Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Basic Res Cardiol ; 115(4): 49, 2020 06 26.
Article in English | MEDLINE | ID: mdl-32592107

ABSTRACT

Heart failure (HF) is a leading cause of morbidity and mortality worldwide. Patients with HF exhibit a loss of junctophilin-2 (JPH2), a structural protein critical in forming junctional membrane complexes in which excitation-contraction takes place. Several mechanisms have been proposed to mediate the loss of JPH2, one being cleavage by the calcium-dependent protease calpain. The downstream mechanisms underlying HF progression after JPH2 cleavage are presently poorly understood. In this study, we used Labcas to bioinformatically predict putative calpain cleavage sites on JPH2. We identified a cleavage site that produces a novel C-terminal JPH2 peptide (JPH2-CTP) using several domain-specific antibodies. Western blotting revealed elevated JPH2-CTP levels in hearts of patients and mice with HF, corresponding to increased levels of calpain-2. Moreover, immunocytochemistry demonstrated nuclear localization of JPH2-CTP within ventricular myocytes isolated from a murine model of pressure overload-induced HF as well as rat ventricular myocytes treated with isoproterenol. Nuclear localization of JPH2-CTP and cellular remodeling were abrogated by a genetic mutation of the nuclear localization sequence within JPH2-CTP. Taken together, our studies identified a novel C-terminal fragment of JPH2 (JPH2-CTP) generated by calpain-2 mediated cleavage which localizes within the cardiomyocyte nucleus during HF. Blocking nuclear localization of JPH2-CTP protects cardiomyocytes from isoproterenol-induced hypertrophy in vitro. Future in vivo studies of the nuclear role of JPH2-CTP may reveal a causal association with adverse remodeling during HF and establish CTP as a therapeutic target.


Subject(s)
Calpain/metabolism , Heart Failure/metabolism , Membrane Proteins/metabolism , Myocytes, Cardiac/metabolism , Animals , Cell Nucleus/metabolism , Female , Heart Failure/physiopathology , Humans , Male , Mice , Mice, Inbred C57BL
2.
Sci Rep ; 9(1): 14677, 2019 10 11.
Article in English | MEDLINE | ID: mdl-31605007

ABSTRACT

Primary aldosteronism (PA) is the most frequent form of secondary arterial hypertension. Mutations in different genes increase aldosterone production in PA, but additional mechanisms may contribute to increased cell proliferation and aldosterone producing adenoma (APA) development. We performed transcriptome analysis in APA and identified retinoic acid receptor alpha (RARα) signaling as a central molecular network involved in nodule formation. To understand how RARα modulates adrenal structure and function, we explored the adrenal phenotype of male and female Rarα knockout mice. Inactivation of Rarα in mice led to significant structural disorganization of the adrenal cortex in both sexes, with increased adrenal cortex size in female mice and increased cell proliferation in males. Abnormalities of vessel architecture and extracellular matrix were due to decreased Vegfa expression and modifications in extracellular matrix components. On the molecular level, Rarα inactivation leads to inhibition of non-canonical Wnt signaling, without affecting the canonical Wnt pathway nor PKA signaling. Our study suggests that Rarα contributes to the maintenance of normal adrenal cortex structure and cell proliferation, by modulating Wnt signaling. Dysregulation of this interaction may contribute to abnormal cell proliferation, creating a propitious environment for the emergence of specific driver mutations in PA.


Subject(s)
Hyperaldosteronism/genetics , Hypertension/genetics , Retinoic Acid Receptor alpha/genetics , Vascular Endothelial Growth Factor A/genetics , Adrenal Cortex/metabolism , Adrenal Cortex/pathology , Adrenocortical Adenoma/genetics , Adrenocortical Adenoma/pathology , Animals , Blood Vessels/metabolism , Blood Vessels/pathology , Cell Proliferation/genetics , Extracellular Matrix/genetics , Humans , Hyperaldosteronism/pathology , Hypertension/pathology , Mice , Mice, Knockout , Mutation/genetics , Wnt Signaling Pathway/genetics
3.
Cardiovasc Res ; 114(11): 1474-1486, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29648621

