GAPVD1 and ANKFY1 Mutations Implicate RAB5 Regulation in Nephrotic Syndrome

Standard

GAPVD1 and ANKFY1 Mutations Implicate RAB5 Regulation in Nephrotic Syndrome. / Hermle, Tobias; Schneider, Ronen; Schapiro, David; Braun, Daniela A; van der Ven, Amelie T; Warejko, Jillian K; Daga, Ankana; Widmeier, Eugen; Nakayama, Makiko; Jobst-Schwan, Tilman; Majmundar, Amar J; Ashraf, Shazia; Rao, Jia; Finn, Laura S; Tasic, Velibor; Hernandez, Joel D; Bagga, Arvind; Jalalah, Sawsan M; El Desoky, Sherif; Kari, Jameela A; Laricchia, Kristen M; Lek, Monkol; Rehm, Heidi L; MacArthur, Daniel G; Mane, Shrikant; Lifton, Richard P; Shril, Shirlee; Hildebrandt, Friedhelm.

in: J AM SOC NEPHROL, Jahrgang 29, Nr. 8, 08.2018, S. 2123-2138.

Publikationen: SCORING: Beitrag in Fachzeitschrift/ZeitungSCORING: ZeitschriftenaufsatzForschungBegutachtung

Harvard

Hermle, T, Schneider, R, Schapiro, D, Braun, DA, van der Ven, AT, Warejko, JK, Daga, A, Widmeier, E, Nakayama, M, Jobst-Schwan, T, Majmundar, AJ, Ashraf, S, Rao, J, Finn, LS, Tasic, V, Hernandez, JD, Bagga, A, Jalalah, SM, El Desoky, S, Kari, JA, Laricchia, KM, Lek, M, Rehm, HL, MacArthur, DG, Mane, S, Lifton, RP, Shril, S & Hildebrandt, F 2018, 'GAPVD1 and ANKFY1 Mutations Implicate RAB5 Regulation in Nephrotic Syndrome', J AM SOC NEPHROL, Jg. 29, Nr. 8, S. 2123-2138. https://doi.org/10.1681/ASN.2017121312

APA

Hermle, T., Schneider, R., Schapiro, D., Braun, D. A., van der Ven, A. T., Warejko, J. K., Daga, A., Widmeier, E., Nakayama, M., Jobst-Schwan, T., Majmundar, A. J., Ashraf, S., Rao, J., Finn, L. S., Tasic, V., Hernandez, J. D., Bagga, A., Jalalah, S. M., El Desoky, S., ... Hildebrandt, F. (2018). GAPVD1 and ANKFY1 Mutations Implicate RAB5 Regulation in Nephrotic Syndrome. J AM SOC NEPHROL, 29(8), 2123-2138. https://doi.org/10.1681/ASN.2017121312

