Comprehensive Physiology Wiley Online Library

Chromaffin Cells of the Adrenal Medulla: Physiology, Pharmacology, and Disease

Full Article on Wiley Online Library



Abstract

Chromaffin cells (CCs) of the adrenal gland and the sympathetic nervous system produce the catecholamines (epinephrine and norepinephrine; EPI and NE) needed to coordinate the bodily “fight‐or‐flight” response to fear, stress, exercise, or conflict. EPI and NE release from CCs is regulated both neurogenically by splanchnic nerve fibers and nonneurogenically by hormones (histamine, corticosteroids, angiotensin, and others) and paracrine messengers [EPI, NE, adenosine triphosphate, opioids, γ‐aminobutyric acid (GABA), etc.]. The “stimulus‐secretion” coupling of CCs is a Ca2+‐dependent process regulated by Ca2+ entry through voltage‐gated Ca2+ channels, Ca2+ pumps, and exchangers and intracellular organelles (RE and mitochondria) and diffusible buffers that provide both Ca2+‐homeostasis and Ca2+‐signaling that ultimately trigger exocytosis. CCs also express Na+ and K+ channels and ionotropic (nAChR and GABAA) and metabotropic receptors (mACh, PACAP, β‐AR, 5‐HT, histamine, angiotensin, and others) that make CCs excitable and responsive to autocrine and paracrine stimuli. To maintain high rates of E/NE secretion during stressful conditions, CCs possess a large number of secretory chromaffin granules (CGs) and members of the soluble NSF‐attachment receptor complex protein family that allow docking, fusion, and exocytosis of CGs at the cell membrane, and their recycling. This article attempts to provide an updated account of well‐established features of the molecular processes regulating CC function, and a survey of the as‐yet‐unsolved but important questions relating to CC function and dysfunction that have been the subject of intense research over the past 15 years. Examples of CCs as a model system to understand the molecular mechanisms associated with neurodegenerative diseases are also provided. Published 2019. Compr Physiol 9:1443‐1502, 2019.

Comprehensive Physiology offers downloadable PowerPoint presentations of figures for non-profit, educational use, provided the content is not modified and full credit is given to the author and publication.

Download a PowerPoint presentation of all images


Figure 1. Figure 1. Different firing modes of spontaneously active chromaffin cells. (A, B) Spontaneous AP trains recorded from two different rat CCs displaying “irregular” and “regular” tonic firings, respectively. (C) Spontaneous AP trains exhibiting slow‐wave bursting recorded from a mouse CC. Below is shown a single burst at an expanded time scale (dashed rectangle) and the overlap of consecutive APs within a burst. Numbers indicate the sequential position in the burst. Adapted, with permission, from Vandael DH, et al., 2015 633.
Figure 2. Figure 2. Time course of Na+, Ca2+, and K+ currents during slow‐wave bursts in mouse CCs. (A) AP‐clamp experiment measuring Kv and BK currents. Top: AP bursts elicited by current steps were recorded at current‐clamp mode and used as voltage command (black trace) in voltage‐clamp experiments. Bottom: Kv currents are shown in red and BK currents in gray. (B, C) As in (A), but the currents isolated were Ca2+ currents (blue), SK (orange), and Na+ (black). (D) Top: BK, SK, and Kv outward current amplitudes versus the spike number of the burst. Bottom: Same, but for the Na+ and Ca2+ inward currents. Adapted, with permission, from Vandael DH, et al., 2015 633.
Figure 3. Figure 3. Low pHo induces burst firing in mouse CCs. Spontaneous firing (no current injection) recorded in mouse CCs at pHo 7.4, 7.0, and 6.6. Bottom: AP recordings on an expanded time scale corresponding to the gray window above. A decrease in pHo results in resting membrane depolarization and the switch of firing modes from tonic (pHo 7.4) to mildly bursting (pHo 7.0), to sustained bursting (pHo 6.6). Intermittent and sustained burst firing are accompanied by a net decrease of AP peak amplitude associated with the slow inactivation of Nav1.3 channels at depolarized potentials. The dotted line indicates the 0‐mV level. Adapted, with permission, from Guarina L, et al., 2017 260.
Figure 4. Figure 4. Spike frequency adaptation during current injections in mouse CCs. (A) Representative current‐clamp recordings from WT mouse CCs in response to 5, 10, or 15 pA current injection from Vh = −70 mV (from top to bottom). (B) Evolution of the instantaneous firing frequency in WT mouse CCs at 15 pA in control (black squares) and in the presence of the SK channel blocker apamin (200 nM; red circles). (C) AP recordings in the presence of 200 nM apamin during 15 pA current injection, to be compared with the control trace to the left. Adapted, with permission, from Vandael DHF, et al., 2012 635.
Figure 5. Figure 5. Voltage‐dependent modulation of CaV2.1 and CaV2.2 in CCs. (A) Acute application of ATP (50 μM) slows down the activation of N‐ and P/Q‐type Ba2+ currents recorded at 0 mV in a bovine CC (for details, see Ref. 98). (B) The delayed activation of N‐ and P/Q‐type Ba2+ currents induced by the acute application of met‐enkephalin (10 μM; black trace; control) is recovered by a 50‐ms prepulse step depolarization to +70 mV (red dot; red trace) (for details, see Ref. 573). (C) Acute application of the soluble vesicle lysate (SVL) containing (EPI, NE, opioids, and ATP) causes N‐ and P/Q‐type current delayed activation in bovine CCs (for details, see Ref. 10). (D, E) Autocrine inhibition of N‐ and P/Q‐type channels revealed by changing cell superfusion from “stop‐flow” to “flow” condition, in bovine CCs. In the “flow” condition, there is no autocrine inhibition. Ba2+ currents at 0 mV are fast activating. In the “stop‐flow” condition, the autocrine‐released material inhibits through GPCRs the opening of CaV2 (N‐ and P/Q‐type) channels, inducing marked activation delay. Adapted, with permission, from Carabelli V, et al., 1998 98. (F) Ba2+ currents recorded from a cell that is part of a cluster undergo robust autocrine modulation (black trace, control). A prepulse to +100 mV is able to rescue the fast channel activation by removing the autocrine inhibition induced by the neurotransmitters released by the surrounding cells (for details, see Ref. 290).
Figure 6. Figure 6. Up‐ and downmodulation of CaV1.2 and CaV1.3 channels by the cAMP/PKA and NO/cGMP/PKG pathways in mouse CCs. The top half illustrates the molecular components and the sequential steps of the β‐AR‐mediated upregulation of CaV1.2 and CaV1.3 L‐type channels through the activation of adenylate cyclase (AC), leading to cAMP production, activation of PKA, and CaV1 channel phosphorylation, causing an increase in open channel probability. The bottom part shows the molecular components and the sequential steps of the NO‐mediated downregulation of CaV1.2 and CaV1.3 L‐type channels through the activation of a soluble guanylate cyclase (sGC), leading to cGMP production, activation of PKG, and CaV1 channels phosphorylation, causing a decrease in open channel probability. NO is produced by membrane NO‐synthases. cGMP is also produced by a membrane guanylate cyclase (mGC). Adapted, with permission, from Mahapatra S, et al., 2012 417.
Figure 7. Figure 7. Synergistic effects of cAMP/PKA and cGMP/PKG pathways on Cav1 currents of WT mouse CCs. The right side is a schematic representation of CaV1 channel α1 subunit with two hypothetical PKA and PKG phosphorylation sites (P). The schemes represent the situation under basal conditions (middle), a synergistic potentiation (top), and a synergistic inhibition (bottom) of CaV1 channel. PKA and PKG P‐sites are partially phosphorylated (P) or unphosphorylated under resting conditions. Phosphorylation and dephosphorylation drove by up‐ and downregulation of PKA and PKG proceed independently of each other and can reach two extreme conditions. In one case, the two PKA P‐sites are dephosphorylated and the two PKG P‐sites are phosphorylated (minimal CaV1 current; cyan trace, on the top left panel). Alternatively, the PKA sites are phosphorylated and PKG P‐sites are dephosphorylated (maximal CaV1 current; cyan trace, on the bottom left panel). The two panels to the left show the time course of synergistic downregulation of CaV1 channels induced by the sequential application of the PKA inhibitor H‐89 and the PKG activator 8‐pCPT‐cGMP (top) and the synergistic upregulation of CaV1 channels by sequential application of the PKA activator forskolin and PKG inhibitor KT 5823 (bottom). Adapted, with permission, from Mahapatra S, et al., 2012 417; Adapted, with permission, from Vandael DHF, et al., 2013 634 (drawing and experimental data).
Figure 8. Figure 8. The concept of functional triads that regulate the generation of local [Ca2+]c transients and exocytosis responses upon stimulation of CCs. (1) After cell depolarization with the physiological neurotransmitter ACh, the voltage‐dependent Ca2+ channels open. (2) Ca2+ enters the cell through a huge electrochemical gradient, giving rise to the formation of a high‐Ca2+ microdomain (HCMD) of about 10 μM or higher near the sub‐plasmalemmal exocytotic sites. (3) High Ca2+ is required to trigger the fast exocytotic release of CAs. (4) The HCMD quickly dissipates initially by mobile Ca2+ buffers (not drawn) and more slowly by Ca2+ uptake by the ER Ca2+‐ATPase (SERCA). (5) The second more relevant pathway for the clearance of the [Ca2+]c transient is the mitochondrial Ca2+ uniporter, which has a low‐affinity high‐capacity for Ca2+ sequestration into the mitochondrial matrix. (6, 7) In both ER and mitochondria (Mito), the matrix [Ca2+]c can reach near half a millimolar. ER Ca2+ can be released back into the cytosol through a CICR mechanism via ryanodine receptors, lnsP3 receptors (6), or through the mitochondrial Na+/Ca2+ exchanger (7). (8) Ca2+ diffusion serves to redistribute Ca2+ at inner areas of the cell core to generate low‐Ca2+ microdomains (LCMD) of around half micromolar that are required for the cytoskeleton‐mediated Ca2+‐dependent vesicle traffic. (9) The Ca2+ levels of the LCMD refills with new vesicles the RRP at sub‐plasmalemmal exocytotic sites, securing that new rounds of exocytosis take place, thus completing the exocytotic process.
Figure 9. Figure 9. Main mechanisms involved in the accumulation of amines and ATP into the CG. A specific vesicular carrier (V‐ATPase) pumps H+ against a concentration and voltage gradient. To reduce the Ψ gradient, Cl channels open, allowing the pH to drop. The pH gradient is also regulated by Na+ (and probably K+) channels. Protons are used for exchange with CAs (dopamine, DA, or NE) or ATP. The synthesis of NE must occur inside the CG by the enzyme dopamine‐β‐hydroxylase (DBH, light blue) using DA as a substrate. In adrenergic cells, NE must leave the granule to be transformed into EPI in the cytosol (by an enzyme called phenylethanolamine‐N‐methyltransferase, PNMT, not shown) Most solutes bind the granule matrix, thus permitting their accumulation by maintaining the isotonicity of CG versus cytosol. Ca2+ is accumulated and released by a combination of H+/Ca2+ exchange, Ca2+ pump, InsP3‐receptors 683, and ryanodine receptors 552. For clarity, Ca2+ turnover is not shown.
Figure 10. Figure 10. Exocytosis and endocytosis of CGs. (A) For clarity, only one SNARE complex is shown, while some of the accessory proteins are omitted: (1) granule proteins are sorted and packaged into the Golgi apparatus; (2) granules are transported to the release sites by tubulin (not shown); (3) actin filaments drive granules to specific tethering points; (4) “granule docking,” the SNARE complex starts to organize and the granule docks to the plasmalemma; (5) priming, mediated by Munc‐18 and complexin, thus allowing a tighter interaction of SNARE complexes. Munc13 acts on syntaxin changing its conformation, thereby leading to SNARE proteins zippering and the formation of fusion pore; (6) the initial fusion pore allows a limited exchange of water and solutes; (7) fusion pore dilatation allowing the partial or complete release of the granule content; depending on the extent of dilatation and duration of the Ω state, more or fewer solutes will be released. The SNARE complex is disassembled; (8) cavicapture. This partial exocytosis occurs when a dilated, but reversible, fusion pore allows the partial release of small molecules like peptides. Whether these granules can go to the SRP or get exocytosed again is unknown; (9) membrane recovery by endocytosis. Two major mechanisms might be involved: depending on clathrin and dynamin 2 or clathrin‐independent that uses dynamin 1 to promote granule fission. The inner content of the granule rapidly acidifies; (10) clathrin is disassembled and the granule can travel deeper inside the cell toward either endosomes or lysosomes from where they can reenter in the secretory cycle after sorting in the Golgi (11). Note that sizes of the resulted endocytotic granules are now smaller, and granule matrices are clearer as a result of protein loss during exocytosis. Note also that some of the steps are reversible (blue double head arrows). (B) Organization of the SNARE complex in the priming state. The presence of cholesterol and lysophospholipids allows the curvature of the cell membrane. Note the lateral disposition of coiled‐coil of proteins and the proposed situation of Munc‐18 and complexin.
Figure 11. Figure 11. Different pools of granules are involved in the secretory responses. (A) Even in the continuous presence of Ach, the secretion of CA from perfused cat adrenals progressively decays (gray bars). Cumulative CA secretion is superimposed (blue trace). Modified, with permission, from Douglas WW, and Rubin RP, 1961 176. (B) Although the time courses are different, a similar situation occurs when secretion is elicited by rising intracellular Ca2+ in permeabilized CCs (leaky cell). Modified, with permission, from Baker PF, and Knight DE, 1981 44. (C) Recordings of whole‐cell capacitance from mouse CCs evidence the presence of at least two exocytotic kinetics: rapid (RRP) and slow (SRP). Authors tested the effect of rising intracellular Ca2+ by flash photolysis of NP‐EGTA (taken from Figure 2A in Ref. 591). Notice the different time course compared with panels A and B. (D) Newly produced granules are the first to be released. Perfused CCs are stimulated with a nicotinic agonist and CA secretion continuously recording by an electrochemical detector. The analysis of the perfusate shows that EGFP (labeling neuropeptide Y) is released only during the first pulses. Modified, with permission, from Estevez‐Herrera J, et al., 2016 214.
Figure 12. Figure 12. Intracellular signaling pathways and physiology of the chromaffin cell. Acetylcholine and PACAP (sympathetic preganglionic) release from splanchnic nerve activates the nicotinic (AChR) and PACAPergic (PAC1R) receptors, triggering catecholamine and neuropeptide secretion from CG, and increased transcription of genes encoding biosynthetic enzymes for catecholamines (TH, PNMA), and prohormones from which processing to mature neuropeptides occurs, and stimulus‐secretion‐synthesis coupling in the chromaffin cell. PAC1R activation promotes Gs coupling to adenylate cyclase (AC), the elevation of cyclic AMP (cAMP), and the activation of three separate cAMP effectors. Protein kinase A (PKA) mediates CREB‐dependent gene transcription; Epac mediates Rap‐dependent activation of the MAP kinase p38, leading to activation of transcription factors including AP1; NCS‐Rapgef2 mediates Rap‐dependent activation of the mixed‐function kinase B‐Raf, allowing MEK upregulation of the MAP kinase ERK (extracellularly regulated kinase) and gene activation through a combination of transcription factors. Stimulus‐secretion‐synthesis coupling also involves on the intercellular level increased expression of connexins that form gap junctions, which help to amplify chromaffin cell secretion from the adrenal gland as a whole. There is as yet no direct proof for PACAP or acetylcholine as the principal mediator of what might be termed the “gap‐junction response.” IFN, TNF, IL‐1, and IL‐6 effects on cognate receptors on chromaffin cells and cellular sequelae in the chromaffin cell are not shown, but synergize with PACAP stimulation to modulate both catecholamine and peptide secretion and chromaffin cell gene transcription. Splanchnic nerve input affords synergistic as well as antagonistic interactions between cytokines and PACAP under physiological conditions in which both stress and inflammation may play a role. Finally, secretory products of the chromaffin cell itself, including substance P, other neuropeptides, and chromogranin‐derived peptides such as catestatin, modulate chromaffin cell secretion via paracrine actions, in part through modulation of AChR function. See text, and references in text, for further details.
Figure 13. Figure 13. The adrenal medulla as a stress transducer and neuroimmunoinflammatory and cardiovascular regulator. Lower figure. a. The “final common pathway” for stress responding in the CNS is the activation of neurons in the paraventricular nucleus (PVN) of the hypothalamus (Hy), which projects both to the cell bodies of sympathetic preganglionic neurons in the intermediolateral column of the spinal cord and to the median eminence: a′. for ACTH release from the pituitary (Pit) to stimulate corticosterone/cortisol (CORT) release from adrenal cortex; b′. cell bodies in the intermediolateral column of the spinal cord innervate the CCs of the adrenal medulla via the splanchnic nerve, and sympathetic postganglionic nerve targets via para‐ and prevertebral postganglionic sympathetic neurons b. via the splanchnic nerve, releasing ACh basally, and ACh and PACAP during stress, with activation of both secretion by ACh and PACAP, and of CC signaling pathways by PACAP increasing expression of genes encoding neuropeptides (NPs), additional mediators, catecholamine biosynthetic enzymes, and adhesion factors and connexins that increase cell‐cell communication among CCs and amplify CA, neuropeptide, and chromogranin output in response to stress; c. NP (neuropeptide) release from CCs has autocrine effects on CA secretion from CCs themselves (e.g. catestatin, substance P, and others), activation of sensory neurons (e.g. BAM22P, acting on specific receptors expressed in sensory nerves), and modulation of CORT secretion from the adrenal cortex (galanin, VIP, and other peptides), as well as hormonal effects on distant organs; d. CA release from CCs into the general circulation, and affecting metabolism, heart rate, blood pressure, and immune cell mobilization; e. Cytokines released as blood‐borne molecules or locally from circulating monocyte macrophages act as inhibitors of CORT secretion in the adrenal cortex, and as modulators of peptide secretion in adrenal cortex via receptors on CCs themselves; f. Sensory inputs to adrenal medulla sense CC secretory activity via release of BAM‐22P, for which sensory neurons express specific receptors. Also depicted are the targets of glucocorticoid (CORT) release from the adrenal cortex at the pituitary g and immune system g′, the latter decreasing in turn cytokine secretion, which affects adrenomedullary function during stress. For a further explication of the figure, see the text.
Figure 14. Figure 14. Divergent responses of rat CCs to GABAA receptor activation. (A) GABA response is excitatory in immature neurons because of a greater functionality of NKCC1. (B) In mature neurons, GABA response is inhibitory because of the dominant activity of the KCC2. (C) GABAA‐Rs‐mediated response is depolarizing with [Ca2+]i elevation in ∼44% of rat adrenal CCs. (D) in ∼26% of CCs, GABA response is hyperpolarizing and causes [Ca2+]i drop. The scheme also represents the anion‐exchanger AE3 (pendrin), which accumulates Cl in exchange for intracellular HCO3 and participates in Cl transport into CCs as a replacement for NKCC1. VDDC: voltage‐dependent Ca2+ channel.
Figure 15. Figure 15. Ryanodine effects on depolarization‐induced CA secretion. (A, B) Burst of amperometric spikes elicited by a 5‐s‐long depolarizing pulse in a WKY (A) and an SHR CCs (B) before (top) and after (bottom) incubation with 10 μM ryanodine. (C, D) Mean cumulative charge in WKY and SHR CCs, respectively, before and after ryanodine treatment. The cumulative charge after RyR blockade was not significantly affected in WKY CCs (20.8 vs. 18.8 pC; p = 0.274), but it was drastically reduced in SHR CCs (from 43.9 to 10.1 pC; p = 0.0001). The number of cells examined is shown in parentheses. Reused, with permission, from Segura‐Chama P, et al., 2015 566.
Figure 16. Figure 16. Cell signaling pathways leading to CACNA1H gene expression and Cav3.2 channels recruitment during chronic or intermittent hypoxia. Schematic pathway of the activation of transcription factors (HIF, CREB, etc.) and CACNA1H gene expression through a NOX, ROS, PLC, and PKC cascade leading to Cav3.2 channels recruitment during chronic/intermittent hypoxia. Adapted, with permission, from Mahapatra S, et al., 2012 417.


Figure 1. Different firing modes of spontaneously active chromaffin cells. (A, B) Spontaneous AP trains recorded from two different rat CCs displaying “irregular” and “regular” tonic firings, respectively. (C) Spontaneous AP trains exhibiting slow‐wave bursting recorded from a mouse CC. Below is shown a single burst at an expanded time scale (dashed rectangle) and the overlap of consecutive APs within a burst. Numbers indicate the sequential position in the burst. Adapted, with permission, from Vandael DH, et al., 2015 633.


Figure 2. Time course of Na+, Ca2+, and K+ currents during slow‐wave bursts in mouse CCs. (A) AP‐clamp experiment measuring Kv and BK currents. Top: AP bursts elicited by current steps were recorded at current‐clamp mode and used as voltage command (black trace) in voltage‐clamp experiments. Bottom: Kv currents are shown in red and BK currents in gray. (B, C) As in (A), but the currents isolated were Ca2+ currents (blue), SK (orange), and Na+ (black). (D) Top: BK, SK, and Kv outward current amplitudes versus the spike number of the burst. Bottom: Same, but for the Na+ and Ca2+ inward currents. Adapted, with permission, from Vandael DH, et al., 2015 633.


Figure 3. Low pHo induces burst firing in mouse CCs. Spontaneous firing (no current injection) recorded in mouse CCs at pHo 7.4, 7.0, and 6.6. Bottom: AP recordings on an expanded time scale corresponding to the gray window above. A decrease in pHo results in resting membrane depolarization and the switch of firing modes from tonic (pHo 7.4) to mildly bursting (pHo 7.0), to sustained bursting (pHo 6.6). Intermittent and sustained burst firing are accompanied by a net decrease of AP peak amplitude associated with the slow inactivation of Nav1.3 channels at depolarized potentials. The dotted line indicates the 0‐mV level. Adapted, with permission, from Guarina L, et al., 2017 260.


Figure 4. Spike frequency adaptation during current injections in mouse CCs. (A) Representative current‐clamp recordings from WT mouse CCs in response to 5, 10, or 15 pA current injection from Vh = −70 mV (from top to bottom). (B) Evolution of the instantaneous firing frequency in WT mouse CCs at 15 pA in control (black squares) and in the presence of the SK channel blocker apamin (200 nM; red circles). (C) AP recordings in the presence of 200 nM apamin during 15 pA current injection, to be compared with the control trace to the left. Adapted, with permission, from Vandael DHF, et al., 2012 635.


Figure 5. Voltage‐dependent modulation of CaV2.1 and CaV2.2 in CCs. (A) Acute application of ATP (50 μM) slows down the activation of N‐ and P/Q‐type Ba2+ currents recorded at 0 mV in a bovine CC (for details, see Ref. 98). (B) The delayed activation of N‐ and P/Q‐type Ba2+ currents induced by the acute application of met‐enkephalin (10 μM; black trace; control) is recovered by a 50‐ms prepulse step depolarization to +70 mV (red dot; red trace) (for details, see Ref. 573). (C) Acute application of the soluble vesicle lysate (SVL) containing (EPI, NE, opioids, and ATP) causes N‐ and P/Q‐type current delayed activation in bovine CCs (for details, see Ref. 10). (D, E) Autocrine inhibition of N‐ and P/Q‐type channels revealed by changing cell superfusion from “stop‐flow” to “flow” condition, in bovine CCs. In the “flow” condition, there is no autocrine inhibition. Ba2+ currents at 0 mV are fast activating. In the “stop‐flow” condition, the autocrine‐released material inhibits through GPCRs the opening of CaV2 (N‐ and P/Q‐type) channels, inducing marked activation delay. Adapted, with permission, from Carabelli V, et al., 1998 98. (F) Ba2+ currents recorded from a cell that is part of a cluster undergo robust autocrine modulation (black trace, control). A prepulse to +100 mV is able to rescue the fast channel activation by removing the autocrine inhibition induced by the neurotransmitters released by the surrounding cells (for details, see Ref. 290).