ABSTRACT

Aims: The pathology of heart failure is characterized by poorly contracting and dilated ventricles. At the cellular level, this is associated with lengthening of individual cardiomyocytes and loss of sarcomeres. While it is known that the transcription factor myocyte enhancer factor-2 (MEF2) is involved in this cardiomyocyte remodelling, the underlying mechanism remains to be elucidated. Here, we aim to mechanistically link MEF2 target genes with loss of sarcomeres during cardiomyocyte remodelling. Methods and results: Neonatal rat cardiomyocytes overexpressing MEF2 elongated and lost their sarcomeric structure. We identified myotonic dystrophy protein kinase (DMPK) as direct MEF2 target gene involved in this process. Adenoviral overexpression of DMPK E, the isoform upregulated in heart failure, resulted in severe loss of sarcomeres in vitro, and transgenic mice overexpressing DMPK E displayed disruption of sarcomere structure and cardiomyopathy in vivo. Moreover, we found a decreased expression of sarcomeric genes following DMPK E gain-of-function. These genes are targets of the transcription factor serum response factor (SRF) and we found that DMPK E acts as inhibitor of SRF transcriptional activity. Conclusion: Our data indicate that MEF2-induced loss of sarcomeres is mediated by DMPK via a decrease in sarcomeric gene expression by interfering with SRF transcriptional activity. Together, these results demonstrate an unexpected role for DMPK as a direct mediator of adverse cardiomyocyte remodelling and heart failure.


Subject(s)
Cardiomyopathies/enzymology , Heart Failure/enzymology , MEF2 Transcription Factors/metabolism , Myocytes, Cardiac/enzymology , Myotonin-Protein Kinase/metabolism , Sarcomeres/enzymology , Ventricular Remodeling , Animals , Animals, Genetically Modified , Animals, Newborn , Cardiomyopathies/genetics , Cardiomyopathies/pathology , Cardiomyopathies/physiopathology , Disease Models, Animal , HEK293 Cells , Heart Failure/genetics , Heart Failure/pathology , Heart Failure/physiopathology , Humans , MEF2 Transcription Factors/genetics , Male , Mice, Inbred C57BL , Myocytes, Cardiac/ultrastructure , Myotonin-Protein Kinase/genetics , Phosphorylation , Rats, Wistar , Sarcomeres/genetics , Sarcomeres/ultrastructure , Serum Response Factor/genetics , Serum Response Factor/metabolism , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic
4.
Hum Mol Genet ; 23(4): 889-905, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24087794

ABSTRACT

Primary aldosteronism (PA) is the main cause of secondary hypertension, resulting from adrenal aldosterone-producing adenomas (APA) or bilateral hyperplasia. Here, we show that constitutive activation of WNT/ß-catenin signalling is the most frequent molecular alteration found in 70% of APA. We provide evidence that decreased expression of the WNT inhibitor SFRP2 may be contributing to deregulated WNT signalling and APA development in patients. This is supported by the demonstration that mice with genetic ablation of Sfrp2 have increased aldosterone production and ectopic differentiation of zona glomerulosa cells. We further show that ß-catenin plays an essential role in the control of basal and Angiotensin II-induced aldosterone secretion, by activating AT1R, CYP21 and CYP11B2 transcription. This relies on both LEF/TCF-dependent activation of AT1R and CYP21 regulatory regions and indirect activation of CYP21 and CYP11B2 promoters, through increased expression of the nuclear receptors NURR1 and NUR77. Altogether, these data show that aberrant WNT/ß-catenin activation is associated with APA development and suggest that WNT pathway may be a good therapeutic target in PA.


Subject(s)
Adrenal Cortex Neoplasms/metabolism , Adrenocortical Adenoma/metabolism , Aldosterone/biosynthesis , Hyperaldosteronism/metabolism , Wnt Signaling Pathway , Adrenal Cortex Neoplasms/complications , Adrenocortical Adenoma/complications , Adult , Aldosterone/blood , Aldosterone/metabolism , Animals , Cell Line, Tumor , Cytochrome P-450 CYP11B2/genetics , Cytochrome P-450 CYP11B2/metabolism , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Humans , Hyperaldosteronism/etiology , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism
5.
Mol Cell Endocrinol ; 371(1-2): 221-7, 2013 May 22.
Article in English | MEDLINE | ID: mdl-23376008

ABSTRACT

Somatic mutations of KCNJ5, coding for the potassium channel GIRK4, have recently been implicated in the formation of aldosterone producing adenoma (APA). While a causal link between KCNJ5 mutations, membrane depolarization and aldosterone production has been established, the precise mechanism by which these mutations promote cell proliferation and APA formation remains unclear. The aim of our study was to correlate KCNJ5 mutation status with morphological and functional characteristics of the adrenal cortex adjacent to APA. While GIRK4 was expressed in APA and in the zona glomerulosa of the adjacent cortex, significantly lower levels were detected in APA harboring a KCNJ5 mutation. There was no correlation between KCNJ5 mutation status and the morphological measures of adrenal cortex remodeling, including nodulation, vascularization and expression of CYP11B2. The cell composition of APA was not significantly different between groups. These results indicate that KCNJ5 mutations are not correlated with adrenal cortex remodeling in APA.


