Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Hypertension ; 79(11): 2552-2564, 2022 11.
Article in English | MEDLINE | ID: mdl-36129175

ABSTRACT

BACKGROUND: Ion channel mutations in calcium regulating genes strongly associate with AngII (angiotensin II)-independent aldosterone production. Here, we used an established mouse model of in vivo aldosterone autonomy, Cyp11b2-driven deletion of TWIK-related acid-sensitive potassium channels (TASK-1 and TASK-3, termed zona glomerulosa [zG]-TASK-loss-of-function), and selective pharmacological TASK channel inhibition to determine whether channel dysfunction in native, electrically excitable zG cell rosette-assemblies: (1) produces spontaneous calcium oscillatory activity and (2) is sufficient to drive substantial aldosterone autonomy. METHODS: We imaged calcium activity in adrenal slices expressing a zG-specific calcium reporter (GCaMP3), an in vitro experimental approach that preserves the native rosette assembly and removes potentially confounding extra-adrenal contributions. In parallel experiments, we measured acute aldosterone production from adrenal slice cultures. RESULTS: Absent from untreated WT slices, we find that either adrenal-specific genetic deletion or acute pharmacological TASK channel inhibition produces spontaneous oscillatory bursting behavior and steroidogenic activity (2.4-fold) that are robust, sustained, and equivalent to activities evoked by 3 nM AngII in WT slices. Moreover, spontaneous activity in zG-TASK-loss-of-function slices and inhibitor-evoked activity in WT slices are unresponsive to AngII regulation over a wide range of concentrations (50 pM to 3 µM). CONCLUSIONS: We provide proof of principle that spontaneous activity of zG cells within classic rosette assemblies evoked solely by a change in an intrinsic, dominant resting-state conductance can be a significant source of AngII-independent aldosterone production from native tissue.


Subject(s)
Aldosterone , Hyperaldosteronism , Mice , Animals , Angiotensin II/pharmacology , Calcium Signaling , Calcium/metabolism , Hyperaldosteronism/genetics , Zona Glomerulosa/metabolism
2.
Mol Cell Endocrinol ; 530: 111287, 2021 06 15.
Article in English | MEDLINE | ID: mdl-33891993

ABSTRACT

How morphology informs function is a fundamental biological question. Here, we review the morphological features of the adrenal zona glomerulosa (zG), highlighting recent cellular and molecular discoveries that govern its formation. The zG consists of glomeruli enwrapped in a Laminin-ß1-enriched basement membrane (BM). Within each glomerulus, zG cells are organized as rosettes, a multicellular structure widely used throughout development to mediate epithelial remodeling, but not often found in healthy adult tissues. Rosettes arise by constriction at a common cellular contact point mediated/facilitated by adherens junctions (AJs). In mice, small, dispersed AJs first appear postnatally and enrich along the entire cell-cell contact around 10 days after birth. Subsequently, these AJ-rich contacts contract, allowing rosettes to form. Concurrently, flat sheet-like domains in the nascent zG, undergo invagination and folding, gradually giving rise to the compact round glomeruli that comprise the adult zG. How these structures impact adrenal function is discussed.


Subject(s)
Zona Glomerulosa/anatomy & histology , Zona Glomerulosa/physiology , Adherens Junctions/metabolism , Animals , Basement Membrane/metabolism , Humans , Laminin/metabolism
3.
Elife ; 102021 01 07.
Article in English | MEDLINE | ID: mdl-33410749

ABSTRACT

Pannexin 1 (Panx1) is a membrane channel implicated in numerous physiological and pathophysiological processes via its ability to support release of ATP and other cellular metabolites for local intercellular signaling. However, to date, there has been no direct demonstration of large molecule permeation via the Panx1 channel itself, and thus the permselectivity of Panx1 for different molecules remains unknown. To address this, we expressed, purified, and reconstituted Panx1 into proteoliposomes and demonstrated that channel activation by caspase cleavage yields a dye-permeable pore that favors flux of anionic, large-molecule permeants (up to ~1 kDa). Large cationic molecules can also permeate the channel, albeit at a much lower rate. We further show that Panx1 channels provide a molecular pathway for flux of ATP and other anionic (glutamate) and cationic signaling metabolites (spermidine). These results verify large molecule permeation directly through caspase-activated Panx1 channels that can support their many physiological roles.


