Cargando…

CTNNB1-Mutant Aldosterone-Producing Adenomas With Somatic Mutations of GNA11/GNAQ Have Distinct Phenotype and Genotype

Background: We report (this meeting) somatic mutation of GNA11/Q in CTNNB1-mutant APAs. The recurrent co-driver mutation causes reversible hypertension in puberty, pregnancy, or menopause. We have investigated the molecular mechanism of this association. Methods: Gene expression profiles in 3 double...

Descripción completa

Detalles Bibliográficos
Autores principales: Zhou, Junhua, Boulkroun, Sheerazed, Cabrera, Claudia P, Azizan, Elena A B, Fernandes-Rosa, Fabio, Cottrell, Emily, Argentesi, Giulia, Wu, Xilin, O’Toole, Sam, Marker, Alison, Jordan, Suzanne, Berney, Daniel M, Lines, Kate, Metherell, Louise, Teo, Ada, Thakker, Rajesh V, Drake, William, Wozniak, Eva, Mein, Charles A, Storr, Helen L, Zennaro, Maria-Christina, Brown, Morris J
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Oxford University Press 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8089757/
http://dx.doi.org/10.1210/jendso/bvab048.133
_version_ 1783687115572248576
author Zhou, Junhua
Boulkroun, Sheerazed
Cabrera, Claudia P
Azizan, Elena A B
Fernandes-Rosa, Fabio
Cottrell, Emily
Argentesi, Giulia
Wu, Xilin
O’Toole, Sam
Marker, Alison
Jordan, Suzanne
Berney, Daniel M
Lines, Kate
Metherell, Louise
Teo, Ada
Thakker, Rajesh V
Drake, William
Wozniak, Eva
Mein, Charles A
Storr, Helen L
Zennaro, Maria-Christina
Brown, Morris J
author_facet Zhou, Junhua
Boulkroun, Sheerazed
Cabrera, Claudia P
Azizan, Elena A B
Fernandes-Rosa, Fabio
Cottrell, Emily
Argentesi, Giulia
Wu, Xilin
O’Toole, Sam
Marker, Alison
Jordan, Suzanne
Berney, Daniel M
Lines, Kate
Metherell, Louise
Teo, Ada
Thakker, Rajesh V
Drake, William
Wozniak, Eva
Mein, Charles A
Storr, Helen L
Zennaro, Maria-Christina
Brown, Morris J
author_sort Zhou, Junhua
collection PubMed
description Background: We report (this meeting) somatic mutation of GNA11/Q in CTNNB1-mutant APAs. The recurrent co-driver mutation causes reversible hypertension in puberty, pregnancy, or menopause. We have investigated the molecular mechanism of this association. Methods: Gene expression profiles in 3 double mutant APAs were studied by unsupervised hierarchical clustering analysis and enrichment analysis of 362 differentially expressed genes and validated by qPCR, IFC and IHC in 10 double mutant APAs or transfected primary adrenal cells. Multiple region biopsies were performed in 7 adrenals adjacent to double-mutant APAs and 4 APAs with KCNJ5 or CACNA1D mutations. The findings of APA mutations in adjacent adrenals were replicated in each case by ddPCR ± NGS. Results: Unsupervised hierarchical clustering analysis showed clustering of the double-mutant APAs, and a high proportion of genes were many-fold upregulated compared to other APAs. LHCGR, TMEM132E, DKK1, C9orf84, FAP, GNRHR and MPP3 are among the genes with high expression. A small number of genes are down-regulated in the double-mutant APAs, including CYP11B1. qPCR confirmed an average of ~10 to 1000-fold higher expression of the hallmark genes in double-mutants. Enrichment analysis showed significant enrichment of features or terms concerned with cell junction and cell adhesion (P<10(–8)). IFC confirmed LHCGR intensity was 31–144 fold higher in primary adrenal cells with GNA11-p.Gln209Pro transfection and high CTNNB1 intensity. LHCGR intensity was qualitatively and quantitatively associated with immunofluorescence for CTNNB1. IHC of double-mutant APAs showed absent CYP11B1 but strong staining of CYP11B2. qPCR confirmed a lower CYP11B1/CYP11B2 ratio and a higher LHCGR expression (P<10(–3), both). IHC confirmed LHCGR positivity in double-mutant APAs but distribution varied both within and between cells. Adjacent ZG was hyperplastic, with absence of both CYP11B1 and CYP11B2 staining, but weak/moderate staining for LHCGR. The same GNA11 ± CTNNB1 somatic mutations were detected in multiple regions of the adjacent adrenals to 3 double mutant APAs. qPCR of hallmark APA genes differed from the APAs. High concordance between ddPCR, NGS and Sanger sequencing of the findings of APA mutations in adjacent adrenals when analysed in the same sample. No mutations were found in 4 adrenals adjacent to APAs with KCNJ5 or CACNA1D mutations, nor in other 4 adrenals adjacent to double-mutant APAs. Conclusions: Patients harboring APAs with somatic mutations in both GNA11/GNAQ Q209 and CTNNB1 have distinct phenotype in both the APAs and their adjacent adrenals. Same GNA11 ± CTNNB1 somatic mutations were found in the adjacent adrenals to double mutant APAs. We infer that a double-hit within related pathways is more likely than a single-hit to cause large increases in expression of LHCGR, and of other genes which may influence clinical presentation.