Subject(s)
Adrenal Cortex Neoplasms/genetics , Adrenocortical Adenoma/genetics , Aldosterone/metabolism , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , Zona Glomerulosa/metabolism , Adrenal Cortex Neoplasms/metabolism , Adrenocortical Adenoma/metabolism , Aldosterone/biosynthesis , Cell Proliferation , Cytochrome P-450 CYP11B2/biosynthesis , Cytochrome P-450 CYP11B2/genetics , Female , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Humans , Male , Mutation , RNA, Messenger/biosynthesis
6.
Nat Genet ; 45(4): 440-4, 444e1-2, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23416519

ABSTRACT

Primary aldosteronism is the most prevalent form of secondary hypertension. To explore molecular mechanisms of autonomous aldosterone secretion, we performed exome sequencing of aldosterone-producing adenomas (APAs). We identified somatic hotspot mutations in the ATP1A1 (encoding an Na(+)/K(+) ATPase α subunit) and ATP2B3 (encoding a Ca(2+) ATPase) genes in three and two of the nine APAs, respectively. These ATPases are expressed in adrenal cells and control sodium, potassium and calcium ion homeostasis. Functional in vitro studies of ATP1A1 mutants showed loss of pump activity and strongly reduced affinity for potassium. Electrophysiological ex vivo studies on primary adrenal adenoma cells provided further evidence for inappropriate depolarization of cells with ATPase alterations. In a collection of 308 APAs, we found 16 (5.2%) somatic mutations in ATP1A1 and 5 (1.6%) in ATP2B3. Mutation-positive cases showed male dominance, increased plasma aldosterone concentrations and lower potassium concentrations compared with mutation-negative cases. In summary, dominant somatic alterations in two members of the ATPase gene family result in autonomous aldosterone secretion.


Subject(s)
Adrenal Cortex Neoplasms/etiology , Adrenocortical Adenoma/etiology , Aldosterone/metabolism , Hypertension/etiology , Mutation/genetics , Plasma Membrane Calcium-Transporting ATPases/genetics , Sodium-Potassium-Exchanging ATPase/genetics , Calcium/metabolism , Cells, Cultured , Electrophysiology , Humans , Immunoenzyme Techniques , Potassium/metabolism , Sodium/metabolism
7.
Hypertension ; 59(3): 592-8, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22275527

ABSTRACT

Primary aldosteronism is the most common form of secondary hypertension. Mutations in the KCNJ5 gene have been described recently in aldosterone-producing adenomas (APAs). The aim of this study was to investigate the prevalence of KCNJ5 mutations in unselected patients with primary aldosteronism and their clinical, biological and molecular correlates. KCNJ5 sequencing was performed on somatic (APA, n=380) and peripheral (APA, n=344; bilateral adrenal hyperplasia, n=174) DNA of patients with primary aldosteronism, collected through the European Network for the Study of Adrenal Tumors. Transcriptome analysis was performed in 102 tumors. Somatic KCNJ5 mutations (p.Gly151Arg or p.Leu168Arg) were found in 34% (129 of 380) of APA. They were significantly more prevalent in females (49%) than males (19%; P<10(-3)) and in younger patients (42.1±1.0 versus 47.6±0.7 years; P<10(-3)) and were associated with higher preoperative aldosterone levels (455±26 versus 376±17 ng/L; P=0.012) but not with therapeutic outcome after surgery. Germline KCNJ5 mutations were found neither in patients with APA nor those with bilateral adrenal hyperplasia. Somatic KCNJ5 mutations were specific for APA, because they were not identified in 25 peritumoral adrenal tissues or 16 cortisol-producing adenomas. Hierarchical clustering of transcriptome profiles showed that APAs with p.Gly151Arg or p.Leu168Arg mutations were indistinguishable from tumors without KCNJ5 mutations. In conclusion, although a large proportion of sporadic APAs harbors somatic KCNJ5 mutations, germline mutations are not similarly causative for bilateral adrenal hyperplasia. KCNJ5 mutation carriers are more likely to be females; younger age and higher aldosterone levels at diagnosis suggest that KCNJ5 mutations may be associated with a more florid phenotype of primary aldosteronism.