Subject(s)
Adenosine Triphosphate/metabolism , Connexins/genetics , Ion Channels/genetics , Nerve Tissue Proteins/genetics , Signal Transduction , Xenopus Proteins/genetics , Animals , Caspases/metabolism , Connexins/metabolism , Humans , Ion Channels/metabolism , Nerve Tissue Proteins/metabolism , Spodoptera/genetics , Spodoptera/metabolism , Xenopus/genetics , Xenopus/metabolism , Xenopus Proteins/metabolism
4.
Annu Rev Physiol ; 83: 451-475, 2021 02 10.
Article in English | MEDLINE | ID: mdl-33176563

ABSTRACT

Aldosterone excess is a pathogenic factor in many hypertensive disorders. The discovery of numerous somatic and germline mutations in ion channels in primary hyperaldosteronism underscores the importance of plasma membrane conductances in determining the activation state of zona glomerulosa (zG) cells. Electrophysiological recordings describe an electrically quiescent behavior for dispersed zG cells. Yet, emerging data indicate that in native rosette structures in situ, zG cells are electrically excitable, generating slow periodic voltage spikes and coordinated bursts of Ca2+ oscillations. We revisit data to understand how a multitude of conductances may underlie voltage/Ca2+ oscillations, recognizing that zG layer self-renewal and cell heterogeneity may complicate this task. We review recent data to understand rosette architecture and apply maxims derived from computational network modeling to understand rosette function. The challenge going forward is to uncover how the rosette orchestrates the behavior of a functional network of conditional oscillators to control zG layer performance and aldosterone secretion.


Subject(s)
Aldosterone/metabolism , Ion Channels/metabolism , Zona Glomerulosa/metabolism , Zona Glomerulosa/physiology , Animals , Calcium/metabolism , Cell Communication/physiology , Humans
5.
Kidney Int ; 98(3): 630-644, 2020 09.
Article in English | MEDLINE | ID: mdl-32446934

ABSTRACT

Kidney function and blood pressure homeostasis are regulated by purinergic signaling mechanisms. These autocrine/paracrine signaling pathways are initiated by the release of cellular ATP, which influences kidney hemodynamics and steady-state renin secretion from juxtaglomerular cells. However, the mechanism responsible for ATP release that supports tonic inputs to juxtaglomerular cells and regulates renin secretion remains unclear. Pannexin 1 (Panx1) channels localize to both afferent arterioles and juxtaglomerular cells and provide a transmembrane conduit for ATP release and ion permeability in the kidney and the vasculature. We hypothesized that Panx1 channels in renin-expressing cells regulate renin secretion in vivo. Using a renin cell-specific Panx1 knockout model, we found that male Panx1 deficient mice exhibiting a heightened activation of the renin-angiotensin-aldosterone system have markedly increased plasma renin and aldosterone concentrations, and elevated mean arterial pressure with altered peripheral hemodynamics. Following ovariectomy, female mice mirrored the male phenotype. Furthermore, constitutive Panx1 channel activity was observed in As4.1 renin-secreting cells, whereby Panx1 knockdown reduced extracellular ATP accumulation, lowered basal intracellular calcium concentrations and recapitulated a hyper-secretory renin phenotype. Moreover, in response to stress stimuli that lower blood pressure, Panx1-deficient mice exhibited aberrant "renin recruitment" as evidenced by reactivation of renin expression in pre-glomerular arteriolar smooth muscle cells. Thus, renin-cell Panx1 channels suppress renin secretion and influence adaptive renin responses when blood pressure homeostasis is threatened.


Subject(s)
Connexins , Renin , Adenosine Triphosphate , Animals , Blood Pressure , Connexins/genetics , Female , Homeostasis , Male , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics
6.
Nat Commun ; 11(1): 1679, 2020 04 03.
Article in English | MEDLINE | ID: mdl-32245948

ABSTRACT

Aldosterone-producing zona glomerulosa (zG) cells of the adrenal gland arrange in distinct multi-cellular rosettes that provide a structural framework for adrenal cortex morphogenesis and plasticity. Whether this cyto-architecture also plays functional roles in signaling remains unexplored. To determine if structure informs function, we generated mice with zG-specific expression of GCaMP3 and imaged zG cells within their native rosette structure. Here we demonstrate that within the rosette, angiotensin II evokes periodic Cav3-dependent calcium events that form bursts that are stereotypic in form. Our data reveal a critical role for angiotensin II in regulating burst occurrence, and a multifunctional role for the rosette structure in activity-prolongation and coordination. Combined our data define the calcium burst as the fundamental unit of zG layer activity evoked by angiotensin II and highlight a novel role for the rosette as a facilitator of cell communication.


