Cargando…

Enhanced Ca(2+) signaling, mild primary aldosteronism, and hypertension in a familial hyperaldosteronism mouse model (Cacna1h(M1560V/+))

Gain-of-function mutations in the CACNA1H gene (encoding the T-type calcium channel Ca(V)3.2) cause autosomal-dominant familial hyperaldosteronism type IV (FH-IV) and early-onset hypertension in humans. We used CRISPR/Cas9 to generate Cacna1h(M1560V/+) knockin mice as a model of the most common FH-I...

Descripción completa

Detalles Bibliográficos
Autores principales: Seidel, Eric, Schewe, Julia, Zhang, Junhui, Dinh, Hoang An, Forslund, Sofia K., Markó, Lajos, Hellmig, Nicole, Peters, Jörg, Muller, Dominik N., Lifton, Richard P., Nottoli, Timothy, Stölting, Gabriel, Scholl, Ute I.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: National Academy of Sciences 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8092574/
https://www.ncbi.nlm.nih.gov/pubmed/33879608
http://dx.doi.org/10.1073/pnas.2014876118
_version_ 1783687665116250112
author Seidel, Eric
Schewe, Julia
Zhang, Junhui
Dinh, Hoang An
Forslund, Sofia K.
Markó, Lajos
Hellmig, Nicole
Peters, Jörg
Muller, Dominik N.
Lifton, Richard P.
Nottoli, Timothy
Stölting, Gabriel
Scholl, Ute I.
author_facet Seidel, Eric
Schewe, Julia
Zhang, Junhui
Dinh, Hoang An
Forslund, Sofia K.
Markó, Lajos
Hellmig, Nicole
Peters, Jörg
Muller, Dominik N.
Lifton, Richard P.
Nottoli, Timothy
Stölting, Gabriel
Scholl, Ute I.
author_sort Seidel, Eric
collection PubMed
description Gain-of-function mutations in the CACNA1H gene (encoding the T-type calcium channel Ca(V)3.2) cause autosomal-dominant familial hyperaldosteronism type IV (FH-IV) and early-onset hypertension in humans. We used CRISPR/Cas9 to generate Cacna1h(M1560V/+) knockin mice as a model of the most common FH-IV mutation, along with corresponding knockout mice (Cacna1h(−/−)). Adrenal morphology of both Cacna1h(M1560V/+) and Cacna1h(−/−) mice was normal. Cacna1h(M1560V/+) mice had elevated aldosterone:renin ratios (a screening parameter for primary aldosteronism). Their adrenal Cyp11b2 (aldosterone synthase) expression was increased and remained elevated on a high-salt diet (relative autonomy, characteristic of primary aldosteronism), but plasma aldosterone was only elevated in male animals. The systolic blood pressure of Cacna1h(M1560V/+) mice was 8 mmHg higher than in wild-type littermates and remained elevated on a high-salt diet. Cacna1h(−/−) mice had elevated renal Ren1 (renin-1) expression but normal adrenal Cyp11b2 levels, suggesting that in the absence of Ca(V)3.2, stimulation of the renin-angiotensin system activates alternative calcium entry pathways to maintain normal aldosterone production. On a cellular level, Cacna1h(M1560V/+) adrenal slices showed increased baseline and peak intracellular calcium concentrations in the zona glomerulosa compared to controls, but the frequency of calcium spikes did not rise. We conclude that FH-IV, on a molecular level, is caused by elevated intracellular Ca(2+) concentrations as a signal for aldosterone production in adrenal glomerulosa cells. We demonstrate that a germline Cacna1h gain-of-function mutation is sufficient to cause mild primary aldosteronism, whereas loss of Ca(V)3.2 channel function can be compensated for in a chronic setting.
format Online
Article
Text
id pubmed-8092574
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher National Academy of Sciences
record_format MEDLINE/PubMed
spelling pubmed-80925742021-05-12 Enhanced Ca(2+) signaling, mild primary aldosteronism, and hypertension in a familial hyperaldosteronism mouse model (Cacna1h(M1560V/+)) Seidel, Eric Schewe, Julia Zhang, Junhui Dinh, Hoang An Forslund, Sofia K. Markó, Lajos Hellmig, Nicole Peters, Jörg Muller, Dominik N. Lifton, Richard P. Nottoli, Timothy Stölting, Gabriel Scholl, Ute I. Proc Natl Acad Sci U S A Biological Sciences Gain-of-function mutations in the CACNA1H gene (encoding the T-type calcium channel Ca(V)3.2) cause autosomal-dominant familial hyperaldosteronism type IV (FH-IV) and early-onset hypertension in humans. We used CRISPR/Cas9 to generate Cacna1h(M1560V/+) knockin mice as a model of the most common FH-IV mutation, along with corresponding knockout mice (Cacna1h(−/−)). Adrenal morphology of both Cacna1h(M1560V/+) and Cacna1h(−/−) mice was normal. Cacna1h(M1560V/+) mice had elevated aldosterone:renin ratios (a screening parameter for primary aldosteronism). Their adrenal Cyp11b2 (aldosterone synthase) expression was increased and remained elevated on a high-salt diet (relative autonomy, characteristic of primary aldosteronism), but plasma