Subject(s)
Aldosterone/blood , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , Hyperaldosteronism/genetics , Mutation , RNA/genetics , Adult , Female , Follow-Up Studies , G Protein-Coupled Inwardly-Rectifying Potassium Channels/blood , Humans , Hyperaldosteronism/blood , Hyperaldosteronism/diagnosis , Magnetic Resonance Imaging , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Prevalence , Real-Time Polymerase Chain Reaction , Retrospective Studies , Tomography, X-Ray Computed
8.
Endocrinology ; 152(12): 4753-63, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21971159

ABSTRACT

Aldosterone producing adenoma (APA) is the most common form of surgically curable hypertension. To further understand mechanisms involved in APA formation, we investigated the expression of molecules linked to adrenal stem/precursor cells [ß-catenin, Sonic hedgehog (Shh), CD56], and nuclear receptors that play key roles in adrenocortical development and function steroidogenic factor 1, dosage-sensitive sex reversal-adrenal hypoplasia congenita critical region on the X chromosome, gene 1) in six control adrenal glands and 14 adrenals with APA and compared their expression with that of specific markers of zona glomerulosa (ZG) [CYP11B2, Disabled 2 (Dab2)]. Both Dab2 and CD56 were expressed in ZG. Although Dab2 associates uniquely with differentiated ZG cells and its expression is lost when cells transdifferentiate to zona fasciculata (ZF) cells, CD56 was also expressed in ZF and in aldosterone-producing cell clusters, confirming that these structures possess an intermediate phenotype between ZG and ZF cells. Shh was barely detectable in cells located to the outer part of the ZG in the control adrenal; in contrast, its expression was detected in the entire APA and was dramatically increased in the hyperplastic peritumoral ZG. Transcriptome profiling revealed differential expression of components of Shh signaling pathway in a subgroup of APA. Similarly, Wnt/ß-catenin signaling was activated in the majority of APA as well as in the entire peritumoral adrenal cortex; however, no mutation was identified in the CTNNB1 gene that could account for ß-catenin activation. Our data suggest that both APA and adjacent ZG present characteristics of stem/precursor cells; the reexpression of genes involved in fetal adrenal development could underlie excessive ZG cell proliferation and APA formation.


Subject(s)
Adenoma/pathology , Adrenal Cortex/pathology , Aldosterone , Stem Cells/pathology , Adenoma/metabolism , Adrenal Cortex/metabolism , Adrenal Glands/pathology , Biomarkers/analysis , Cell Proliferation , Gene Expression Profiling , Humans , Hypertension , In Vitro Techniques , Stem Cells/metabolism , Zona Glomerulosa
9.
Hypertension ; 56(5): 885-92, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20937967

ABSTRACT

Primary aldosteronism is the most common form of secondary hypertension with hypokalemia and suppressed renin-angiotensin system caused by autonomous aldosterone production. Our aim was to compare zona glomerulosa (ZG) structure and function between control adrenals and the peritumoral tissue from patients operated on for aldosterone-producing adenoma. ZG morphology and CYP11B1, CYP11B2, and disabled 2 expression were studied in 15 control adrenals and 25 adrenals with aldosterone-producing adenoma. A transcriptome analysis was done using publicly available data sets. In control adrenals, ZG was discontinuous, and CYP11B2 expression was focal or partly continuous and localized to 3 structures, foci, megafoci, and aldosterone-producing cell clusters. CYP11B2 expression was restricted to a limited number of ZG cells expressing Dab2 but not CYP11B1; aldosterone-producing cell clusters were composed of cells with an intermediate phenotype expressing CYP11B2 but not disabled 2 or CYP11B1. In peritumoral tissue, large remodeling of the adrenal cortex was observed with increased nodulation and decreased vascularization that were not correlated with CYP11B2 expression. In 17 out of 25 adrenals, hyperplasia of adjacent ZG was observed with persistent expression of CYP11B2 that was extended to the entire ZG. In all of the adrenals from patients with aldosterone-producing adenoma, CYP11B2 expression was present in foci, megafoci, and aldosterone-producing cell clusters. Transcriptome profiling indicates a close relationship between peritumoral and control adrenal cortex. In conclusion, adrenal cortex remodeling, reduced vascularization, and ZG hyperplasia are major features of adrenals with aldosterone-producing adenoma. Transcriptional phenotyping is not in favor of this being an intermediate step toward the formation of aldosterone-producing adenoma.


Subject(s)
Adenoma/pathology , Adrenal Cortex Neoplasms/pathology , Adrenal Cortex/pathology , Hyperaldosteronism/pathology , Zona Glomerulosa/pathology , Adenoma/genetics , Adenoma/metabolism , Adrenal Cortex/metabolism , Adrenal Cortex Neoplasms/genetics , Adrenal Cortex Neoplasms/metabolism , Adult , Aldosterone/biosynthesis , Cytochrome P-450 CYP11B2/genetics , Cytochrome P-450 CYP11B2/metabolism , Female , Humans , Hyperaldosteronism/genetics , Hyperaldosteronism/metabolism , Hyperplasia/pathology , Immunohistochemistry , In Situ Hybridization , Male , Middle Aged , Steroid 11-beta-Hydroxylase/genetics , Steroid 11-beta-Hydroxylase/metabolism , Zona Glomerulosa/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...