Subject(s)
Aldosterone/metabolism , Angiotensin II/metabolism , Calcium/metabolism , Zona Glomerulosa/metabolism , Animals , Calcium-Binding Proteins/genetics , Cytochrome P-450 CYP11B2/genetics , Cytochrome P-450 CYP11B2/metabolism , Female , Genes, Reporter/genetics , Green Fluorescent Proteins/genetics , Intravital Microscopy , Male , Mice , Mice, Transgenic , Microscopy, Fluorescence , Tissue Culture Techniques
7.
Nat Commun ; 11(1): 1680, 2020 04 03.
Article in English | MEDLINE | ID: mdl-32245949

ABSTRACT

Rosettes are widely used in epithelial morphogenesis during embryonic development and organogenesis. However, their role in postnatal development and adult tissue maintenance remains largely unknown. Here, we show zona glomerulosa cells in the adult adrenal cortex organize into rosettes through adherens junction-mediated constriction, and that rosette formation underlies the maturation of adrenal glomerular structure postnatally. Using genetic mouse models, we show loss of ß-catenin results in disrupted adherens junctions, reduced rosette number, and dysmorphic glomeruli, whereas ß-catenin stabilization leads to increased adherens junction abundance, more rosettes, and glomerular expansion. Furthermore, we uncover numerous known regulators of epithelial morphogenesis enriched in ß-catenin-stabilized adrenals. Among these genes, we show Fgfr2 is required for adrenal rosette formation by regulating adherens junction abundance and aggregation. Together, our data provide an example of rosette-mediated postnatal tissue morphogenesis and a framework for studying the role of rosettes in adult zona glomerulosa tissue maintenance and function.


Subject(s)
Adherens Junctions/metabolism , Morphogenesis , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Zona Glomerulosa/growth & development , beta Catenin/metabolism , Adherens Junctions/genetics , Adherens Junctions/ultrastructure , Adrenal Gland Neoplasms/surgery , Animals , Animals, Newborn , Female , Humans , Male , Mice , Mice, Knockout , Microscopy, Electron, Transmission , Receptor, Fibroblast Growth Factor, Type 2/genetics , Zona Glomerulosa/cytology , Zona Glomerulosa/metabolism , Zona Glomerulosa/ultrastructure , beta Catenin/genetics
8.
Cell Rep ; 31(3): 107524, 2020 04 21.
Article in English | MEDLINE | ID: mdl-32320669

ABSTRACT

Activating mutations in the canonical Wnt/ß-catenin pathway are key drivers of hyperplasia, the gateway for tumor development. In a wide range of tissues, this occurs primarily through enhanced effects on cellular proliferation. Whether additional mechanisms contribute to ß-catenin-driven hyperplasia remains unknown. The adrenal cortex is an ideal system in which to explore this question, as it undergoes hyperplasia following somatic ß-catenin gain-of-function (ßcat-GOF) mutations. Targeting ßcat-GOF to zona Glomerulosa (zG) cells leads to a progressive hyperplastic expansion in the absence of increased proliferation. Instead, we find that hyperplasia results from a functional block in the ability of zG cells to transdifferentiate into zona Fasciculata (zF) cells. Mechanistically, zG cells demonstrate an upregulation of Pde2a, an inhibitor of zF-specific cAMP/PKA signaling. Hyperplasia is further exacerbated by trophic factor stimulation leading to organomegaly. Together, these data indicate that ß-catenin drives adrenal hyperplasia through both proliferation-dependent and -independent mechanisms.


Subject(s)
Adrenal Hyperplasia, Congenital/metabolism , Adrenal Hyperplasia, Congenital/pathology , beta Catenin/metabolism , Adrenal Hyperplasia, Congenital/genetics , Animals , Cell Transdifferentiation/physiology , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , beta Catenin/genetics
9.
Front Cell Neurosci ; 14: 605312, 2020.
Article in English | MEDLINE | ID: mdl-33384586

