Chapman, Arlene B., additional, Cherney, David, additional, Coca, Steven G., additional, Cohen, Debbie L., additional, Crawford, Brendan D., additional, Curhan, Gary C., additional, Dad, Taimur, additional, D'Agati, Vivette D., additional, Derebail, Vimal K., additional, De Vriese, An S., additional, de Zeeuw, Dick, additional, DuBose, Thomas D., additional, Emmett, Michael, additional, Evenepoel, Pieter, additional, Fairhead, Todd, additional, Falk, Ronald J., additional, Fervenza, Fernando C., additional, Finkel, Kevin W., additional, Födinger, Manuela, additional, Gbadegesin, Rasheed A., additional, Gehr, Todd W.B., additional, Gilbert, Scott J., additional, Gill, Jagbir S., additional, Gonwa, Thomas A., additional, Greenberg, Arthur, additional, Gregory, Martin C., additional, Herlitz, Leal, additional, Hildebrandt, Friedhelm, additional, Hladik, Gerald A., additional, Hladunewich, Michelle A., additional, Hoenig, Melanie P., additional, Hogan, Jonathan, additional, House, Andrew A., additional, Hutchison, Alastair J., additional, Ikizler, T. Alp, additional, Inker, Lesley A., additional, Ison, Michael G., additional, James, Matthew T., additional, Jennette, J. Charles, additional, Kain, Renate, additional, Kala, Jaya, additional, Kalantar-Zadeh, Kamyar, additional, Kalb, Bobby, additional, Kopp, Jeffrey B., additional, Knoll, Greg, additional, Kulkarni, Dhananjay P., additional, Lan, James, additional, Levey, Andrew S., additional, Lewis, Ed, additional, Linas, Stuart L., additional, Luciano, Randy L., additional, Lytvyn, Yuliya, additional, Macedo, Etienne, additional, Madias, Nicolaos E., additional, Martin, Diego R., additional, Matzke, Gary R., additional, Mehrotra, Rajnish, additional, Mehta, Ankit N., additional, Mehta, Ravindra L., additional, Meyers, Catherine M., additional, Misra, Madhukar, additional, Moe, Sharon M., additional, Nachman, Patrick H., additional, Nicolle, Lindsay E., additional, Nolin, Thomas D., additional, O'Hare, Ann M., additional, Pannu, Neesh, additional, Peixoto, Aldo J., additional, Perazella, Mark A., additional, Prochaska, Megan, additional, Provenzano, Laura Ferreira, additional, Quarles, L. Darryl, additional, Radhakrishnan, Jai, additional, Reddy, Bharathi, additional, Rizk, Dana V., additional, Ronco, Claudio, additional, Rosenberg, Avi Z., additional, Rosenblum, Norman D., additional, Sampson, Matthew G., additional, Sanders, Paul W., additional, Sarnak, Mark J., additional, Scheinman, Steven J., additional, Schnaper, H. William, additional, Schrauben, Sarah, additional, Semelka, Richard C., additional, Shirali, Anushree C., additional, Sica, Domenic A., additional, Sunder-Plassmann, Gere, additional, Sutherland, Richard W., additional, Szerlip, Harold M., additional, Kurella Tamura, Manjula, additional, Tangren, Jessica Sheehan, additional, Thurman, Joshua M., additional, Tonelli, Marcello, additional, Townsend, Raymond R., additional, Trachtman, Howard, additional, Turner, Jeffrey M., additional, Vardhan, Anand, additional, Verbalis, Joseph G., additional, Vincenti, Flavio G., additional, Vivarelli, Marina, additional, Voora, Raven, additional, Wadei, Hani M., additional, Warady, Bradley A., additional, Weidemann, Darcy K., additional, Weiner, Daniel E., additional, Whittier, William L., additional, Wilcox, Christopher S., additional, Wish, Jay B., additional, and Yeo, See Cheng, additional
- Published
- 2018
- Full Text
- View/download PDF
5. Contributors
- Author
-
Adler, Sharon, primary, Adrogué, Horacio J., additional, Allon, Michael, additional, Arif-Tiwari, Hina, additional, Arroyo, Vincente, additional, Avery, Robin K., additional, Avila-Casado, Carmen, additional, Barratt, Jonathan, additional, Berns, Jeffrey S., additional, Bomback, Andrew S., additional, Bonventre, Joseph V., additional, Bowling, C. Barrett, additional, Brewster, Ursula C., additional, Briggs, Josephine P., additional, Cattran, Daniel C., additional, Chandran, Sindhu, additional, Chapman, Arlene B., additional, Coca, Steven G., additional, Conlon, Peter J., additional, Copelovitch, Lawrence A., additional, Curhan, Gary, additional, D’Agati, Vivette D., additional, Daoud, Jacques R., additional, de Zeeuw, Dick, additional, Dennen, Paula, additional, Derebail, Vimal K., additional, DuBose, Thomas D., additional, Emmett, Michael, additional, Fairhead, Todd, additional, Falk, Ronald J., additional, Feehally, John, additional, Fernández, Javier, additional, Fervenza, Fernando C., additional, Fioretto, Paola, additional, Födinger, Manuela, additional, Furth, Susan L., additional, Gehr, Todd W.B., additional, Gilbert, Scott J., additional, Gill, Jagbir S., additional, Gipson, Debbie S., additional, Goldstein-Fuchs, D. Jordi, additional, Greenberg, Arthur, additional, Gregory, Martin C., additional, Gunaratnam, Lakshman, additional, Hakim, Raymond M., additional, Hildebrandt, Friedhelm, additional, Hladunewich, Michelle A., additional, Hogan, Jonathan, additional, Hou, Susan, additional, House, Andrew A., additional, Huan, Yonghong, additional, Hutchison, Alastair J., additional, Inker, Lesley A., additional, James, Matthew T., additional, Jayne, David, additional, Jennette, J. Charles, additional, Jiménez, Wladimiro, additional, Kain, Renate, additional, Kalantar-Zadeh, Kamyar, additional, Kalb, Bobby, additional, Knoll, Greg, additional, Kriz, Wilhelm, additional, Tamura, Manjula Kurella, additional, LaPierre, Amy Frances, additional, Lambers Heerspink, Hiddo J., additional, Levey, Andrew S., additional, Lewis, Edmund J., additional, Linas, Stuart L., additional, Macedo, Etienne, additional, Madias, Nicolaos E., additional, Magee, Colm, additional, Mariani, Laura H., additional, Martin, Diego R., additional, Matzke, Gary R., additional, McQuillan, Rory F., additional, Mehrotra, Rajnish, additional, Mehta, Ankit N., additional, Mehta, Ravindra L., additional, Meyers, Catherine M., additional, Meyrier, Alain, additional, Moe, Sharon M., additional, Nast, Cynthia C., additional, Nicolle, Lindsay E., additional, Nolin, Thomas D., additional, O’Hare, Ann M., additional, O’Toole, John F., additional, Pannu, Neesh, additional, Perazella, Mark A., additional, Pusey, Charles D., additional, Quarles, L. Darryl, additional, Radhakrishnan, Jai, additional, Rastegar, Asghar, additional, Redahan, Lynn, additional, Rizk, Dana V., additional, Ronco, Claudio, additional, Rosenblum, Norman D., additional, Salama, Alan D., additional, Sanders, Paul W., additional, Sarnak, Mark J., additional, Scheinman, Steven J., additional, Schnermann, Jurgen B., additional, Semelka, Richard C., additional, Shirali, Anushree, additional, Sica, Domenic A., additional, Sunder-Plassmann, Gere, additional, Sutherland, Richard W., additional, Szerlip, Harold M., additional, Tonelli, Marcello, additional, Townsend, Raymond R., additional, Trachtman, Howard, additional, Turner, Jeffrey M., additional, Vardhan, Anand, additional, Vellanki, Kavitha, additional, Verbalis, Joseph G., additional, Vincenti, Flavio G., additional, Waikar, Sushrut S., additional, Weiner, Daniel E., additional, White, Colin T., additional, Whittier, William L., additional, Wilcox, Christopher S., additional, Wish, Jay B., additional, and Yiu, Vivian, additional
- Published
- 2014
- Full Text
- View/download PDF
6. Nephronophthisis and Medullary Cystic Kidney Disease
- Author
-
O’Toole, John F., primary and Hildebrandt, Friedhelm, additional
- Published
- 2014
- Full Text
- View/download PDF
7. Contributors
- Author
-
Antignac, Corinne, primary, Aronson, Peter S., additional, Asch, William S., additional, Baum, Michel, additional, Beerman, Isabel, additional, Bergeron, Anne, additional, Biber, Jürg, additional, Bichet, Daniel G., additional, Bleyer, Anthony J., additional, Bowden, Donald W., additional, Boyden, Lynn M., additional, Brown, Edward M., additional, Cantley, Lloyd G., additional, Chou, Janice Y., additional, Coca, Steven G., additional, Cox, Timothy M., additional, Cramer, Scott D., additional, Cremers, Cor WJR., additional, Deen, Peter M.T., additional, Desnick, Robert J., additional, Endou, Hitoshi, additional, Esquivel, Ernie L., additional, Forster, Ian C., additional, Freedman, Barry I., additional, Geller, David S., additional, Gharavi, Ali, additional, Giebisch, Gerhard H., additional, Grubb, Robert L., additional, Guay-Woodford, Lisa M., additional, Hariri, Ali, additional, Hart, Thomas C., additional, Hartwig, Sunny, additional, Hebert, Steven C., additional, Hernando, Nati, additional, Hildebrandt, Friedhelm, additional, Hosoya, Tatsuo, additional, Hosoyamada, Makoto, additional, Ichida, Kimiyoshi, additional, Igarashi, Peter, additional, Jorquera, Rossana, additional, Kahle, Kristopher T., additional, Kalatzis, Vasiliki, additional, Karet, Fiona E., additional, Klotman, Paul E., additional, Knoers, Nine V.A.M., additional, Kreidberg, Jordan A., additional, Kwiatkowski, David Joseph, additional, Lee, Brendan, additional, Lifton, Richard P., additional, Lin, Fangming, additional, Marston Linehan, W., additional, Mansfield, Brian C., additional, Marlier, Arnaud, additional, Mistry, Pramod Kumar, additional, Moe, Orson W., additional, Mohan, Chandra, additional, Morello, Roy, additional, Murer, Heini, additional, Palacin, Manuel, additional, Patel, Vishal, additional, Patrakka, Jaakko, additional, Pollak, Martin R., additional, Polu, Krishna R., additional, Reilly, Robert F., additional, Rich, Stephen S., additional, Rossier, Bernard C., additional, Scheinman, Steven J., additional, Schild, Laurent, additional, Schilsky, Michael L., additional, Scholl, Ute I., additional, Scolari, Francesco, additional, Scott, Daryl, additional, Seldin, Donald W., additional, Somlo, Stefan, additional, Takayama, Tatsuya, additional, Tanguay, Robert M., additional, Tryggvason, Karl, additional, Wagner, Carsten A., additional, Wakeland, Edward K., additional, Walther, McClellan M., additional, Wang, Andrew, additional, Weinstein, David A., additional, White, Perrin C., additional, Wilson, Frederick H., additional, Wright, Ernest M., additional, Wyatt, Christina M., additional, and van Os, Carel H., additional
- Published
- 2009
- Full Text
- View/download PDF
8. Nephronophthisis and Medullary Cystic Kidney Disease
- Author
-
O'Toole, John F., primary and Hildebrandt, Friedhelm, additional
- Published
- 2009
- Full Text
- View/download PDF
9. Nephronophthisis
- Author
-
Hildebrandt, Friedhelm, primary
- Published
- 2009
- Full Text
- View/download PDF
10. Contributors
- Author
-