Figure 6. Up‐ and downmodulation of CaV1.2 and CaV1.3 channels by the cAMP/PKA and NO/cGMP/PKG pathways in mouse CCs. The top half illustrates the molecular components and the sequential steps of the β‐AR‐mediated upregulation of CaV1.2 and CaV1.3 L‐type channels through the activation of adenylate cyclase (AC), leading to cAMP production, activation of PKA, and CaV1 channel phosphorylation, causing an increase in open channel probability. The bottom part shows the molecular components and the sequential steps of the NO‐mediated downregulation of CaV1.2 and CaV1.3 L‐type channels through the activation of a soluble guanylate cyclase (sGC), leading to cGMP production, activation of PKG, and CaV1 channels phosphorylation, causing a decrease in open channel probability. NO is produced by membrane NO‐synthases. cGMP is also produced by a membrane guanylate cyclase (mGC). Adapted, with permission, from Mahapatra S, et al., 2012 417.


Figure 7. Synergistic effects of cAMP/PKA and cGMP/PKG pathways on Cav1 currents of WT mouse CCs. The right side is a schematic representation of CaV1 channel α1 subunit with two hypothetical PKA and PKG phosphorylation sites (P). The schemes represent the situation under basal conditions (middle), a synergistic potentiation (top), and a synergistic inhibition (bottom) of CaV1 channel. PKA and PKG P‐sites are partially phosphorylated (P) or unphosphorylated under resting conditions. Phosphorylation and dephosphorylation drove by up‐ and downregulation of PKA and PKG proceed independently of each other and can reach two extreme conditions. In one case, the two PKA P‐sites are dephosphorylated and the two PKG P‐sites are phosphorylated (minimal CaV1 current; cyan trace, on the top left panel). Alternatively, the PKA sites are phosphorylated and PKG P‐sites are dephosphorylated (maximal CaV1 current; cyan trace, on the bottom left panel). The two panels to the left show the time course of synergistic downregulation of CaV1 channels induced by the sequential application of the PKA inhibitor H‐89 and the PKG activator 8‐pCPT‐cGMP (top) and the synergistic upregulation of CaV1 channels by sequential application of the PKA activator forskolin and PKG inhibitor KT 5823 (bottom). Adapted, with permission, from Mahapatra S, et al., 2012 417; Adapted, with permission, from Vandael DHF, et al., 2013 634 (drawing and experimental data).


Figure 8. The concept of functional triads that regulate the generation of local [Ca2+]c transients and exocytosis responses upon stimulation of CCs. (1) After cell depolarization with the physiological neurotransmitter ACh, the voltage‐dependent Ca2+ channels open. (2) Ca2+ enters the cell through a huge electrochemical gradient, giving rise to the formation of a high‐Ca2+ microdomain (HCMD) of about 10 μM or higher near the sub‐plasmalemmal exocytotic sites. (3) High Ca2+ is required to trigger the fast exocytotic release of CAs. (4) The HCMD quickly dissipates initially by mobile Ca2+ buffers (not drawn) and more slowly by Ca2+ uptake by the ER Ca2+‐ATPase (SERCA). (5) The second more relevant pathway for the clearance of the [Ca2+]c transient is the mitochondrial Ca2+ uniporter, which has a low‐affinity high‐capacity for Ca2+ sequestration into the mitochondrial matrix. (6, 7) In both ER and mitochondria (Mito), the matrix [Ca2+]c can reach near half a millimolar. ER Ca2+ can be released back into the cytosol through a CICR mechanism via ryanodine receptors, lnsP3 receptors (6), or through the mitochondrial Na+/Ca2+ exchanger (7). (8) Ca2+ diffusion serves to redistribute Ca2+ at inner areas of the cell core to generate low‐Ca2+ microdomains (LCMD) of around half micromolar that are required for the cytoskeleton‐mediated Ca2+‐dependent vesicle traffic. (9) The Ca2+ levels of the LCMD refills with new vesicles the RRP at sub‐plasmalemmal exocytotic sites, securing that new rounds of exocytosis take place, thus completing the exocytotic process.


Figure 9. Main mechanisms involved in the accumulation of amines and ATP into the CG. A specific vesicular carrier (V‐ATPase) pumps H+ against a concentration and voltage gradient. To reduce the Ψ gradient, Cl channels open, allowing the pH to drop. The pH gradient is also regulated by Na+ (and probably K+) channels. Protons are used for exchange with CAs (dopamine, DA, or NE) or ATP. The synthesis of NE must occur inside the CG by the enzyme dopamine‐β‐hydroxylase (DBH, light blue) using DA as a substrate. In adrenergic cells, NE must leave the granule to be transformed into EPI in the cytosol (by an enzyme called phenylethanolamine‐N‐methyltransferase, PNMT, not shown) Most solutes bind the granule matrix, thus permitting their accumulation by maintaining the isotonicity of CG versus cytosol. Ca2+ is accumulated and released by a combination of H+/Ca2+ exchange, Ca2+ pump, InsP3‐receptors 683, and ryanodine receptors 552. For clarity, Ca2+ turnover is not shown.


Figure 10. Exocytosis and endocytosis of CGs. (A) For clarity, only one SNARE complex is shown, while some of the accessory proteins are omitted: (1) granule proteins are sorted and packaged into the Golgi apparatus; (2) granules are transported to the release sites by tubulin (not shown); (3) actin filaments drive granules to specific tethering points; (4) “granule docking,” the SNARE complex starts to organize and the granule docks to the plasmalemma; (5) priming, mediated by Munc‐18 and complexin, thus allowing a tighter interaction of SNARE complexes. Munc13 acts on syntaxin changing its conformation, thereby leading to SNARE proteins zippering and the formation of fusion pore; (6) the initial fusion pore allows a limited exchange of water and solutes; (7) fusion pore dilatation allowing the partial or complete release of the granule content; depending on the extent of dilatation and duration of the Ω state, more or fewer solutes will be released. The SNARE complex is disassembled; (8) cavicapture. This partial exocytosis occurs when a dilated, but reversible, fusion pore allows the partial release of small molecules like peptides. Whether these granules can go to the SRP or get exocytosed again is unknown; (9) membrane recovery by endocytosis. Two major mechanisms might be involved: depending on clathrin and dynamin 2 or clathrin‐independent that uses dynamin 1 to promote granule fission. The inner content of the granule rapidly acidifies; (10) clathrin is disassembled and the granule can travel deeper inside the cell toward either endosomes or lysosomes from where they can reenter in the secretory cycle after sorting in the Golgi (11). Note that sizes of the resulted endocytotic granules are now smaller, and granule matrices are clearer as a result of protein loss during exocytosis. Note also that some of the steps are reversible (blue double head arrows). (B) Organization of the SNARE complex in the priming state. The presence of cholesterol and lysophospholipids allows the curvature of the cell membrane. Note the lateral disposition of coiled‐coil of proteins and the proposed situation of Munc‐18 and complexin.


Figure 11. Different pools of granules are involved in the secretory responses. (A) Even in the continuous presence of Ach, the secretion of CA from perfused cat adrenals progressively decays (gray bars). Cumulative CA secretion is superimposed (blue trace). Modified, with permission, from Douglas WW, and Rubin RP, 1961 176. (B) Although the time courses are different, a similar situation occurs when secretion is elicited by rising intracellular Ca2+ in permeabilized CCs (leaky cell). Modified, with permission, from Baker PF, and Knight DE, 1981 44. (C) Recordings of whole‐cell capacitance from mouse CCs evidence the presence of at least two exocytotic kinetics: rapid (RRP) and slow (SRP). Authors tested the effect of rising intracellular Ca2+ by flash photolysis of NP‐EGTA (taken from Figure 2A in Ref. 591). Notice the different time course compared with panels A and B. (D) Newly produced granules are the first to be released. Perfused CCs are stimulated with a nicotinic agonist and CA secretion continuously recording by an electrochemical detector. The analysis of the perfusate shows that EGFP (labeling neuropeptide Y) is released only during the first pulses. Modified, with permission, from Estevez‐Herrera J, et al., 2016 214.


Figure 12. Intracellular signaling pathways and physiology of the chromaffin cell. Acetylcholine and PACAP (sympathetic preganglionic) release from splanchnic nerve activates the nicotinic (AChR) and PACAPergic (PAC1R) receptors, triggering catecholamine and neuropeptide secretion from CG, and increased transcription of genes encoding biosynthetic enzymes for catecholamines (TH, PNMA), and prohormones from which processing to mature neuropeptides occurs, and stimulus‐secretion‐synthesis coupling in the chromaffin cell. PAC1R activation promotes Gs coupling to adenylate cyclase (AC), the elevation of cyclic AMP (cAMP), and the activation of three separate cAMP effectors. Protein kinase A (PKA) mediates CREB‐dependent gene transcription; Epac mediates Rap‐dependent activation of the MAP kinase p38, leading to activation of transcription factors including AP1; NCS‐Rapgef2 mediates Rap‐dependent activation of the mixed‐function kinase B‐Raf, allowing MEK upregulation of the MAP kinase ERK (extracellularly regulated kinase) and gene activation through a combination of transcription factors. Stimulus‐secretion‐synthesis coupling also involves on the intercellular level increased expression of connexins that form gap junctions, which help to amplify chromaffin cell secretion from the adrenal gland as a whole. There is as yet no direct proof for PACAP or acetylcholine as the principal mediator of what might be termed the “gap‐junction response.” IFN, TNF, IL‐1, and IL‐6 effects on cognate receptors on chromaffin cells and cellular sequelae in the chromaffin cell are not shown, but synergize with PACAP stimulation to modulate both catecholamine and peptide secretion and chromaffin cell gene transcription. Splanchnic nerve input affords synergistic as well as antagonistic interactions between cytokines and PACAP under physiological conditions in which both stress and inflammation may play a role. Finally, secretory products of the chromaffin cell itself, including substance P, other neuropeptides, and chromogranin‐derived peptides such as catestatin, modulate chromaffin cell secretion via paracrine actions, in part through modulation of AChR function. See text, and references in text, for further details.


Figure 13. The adrenal medulla as a stress transducer and neuroimmunoinflammatory and cardiovascular regulator. Lower figure. a. The “final common pathway” for stress responding in the CNS is the activation of neurons in the paraventricular nucleus (PVN) of the hypothalamus (Hy), which projects both to the cell bodies of sympathetic preganglionic neurons in the intermediolateral column of the spinal cord and to the median eminence: a′. for ACTH release from the pituitary (Pit) to stimulate corticosterone/cortisol (CORT) release from adrenal cortex; b′. cell bodies in the intermediolateral column of the spinal cord innervate the CCs of the adrenal medulla via the splanchnic nerve, and sympathetic postganglionic nerve targets via para‐ and prevertebral postganglionic sympathetic neurons b. via the splanchnic nerve, releasing ACh basally, and ACh and PACAP during stress, with activation of both secretion by ACh and PACAP, and of CC signaling pathways by PACAP increasing expression of genes encoding neuropeptides (NPs), additional mediators, catecholamine biosynthetic enzymes, and adhesion factors and connexins that increase cell‐cell communication among CCs and amplify CA, neuropeptide, and chromogranin output in response to stress; c. NP (neuropeptide) release from CCs has autocrine effects on CA secretion from CCs themselves (e.g. catestatin, substance P, and others), activation of sensory neurons (e.g. BAM22P, acting on specific receptors expressed in sensory nerves), and modulation of CORT secretion from the adrenal cortex (galanin, VIP, and other peptides), as well as hormonal effects on distant organs; d. CA release from CCs into the general circulation, and affecting metabolism, heart rate, blood pressure, and immune cell mobilization; e. Cytokines released as blood‐borne molecules or locally from circulating monocyte macrophages act as inhibitors of CORT secretion in the adrenal cortex, and as modulators of peptide secretion in adrenal cortex via receptors on CCs themselves; f. Sensory inputs to adrenal medulla sense CC secretory activity via release of BAM‐22P, for which sensory neurons express specific receptors. Also depicted are the targets of glucocorticoid (CORT) release from the adrenal cortex at the pituitary g and immune system g′, the latter decreasing in turn cytokine secretion, which affects adrenomedullary function during stress. For a further explication of the figure, see the text.


Figure 14. Divergent responses of rat CCs to GABAA receptor activation. (A) GABA response is excitatory in immature neurons because of a greater functionality of NKCC1. (B) In mature neurons, GABA response is inhibitory because of the dominant activity of the KCC2. (C) GABAA‐Rs‐mediated response is depolarizing with [Ca2+]i elevation in ∼44% of rat adrenal CCs. (D) in ∼26% of CCs, GABA response is hyperpolarizing and causes [Ca2+]i drop. The scheme also represents the anion‐exchanger AE3 (pendrin), which accumulates Cl in exchange for intracellular HCO3 and participates in Cl transport into CCs as a replacement for NKCC1. VDDC: voltage‐dependent Ca2+ channel.


Figure 15. Ryanodine effects on depolarization‐induced CA secretion. (A, B) Burst of amperometric spikes elicited by a 5‐s‐long depolarizing pulse in a WKY (A) and an SHR CCs (B) before (top) and after (bottom) incubation with 10 μM ryanodine. (C, D) Mean cumulative charge in WKY and SHR CCs, respectively, before and after ryanodine treatment. The cumulative charge after RyR blockade was not significantly affected in WKY CCs (20.8 vs. 18.8 pC; p = 0.274), but it was drastically reduced in SHR CCs (from 43.9 to 10.1 pC; p = 0.0001). The number of cells examined is shown in parentheses. Reused, with permission, from Segura‐Chama P, et al., 2015 566.