ABSTRACT

Our previous studies implicated glycosylation of the CaV3.2 isoform of T-type Ca2+ channels (T-channels) in the development of Type 2 painful peripheral diabetic neuropathy (PDN). Here we investigated biophysical mechanisms underlying the modulation of recombinant CaV3.2 channel by de-glycosylation enzymes such as neuraminidase (NEU) and PNGase-F (PNG), as well as their behavioral and biochemical effects in painful PDN Type 1. In our in vitro study we used whole-cell recordings of current-voltage relationships to confirm that CaV3.2 current densities were decreased ~2-fold after de-glycosylation. Furthermore, de-glycosylation induced a significant depolarizing shift in the steady-state relationships for activation and inactivation while producing little effects on the kinetics of current deactivation and recovery from inactivation. PDN was induced in vivo by injections of streptozotocin (STZ) in adult female C57Bl/6j wild type (WT) mice, adult female Sprague Dawley rats and CaV3.2 knock-out (KO mice). Either NEU or vehicle (saline) were locally injected into the right hind paws or intrathecally. We found that injections of NEU, but not vehicle, completely reversed thermal and mechanical hyperalgesia in diabetic WT rats and mice. In contrast, NEU did not alter baseline thermal and mechanical sensitivity in the CaV3.2 KO mice which also failed to develop painful PDN. Finally, we used biochemical methods with gel-shift analysis to directly demonstrate that N-terminal fragments of native CaV3.2 channels in the dorsal root ganglia (DRG) are glycosylated in both healthy and diabetic animals. Our results demonstrate that in sensory neurons glycosylation-induced alterations in CaV3.2 channels in vivo directly enhance diabetic hyperalgesia, and that glycosylation inhibitors can be used to ameliorate painful symptoms in Type 1 diabetes. We expect that our studies may lead to a better understanding of the molecular mechanisms underlying painful PDN in an effort to facilitate the discovery of novel treatments for this intractable disease.

10.
J Endocrinol ; 244(1): 237-247, 2020 01.
Article in English | MEDLINE | ID: mdl-31652415

ABSTRACT

Aldosterone, which plays a key role in the regulation of blood pressure, is produced by zona glomerulosa (ZG) cells of the adrenal cortex. Exaggerated overproduction of aldosterone from ZG cells causes primary hyperaldosteronism. In ZG cells, calcium entry through voltage-gated calcium channels plays a central role in the regulation of aldosterone secretion. Previous studies in animal adrenals and human adrenal adrenocortical cell lines suggest that the T-type but not the L-type calcium channel activity drives aldosterone production. However, recent clinical studies show that somatic mutations in L-type calcium channels are the second most prevalent cause of aldosterone-producing adenoma. Our objective was to define the roles of T and L-type calcium channels in regulating aldosterone secretion from human adrenals. We find that human adrenal ZG cells mainly express T-type CaV3.2/3.3 and L-type CaV1.2/1.3 calcium channels. TTA-P2, a specific inhibitor of T-type calcium channel subtypes, reduced basal aldosterone secretion from acutely prepared slices of human adrenals. Surprisingly, nifedipine, the prototypic inhibitor of L-type calcium channels, also decreased basal aldosterone secretion, suggesting that L-type calcium channels are active under basal conditions. In addition, TTA-P2 or nifedipine also inhibited aldosterone secretion stimulated by angiotensin II- or elevations in extracellular K+. Remarkably, blockade of either L- or T-type calcium channels inhibits basal and stimulated aldosterone production to a similar extent. Low concentrations of TTA-P2 and nifedipine showed additive inhibitory effect on aldosterone secretion. We conclude that T- and L-type calcium channels play equally important roles in controlling aldosterone production from human adrenals.


Subject(s)
Adrenal Glands/metabolism , Aldosterone/biosynthesis , Calcium Channels, L-Type/metabolism , Calcium Channels, T-Type/metabolism , Benzamides/metabolism , Cell Line , Humans , Nifedipine/metabolism , Piperidines/metabolism
11.
Hypertension ; 73(2): 407-414, 2019 02.
Article in English | MEDLINE | ID: mdl-30580687

ABSTRACT

The renin-angiotensin system tightly controls aldosterone synthesis. Dysregulation is evident in hypertension (primary aldosteronism), low renin, and resistant hypertension) but also can exist in normotension. Whether chronic, mild aldosterone autonomy can elicit hypertension remains untested. Previously, we reported that global genetic deletion of 2 pore-domain TWIK-relative acid-sensitive potassium channels, TASK-1 and TASK-3, from mice produces striking aldosterone excess, low renin, and hypertension. Here, we deleted TASK-1 and TASK-3 channels selectively from zona glomerulosa cells and generated a model of mild aldosterone autonomy with attendant hypertension that is aldosterone-driven and Ang II (angiotensin II)-independent. This study shows that a zona glomerulosa-specific channel defect can produce mild autonomous hyperaldosteronism sufficient to cause chronic blood pressure elevation.