Adler, Sharon, primary, Adrogué, Horacio J., additional, Aggarwal, Nidhi, additional, Allon, Michael, additional, Anderson, Sharon, additional, Andreoli, Sharon Phillips, additional, Appel, Gerald B., additional, Arroyo, Vicente, additional, August, Phyllis, additional, Bakris, George L., additional, Balow, James E., additional, Beddhu, Srinivasan, additional, Berns, Jeffrey S., additional, Bonventre, Joseph V., additional, Braga, Larissa, additional, Briggs, Josephine P., additional, Caramori, Maria Luiza, additional, Cattran, Daniel C., additional, Chapman, Arlene B., additional, Chertow, Glenn M., additional, Cheung, Alfred K., additional, Cho, Kerry C., additional, Coffman, Thomas M., additional, Conlon, Peter J., additional, Connaire, Jeffrey J., additional, Curhan, Gary, additional, Dennen, Paula, additional, DuBose, Thomas D., additional, Ellison, David H., additional, Emmett, Michael, additional, Falk, Ronald J., additional, Fernández, Javier, additional, Goeddeke-Merickel, Catherine M., additional, Goldstein-Fuchs, D. Jordi, additional, Greenberg, Arthur, additional, Gregory, Martin C., additional, Guasch, Antonio, additional, Gunaratnam, Lakshman, additional, Henrich, William L., additional, Hildebrandt, Friedhelm, additional, Hogg, Ronald J., additional, Holley, Jean L., additional, Hsu, Chi-yuan, additional, Hutchison, Alastair J., additional, Jayne, A. David, additional, Jennette, J. Charles, additional, Jiménez, Wladimiro, additional, Kasiske, Bertram L., additional, Khosla, Nitin, additional, Klotman, Paul E., additional, Kovalik, Eugene C., additional, Kovalik, Jean-Paul, additional, Krause, Michelle Whittier, additional, Kriz, Wilhelm, additional, Levey, Andrew S., additional, Lin, Fang-Ying, additional, Linas, Stuart, additional, Madias, Nicolaos E., additional, Mannon, Roslyn B., additional, Martin, Diego R., additional, MatzkePharmD, Gary R., additional, Maver, Michael, additional, McQuillan, Rory, additional, Mehta, Ankit N., additional, Meyers, Catherine M., additional, Meyrier, Alain, additional, Moe, Sharon M., additional, Monahan, Marianne, additional, Murali, Narayana S., additional, Nast, Cynthia C., additional, Nath, Karl A., additional, Nicolle, Lindsay E., additional, O'Toole, John F., additional, Palmer, Biff F., additional, Pisoni, Roberto, additional, Podymow, Tiina, additional, Pusey, Charles D., additional, Quarles, L. Darryl, additional, Rao, Maya K., additional, Remuzzi, Giuseppe, additional, Ritz, Eberhard, additional, Saifullah, Akber, additional, Salama, Alan D., additional, Sanders, Paul W., additional, Sarnak, Mark J., additional, Scheinman, Steven J., additional, Schieppati, Arrigo, additional, Schnermann, Jürgen B., additional, Semelka, Richard C., additional, Stevens, Lesley A., additional, Stoycheff, Nicholas, additional, Szerlip, Harold M., additional, Tanenbaum, Nadine D., additional, Trachtman, Howard, additional, Verbalis, Joseph G., additional, Vardhan, Anand, additional, Weiner, Daniel E., additional, Wilcox, Christopher S., additional, Wish, Jay B., additional, Wyatt, Christina M., additional, and Ziyadeh, Fuad N., additional
- Published
- 2009
- Full Text
- View/download PDF
11. Defects in the IFT-B Component IFT172 Cause Jeune and Mainzer-Saldino Syndromes in Humans
- Author
-
Halbritter, Jan, Bizet, Albane A, Schmidts, Miriam, Porath, Jonathan D, Braun, Daniela A, Gee, Heon Yung, McInerney-Leo, Aideen M, Krug, Pauline, Filhol, Emilie, Davis, Erica E, Airik, Rannar, Czarnecki, Peter G, Lehman, Anna M, Trnka, Peter, Nitschké, Patrick, Bole-Feysot, Christine, Schueler, Markus, Knebelmann, Bertrand, Burtey, Stéphane, Szabó, Attila J, Tory, Kálmán, Leo, Paul J, Gardiner, Brooke, McKenzie, Fiona A, Zankl, Andreas, Brown, Matthew A, Hartley, Jane L, Maher, Eamonn R, Li, Chunmei, Leroux, Michel R, Scambler, Peter J, Zhan, Shing H, Jones, Steven J, Kayserili, Hülya, Tuysuz, Beyhan, Moorani, Khemchand N, Constantinescu, Alexandru, Krantz, Ian D, Kaplan, Bernard S, Shah, Jagesh V, Hurd, Toby W, Doherty, Dan, Katsanis, Nicholas, Duncan, Emma L, Otto, Edgar A, Beales, Philip L, Mitchison, Hannah M, Saunier, Sophie, and Hildebrandt, Friedhelm
- Subjects
Cytoplasmic Dyneins ,Male ,Cerebellar Ataxia ,Ellis-Van Creveld Syndrome ,Molecular Sequence Data ,Bone and Bones ,White People ,Craniosynostoses ,Asian People ,Ectodermal Dysplasia ,Report ,Genetics ,Animals ,Humans ,Genetics(clinical) ,Amino Acid Sequence ,Alleles ,Zebrafish ,Intracellular Signaling Peptides and Proteins ,Dyneins ,Epistasis, Genetic ,Fibroblasts ,Kidney Diseases, Cystic ,Phenotype ,Gene Knockdown Techniques ,Mutation ,Female ,sense organs ,Retinitis Pigmentosa - Abstract
Intraflagellar transport (IFT) depends on two evolutionarily conserved modules, subcomplexes A (IFT-A) and B (IFT-B), to drive ciliary assembly and maintenance. All six IFT-A components and their motor protein, DYNC2H1, have been linked to human skeletal ciliopathies, including asphyxiating thoracic dystrophy (ATD; also known as Jeune syndrome), Sensenbrenner syndrome, and Mainzer-Saldino syndrome (MZSDS). Conversely, the 14 subunits in the IFT-B module, with the exception of IFT80, have unknown roles in human disease. To identify additional IFT-B components defective in ciliopathies, we independently performed different mutation analyses: candidate-based sequencing of all IFT-B-encoding genes in 1,467 individuals with a nephronophthisis-related ciliopathy or whole-exome resequencing in 63 individuals with ATD. We thereby detected biallelic mutations in the IFT-B-encoding gene IFT172 in 12 families. All affected individuals displayed abnormalities of the thorax and/or long bones, as well as renal, hepatic, or retinal involvement, consistent with the diagnosis of ATD or MZSDS. Additionally, cerebellar aplasia or hypoplasia characteristic of Joubert syndrome was present in 2 out of 12 families. Fibroblasts from affected individuals showed disturbed ciliary composition, suggesting alteration of ciliary transport and signaling. Knockdown of ift172 in zebrafish recapitulated the human phenotype and demonstrated a genetic interaction between ift172 and ift80. In summary, we have identified defects in IFT172 as a cause of complex ATD and MZSDS. Our findings link the group of skeletal ciliopathies to an additional IFT-B component, IFT172, similar to what has been shown for IFT-A.
- Published
- 2013
- Full Text
- View/download PDF
12. Pathogenic PHIP Variants are Variably Associated With CAKUT.
- Author
-
de Fallois J, Sieckmann T, Schönauer R, Petzold F, Münch J, Pauly M, Vasileiou G, Findeisen C, Kampmeier A, Kuechler A, Reis A, Decker E, Bergmann C, Platzer K, Tasic V, Kirschner KM, Shril S, Hildebrandt F, Chung WK, and Halbritter J
- Abstract
Introduction: Congenital anomalies of the kidney and urinary tract (CAKUT) represent the most common cause of chronic kidney disease in children. Although only 20% of cases can be genetically explained, the majority remain without an identified underlying etiology. The neurodevelopmental disorder Chung-Jansen syndrome (CHUJANS) is caused by haploinsufficiency of Pleckstrin homology domain-interacting protein (PHIP) and was previously associated with genital malformations. Anecdotal coincidence of CHUJANS and CAKUT prompted us to investigate whether urorenal malformations are part of the phenotypic spectrum of CHUJANS., Methods: Analysis of existing CHUJANS and CAKUT cohorts, consulting matchmaking platforms, and systematic literature review to look for additional patients with both CHUJANS and CAKUT. Prenatal expression studies in murine and human renal tissues to investigate the role for PHIP in kidney development., Results: We identified 4 novel and 8 published cases, indicating variable expressivity with a urorenogenital trait frequency of 5% to 35%. The prenatal expression studies supported a role for PHIP in normal kidney and urinary tract development., Conclusion: Pathogenic PHIP gene variants should be considered as causative in patients with syndromal CAKUT. Conversely, patients with CHUJANS should be clinically evaluated for urorenogenital manifestations. Because neurodevelopmental disorders are often associated with kidney phenotypes, an interdisciplinary re-evaluation offers promise in identifying incompletely penetrant kidney associations and uncovering novel molecular mechanisms of disturbed nephrogenesis., (© 2024 International Society of Nephrology. Published by Elsevier Inc.)
- Published
- 2024
- Full Text
- View/download PDF
13. Implication of transcription factor FOXD2 dysfunction in syndromic congenital anomalies of the kidney and urinary tract (CAKUT).
- Author
-
Riedhammer KM, Nguyen TT, Koşukcu C, Calzada-Wack J, Li Y, Assia Batzir N, Saygılı S, Wimmers V, Kim GJ, Chrysanthou M, Bakey Z, Sofrin-Drucker E, Kraiger M, Sanz-Moreno A, Amarie OV, Rathkolb B, Klein-Rodewald T, Garrett L, Hölter SM, Seisenberger C, Haug S, Schlosser P, Marschall S, Wurst W, Fuchs H, Gailus-Durner V, Wuttke M, Hrabe de Angelis M, Ćomić J, Akgün Doğan Ö, Özlük Y, Taşdemir M, Ağbaş A, Canpolat N, Orenstein N, Çalışkan S, Weber RG, Bergmann C, Jeanpierre C, Saunier S, Lim TY, Hildebrandt F, Alhaddad B, Basel-Salmon L, Borovitz Y, Wu K, Antony D, Matschkal J, Schaaf CW, Renders L, Schmaderer C, Rogg M, Schell C, Meitinger T, Heemann U, Köttgen A, Arnold SJ, Ozaltin F, Schmidts M, and Hoefele J
- Subjects
- Adult, Animals, Humans, Mice, Genome-Wide Association Study, Mice, Knockout, Transcription Factors genetics, Embryonic Structures, Kidney abnormalities, Kidney embryology, Kidney Diseases genetics, Nephrons embryology, Urinary Tract, Urogenital Abnormalities genetics, Vesico-Ureteral Reflux genetics, Forkhead Transcription Factors deficiency, Forkhead Transcription Factors metabolism
- Abstract
Congenital anomalies of the kidney and urinary tract (CAKUT) are the predominant cause for chronic kidney disease below age 30 years. Many monogenic forms have been discovered due to comprehensive genetic testing like exome sequencing. However, disease-causing variants in known disease-associated genes only explain a proportion of cases. Here, we aim to unravel underlying molecular mechanisms of syndromic CAKUT in three unrelated multiplex families with presumed autosomal recessive inheritance. Exome sequencing in the index individuals revealed three different rare homozygous variants in FOXD2, encoding a transcription factor not previously implicated in CAKUT in humans: a frameshift in the Arabic and a missense variant each in the Turkish and the Israeli family with segregation patterns consistent with autosomal recessive inheritance. CRISPR/Cas9-derived Foxd2 knockout mice presented with a bilateral dilated kidney pelvis accompanied by atrophy of the kidney papilla and mandibular, ophthalmologic, and behavioral anomalies, recapitulating the human phenotype. In a complementary approach to study pathomechanisms of FOXD2-dysfunction-mediated developmental kidney defects, we generated CRISPR/Cas9-mediated knockout of Foxd2 in ureteric bud-induced mouse metanephric mesenchyme cells. Transcriptomic analyses revealed enrichment of numerous differentially expressed genes important for kidney/urogenital development, including Pax2 and Wnt4 as well as gene expression changes indicating a shift toward a stromal cell identity. Histology of Foxd2 knockout mouse kidneys confirmed increased fibrosis. Further, genome-wide association studies suggest that FOXD2 could play a role for maintenance of podocyte integrity during adulthood. Thus, our studies help in genetic diagnostics of monogenic CAKUT and in understanding of monogenic and multifactorial kidney diseases., (Copyright © 2023 International Society of Nephrology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