Figure 16. Cell signaling pathways leading to CACNA1H gene expression and Cav3.2 channels recruitment during chronic or intermittent hypoxia. Schematic pathway of the activation of transcription factors (HIF, CREB, etc.) and CACNA1H gene expression through a NOX, ROS, PLC, and PKC cascade leading to Cav3.2 channels recruitment during chronic/intermittent hypoxia. Adapted, with permission, from Mahapatra S, et al., 2012 417.
References
 1. Adams MB , Simonetta G , McMillen IC . The non‐neurogenic catecholamine response of the fetal adrenal to hypoxia is dependent on activation of voltage sensitive Ca2+ channels. Brain Res Dev Brain Res 94: 182‐189, 1996.
 2. Adrian ED , Bronk DW , Phillips G . Discharges in mammalian sympathetic nerves. J Physiol 74: 115‐133, 1932.
 3. Affolter H‐U , Giraud P , Hotchkiss AJ , Eiden LE . Stimulus‐secretion‐synthesis coupling: A model for cholinergic regulation of enkephalin secretion and gene transcription in adrenomedullary chromaffin cells. In: Fraioli F , editor. Opiate Peptides in the Periphery. Amsterdam: Elsevier, 1984, p. 23‐30.
 4. Ait‐Ali D , Turquier V , Grumolato L , Yon L , Jourdain M , Alexandre D , Eiden LE , Vaudry H , Anouar Y . The proinflammatory cytokines tumor necrosis factor‐alpha and interleukin‐1 stimulate neuropeptide gene transcription and secretion in adrenochromaffin cells via activation of extracellularly regulated kinase 1/2 and p38 protein kinases, and activator protein‐1 transcription factors. Mol Endocrinol 18: 1721‐1739, 2004.
 5. Ait‐Ali D , Turquier V , Tanguy Y , Thouennon E , Ghzili H , Mounien L , Derambure C , Jegou S , Salier JP , Vaudry H , Eiden LE , Anouar Y . Tumor necrosis factor (TNF)‐alpha persistently activates nuclear factor‐kappaB signaling through the type 2 TNF receptor in chromaffin cells: Implications for long‐term regulation of neuropeptide gene expression in inflammation. Endocrinology 149: 2840‐2852, 2008.
 6. Akiyama T , Yamazaki T , Mori H , Sunagawa K . Effects of Ca2+ channel antagonists on acetylcholine and catecholamine releases in the in vivo rat adrenal medulla. Am J Physiol Regul Integr Comp Physiol 287: R161‐R166, 2004.
 7. Albillos A , Artalejo AR , Lopez MG , Gandia L , Garcia AG , Carbone E . Calcium channel subtypes in cat chromaffin cells. J Physiol 477 (Pt 2): 197‐213, 1994.
 8. Albillos A , Carbone E , Gandia L , Garcia AG , Pollo A . Opioid inhibition of Ca2+ channel subtypes in bovine chromaffin cells: Selectivity of action and voltage dependence. Eur J Neurosci 8: 1561‐1570, 1996.
 9. Albillos A , Dernick G , Horstmann H , Almers W , Alvarez de Toledo G , Lindau M . The exocytotic event in chromaffin cells revealed by patch amperometry. Nature 389: 509‐512, 1997.
 10. Albillos A , Gandia L , Michelena P , Gilabert JA , delValle M , Carbone E , Garcia AG . The mechanism of calcium channel facilitation in bovine chromaffin cells. J Physiol 494: 687‐695, 1996.
 11. Albillos A , Neher E , Moser T . R‐Type Ca2+ channels are coupled to the rapid component of secretion in mouse adrenal slice chromaffin cells. J Neurosci 20: 8323‐8330, 2000.
 12. Albinana E , Segura‐Chama P , Baraibar AM , Hernandez‐Cruz A , Hernandez‐Guijo JM . Different contributions of calcium channel subtypes to electrical excitability of chromaffin cells in rat adrenal slices. J Neurochem 133: 511‐521, 2015.
 13. Alejandre‐Garcia T , Pena‐Del Castillo JG , Hernandez‐Cruz A . GABAA receptor: A unique modulator of excitability, Ca(2+) signaling, and catecholamine release of rat chromaffin cells. Pflug Archiv: Eur J Physiol 470: 67‐77, 2018.
 14. Alejandre‐Garcia T , Segura‐Chama P , Perez‐Armendariz EM , Delgado‐Lezama R , Hernandez‐Cruz A . Erratum to: Modulation of spontaneous intracellular Ca(2+) fluctuations and spontaneous cholinergic transmission in rat chromaffin cells in situ by endogenous GABA acting on GABAA receptors. Pflug Archiv: Eur J Physiol 469: 1413, 2017.
 15. Ales E , Tabares L , Poyato JM , Valero V , Lindau M , Alvarez de Toledo G . High calcium concentrations shift the mode of exocytosis to the kiss‐and‐run mechanism. Nat Cell Biol 1: 40‐44, 1999.
 16. Aloe L , Levi‐Montalcini R . Nerve growth factor‐induced transformation of immature chromaffin cells in vivo into sympathetic neurons: Effect of antiserum to nerve growth factor. Proc Natl Acad Sci USA 76: 1246‐1250, 1979.
 17. Alonso MT , Barrero MJ , Michelena P , Carnicero E , Cuchillo I , Garcia AG , Garcia‐Sancho J , Montero M , Alvarez J . Ca2+‐induced Ca2+ release in chromaffin cells seen from inside the ER with targeted aequorin. J Cell Biol 144: 241‐254, 1999.
 18. Alvarez de Toledo G , Fernandez‐Chacon R , Fernandez JM . Release of secretory products during transient vesicle fusion. Nature 363: 554‐558, 1993.
 19. Alvarez de Toledo G , Montes MA , Montenegro P , Borges R . Phases of the exocytotic fusion pore. FEBS Lett 592: 3532‐3541, 2018.
 20. Alvarez YD , Ibanez LI , Uchitel OD , Marengo FD . P/Q Ca2+ channels are functionally coupled to exocytosis of the immediately releasable pool in mouse chromaffin cells. Cell Calcium 43: 155‐164, 2008.
 21. Amar L , Servais A , Gimenez‐Roqueplo AP , Zinzindohoue F , Chatellier G , Plouin PF . Year of diagnosis, features at presentation, and risk of recurrence in patients with pheochromocytoma or secreting paraganglioma. J Clin Endocrinol Metab 90: 2110‐2116, 2005.
 22. Amenta F , Collier WL , Erdo SL , Giuliani S , Maggi CA , Meli A . GABAA receptor sites modulating catecholamine secretion in the rat adrenal gland: Evidence from 3H‐muscimol autoradiography and in vivo functional studies. Pharmacology 37: 394‐402, 1988.
 23. Anderson EA , Sinkey CA , Lawton WJ , Mark AL . Elevated sympathetic nerve activity in borderline hypertensive humans. Evidence from direct intraneural recordings. Hypertension 14: 177‐183, 1989.
 24. Anderson K , Robinson PJ , Marley PD . Cholinoceptor regulation of cyclic AMP levels in bovine adrenal medullary cells. Br J Pharmacol 106: 360‐366, 1992.
 25. Andreis PG , Neri G , Prayer‐Galetti T , Rossi GP , Gottardo G , Malendowicz LK , Nussdorfer GG . Effects of adrenomedullin on the human adrenal glands: An in vitro study. J Clin Endocrinol Metab 82: 1167‐1170, 1997.
 26. Arimura A . Pituitary adenylate cyclase‐activating polypeptide (PACAP): Discovery and current status of research. Regul Pept 37: 287‐303, 1992.
 27. Ariton M , Juan CS , AvRuskin TW . Pheochromocytoma: Clinical observations from a Brooklyn tertiary hospital. Endocr Prac: Off J Am Coll Endocrinol Am Assoc Clin Endocrinol 6: 249‐252, 2000.
 28. Arribas‐Blazquez M , Olivos‐Ore LA , Barahona MV , Sanchez de la Muela M , Solar V , Jimenez E , Gualix J , McIntosh JM , Ferrer‐Montiel A , Miras‐Portugal MT , Artalejo AR . Overexpression of P2X3 and P2X7 receptors and TRPV1 channels in adrenomedullary chromaffin cells in a rat model of neuropathic pain. Int J Mol Sci 20 (1): 155, 2019.
 29. Artalejo AR , Garcia AG , Neher E . Small‐conductance Ca(2+)‐activated K+ channels in bovine chromaffin cells. Pflug Archiv: Eur J Physiol 423: 97‐103, 1993.
 30. Artalejo CR , Adams ME , Fox AP . Three types of Ca2+ channel trigger secretion with different efficacies in chromaffin cells. Nature 367: 72‐76, 1994.
 31. Artalejo CR , Dahmer MK , Perlman RL , Fox AP . Two types of Ca2+ currents are found in bovine chromaffin cells: Facilitation is due to the recruitment of one type. J Physiol 432: 681‐707, 1991.
 32. Artalejo CR , Garcia AG , Aunis D . Chromaffin cell calcium channel kinetics measured isotopically through fast calcium, strontium, and barium fluxes. J Biol Chem 262: 915‐926, 1987.
 33. Artalejo CR , Henley JR , McNiven MA , Palfrey HC . Rapid endocytosis coupled to exocytosis in adrenal chromaffin cells involves Ca2+, GTP, and dynamin but not clathrin. Proc Natl Acad Sci USA 92: 8328‐8332, 1995.
 34. Artalejo CR , Lemmon MA , Schlessinger J , Palfrey HC . Specific role for the PH domain of dynamin‐1 in the regulation of rapid endocytosis in adrenal chromaffin cells. EMBO J 16: 1565‐1574, 1997.
 35. Artalejo CR , Mogul DJ , Perlman RL , Fox AP . Three types of bovine chromaffin cell Ca2+ channels: Facilitation increases the opening probability of a 27 pS channel. J Physiol 444: 213‐240, 1991.
 36. Artalejo CR , Perlman RL , Fox AP . Omega‐conotoxin GVIA blocks a Ca2+ current in bovine chromaffin cells that is not of the “classic” N type. Neuron 8: 85‐95, 1992.
 37. Arvan P , Castle D . Sorting and storage during secretory granule biogenesis: Looking backward and looking forward. Biochem J 332 (Pt 3): 593‐610, 1998.
 38. Augustine GJ , Neher E . Calcium requirements for secretion in bovine chromaffin cells. J Physiol 450: 247‐271, 1992.
 39. Aunis D , Bader MF . The cytoskeleton as a barrier to exocytosis in secretory cells. J Exp Biol 139: 253‐266, 1988.
 40. Babcock DF , Herrington J , Goodwin PC , Park YB , Hille B . Mitochondrial participation in the intracellular Ca2+ network. J Cell Biol 136: 833‐844, 1997.
 41. Bader PL , Faizi M , Kim LH , Owen SF , Tadross MR , Alfa RW , Bett GC , Tsien RW , Rasmusson RL , Shamloo M . Mouse model of Timothy syndrome recapitulates triad of autistic traits. Proc Natl Acad Sci USA 108: 15432‐15437, 2011.
 42. Baker PF , Blaustein MP , Hodgkin AL , Steinhardt RA . The influence of calcium on sodium efflux in squid axons. J Physiol 200: 431‐458, 1969.
 43. Baker PF , Knight DE . Calcium‐dependent exocytosis in bovine adrenal medullary cells with leaky plasma membranes. Nature 276: 620‐622, 1978.
 44. Baker PF , Knight DE . Calcium control of exocytosis and endocytosis in bovine adrenal medullary cells. Philos Trans R Soc Lond B Biol Sci 296: 83‐103, 1981.
 45. Baker PF , Rink TJ . Catecholamine release from bovine adrenal medulla in response to maintained depolarization. J Physiol 253: 593‐620, 1975.
 46. Baldelli P , Hernandez‐Guijo JM , Carabelli V , Novara M , Cesetti T , Andres‐Mateos E , Montiel C , Carbone E . Direct and remote modulation of L‐channels in chromaffin cells – Distinct actions on alpha(1C) and alpha(1D) subunits? Mol Neurobiol 29: 73‐96, 2004.
 47. Barbara JG , Poncer JC , McKinney RA , Takeda K . An adrenal slice preparation for the study of chromaffin cells and their cholinergic innervation. J Neurosci Methods 80: 181‐189, 1998.
 48. Barbara JG , Takeda K . Quantal release at a neuronal nicotinic synapse from rat adrenal gland. Proc Natl Acad Sci USA 93: 9905‐9909, 1996.
 49. Barg S , Machado JD . Compensatory endocytosis in chromaffin cells. Acta Physiol (Oxf) 192: 195‐201, 2008.
 50. Barman SM , Gebber GL . Sympathetic nerve rhythm of brain stem origin. Am J Physiol 239: R42‐R47, 1980.
 51. Bean BP . The action potential in mammalian central neurons. Nat Rev Neurosci 8: 451‐465, 2007.
 52. Bean BP . Neurotransmitter inhibition of neuronal calcium currents by changes in channel voltage dependence. Nature 340: 153‐156, 1989.
 53. Beaudet MM , Parsons RL , Braas KM , May V . Mechanisms mediating pituitary adenylate cyclase‐activating polypeptide depolarization of rat sympathetic neurons. J Neurosci 20: 7353‐7361, 2000.
 54. Becherer U , Moser T , Stuhmer W , Oheim M . Calcium regulates exocytosis at the level of single vesicles. Nat Neurosci 6: 846‐853, 2003.
 55. Ben‐Ari Y , Gaiarsa JL , Tyzio R , Khazipov R . GABA: A pioneer transmitter that excites immature neurons and generates primitive oscillations. Physiol Rev 87: 1215‐1284, 2007.
 56. Benedeczky I , Somogyi P . Cytochemical localization of exogenous peroxidase in adrenal medullary cells of hamster. Acta Biol Acad Sci Hung 29: 155‐163, 1978.
 57. Benfante R , Flora A , Di Lascio S , Cargnin F , Longhi R , Colombo S , Clementi F , Fornasari D . Transcription factor PHOX2A regulates the human alpha3 nicotinic receptor subunit gene promoter. J Biol Chem 282: 13290‐13302, 2007.
 58. Bennett MR , McLachlan EM . An electrophysiological analysis of the synthesis of acetylcholine in preganglionic nerve terminals. J Physiol 221: 669‐682, 1972.
 59. Bereiter DA , Engeland WC , Gann DS . Adrenal secretion of epinephrine after stimulation of trigeminal nucleus caudalis depends on stimulus pattern. Neuroendocrinology 45: 54‐61, 1987.
 60. Bernard C . Decons sur les phénomenes de la vie communs aux animaux et auc végétaux. Paris: Bailliere, 1878.
 61. Biales B , Dichter M , Tischler A . Electrical excitability of cultured adrenal chromaffin cells. J Physiol 262: 743‐753, 1976.
 62. Blaschko H , Comline RS , Schneider FH , Silver M , Smith AD . Secretion of a chromaffin granule protein, chromogranin, from the adrenal gland after splanchnic stimulation. Nature 215: 58‐59, 1967.
 63. Blaustein MP , Lederer WJ . Sodium/calcium exchange: Its physiological implications. Physiol Rev 79: 763‐854, 1999.
 64. Bödding M . Histamine evoked sustained elevations of cytosolic Ca2+ in bovine adrenal chromaffin cells independently of Ca2+ entry. Cell Calcium 27: 139‐151, 2000.
 65. Bomfim GHS , Mendez‐Lopez I , Fernandez‐Morales JC , Padin JF , Jurkiewicz A , Jurkiewicz NH , Garcia AG . Electrophysiological properties and augmented catecholamine release from chromaffin cells of WKY and SHR rats contributing to the hypertension development elicited by chronic EtOH consumption. Eur J Pharmacol 803: 65‐77, 2017.
 66. Borchelt DR , Ratovitski T , van Lare J , Lee MK , Gonzales V , Jenkins NA , Copeland NG , Price DL , Sisodia SS . Accelerated amyloid deposition in the brains of transgenic mice coexpressing mutant presenilin 1 and amyloid precursor proteins. Neuron 19: 939‐945, 1997.
 67. Borges R . The ATP or the natural history of neurotransmission. Purinergic Signal 9: 5‐6, 2013.
 68. Borges R . Histamine H1 receptor activation mediates the preferential release of adrenaline in the rat adrenal gland. Life Sci 54: 631‐640, 1994.
 69. Borges R , Camacho M , Gillis KD . Measuring secretion in chromaffin cells using electrophysiological and electrochemical methods. Acta Physiol (Oxf) 192: 173‐184, 2008.
 70. Borges R , Gandia L , Carbone E . Old and emerging concepts on adrenal chromaffin cell stimulus‐secretion coupling. Pflug Archiv: Eur J Physiol 470: 1‐6, 2018.
 71. Bormann J . Electrophysiology of GABAA and GABAB receptor subtypes. Trends Neurosci 11: 112‐116, 1988.
 72. Bos JL . Epac: A new cAMP target and new avenues in cAMP research. Nat Rev Mol Cell Biol 4: 733‐738, 2003.
 73. Bournaud R , Hidalgo J , Yu H , Girard E , Shimahara T . Catecholamine secretion from rat foetal adrenal chromaffin cells and hypoxia sensitivity. Pflug Archiv: Eur J Physiol 454: 83‐92, 2007.
 74. Bournaud R , Hidalgo J , Yu H , Jaimovich E , Shimahara T . Low threshold T‐type calcium current in rat embryonic chromaffin cells. J Physiol 537: 35‐44, 2001.
 75. Bowery N . GABAB receptors and their significance in mammalian pharmacology. Trends Pharmacol Sci 10: 401‐407, 1989.
 76. Brandt BL , Hagiwara S , Kidokoro Y , Miyazaki S . Action potentials in the rat chromaffin cell and effects of acetylcholine. J Physiol 263: 417‐439, 1976.
 77. Bravo EL , Tagle R . Pheochromocytoma: State‐of‐the‐art and future prospects. Endocr Rev 24: 539‐553, 2003.
 78. Brede M , Nagy G , Philipp M , Sorensen JB , Lohse MJ , Hein L . Differential control of adrenal and sympathetic catecholamine release by alpha 2‐adrenoceptor subtypes. Mol Endocrinol (Baltimore, Md) 17: 1640‐1646, 2003.
 79. Brini M , Carafoli E . The plasma membrane Ca(2)+ ATPase and the plasma membrane sodium calcium exchanger cooperate in the regulation of cell calcium. Cold Spring Harb Perspect Biol 3, 2011. DOI: 10.1101/cshperspect.a004168.
 80. Brown CH , Scott V , Ludwig M , Leng G , Bourque CW . Somatodendritic dynorphin release: Orchestrating activity patterns of vasopressin neurons. Biochem Soc Trans 35: 1236‐1242, 2007.
 81. Brown RL , Strassmaier T , Brady JD , Karpen JW . The pharmacology of cyclic nucleotide‐gated channels: Emerging from the darkness. Curr Pharm Des 12: 3597‐3613, 2006.
 82. Bunn SJ , Ait‐Ali D , Eiden LE . Immune‐neuroendocrine integration at the adrenal gland: Cytokine control of the adrenomedullary transcriptome. J Mol Neurosci 48: 413‐419, 2012.
 83. Bunn SJ , Boyd TL . Characterization of histamine‐induced catecholamine secretion from bovine adrenal medullary chromaffin cells. J Neurochem 58: 1602‐1610, 1992.
 84. Bunn SJ , Marley PD , Livett BG . Receptor stimulated formation of inositol phosphates in cultures of bovine adrenal medullary cells: The effects of bradykinin, bombesin and neurotensin. Neuropeptides 15: 187‐194, 1990.
 85. Burgoyne RD . Fast exocytosis and endocytosis triggered by depolarisation in single adrenal chromaffin cells before rapid Ca2+ current run‐down. Pflug Archiv: Eur J Physiol 430: 213‐219, 1995.
 86. Burgoyne RD . Mechanisms of catecholamine secretion from adrenal chromaffin cells. J Physiol Pharmacol 46: 273‐283, 1995.
 87. Busik J , Nakamura M , Abe Y , Shibuya I , Kanno T . Effects of GABA on spontaneous [Ca2+]c dynamics and electrical properties of rat adrenal chromaffin cells. Brain Res 739: 97‐103, 1996.
 88. Byrne CJ , Khurana S , Kumar A , Tai TC . Inflammatory signaling in hypertension: Regulation of adrenal catecholamine biosynthesis. Front Endocrinol 9: 343, 2018.
 89. Calbet JA . Chronic hypoxia increases blood pressure and noradrenaline spillover in healthy humans. J Physiol 551: 379‐386, 2003.
 90. Calhoun DA , Jones D , Textor S , Goff DC , Murphy TP , Toto RD , White A , Cushman WC , White W , Sica D , Ferdinand K , Giles TD , Falkner B , Carey RM . Resistant hypertension: Diagnosis, evaluation, and treatment. A scientific statement from the American Heart Association Professional Education Committee of the Council for High Blood Pressure Research. Hypertension 51: 1403‐1419, 2008.
 91. Calorio C , Gavello D , Guarina L , Salio C , Sassoe‐Pognetto M , Riganti C , Bianchi FT , Hofer NT , Tuluc P , Obermair GJ , Defilippi P , Balzac F , Turco E , Bett GC , Rasmusson RL , Carbone E . Impaired chromaffin cell excitability and exocytosis in autistic Timothy syndrome TS2‐neo mouse rescued by L‐type calcium channel blockers. J Physiol 597: 1705‐1733, 2019.
 92. Calvo‐Gallardo E , de Pascual R , Fernandez‐Morales JC , Arranz‐Tagarro JA , Maroto M , Nanclares C , Gandia L , de Diego AM , Padin JF , Garcia AG . Depressed excitability and ion currents linked to slow exocytotic fusion pore in chromaffin cells of the SOD1(G93A) mouse model of amyotrophic lateral sclerosis. Am J Physiol Cell Physiol 308: C1‐C19, 2015.
 93. Camacho M , Machado JD , Alvarez J , Borges R . Intravesicular calcium release mediates the motion and exocytosis of secretory organelles: A study with adrenal chromaffin cells. J Biol Chem 283: 22383‐22389, 2008.
 94. Camacho M , Machado JD , Montesinos MS , Criado M , Borges R . Intragranular pH rapidly modulates exocytosis in adrenal chromaffin cells. J Neurochem 96: 324‐334, 2006.
 95. Cannon WB . Organization for physiological homeostasis. Physiol Rev 9: 399‐431, 1929.
 96. Cannon WB . Wisdom of the Body. New York: W. W. Norton & Company, 1932.
 97. Cannon WB , Hoskins RG . The effects of sphyxia, hyperpnea, and sensory stimulation on adrenal secretion. Am J Physiol 29: 274‐279, 1911.
 98. Carabelli V , Carra I , Carbone E . Localized secretion of ATP and opioids revealed through single Ca2+ channel modulation in bovine chromaffin cells. Neuron 20: 1255‐1268, 1998.
 99. Carabelli V , D'Ascenzo M , Carbone E , Grassi C . Nitric oxide inhibits neuroendocrine Ca(v)1 L‐channel gating via cGMP‐dependent protein kinase in cell‐attached patches of bovine chromaffin cells. J Physiol‐Lond 541: 351‐366, 2002.
 100. Carabelli V , Giancippoli A , Baldelli P , Carbone E , Artalejo AR . Distinct potentiation of L‐type currents and secretion by cAMP in rat chromaffin cells. Biophys J 85: 1326‐1337, 2003.
 101. Carabelli V , Hernandez‐Guijo JM , Baldelli P , Carbone E . Direct autocrine inhibition and cAMP‐dependent potentiation of single L‐type Ca2+ channels in bovine chromaffin cells. J Physiol‐Lond 532: 73‐90, 2001.
 102. Carabelli V , Lovallo M , Magnelli V , Zucker H , Carbone E . Voltage‐dependent modulation of single N‐type Ca2+ channel kinetics by receptor agonists in IMR32 cells. Biophys J 70: 2144‐2154, 1996.
 103. Carabelli V , Marcantoni A , Comunanza V , Carbone E . Fast exocytosis mediated by T‐ and L‐type channels in chromaffin cells: Distinct voltage‐dependence but similar Ca2+‐dependence. Eur Biophys J Biophys Lett 36: 753‐762, 2007.
 104. Carabelli V , Marcantoni A , Comunanza V , De Luca A , Diaz J , Borges R , Carbone E . Chronic hypoxia up‐regulates alpha(1H) T‐type channels and low‐threshold catecholamine secretion in rat chromaffin cells. J Physiol 584: 149‐165, 2007.
 105. Carafoli E . The calcium cycle of mitochondria. FEBS Lett 104: 1‐5, 1979.