Subject(s)
Aldosterone/physiology , Angiotensin II/physiology , Hypertension/etiology , Nerve Tissue Proteins/physiology , Potassium Channels, Tandem Pore Domain/physiology , Potassium Channels/physiology , Zona Glomerulosa/physiology , Animals , Male , Mice , Mice, Knockout , Renin-Angiotensin System/physiology
12.
Nat Biotechnol ; 36(8): 726-737, 2018 09.
Article in English | MEDLINE | ID: mdl-29985477

ABSTRACT

The neurotransmitter acetylcholine (ACh) regulates a diverse array of physiological processes throughout the body. Despite its importance, cholinergic transmission in the majority of tissues and organs remains poorly understood owing primarily to the limitations of available ACh-monitoring techniques. We developed a family of ACh sensors (GACh) based on G-protein-coupled receptors that has the sensitivity, specificity, signal-to-noise ratio, kinetics and photostability suitable for monitoring ACh signals in vitro and in vivo. GACh sensors were validated with transfection, viral and/or transgenic expression in a dozen types of neuronal and non-neuronal cells prepared from multiple animal species. In all preparations, GACh sensors selectively responded to exogenous and/or endogenous ACh with robust fluorescence signals that were captured by epifluorescence, confocal, and/or two-photon microscopy. Moreover, analysis of endogenous ACh release revealed firing-pattern-dependent release and restricted volume transmission, resolving two long-standing questions about central cholinergic transmission. Thus, GACh sensors provide a user-friendly, broadly applicable tool for monitoring cholinergic transmission underlying diverse biological processes.


Subject(s)
Acetylcholine/metabolism , Fluorescent Dyes/chemistry , Receptors, G-Protein-Coupled/genetics , Animals , Animals, Genetically Modified , Brain/cytology , Brain/metabolism , Drosophila/physiology , HEK293 Cells , Humans , In Vitro Techniques , Limit of Detection , Mice , Mutagenesis, Site-Directed , Neurons/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal-To-Noise Ratio
13.
Hypertension ; 70(2): 347-356, 2017 08.
Article in English | MEDLINE | ID: mdl-28630209

ABSTRACT

Ca2+ drives aldosterone synthesis in the cytosolic and mitochondrial compartments of the adrenal zona glomerulosa cell. Membrane potential across each of these compartments regulates the amplitude of the Ca2+ signal; yet, only plasma membrane ion channels and their role in regulating cell membrane potential have garnered investigative attention as pathological causes of human hyperaldosteronism. Previously, we reported that genetic deletion of TASK-3 channels (tandem pore domain acid-sensitive K+ channels) from mice produces aldosterone excess in the absence of a change in the cell membrane potential of zona glomerulosa cells. Here, we report using yeast 2-hybrid, immunoprecipitation, and electron microscopic analyses that TASK-3 channels are resident in mitochondria, where they regulate mitochondrial morphology, mitochondrial membrane potential, and aldosterone production. This study provides proof of principle that mitochondrial K+ channels, by modulating inner mitochondrial membrane morphology and mitochondrial membrane potential, have the ability to play a pathological role in aldosterone dysregulation in steroidogenic cells.


Subject(s)
Aldosterone/biosynthesis , Hyperaldosteronism/metabolism , Mitochondria/physiology , Potassium Channels, Tandem Pore Domain/physiology , Zona Glomerulosa/physiology , Cells, Cultured , Humans , Membrane Potential, Mitochondrial/physiology
14.
PLoS One ; 11(10): e0164132, 2016.
Article in English | MEDLINE | ID: mdl-27736895