14. OXGR1 is a candidate disease gene for human calcium oxalate nephrolithiasis.
- Author
-
Majmundar AJ, Widmeier E, Heneghan JF, Daga A, Wu CW, Buerger F, Hugo H, Ullah I, Amar A, Ottlewski I, Braun DA, Jobst-Schwan T, Lawson JA, Zahoor MY, Rodig NM, Tasic V, Nelson CP, Khaliq S, Schönauer R, Halbritter J, Sayer JA, Fathy HM, Baum MA, Shril S, Mane S, Alper SL, and Hildebrandt F
- Subjects
- Humans, Calcium Oxalate, Mutation, Missense genetics, Sulfate Transporters genetics, Nephrolithiasis genetics, Receptors, Purinergic P2 genetics, Receptors, Purinergic P2 metabolism
- Abstract
Purpose: Nephrolithiasis (NL) affects 1 in 11 individuals worldwide, leading to significant patient morbidity. NL is associated with nephrocalcinosis (NC), a risk factor for chronic kidney disease. Causative genetic variants are detected in 11% to 28% of NL and/or NC, suggesting that additional NL/NC-associated genetic loci await discovery. Therefore, we employed genomic approaches to discover novel genetic forms of NL/NC., Methods: Exome sequencing and directed sequencing of the OXGR1 locus were performed in a worldwide NL/NC cohort. Putatively deleterious, rare OXGR1 variants were functionally characterized., Results: Exome sequencing revealed a heterozygous OXGR1 missense variant (c.371T>G, p.L124R) cosegregating with calcium oxalate NL and/or NC disease in an autosomal dominant inheritance pattern within a multigenerational family with 5 affected individuals. OXGR1 encodes 2-oxoglutarate (α-ketoglutarate [AKG]) receptor 1 in the distal nephron. In response to its ligand AKG, OXGR1 stimulates the chloride-bicarbonate exchanger, pendrin, which also regulates transepithelial calcium transport in cortical connecting tubules. Strong amino acid conservation in orthologs and paralogs, severe in silico prediction scores, and extreme rarity in exome population databases suggested that the variant was deleterious. Interrogation of the OXGR1 locus in 1107 additional NL/NC families identified 5 additional deleterious dominant variants in 5 families with calcium oxalate NL/NC. Rare, potentially deleterious OXGR1 variants were enriched in patients with NL/NC compared with Exome Aggregation Consortium controls (χ
2 = 7.117, P = .0076). Wild-type OXGR1-expressing Xenopus oocytes exhibited AKG-responsive Ca2+ uptake. Of 5 NL/NC-associated missense variants, 5 revealed impaired AKG-dependent Ca2+ uptake, demonstrating loss of function., Conclusion: Rare, dominant loss-of-function OXGR1 variants are associated with recurrent calcium oxalate NL/NC disease., Competing Interests: Conflict of Interest F.H. is a cofounder of Goldfinch Bio, Inc. S.L.A. is a consultant to and received funding from Quest Diagnostics, Inc. All other authors declare no conflicts of interest., (Copyright © 2022 American College of Medical Genetics and Genomics. Published by Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
15. Inhibition of endoplasmic reticulum stress signaling rescues cytotoxicity of human apolipoprotein-L1 risk variants in Drosophila.
- Author
-
Gerstner L, Chen M, Kampf LL, Milosavljevic J, Lang K, Schneider R, Hildebrandt F, Helmstädter M, Walz G, and Hermle T
- Subjects
- Animals, Apolipoprotein L1 genetics, Drosophila genetics, Endoplasmic Reticulum Stress genetics, Humans, Kidney Diseases pathology, Podocytes pathology
- Abstract
Risk variants of the apolipoprotein-L1 (APOL1) gene are associated with severe kidney disease, putting homozygous carriers at risk. Since APOL1 lacks orthologs in all major model organisms, a wide range of mechanisms frequently in conflict have been described for APOL1-associated nephropathies. The genetic toolkit in Drosophila allows unique in vivo insights into disrupted cellular homeostasis. To perform a mechanistic analysis, we expressed human APOL1 control and gain-of-function kidney risk variants in the podocyte-like garland cells of Drosophila nephrocytes and a wing precursor tissue. Expression of APOL1 risk variants was found to elevate endocytic function of garland cell nephrocytes that simultaneously showed early signs of cell death. Wild-type APOL1 had a significantly milder effect, while a control transgene with deletion of the short BH3 domain showed no overt phenotype. Nephrocyte endo-lysosomal function and slit diaphragm architecture remained unaffected by APOL1 risk variants, but endoplasmic reticulum (ER) swelling, chaperone induction, and expression of the reporter Xbp1-EGFP suggested an ER stress response. Pharmacological inhibition of ER stress diminished APOL1-mediated cell death and direct ER stress induction enhanced nephrocyte endocytic function similar to expression of APOL1 risk variants. We confirmed APOL1-dependent ER stress in the Drosophila wing precursor where silencing the IRE1-dependent branch of ER stress signaling by inhibition with Xbp1-RNAi abrogated cell death, representing the first rescue of APOL1-associated cytotoxicity in vivo. Thus, we uncovered ER stress as an essential consequence of APOL1 risk variant expression in vivo in Drosophila, suggesting a central role of this pathway in the pathogenesis of APOL1-associated nephropathies., (Copyright © 2022 International Society of Nephrology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
16. Reverse phenotyping facilitates disease allele calling in exome sequencing of patients with CAKUT.
- Author
-
Seltzsam S, Wang C, Zheng B, Mann N, Connaughton DM, Wu CW, 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, and Hildebrandt F
- Subjects
- Alleles, Exome genetics, Humans, Kidney abnormalities, Vesico-Ureteral Reflux, Urinary Tract, Urogenital Abnormalities genetics
- 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., Competing Interests: Conflict of Interest F.H. is a cofounder and Scientific Advisory Committee member of and holds stocks in Goldfinch-Bio. All other authors declare no conflicts of interest., (Copyright © 2021 American College of Medical Genetics and Genomics. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
17. A recurrent, homozygous EMC10 frameshift variant is associated with a syndrome of developmental delay with variable seizures and dysmorphic features.
- Author
-
Shao DD, Straussberg R, Ahmed H, Khan A, Tian S, Hill RS, Smith RS, Majmundar AJ, Ameziane N, Neil JE, Yang E, Al Tenaiji A, Jamuar SS, Schlaeger TM, Al-Saffar M, Hovel I, Al-Shamsi A, Basel-Salmon L, Amir AZ, Rento LM, Lim JY, Ganesan I, Shril S, Evrony G, Barkovich AJ, Bauer P, Hildebrandt F, Dong M, Borck G, Beetz C, Al-Gazali L, Eyaid W, and Walsh CA
- Subjects
- Child, Frameshift Mutation, Homozygote, Humans, Membrane Proteins genetics, Pedigree, Phenotype, Seizures genetics, Developmental Disabilities genetics, Intellectual Disability genetics
- Abstract
Purpose: The endoplasmic reticulum membrane complex (EMC) is a highly conserved, multifunctional 10-protein complex related to membrane protein biology. In seven families, we identified 13 individuals with highly overlapping phenotypes who harbor a single identical homozygous frameshift variant in EMC10., Methods: Using exome, genome, and Sanger sequencing, a recurrent frameshift EMC10 variant was identified in affected individuals in an international cohort of consanguineous families. Multiple families were independently identified and connected via Matchmaker Exchange and internal databases. We assessed the effect of the frameshift variant on EMC10 RNA and protein expression and evaluated EMC10 expression in normal human brain tissue using immunohistochemistry., Results: A homozygous variant EMC10 c.287delG (Refseq NM_206538.3, p.Gly96Alafs*9) segregated with affected individuals in each family, who exhibited a phenotypic spectrum of intellectual disability (ID) and global developmental delay (GDD), variable seizures and variable dysmorphic features (elongated face, curly hair, cubitus valgus, and arachnodactyly). The variant arose on two founder haplotypes and results in significantly reduced EMC10 RNA expression and an unstable truncated EMC10 protein., Conclusion: We propose that a homozygous loss-of-function variant in EMC10 causes a novel syndromic neurodevelopmental phenotype. Remarkably, the recurrent variant is likely the result of a hypermutable site and arose on distinct founder haplotypes.
- Published
- 2021
- Full Text
- View/download PDF
18. Generation of Monogenic Candidate Genes for Human Nephrotic Syndrome Using 3 Independent Approaches.
- Author
-
Klämbt V, Mao Y, Schneider R, Buerger F, Shamseldin H, Onuchic-Whitford AC, Deutsch K, Kitzler TM, Nakayama M, Majmundar AJ, Mann N, Hugo H, Widmeier E, Tan W, Rehm HL, Mane S, Lifton RP, Alkuraya FS, Shril S, and Hildebrandt F
- Abstract
Introduction: Steroid-resistant nephrotic syndrome (SRNS) is the second most common cause of chronic kidney disease during childhood. Identification of 63 monogenic human genes has delineated 12 distinct pathogenic pathways., Methods: Here, we generated 2 independent sets of nephrotic syndrome (NS) candidate genes to augment the discovery of additional monogenic causes based on whole-exome sequencing (WES) data from 1382 families with NS., Results: We first identified 63 known monogenic causes of NS in mice from public databases and scientific publications, and 12 of these genes overlapped with the 63 known human monogenic SRNS genes. Second, we used a set of 64 genes that are regulated by the transcription factor Wilms tumor 1 (WT1), which causes SRNS if mutated. Thirteen of these WT1-regulated genes overlapped with human or murine NS genes. Finally, we overlapped these lists of murine and WT1 candidate genes with our list of 120 candidate genes generated from WES in 1382 NS families, to identify novel candidate genes for monogenic human SRNS. Using this approach, we identified 7 overlapping genes, of which 3 genes were shared by all datasets, including SYNPO . We show that loss-of-function of SYNPO leads to decreased CDC42 activity and reduced podocyte migration rate, both of which are rescued by overexpression of wild-type complementary DNA (cDNA), but not by cDNA representing the patient mutation., Conclusion: Thus, we identified 3 novel candidate genes for human SRNS using 3 independent, nonoverlapping hypotheses, and generated functional evidence for SYNPO as a novel potential monogenic cause of NS., (© 2020 International Society of Nephrology. Published by Elsevier Inc.)