aldosterone was only elevated in male animals. The systolic blood pressure of Cacna1h(M1560V/+) mice was 8 mmHg higher than in wild-type littermates and remained elevated on a high-salt diet. Cacna1h(−/−) mice had elevated renal Ren1 (renin-1) expression but normal adrenal Cyp11b2 levels, suggesting that in the absence of Ca(V)3.2, stimulation of the renin-angiotensin system activates alternative calcium entry pathways to maintain normal aldosterone production. On a cellular level, Cacna1h(M1560V/+) adrenal slices showed increased baseline and peak intracellular calcium concentrations in the zona glomerulosa compared to controls, but the frequency of calcium spikes did not rise. We conclude that FH-IV, on a molecular level, is caused by elevated intracellular Ca(2+) concentrations as a signal for aldosterone production in adrenal glomerulosa cells. We demonstrate that a germline Cacna1h gain-of-function mutation is sufficient to cause mild primary aldosteronism, whereas loss of Ca(V)3.2 channel function can be compensated for in a chronic setting. National Academy of Sciences 2021-04-27 2021-04-20 /pmc/articles/PMC8092574/ /pubmed/33879608 http://dx.doi.org/10.1073/pnas.2014876118 Text en Copyright © 2021 the Author(s). Published by PNAS. https://creativecommons.org/licenses/by-nc-nd/4.0/This open access article is distributed under Creative Commons Attribution-NonCommercial-NoDerivatives License 4.0 (CC BY-NC-ND) (https://creativecommons.org/licenses/by-nc-nd/4.0/) .
spellingShingle Biological Sciences
Seidel, Eric
Schewe, Julia
Zhang, Junhui
Dinh, Hoang An
Forslund, Sofia K.
Markó, Lajos
Hellmig, Nicole
Peters, Jörg
Muller, Dominik N.
Lifton, Richard P.
Nottoli, Timothy
Stölting, Gabriel
Scholl, Ute I.
Enhanced Ca(2+) signaling, mild primary aldosteronism, and hypertension in a familial hyperaldosteronism mouse model (Cacna1h(M1560V/+))
title Enhanced Ca(2+) signaling, mild primary aldosteronism, and hypertension in a familial hyperaldosteronism mouse model (Cacna1h(M1560V/+))
title_full Enhanced Ca(2+) signaling, mild primary aldosteronism, and hypertension in a familial hyperaldosteronism mouse model (Cacna1h(M1560V/+))
title_fullStr Enhanced Ca(2+) signaling, mild primary aldosteronism, and hypertension in a familial hyperaldosteronism mouse model (Cacna1h(M1560V/+))
title_full_unstemmed Enhanced Ca(2+) signaling, mild primary aldosteronism, and hypertension in a familial hyperaldosteronism mouse model (Cacna1h(M1560V/+))
title_short Enhanced Ca(2+) signaling, mild primary aldosteronism, and hypertension in a familial hyperaldosteronism mouse model (Cacna1h(M1560V/+))
title_sort enhanced ca(2+) signaling, mild primary aldosteronism, and hypertension in a familial hyperaldosteronism mouse model (cacna1h(m1560v/+))
topic Biological Sciences
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8092574/
https://www.ncbi.nlm.nih.gov/pubmed/33879608
http://dx.doi.org/10.1073/pnas.2014876118
work_keys_str_mv AT seideleric enhancedca2signalingmildprimaryaldosteronismandhypertensioninafamilialhyperaldosteronismmousemodelcacna1hm1560v
AT schewejulia enhancedca2signalingmildprimaryaldosteronismandhypertensioninafamilialhyperaldosteronismmousemodelcacna1hm1560v
AT zhangjunhui enhancedca2signalingmildprimaryaldosteronismandhypertensioninafamilialhyperaldosteronismmousemodelcacna1hm1560v
AT dinhhoangan enhancedca2signalingmildprimaryaldosteronismandhypertensioninafamilialhyperaldosteronismmousemodelcacna1hm1560v
AT forslundsofiak enhancedca2signalingmildprimaryaldosteronismandhypertensioninafamilialhyperaldosteronismmousemodelcacna1hm1560v
AT markolajos enhancedca2signalingmildprimaryaldosteronismandhypertensioninafamilialhyperaldosteronismmousemodelcacna1hm1560v
AT hellmignicole enhancedca2signalingmildprimaryaldosteronismandhypertensioninafamilialhyperaldosteronismmousemodelcacna1hm1560v
AT petersjorg enhancedca2signalingmildprimaryaldosteronismandhypertensioninafamilialhyperaldosteronismmousemodelcacna1hm1560v
AT mullerdominikn enhancedca2signalingmildprimaryaldosteronismandhypertensioninafamilialhyperaldosteronismmousemodelcacna1hm1560v
AT liftonrichardp enhancedca2signalingmildprimaryaldosteronismandhypertensioninafamilialhyperaldosteronismmousemodelcacna1hm1560v
AT nottolitimothy enhancedca2signalingmildprimaryaldosteronismandhypertensioninafamilialhyperaldosteronismmousemodelcacna1hm1560v
AT stoltinggabriel enhancedca2signalingmildprimaryaldosteronismandhypertensioninafamilialhyperaldosteronismmousemodelcacna1hm1560v
AT schollutei enhancedca2signalingmildprimaryaldosteronismandhypertensioninafamilialhyperaldosteronismmousemodelcacna1hm1560v