Vancouver

Bibtex

@article{4896f002d84a4c18a56f77b097d33892,
title = "GAPVD1 and ANKFY1 Mutations Implicate RAB5 Regulation in Nephrotic Syndrome",
abstract = "BACKGROUND: Steroid-resistant nephrotic syndrome (SRNS) is a frequent cause of CKD. The discovery of monogenic causes of SRNS has revealed specific pathogenetic pathways, but these monogenic causes do not explain all cases of SRNS.METHODS: To identify novel monogenic causes of SRNS, we screened 665 patients by whole-exome sequencing. We then evaluated the in vitro functional significance of two genes and the mutations therein that we discovered through this sequencing and conducted complementary studies in podocyte-like Drosophila nephrocytes.RESULTS: We identified conserved, homozygous missense mutations of GAPVD1 in two families with early-onset NS and a homozygous missense mutation of ANKFY1 in two siblings with SRNS. GAPVD1 and ANKFY1 interact with the endosomal regulator RAB5. Coimmunoprecipitation assays indicated interaction between GAPVD1 and ANKFY1 proteins, which also colocalized when expressed in HEK293T cells. Silencing either protein diminished the podocyte migration rate. Compared with wild-type GAPVD1 and ANKFY1, the mutated proteins produced upon ectopic expression of GAPVD1 or ANKFY1 bearing the patient-derived mutations exhibited altered binding affinity for active RAB5 and reduced ability to rescue the knockout-induced defect in podocyte migration. Coimmunoprecipitation assays further demonstrated a physical interaction between nephrin and GAPVD1, and immunofluorescence revealed partial colocalization of these proteins in rat glomeruli. The patient-derived GAPVD1 mutations reduced nephrin-GAPVD1 binding affinity. In Drosophila, silencing Gapvd1 impaired endocytosis and caused mistrafficking of the nephrin ortholog.CONCLUSIONS: Mutations in GAPVD1 and probably in ANKFY1 are novel monogenic causes of NS. The discovery of these genes implicates RAB5 regulation in the pathogenesis of human NS.",
keywords = "Animals, Cell Movement/genetics, Cells, Cultured, Cohort Studies, Disease Progression, Drosophila melanogaster, Female, Gene Expression Regulation, Genetic Predisposition to Disease, Humans, Male, Mass Screening/methods, Membrane Proteins/genetics, Mutation, Missense, Nephrotic Syndrome/genetics, Pedigree, Podocytes/metabolism, RNA, Small Interfering/genetics, Real-Time Polymerase Chain Reaction/methods, Renal Insufficiency, Chronic/genetics, Whole Exome Sequencing, rab5 GTP-Binding Proteins/genetics",
author = "Tobias Hermle and Ronen Schneider and David Schapiro and Braun, {Daniela A} and {van der Ven}, {Amelie T} and Warejko, {Jillian K} and Ankana Daga and Eugen Widmeier and Makiko Nakayama and Tilman Jobst-Schwan and Majmundar, {Amar J} and Shazia Ashraf and Jia Rao and Finn, {Laura S} and Velibor Tasic and Hernandez, {Joel D} and Arvind Bagga and Jalalah, {Sawsan M} and {El Desoky}, Sherif and Kari, {Jameela A} and Laricchia, {Kristen M} and Monkol Lek and Rehm, {Heidi L} and MacArthur, {Daniel G} and Shrikant Mane and Lifton, {Richard P} and Shirlee Shril and Friedhelm Hildebrandt",
note = "Copyright {\textcopyright} 2018 by the American Society of Nephrology.",
year = "2018",
month = aug,
doi = "10.1681/ASN.2017121312",
language = "English",
volume = "29",
pages = "2123--2138",
journal = "J AM SOC NEPHROL",
issn = "1046-6673",
publisher = "American Society of Nephrology",
number = "8",

}

RIS

TY - JOUR

T1 - GAPVD1 and ANKFY1 Mutations Implicate RAB5 Regulation in Nephrotic Syndrome

AU - Hermle, Tobias

AU - Schneider, Ronen

AU - Schapiro, David

AU - Braun, Daniela A

AU - van der Ven, Amelie T

AU - Warejko, Jillian K

AU - Daga, Ankana

AU - Widmeier, Eugen

AU - Nakayama, Makiko

AU - Jobst-Schwan, Tilman

AU - Majmundar, Amar J

AU - Ashraf, Shazia

AU - Rao, Jia

AU - Finn, Laura S

AU - Tasic, Velibor

AU - Hernandez, Joel D

AU - Bagga, Arvind

AU - Jalalah, Sawsan M

AU - El Desoky, Sherif

AU - Kari, Jameela A

AU - Laricchia, Kristen M

AU - Lek, Monkol

AU - Rehm, Heidi L

AU - MacArthur, Daniel G

AU - Mane, Shrikant

AU - Lifton, Richard P

AU - Shril, Shirlee

AU - Hildebrandt, Friedhelm

N1 - Copyright © 2018 by the American Society of Nephrology.