 106. Carafoli E , Fedrizzi L , Domi T , Di Leva F , Brini M . Caclium pumps part II. Transmission: Effectors and cytosolic events. In: Handbook of Cell Signaling (2nd ed). New York: Academic Press, 2010, p. 943‐947.
 107. Carbone E , Calorio C , Vandael DH . T‐type channel‐mediated neurotransmitter release. Pflug Archiv: Eur J Physiol 466: 677‐687, 2014.
 108. Carbone E , Carabelli V . O2 sensing in chromaffin cells: New duties for T‐type channels. J Physiol 587: 1859‐1860, 2009.
 109. Carbone E , Carabelli V , Cesetti T , Baldelli P , Hernandez‐Guijo JM , Giusta L . G‐protein‐ and cAMP‐dependent L‐channel gating modulation: A manyfold system to control calcium entry in neurosecretory cells. Pflug Archiv: Eur J Physiol 442: 801‐813, 2001.
 110. Carbone E , Giancippoli A , Marcantoni A , Guido D , Carabelli V . A new role for T‐type channels in fast “low‐threshold” exocytosis. Cell Calcium 40: 147‐154, 2006.
 111. Cardenas AM , Marengo FD . Rapid endocytosis and vesicle recycling in neuroendocrine cells. Cell Mol Neurobiol 30: 1365‐1370, 2010.
 112. Caricati‐Neto A , Padin JF , Silva‐Junior ED , Fernandez‐Morales JC , de Diego AM , Jurkiewicz A , Garcia AG . Novel features on the regulation by mitochondria of calcium and secretion transients in chromaffin cells challenged with acetylcholine at 37 °C. Physiol Rep 1: e00182, 2013.
 113. Carmichael SW , Winkler H . The adrenal chromaffin cell. Sci Am 253: 39‐49, 1985.
 114. Carrasco‐Serrano C , Campos‐Caro A , Viniegra S , Ballesta JJ , Criado M . GC‐ and E‐box motifs as regulatory elements in the proximal promoter region of the neuronal nicotinic receptor alpha7 subunit gene. J Biol Chem 273: 20021‐20028, 1998.
 115. Catterall WA . Interactions of presynaptic Ca2+ channels and snare proteins in neurotransmitter release. Ann NY Acad Sci 868: 144‐159, 1999.
 116. Catterall WA . Voltage‐gated calcium channels. Cold Spring Harbor Perspect Biol 3: 23, 2011.
 117. Catterall WA , Goldin AL , Waxman SG . International Union of Pharmacology. XLVII. Nomenclature and structure‐function relationships of voltage‐gated sodium channels. Pharmacol Rev 57: 397‐409, 2005.
 118. Ceccarelli B , Hurlbut WP . Ca2+‐dependent recycling of synaptic vesicles at the frog neuromuscular junction. J Cell Biol 87: 297‐303, 1980.
 119. Ceccarelli B , Hurlbut WP , Mauro A . Turnover of transmitter and synaptic vesicles at the frog neuromuscular junction. J Cell Biol 57: 499‐524, 1973.
 120. Cena V , Nicolas GP , Sanchez‐Garcia P , Kirpekar SM , Garcia AG . Pharmacological dissection of receptor‐associated and voltage‐sensitive ionic channels involved in catecholamine release. Neuroscience 10: 1455‐1462, 1983.
 121. Cesetti T , Hernandez‐Guijo JM , Baldelli P , Carabelli V , Carbone E . Opposite action of beta 1‐ and beta 2‐adrenergic receptors on Ca(V)1 L‐channel current in rat adrenal chromaffin cells. J Neurosci 23: 73‐83, 2003.
 122. Chan SA , Polo‐Parada L , Smith C . Action potential stimulation reveals an increased role for P/Q‐calcium channel‐dependent exocytosis in mouse adrenal tissue slices. Arch Biochem Biophys 435: 65‐73, 2005.
 123. Cheek TR , Barry VA , Berridge MJ , Missiaen L . Bovine adrenal chromaffin cells contain an inositol 1,4,5‐trisphosphate‐insensitive but caffeine‐sensitive Ca2+ store that can be regulated by intraluminal free Ca2+ . Biochem J 275 (Pt 3): 697‐701, 1991.
 124. Cheek TR , Berridge MJ , Moreton RB , Stauderman KA , Murawsky MM , Bootman MD . Quantal Ca2+ mobilization by ryanodine receptors is due to all‐or‐none release from functionally discrete intracellular stores. Biochem J 301 (Pt 3): 879‐883, 1994.
 125. Cheek TR , Burgoyne RD . Nicotine‐evoked disassembly of cortical actin filaments in adrenal chromaffin cells. FEBS Lett 207: 110‐114, 1986.
 126. Cheek TR , Moreton RB , Berridge MJ , Stauderman KA , Murawsky MM , Bootman MD . Quantal Ca2+ release from caffeine‐sensitive stores in adrenal chromaffin cells. J Biol Chem 268: 27076‐27083, 1993.
 127. Cheek TR , Murawsky MM , Stauderman KA . Histamine‐induced Ca2+ entry precedes Ca2+ mobilization in bovine adrenal chromaffin cells. Biochem J 304 (Pt 2): 469‐476, 1994.
 128. Chen TT , Li L , Chung DH , Allen CD , Torti SV , Torti FM , Cyster JG , Chen CY , Brodsky FM , Niemi EC , Nakamura MC , Seaman WE , Daws MR . TIM‐2 is expressed on B cells and in liver and kidney and is a receptor for H‐ferritin endocytosis. J Exp Med 202: 955‐965, 2005.
 129. Chow RH , Klingauf J , Neher E . Time course of Ca2+ concentration triggering exocytosis in neuroendocrine cells. Proc Natl Acad Sci USA 91: 12765‐12769, 1994.
 130. Chow RH , von Ruden L , Neher E . Delay in vesicle fusion revealed by electrochemical monitoring of single secretory events in adrenal chromaffin cells. Nature 356: 60‐63, 1992.
 131. Chuang D‐M , Costa E . Biosynthesis of tyrosine hydroxylase in rat adrenal medulla after exposure to cold. Proc Natl Acad Sci USA 71: 4570‐4574, 1974.
 132. Clapham DE , Neher E . Substance P reduces acetylcholine‐induced currents in isolated bovine chromaffin cells. J Physiol 347: 255‐277, 1984.
 133. Colomer C , Martin AO , Desarmenien MG , Guerineau NC . Gap junction‐mediated intercellular communication in the adrenal medulla: An additional ingredient of stimulus‐secretion coupling regulation. Biochim Biophys Acta 1818: 1937‐1951, 2011.
 134. Colomer C , Olivos Ore LA , Coutry N , Mathieu MN , Arthaud S , Fontanaud P , Iankova I , Macari F , Thouennon E , Yon L , Anouar Y , Guerineau NC . Functional remodeling of gap junction‐mediated electrical communication between adrenal chromaffin cells in stressed rats. J Neurosci 28: 6616‐6626, 2008.
 135. Comb M , Seeburg PH , Adelman J , Eiden L , Herbert E . Primary structure of the human Met‐ and Leu‐enkephalin precursor and its mRNA. Nature 295: 663‐666, 1982.
 136. Comunanza V , Marcantoni A , Vandael DHF , Mahapatra S , Gavello D , Navarro‐Tableros V , Carabelli V , Carbone E . Ca(V)1.3 as pacemaker channels in adrenal chromaffin cells. Channels 4: 440‐446, 2010.
 137. Conrad CH , Brooks WW , Hayes JA , Sen S , Robinson KG , Bing OH . Myocardial fibrosis and stiffness with hypertrophy and heart failure in the spontaneously hypertensive rat. Circulation 91: 161‐170, 1995.
 138. Cortez V , Santana M , Marques AP , Mota A , Rosmaninho‐Salgado J , Cavadas C . Regulation of catecholamine release in human adrenal chromaffin cells by beta‐adrenoceptors. Neurochem Int 60: 387‐393, 2012.
 139. Coupland RE . Electron microscopic observations on the structure of the rat adrenal medulla. I. The ultrastructure and organization of chromaffin cells in the normal adrenal medulla. J Anat 99: 231‐254, 1965.
 140. Coupland RE , Pyper AS , Hopwood D . A method for differentiating between noradrenaline‐ and adrenaline‐storing cells in the light and electron microscope. Nature 201: 1240‐1242, 1964.
 141. Couratier P , Hugon J , Sindou P , Vallat JM , Dumas M . Cell culture evidence for neuronal degeneration in amyotrophic lateral sclerosis being linked to glutamate AMPA/kainate receptors. Lancet 341: 265‐268, 1993.
 142. Criado M . Acetylcholine nicotinic receptor subtypes in chromaffin cells. Pflug Archiv: Eur J Physiol 470: 13‐20, 2018.
 143. Crill WE . Persistent sodium current in mammalian central neurons. Annu Rev Physiol 58: 349‐362, 1996.
 144. Cuchillo‐Ibanez I , Lejen T , Albillos A , Rose SD , Olivares R , Villarroya M , Garcia AG , Trifaro JM . Mitochondrial calcium sequestration and protein kinase C cooperate in the regulation of cortical F‐actin disassembly and secretion in bovine chromaffin cells. J Physiol 560: 63‐76, 2004.
 145. Cuchillo‐Ibanez I , Olivares R , Aldea M , Villarroya M , Arroyo G , Fuentealba J , Garcia AG , Albillos A . Acetylcholine and potassium elicit different patterns of exocytosis in chromaffin cells when the intracellular calcium handling is disturbed. Pflug Archiv: Eur J Physiol 444: 133‐142, 2002.
 146. Cummins TR , Aglieco F , Renganathan M , Herzog RI , Dib‐Hajj SD , Waxman SG . Nav1.3 sodium channels: Rapid repriming and slow closed‐state inactivation display quantitative differences after expression in a mammalian cell line and in spinal sensory neurons. J Neurosci 21: 5952‐5961, 2001.
 147. Currie G , Freel EM , Perry CG , Dominiczak AF . Disorders of blood pressure regulation‐role of catecholamine biosynthesis, release, and metabolism. Curr Hypertens Rep 14: 38‐45, 2012.
 148. Currie KP . Inhibition of Ca2+ channels and adrenal catecholamine release by G protein coupled receptors. Cell Mol Neurobiol 30: 1201‐1208, 2010.
 149. Currie KP , Fox AP . ATP serves as a negative feedback inhibitor of voltage‐gated Ca2+ channel currents in cultured bovine adrenal chromaffin cells. Neuron 16: 1027‐1036, 1996.
 150. Currie KP , Fox AP . Differential facilitation of N‐ and P/Q‐type calcium channels during trains of action potential‐like waveforms. J Physiol 539: 419‐431, 2002.
 151. Currie KP , Fox AP . Voltage‐dependent, pertussis toxin insensitive inhibition of calcium currents by histamine in bovine adrenal chromaffin cells. J Neurophysiol 83: 1435‐1442, 2000.
 152. Dani JA . Neuronal nicotinic acetylcholine receptor structure and function and response to nicotine. Int Rev Neurobiol 124: 3‐19, 2015.
 153. de Diego AM . Electrophysiological and morphological features underlying neurotransmission efficacy at the splanchnic nerve‐chromaffin cell synapse of bovine adrenal medulla. Am J Physiol Cell Physiol 298: C397‐C405, 2010.
 154. de Diego AM , Arnaiz‐Cot JJ , Hernandez‐Guijo JM , Gandia L , Garcia AG . Differential variations in Ca2+ entry, cytosolic Ca2+ and membrane capacitance upon steady or action potential depolarizing stimulation of bovine chromaffin cells. Acta Physiol (Oxford) 194: 97‐109, 2008.
 155. de Diego AM , Gandia L , Garcia AG . A physiological view of the central and peripheral mechanisms that regulate the release of catecholamines at the adrenal medulla. Acta Physiol (Oxford, England) 192: 287‐301, 2007.
 156. de Diego AM , Lorrio S , Calvo‐Gallardo E , Garcia AG . Smaller quantal size and faster kinetics of single exocytotic events in chromaffin cells from the APP/PS1 mouse model of Alzheimer's disease. Biochem Biophys Res Commun 428: 482‐486, 2012.
 157. de Diego AMG , Garcia AG . Altered exocytosis in chromaffin cells from mouse models of neurodegenerative diseases. Acta Physiol (Oxford, England) 224: e13090, 2018.
 158. de Rooij J , Zwartkruis FJT , Verheijen MHG , Cool RH , Jijman SMB , Witinghofer A , Bos JL . Epac is a Rap1 guanine‐nucleotide‐exchange factor directly activated by cyclic AMP. Nature 396: 474‐477, 1998.
 159. del Barrio L , Egea J , Leon R , Romero A , Ruiz A , Montero M , Alvarez J , Lopez MG . Calcium signalling mediated through alpha7 and non‐alpha7 nAChR stimulation is differentially regulated in bovine chromaffin cells to induce catecholamine release. Br J Pharmacol 162: 94‐110, 2011.
 160. Del Toro R , Levitsky KL , Lopez‐Barneo J , Chiara MD . Induction of T‐type calcium channel gene expression by chronic hypoxia. J Biol Chem 278: 22316‐22324, 2003.
 161. Delmas P , Brown DA . Pathways modulating neural KCNQ/M (Kv7) potassium channels. Nat Rev Neurosci 6: 850‐862, 2005.
 162. Desarmenien MG , Jourdan C , Toutain B , Vessieres E , Hormuzdi SG , Guerineau NC . Gap junction signalling is a stress‐regulated component of adrenal neuroendocrine stimulus‐secretion coupling in vivo. Nat Commun 4: 2938, 2013.
 163. Dhara M , Mohrmann R , Bruns D . v‐SNARE function in chromaffin cells. Pflug Archiv: Eur J Physiol 470: 169‐180, 2018.
 164. Di Angelantonio S , Giniatullin R , Costa V , Sokolova E , Nistri A . Modulation of neuronal nicotinic receptor function by the neuropeptides CGRP and substance P on autonomic nerve cells. Br J Pharmacol 139: 1061‐1073, 2003.
 165. Di Leva F , Domi T , Fedrizzi L , Lim D , Carafoli E . The plasma membrane Ca2+ ATPase of animal cells: Structure, function and regulation. Arch Biochem Biophys 476: 65‐74, 2008.
 166. Diaz‐Vera J , Camacho M , Machado JD , Dominguez N , Montesinos MS , Hernandez‐Fernaud JR , Lujan R , Borges R . Chromogranins A and B are key proteins in amine accumulation, but the catecholamine secretory pathway is conserved without them. FASEB J 26: 430‐438, 2012.
 167. Ding JP , Li ZW , Lingle CJ . Inactivating BK channels in rat chromaffin cells may arise from heteromultimeric assembly of distinct inactivation‐competent and noninactivating subunits. Biophys J 74: 268‐289, 1998.
 168. Dinkelacker V , Voets T , Neher E , Moser T . The readily releasable pool of vesicles in chromaffin cells is replenished in a temperature‐dependent manner and transiently overfills at 37 °C. J Neurosci 20: 8377‐8383, 2000.
 169. Dodge FA Jr. , Rahamimoff R . Co‐operative action a calcium ions in transmitter release at the neuromuscular junction. J Physiol 193: 419‐432, 1967.
 170. Dominguez N , Estevez‐Herrera J , Borges R , Machado JD . The interaction between chromogranin A and catecholamines governs exocytosis. FASEB J 28: 4657‐4667, 2014.
 171. Douglas SA , Sreenivasan D , Carman FH , Bunn SJ . Cytokine interactions with adrenal medullary chromaffin cells. Cell Mol Neurobiol 30: 1467‐1475, 2010.
 172. Douglas WW . Stimulus‐secretion coupling: The concept and clues from chromaffin and other cells. Br J Pharmacol 34: 451‐474, 1968.
 173. Douglas WW , Poisner AM . Preferential release of adrenaline from the adrenal medulla by muscarine and pilocarpine. Nature 208: 1102‐1103, 1965.
 174. Douglas WW , Poisner AM . Stimulation of uptake of calcium‐45 in the adrenal gland by acetylcholine. Nature 192: 1299, 1961.
 175. Douglas WW , Rubin RP . Mechanism of nicotinic action at the adrenal medulla: Calcium as a link in stimulus‐secretion coupling. Nature 192: 1087‐1089, 1961.
 176. Douglas WW , Rubin RP . The role of calcium in the secretory response of the adrenal medulla to acetylcholine. J Physiol 159: 40‐57, 1961.
 177. Dragatsis I , Dietrich P , Zeitlin S . Expression of the Huntingtin‐associated protein 1 gene in the developing and adult mouse. Neurosci Lett 282: 37‐40, 2000.
 178. Duan K , Yu X , Zhang C , Zhou Z . Control of secretion by temporal patterns of action potentials in adrenal chromaffin cells. J Neurosci: Off J Soc Neurosci 23: 11235‐11243, 2003.
 179. Duerr JS , Frisby DL , Gaskin J , Duke A , Asermely K , Huddleston D , Eiden LE , Rand JB . The cat‐1 gene of Caenorhabditis elegans encodes a vesicular monoamine transporter required for specific monoamine‐dependent behaviors. J Neurosci 19: 72‐84, 1999.
 180. Duncan RR , Greaves J , Wiegand UK , Matskevich I , Bodammer G , Apps DK , Shipston MJ , Chow RH . Functional and spatial segregation of secretory vesicle pools according to vesicle age. Nature 422: 176‐180, 2003.
 181. Dunevall J , Fathali H , Najafinobar N , Lovric J , Wigstrom J , Cans AS , Ewing AG . Characterizing the catecholamine content of single mammalian vesicles by collision‐adsorption events at an electrode. J Am Chem Soc 137: 4344‐4346, 2015.
 182. Dunlap K , Fischbach GD . Neurotransmitters decrease the calcium conductance activated by depolarization of embryonic chick sensory neurones. J Physiol 317: 519‐535, 1981.
 183. Dunlap K , Fischbach GD . Neurotransmitters decrease the calcium component of sensory neurone action potentials. Nature 276: 837‐839, 1978.
 184. Dunn LA , Holz RW . Catecholamine secretion from digitonin‐treated adrenal medullary chromaffin cells. J Biol Chem 258: 4989‐4993, 1983.
 185. Ebert SN , Balt SL , Hunter JP , Gashler A , Sukhatme V , Wong DL . Egr‐1 activation of rat adrenal phenylethanolamine N‐methyltransferase gene. J Biol Chem 269: 20885‐20898, 1994.
 186. Edwards RM , Trizna W , Aiyar N . Adrenomedullin: A new peptide involved in cardiorenal homeostasis? Exp Nephrol 5: 18‐22, 1997.
 187. Edwards SL , Anderson CR , Southwell BR , McAllen RM . Distinct preganglionic neurons innervate noradrenaline and adrenaline cells in the cat adrenal medulla. Neuroscience 70: 825‐832, 1996.
 188. Eiden LE . Is chromogranin a prohormone? Nature 325: 301, 1987.
 189. Eiden LE , Emery AC , Zhang L , Smith CB . PACAP signaling in stress: Insights from the chromaffin cell. Pflug Archiv: Eur J Physiol 470: 79‐88, 2018.
 190. Eiden LE , Giraud P , Dave J , Hotchkiss JA , Affolter H‐U . Nicotinic receptor stimulation activates both enkephalin release and biosynthesis in adrenal chromaffin cells. Nature 312: 661‐663, 1984.
 191. Eiden LE , Iacangelo A , Hsu C‐M , Hotchkiss AJ , Bader M‐F , Aunis D . Chromogranin A synthesis and secretion in chromaffin cells. J Neurochem 49: 65‐74, 1987.
 192. Eipper BA , Milgram SL , Husten EJ , Yun HY , Mains RE . Peptidylglycine alpha‐amidating monooxygenase: A multifunctional protein with catalytic, processing, and routing domains. Protein Sci: Publ Protein Soc 2: 489‐497, 1993.
 193. Eisenhofer G . The role of neuronal and extraneuronal plasma membrane transporters in the inactivation of peripheral catecholamines. Pharmacol Ther 91: 35‐62, 2001.
 194. Eisenhofer G , Huynh TT , Hiroi M , Pacak K . Understanding catecholamine metabolism as a guide to the biochemical diagnosis of pheochromocytoma. Rev Endocr Metab Disord 2: 297‐311, 2001.
 195. Eisenhofer G , Keiser H , Friberg P , Mezey E , Huynh TT , Hiremagalur B , Ellingson T , Duddempudi S , Eijsbouts A , Lenders JW . Plasma metanephrines are markers of pheochromocytoma produced by catechol‐O‐methyltransferase within tumors. J Clin Endocrinol Metab 83: 2175‐2185, 1998.
 196. Eisenhofer G , Kopin IJ , Goldstein DS . Catecholamine metabolism: A contemporary view with implications for physiology and medicine. Pharmacol Rev 56: 331‐349, 2004.
 197. Eisenhofer G , Lenders JW , Siegert G , Bornstein SR , Friberg P , Milosevic D , Mannelli M , Linehan WM , Adams K , Timmers HJ , Pacak K . Plasma methoxytyramine: A novel biomarker of metastatic pheochromocytoma and paraganglioma in relation to established risk factors of tumour size, location and SDHB mutation status. Eur J Cancer (Oxford, England: 1990) 48: 1739‐1749, 2012.
 198. Eisenhofer G , Rundquist B , Aneman A , Friberg P , Dakak N , Kopin IJ , Jacobs MC , Lenders JW . Regional release and removal of catecholamines and extraneuronal metabolism to metanephrines. J Clin Endocrinol Metab 80: 3009‐3017, 1995.
 199. Elhamdani A , Azizi F , Artalejo CR . Double patch clamp reveals that transient fusion (kiss‐and‐run) is a major mechanism of secretion in calf adrenal chromaffin cells: High calcium shifts the mechanism from kiss‐and‐run to complete fusion. J Neurosci: Off J Soc Neurosci 26: 3030‐3036, 2006.
 200. Elhamdani A , Azizi F , Solomaha E , Palfrey HC , Artalejo CR . Two mechanistically distinct forms of endocytosis in adrenal chromaffin cells: Differential effects of SH3 domains and amphiphysin antagonism. FEBS Lett 580: 3263‐3269, 2006.
 201. Elmslie KS , Zhou W , Jones SW . LHRH and GTP‐gamma‐S modify calcium current activation in bullfrog sympathetic neurons. Neuron 5: 75‐80, 1990.
 202. Emery A , Eiden MV , Mustafa T , Eiden LE . GPCR‐Gs signaling to ERK is controlled by the cAMP‐sensing guanine nucleotide exchange factor NCS/Rapgef2 in neuronal and endocrine cells. Sci Signal 6: ra51, 2013.
 203. Emery AC , Eiden LE . Signaling through the neuropeptide GPCR PAC1 induces neuritogenesis via a single linear cAMP‐ and ERK‐dependent pathway using a novel cAMP sensor. FASEB J 26: 3199‐3211, 2012.
 204. Emery AC , Liu XH , Xu W , Eiden MV , Eiden LE . Cyclic adenosine 3′,5′‐monophosphate elevation and biological signaling through a secretin family Gs‐coupled G protein‐coupled receptor are restricted to a single adenylate cyclase isoform. Mol Pharmacol 87: 928‐935, 2015.
 205. Emery AC , Xu W , Eiden MV , Eiden LE . Guanine nucleotide exchange factor Epac2‐dependent activation of the GTP‐binding protein Rap2A mediates cAMP‐dependent growth arrest in neuroendocrine cells. J Biol Chem 292: 12220‐12231, 2017.
 206. Engisch KL , Nowycky MC . Calcium dependence of large dense‐cored vesicle exocytosis evoked by calcium influx in bovine adrenal chromaffin cells. J Neurosci: Off J Soc Neurosci 16: 1359‐1369, 1996.
 207. Engisch KL , Nowycky MC . Compensatory and excess retrieval: Two types of endocytosis following single step depolarizations in bovine adrenal chromaffin cells. J Physiol 506 (Pt 3): 591‐608, 1998.
 208. Erickson JD , Eiden LE , Hoffman B . Expression cloning of a reserpine‐sensitive vesicular monoamine transporter. Proc Natl Acad Sci USA 89: 10993‐10997, 1992.
 209. Erickson JD , Eiden LE , Schäfer MK‐H , Weihe E . Reserpine‐ and tetrabenazine‐sensitive transport of 3H‐histamine by the neuronal isoform of the vesicular monoamine transporter. J Mol Neurosci 6: 277‐287, 1995.
 210. Erickson JD , Schäfer MK‐H , Bonner TI , Eiden LE , Weihe E . Distinct pharmacological properties and distribution in neurons and endocrine cells of two isoforms of the human vesicular monoamine transporter. Proc Natl Acad Sci USA 93: 5166‐5171, 1996.