ABSTRACT

Despite the substantial burden of hypertension in US minority populations, few genetic studies of blood pressure have been conducted in Hispanics and African Americans, and it is unclear whether many of the established loci identified in European-descent populations contribute to blood pressure variation in non-European descent populations. Using the Metabochip array, we sought to characterize the genetic architecture of previously identified blood pressure loci, and identify novel cardiometabolic variants related to systolic and diastolic blood pressure in a multi-ethnic US population including Hispanics (n = 19,706) and African Americans (n = 18,744). Several known blood pressure loci replicated in African Americans and Hispanics. Fourteen variants in three loci (KCNK3, FGF5, ATXN2-SH2B3) were significantly associated with blood pressure in Hispanics. The most significant diastolic blood pressure variant identified in our analysis, rs2586886/KCNK3 (P = 5.2 x 10-9), also replicated in independent Hispanic and European-descent samples. African American and trans-ethnic meta-analysis data identified novel variants in the FGF5, ULK4 and HOXA-EVX1 loci, which have not been previously associated with blood pressure traits. Our identification and independent replication of variants in KCNK3, a gene implicated in primary hyperaldosteronism, as well as a variant in HOTTIP (HOXA-EVX1) suggest that further work to clarify the roles of these genes may be warranted. Overall, our findings suggest that loci identified in European descent populations also contribute to blood pressure variation in diverse populations including Hispanics and African Americans-populations that are understudied for hypertension genetic risk factors.


Subject(s)
Black or African American/genetics , Blood Pressure/genetics , Genome-Wide Association Study/methods , Hispanic or Latino/genetics , Quantitative Trait Loci , Genetic Variation , Humans , Nerve Tissue Proteins/genetics , Potassium Channels, Tandem Pore Domain/genetics , RNA, Long Noncoding/genetics
15.
Hypertension ; 68(3): 785-95, 2016 09.
Article in English | MEDLINE | ID: mdl-27432863

ABSTRACT

Aldosterone, which plays a key role in maintaining water and electrolyte balance, is produced by zona glomerulosa cells of the adrenal cortex. Autonomous overproduction of aldosterone from zona glomerulosa cells causes primary hyperaldosteronism. Recent clinical studies have highlighted the pathological role of the KCNJ5 potassium channel in primary hyperaldosteronism. Our objective was to determine whether small-conductance Ca(2+)-activated potassium (SK) channels may also regulate aldosterone secretion in human adrenocortical cells. We found that apamin, the prototypic inhibitor of SK channels, decreased membrane voltage, raised intracellular Ca(2+) and dose dependently increased aldosterone secretion from human adrenocortical H295R cells. By contrast, 1-Ethyl-2-benzimidazolinone, an agonist of SK channels, antagonized apamin's action and decreased aldosterone secretion. Commensurate with an increase in aldosterone production, apamin increased mRNA expression of steroidogenic acute regulatory protein and aldosterone synthase that control the early and late rate-limiting steps in aldosterone biosynthesis, respectively. In addition, apamin increased angiotensin II-stimulated aldosterone secretion, whereas 1-Ethyl-2-benzimidazolinone suppressed both angiotensin II- and high K(+)-stimulated production of aldosterone in H295R cells. These findings were supported by apamin-modulation of basal and angiotensin II-stimulated aldosterone secretion from acutely prepared slices of human adrenals. We conclude that SK channel activity negatively regulates aldosterone secretion in human adrenocortical cells. Genetic association studies are necessary to determine whether mutations in SK channel subtype 2 genes may also drive aldosterone excess in primary hyperaldosteronism.


Subject(s)
Adrenal Cortex/cytology , Aldosterone/metabolism , Calcium Channel Agonists/pharmacology , Hyperaldosteronism/physiopathology , Potassium Channels/metabolism , Adrenal Cortex/metabolism , Adult , Aged , Analysis of Variance , Angiotensin II/administration & dosage , Apamin/administration & dosage , Cells, Cultured/drug effects , Female , Humans , Hyperaldosteronism/metabolism , Male , Middle Aged , Potassium Channels/drug effects , RNA, Messenger/metabolism , Sampling Studies
16.
Hypertension ; 68(2): 356-64, 2016 08.
Article in English | MEDLINE | ID: mdl-27296998