- Published
- 2020
- Full Text
- View/download PDF
19. Recessive Mutations in SYNPO2 as a Candidate of Monogenic Nephrotic Syndrome.
- Author
-
Mao Y, Schneider R, van der Ven PFM, Assent M, Lohanadan K, Klämbt V, Buerger F, Kitzler TM, Deutsch K, Nakayama M, Majmundar AJ, Mann N, Hermle T, Onuchic-Whitford AC, Zhou W, Margam NN, Duncan R, Marquez J, Khokha M, Fathy HM, Kari JA, El Desoky S, Eid LA, Awad HS, Al-Saffar M, Mane S, Lifton RP, Fürst DO, Shril S, and Hildebrandt F
- Abstract
Introduction: Most of the approximately 60 genes that if mutated cause steroid-resistant nephrotic syndrome (SRNS) are highly expressed in the glomerular podocyte, rendering SRNS a "podocytopathy.", Methods: We performed whole-exome sequencing (WES) in 1200 nephrotic syndrome (NS) patients., Results: We discovered homozygous truncating and homozygous missense mutation in SYNPO2 (synaptopodin-2) (p.Lys1124∗ and p.Ala1134Thr) in 2 patients with childhood-onset NS. We found SYNPO2 expression in both podocytes and mesangial cells; however, notably, immunofluorescence staining of adult human and rat kidney cryosections indicated that SYNPO2 is localized mainly in mesangial cells. Subcellular localization studies reveal that in these cells SYNPO2 partially co-localizes with α-actinin and filamin A-containing F-actin filaments. Upon transfection in mesangial cells or podocytes, EGFP-SYNPO2 co-localized with α-actinin-4, which gene is mutated in autosomal dominant SRNS in humans. SYNPO2 overexpression increases mesangial cell migration rate (MMR), whereas shRNA knockdown reduces MMR. Decreased MMR was rescued by transfection of wild-type mouse Synpo2 cDNA but only partially by cDNA representing mutations from the NS patients. The increased mesangial cell migration rate (MMR) by SYNPO2 overexpression was inhibited by ARP complex inhibitor CK666. SYNPO2 shRNA knockdown in podocytes decreased active Rac1, which was rescued by transfection of wild-type SYNPO2 cDNA but not by cDNA representing any of the 2 mutant variants., Conclusion: We show that SYNPO2 variants may lead to Rac1-ARP3 dysregulation, and may play a role in the pathogenesis of nephrotic syndrome., (© 2020 International Society of Nephrology. Published by Elsevier Inc.)
- Published
- 2020
- Full Text
- View/download PDF
20. Phenotype expansion of heterozygous FOXC1 pathogenic variants toward involvement of congenital anomalies of the kidneys and urinary tract (CAKUT).
- Author
-
Wu CW, Mann N, Nakayama M, Connaughton DM, Dai R, Kolvenbach CM, Kause F, Ottlewski I, Wang C, Klämbt V, Seltzsam S, Lai EW, Selvin A, Senguttuva P, Bodamer O, Stein DR, El Desoky S, Kari JA, Tasic V, Bauer SB, Shril S, and Hildebrandt F
- Subjects
- Child, Forkhead Transcription Factors genetics, Heterozygote, Humans, Kidney, Phenotype, Eye Abnormalities, Urinary Tract
- Abstract
Purpose: Congenital anomalies of the kidney and urinary tract (CAKUT) are the most common cause of chronic kidney disease in childhood and adolescence. We aim to identify novel monogenic causes of CAKUT., Methods: Exome sequencing was performed in 550 CAKUT-affected families., Results: We discovered seven FOXC1 heterozygous likely pathogenic variants within eight CAKUT families. These variants are either never reported, or present in <5 alleles in the gnomAD database with ~141,456 controls. FOXC1 is a causal gene for Axenfeld-Rieger syndrome type 3 and anterior segment dysgenesis 3. Pathogenic variants in FOXC1 have not been detected in patients with CAKUT yet. Interestingly, mouse models for Foxc1 show severe CAKUT phenotypes with incomplete penetrance and variable expressivity. The FOXC1 variants are enriched in the CAKUT cohort compared with the control. Genotype-phenotype correlations showed that Axenfeld-Rieger syndrome or anterior segment dysgenesis can be caused by both truncating and missense pathogenic variants, and the missense variants are located at the forkhead domain. In contrast, for CAKUT, there is no truncating pathogenic variant, and all variants except one are located outside the forkhead domain., Conclusion: We thereby expanded the phenotype of FOXC1 pathogenic variants toward involvement of CAKUT, which can potentially be explained by allelism.
- Published
- 2020
- Full Text
- View/download PDF
21. Novel nephronophthisis-associated variants reveal functional importance of MAPKBP1 dimerization for centriolar recruitment.
- Author
-
Schönauer R, Jin W, Ertel A, Nemitz-Kliemchen M, Panitz N, Hantmann E, Seidel A, Braun DA, Shril S, Hansen M, Shahzad K, Sandford R, Saunier S, Benmerah A, Bergmann C, Hildebrandt F, and Halbritter J
- Subjects
- Adult, Cell Cycle Proteins genetics, Cell Cycle Proteins metabolism, Cilia metabolism, Dimerization, Fibrosis, Humans, Intracellular Signaling Peptides and Proteins, Nerve Tissue Proteins metabolism, Centrosome metabolism, Polycystic Kidney Diseases metabolism
- Abstract
Biallelic mutations in MAPKBP1 were recently associated with late-onset cilia-independent nephronophthisis. MAPKBP1 was found at mitotic spindle poles but could not be detected at primary cilia or centrosomes. Here, by identification and characterization of novel MAPKBP1 variants, we aimed at further investigating its role in health and disease. Genetic analysis was done by exome sequencing, homozygosity mapping, and a targeted kidney gene panel while coimmunoprecipitation was used to explore wild-type and mutant protein-protein interactions. Expression of MAPKBP1 in non-ciliated HeLa and ciliated inner medullary collecting duct cells enabled co-localization studies by fluorescence microscopy. By next generation sequencing, we identified two novel homozygous MAPKBP1 splice-site variants in patients with nephronophthisis-related chronic kidney disease. Splice-site analyses revealed truncation of C-terminal coiled-coil domains and patient-derived deletion constructs lost their ability to homodimerize and heterodimerize with paralogous WDR62. While wild-type MAPKBP1 exhibited centrosomal, basal body, and microtubule association, mutant proteins lost the latter and showed reduced recruitment to cell cycle dependent centriolar structures. Wild-type and mutant proteins had no reciprocal influence upon co-expression excluding dominant negative effects. Thus, MAPKBP1 appears to be a novel microtubule-binding protein with cell cycle dependent centriolar localization. Truncation of its coiled-coil domain is enough to abrogate its dimerization and results in severely disturbed intracellular localizations. Delineating the impact of impaired dimerization on cell cycle regulation and intracellular kidney signaling may provide new insights into common mechanisms of kidney degeneration. Thus, due to milder clinical presentation, MAPKBP1-associated nephronophthisis should be considered in adult patients with otherwise unexplained chronic kidney disease., (Copyright © 2020 International Society of Nephrology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
22. Whole exome sequencing identified ATP6V1C2 as a novel candidate gene for recessive distal renal tubular acidosis.
- Author
-
Jobst-Schwan T, Klämbt V, Tarsio M, Heneghan JF, Majmundar AJ, Shril S, Buerger F, Ottlewski I, Shmukler BE, Topaloglu R, Hashmi S, Hafeez F, Emma F, Greco M, Laube GF, Fathy HM, Pohl M, Gellermann J, Milosevic D, Baum MA, Mane S, Lifton RP, Kane PM, Alper SL, and Hildebrandt F
- Subjects
- Anion Exchange Protein 1, Erythrocyte, Child, Chloride-Bicarbonate Antiporters, DNA Mutational Analysis, Forkhead Transcription Factors, Humans, Mutation, Exome Sequencing, Acidosis, Renal Tubular genetics, Vacuolar Proton-Translocating ATPases genetics
- Abstract
Distal renal tubular acidosis is a rare renal tubular disorder characterized by hyperchloremic metabolic acidosis and impaired urinary acidification. Mutations in three genes (ATP6V0A4, ATP6V1B1 and SLC4A1) constitute a monogenic causation in 58-70% of familial cases of distal renal tubular acidosis. Recently, mutations in FOXI1 have been identified as an additional cause. Therefore, we hypothesized that further monogenic causes of distal renal tubular acidosis remain to be discovered. Panel sequencing and/or whole exome sequencing was performed in a cohort of 17 families with 19 affected individuals with pediatric onset distal renal tubular acidosis. A causative mutation was detected in one of the three "classical" known distal renal tubular acidosis genes in 10 of 17 families. The seven unsolved families were then subjected to candidate whole exome sequencing analysis. Potential disease causing mutations in three genes were detected: ATP6V1C2, which encodes another kidney specific subunit of the V-type proton ATPase (1 family); WDR72 (2 families), previously implicated in V-ATPase trafficking in cells; and SLC4A2 (1 family), a paralog of the known distal renal tubular acidosis gene SLC4A1. Two of these mutations were assessed for deleteriousness through functional studies. Yeast growth assays for ATP6V1C2 revealed loss-of-function for the patient mutation, strongly supporting ATP6V1C2 as a novel distal renal tubular acidosis gene. Thus, we provided a molecular diagnosis in a known distal renal tubular acidosis gene in 10 of 17 families (59%) with this disease, identified mutations in ATP6V1C2 as a novel human candidate gene, and provided further evidence for phenotypic expansion in WDR72 mutations from amelogenesis imperfecta to distal renal tubular acidosis., (Copyright © 2019 International Society of Nephrology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
23. Mutations in KIRREL1, a slit diaphragm component, cause steroid-resistant nephrotic syndrome.
- Author
-
Solanki AK, Widmeier E, Arif E, Sharma S, Daga A, Srivastava P, Kwon SH, Hugo H, Nakayama M, Mann N, Majmundar AJ, Tan W, Gee HY, Sadowski CE, Rinat C, Becker-Cohen R, Bergmann C, Rosen S, Somers M, Shril S, Huber TB, Mane S, Hildebrandt F, and Nihalani D
- Subjects
- Adolescent, Age of Onset, Cell Line, Child, Child, Preschool, Consanguinity, DNA Mutational Analysis, Disease Progression, Female, Follow-Up Studies, Gene Frequency, Glomerular Basement Membrane pathology, Glomerular Basement Membrane ultrastructure, Glucocorticoids therapeutic use, Homozygote, Humans, Male, Membrane Proteins metabolism, Microscopy, Electron, Transmission, Mutation, Nephrotic Syndrome drug therapy, Nephrotic Syndrome pathology, Pedigree, Podocytes, Renal Insufficiency, Chronic pathology, Exome Sequencing, Drug Resistance genetics, Glucocorticoids pharmacology, Membrane Proteins genetics, Nephrotic Syndrome genetics, Renal Insufficiency, Chronic genetics
- Abstract
Steroid-resistant nephrotic syndrome is a frequent cause of chronic kidney disease almost inevitably progressing to end-stage renal disease. More than 58 monogenic causes of SRNS have been discovered and majority of known steroid-resistant nephrotic syndrome causing genes are predominantly expressed in glomerular podocytes, placing them at the center of disease pathogenesis. Herein, we describe two unrelated families with steroid-resistant nephrotic syndrome with homozygous mutations in the KIRREL1 gene. One mutation showed high frequency in the European population (minor allele frequency 0.0011) and this patient achieved complete remission following treatment, but later progressed to chronic kidney disease. We found that mutant KIRREL1 proteins failed to localize to the podocyte cell membrane, indicating defective trafficking and impaired podocytes function. Thus, the KIRREL1 gene product has an important role in modulating the integrity of the slit diaphragm and maintaining glomerular filtration function., (Copyright © 2019 International Society of Nephrology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
24. Disruption of MAGI2-RapGEF2-Rap1 signaling contributes to podocyte dysfunction in congenital nephrotic syndrome caused by mutations in MAGI2.