PY - 2018/8

Y1 - 2018/8

N2 - BACKGROUND: Steroid-resistant nephrotic syndrome (SRNS) is a frequent cause of CKD. The discovery of monogenic causes of SRNS has revealed specific pathogenetic pathways, but these monogenic causes do not explain all cases of SRNS.METHODS: To identify novel monogenic causes of SRNS, we screened 665 patients by whole-exome sequencing. We then evaluated the in vitro functional significance of two genes and the mutations therein that we discovered through this sequencing and conducted complementary studies in podocyte-like Drosophila nephrocytes.RESULTS: We identified conserved, homozygous missense mutations of GAPVD1 in two families with early-onset NS and a homozygous missense mutation of ANKFY1 in two siblings with SRNS. GAPVD1 and ANKFY1 interact with the endosomal regulator RAB5. Coimmunoprecipitation assays indicated interaction between GAPVD1 and ANKFY1 proteins, which also colocalized when expressed in HEK293T cells. Silencing either protein diminished the podocyte migration rate. Compared with wild-type GAPVD1 and ANKFY1, the mutated proteins produced upon ectopic expression of GAPVD1 or ANKFY1 bearing the patient-derived mutations exhibited altered binding affinity for active RAB5 and reduced ability to rescue the knockout-induced defect in podocyte migration. Coimmunoprecipitation assays further demonstrated a physical interaction between nephrin and GAPVD1, and immunofluorescence revealed partial colocalization of these proteins in rat glomeruli. The patient-derived GAPVD1 mutations reduced nephrin-GAPVD1 binding affinity. In Drosophila, silencing Gapvd1 impaired endocytosis and caused mistrafficking of the nephrin ortholog.CONCLUSIONS: Mutations in GAPVD1 and probably in ANKFY1 are novel monogenic causes of NS. The discovery of these genes implicates RAB5 regulation in the pathogenesis of human NS.

AB - BACKGROUND: Steroid-resistant nephrotic syndrome (SRNS) is a frequent cause of CKD. The discovery of monogenic causes of SRNS has revealed specific pathogenetic pathways, but these monogenic causes do not explain all cases of SRNS.METHODS: To identify novel monogenic causes of SRNS, we screened 665 patients by whole-exome sequencing. We then evaluated the in vitro functional significance of two genes and the mutations therein that we discovered through this sequencing and conducted complementary studies in podocyte-like Drosophila nephrocytes.RESULTS: We identified conserved, homozygous missense mutations of GAPVD1 in two families with early-onset NS and a homozygous missense mutation of ANKFY1 in two siblings with SRNS. GAPVD1 and ANKFY1 interact with the endosomal regulator RAB5. Coimmunoprecipitation assays indicated interaction between GAPVD1 and ANKFY1 proteins, which also colocalized when expressed in HEK293T cells. Silencing either protein diminished the podocyte migration rate. Compared with wild-type GAPVD1 and ANKFY1, the mutated proteins produced upon ectopic expression of GAPVD1 or ANKFY1 bearing the patient-derived mutations exhibited altered binding affinity for active RAB5 and reduced ability to rescue the knockout-induced defect in podocyte migration. Coimmunoprecipitation assays further demonstrated a physical interaction between nephrin and GAPVD1, and immunofluorescence revealed partial colocalization of these proteins in rat glomeruli. The patient-derived GAPVD1 mutations reduced nephrin-GAPVD1 binding affinity. In Drosophila, silencing Gapvd1 impaired endocytosis and caused mistrafficking of the nephrin ortholog.CONCLUSIONS: Mutations in GAPVD1 and probably in ANKFY1 are novel monogenic causes of NS. The discovery of these genes implicates RAB5 regulation in the pathogenesis of human NS.

KW - Animals

KW - Cell Movement/genetics

KW - Cells, Cultured

KW - Cohort Studies

KW - Disease Progression

KW - Drosophila melanogaster

KW - Female

KW - Gene Expression Regulation

KW - Genetic Predisposition to Disease

KW - Humans

KW - Male

KW - Mass Screening/methods

KW - Membrane Proteins/genetics

KW - Mutation, Missense

KW - Nephrotic Syndrome/genetics

KW - Pedigree

KW - Podocytes/metabolism

KW - RNA, Small Interfering/genetics

KW - Real-Time Polymerase Chain Reaction/methods

KW - Renal Insufficiency, Chronic/genetics

KW - Whole Exome Sequencing

KW - rab5 GTP-Binding Proteins/genetics

U2 - 10.1681/ASN.2017121312

DO - 10.1681/ASN.2017121312

M3 - SCORING: Journal article

C2 - 29959197

VL - 29

SP - 2123

EP - 2138

JO - J AM SOC NEPHROL

JF - J AM SOC NEPHROL

SN - 1046-6673

IS - 8

ER -