 211. Eskay RL , Eiden LE . Interleukin‐1 alpha and tumor necrosis factor alpha differentially regulate enkephalin, vasoactive intestinal polypeptide, neurotensin, and substance P biosynthesis in chromaffin cells. Endocrinology 130: 2252‐2258, 1992.
 212. Esler M , Rumantir M , Kaye D , Jennings G , Hastings J , Socratous F , Lambert G . Sympathetic nerve biology in essential hypertension. Clin Exp Pharmacol Physiol 28: 986‐989, 2001.
 213. Esquerro E , Garcia AG , Herandez M , Kirpekar SM , Prat JC . Catecholamine secretory response to calcium reintroduction in the perfused cat adrenal gland treated with ouabain. Biochem Pharmacol 29: 2669‐2673, 1980.
 214. Estevez‐Herrera J , Dominguez N , Pardo MR , Gonzalez‐Santana A , Westhead EW , Borges R , Machado JD . ATP: The crucial component of secretory vesicles. Proc Natl Acad Sci USA 113: E4098‐E4106, 2016.
 215. Estevez‐Herrera J , Gonzalez‐Santana A , Baz‐Davila R , Machado JD , Borges R . The intravesicular cocktail and its role in the regulation of exocytosis. J Neurochem 137: 897‐903, 2016.
 216. Evans RM , Zamponi GW . Presynaptic Ca2+ channels – integration centers for neuronal signaling pathways. Trends Neurosci 29: 617‐624, 2006.
 217. Evinger MJ , Ernsberger P , Regunathan S , Joh TH , Reis DJ . A single transmitter regulates gene expression through two separate mechanisms: Cholinergic regulation of phenylethanolamine N‐methyltransferase mRNA via nicotinic and muscarinic pathways. J Neurosci 14: 2106‐2116, 1994.
 218. Fakler B , Adelman JP . Control of K‐Ca channels by calcium nano/microdomains. Neuron 59: 873‐881, 2008.
 219. Farnsworth NL , Benninger RK . New insights into the role of connexins in pancreatic islet function and diabetes. FEBS Lett 588: 1278‐1287, 2014.
 220. Feldberg W , Gaddum JH . The chemical transmitter at synapses in a sympathetic ganglion. J Physiol 81: 305‐319, 1934.
 221. Fenwick EM , Marty A , Neher E . A patch‐clamp study of bovine chromaffin cells and of their sensitivity to acetylcholine. J Physiol 331: 577‐597, 1982.
 222. Fenwick EM , Marty A , Neher E . Sodium and calcium channels in bovine chromaffin cells. J Physiol 331: 599‐635, 1982.
 223. Fernandez‐Aguera MC , Gao L , Gonzalez‐Rodriguez P , Pintado CO , Arias‐Mayenco I , Garcia‐Flores P , Garcia‐Perganeda A , Pascual A , Ortega‐Saenz P , Lopez‐Barneo J . Oxygen sensing by arterial chemoreceptors depends on mitochondrial complex I signaling. Cell Metab 22: 825‐837, 2015.
 224. Fernandez‐Morales JC , Padin JF , Arranz‐Tagarro JA , Vestring S , Garcia AG , de Diego AM . Hypoxia‐elicited catecholamine release is controlled by L‐type as well as N/PQ types of calcium channels in rat embryo chromaffin cells. Am J Physiol Cell Physiol 307: C455‐C465, 2014.
 225. Feuerstein G , Gutman Y . Preferential secretion of adrenaline or noradrenaline by the cat adrenal in vivo in response to different stimuli. Br J Pharmacol 43: 764‐775, 1971.
 226. Finn JP, 3rd , Edwards RH . Individual residues contribute to multiple differences in ligand recognition between vesicular monoamine transporters 1 and 2. J Biol Chem 272: 16301‐16307, 1997.
 227. Fischer‐Colbrie R , Iacangelo A , Eiden LE . Neural and humoral factors separately regulate neuropeptide Y, enkephalin, and chromogranin A and B mRNA levels in rat adrenal medulla. Proc Natl Acad Sci USA 85: 3240‐3244, 1988.
 228. Folkow B , Von Euler US . Selective activation of noradrenaline and adrenaline producing cells in the cat's adrenal gland by hypothalamic stimulation. Circ Res 2: 191‐195, 1954.
 229. Foucart S , Nadeau R , Dechamplain J . Local modulation of adrenal catecholamines release by beta‐2 adrenoceptors in the anesthetized dog. Naunyn‐Schmiedeberg's Arch Pharmacol 337: 29‐34, 1988.
 230. Fox AP , Cahill AL , Currie KP , Grabner C , Harkins AB , Herring B , Hurley JH , Xie Z . N‐ and P/Q‐type Ca2+ channels in adrenal chromaffin cells. Acta Physiol (Oxf) 192: 247‐261, 2008.
 231. Fricker LD , Snyder SH . Enkephalin convertase: Purification and characterization of a specific enkephalin‐synthesizing carboxypeptidase localized to adrenal chromaffin granules. Proc Natl Acad Sci USA 79: 3886‐3890, 1982.
 232. Fuentealba J , Olivares R , Ales E , Tapia L , Rojo J , Arroyo G , Aldea M , Criado M , Gandia L , Garcia AG . A choline‐evoked [Ca2+]c signal causes catecholamine release and hyperpolarization of chromaffin cells. FASEB J 18: 1468‐1470, 2004.
 233. Fulop T , Doreian B , Smith C . Dynamin I plays dual roles in the activity‐dependent shift in exocytic mode in mouse adrenal chromaffin cells. Arch Biochem Biophys 477: 146‐154, 2008.
 234. Fumimura Y , Ikemura M , Saito Y , Sengoku R , Kanemaru K , Sawabe M , Arai T , Ito G , Iwatsubo T , Fukayama M , Mizusawa H , Murayama S . Analysis of the adrenal gland is useful for evaluating pathology of the peripheral autonomic nervous system in lewy body disease. J Neuropathol Exp Neurol 66: 354‐362, 2007.
 235. Gandia L , Garcia AG , Morad M . ATP modulation of calcium channels in chromaffin cells. J Physiol 470: 55‐72, 1993.
 236. Gandia L , Mayorgas I , Michelena P , Cuchillo I , de Pascual R , Abad F , Novalbos JM , Larranaga E , Garcia AG . Human adrenal chromaffin cell calcium channels: Drastic current facilitation in cell clusters, but not in isolated cells. Pflug Archiv: Eur J Physiol 436: 696‐704, 1998.
 237. Gandia L , Villarroya M , Lara B , Olmos V , Gilabert JA , Lopez MG , Martinez‐Sierra R , Borges R , Garcia AG . Otilonium: A potent blocker of neuronal nicotinic ACh receptors in bovine chromaffin cells. Br J Pharmacol 117: 463‐470, 1996.
 238. Gao L , Bonilla‐Henao V , Garcia‐Flores P , Arias‐Mayenco I , Ortega‐Saenz P , Lopez‐Barneo J . Gene expression analyses reveal metabolic specifications in acute O2‐sensing chemoreceptor cells. J Physiol 595: 6091‐6120, 2017.
 239. Garcia‐Fernandez M , Mejias R , Lopez‐Barneo J . Developmental changes of chromaffin cell secretory response to hypoxia studied in thin adrenal slices. Pflug Archiv: Eur J Physiol 454: 93‐100, 2007.
 240. Garcia AG , Garcia‐De‐Diego AM , Gandia L , Borges R , Garcia‐Sancho J . Calcium signaling and exocytosis in adrenal chromaffin cells. Physiol Rev 86: 1093‐1131, 2006.
 241. Garcia AG , Garcia‐Lopez E , Horga JF , Kirpekar SM , Montiel C , Sanchez‐Garcia P . Potentiation of K+‐evoked catecholamine release in the cat adrenal gland treated with ouabain. Br J Pharmacol 74: 673‐680, 1981.
 242. Garcia AG , Padin F , Fernandez‐Morales JC , Maroto M , Garcia‐Sancho J . Cytosolic organelles shape calcium signals and exo‐endocytotic responses of chromaffin cells. Cell Calcium 51: 309‐320, 2012.
 243. Garcia AG , Sala F , Reig JA , Viniegra S , Frias J , Fonteriz R , Gandia L . Dihydropyridine BAY‐K‐8644 activates chromaffin cell calcium channels. Nature 309: 69‐71, 1984.
 244. Gavello D , Vandael D , Gosso S , Carbone E , Carabelli V . Dual action of leptin on rest‐firing and stimulated catecholamine release via phosphoinositide 3‐kinase‐driven BK channel up‐regulation in mouse chromaffin cells. J Physiol 593: 4835‐4853, 2015.
 245. Geisow MJ , Childs J , Burgoyne RD . Cholinergic stimulation of chromaffin cells induces rapid coating of the plasma membrane. Eur J Cell Biol 38: 51‐56, 1985.
 246. Gengler S , Hamilton A , Holscher C . Synaptic plasticity in the hippocampus of a APP/PS1 mouse model of Alzheimer's disease is impaired in old but not young mice. PLoS One 5: e9764, 2010.
 247. Gerra G , Zaimovic A , Mascetti GG , Gardini S , Zambelli U , Timpano M , Raggi MA , Brambilla F . Neuroendocrine responses to experimentally‐induced psychological stress in healthy humans. Psychoneuroendocrinology 26: 91‐107, 2001.
 248. Giancippoli A , Novara M , de Luca A , Baldelli P , Marcantoni A , Carbone E , Carabelli V . Low‐threshold exocytosis induced by cAMP‐recruited Ca(V)3.2 (alpha(1H)) channels in rat chromaffin cells. Biophys J 90: 1830‐1841, 2006.
 249. Goldstein DS . Catecholamines and stress. Endocr Regul 37: 69‐80, 2003.
 250. Goldstein DS . Concepts of scientific integrative medicine applied to the physiology and pathophysiology of catecholamine systems. Compr Physiol 3: 1569‐1610, 2013.
 251. Goldstein DS . Plasma catecholamines and essential hypertension. An analytical review. Hypertension 5: 86‐99, 1983.
 252. Goldstein DS , Kopin IJ . Adrenomedullary, adrenocortical, and sympathoneural responses to stressors: A meta‐analysis. Endocr Regul 42: 111‐119, 2008.
 253. Goldstein DS , Sullivan P , Holmes C , Miller GW , Alter S , Strong R , Mash DC , Kopin IJ , Sharabi Y . Determinants of buildup of the toxic dopamine metabolite DOPAL in Parkinson's disease. J Neurochem 126: 591‐603, 2013.
 254. Gong LW , Hafez I , Alvarez de Toledo G , Lindau M . Secretory vesicles membrane area is regulated in tandem with quantal size in chromaffin cells. J Neurosci 23: 7917‐7921, 2003.
 255. Gonzalez‐Jamett AM , Momboisse F , Guerra MJ , Ory S , Baez‐Matus X , Barraza N , Calco V , Houy S , Couve E , Neely A , Martinez AD , Gasman S , Cardenas AM . Dynamin‐2 regulates fusion pore expansion and quantal release through a mechanism that involves actin dynamics in neuroendocrine chromaffin cells. PLoS One 8: e70638, 2013.
 256. Gonzalez‐Rubio JM , Garcia de Diego AM , Egea J , Olivares R , Rojo J , Gandia L , Garcia AG , Hernandez‐Guijo JM . Blockade of nicotinic receptors of bovine adrenal chromaffin cells by nanomolar concentrations of atropine. Eur J Pharmacol 535: 13‐24, 2006.
 257. Greene LA , Tischler AS . Establishment of a noradrenergic clonal line of rat adrenal pheochromocytoma cells which respond to nerve growth factor. Proc Natl Acad Sci USA 73: 2424‐2428, 1976.
 258. Greten‐Harrison B , Polydoro M , Morimoto‐Tomita M , Diao L , Williams AM , Nie EH , Makani S , Tian N , Castillo PE , Buchman VL , Chandra SS . αβγ‐Synuclein triple knockout mice reveal age‐dependent neuronal dysfunction. Proc Natl Acad Sci USA 107: 19573‐19578, 2010.
 259. Grynszpan‐Winograd O . Adrenaline and noradrenaline cells in the adrenal medulla of the hamster: A morphological study of their innervation. J Neurocytol 3: 341‐361, 1974.
 260. Guarina L , Vandael DH , Carabelli V , Carbone E . Low pHo boosts burst firing and catecholamine release by blocking TASK‐1 and BK channels while preserving Cav1 channels in mouse chromaffin cells. J Physiol 595: 2587‐2609, 2017.
 261. Guerineau NC . Gap junction communication between chromaffin cells: The hidden face of adrenal stimulus‐secretion coupling. Pflug Archiv: Eur J Physiol 470: 89‐96, 2018.
 262. Guerineau NC , Desarmenien MG , Carabelli V , Carbone E . Functional chromaffin cell plasticity in response to stress: Focus on nicotinic, gap junction, and voltage‐gated Ca2+ channels. J Mol Neurosci 48: 368‐386, 2012.
 263. Gullo F , Ales E , Rosati B , Lecchi M , Masi A , Guasti L , Cano‐Abad MF , Arcangeli A , Lopez MG , Wanke E . ERG K+ channel blockade enhances firing and epinephrine secretion in rat chromaffin cells: The missing link to LQT2‐related sudden death? FASEB J 17: 330‐332, 2003.
 264. Gunter TE , Pfeiffer DR . Mechanisms by which mitochondria transport calcium. Am J Physiol 258: C755‐C786, 1990.
 265. Guo X , Wakade AR . Differential secretion of catecholamines in response to peptidergic and cholinergic transmitters in rat adrenals. J Physiol 475: 539‐545, 1994.
 266. Gureviciene I , Ikonen S , Gurevicius K , Sarkaki A , van Groen T , Pussinen R , Ylinen A , Tanila H . Normal induction but accelerated decay of LTP in APP + PS1 transgenic mice. Neurobiol Dis 15: 188‐195, 2004.
 267. Gurney ME , Pu H , Chiu AY , Dal Canto MC , Polchow CY , Alexander DD , Caliendo J , Hentati A , Kwon YW , Deng HX , et al. Motor neuron degeneration in mice that express a human Cu. Zn superoxide dismutase mutation. Science 264: 1772‐1775, 1994.
 268. Guyton AC , Hall JE . The autonomic nervous system and the adrenal medulla. In: Textbook of Medical Physiology, 13th ed. New York: Saunders, 2016.
 269. Hamelink C , Tjurmina O , Damadzic R , Young WS , Weihe E , Lee H‐W , Eiden LE . Pituitary adenylate cyclase activating polypeptide is a sympathoadrenal neurotransmitter involved in catecholamine regulation and glucohomeostasis. Proc Natl Acad Sci USA 99: 461‐466, 2002.
 270. Hamelink C , Weihe E , Eiden LE . PACAP: an ‘emergency response’ co‐transmitter in the adrenal medulla. In: Vaudry H , Arimura A , editors. Pituitary Adenylate Cyclase‐Activating Polypeptide. Norwell, MA: Kluwer‐Academic Press, 2003, p. 227‐250.
 271. Hamill OP , Marty A , Neher E , Sakmann B , Sigworth FJ . Improved patch‐clamp techhniques for high‐resolution current recording from cells and cell‐free membrane patches. Pflug Arch 391: 85‐100, 1981.
 272. Hansson O , Guatteo E , Mercuri NB , Bernardi G , Li XJ , Castilho RF , Brundin P . Resistance to NMDA toxicity correlates with appearance of nuclear inclusions, behavioural deficits and changes in calcium homeostasis in mice transgenic for exon 1 of the huntington gene. Eur J Neurosci 14: 1492‐1504, 2001.
 273. Harada K , Matsuoka H , Fujihara H , Ueta Y , Yanagawa Y , Inoue M . GABA signaling and neuroactive steroids in adrenal medullary chromaffin cells. Front Cell Neurosci 10: 100, 2016.
 274. Harada K , Matsuoka H , Nakamura J , Fukuda M , Inoue M . Storage of GABA in chromaffin granules and not in synaptic‐like microvesicles in rat adrenal medullary cells. J Neurochem 114: 617‐626, 2010.
 275. Hardy J , Selkoe DJ . The amyloid hypothesis of Alzheimer's disease: Progress and problems on the road to therapeutics. Science 297: 353‐356, 2002.
 276. Haycock JW , Wakade AR . Activation and multiple‐site phosphorylation of tyrosine hydroxylase in perfused rat adrenal glands. J Neurochem 58: 57‐64, 1992.
 277. Heinemann C , von Ruden L , Chow RH , Neher E . A two‐step model of secretion control in neuroendocrine cells. Pflug Archiv: Eur J Physiol 424: 105‐112, 1993.
 278. Helan M , Aravamudan B , Hartman WR , Thompson MA , Johnson BD , Pabelick CM , Prakash YS . BDNF secretion by human pulmonary artery endothelial cells in response to hypoxia. J Mol Cell Cardiol 68: 89‐97, 2014.
 279. Helle KB . The chromogranin A‐derived peptides vasostatin‐I and catestatin as regulatory peptides for cardiovascular functions. Cardiovasc Res, 2009.
 280. Helle KB . The granin family of uniquely acidic proteins of the diffuse neuroendocrine system: Comparative and functional aspects. Biol Rev Camb Philos Soc 79: 769‐794, 2004.
 281. Helle KB , Angeletti RH . Chromogranin A: A multipurpose prohormone? Acta Physiol Scand 152: 1‐10, 1994.
 282. Helle KB , Reed RK , Pihl KE , Serck‐Hanssen G . Osmotic properties of the chromogranins and relation to osmotic pressure in catecholamine storage granules. Acta Physiol Scand 123: 21‐33, 1985.
 283. Helle KB , Serck‐Hanssen G . The adrenal medulla: A model for studies of hormonal and neuronal storage and release mechanisms. Mol Cell Biochem 6: 127‐146, 1975.
 284. Henkel AW , Almers W . Fast steps in exocytosis and endocytosis studied by capacitance measurements in endocrine cells. Curr Opin Neurobiol 6: 350‐357, 1996.
 285. Henkel AW , Meiri H , Horstmann H , Lindau M , Almers W . Rhythmic opening and closing of vesicles during constitutive exo‐ and endocytosis in chromaffin cells. Embo J 19: 84‐93, 2000.
 286. Henry JP , Botton D , Sagne C , Isambert MF , Desnos C , Blanchard V , Raisman‐Vozari R , Krejci E , Massoulie J , Gasnier B . Biochemistry and molecular biology of the vesicular monoamine transporter from chromaffin granules. J Exp Biol 196: 251‐262, 1994.
 287. Hernandez‐Cruz A , Sala F , Adams PR . Subcellular calcium transients visualized by confocal microscopy in a voltage‐clamped vertebrate neuron. Science 247: 858‐862, 1990.
 288. Hernandez‐Guijo JM , Carabelli V , Gandia L , Garcia AG , Carbone E . Voltage‐independent autocrine modulation of L‐type channels mediated by ATP, opioids and catecholamines in rat chromaffin cells. Eur J Neurosci 11: 3574‐3584, 1999.
 289. Hernandez‐Guijo JM , Gandia L , Cuchillo‐Ibanez I , Albillos A , Novalbos J , Gilsanz F , Larranaga E , de Pascual R , Abad F , Garcia AG . Altered regulation of calcium channels and exocytosis in single human pheochromocytoma cells. Pflug Archiv: Eur J Physiol 440: 253‐263, 2000.
 290. Hernandez‐Guijo JM , Gandia L , Lara B , Garcia AG . Autocrine/paracrine modulation of calcium channels in bovine chromaffin cells. Pflug Archiv: Eur J Physiol 437: 104‐113, 1998.
 291. Hernandez‐Guijo JM , Maneu‐Flores VE , Ruiz‐Nuno A , Villarroya M , Garcia AG , Gandia L . Calcium‐dependent inhibition of L, N, and P/Q Ca2+ channels in chromaffin cells: Role of mitochondria. J Neurosci 21: 2553‐2560, 2001.
 292. Hernandez A , Segura‐Chama P , Jimenez N , Garcia AG , Hernandez‐Guijo JM , Hernandez‐Cruz A . Modulation by endogenously released ATP and opioids of chromaffin cell calcium channels in mouse adrenal slices. Am J Physiol Cell Physiol 300: C610‐C623, 2011.
 293. Herrera A , Munoz P , Steinbusch HWM , Segura‐Aguilar J . Are dopamine oxidation metabolites involved in the loss of dopaminergic neurons in the nigrostriatal system in Parkinson's disease? ACS Chem Neurosci 8: 702‐711, 2017.
 294. Herrero CJ , Ales E , Pintado AJ , Lopez MG , Garcia‐Palomero E , Mahata SK , O'Connor DT , Garcia AG , Montiel C . Modulatory mechanism of the endogenous peptide catestatin on neuronal nicotinic acetylcholine receptors and exocytosis. J Neurosci: Off J Soc Neurosci 22: 377‐388, 2002.
 295. Herrington J , Park YB , Babcock DF , Hille B . Dominant role of mitochondria in clearance of large Ca2+ loads from rat adrenal chromaffin cells. Neuron 16: 219‐228, 1996.
 296. Hervonen A , Partanen S , Vaalasti A , Partanen M , Kanerva L , Alho H . The distribution and endocrine nature of the abdominal paraganglia of adult man. Am J Anat 153: 563‐572, 1978.
 297. Herzog RI , Cummins TR , Ghassemi F , Dib‐Hajj SD , Waxman SG . Distinct repriming and closed‐state inactivation kinetics of Nav1.6 and Nav1.7 sodium channels in mouse spinal sensory neurons. J Physiol 551: 741‐750, 2003.
 298. Hill J , Chan SA , Kuri B , Smith C . Pituitary adenylate cyclase‐activating peptide (PACAP) recruits low voltage‐activated T‐type calcium influx under acute sympathetic stimulation in mouse adrenal chromaffin cells. J Biol Chem 286: 42459‐42469, 2011.
 299. Hillarp NA . Isolation and some biochemical properties of the catechol amine granules in the cow adrenal medulla. Acta Physiol Scand 43: 82‐96, 1958.
 300. Hillarp NA , Hokfelt B . Evidence of adrenaline and noradrenaline in separate adrenal medullary cells. Acta Physiol Scand 30: 55‐68, 1953.
 301. Hohne‐Zell B , Gratzl M . Adrenal chromaffin cells contain functionally different SNAP‐25 monomers and SNAP‐25/syntaxin heterodimers. FEBS Lett 394: 109‐116, 1996.
 302. Holgert H , Holmberg K , Hannibal J , Fahrenkrug J , Brimijoin S , Hartman BK , Hökfelt T . PACAP in the adrenal gland – relationship with choline acetyltransferase, enkephalin and chromaffin cells and effects of immunological sympathectomy. NeuroReport 20: 297‐301, 1996.
 303. Holman ME , Coleman HA , Tonta MA , Parkington HC . Synaptic transmission from splanchnic nerves to the adrenal medulla of guinea‐pigs. J Physiol 478 (Pt 1): 115‐124, 1994.
 304. Holman ME , Tonta MA , Coleman HA , Parkington HC . Currents caused by the spontaneous release of quanta of acetylcholine onto chromaffin cells in guinea‐pig adrenal medulla. Neurosci Lett 184: 75‐78, 1995.
 305. Holz GG , Kang G , Harbeck M , Roe MW , Chepurny OG . Cell physiology of cAMP sensor Epac. J Physiol 577: 5‐15, 2006.
 306. Honigmann A , van den Bogaart G , Iraheta E , Risselada HJ , Milovanovic D , Mueller V , Mullar S , Diederichsen U , Fasshauer D , Grubmuller H , Hell SW , Eggeling C , Kuhnel K , Jahn R . Phosphatidylinositol 4,5‐bisphosphate clusters act as molecular beacons for vesicle recruitment. Nat Struct Mol Biol 20: 679‐686, 2013.
 307. Hook VY , Eiden LE , Brownstein MJ . A carboxypeptidase processing enzyme for enkephalin precursors. Nature 295: 341‐342, 1982.
 308. Horrigan FT , Bookman RJ . Releasable pools and the kinetics of exocytosis in adrenal chromaffin cells. Neuron 13: 1119‐1129, 1994.
 309. Hoshi T , Rothlein J , Smith SJ . Facilitation of Ca2+‐channel currents in bovine adrenal chromaffin cells. Proc Natl Acad Sci USA 81: 5871‐5875, 1984.
 310. Houchi H , Teraoka K , Oka M , Murakumo Y , Morita K . Substance P inhibits catecholamine biosynthesis stimulated by carbamylcholine in cultured adrenal chromaffin cells. Biochem Pharmacol 45: 1165‐1167, 1993.
 311. Houy S , Croise P , Gubar O , Chasserot‐Golaz S , Tryoen‐Toth P , Bailly Y , Ory S , Bader MF , Gasman S . Exocytosis and endocytosis in neuroendocrine cells: Inseparable membranes! Front Endocrinol 4: 135, 2013.