- Author
-
Zhu B, Cao A, Li J, Young J, Wong J, Ashraf S, Bierzynska A, Menon MC, Hou S, Sawyers C, Campbell KN, Saleem MA, He JC, Hildebrandt F, D'Agati VD, Peng W, and Kaufman L
- Subjects
- Adaptor Proteins, Signal Transducing metabolism, Animals, Cell Line, Cyclic AMP analogs & derivatives, Cyclic AMP pharmacology, Guanine Nucleotide Exchange Factors genetics, Guanylate Kinases metabolism, Humans, Mice, Mice, Knockout, Mutation, Nephrotic Syndrome pathology, Shelterin Complex, Signal Transduction drug effects, Signal Transduction genetics, Telomere-Binding Proteins agonists, rap1 GTP-Binding Proteins metabolism, Adaptor Proteins, Signal Transducing genetics, Guanine Nucleotide Exchange Factors metabolism, Guanylate Kinases genetics, Nephrotic Syndrome genetics, Nerve Tissue Proteins metabolism, Podocytes pathology, Telomere-Binding Proteins metabolism
- Abstract
The essential role of membrane associated guanylate kinase 2 (MAGI2) in podocytes is indicated by the phenotypes of severe glomerulosclerosis of both MAGI2 knockout mice and in patients with congenital nephrotic syndrome (CNS) caused by mutations in MAGI2. Here, we show that MAGI2 forms a complex with the Rap1 guanine nucleotide exchange factor, RapGEF2, and that this complex is lost when expressing MAGI2 CNS variants. Co-expression of RapGEF2 with wild-type MAGI2, but not MAGI2 CNS variants, enhanced activation of the small GTPase Rap1, a central signaling node in podocytes. In mice, podocyte-specific RapGEF2 deletion resulted in spontaneous glomerulosclerosis, with qualitative glomerular features comparable to MAGI2 knockout mice. Knockdown of RapGEF2 or MAGI2 in human podocytes caused similar reductions in levels of Rap1 activation and Rap1-mediated downstream signaling. Furthermore, human podocytes expressing MAGI2 CNS variants show severe abnormalities of cellular morphology and dramatic loss of actin cytoskeletal organization, features completely rescued by pharmacological activation of Rap1 via a non-MAGI2 dependent upstream pathway. Finally, immunostaining of kidney sections from patients with congenital nephrotic syndrome and MAGI2 mutations showed reduced podocyte Rap1-mediated signaling. Thus, MAGI2-RapGEF2-Rap1 signaling is essential for normal podocyte function. Hence, disruption of this pathway is an important cause of the renal phenotype induced by MAGI2 CNS mutations., (Copyright © 2019 International Society of Nephrology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
25. Roscovitine blocks collecting duct cyst growth in Cep164-deficient kidneys.
- Author
-
Airik R, Airik M, Schueler M, Bates CM, and Hildebrandt F
- Subjects
- Animals, Animals, Newborn, Cell Cycle drug effects, Cilia pathology, Ciliopathies genetics, Ciliopathies pathology, Cyclin-Dependent Kinases antagonists & inhibitors, Cyclin-Dependent Kinases metabolism, Disease Models, Animal, Drug Evaluation, Preclinical, Embryo, Mammalian, Epithelium drug effects, Epithelium pathology, Female, Humans, Kidney Diseases, Cystic genetics, Kidney Tubules, Collecting cytology, Kidney Tubules, Collecting growth & development, Kidney Tubules, Collecting pathology, Male, Mice, Mice, Knockout, Microtubule Proteins genetics, Organoselenium Compounds, Proof of Concept Study, Ciliopathies drug therapy, Kidney Diseases, Cystic drug therapy, Kidney Tubules, Collecting drug effects, Microtubule Proteins deficiency, Protein Kinase Inhibitors administration & dosage, Roscovitine administration & dosage
- Abstract
Nephronophthisis is an autosomal recessive kidney disease with high genetic heterogeneity. Understanding the functions of the individual genes contributing to this disease is critical for delineating the pathomechanisms of this disorder. Here, we investigated kidney function of a novel gene associated with nephronophthisis, CEP164, coding a centriolar distal appendage protein, using a Cep164 knockout mouse model. Collecting duct-specific deletion of Cep164 abolished primary cilia from the collecting duct epithelium and led to rapid postnatal cyst growth in the kidneys. Cell cycle and biochemical studies revealed that tubular hyperproliferation is the primary mechanism that drives cystogenesis in the kidneys of these mice. Administration of roscovitine, a cell cycle inhibitor, blocked cyst growth in the cortical collecting ducts and preserved kidney parenchyma in Cep164 knockout mice. Thus, our findings provide evidence that therapeutic modulation of cell cycle activity can be an effective approach to prevent cyst progression in the kidney., (Copyright © 2019 International Society of Nephrology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
26. Corticosteroid treatment exacerbates nephrotic syndrome in a zebrafish model of magi2a knockout.
- Author
-
Jobst-Schwan T, Hoogstraten CA, Kolvenbach CM, Schmidt JM, Kolb A, Eddy K, Schneider R, Ashraf S, Widmeier E, Majmundar AJ, and Hildebrandt F
- Subjects
- Animals, Animals, Genetically Modified, Cyclosporine pharmacology, Cyclosporine therapeutic use, Disease Models, Animal, Disease Progression, Drug Resistance, Gene Knockout Techniques, Glucocorticoids therapeutic use, Humans, Immunosuppressive Agents therapeutic use, Monomeric GTP-Binding Proteins metabolism, Nephrotic Syndrome genetics, Nephrotic Syndrome pathology, Podocytes drug effects, Podocytes pathology, Proteinuria genetics, Proteinuria pathology, Signal Transduction drug effects, Tacrolimus pharmacology, Tacrolimus therapeutic use, Treatment Outcome, Zebrafish, Zebrafish Proteins metabolism, Glucocorticoids pharmacology, Immunosuppressive Agents pharmacology, Membrane Proteins genetics, Nephrotic Syndrome drug therapy, Proteinuria drug therapy, Zebrafish Proteins genetics
- Abstract
Recently, recessive mutations of MAGI2 were identified as a cause of steroid-resistant nephrotic syndrome (SRNS) in humans and mice. To further delineate the pathogenesis of MAGI2 loss of function, we generated stable knockout lines for the two zebrafish orthologues magi2a and magi2b by CRISPR/Cas9. We also developed a novel assay for the direct detection of proteinuria in zebrafish independent of transgenic background. Whereas knockout of magi2b did not yield a nephrotic syndrome phenotype, magi2a
-/- larvae developed ascites, periorbital edema, and proteinuria, as indicated by increased excretion of low molecular weight protein. Electron microscopy demonstrated extensive podocyte foot process effacement. As in human SRNS, we observed genotype/phenotype correlation, with edema onset occurring earlier in zebrafish with truncating alleles (5-6 days post fertilization) versus hypomorphic alleles (19-20 days post fertilization). Paradoxically, corticosteroid treatment exacerbated the phenotype, with earlier onset of edema. In contrast, treatment with cyclosporine A or tacrolimus had no significant effect. Although RhoA signaling has been implicated as a downstream mediator of MAGI2 activity, targeting of the RhoA pathway did not modify the nephrotic syndrome phenotype. In the first CRISPR/Cas9 zebrafish knockout model of SRNS, we found that corticosteroids may have a paradoxical effect in the setting of specific genetic mutations., (Copyright © 2019 International Society of Nephrology. Published by Elsevier Inc. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
27. Monogenic causes of chronic kidney disease in adults.
- Author
-
Connaughton DM, Kennedy C, Shril S, Mann N, Murray SL, Williams PA, Conlon E, Nakayama M, van der Ven AT, Ityel H, Kause F, Kolvenbach CM, Dai R, Vivante A, Braun DA, Schneider R, Kitzler TM, Moloney B, Moran CP, Smyth JS, Kennedy A, Benson K, Stapleton C, Denton M, Magee C, O'Seaghdha CM, Plant WD, Griffin MD, Awan A, Sweeney C, Mane SM, Lifton RP, Griffin B, Leavey S, Casserly L, de Freitas DG, Holian J, Dorman A, Doyle B, Lavin PJ, Little MA, Conlon PJ, and Hildebrandt F
- Subjects
- Adolescent, Adult, Age of Onset, Aged, Aged, 80 and over, Cohort Studies, Exome genetics, Female, Humans, Ireland, Kidney, Male, Medical History Taking, Middle Aged, Mutation, Pedigree, Precision Medicine, Renal Insufficiency, Chronic diagnosis, Renal Insufficiency, Chronic therapy, Young Adult, Genetic Predisposition to Disease, Genetic Testing methods, Renal Insufficiency, Chronic genetics, Exome Sequencing
- Abstract
Approximately 500 monogenic causes of chronic kidney disease (CKD) have been identified, mainly in pediatric populations. The frequency of monogenic causes among adults with CKD has been less extensively studied. To determine the likelihood of detecting monogenic causes of CKD in adults presenting to nephrology services in Ireland, we conducted whole exome sequencing (WES) in a multi-centre cohort of 114 families including 138 affected individuals with CKD. Affected adults were recruited from 78 families with a positive family history, 16 families with extra-renal features, and 20 families with neither a family history nor extra-renal features. We detected a pathogenic mutation in a known CKD gene in 42 of 114 families (37%). A monogenic cause was identified in 36% of affected families with a positive family history of CKD, 69% of those with extra-renal features, and only 15% of those without a family history or extra-renal features. There was no difference in the rate of genetic diagnosis in individuals with childhood versus adult onset CKD. Among the 42 families in whom a monogenic cause was identified, WES confirmed the clinical diagnosis in 17 (40%), corrected the clinical diagnosis in 9 (22%), and established a diagnosis for the first time in 16 families referred with CKD of unknown etiology (38%). In this multi-centre study of adults with CKD, a molecular genetic diagnosis was established in over one-third of families. In the evolving era of precision medicine, WES may be an important tool to identify the cause of CKD in adults., (Copyright © 2019 International Society of Nephrology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
28. Whole exome sequencing frequently detects a monogenic cause in early onset nephrolithiasis and nephrocalcinosis.
- Author
-
Daga A, Majmundar AJ, Braun DA, Gee HY, Lawson JA, Shril S, Jobst-Schwan T, Vivante A, Schapiro D, Tan W, Warejko JK, Widmeier E, Nelson CP, Fathy HM, Gucev Z, Soliman NA, Hashmi S, Halbritter J, Halty M, Kari JA, El-Desoky S, Ferguson MA, Somers MJG, Traum AZ, Stein DR, Daouk GH, Rodig NM, Katz A, Hanna C, Schwaderer AL, Sayer JA, Wassner AJ, Mane S, Lifton RP, Milosevic D, Tasic V, Baum MA, and Hildebrandt F
- Subjects
- Adolescent, Age of Onset, Child, Child, Preschool, Disease Progression, Female, Genetic Association Studies, Genetic Markers, Genetic Predisposition to Disease, Heredity, Humans, Infant, Male, Nephrocalcinosis diagnostic imaging, Nephrocalcinosis epidemiology, Nephrolithiasis diagnostic imaging, Nephrolithiasis epidemiology, Pedigree, Phenotype, Predictive Value of Tests, Prognosis, Risk Factors, Tomography, X-Ray Computed, Ultrasonography, Young Adult, Mutation, Nephrocalcinosis genetics, Nephrolithiasis genetics, Exome Sequencing
- Abstract
The incidence of nephrolithiasis continues to rise. Previously, we showed that a monogenic cause could be detected in 11.4% of individuals with adult-onset nephrolithiasis or nephrocalcinosis and in 16.7-20.8% of individuals with onset before 18 years of age, using gene panel sequencing of 30 genes known to cause nephrolithiasis/nephrocalcinosis. To overcome the limitations of panel sequencing, we utilized whole exome sequencing in 51 families, who presented before age 25 years with at least one renal stone or with a renal ultrasound finding of nephrocalcinosis to identify the underlying molecular genetic cause of disease. In 15 of 51 families, we detected a monogenic causative mutation by whole exome sequencing. A mutation in seven recessive genes (AGXT, ATP6V1B1, CLDN16, CLDN19, GRHPR, SLC3A1, SLC12A1), in one dominant gene (SLC9A3R1), and in one gene (SLC34A1) with both recessive and dominant inheritance was detected. Seven of the 19 different mutations were not previously described as disease-causing. In one family, a causative mutation in one of 117 genes that may represent phenocopies of nephrolithiasis-causing genes was detected. In nine of 15 families, the genetic diagnosis may have specific implications for stone management and prevention. Several factors that correlated with the higher detection rate in our cohort were younger age at onset of nephrolithiasis/nephrocalcinosis, presence of multiple affected members in a family, and presence of consanguinity. Thus, we established whole exome sequencing as an efficient approach toward a molecular genetic diagnosis in individuals with nephrolithiasis/nephrocalcinosis who manifest before age 25 years., (Copyright © 2017 International Society of Nephrology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
29. Whole exome sequencing identifies causative mutations in the majority of consanguineous or familial cases with childhood-onset increased renal echogenicity.