format Online
Article
Text
id pubmed-8089757
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Oxford University Press
record_format MEDLINE/PubMed
spelling pubmed-80897572021-05-06 CTNNB1-Mutant Aldosterone-Producing Adenomas With Somatic Mutations of GNA11/GNAQ Have Distinct Phenotype and Genotype Zhou, Junhua Boulkroun, Sheerazed Cabrera, Claudia P Azizan, Elena A B Fernandes-Rosa, Fabio Cottrell, Emily Argentesi, Giulia Wu, Xilin O’Toole, Sam Marker, Alison Jordan, Suzanne Berney, Daniel M Lines, Kate Metherell, Louise Teo, Ada Thakker, Rajesh V Drake, William Wozniak, Eva Mein, Charles A Storr, Helen L Zennaro, Maria-Christina Brown, Morris J J Endocr Soc Adrenal Background: We report (this meeting) somatic mutation of GNA11/Q in CTNNB1-mutant APAs. The recurrent co-driver mutation causes reversible hypertension in puberty, pregnancy, or menopause. We have investigated the molecular mechanism of this association. Methods: Gene expression profiles in 3 double mutant APAs were studied by unsupervised hierarchical clustering analysis and enrichment analysis of 362 differentially expressed genes and validated by qPCR, IFC and IHC in 10 double mutant APAs or transfected primary adrenal cells. Multiple region biopsies were performed in 7 adrenals adjacent to double-mutant APAs and 4 APAs with KCNJ5 or CACNA1D mutations. The findings of APA mutations in adjacent adrenals were replicated in each case by ddPCR ± NGS. Results: Unsupervised hierarchical clustering analysis showed clustering of the double-mutant APAs, and a high proportion of genes were many-fold upregulated compared to other APAs. LHCGR, TMEM132E, DKK1, C9orf84, FAP, GNRHR and MPP3 are among the genes with high expression. A small number of genes are down-regulated in the double-mutant APAs, including CYP11B1. qPCR confirmed an average of ~10 to 1000-fold higher expression of the hallmark genes in double-mutants. Enrichment analysis showed significant enrichment of features or terms concerned with cell junction and cell adhesion (P<10(–8)). IFC confirmed LHCGR intensity was 31–144 fold higher in primary adrenal cells with GNA11-p.Gln209Pro transfection and high CTNNB1 intensity. LHCGR intensity was qualitatively and quantitatively associated with immunofluorescence for CTNNB1. IHC of double-mutant APAs showed absent CYP11B1 but strong staining of CYP11B2. qPCR confirmed a lower CYP11B1/CYP11B2 ratio and a higher LHCGR expression (P<10(–3), both). IHC confirmed LHCGR positivity in double-mutant APAs but distribution varied both within and between cells. Adjacent ZG was hyperplastic, with absence of both CYP11B1 and CYP11B2 staining, but weak/moderate staining for LHCGR. The same GNA11 ± CTNNB1 somatic mutations were detected in multiple regions of the adjacent adrenals to 3 double mutant APAs. qPCR of hallmark APA genes differed from the APAs. High concordance between ddPCR, NGS and Sanger sequencing of the findings of APA mutations in adjacent adrenals when analysed in the same sample. No mutations were found in 4 adrenals adjacent to APAs with KCNJ5 or CACNA1D mutations, nor in other 4 adrenals adjacent to double-mutant APAs. Conclusions: Patients harboring APAs with somatic mutations in both GNA11/GNAQ Q209 and CTNNB1 have distinct phenotype in both the APAs and their adjacent adrenals. Same GNA11 ± CTNNB1 somatic mutations were found in the adjacent adrenals to double mutant APAs. We infer that a double-hit within related pathways is more likely than a single-hit to cause large increases in expression of LHCGR, and of other genes which may influence clinical presentation. Oxford University Press 2021-05-03 /pmc/articles/PMC8089757/ http://dx.doi.org/10.1210/jendso/bvab048.133 Text en © The Author(s) 2021. Published by Oxford University Press on behalf of the Endocrine Society. https://creativecommons.org/licenses/by-nc-nd/4.0/This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivs licence (http://creativecommons.org/licenses/by-nc-nd/4.0/ (https://creativecommons.org/licenses/by-nc-nd/4.0/) ), which permits non-commercial reproduction and distribution of the work, in any medium, provided the original work is not altered or transformed in any way, and that the work is properly cited. For commercial re-use, please contact journals.permissions@oup.com