 312. Huttner WB , Gerdes HH , Rosa P . The granin (chromogranin/secretogranin) family. Trends Biochem Sci 16: 27‐30, 1991.
 313. Iacangelo AL , Eiden LE . Chromogranin A: Current status as a precursor for bioactive peptides and a granulogenic/sorting factor in the regulated secretory pathway. Regul Pept 58: 65‐88, 1995.
 314. Ikeda SR , Dunlap K . Voltage‐dependent modulation of N‐type calcium channels: Role of G protein subunits. Adv Second Messenger Phosphoprotein Res 33: 131‐151, 1999.
 315. Inoue M , Fujishiro N , Imanaga I . Hypoxia and cyanide induce depolarization and catecholamine release in dispersed guinea‐pig chromaffin cells. J Physiol 507 (Pt 3): 807‐818, 1998.
 316. Inoue M , Harada K , Matsuoka H , Sata T , Warashina A . Inhibition of TASK1‐like channels by muscarinic receptor stimulation in rat adrenal medullary cells. J Neurochem 106: 1804‐1814, 2008.
 317. Inoue M , Harada K , Matsuoka H , Warashina A . Paracrine role of GABA in adrenal chromaffin cells. Cell Mol Neurobiol 30: 1217‐1224, 2010.
 318. Ip NY , Perlman RL , Zigmond RE . Acute transsynaptic regulation of tyrosine 3‐monooxygenase activity in the rat superior cervical ganglion: Evidence for both cholinergic and noncholinergic mechanisms. Proc Natl Acad Sci USA 80: 2081‐2085, 1983.
 319. Ishii TM , Maylie J , Adelman JP . Determinants of apamin and d‐tubocurarine block in SK potassium channels. J Biol Chem 272: 23195‐23200, 1997.
 320. Islas‐Suarez L , Gomez‐Chavarin M , Drucker‐Colin R , Hernandez‐Cruz A . Properties of the sodium current in rat chromaffin cells exposed to nerve growth factor in vitro. J Neurophysiol 72: 1938‐1948, 1994.
 321. Ito Y , Fujimoto Y , Obara T . The role of epinephrine, norepinephrine, and dopamine in blood pressure disturbances in patients with pheochromocytoma. World J Surg 16: 759‐763; discussion 763‐754, 1992.
 322. Jackson LP , Kelly BT , McCoy AJ , Gaffry T , James LC , Collins BM , Honing S , Evans PR , Owen DJ . A large‐scale conformational change couples membrane recruitment to cargo binding in the AP2 clathrin adaptor complex. Cell 141: 1220‐1229, 2010.
 323. Jansen AS , Nguyen XV , Karpitskiy V , Mettenleiter TC , Loewy AD . Central command neurons of the sympathetic nervous system: Basis of the fight‐or‐flight response. Science 270: 644‐646, 1995.
 324. Jiang SZ , Xu W , Emery AC , Gerfen CR , Eiden MV , Eiden LE . NCS‐Rapgef2, the protein product of the neuronal Rapgef2 gene, is a specific activator of D1 dopamine receptor‐dependent ERK phosphorylation in mouse brain. eNeuro 4, 2017. DOI: 10.1523/ENEURO.0248‐17.2017.
 325. Jimenez RR , Lopez MG , Sancho C , Maroto R , Garcia AG . A component of the catecholamine secretory response in the bovine adrenal gland is resistant to dihydropyridines and omega‐conotoxin. Biochem Biophys Res Commun 191: 1278‐1283, 1993.
 326. Johns LM , Levitan ES , Shelden EA , Holz RW , Axelrod D . Restriction of secretory granule motion near the plasma membrane of chromaffin cells. J Cell Biol 153: 177‐190, 2001.
 327. Johnson MA , Rajan V , Miller CE , Wightman RM . Dopamine release is severely compromised in the R6/2 mouse model of Huntington's disease. J Neurochem 97: 737‐746, 2006.
 328. Johnson MA , Villanueva M , Haynes CL , Seipel AT , Buhler LA , Wightman RM . Catecholamine exocytosis is diminished in R6/2 Huntington's disease model mice. J Neurochem 103: 2102‐2110, 2007.
 329. Johnson RG , Carty SE , Scarpa A . Proton: Substrate stoichiometries during active transport of biogenic amines in chromaffin ghosts. J Biol Chem 256: 5773‐5780, 1981.
 330. Johnson RG , Pfister D , Carty SE , Scarpa A . Biological amine transport in chromaffin ghosts. Coupling to the transmembrane proton and potential gradients. J Biol Chem 254: 10963‐10972, 1979.
 331. Johnson RG , Scarpa A . Protonmotive force and catecholamine transport in isolated chromaffin granules. J Biol Chem 254: 3750‐3760, 1979.
 332. Johnson TS , Young JB , Landsberg L . Sympathoadrenal responses to acute and chronic hypoxia in the rat. J Clin Invest 71: 1263‐1272, 1983.
 333. Kahle KT , Rinehart J , Lifton RP . Phosphoregulation of the Na‐K‐2Cl and K‐Cl cotransporters by the WNK kinases. Biochim Biophys Acta 1802: 1150‐1158, 2010.
 334. Kajiwara R , Sand O , Kidokoro Y , Barish ME , Iijima T . Functional organization of chromaffin cells and cholinergic synaptic transmission in rat adrenal medulla. Jpn J Physiol 47: 449‐464, 1997.
 335. Kao LS , Cheung NS . Mechanism of calcium transport across the plasma membrane of bovine chromaffin cells. J Neurochem 54: 1972‐1979, 1990.
 336. Kataoka Y , Fujimoto M , Alho H , Guidotti A , Geffard M , Kelly GD , Hanbauer I . Intrinsic gamma aminobutyric acid receptors modulate the release of catecholamine from canine adrenal gland in situ. J Pharmacol Exp Ther 239: 584‐590, 1986.
 337. Katz B , Miledi R . Ionic requirements of synaptic transmitter release. Nature 215: 651, 1967.
 338. Katz B , Miledi R . A study of synaptic transmission in the absence of nerve impulses. J Physiol 192: 407‐436, 1967.
 339. Kawahara Y , Ito K , Sun H , Aizawa H , Kanazawa I , Kwak S . Glutamate receptors: RNA editing and death of motor neurons. Nature 427: 801, 2004.
 340. Kawaida M , Abe T , Nakanishi T , Miyahara Y , Yamagishi H , Sakamoto M , Yamada T . A case of Timothy syndrome with adrenal medullary dystrophy. Pathol Int 66: 587‐592, 2016.
 341. Kawasaki H , Springett GM , Mochizuki N , Toki S , Nakaya M , Matsuda M , Housman DE , Graybiel AM . A family of cAMP‐binding proteins that directly activate Rap1. Science 282: 2275‐2279, 1998.
 342. Kayaalp SO , McIsaac RJ . Muscarinic component of splanchnic‐adrenal transmission in the dog. Br J Pharmacol 36: 286‐293, 1969.
 343. Keating DJ , Rychkov GY , Adams MB , Holgert H , McMillen IC , Roberts ML . Opioid receptor stimulation suppresses the adrenal medulla hypoxic response in sheep by actions on Ca(2+) and K(+) channels. J Physiol 555: 489‐502, 2004.
 344. Keating DJ , Rychkov GY , Giacomin P , Roberts ML . Oxygen‐sensing pathway for SK channels in the ovine adrenal medulla. Clin Exp Pharmacol Physiol 32: 882‐887, 2005.
 345. Keating DJ , Rychkov GY , Roberts ML . The contribution of voltage‐gated Ca2+ currents to K+ channel activation during ovine adrenal chromaffin cell development. Int J Dev Neurosci 27: 357‐363, 2009.
 346. Keating DJ , Rychkov GY , Roberts ML . Oxygen sensitivity in the sheep adrenal medulla: Role of SK channels. Am J Physiol Cell Physiol 281: C1434‐C1441, 2001.
 347. Keiper M , Stope MB , Szatkowski D , Bohm A , Tysack K , Vom Dorp F , Saur O , Oude Weernink PA , Evellin S , Jakobs KH , Schmidt M . Epac‐ and Ca2+‐controlled activation of Ras and extracellular signal‐regulated kinases by Gs‐coupled receptors. J Biol Chem 279: 46497‐46508, 2004.
 348. Kennedy BP , Mahata SK , O'Connor DT , Ziegler MG . Mechanism of cardiovascular actions of the chromogranin A fragment catestatin in vivo. Peptides 19: 1241‐1248, 1998.
 349. Kidokoro Y , Miyazaki S , Ozawa S . Acetylcholine‐induced membrane depolarization and potential fluctuations in the rat adrenal chromaffin cell. J Physiol 324: 203‐220, 1982.
 350. Kidokoro Y , Ritchie AK . Chromaffin cell action potentials and their possible role in adrenaline secretion from rat adrenal medulla. J Physiol 307: 199‐216, 1980.
 351. Kim D , Kang D . Role of K(2)p channels in stimulus‐secretion coupling. Pflug Archiv: Eur J Physiol 467: 1001‐1011, 2015.
 352. Kim SJ , Lim W , Kim J . Contribution of L‐ and N‐type calcium currents to exocytosis in rat adrenal medullary chromaffin cells. Brain Res 675: 289‐296, 1995.
 353. Kim T , Tao‐Cheng J‐H , Eiden LE , Loh YP . Chromogranin A, an “on/off” switch controlling dense‐core secretory granule biogenesis. Cell 106: 499‐509, 2001.
 354. Kitamura K , Kangawa K , Kawamoto M , Ichiki Y , Nakamura S , Matsuo H , Eto T . Adrenomedullin: A novel hypotensive peptide isolated from human pheochromocytoma. Biochem Biophys Res Commun 192: 553‐560, 1993.
 355. Kleppisch T , Ahnert‐Hilger G , Gollasch M , Spicher K , Hescheler J , Schultz G , Rosenthal W . Inhibition of voltage‐dependent Ca2+ channels via alpha 2‐adrenergic and opioid receptors in cultured bovine adrenal chromaffin cells. Pflug Archiv: Eur J Physiol 421: 131‐137, 1992.
 356. Klevans LR , Gebber GL . Comparison of differential secretion of adrenal catecholamines by splanchnic nerve stimulation and cholinergic agents. J Pharmacol Exp Therap 172: 69‐76, 1970.
 357. Klingauf J , Neher E . Modeling buffered Ca2+ diffusion near the membrane: Implications for secretion in neuroendocrine cells. Biophys J 72: 674‐690, 1997.
 358. Klugbauer N , Lacinova L , Flockerzi V , Hofmann F . Structure and functional expression of a new member of the tetrodotoxin‐sensitive voltage‐activated sodium channel family from human neuroendocrine cells. EMBO J 14: 1084‐1090, 1995.
 359. Knight DE . Botulinum toxin types A, B and D inhibit catecholamine secretion from bovine adrenal medullary cells. FEBS Lett 207: 222‐226, 1986.
 360. Knight DE , Baker PF . Calcium‐dependence of catecholamine release from bovine adrenal medullary cells after exposure to intense electric fields. J Membr Biol 68: 107‐140, 1982.
 361. Kopell WN , Westhead EW . Osmotic pressures of solutions of ATP and catecholamines relating to storage in chromaffin granules. J Biol Chem 257: 5707‐5710, 1982.
 362. Koschak A , Reimer D , Huber I , Grabner M , Glossmann H , Engel J , Striessnig J . alpha 1D (Cav1.3) subunits can form L‐type Ca2+ channels activating at negative voltages. J Biol Chem 276: 22100‐22106, 2001.
 363. Koseoglu S , Love SA , Haynes CL . Cholesterol effects on vesicle pools in chromaffin cells revealed by carbon‐fiber microelectrode amperometry. Anal Bioanal Chem 400: 2963‐2971, 2011.
 364. Kovacic B , Robinson RL . Drug‐induced secretion of catecholamines by the perfused adrenal gland of the dog during nicotine blockade. J Pharmacol Exp Therap 175: 178‐182, 1970.
 365. Kremer HP , Roos RA , Dingjan G , Marani E , Bots GT . Atrophy of the hypothalamic lateral tuberal nucleus in Huntington's disease. J Neuropathol Exp Neurol 49: 371‐382, 1990.
 366. Kummer W , Neuhuber WL . Vagal paraganglia of the rat. J Electr Microsc Techn 12: 343‐355, 1989.
 367. Kuo JF , Greengard P . Cyclic nucleotide‐dependent protein kinases. IV. Widespread occurrence of adenosine 3′,5′‐monophosphate‐dependent protein kinase in various tissues and phyla of the animal kingdom. Proc Natl Acad Sci USA 64: 1349‐1355, 1969.
 368. Kuri BA , Chan SA , Smith CB . PACAP regulates immediate catecholamine release from adrenal chromaffin cells in an activity‐dependent manner through a protein kinase C‐dependent pathway. J Neurochem 110: 1214‐1225, 2009.
 369. Lang T , Wacker I , Wunderlich I , Rohrbach A , Giese G , Soldati T , Almers W . Role of actin cortex in the subplasmalemmal transport of secretory granules in PC‐12 cells. Biophys J 78: 2863‐2877, 2000.
 370. Lara B , Lopez MG , Villarroya M , Gandia L , Cleeman L , Morad M , Garcia AG . A caffeine‐sensitive Ca2+ store modulates K+‐evoked secretion in chromaffin cells. Am J Physiol 272: C1211‐C1221, 1997.
 371. Lara B , Zapater P , Montiel C , de la Fuente MT , Martinez‐Sierra R , Ballesta JJ , Gandia L , Garcia AG . Density of apamin‐sensitive Ca(2+)‐dependent K+ channels in bovine chromaffin cells: Relevance to secretion. Biochem Pharmacol 49: 1459‐1468, 1995.
 372. Larsen KE , Schmitz Y , Troyer MD , Mosharov E , Dietrich P , Quazi AZ , Savalle M , Nemani V , Chaudhry FA , Edwards RH , Stefanis L , Sulzer D . Alpha‐synuclein overexpression in PC12 and chromaffin cells impairs catecholamine release by interfering with a late step in exocytosis. J Neurosci 26: 11915‐11922, 2006.
 373. Lazo‐Fernandez Y , Aguilera G , Pham TD , Park AY , Beierwaltes WH , Sutliff RL , Verlander JW , Pacak K , Osunkoya AO , Ellis CL , Kim YH , Shipley GL , Wynne BM , Hoover RS , Sen SK , Plotsky PM , Wall SM . Pendrin localizes to the adrenal medulla and modulates catecholamine release. Am J Physiol Endocrinol Metab 309: E534‐E545, 2015.
 374. Lee FL , Trendelenburg U . Muscarinic transmission of preganglionic impulses to the adrenal medulla of the cat. J Pharmacol Exp Therap 158: 73‐79, 1967.
 375. Lee J , Lim W , Eun SY , Kim SJ , Kim J . Inhibition of apamin‐sensitive K+ current by hypoxia in adult rat adrenal chromaffin cells. Pflug Archiv: Eur J Physiol 439: 700‐704, 2000.
 376. Lee RM , Borkowski KR , Leenen FH , Tsoporis J , Coughlin M . Combined effect of neonatal sympathectomy and adrenal demedullation on blood pressure and vascular changes in spontaneously hypertensive rats. Circulat Res 69: 714‐721, 1991.
 377. Lembo PM , Grazzini E , Groblewski T , O'Donnell D , Roy MO , Zhang J , Hoffert C , Cao J , Schmidt R , Pelletier M , Labarre M , Gosselin M , Fortin Y , Banville D , Shen SH , Strom P , Payza K , Dray A , Walker P , Ahmad S . Proenkephalin A gene products activate a new family of sensory neuron – specific GPCRs. Nat Neurosci 5: 201‐209, 2002.
 378. Lenders JW , Eisenhofer G . Pathophysiology and diagnosis of disorders of the adrenal medulla: Focus on pheochromocytoma. Compr Physiol 4: 691‐713, 2014.
 379. Levitsky KL , Lopez‐Barneo J . Developmental change of T‐type Ca2+ channel expression and its role in rat chromaffin cell responsiveness to acute hypoxia. J Physiol 587: 1917‐1929, 2009.
 380. Lewis RV , Stern AS , Kilpatrick DL , Gerber LD , Rossier J , Stern S , Udenfriend S . Marked increases in large enkephalin‐containing polypeptides in the rat adrenal gland following denervation. J Neurosci 1: 80‐82, 1981.
 381. Lewis RV , Stern AS , Kimura S , Rossier J , Stein S , Udenfriend S . An about 50,000‐dalton protein in adrenal medulla: A common precursor of [Met]‐ and [Leu]enkephalin. Science 208: 1459‐1461, 1980.
 382. Li H , Li SH , Cheng AL , Mangiarini L , Bates GP , Li XJ . Ultrastructural localization and progressive formation of neuropil aggregates in Huntington's disease transgenic mice. Hum Mol Genet 8: 1227‐1236, 1999.
 383. Li H , Wyman T , Yu ZX , Li SH , Li XJ . Abnormal association of mutant huntingtin with synaptic vesicles inhibits glutamate release. Hum Mol Genet 12: 2021‐2030, 2003.
 384. Liao M , Shen J , Zhang Y , Li SH , Li XJ , Li H . Immunohistochemical localization of huntingtin‐associated protein 1 in endocrine system of the rat. J Histochem Cytochem 53: 1517‐1524, 2005.
 385. Lim DY , Jang SJ , Park DG . Comparison of catecholamine release in the isolated adrenal glands of SHR and WKY rats. Auton Autacoid Pharmacol 22: 225‐232, 2002.
 386. Lindau M . Time‐resolved capacitance measurements: Monitoring exocytosis in single cells. Q Rev Biophys 24: 75‐101, 1991.
 387. Lindau M , Neher E . Patch‐clamp techniques for time‐resolved capacitance measurements in single cells. Pflug Arch 411: 137‐146, 1988.
 388. Lindgren P , Rosen A , Uvnas B . The release of catechols from the adrenal medulla on activation of the bulbar part of the sympathetic vasodilator outflow in cats. Acta Physiol Scand 47: 233‐242, 1959.
 389. Lingle CJ , Martinez‐Espinosa PL , Guarina L , Carbone E . Roles of Na(+), Ca(2+), and K(+) channels in the generation of repetitive firing and rhythmic bursting in adrenal chromaffin cells. Pflug Archiv: Eur J Physiol 470: 39‐52, 2018.
 390. Lingle CJ , Solaro CR , Prakriya M , Ding JP . Calcium‐activated potassium channels in adrenal chromaffin cells. Ion Channels 4: 261‐301, 1996.
 391. Liu PS , Kao LS . Na(+)‐dependent Ca2+ influx in bovine adrenal chromaffin cells. Cell Calcium 11: 573‐579, 1990.
 392. Liu PS , Lin YJ , Kao LS . Caffeine‐sensitive calcium stores in bovine adrenal chromaffin cells. J Neurochem 56: 172‐177, 1991.
 393. Liu Y , Peter D , Roghani A , Schuldiner S , Prive GG , Eisenberg D , Brecha N , Edwards RH . A cDNA that suppresses MPP+ toxicity encodes a vesicular amine transporter. Cell 70: 539‐551, 1992.
 394. Livett BG . Chromaffin cells: Model cells for neuronal cell biology. In: Squire LR , editor. Encyclopedia of Neuroscience. Burlington, MA: Elsevier, vol. 2, 2009.
 395. Livett BG , Dean DM , Whelan LG , Udenfriend S , Rossier J . Co‐release of enkephalin and catecholamines from cultured chromaffin cells. Nature 289: 317‐319, 1981.
 396. Livett BG , Kozousek V , Mizobe F , Dean DM . Substance P inhibits nicotinic activation of chromaffin cells. Nature 278: 256‐257, 1979.
 397. Livett BG , Marley PD . Effects of opioid peptides and morphine on histamine‐induced catecholamine secretion from cultured, bovine adrenal chromaffin cells. Br J Pharmacol 89: 327‐334, 1986.
 398. Llinas R , Steinberg IZ , Walton K . Relationship between presynaptic calcium current and postsynaptic potential in squid giant synapse. Biophys J 33: 323‐351, 1981.
 399. Logan T , Bendor J , Toupin C , Thorn K , Edwards RH . alpha‐Synuclein promotes dilation of the exocytotic fusion pore. Nat Neurosci 20: 681‐689, 2017.
 400. Lomax RB , Michelena P , Nunez L , Garcia‐Sancho J , Garcia AG , Montiel C . Different contributions of L‐ and Q‐type Ca2+ channels to Ca2+ signals and secretion in chromaffin cell subtypes. Am J Physiol 272: C476‐C484, 1997.
 401. Lopez‐Barneo J , Gonzalez‐Rodriguez P , Gao L , Fernandez‐Aguera MC , Pardal R , Ortega‐Saenz P . Oxygen sensing by the carotid body: Mechanisms and role in adaptation to hypoxia. Am J Physiol Cell Physiol 310: C629‐C642, 2016.
 402. Lopez‐Gil A , Nanclares C , Mendez‐Lopez I , Martinez‐Ramirez C , de Los RC , Padin‐Nogueira JF , Montero M , Gandia L , Garcia AG . The quantal catecholamine release from mouse chromaffin cells challenged with repeated ACh pulses is regulated by the mitochondrial Na(+)/Ca(2+) exchanger. J Physiol 595: 2129‐2146, 2017.
 403. Lopez De Jesus M , Stope MB , Oude Weernink PA , Mahlke Y , Borgermann C , Ananaba VN , Rimmbach C , Rosskopf D , Michel MC , Jakobs KH , Schmidt M . Cyclic AMP‐dependent and Epac‐mediated activation of R‐Ras by G protein‐coupled receptors leads to phospholipase D stimulation. J Biol Chem 281: 21837‐21847, 2006.
 404. Lopez MG , Albillos A , de la Fuente MT , Borges R , Gandia L , Carbone E , Garcia AG , Artalejo AR . Localized L‐type calcium channels control exocytosis in cat chromaffin cells. Pflug Archiv: Eur J Physiol 427: 348‐354, 1994.
 405. Lopez MG , Montiel C , Herrero CJ , Garcia‐Palomero E , Mayorgas I , Hernandez‐Guijo JM , Villarroya M , Olivares R , Gandia L , McIntosh JM , Olivera BM , Garcia AG . Unmasking the functions of the chromaffin cell alpha7 nicotinic receptor by using short pulses of acetylcholine and selective blockers. Proc Natl Acad Sci USA 95: 14184‐14189, 1998.
 406. Lopez MG , Villarroya M , Lara B , Martinez Sierra R , Albillos A , Garcia AG , Gandia L . Q‐ and L‐type Ca2+ channels dominate the control of secretion in bovine chromaffin cells. FEBS Lett 349: 331‐337, 1994.
 407. Lou XL , Yu X , Chen XK , Duan KL , He LM , Qu AL , Xu T , Zhou Z . Na+ channel inactivation: A comparative study between pancreatic islet beta‐cells and adrenal chromaffin cells in rat. J Physiol 548: 191‐202, 2003.
 408. Lovell PV , James DG , McCobb DP . Bovine versus rat adrenal chromaffin cells: Big differences in BK potassium channel properties. J Neurophysiol 83: 3277‐3286, 2000.
 409. Lu BX , Su YH , Das S , Liu J , Xia JS , Ren DJ . The neuronal channel NALCN contributes resting sodium permeability and is required for normal respiratory rhythm. Cell 129: 371‐383, 2007.
 410. Lugnier C . Cyclic nucleotide phosphodiesterase (PDE) superfamily: A new target for the development of specific therapeutic agents. Pharmacol Ther 109: 366‐398, 2006.
 411. Lukyanetz EA , Neher E . Different types of calcium channels and secretion from bovine chromaffin cells. Eur J Neurosci 11: 2865‐2873, 1999.
 412. Machado JD , Montesinos MS , Borges R . Good practices in single cell amperometry. Methods Mol Biol 440: 297‐313, 2008.
 413. Machado JD , Morales A , Gomez JF , Borges R . cAMP modulates exocytotic kinetics and increases quantal size in chromaffin cells. Mol Pharmacol 60: 514‐520, 2001.
 414. Mackenzie KD , Duffield MD , Peiris H , Phillips L , Zanin MP , Teo EH , Zhou XF , Keating DJ . Huntingtin‐associated protein 1 regulates exocytosis, vesicle docking, readily releasable pool size and fusion pore stability in mouse chromaffin cells. J Physiol 592: 1505‐1518, 2014.
 415. Mackenzie KD , Lim Y , Duffield MD , Chataway T , Zhou XF , Keating DJ . Huntingtin‐associated protein‐1 (HAP1) regulates endocytosis and interacts with multiple trafficking‐related proteins. Cell Signal 35: 176‐187, 2017.
 416. Mahapatra NR , O'Connor DT , Vaingankar SM , Hikim AP , Mahata M , Ray S , Staite E , Wu H , Gu Y , Dalton N , Kennedy BP , Ziegler MG , Ross J , Mahata SK . Hypertension from targeted ablation of chromogranin A can be rescued by the human ortholog. J Clin Invest 115: 1942‐1952, 2005.