- Author
-
Braun DA, Schueler M, Halbritter J, Gee HY, Porath JD, Lawson JA, Airik R, Shril S, Allen SJ, Stein D, Al Kindy A, Beck BB, Cengiz N, Moorani KN, Ozaltin F, Hashmi S, Sayer JA, Bockenhauer D, Soliman NA, Otto EA, Lifton RP, and Hildebrandt F
- Subjects
- Age of Onset, Cohort Studies, DNA Mutational Analysis, Exome, Humans, Kidney Diseases, Cystic congenital, Kidney Diseases, Cystic genetics, Renal Insufficiency, Chronic diagnostic imaging, Renal Insufficiency, Chronic genetics
- Abstract
Chronically increased echogenicity on renal ultrasound is a sensitive early finding of chronic kidney disease that can be detected before manifestation of other symptoms. Increased echogenicity, however, is not specific for a certain etiology of chronic kidney disease. Here, we performed whole exome sequencing in 79 consanguineous or familial cases of suspected nephronophthisis in order to determine the underlying molecular disease cause. In 50 cases, there was a causative mutation in a known monogenic disease gene. In 32 of these cases whole exome sequencing confirmed the diagnosis of a nephronophthisis-related ciliopathy. In 8 cases it revealed the diagnosis of a renal tubulopathy. The remaining 10 cases were identified as Alport syndrome (4), autosomal-recessive polycystic kidney disease (2), congenital anomalies of the kidney and urinary tract (3), and APECED syndrome (1). In 5 families, in whom mutations in known monogenic genes were excluded, we applied homozygosity mapping for variant filtering and identified 5 novel candidate genes (RBM48, FAM186B, PIAS1, INCENP, and RCOR1) for renal ciliopathies. Thus, whole exome sequencing allows the detection of the causative mutation in 2/3 of affected individuals, thereby presenting the etiologic diagnosis, and allows identification of novel candidate genes.
- Published
- 2016
- Full Text
- View/download PDF
30. Whole-exome resequencing reveals recessive mutations in TRAP1 in individuals with CAKUT and VACTERL association.
- Author
-
Saisawat P, Kohl S, Hilger AC, Hwang DY, Yung Gee H, Dworschak GC, Tasic V, Pennimpede T, Natarajan S, Sperry E, Matassa DS, Stajić N, Bogdanovic R, de Blaauw I, Marcelis CL, Wijers CH, Bartels E, Schmiedeke E, Schmidt D, Märzheuser S, Grasshoff-Derr S, Holland-Cunz S, Ludwig M, Nöthen MM, Draaken M, Brosens E, Heij H, Tibboel D, Herrmann BG, Solomon BD, de Klein A, van Rooij IA, Esposito F, Reutter HM, and Hildebrandt F
- Subjects
- Age Factors, Animals, Europe, Female, Gene Expression Regulation, Developmental, Genetic Predisposition to Disease, Gestational Age, Heart Defects, Congenital diagnosis, High-Throughput Nucleotide Sequencing, Homozygote, Humans, Kidney embryology, Kidney metabolism, Limb Deformities, Congenital diagnosis, Male, Mice, Multiplex Polymerase Chain Reaction, Pedigree, Predictive Value of Tests, Risk Factors, United States, Urogenital Abnormalities, Vesico-Ureteral Reflux diagnosis, Anal Canal abnormalities, DNA Mutational Analysis methods, Esophagus abnormalities, Exosomes, Genetic Testing methods, HSP90 Heat-Shock Proteins genetics, HSP90 Heat-Shock Proteins metabolism, Heart Defects, Congenital genetics, Kidney abnormalities, Limb Deformities, Congenital genetics, Mutation, Spine abnormalities, Trachea abnormalities, Vesico-Ureteral Reflux genetics
- Abstract
Congenital abnormalities of the kidney and urinary tract (CAKUT) account for approximately half of children with chronic kidney disease and they are the most frequent cause of end-stage renal disease in children in the US. However, its genetic etiology remains mostly elusive. VACTERL association is a rare disorder that involves congenital abnormalities in multiple organs including the kidney and urinary tract in up to 60% of the cases. By homozygosity mapping and whole-exome resequencing combined with high-throughput mutation analysis by array-based multiplex PCR and next-generation sequencing, we identified recessive mutations in the gene TNF receptor-associated protein 1 (TRAP1) in two families with isolated CAKUT and three families with VACTERL association. TRAP1 is a heat-shock protein 90-related mitochondrial chaperone possibly involved in antiapoptotic and endoplasmic reticulum stress signaling. Trap1 is expressed in renal epithelia of developing mouse kidney E13.5 and in the kidney of adult rats, most prominently in proximal tubules and in thick medullary ascending limbs of Henle's loop. Thus, we identified mutations in TRAP1 as highly likely causing CAKUT or VACTERL association with CAKUT.
- Published
- 2014
- Full Text
- View/download PDF
31. Mutations in 12 known dominant disease-causing genes clarify many congenital anomalies of the kidney and urinary tract.
- Author
-
Hwang DY, Dworschak GC, Kohl S, Saisawat P, Vivante A, Hilger AC, Reutter HM, Soliman NA, Bogdanovic R, Kehinde EO, Tasic V, and Hildebrandt F
- Subjects
- DNA Mutational Analysis, Female, Genetic Predisposition to Disease, Genetic Testing methods, Heredity, Heterozygote, Humans, Male, Pedigree, Phenotype, Predictive Value of Tests, Risk Factors, Urogenital Abnormalities, Genes, Dominant, Mutation, Vesico-Ureteral Reflux genetics
- Abstract
Congenital anomalies of the kidney and urinary tract (CAKUT) account for approximately half of children with chronic kidney disease. CAKUT can be caused by monogenic mutations; however, data are lacking on their frequency. Genetic diagnosis has been hampered by genetic heterogeneity and lack of genotype-phenotype correlation. To determine the percentage of cases with CAKUT that can be explained by mutations in known CAKUT genes, we analyzed the coding exons of the 17 known dominant CAKUT-causing genes in a cohort of 749 individuals from 650 families with CAKUT. The most common phenotypes in this CAKUT cohort were vesicoureteral reflux in 288 patients, renal hypodysplasia in 120 patients, and unilateral renal agenesis in 90 patients. We identified 37 different heterozygous mutations (33 novel) in 12 of the 17 known genes in 47 patients from 41 of the 650 families (6.3%). These mutations include (number of families): BMP7 (1), CDC5L (1), CHD1L (5), EYA1 (3), GATA3 (2), HNF1B (6), PAX2 (5), RET (3), ROBO2 (4), SALL1 (9), SIX2 (1), and SIX5 (1). Furthermore, several mutations previously reported to be disease-causing are most likely benign variants. Thus, in a large cohort over 6% of families with isolated CAKUT are caused by a mutation in 12 of 17 dominant CAKUT genes. Our report represents one of the most in-depth diagnostic studies of monogenic causes of isolated CAKUT in children.
- Published
- 2014
- Full Text
- View/download PDF
32. Whole-exome resequencing distinguishes cystic kidney diseases from phenocopies in renal ciliopathies.
- Author
-
Gee HY, Otto EA, Hurd TW, Ashraf S, Chaki M, Cluckey A, Vega-Warner V, Saisawat P, Diaz KA, Fang H, Kohl S, Allen SJ, Airik R, Zhou W, Ramaswami G, Janssen S, Fu C, Innis JL, Weber S, Vester U, Davis EE, Katsanis N, Fathy HM, Jeck N, Klaus G, Nayir A, Rahim KA, Al Attrach I, Al Hassoun I, Ozturk S, Drozdz D, Helmchen U, O'Toole JF, Attanasio M, Lewis RA, Nürnberg G, Nürnberg P, Washburn J, MacDonald J, Innis JW, Levy S, and Hildebrandt F
- Subjects
- Adolescent, Adult, DNA Mutational Analysis, Early Diagnosis, Exome, Genes, Recessive, Humans, Infant, Male, Mutation, Phenotype, Young Adult, Genetic Testing methods, Kidney Diseases, Cystic diagnosis, Kidney Diseases, Cystic genetics
- Abstract
Rare single-gene disorders cause chronic disease. However, half of the 6000 recessive single gene causes of disease are still unknown. Because recessive disease genes can illuminate, at least in part, disease pathomechanism, their identification offers direct opportunities for improved clinical management and potentially treatment. Rare diseases comprise the majority of chronic kidney disease (CKD) in children but are notoriously difficult to diagnose. Whole-exome resequencing facilitates identification of recessive disease genes. However, its utility is impeded by the large number of genetic variants detected. We here overcome this limitation by combining homozygosity mapping with whole-exome resequencing in 10 sib pairs with a nephronophthisis-related ciliopathy, which represents the most frequent genetic cause of CKD in the first three decades of life. In 7 of 10 sibships with a histologic or ultrasonographic diagnosis of nephronophthisis-related ciliopathy, we detect the causative gene. In six sibships, we identify mutations of known nephronophthisis-related ciliopathy genes, while in two additional sibships we found mutations in the known CKD-causing genes SLC4A1 and AGXT as phenocopies of nephronophthisis-related ciliopathy. Thus, whole-exome resequencing establishes an efficient, noninvasive approach towards early detection and causation-based diagnosis of rare kidney diseases. This approach can be extended to other rare recessive disorders, thereby providing accurate diagnosis and facilitating the study of disease mechanisms.
- Published
- 2014
- Full Text
- View/download PDF
33. Karyomegalic interstitial nephritis.
- Author
-
Zschiedrich S, Huber TB, Hildebrandt F, Mihatsch MJ, and Wiech T
- Subjects
- Adult, Epithelial Cells pathology, Humans, Male, Kidney Failure, Chronic pathology, Kidney Tubules, Proximal pathology, Nephritis, Interstitial pathology
- Published
- 2013
- Full Text
- View/download PDF
34. The Case. Unusual cause of chronic renal failure with elevated liver enzymes.
- Author
-
Artunc F, Hildebrandt F, and Amann K
- Subjects
- Humans, Kidney Failure, Chronic enzymology, Liver enzymology, Male, Middle Aged, Nephritis, Interstitial complications, Nephritis, Interstitial enzymology, Kidney Failure, Chronic etiology, Nephritis, Interstitial diagnosis
- Published
- 2012
- Full Text
- View/download PDF
35. Identification of two novel CAKUT-causing genes by massively parallel exon resequencing of candidate genes in patients with unilateral renal agenesis.
- Author
-
Saisawat P, Tasic V, Vega-Warner V, Kehinde EO, Günther B, Airik R, Innis JW, Hoskins BE, Hoefele J, Otto EA, and Hildebrandt F
- Subjects
- Female, Heterozygote, High-Throughput Nucleotide Sequencing, Humans, Kidney abnormalities, Kidney Diseases genetics, Male, Mutation, Missense, Polymerase Chain Reaction, Sequence Analysis, DNA, Congenital Abnormalities genetics, Exons, Extracellular Matrix Proteins genetics, Kidney Diseases congenital
- Abstract
Congenital abnormalities of the kidney and urinary tract (CAKUT) are the most frequent cause of chronic kidney disease in children, accounting for about half of all cases. Although many forms of CAKUT are likely caused by single-gene defects, mutations in only a few genes have been identified. In order to detect new contributing genes we pooled DNA from 20 individuals to amplify all 313 exons of 30 CAKUT candidate genes by PCR analysis and massively parallel exon resequencing. Mutation carriers were identified by Sanger sequencing. We repeated the analysis with 20 new patients to give a total of 29 with unilateral renal agenesis and 11 with other CAKUT phenotypes. Five heterozygous missense mutations were detected in 2 candidate genes (4 mutations in FRAS1 and 1 in FREM2) not previously implicated in non-syndromic CAKUT in humans. All of these mutations were absent from 96 healthy control individuals and had a PolyPhen score over 1.4, predicting possible damaging effects of the mutation on protein function. Recessive truncating mutations in FRAS1 and FREM2 were known to cause Fraser syndrome in humans and mice; however, a phenotype in heterozygous carriers has not been described. Thus, heterozygous missense mutations in FRAS1 and FREM2 cause non-syndromic CAKUT in humans.