ABSTRACT

Blood pressure (BP) is a complex trait that is the consequence of an interaction between genetic and environmental determinants. Previous studies have demonstrated increased BP in mice with global deletion of TASK-1 channels contemporaneous with diverse dysregulation of aldosterone production. In humans, genome-wide association studies in ≈100 000 individuals of European, East Asian, and South Asian ancestry identified a single nucleotide polymorphism (SNP) in KCNK3 (the gene encoding TASK-1) associated with mean arterial pressure. The current study was motivated by the hypotheses that (1) association of KCNK3 SNPs with BP and related traits extends to blacks and Hispanics, and (2) KCNK3 SNPs exhibit associations with plasma renin activity and aldosterone levels. We examined baseline BP measurements for 7840 participants from the Multi-Ethnic Study of Atherosclerosis (MESA), and aldosterone levels and plasma renin activity in a subset of 1653 MESA participants. We identified statistically significant association of the previously reported KCNK3 SNP (rs1275988) with mean arterial pressure in MESA blacks (P=0.024) and a nearby SNP (rs13394970) in MESA Hispanics (P=0.031). We discovered additional KCNK3 SNP associations with systolic BP, mean arterial pressure, and hypertension. We also identified statistically significant association of KCNK3 rs2586886 with plasma aldosterone level in MESA and demonstrated that global deletion of TASK-1 channels in mice produces a mild-hyperaldosteronism, not associated with a decrease in renin. Our results suggest that genetic variation in the KCNK3 gene may contribute to BP variation and less severe hypertensive disorders in which aldosterone may be one of several causative factors.


Subject(s)
Blood Pressure/genetics , Hyperaldosteronism , Hypertension , Nerve Tissue Proteins/genetics , Potassium Channels, Tandem Pore Domain/genetics , Aldosterone/blood , Blood Pressure Determination/methods , California/epidemiology , Ethnicity , Female , Genetic Predisposition to Disease , Humans , Hyperaldosteronism/diagnosis , Hyperaldosteronism/ethnology , Hyperaldosteronism/genetics , Hypertension/diagnosis , Hypertension/ethnology , Hypertension/genetics , Longitudinal Studies , Male , Middle Aged , Polymorphism, Single Nucleotide , Renin/blood , Risk Factors
17.
J Physiol ; 594(20): 5851-5860, 2016 10 15.
Article in English | MEDLINE | ID: mdl-26845064

ABSTRACT

Zona glomerulosa cells (ZG) of the adrenal gland constantly integrate fluctuating ionic, hormonal and paracrine signals to control the synthesis and secretion of aldosterone. These signals modulate Ca2+ levels, which provide the critical second messenger to drive steroid hormone production. Angiotensin II is a hormone known to modulate the activity of voltage-dependent L- and T-type Ca2+ channels that are expressed on the plasma membrane of ZG cells in many species. Because the ZG cell maintains a resting membrane voltage of approximately -85 mV and has been considered electrically silent, low voltage-activated T-type Ca2+ channels are assumed to provide the primary Ca2+ signal that drives aldosterone production. However, this view has recently been challenged by human genetic studies identifying somatic gain-of-function mutations in L-type CaV 1.3 channels in aldosterone-producing adenomas of patients with primary hyperaldosteronism. We provide a review of these assumptions and challenges, and update our understanding of the state of the ZG cell in a layer in which native cellular associations are preserved. This updated view of Ca2+ signalling in ZG cells provides a unifying mechanism that explains how transiently activating CaV 3.2 channels can generate a significant and recurring Ca2+ signal, and how CaV 1.3 channels may contribute to the Ca2+ signal that drives aldosterone production.


Subject(s)
Adrenal Cortex/metabolism , Aldosterone/metabolism , Calcium Channels/metabolism , Zona Glomerulosa/metabolism , Animals , Calcium/metabolism , Humans
18.
Dev Cell ; 26(6): 666-673, 2013 Sep 30.
Article in English | MEDLINE | ID: mdl-24035414

ABSTRACT

Lineage conversion of differentiated cells in response to hormonal feedback has yet to be described. To investigate this, we studied the adrenal cortex, which is composed of functionally distinct concentric layers that develop postnatally, the outer zona glomerulosa (zG) and the inner zona fasciculata (zF). These layers have separate functions, are continuously renewed in response to physiological demands, and are regulated by discrete hormonal feedback loops. Their cellular origin, lineage relationship, and renewal mechanism, however, remain poorly understood. Cell-fate mapping and gene-deletion studies using zG-specific Cre expression demonstrate that differentiated zG cells undergo lineage conversion into zF cells. In addition, zG maintenance is dependent on the master transcriptional regulator Steroidogenic Factor 1 (SF-1), and zG-specific Sf-1 deletion prevents lineage conversion. These findings demonstrate that adrenocortical zonation and regeneration result from lineage conversion and may provide a paradigm for homeostatic cellular renewal in other tissues.