 417. Mahapatra S , Calorio C , Vandael DHF , Marcantoni A , Carabelli V , Carbone E . Calcium channel types contributing to chromaffin cell excitability, exocytosis and endocytosis. Cell Calcium 51: 321‐330, 2012.
 418. Mahapatra S , Marcantoni A , Vandael DHF , Striessnig J , Carbone E . Are Ca(v)1.3 pacemaker channels in chromaffin cells? Possible bias from resting cell conditions and DHP blockers usage. Channels 5: 219‐224, 2011.
 419. Mahapatra S , Marcantoni A , Zuccotti A , Carabelli V , Carbone E . Equal sensitivity of Cav1.2 and Cav1.3 channels to the opposing modulations of PKA and PKG in mouse chromaffin cells. J Physiol‐Lond 590: 5053‐5073, 2012.
 420. Mahata SK , Mahata M , Parmer RJ , O'Connor DT . Desensitizaton of catecholamine release. The novel catecholammine release‐inhibitory peptide catestatin (chromogranin A344‐364) acts at the receptor to prevent nicotinic cholinergic tolerance. J Biol Chem 274: 2920‐2928, 1999.
 421. Majdi S , Larsson A , Najafinobar N , Borges R , Ewing AG . Extracellular ATP regulates the vesicular pore opening in chromaffin cells and increases the fraction released during individual exocytosis events. ACS Chem Neurosci 10 (5): 2459‐2466, 2019.
 422. Malhotra RK , Wakade AR . Non‐cholinergic component of rat splanchnic nerves predominates at low neuronal activity and is eliminated by naloxone. J Physiol 383: 639‐652, 1987.
 423. Man KN , Imig C , Walter AM , Pinheiro PS , Stevens DR , Rettig J , Sorensen JB , Cooper BH , Brose N , Wojcik SM . Identification of a Munc13‐sensitive step in chromaffin cell large dense‐core vesicle exocytosis. eLife 4, 2015. DOI: 10.7554/eLife.10635.
 424. Manger WM , Eisenhofer G . Pheochromocytoma: Diagnosis and management update. Curr Hypertension Rep 6: 477‐484, 2004.
 425. Mangiarini L , Sathasivam K , Seller M , Cozens B , Harper A , Hetherington C , Lawton M , Trottier Y , Lehrach H , Davies SW , Bates GP . Exon 1 of the HD gene with an expanded CAG repeat is sufficient to cause a progressive neurological phenotype in transgenic mice. Cell 87: 493‐506, 1996.
 426. Marcantoni A , Baldelli P , Hernandez‐Guijo JM , Comunanza V , Carabelli V , Carbone E . L‐type calcium channels in adrenal chromaffin cells: Role in pace‐making and secretion. Cell Calcium 42: 397‐408, 2007.
 427. Marcantoni A , Carabelli V , Comunanza V , Hoddah H , Carbone E . Calcium channels in chromaffin cells: Focus on L and T types. Acta Physiol 192: 233‐246, 2008.
 428. Marcantoni A , Carabelli V , Vandael DH , Comunanza V , Carbone E . PDE type‐4 inhibition increases L‐type Ca2+ currents, action potential firing, and quantal size of exocytosis in mouse chromaffin cells. Pflug Archiv: Eur J Physiol 457: 1093‐1110, 2009.
 429. Marcantoni A , Vandael DHF , Mahapatra S , Carabelli V , Sinnegger‐Brauns MJ , Striessnig J , Carbone E . Loss of Cav1.3 channels reveals the critical role of L‐type and BK channel coupling in pacemaking mouse adrenal chromaffin cells. J Neurosci 30: 491‐504, 2010.
 430. Marchetti C , Carbone E , Lux HD . Effects of dopamine and noradrenaline on Ca channels of cultured sensory and sympathetic neurons of chick. Pflug Archiv: Eur J Physiol 406: 104‐111, 1986.
 431. Marley PD . Mechanisms in histamine‐mediated secretion from adrenal chromaffin cells. Pharmacol Ther 98: 1‐34, 2003.
 432. Marshall JM . Peripheral chemoreceptors and cardiovascular regulation. Physiol Rev 74: 543‐594, 1994.
 433. Martin AO , Mathieu MN , Chevillard C , Guerineau NC . Gap junctions mediate electrical signaling and ensuing cytosolic Ca2+ increases between chromaffin cells in adrenal slices: A role in catecholamine release. J Neurosci 21: 5397‐5405, 2001.
 434. Martinez‐Espinosa PL , Yang C , Gonzalez‐Perez V , Xia XM , Lingle CJ . Knockout of the BK beta2 subunit abolishes inactivation of BK currents in mouse adrenal chromaffin cells and results in slow‐wave burst activity. J Gen Physiol 144: 275‐295, 2014.
 435. Martinez‐Ramirez C , Baraibar AM , Nanclares C , Mendez‐Lopez I , Gomez A , Munoz MP , de Diego AMG , Gandia L , Casarejos MJ , Garcia AG . Altered excitability and exocytosis in chromaffin cells from the R6/1 mouse model of Huntington's disease is linked to over‐expression of mutated huntingtin. J Neurochem, 2018.
 436. Marty A , Neher E . Potassium channels in cultured bovine adrenal chromaffin cells. J Physiol 367: 117‐141, 1985.
 437. Mascorro JA , Yates RD , Chen IL . A glutaraldehyde/potassium dichromate tracing method for the localization and preservation of abdominal extra‐adrenal chromaffin tissues. Stain Technol 50: 391‐396, 1975.
 438. Matsui H . Effect of myelencephalic stimulation on the secretion of noradrenaline and adrenaline of the adrenal gland in the cat. Tohoku J Exp Med 87: 332‐337, 1965.
 439. Matsuoka H , Harada K , Endo Y , Warashina A , Doi Y , Nakamura J , Inoue M . Molecular mechanisms supporting a paracrine role of GABA in rat adrenal medullary cells. J Physiol 586: 4825‐4842, 2008.
 440. May V , Beaudet MM , Parsons RL , Hardwick JC , Gauthier EA , Durda JP , Braas KM . Mechanisms of pituitary adenylate cyclase activating polypeptide (PACAP)‐induced depolarization of sympathetic superior cervical ganglion (SCG) neurons. Ann N Y Acad Sci 865: 164‐175, 1998.
 441. Medbo JI , Sejersted OM . Plasma potassium changes with high‐intensity exercise. J Physiol‐Lond 421: 105‐122, 1990.
 442. Michelena P , Vega T , Montiel C , Lopez MG , Garcia‐Perez LE , Gandia L , Garcia AG . Effects of tyramine and calcium on the kinetics of secretion in intact and electroporated chromaffin cells superfused at high speed. Pflug Archiv: EurJ Physiol 431: 283‐296, 1995.
 443. Milla J , Montesinos MS , Machado JD , Borges R , Alonso E , Moreno‐Ortega AJ , Cano‐Abad MF , Garcia AG , Ruiz‐Nuno A . Ouabain enhances exocytosis through the regulation of calcium handling by the endoplasmic reticulum of chromaffin cells. Cell Calcium 50: 332‐342, 2011.
 444. Miranda‐Ferreira R , de Pascual R , Caricati‐Neto A , Gandia L , Jurkiewicz A , Garcia AG . Role of the endoplasmic reticulum and mitochondria on quantal catecholamine release from chromaffin cells of control and hypertensive rats. J Pharmacol Exp Therap 329: 231‐240, 2009.
 445. Miranda‐Ferreira R , de Pascual R , de Diego AM , Caricati‐Neto A , Gandia L , Jurkiewicz A , Garcia AG . Single‐vesicle catecholamine release has greater quantal content and faster kinetics in chromaffin cells from hypertensive, as compared with normotensive, rats. J Pharmacol Exp Therap 324: 685‐693, 2008.
 446. Miranda‐Ferreira R , de Pascual R , Smaili SS , Caricati‐Neto A , Gandia L , Garcia AG , Jurkiewicz A . Greater cytosolic and mitochondrial calcium transients in adrenal medullary slices of hypertensive, compared with normotensive rats. Eur J Pharmacol 636: 126‐136, 2010.
 447. Miyata A , Arimura A , Dahl RR , Minamino N , Uehara A , Jiang L , Culler MD , Coy DH . Isolation of a novel 38 residue‐hypothalamic polypeptide which stimulates adenylate cyclase in pituitary cells. Biochem Biophys Res Commun 164: 567‐574, 1989.
 448. Mizuno K , Minamino N , Kangawa K , Matsuo H . A new family of endogenous “big” Met‐enkephalins from bovine adrenal medulla: Purification and structure of docosa‐ (BAM‐22P) and eicosapeptide (BAM‐20P) with very potent opiate activity. Biochem Biophys Res Commun 97: 1283‐1290, 1980.
 449. Mochizuki‐Oda N , Takeuchi Y , Matsumura K , Oosawa Y , Watanabe Y . Hypoxia‐induced catecholamine release and intracellular Ca2+ increase via suppression of K+ channels in cultured rat adrenal chromaffin cells. J Neurochem 69: 377‐387, 1997.
 450. Mojet MH , Mills E , Duchen MR . Hypoxia‐induced catecholamine secretion in isolated newborn rat adrenal chromaffin cells is mimicked by inhibition of mitochondrial respiration. J Physiol 504 (Pt 1): 175‐189, 1997.
 451. Mollard P , Seward EP , Nowycky MC . Activation of nicotinic receptors triggers exocytosis from bovine chromaffin cells in the absence of membrane depolarization. Proc Natl Acad Sci USA 92: 3065‐3069, 1995.
 452. Montero M , Alonso MT , Albillos A , Cuchillo‐Ibanez I , Olivares R , Garcia AG , Garcia‐Sancho J , Alvarez J . Control of secretion by mitochondria depends on the size of the local [Ca2+] after chromaffin cell stimulation. Eur J Neurosci 13: 2247‐2254, 2001.
 453. Montero M , Alonso MT , Carnicero E , Cuchillo‐Ibanez I , Albillos A , Garcia AG , Garcia‐Sancho J , Alvarez J . Chromaffin‐cell stimulation triggers fast millimolar mitochondrial Ca2+ transients that modulate secretion. Nat Cell Biol 2: 57‐61, 2000.
 454. Montesinos MS , Camacho M , Machado JD , Viveros OH , Beltran B , Borges R . The quantal secretion of catecholamines is impaired by the accumulation of beta‐adrenoceptor antagonists into chromaffin cell vesicles. Br J Pharmacol 159: 1548‐1556, 2010.
 455. Montesinos MS , Machado JD , Camacho M , Diaz J , Morales YG , Alvarez de la Rosa D , Carmona E , Castaneyra A , Viveros OH , O'Connor DT , Mahata SK , Borges R . The crucial role of chromogranins in storage and exocytosis revealed using chromaffin cells from chromogranin A null mouse. J Neurosci:Off J Soc Neurosci 28: 3350‐3358, 2008.
 456. Moro MA , Garcia AG , Langley OK . Characterization of two chromaffin cell populations isolated from bovine adrenal medulla. J Neurochem 57: 363‐369, 1991.
 457. Morrison SF , Cao WH . Different adrenal sympathetic preganglionic neurons regulate epinephrine and norepinephrine secretion. Am J Physiol Regul, Integr Comp Physiol 279: R1763‐R1775, 2000.
 458. Mosharov EV , Gong LW , Khanna B , Sulzer D , Lindau M . Intracellular patch electrochemistry: Regulation of cytosolic catecholamines in chromaffin cells. J Neurosci: Off J Soc Neurosci 23: 5835‐5845, 2003.
 459. Muller L , Lindberg I . The cell biology of the prohormone convertases PC1 and PC2. Progr Nucl Acid Res Mol Biol 63: 69‐108, 1999.
 460. Musial DC , Bomfim GH , Arranz‐Tagarro JA , Mendez‐Lopez I , Miranda‐Ferreira R , Jurkiewicz A , Jurkiewicz NH , Garcia AG , Padin JF . Altered mitochondrial function, calcium signaling, and catecholamine release in chromaffin cells of diabetic and SHR rats. Eur J Pharmacol 815: 416‐426, 2017.
 461. Nagayama T , Matsumoto T , Kuwakubo F , Fukushima Y , Yoshida M , Suzuki‐Kusaba M , Hisa H , Kimura T , Satoh S . Role of calcium channels in catecholamine secretion in the rat adrenal gland. J Physiol 520 (Pt 2): 503‐512, 1999.
 462. Nakada T , Furuta H , Katayama T . Catecholamine metabolism in pheochromocytoma and normal adrenal medullae. J Urol 140: 1348‐1351, 1988.
 463. Nanclares C , Baraibar AM , Gandia L . L‐type calcium channels in exocytosis and endocytosis of chromaffin cells. Pflug Archiv: Eur J Physiol 470: 53‐60, 2018.
 464. Nassar‐Gentina V , Pollard HB , Rojas E . Electrical activity in chromaffin cells of intact mouse adrenal gland. Am J Physiol 254: C675‐C683, 1988.
 465. Neale EA , Bowers LM , Jia M , Bateman KE , Williamson LC . Botulinum neurotoxin A blocks synaptic vesicle exocytosis but not endocytosis at the nerve terminal. J Cell Biol 147: 1249‐1260, 1999.
 466. Neco P , Giner D , Viniegra S , Borges R , Villarroel A , Gutierrez LM . New roles of myosin II during the vesicle transport and fusion in chromaffin cells. J Biol Chem, 2004.
 467. Neely A , Lingle CJ . Effects of muscarine on single‐rat adrenal chromaffin cells. J Physiol‐Lond 453: 133‐166, 1992.
 468. Neely A , Lingle CJ . Two components of calcium‐activated potassium current in rat adrenal chromaffin cells. J Physiol 453: 97‐131, 1992.
 469. Neher E . A comparison between exocytic control mechanisms in adrenal chromaffin cells and a glutamatergic synapse. Pflug Archiv: Eur J Physiol 453: 261‐268, 2006.
 470. Neher E . Neurosecretion: What can we learn from chromaffin cells. Pflug Archiv: Eur J Physiol 470: 7‐11, 2018.
 471. Neher E . Usefulness and limitations of linear approximations to the understanding of Ca++ signals. Cell Calcium 24: 345‐357, 1998.
 472. Neher E . Vesicle pools and Ca2+ microdomains: New tools for understanding their roles in neurotransmitter release. Neuron 20: 389‐399, 1998.
 473. Neher E , Augustine GJ . Calcium gradients and buffers in bovine chromaffin cells. J Physiol 450: 273‐301, 1992.
 474. Nelson O , Tu H , Lei T , Bentahir M , de Strooper B , Bezprozvanny I . Familial Alzheimer disease‐linked mutations specifically disrupt Ca2+ leak function of presenilin 1. J Clin Invest 117: 1230‐1239, 2007.
 475. Nemani VM , Lu W , Berge V , Nakamura K , Onoa B , Lee MK , Chaudhry FA , Nicoll RA , Edwards RH . Increased expression of alpha‐synuclein reduces neurotransmitter release by inhibiting synaptic vesicle reclustering after endocytosis. Neuron 65: 66‐79, 2010.
 476. Noble EP , Bommer M , Liebisch D , Herz A . H1‐histaminergic activation of catecholamine release by chromaffin cells. Biochem Pharmacol 37: 221‐228, 1988.
 477. Noble EP , Bommer M , Sincini E , Costa T , Herz A . H1‐histaminergic activation stimulates inositol‐1‐phosphate accumulation in chromaffin cells. Biochem Biophys Res Commun 135: 566‐573, 1986.
 478. Nooney JM , Feltz A . Inhibition by cyclothiazide of neuronal nicotinic responses in bovine chromaffin cells. Br J Pharmacol 114: 648‐655, 1995.
 479. Novara M , Baldelli P , Cavallari D , Carabelli V , Giancippoli A , Carbone E . Exposure to cAMP and beta‐adrenergic stimulation recruits Ca(V)3 T‐type channels in rat chromaffin cells through Epac cAMP‐receptor proteins. J Physiol 558: 433‐449, 2004.
 480. Nowycky MC , Pinter MJ . Time courses of calcium and calcium‐bound buffers following calcium influx in a model cell. Biophys J 64: 77‐91, 1993.
 481. Nucifora PG , Fox AP . Tyrosine phosphorylation regulates rapid endocytosis in adrenal chromaffin cells. J Neurosci: Off J Soc Neurosci 19: 9739‐9746, 1999.
 482. Nurse CA , Salman S , Scott AL . Hypoxia‐regulated catecholamine secretion in chromaffin cells. Cell Tissue Res 372: 433‐441, 2018.
 483. Nussdorfer GG . Paracrine control of adrenal cortical function by medullary chromaffin cells. Pharmacol Rev 48: 495‐530, 1996.
 484. O'Farrell M , Marley PD . Different contributions of voltage‐sensitive Ca2+ channels to histamine‐induced catecholamine release and tyrosine hydroxylase activation in bovine adrenal chromaffin cells. Cell Calcium 25: 209‐217, 1999.
 485. O'Sullivan AJ , Burgoyne RD . A comparison of bradykinin, angiotensin II and muscarinic stimulation of cultured bovine adrenal chromaffin cells. Biosci Rep 9: 243‐252, 1989.
 486. Oheim M , Stuhmer W . Tracking chromaffin granules on their way through the actin cortex. Eur Biophys J 29: 67‐89, 2000.
 487. Ohta T , Ito S , Nakazato Y . Ca2+‐dependent K+ currents induced by muscarinic receptor activation in guinea pig adrenal chromaffin cells. J Neurochem 70: 1280‐1288, 1998.
 488. Okamoto K , Aoki K . Development of a strain of spontaneously hypertensive rats. Jpn Circul J 27: 282‐293, 1963.
 489. Olivos L , Artalejo AR . Muscarinic excitation‐secretion coupling in chromaffin cells. Acta Physiol (Oxford, England) 192: 213‐220, 2007.
 490. Olivos Ore L , Artalejo AR . Intracellular Ca2+ microdomain‐triggered exocytosis in neuroendocrine cells. Trends Neurosci 27: 113‐115, 2004.
 491. Omiatek DM , Dong Y , Heien ML , Ewing AG . Only a fraction of quantal content is released during exocytosis as revealed by electrochemical cytometry of secretory vesicles. ACS Chem Neurosci 1: 234‐245, 2010.
 492. Omura M , Saito J , Yamaguchi K , Kakuta Y , Nishikawa T . Prospective study on the prevalence of secondary hypertension among hypertensive patients visiting a general outpatient clinic in Japan. Hypertens Res: Off J Jpn Soc Hypertens 27: 193‐202, 2004.
 493. Oset‐Gasque MJ , Castro E , Gonzalez MP . Mechanisms of [3H] gamma‐aminobutyric acid release by chromaffin cells in primary culture. J Neurosci Res 26: 181‐187, 1990.
 494. Oset‐Gasque MJ , Parramon M , Hortelano S , Bosca L , Gonzalez MP . Nitric oxide implication in the control of neurosecretion by chromaffin cells. J Neurochem 63: 1693‐1700, 1994.
 495. Padin JF , Fernandez‐Morales JC , de Diego AM , Garcia AG . Calcium channel subtypes and exocytosis in chromaffin cells at early life. Curr Mol Pharmacol 8: 81‐86, 2015.
 496. Pak CH . Plasma adrenaline and noradrenaline concentrations of the spontaneously hypertensive rat. Jpn Heart J 22: 987‐995, 1981.
 497. Pan CY , Chu YS , Kao LS . Molecular study of the Na+/Ca2+ exchanger in bovine adrenal chromaffin cells. Biochem J 336 (Pt 2): 305‐310, 1998.
 498. Pan CY , Fox AP . Rundown of secretion after depletion of intracellular calcium stores in bovine adrenal chromaffin cells. J Neurochem 75: 1132‐1139, 2000.
 499. Pan CY , Huang CH , Lee CH . Calcium elevation elicited by reverse mode Na+/Ca2+ exchange activity is facilitated by intracellular calcium stores in bovine chromaffin cells. Biochem Biophys Res Commun 342: 589‐595, 2006.
 500. Pan CY , Jeromin A , Lundstrom K , Yoo SH , Roder J , Fox AP . Alterations in exocytosis induced by neuronal Ca2+ sensor‐1 in bovine chromaffin cells. J Neurosci 22: 2427‐2433, 2002.
 501. Pan CY , Kao LS . Catecholamine secretion from bovine adrenal chromaffin cells: The role of the Na+/Ca2+ exchanger and the intracellular Ca2+ pool. J Neurochem 69: 1085‐1092, 1997.
 502. Pape HC . Queer current and pacemaker: The hyperpolarization‐activated cation current in neurons. Annu Rev Physiol 58: 299‐327, 1996.
 503. Park YB . Ion selectivity and gating of small conductance Ca(2+)‐activated K+ channels in cultured rat adrenal chromaffin cells. J Physiol 481 (Pt 3). 555‐570, 1994.
 504. Park YB , Herrington J , Babcock DF , Hille B . Ca2+ clearance mechanisms in isolated rat adrenal chromaffin cells. J Physiol 492 (Pt 2): 329‐346, 1996.
 505. Parker TL , Kesse WK , Mohamed AA , Afework M . The innervation of the mammalian adrenal gland. J Anat 183 (Pt 2): 265‐276, 1993.
 506. Patel JN , Coppack SW , Goldstein DS , Miles JM , Eisenhofer G . Norepinephrine spillover from human adipose tissue before and after a 72‐hour fast. J Clin Endocrinol Metab 87: 3373‐3377, 2002.
 507. Patzak A , Winkler H . Exocytotic exposure and recycling of membrane antigens of chromaffin granules: Ultrastructural evaluation after immunolabeling. J Cell Biol 102: 510‐515, 1986.
 508. Perez‐Alvarez A , Hernandez‐Vivanco A , Alonso YGS , Tabernero A , McIntosh JM , Albillos A . Pharmacological characterization of native alpha7 nicotinic ACh receptors and their contribution to depolarization‐elicited exocytosis in human chromaffin cells. Br J Pharmacol 165: 908‐921, 2012.
 509. Perez‐Alvarez A , Hernandez‐Vivanco A , Caba‐Gonzalez JC , Albillos A . Different roles attributed to Cav1 channel subtypes in spontaneous action potential firing and fine tuning of exocytosis in mouse chromaffin cells. J Neurochem 116: 105‐121, 2011.
 510. Perez‐Alvarez A , Hernandez‐Vivanco A , Cano‐Abad M , Albillos A . Pharmacological and biophysical properties of Ca2+ channels and subtype distributions in human adrenal chromaffin cells. Pflug Archiv: Eur J Physiol 456: 1149‐1162, 2008.
 511. Perez Bay AE , Belingheri AV , Alvarez YD , Marengo FD . Membrane cycling after the excess retrieval mode of rapid endocytosis in mouse chromaffin cells. Acta Physiol (Oxford, England) 204: 403‐418, 2012.
 512. Perrais D , Kleppe IC , Taraska JW , Almers W . Recapture after exocytosis causes differential retention of protein in granules of bovine chromaffin cells. J Physiol 560: 413‐428, 2004.
 513. Peters JA , Lambert JJ , Cottrell GA . An electrophysiological investigation of the characteristics and function of GABAA receptors on bovine adrenomedullary chromaffin cells. Pflug Archiv: Eur J Physiol 415: 95‐103, 1989.
 514. Picones A , Loza‐Huerta A , Segura‐Chama P , Lara‐Figueroa CO . Contribution of automated technologies to ion channel drug discovery. Adv Protein Chem Struct Biol 104: 357‐378, 2016.
 515. Pitts RF , Bronk DW . Excitability cycle of the hypothalamus‐sympathetic neurone system. Am J Physiol 135 (2): 504‐522, 1941.
 516. Platzer J , Engel J , Schrott‐Fischer A , Stephan K , Bova S , Chen H , Zheng H , Striessnig J . Congenital deafness and sinoatrial node dysfunction in mice lacking class D L‐type Ca2+ channels. Cell 102: 89‐97, 2000.
 517. Pletscher A , Da Prada M , Berneis KH , Steffen H , Lütold B , Weder HG . Molecular organization of amine storage organelles of blood platelets and adrenal medulla. In: Ceccarelli B , Clementi F , Meldolesi J , editors. Advances in Cytopharmacology. New York: Raven Press, 1974, p. 257‐264.
 518. Plevin R , Boarder MR . Stimulation of formation of inositol phosphates in primary cultures of bovine adrenal chromaffin cells by angiotensin II, histamine, bradykinin, and carbachol. J Neurochem 51: 634‐641, 1988.
 519. Powell AD , Teschemacher AG , Seward EP . P2Y purinoceptors inhibit exocytosis in adrenal chromaffin cells via modulation of voltage‐operated calcium channels. J Neurosci: Off J Soc Neurosci 20: 606‐616, 2000.