- Published
- 2012
- Full Text
- View/download PDF
36. Genotype-phenotype correlation in 440 patients with NPHP-related ciliopathies.
- Author
-
Chaki M, Hoefele J, Allen SJ, Ramaswami G, Janssen S, Bergmann C, Heckenlively JR, Otto EA, and Hildebrandt F
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Alleles, Cytoskeletal Proteins, Family, Humans, Kidney Diseases, Cystic epidemiology, Kidney Diseases, Cystic genetics, Kidney Failure, Chronic epidemiology, Kidney Failure, Chronic genetics, Membrane Proteins genetics, Mutation, Genetic Association Studies, Kidney Diseases, Cystic congenital
- Abstract
Nephronophthisis (NPHP), an autosomal recessive cystic kidney disease, is the most frequent genetic cause for end-stage renal failure in the first three decades of life. Mutations in 13 genes (NPHP1-NPHP11, AHI1, and CC2D2A) cause NPHP with ubiquitous expression of the corresponding proteins consistent with the multiorgan involvement of NPHP-related diseases. The genotype-phenotype correlation in these ciliopathies can be explained by gene locus heterogeneity, allelism, and the impact of modifier genes. In some NPHP-related ciliopathies, the nature of the recessive mutations determines disease severity. In order to define the genotype-phenotype correlation more clearly, we evaluated a worldwide cohort of 440 patients from 365 families with NPHP-related ciliopathies, in whom both disease-causing alleles were identified. The phenotypes were ranked in the order of severity from degenerative to degenerative/dysplastic to dysplastic. A genotype of two null alleles caused a range of phenotypes, with an increasing order of severity of NPHP1, NPHP3, NPHP4, NPHP5, NPHP2, NPHP10, NPHP6, to AHI1. Only NPHP6 showed allelic influences on the phenotypes; the presence of two null mutations caused dysplastic phenotypes, whereas at least one missense allele rescued it to a milder degenerative phenotype. We also found nine novel mutations in the NPHP genes. Thus, our studies have important implications for genetic counseling and planning of renal replacement therapy.
- Published
- 2011
- Full Text
- View/download PDF
37. Exome capture and massively parallel sequencing identifies a novel HPSE2 mutation in a Saudi Arabian child with Ochoa (urofacial) syndrome.
- Author
-
Al Badr W, Al Bader S, Otto E, Hildebrandt F, Ackley T, Peng W, Xu J, Li J, Owens KM, Bloom D, and Innis JW
- Subjects
- Child, Chromosome Mapping, DNA Mutational Analysis, Diagnosis, Differential, Facies, Female, Follow-Up Studies, Glucuronidase metabolism, Homozygote, Humans, Pedigree, Saudi Arabia, Urologic Diseases diagnosis, Urologic Diseases enzymology, Exome genetics, Glucuronidase genetics, Mutation, Urologic Diseases genetics
- Abstract
We describe a child of Middle Eastern descent by first-cousin coupling with idiopathic neurogenic bladder and high-grade vesicoureteral reflux at 1 year of age, whose characteristic facial grimace led to the diagnosis of Ochoa (urofacial) syndrome at age 5 years. We used homozygosity mapping, exome capture and paired-end sequencing to identify the disease causing mutation in the proband. We reviewed the literature with respect to the urologic manifestations of Ochoa syndrome. A large region of marker homozygosity was observed at 10q24, consistent with known autosomal recessive inheritance, family consanguinity and previous genetic mapping in other families with Ochoa syndrome. A homozygous mutation was identified in the proband in HPSE2: c.1374_1378delTGTGC, a deletion of 5 nucleotides in exon 10 that is predicted to lead to a frameshift followed by replacement of 132 C-terminal amino acids with 153 novel amino acids (p.Ala458Alafsdel132ins153). This mutation is novel relative to very recently published mutations in HPSE2 in other families. Early intervention and recognition of Ochoa syndrome with control of risk factors and close surveillance will decrease complications and renal failure., (Copyright © 2011 Journal of Pediatric Urology Company. Published by Elsevier Ltd. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
38. Genetic kidney diseases.
- Author
-
Hildebrandt F
- Subjects
- Humans, Kidney abnormalities, Kidney Diseases diagnosis, Mutation, Urinary Tract abnormalities, Kidney Diseases genetics
- Abstract
Knowledge of the primary cause of a disease is essential for elucidation of its mechanisms, and for adequate classification, prognosis, and treatment. Recently, the causes of many kidney diseases have been shown to be single-gene defects-eg, steroid-resistant nephrotic syndrome, which is caused by podocin mutations in about 25% of children and nearly 15% of adults with the disease. Knowledge of a disease-causing mutation in a single-gene disorder represents one of the most robust diagnostic examples of personalised medicine because the mutation conveys an almost 100% risk of developing the disease by a defined age. Whereas single-gene diseases are rare disorders, polygenic risk alleles arise in common adult-onset diseases. In this Review, I will discuss prominent renal single-gene kidney disorders, and polygenic risk alleles of common disorders. I delineate how emerging techniques of total exome capture and large-scale sequencing will assist molecular genetic diagnosis, prognosis, and specific treatment, and lead to an improved elucidation of disease mechanisms, thus enabling development of new targeted drugs., (Copyright 2010 Elsevier Ltd. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
39. Molecular cloning and expression of phospholipase C epsilon 1 in zebrafish.
- Author
-
Zhou W and Hildebrandt F
- Subjects
- Amino Acid Sequence, Animals, Cloning, Molecular, DNA, Complementary chemistry, DNA, Complementary genetics, Embryo, Nonmammalian embryology, Embryo, Nonmammalian metabolism, Female, Gene Expression Regulation, Developmental, Gene Expression Regulation, Enzymologic, Immunohistochemistry, In Situ Hybridization, Male, Molecular Sequence Data, Phosphoinositide Phospholipase C metabolism, Reverse Transcriptase Polymerase Chain Reaction, Sequence Analysis, DNA, Sequence Homology, Amino Acid, Zebrafish embryology, Zebrafish Proteins metabolism, Gene Expression Profiling, Phosphoinositide Phospholipase C genetics, Zebrafish genetics, Zebrafish Proteins genetics
- Abstract
Phospholipase C (PLC) is a key enzyme that generates inositol 1,4,5-triphosphate (IP(3)) and diacylglycerol (DAG), two second messengers in signal transduction. Phospholipase Cepsilon (PLCE1) is a unique member of the PLC family in that it carries both RasGEF and Ras-associating (RA) domains and may serve as an activator and an effector of small GTPases. Recently, mutations in PLCE1 have been associated with early-onset nephrotic syndrome. We have identified the zebrafish ortholog of PLCE1 and characterized its expression pattern in zebrafish embryos by in situ hybridization. Zebrafish plce1 gene encodes a protein of 2248 amino acids that shares 56% identity with mammalian PLCepsilon. During zebrafish embryonic development, plce1 is expressed in a dynamic pattern in a variety of organs, such as brain, muscle, liver, and pronephric glomeruli. Our results suggest that zebrafish may be used as a model organism to address the function of PLCepsilon during the development of these organs.
- Published
- 2009
- Full Text
- View/download PDF
40. Specific podocin mutations determine age of onset of nephrotic syndrome all the way into adult life.
- Author
-
Hildebrandt F and Heeringa SF
- Subjects
- Adult, Age of Onset, Drug Resistance, Humans, Nephrotic Syndrome diagnosis, Prognosis, Intracellular Signaling Peptides and Proteins genetics, Membrane Proteins genetics, Mutation physiology, Nephrotic Syndrome epidemiology, Nephrotic Syndrome genetics
- Abstract
In steroid-resistant nephrotic syndrome (SRNS) Machuca et al. report that mutations of the recessive podocin gene cause adult-onset SRNS if the R229Q genetic variant occurs in a compound heterozygous state with another podocin mutation. Learning to tell apart the specific allele combinations of podocin mutations will be important for prognosis, genetic counseling in living related kidney donation, accurate etiologic classification within treatment studies, and the understanding of podocin function.
- Published
- 2009
- Full Text
- View/download PDF
41. Jouberin localizes to collecting ducts and interacts with nephrocystin-1.
- Author
-
Eley L, Gabrielides C, Adams M, Johnson CA, Hildebrandt F, and Sayer JA
- Subjects
- Adaptor Proteins, Vesicular Transport, Animals, Aquaporin 2 metabolism, Cell Line, Centrosome chemistry, Cilia chemistry, Cytoskeletal Proteins, Dogs, Humans, Immunohistochemistry, Intercellular Junctions chemistry, Kidney Tubules, Collecting metabolism, Mice, Protein Binding, Tissue Distribution, Adaptor Proteins, Signal Transducing metabolism, Kidney Tubules, Collecting chemistry, Membrane Proteins metabolism, Proto-Oncogene Proteins metabolism
- Abstract
Joubert syndrome and related disorders are autosomal recessive multisystem diseases characterized by cerebellar vermis aplasia/hypoplasia, retinal degeneration and cystic kidney disease. There are five known genes; mutations of which give rise to a spectrum of renal cystic diseases the most common of which is nephronophthisis, a disorder characterized by early loss of urinary concentrating ability, renal fibrosis, corticomedullary cyst formation and renal failure. Many of the proteins encoded by these genes interact with one another and are located at adherens junctions or the primary cilia and or basal bodies. Here we characterize Jouberin, a multi-domain protein encoded by the AHI1 gene. Immunohistochemistry with a novel antibody showed that endogenous Jouberin is expressed in brain, kidney and HEK293 cells. In the kidney, Jouberin co-localized with aquaporin-2 in the collecting ducts. We show that Jouberin interacts with nephrocystin-1 as determined by yeast-2-hybrid system and this was confirmed by exogenous and endogenous co-immunoprecipitation in HEK293 cells. Jouberin is expressed at cell-cell junctions, primary cilia and basal body of mIMCD3 cells while a Jouberin-GFP construct localized to centrosomes in subconfluent and dividing MDCK cells. Our results suggest that Jouberin is a protein whose expression pattern supports both the adherens junction and the ciliary hypotheses for abnormalities leading to nephronophthisis.
- Published
- 2008
- Full Text
- View/download PDF
42. Nephrocystin-1 interacts directly with Ack1 and is expressed in human collecting duct.
- Author
-
Eley L, Moochhala SH, Simms R, Hildebrandt F, and Sayer JA
- Subjects
- Adaptor Proteins, Signal Transducing, Cell Line, Cytoskeletal Proteins, Humans, Immunoprecipitation, Intercellular Junctions metabolism, Kidney Tubules, Collecting cytology, Membrane Proteins, Proteins genetics, Two-Hybrid System Techniques, src Homology Domains, Kidney Tubules, Collecting metabolism, Protein-Tyrosine Kinases metabolism, Proteins metabolism
- Abstract
Nephronophthisis is characterised by renal fibrosis, tubular basement membrane disruption and corticomedullary cyst formation leading to end stage renal failure. Mutations in NPHP1 account for the underlying genetic defect in 25% of patients with nephronophthisis. Loss of urine concentration ability may be an early feature of nephronophthisis. Using yeast-2-library screening with the SH3 domain of nephrocystin-1 as bait, we identify Ack1 as a novel interaction partner. This interaction is confirmed using exogenous over-expression followed by co-immunoprecipitation. Ack1 is an activated Cdc42-associated kinase, and like nephrocystin-1, is a known interactor of p130Cas. Nephrocystin-1 partially colocalises with Ack1 at cell-cell contacts in IMCD3 cells. In human kidney, nephrocystin-1 expression is limited to cell-cell junctions in renal collecting duct cells. These data define Ack1 as a novel interaction partner of nephrocystin-1 and implicate cell-cell junctions and the renal collecting duct in the pathology of nephronophthisis.