spellingShingle Adrenal
Zhou, Junhua
Boulkroun, Sheerazed
Cabrera, Claudia P
Azizan, Elena A B
Fernandes-Rosa, Fabio
Cottrell, Emily
Argentesi, Giulia
Wu, Xilin
O’Toole, Sam
Marker, Alison
Jordan, Suzanne
Berney, Daniel M
Lines, Kate
Metherell, Louise
Teo, Ada
Thakker, Rajesh V
Drake, William
Wozniak, Eva
Mein, Charles A
Storr, Helen L
Zennaro, Maria-Christina
Brown, Morris J
CTNNB1-Mutant Aldosterone-Producing Adenomas With Somatic Mutations of GNA11/GNAQ Have Distinct Phenotype and Genotype
title CTNNB1-Mutant Aldosterone-Producing Adenomas With Somatic Mutations of GNA11/GNAQ Have Distinct Phenotype and Genotype
title_full CTNNB1-Mutant Aldosterone-Producing Adenomas With Somatic Mutations of GNA11/GNAQ Have Distinct Phenotype and Genotype
title_fullStr CTNNB1-Mutant Aldosterone-Producing Adenomas With Somatic Mutations of GNA11/GNAQ Have Distinct Phenotype and Genotype
title_full_unstemmed CTNNB1-Mutant Aldosterone-Producing Adenomas With Somatic Mutations of GNA11/GNAQ Have Distinct Phenotype and Genotype
title_short CTNNB1-Mutant Aldosterone-Producing Adenomas With Somatic Mutations of GNA11/GNAQ Have Distinct Phenotype and Genotype
title_sort ctnnb1-mutant aldosterone-producing adenomas with somatic mutations of gna11/gnaq have distinct phenotype and genotype
topic Adrenal
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8089757/
http://dx.doi.org/10.1210/jendso/bvab048.133
work_keys_str_mv AT zhoujunhua ctnnb1mutantaldosteroneproducingadenomaswithsomaticmutationsofgna11gnaqhavedistinctphenotypeandgenotype
AT boulkrounsheerazed ctnnb1mutantaldosteroneproducingadenomaswithsomaticmutationsofgna11gnaqhavedistinctphenotypeandgenotype
AT cabreraclaudiap ctnnb1mutantaldosteroneproducingadenomaswithsomaticmutationsofgna11gnaqhavedistinctphenotypeandgenotype
AT azizanelenaab ctnnb1mutantaldosteroneproducingadenomaswithsomaticmutationsofgna11gnaqhavedistinctphenotypeandgenotype
AT fernandesrosafabio ctnnb1mutantaldosteroneproducingadenomaswithsomaticmutationsofgna11gnaqhavedistinctphenotypeandgenotype
AT cottrellemily ctnnb1mutantaldosteroneproducingadenomaswithsomaticmutationsofgna11gnaqhavedistinctphenotypeandgenotype
AT argentesigiulia ctnnb1mutantaldosteroneproducingadenomaswithsomaticmutationsofgna11gnaqhavedistinctphenotypeandgenotype
AT wuxilin ctnnb1mutantaldosteroneproducingadenomaswithsomaticmutationsofgna11gnaqhavedistinctphenotypeandgenotype
AT otoolesam ctnnb1mutantaldosteroneproducingadenomaswithsomaticmutationsofgna11gnaqhavedistinctphenotypeandgenotype
AT markeralison ctnnb1mutantaldosteroneproducingadenomaswithsomaticmutationsofgna11gnaqhavedistinctphenotypeandgenotype
AT jordansuzanne ctnnb1mutantaldosteroneproducingadenomaswithsomaticmutationsofgna11gnaqhavedistinctphenotypeandgenotype
AT berneydanielm ctnnb1mutantaldosteroneproducingadenomaswithsomaticmutationsofgna11gnaqhavedistinctphenotypeandgenotype
AT lineskate ctnnb1mutantaldosteroneproducingadenomaswithsomaticmutationsofgna11gnaqhavedistinctphenotypeandgenotype
AT metherelllouise ctnnb1mutantaldosteroneproducingadenomaswithsomaticmutationsofgna11gnaqhavedistinctphenotypeandgenotype
AT teoada ctnnb1mutantaldosteroneproducingadenomaswithsomaticmutationsofgna11gnaqhavedistinctphenotypeandgenotype
AT thakkerrajeshv ctnnb1mutantaldosteroneproducingadenomaswithsomaticmutationsofgna11gnaqhavedistinctphenotypeandgenotype
AT drakewilliam ctnnb1mutantaldosteroneproducingadenomaswithsomaticmutationsofgna11gnaqhavedistinctphenotypeandgenotype
AT wozniakeva ctnnb1mutantaldosteroneproducingadenomaswithsomaticmutationsofgna11gnaqhavedistinctphenotypeandgenotype
AT meincharlesa ctnnb1mutantaldosteroneproducingadenomaswithsomaticmutationsofgna11gnaqhavedistinctphenotypeandgenotype
AT storrhelenl ctnnb1mutantaldosteroneproducingadenomaswithsomaticmutationsofgna11gnaqhavedistinctphenotypeandgenotype
AT zennaromariachristina ctnnb1mutantaldosteroneproducingadenomaswithsomaticmutationsofgna11gnaqhavedistinctphenotypeandgenotype
AT brownmorrisj ctnnb1mutantaldosteroneproducingadenomaswithsomaticmutationsofgna11gnaqhavedistinctphenotypeandgenotype