Subject(s)
Cell Lineage , Zona Fasciculata/cytology , Zona Glomerulosa/cytology , Animals , Cell Differentiation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Feedback, Physiological , Gene Deletion , Gene Expression Regulation, Developmental , Homeostasis , Hormones/metabolism , Mice , Steroidogenic Factor 1/genetics , Steroidogenic Factor 1/metabolism , Transcription, Genetic , Zona Fasciculata/growth & development , Zona Fasciculata/metabolism , Zona Glomerulosa/growth & development , Zona Glomerulosa/metabolism
19.
Diabetes ; 62(11): 3828-38, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23835327

ABSTRACT

It has been established that Ca(V)3.2 T-type voltage-gated calcium channels (T-channels) play a key role in the sensitized (hyperexcitable) state of nociceptive sensory neurons (nociceptors) in response to hyperglycemia associated with diabetes, which in turn can be a basis for painful symptoms of peripheral diabetic neuropathy (PDN). Unfortunately, current treatment for painful PDN has been limited by nonspecific systemic drugs with significant side effects or potential for abuse. We studied in vitro and in vivo mechanisms of plasticity of Ca(V)3.2 T-channel in a leptin-deficient (ob/ob) mouse model of PDN. We demonstrate that posttranslational glycosylation of specific extracellular asparagine residues in Ca(V)3.2 channels accelerates current kinetics, increases current density, and augments channel membrane expression. Importantly, deglycosylation treatment with neuraminidase inhibits native T-currents in nociceptors and in so doing completely and selectively reverses hyperalgesia in diabetic ob/ob mice without altering baseline pain responses in healthy mice. Our study describes a new mechanism for the regulation of Ca(V)3.2 activity and suggests that modulating the glycosylation state of T-channels in nociceptors may provide a way to suppress peripheral sensitization. Understanding the details of this regulatory pathway could facilitate the development of novel specific therapies for the treatment of painful PDN.


Subject(s)
Calcium Channels, T-Type/physiology , Diabetic Neuropathies/drug therapy , Glycosylation/drug effects , Hyperalgesia/drug therapy , Neuralgia/drug therapy , Animals , Asparagine/metabolism , Calcium Channels, T-Type/drug effects , Calcium Channels, T-Type/genetics , HEK293 Cells , Humans , Mice , Mice, Obese , Neuraminidase/metabolism , Nociceptors/drug effects , Patch-Clamp Techniques , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/metabolism , Recombinant Proteins , Transfection
20.
J Clin Endocrinol Metab ; 97(11): E2160-7, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22893713

ABSTRACT

CONTEXT: Two potassium (K) channel genes, Kcnk3 and Kcnk9, when deleted in mice, produced a model of hyperaldosteronism and hypertension. OBJECTIVE: Our objective was to explore genetic variation [single-nucleotide polymorphisms (SNP)] in KCNK3 and KCNK9 in relation to blood pressure (BP) and aldosterone production in humans. SUBJECTS AND STUDY DESIGN: Two groups of healthy European Americans (EA) and African Americans (AA) were studied: 1) a longitudinal study group (age ∼14 yr when enrolled, 444 EA and 351 AA) and 2) an inpatient cross-sectional study group (age ∼23 yr, 85 EA and 109 AA). Plasma renin activity, plasma aldosterone concentration, and level of serum K were measured cross-sectionally; BP was measured semiannually in the longitudinal study. SNP were selected to provide coverage of the genes for both EA and AA (15 in KCNK3 and 74 in KCNK9). RESULTS: No associations with KCNK3 were observed. In the longitudinal study, multiple SNP in KCNK9 associated with systolic BP in AA, whereas associations were primarily with aldosterone production in EA. The direction of the changes was the same for aldosterone production and BP, whereas serum K changed in the opposite direction. In the cross-sectional study, associations were observed only in AA. Combining the two studies, one SNP in particular, rs888345, was strongly associated with BP in AA and with indices of aldosterone production in AA and EA. CONCLUSION: Results of an exploratory study suggest that BP and aldosterone production may be affected by variations in KCNK9. The findings could have relevance to risk for hypertension.


Subject(s)
Aldosterone/blood , Blood Pressure/genetics , Nerve Tissue Proteins/genetics , Potassium Channels, Tandem Pore Domain/genetics , Adolescent , Adult , Cross-Sectional Studies , Female , Genetic Association Studies , Humans , Longitudinal Studies , Male , Polymorphism, Single Nucleotide , Potassium/blood , Renin/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...