- Published
- 2008
- Full Text
- View/download PDF
43. Prevalence of WT1 mutations in a large cohort of patients with steroid-resistant and steroid-sensitive nephrotic syndrome.
- Author
-
Ruf RG, Schultheiss M, Lichtenberger A, Karle SM, Zalewski I, Mucha B, Everding AS, Neuhaus T, Patzer L, Plank C, Haas JP, Ozaltin F, Imm A, Fuchshuber A, Bakkaloglu A, and Hildebrandt F
- Subjects
- Child, Child, Preschool, Cohort Studies, Drug Resistance, Exons, Female, Genotype, Gonadoblastoma epidemiology, Gonadoblastoma genetics, Humans, Incidence, Infant, Male, Mutation, Missense, Neoplasms, Gonadal Tissue epidemiology, Neoplasms, Gonadal Tissue genetics, Nephrotic Syndrome drug therapy, Phenotype, Prevalence, Steroids therapeutic use, Urogenital Abnormalities drug therapy, Urogenital Abnormalities epidemiology, Urogenital Abnormalities genetics, Nephrotic Syndrome epidemiology, Nephrotic Syndrome genetics, WT1 Proteins genetics
- Abstract
Background: Nephrotic syndrome (NS) represents the association of proteinuria, hypoalbuminemia, edema, and hyperlipidemia. Steroid-resistant nephrotic syndrome (SRNS) is defined by primary resistance to standard steroid therapy. It remains one of the most intractable causes for end-stage renal disease (ESRD) in the first two decades of life. Sporadic mutations in the Wilms' tumor suppressor gene WT1 have been found to be present in patients with SRNS in association with Wilms' tumor (WT) and urinary or genital malformations, as well as in patients with isolated SRNS., Methods: To further evaluate the incidence of WT1 mutations in patients with NS we performed mutational analysis in 115 sporadic cases of SRNS and in 110 sporadic cases of steroid-sensitive nephrotic syndrome (SSNS) as a control group. Sixty out of 115 (52%) patients with sporadic SRNS were male, 55/115 (48%) were female. Sex genotype was verified by haplotype analysis. Mutational analysis was performed by direct sequencing and by denaturing high-performance liquid chromatography (DHPLC)., Results: Mutations in WT1 were found in 3/60 (5%) male (sex genotype) cases and 5/55 (9%) female (sex genotype) cases of sporadic SRNS, and 0/110 (0%) sporadic cases of SSNS. One out of five female patients with mutations in WT1 developed a WT, 2/3 male patients presented with the association of urinary and genital malformations, 1/3 male patients presented with sexual reversal (female phenotype) and bilateral gonadoblastoma, and 4/5 female patients presented with isolated SRNS., Conclusion: According to the data acquired in this study, patients presenting with a female phenotype and SRNS and male patients presenting with genital abnormalities should especially be screened to take advantage of the important genetic information on potential Wilms' tumor risk and differential therapy. This will also help to provide more data on the phenotype/genotype correlation in this patient population.
- Published
- 2004
- Full Text
- View/download PDF
44. Telomeric refinement of the MCKD1 locus on chromosome 1q21.
- Author
-
Wolf MT, van Vlem B, Hennies HC, Zalewski I, Karle SM, Puetz M, Panther F, Otto E, Fuchshuber A, Lameire N, Loeys B, and Hildebrandt F
- Subjects
- Adult, Female, Haplotypes, Humans, Male, Middle Aged, Pedigree, Chromosomes, Human, Pair 1, Genetic Linkage, Polycystic Kidney, Autosomal Dominant genetics, Telomere genetics
- Abstract
Background: Autosomal-dominant medullary cystic kidney disease type 1 (MCKD1) is a tubulointerstitial nephropathy that causes renal salt wasting and end-stage renal failure in the sixth decade of life. The chromosomal locus for MCKD1 was localized to chromosome 1q21 in a Cyprotic kindred. In this report we describe further refinement of the critical genetic region by a recombination in a Belgian kindred., Methods: Clinical data and blood samples of 33 individuals from a large Belgian kindred were collected and high-resolution haplotype analysis was performed., Results: In the Belgian kindred linkage to the MCKD1 locus on chromosome 1q21 was found with a logarithm of odds (LOD) score significant for linkage. A recombination in individual III:7 for marker D1S2624 refines the critical genetic region to 2.1 Mb. In this kindred a wide variety of clinical symptoms and age of onset of renal failure was detected., Conclusion: We confirm the MCKD1 locus on chromosome 1q21 and show further refinement of the MCKD1 locus to 2.1 Mb. This allowed us to exclude another 17 genes as positional candidate genes.
- Published
- 2004
- Full Text
- View/download PDF
45. Mutations of the Uromodulin gene in MCKD type 2 patients cluster in exon 4, which encodes three EGF-like domains.
- Author
-
Wolf MT, Mucha BE, Attanasio M, Zalewski I, Karle SM, Neumann HP, Rahman N, Bader B, Baldamus CA, Otto E, Witzgall R, Fuchshuber A, and Hildebrandt F
- Subjects
- Adolescent, Amino Acid Sequence, Base Sequence, Child, Child, Preschool, Epidermal Growth Factor chemistry, Exons genetics, Female, Genetic Linkage, Haplotypes, Humans, Male, Molecular Sequence Data, Mucoproteins chemistry, Multigene Family, Pedigree, Phenotype, Protein Structure, Tertiary, Uromodulin, Epidermal Growth Factor genetics, Mucoproteins genetics, Polycystic Kidney, Autosomal Dominant genetics
- Abstract
Background: Autosomal-dominant medullary cystic kidney disease type 2 (MCKD2) is a tubulointerstitial nephropathy that causes renal salt wasting, hyperuricemia, gout, and end-stage renal failure in the fifth decade of life. The chromosomal locus for MCKD2 was localized on chromosome 16p12. Within this chromosomal region, Uromodulin (UMOD) was located as a candidate gene. UMOD encodes the Tamm-Horsfall protein. By sequence analysis, one group formerly excluded UMOD as the disease-causing gene. In contrast, recently, another group described mutations in the UMOD gene as responsible for MCKD2 and familial juvenile hyperuricemic nephropathy (FJHN)., Methods: Haplotype analysis for linkage to MCKD2 was performed in 25 MCKD families. In the kindreds showing linkage to the MCKD2 locus on chromosome 16p12, mutational analysis of the UMOD gene was performed by exon polymerase chain reaction (PCR) and direct sequencing., Results: In 19 families, haplotype analysis was compatible with linkage to the MCKD2 locus. All these kindreds were examined for mutations in the UMOD gene. In three different families, three novel heterozygous mutations in the UMOD gene were found and segregated with the phenotype in affected individuals. Mutations were found only in exon 4., Conclusion: We confirm the UMOD gene as the disease-causing gene for MCKD2. All three novel mutations were found in the fourth exon of UMOD, in which all mutations except one (this is located in the neighboring exon 5) published so far are located. These data point to a specific role of exon 4 encoded sequence of UMOD in the generation of the MCKD2 renal phenotype.
- Published
- 2003
- Full Text
- View/download PDF
46. Refinement of the critical region for MCKD1 by detection of transcontinental haplotype sharing.
- Author
-
Wolf MT, Karle SM, Schwarz S, Anlauf M, Anlauf M, Glaeser L, Kroiss S, Burton C, Feest T, Otto E, Fuchshuber A, and Hildebrandt F
- Subjects
- Humans, Microsatellite Repeats, Polymorphism, Genetic, Protein Structure, Tertiary genetics, Haplotypes, Multicystic Dysplastic Kidney genetics
- Abstract
Background: Autosomal-dominant medullary cystic kidney disease type 1 (MCKD1) [OMIM 174000] is a hereditary nephropathy that leads to renal salt wasting and end-stage renal failure at a median age of 62 years. In a Welsh MCKD1 kindred we have recently demonstrated linkage to the MCKD1 locus on chromosome 1q23.1 and refined the critical MCKD1 region to <3.3 Mb., Methods: In order to refine the candidate gene region for MCKD1, high-resolution haplotype analysis in three large kindreds with MCKD1 was performed., Results: We report here on high-resolution haplotype analysis in this Welsh kindred, as well as in the Arizona kindred, which was used for the first definition of MCKD as a disease entity, and in a kindred from the Dutch/German border. We detected extensive haplotype sharing among all affected individuals of all three kindreds. Scrutinization of the genealogy of the Arizona kindred revealed an origin from Germany in the 17th century, thereby providing historical data for haplotype sharing by descent at the MCKD1 locus., Conclusion: Under the hypothesis of haplotype sharing by descent, we refined the critical genetic interval to <650 kb, thus enabling candidate gene analysis.
- Published
- 2003
- Full Text
- View/download PDF
47. Autosomal-dominant medullary cystic kidney disease type 1: clinical and molecular findings in six large Cypriot families.
- Author
-
Stavrou C, Koptides M, Tombazos C, Psara E, Patsias C, Zouvani I, Kyriacou K, Hildebrandt F, Christofides T, Pierides A, and Deltas CC
- Subjects
- Cyprus, Family Characteristics, Family Health, Female, Heterozygote, Humans, Hypertension, Renal genetics, Hypertension, Renal pathology, Hyperuricemia genetics, Hyperuricemia pathology, Male, Pedigree, Polycystic Kidney, Autosomal Dominant pathology, Sodium metabolism, Specific Gravity, Urine, Genetic Linkage, Polycystic Kidney, Autosomal Dominant genetics
- Abstract
Background: Autosomal-dominant medullary cystic kidney disease (ADMCKD), a hereditary chronic interstitial nephropathy, recently attracted attention because of the cloning or mapping of certain gene loci, namely NPHP1, NPHP2 and NPHP3 for familial juvenile nephronophthisis (NPH) and MCKD1 and MCKD2 for the adult form of medullary cystic kidney disease. Our aim was to present and discuss the clinical, biochemical, sonographic and histopathological findings in six large Cypriot families in whom molecular analysis has confirmed linkage to the MCKD1 locus on chromosome 1q21., Methods: The clinical, biochemical, sonographic and histopathological findings in 186 members of six large Cypriot families with ADMCKD-1 are presented. Creatinine clearance was calculated according to the Cockroft-Gault formula and was corrected to a body surface area (BSA) of 1.73 m2. DNA linkage analysis was performed with previously identified flanking polymorphic markers., Results: This disease is characterized by the absence of urinary findings in the vast majority of patients, leading to end-stage renal failure (ESRF) at a mean age of 53.7 years. Hypertension and hyperuricemia are common, especially in males, the former encountered more frequently in advanced chronic renal failure (CRF). Gout has been noted in a small percentage of male patients. Loss of urinary concentrating ability was not a prominent early feature of the disease, while severe natriuresis was observed in a few males toward ESRF. Renal cysts are mainly corticomedullary or medullary, and they are present in about 40.3% of patients and appear more frequently near ESRF., Conclusion: ADMCKD type 1 is a common cause of ESRF among our dialysis population. The disease is difficult to diagnose clinically, particularly in the early stage when renal cysts are not usually present, making them a weak diagnostic finding. A dominant pattern of inheritance and DNA linkage analysis are helpful in the diagnosis of this disease.
- Published
- 2002
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.