 520. Powis DA , O'Brien KJ , Von Grafenstein HR . Calcium export by sodium‐calcium exchange in bovine chromaffin cells. Cell Calcium 12: 493‐504, 1991.
 521. Prabhakar NR , Kumar GK , Peng YJ . Sympatho‐adrenal activation by chronic intermittent hypoxia. J Appl Physiol 113: 1304‐1310, 2012, 1985.
 522. Prakriya M , Lingle CJ . Activation of BK channels in rat chromaffin cells requires summation of Ca(2+) influx from multiple Ca(2+) channels. J Neurophysiol 84: 1123‐1135, 2000.
 523. Prakriya M , Lingle CJ . BK channel activation by brief depolarizations requires Ca2+ influx through L‐ and Q‐type Ca2+ channels in rat chromaffin cells. J Neurophysiol 81: 2267‐2278, 1999.
 524. Priller C , Mitteregger G , Paluch S , Vassallo N , Staufenbiel M , Kretzschmar HA , Jucker M , Herms J . Excitatory synaptic transmission is depressed in cultured hippocampal neurons of APP/PS1 mice. Neurobiol Aging 30: 1227‐1237, 2009.
 525. Proye C , Fossati P , Fontaine P , Lefebvre J , Decoulx M , Wemeau JL , Dewailly D , Rwamasirabo E , Cecat P . Dopamine‐secreting pheochromocytoma: An unrecognized entity? Classification of pheochromocytomas according to their type of secretion. Surgery 100: 1154‐1162, 1986.
 526. Przywara DA , Guo X , Angelilli ML , Wakade TD , Wakade AR . A non‐cholinergic transmitter, pituitary adenylate cyclase‐activating polypeptide, utilizes a novel mechanism to evoke catecholamine secretion in rat adrenal chromaffin cells. J Biol Chem 271: 10545‐10550, 1996.
 527. Raman IM , Bean BP . Resurgent sodium current and action potential formation in dissociated cerebellar Purkinje neurons. J Neurosci: Off J Soc Neurosci 17: 4517‐4526, 1997.
 528. Rao TC , Santana Rodriguez Z , Bradberry MM , Ranski AH , Dahl PJ , Schmidtke MW , Jenkins PM , Axelrod D , Chapman ER , Giovannucci DR , Anantharam A . Synaptotagmin isoforms confer distinct activation kinetics and dynamics to chromaffin cell granules. J Gen Physiol 149: 763‐780, 2017.
 529. Ream MA , Chandra R , Peavey M , Ray AM , Roffler‐Tarlov S , Kim HG , Wetsel WC , Rockman HA , Chikaraishi DM . High oxygen prevents fetal lethality due to lack of catecholamines. Am J Physiol Regul Integr Comp Physiol 295: R942‐R953, 2008.
 530. Reid CA , Bekkers JM , Clements JD . Presynaptic Ca2+ channels: A functional patchwork. Trends Neurosci 26: 683‐687, 2003.
 531. Reuter H . The dependence of slow inward current in Purkinje fibres on the extracellular calcium‐concentration. J Physiol 192: 479‐492, 1967.
 532. Reynafarje B , Lehninger AL . Electric charge stoichiometry of calcium translocation in mitochondria. Biochem Biophys Res Commun 77: 1273‐1279, 1977.
 533. Richards DA , Guatimosim C , Betz WJ . Two endocytic recycling routes selectively fill two vesicle pools in frog motor nerve terminals. Neuron 27: 551‐559, 2000.
 534. Rico AJ , Prieto‐Lloret J , Gonzalez C , Rigual R . Hypoxia and acidosis increase the secretion of catecholamines in the neonatal rat adrenal medulla: An in vitro study. Am J Physiol Cell Physiol 289: C1417‐C1425, 2005.
 535. Rigual R , Montero M , Rico AJ , Prieto‐Lloret J , Alonso MT , Alvarez J . Modulation of secretion by the endoplasmic reticulum in mouse chromaffin cells. Eur J Neurosci 16: 1690‐1696, 2002.
 536. Rink TJ . The influence of sodium on calcium movements and catecholamine release in thin slices of bovine adrenal medulla. J Physiol 266: 297‐325, 1977.
 537. Rizzuto R , Brini M , Murgia M , Pozzan T . Microdomains with high Ca2+ close to IP3‐sensitive channels that are sensed by neighboring mitochondria. Science 262: 744‐747, 1993.
 538. Robinson IM , Burgoyne RD . Characterisation of distinct inositol 1,4,5‐trisphosphate‐sensitive and caffeine‐sensitive calcium stores in digitonin‐permeabilised adrenal chromaffin cells. J Neurochem 56: 1587‐1593, 1991.
 539. Robinson IM , Yamada M , Carrion‐Vazquez M , Lennon VA , Fernandez JM . Specialized release zones in chromaffin cells examined with pulsed‐laser imaging. Cell Calcium 20: 181‐201, 1996.
 540. Robinson RL , Culberson JL , Carmichael SW . Influence of hypothalamic stimulation on the secretion of adrenal medullary catecholamines. J Auton Nerv Syst 8: 89‐96, 1983.
 541. Rosa JM , de Diego AM , Gandia L , Garcia AG . L‐type calcium channels are preferentially coupled to endocytosis in bovine chromaffin cells. Biochem Biophys Res Commun 357: 834‐839, 2007.
 542. Rosa JM , Gandia L , Garcia AG . Inhibition of N and PQ calcium channels by calcium entry through L channels in chromaffin cells. Pflug Archiv: Eur J Physiol 458: 795‐807, 2009.
 543. Rosa JM , Torregrosa‐Hetland CJ , Colmena I , Gutierrez LM , Garcia AG , Gandia L . Calcium entry through slow‐inactivating L‐type calcium channels preferentially triggers endocytosis rather than exocytosis in bovine chromaffin cells. Am J Physiol Cell Physiol 301: C86‐C98, 2011.
 544. Roscioni SS , Elzinga CR , Schmidt M . Epac: Effectors and biological functions. Naunyn Schmiedeberg's Arch Pharmacol 377: 345‐357, 2008.
 545. Rose SD , Lejen T , Casaletti L , Larson RE , Pene TD , Trifaro JM . Molecular motors involved in chromaffin cell secretion. Ann N Y Acad Sci 971: 222‐231, 2002.
 546. Rowland LP , Shneider NA . Amyotrophic lateral sclerosis. N Engl J Med 344: 1688‐1700, 2001.
 547. Rozas JL , Gomez‐Sanchez L , Tomas‐Zapico C , Lucas JJ , Fernandez‐Chacon R . Increased neurotransmitter release at the neuromuscular junction in a mouse model of polyglutamine disease. J Neurosci 31: 1106‐1113, 2011.
 548. Rubio I , Rodriguez‐Navarro JA , Tomas‐Zapico C , Ruiz C , Casarejos MJ , Perucho J , Gomez A , Rodal I , Lucas JJ , Mena MA , de Yebenes JG . Effects of partial suppression of parkin on huntingtin mutant R6/1 mice. Brain Res 1281: 91‐100, 2009.
 549. Sahu BS , Mahata S , Bandyopadhyay K , Mahata M , Avolio E , Pasqua T , Sahu C , Bandyopadhyay GK , Bartolomucci A , Webster NJG , Van Den Bogaart G , Fischer‐Colbrie R , Corti A , Eiden LE , Mahata SK . Catestatin regulates vesicular quanta through modulation of cholinergic and peptidergic (PACAPergic) stimulation in PC12 cells. Cell Tissue Res, 2018.
 550. Salman S , Buttigieg J , Zhang M , Nurse CA . Chronic exposure of neonatal rat adrenomedullary chromaffin cells to opioids in vitro blunts both hypoxia and hypercapnia chemosensitivity. J Physiol 591: 515‐529, 2013.
 551. Salvador JM , Inesi G , Rigaud JL , Mata AM . Ca2+ transport by reconstituted synaptosomal ATPase is associated with H+ countertransport and net charge displacement. J Biol Chem 273: 18230‐18234, 1998.
 552. Santodomingo J , Vay L , Camacho M , Hernandez‐Sanmiguel E , Fonteriz RI , Lobaton CD , Montero M , Moreno A , Alvarez J . Calcium dynamics in bovine adrenal medulla chromaffin cell secretory granules. Eur J Neurosci 28: 1265‐1274, 2008.
 553. Sasakawa N , Nakaki T , Kato R . Formation of inositol polyphosphates in cultured adrenal chromaffin cells. Adv Exp Med Biol 287: 111‐123, 1991.
 554. Sawada K , Echigo N , Juge N , Miyaji T , Otsuka M , Omote H , Yamamoto A , Moriyama Y . Identification of a vesicular nucleotide transporter. Proc Natl Acad Sci USA 105: 5683‐5686, 2008.
 555. Schapiro FB , Grinstein S . Determinants of the pH of the Golgi complex. J Biol Chem 275: 21025‐21032, 2000.
 556. Scharinger A , Eckrich S , Vandael DH , Schonig K , Koschak A , Hecker D , Kaur G , Lee A , Sah A , Bartsch D , Benedetti B , Lieb A , Schick B , Singewald N , Sinnegger‐Brauns MJ , Carbone E , Engel J , Steiessnig J . Cell‐type‐specific tuning of Cav1.3 Ca2+‐channels by a C‐terminal automodulatory domain. Front Cellular Neurosci 9: 18, 2015.
 557. Schonn JS , Maximov A , Lao Y , Sudhof TC , Sorensen JB . Synaptotagmin‐1 and ‐7 are functionally overlapping Ca2+ sensors for exocytosis in adrenal chromaffin cells. Proc Natl Acad Sci USA 105: 3998‐4003, 2008.
 558. Schroeder TJ , Borges R , Finnegan JM , Pihel K , Amatore C , Wightman RM . Temporally resolved, independent stages of individual exocytotic secretion events. Biophys J 70: 1061‐1068, 1996.
 559. Schuldiner S . A molecular glimpse of vesicular transporters. J Neurochem 62: 2067‐2078, 1994.
 560. Schuldiner S , Shirvan A , Stern‐Bach Y , Steiner‐Mordoch S , Yelin R , Laskar O . From bacterial antibiotic resistance to neurotransmitter uptake. A common theme of cell survival. Ann N Y Acad Sci 733: 174‐184, 1994.
 561. Schwarz PM , Rodriguez‐Pascual F , Koesling D , Torres M , Forstermann U . Functional coupling of nitric oxide synthase and soluble guanylyl cyclase in controlling catecholamine secretion from bovine chromaffin cells. Neuroscience 82: 255‐265, 1998.
 562. Scott AL , Zhang M , Nurse CA . Enhanced BDNF signalling following chronic hypoxia potentiates catecholamine release from cultured rat adrenal chromaffin cells. J Physiol 593: 3281‐3299, 2015.
 563. Scott DA , Tabarean I , Tang Y , Cartier A , Masliah E , Roy S . A pathologic cascade leading to synaptic dysfunction in alpha‐synuclein‐induced neurodegeneration. J Neurosci 30: 8083‐8095, 2010.
 564. Scott RS , Bustillo D , Olivos‐Oré LA , Cuchillo‐Ibanez I , Barahona MV , Carbone E , Artalejo AR . Contribution of BK channels to action potential repolarisation at minimal cytosolic Ca2+ concentration in chromaffin cells. Pflug Archiv: Eur J Physiol 462: 545‐557, 2011.
 565. Segura‐Chama P , Hernandez A , Jimenez‐Perez N , Alejandre‐Garcia T , Rivera‐Cerecedo CV , Hernandez‐Guijo J , Hernandez‐Cruz A . Comparison of Ca2+ currents of chromaffin cells from normotensive Wistar Kyoto and spontaneously hypertensive rats. Cell Mol Neurobiol 30: 1243‐1250, 2010.
 566. Segura‐Chama P , Lopez‐Bistrain P , Perez‐Armendariz EM , Jimenez‐Perez N , Millan‐Aldaco D , Hernandez‐Cruz A . Enhanced Ca‐induced Ca release from intracellular stores contributes to catecholamine hypersecretion in adrenal chromaffin cells from spontaneously hypertensive rats. Pflug Archiv: Eur J Physiol, 2015.
 567. Seidler FJ , Slotkin TA . Adrenomedullary function in the neonatal rat: Responses to acute hypoxia. J Physiol 358: 1‐16, 1985.
 568. Selkoe DJ . Alzheimer's disease is a synaptic failure. Science 298: 789‐791, 2002.
 569. Selye H . The evolution of the stress concept. Am Scientist 61: 692‐699, 1973.
 570. Selye H . Stress In Health and Disease. Butterworths, 1976.
 571. Senior SL , Ninkina N , Deacon R , Bannerman D , Buchman VL , Cragg SJ , Wade‐Martins R . Increased striatal dopamine release and hyperdopaminergic‐like behaviour in mice lacking both alpha‐synuclein and gamma‐synuclein. Eur J Neurosci 27: 947‐957, 2008.
 572. Shahar E , Whitney CW , Redline S , Lee ET , Newman AB , Nieto FJ , O'Connor GT , Boland LL , Schwartz JE , Samet JM . Sleep‐disordered breathing and cardiovascular disease: Cross‐sectional results of the Sleep Heart Health Study. Am J Respir Crit Care Med 163: 19‐25, 2001.
 573. Sher E , Cesare P , Codignola A , Clementi F , Tarroni P , Pollo A , Magnelli V , Carbone E . Activation of delta‐opioid receptors inhibits neuronal‐like calcium channels and distal steps of Ca2+‐dependent secretion in human small‐cell lung carcinoma cells. J Neurosci 16: 3672‐3684, 1996.
 574. Shin W , Ge L , Arpino G , Villarreal SA , Hamid E , Liu H , Zhao WD , Wen PJ , Chiang HC , Wu LG . Visualization of membrane pore in live cells reveals a dynamic‐pore theory governing fusion and endocytosis. Cell 173: 934‐945, e912, 2018.
 575. Shukla R , Wakade AR . Functional aspects of calcium channels of splanchnic neurons and chromaffin cells of the rat adrenal medulla. J Neurochem 56: 753‐758, 1991.
 576. Silver M . The output of adrenaline and noradrenaline from the adrenal medulla of the calf. J Physiol 152: 14‐29, 1960.
 577. Singleton AB , Farrer M , Johnson J , Singleton A , Hague S , Kachergus J , Hulihan M , Peuralinna T , Dutra A , Nussbaum R , Lincoln S , Crawley A , Hanson M , Maraganore D , Adler C , Cookson MR , Muenter M , Baptista M , Miller D , Blancato J , Hardy J , Gwinn‐Hardy K . alpha‐Synuclein locus triplication causes Parkinson's disease. Science 302: 841, 2003.
 578. Slotkin TA , Seidler FJ . Adrenomedullary catecholamine release in the fetus and newborn: Secretory mechanisms and their role in stress and survival. J Dev Physiol 10: 1‐16, 1988.
 579. Smith C , Neher E . Multiple forms of endocytosis in bovine adrenal chromaffin cells. J Cell Biol 139: 885‐894, 1997.
 580. Smith CB , Eiden LE . Is PACAP the major neurotransmitter for stress transduction at the adrenomedullary synapse? J Mol Neurosci 48: 403‐412, 2012.
 581. Solaro CR , Prakriya M , Ding JP , Lingle CJ . Inactivating and noninactivating Ca2+‐ and voltage‐dependent K+ current in rat adrenal chromaffin cells. J Neurosci 15: 6110‐6123, 1995.
 582. Sorensen JB . Formation, stabilisation and fusion of the readily releasable pool of secretory vesicles. Pflug Archiv: Eur J Physiol 448: 347‐362, 2004.
 583. Sorensen JB , Matti U , Wei SH , Nehring RB , Voets T , Ashery U , Binz T , Neher E , Rettig J . The SNARE protein SNAP‐25 is linked to fast calcium triggering of exocytosis. Proc Natl Acad Sci USA 99: 1627‐1632, 2002.
 584. Souvannakitti D , Kumar GK , Fox A , Prabhakar NR . Neonatal intermittent hypoxia leads to long‐lasting facilitation of acute hypoxia‐evoked catecholamine secretion from rat chromaffin cells. J Neurophysiol 101: 2837‐2846, 2009.
 585. Souvannakitti D , Nanduri J , Yuan G , Kumar GK , Fox AP , Prabhakar NR . NADPH oxidase‐dependent regulation of T‐type Ca2+ channels and ryanodine receptors mediate the augmented exocytosis of catecholamines from intermittent hypoxia‐treated neonatal rat chromaffin cells. J Neurosci 30: 10763‐10772, 2010.
 586. Splawski I , Timothy KW , Decher N , Kumar P , Sachse FB , Beggs AH , Sanguinetti MC , Keating MT . Severe arrhythmia disorder caused by cardiac L‐type calcium channel mutations. Proc Natl Acad Sci USA 102: 8089‐8096, 2005.
 587. Splawski I , Timothy KW , Sharpe LM , Decher N , Kumar P , Bloise R , Napolitano C , Schwartz PJ , Joseph RM , Condouris K , Tager‐Flusberg H , Priori SG , Sanguinetti MC , Keating MT . Ca(v)1.2 calcium channel dysfunction causes a multisystem disorder including arrhythmia and autism. Cell 119: 19‐31, 2004.
 588. Stadinski BD , Delong T , Reisdorph N , Reisdorph R , Powell RL , Armstrong M , Piganelli JD , Barbour G , Bradley B , Crawford F , Marrack P , Mahata SK , Kappler JW , Haskins K . Chromogranin A is an autoantigen in type 1 diabetes. Nat Immunol 11: 225‐231.
 589. Stauderman KA , Murawsky MM , Pruss RM . Agonist‐dependent patterns of cytosolic Ca2+ changes in single bovine adrenal chromaffin cells: Relationship to catecholamine release. Cell Regul 1: 683‐691, 1990.
 590. Stauderman KA , Pruss RM . Different patterns of agonist‐stimulated increases of 3H‐inositol phosphate isomers and cytosolic Ca2+ in bovine chromaffin cells: Comparison of the effects of histamine and angiotensin II. J Neurochem 54: 946‐953, 1990.
 591. Stevens DR , Schirra C , Becherer U , Rettig J . Vesicle pools: Lessons from adrenal chromaffin cells. Front Synap Neurosci 3: 2, 2011.
 592. Steyer JA , Almers W . Tracking single secretory granules in live chromaffin cells by evanescent‐field fluorescence microscopy. Biophys J 76: 2262‐2271, 1999.
 593. Stroth N , Kuri BA , Mustafa T , Chan SA , Smith CB , Eiden LE . PACAP controls adrenomedullary catecholamine secretion and expression of catecholamine biosynthetic enzymes at high splanchnic nerve firing rates characteristic of stress transduction in male mice. Endocrinology 154: 330‐339, 2013.
 594. Sudhof TC , Rizo J . Synaptic vesicle exocytosis. Cold Spring Harb Perspect Biol 3, 2011.
 595. Sudhof TC , Rothman JE . Membrane fusion: Grappling with SNARE and SM proteins. Science 323: 474‐477, 2009.
 596. Suh BC , Leal K , Hille B . Modulation of high‐voltage activated Ca(2+) channels by membrane phosphatidylinositol 4,5‐bisphosphate. Neuron 67: 224‐238, 2010.
 597. Sun CL , Thoa NB , Kopin IJ . Comparison of the effects of 2‐deoxyglucose and immobilization on plasma levels of catecholamines and corticosterone in awake rats. Endocrinology 105: 306‐311, 1979.
 598. Sun L , Xiong Y , Zeng X , Wu Y , Pan N , Lingle CJ , Qu A , Ding J . Differential regulation of action potentials by inactivating and noninactivating BK channels in rat adrenal chromaffin cells. Biophys J 97: 1832‐1842, 2009.
 599. Tabares L , Ales E , Lindau M , Alvarez de Toledo G . Exocytosis of catecholamine (CA)‐containing and CA‐free granules in chromaffin cells. J Biol Chem 276: 39974‐39979, 2001.
 600. Tabarin A , Chen D , Hakanson R , Sundler F . Pituitary adenylate cyclase‐activating peptide in the adrenal gland of mammals: Distribution, characterization and responses to drugs. Neuroendocrinology 59: 113‐119, 1994.
 601. Tagaya M , Toyonaga S , Takahashi M , Yamamoto A , Fujiwara T , Akagawa K , Moriyama Y , Mizushima S . Syntaxin 1 (HPC‐1) is associated with chromaffin granules. J Biol Chem 270: 15930‐15933, 1995.
 602. Takeuchi Y , Mochizuki‐Oda N , Yamada H , Kurokawa K , Watanabe Y . Nonneurogenic hypoxia sensitivity in rat adrenal slices. Biochem Biophys Res Commun 289: 51‐56, 2001.
 603. Taleat Z , Estevez‐Herrera J , Machado JD , Dunevall J , Ewing AG , Borges R . Electrochemical investigation of the interaction between catecholamines and ATP. Anal Chem 90: 1601‐1607, 2018.
 604. Tanguy E , Carmon O , Wang Q , Jeandel L , Chasserot‐Golaz S , Montero‐Hadjadje M , Vitale N . Lipids implicated in the journey of a secretory granule: From biogenesis to fusion. J Neurochem 137: 904‐912, 2016.
 605. Taraska JW , Almers W . Bilayers merge even when exocytosis is transient. Proc Natl Acad Sci USA 101: 8780‐8785, 2004.
 606. Taraska JW , Perrais D , Ohara‐Imaizumi M , Nagamatsu S , Almers W . Secretory granules are recaptured largely intact after stimulated exocytosis in cultured endocrine cells. Proc Natl Acad Sci USA 100: 2070‐2075, 2003.
 607. Tateno M , Sadakata H , Tanaka M , Itohara S , Shin RM , Miura M , Masuda M , Aosaki T , Urushitani M , Misawa H , Takahashi R . Calcium‐permeable AMPA receptors promote misfolding of mutant SOD1 protein and development of amyotrophic lateral sclerosis in a transgenic mouse model. Hum Mol Genet 13: 2183‐2196, 2004.
 608. Taylor CV , Taupenot L , Mahata SK , Mahata M , Wu H , Yasothornsrikul S , Toneff T , Caporale C , Jiang Q , Parmer RJ , Hook VY , O'Connor DT . Formation of the catecholamine release‐inhibitory peptide catestatin from chromogranin A. Determination of proteolytic cleavage sites in hormone storage granules. J Biol Chem 275: 22905‐22915, 2000.
 609. Thiagarajan R , Tewolde T , Li Y , Becker PL , Rich MM , Engisch KL . Rab3A negatively regulates activity‐dependent modulation of exocytosis in bovine adrenal chromaffin cells. J Physiol 555: 439‐457, 2003.
 610. Thoenen H , Mueller RA , Axelrod J . Trans‐synaptic induction of adrenal tyrosine hydroxylase. J Pharm Exp Ther 169: 249‐254, 1969.
 611. Thomas P , Lee AK , Wong JG , Almers W . A triggered mechanism retrieves membrane in seconds after Ca(2+)‐stimulated exocytosis in single pituitary cells. J Cell Biol 124: 667‐675, 1994.
 612. Thomas RC . The plasma membrane calcium ATPase (PMCA) of neurones is electroneutral and exchanges 2 H+ for each Ca2+ or Ba2+ ion extruded. J Physiol 587: 315‐327, 2009.
 613. Thompson RJ , Farragher SM , Cutz E , Nurse CA . Developmental regulation of O(2) sensing in neonatal adrenal chromaffin cells from wild‐type and NADPH‐oxidase‐deficient mice. Pflug Archiv: Eur J Physiol 444: 539‐548, 2002.
 614. Thompson RJ , Jackson A , Nurse CA . Developmental loss of hypoxic chemosensitivity in rat adrenomedullary chromaffin cells. J Physiol 498 (Pt 2): 503‐510, 1997.
 615. Thompson RJ , Nurse CA . Anoxia differentially modulates multiple K+ currents and depolarizes neonatal rat adrenal chromaffin cells. J Physiol 512 (Pt 2): 421‐434, 1998.
 616. Tillinger A , Sollas A , Serova LI , Kvetnansky R , Sabban EL . Vesicular monoamine transporters (VMATs) in adrenal chromaffin cells: Stress‐triggered induction of VMAT2 and expression in epinephrine synthesizing cells. Cell Mol Neurobiol 30: 1459‐1465, 2010.
 617. Tischler AS . Pheochromocytoma and extra‐adrenal paraganglioma: Updates. Arch Pathol Lab Med 132: 1272‐1284, 2008.
 618. Tomatis VM , Papadopulos A , Malintan NT , Martin S , Wallis T , Gormal RS , Kendrick‐Jones J , Buss F , Meunier FA . Myosin VI small insert isoform maintains exocytosis by tethering secretory granules to the cortical actin. J Cell Biol 200: 301‐320, 2013.
 619. Tomlinson A , Coupland RE . The innervation of the adrenal gland. IV. Innervation of the rat adrenal medulla from birth to old age. A descriptive and quantitative morphometric and biochemical study of the innervation of chromaffin cells and adrenomedullary neurons in Wistar rats. J Anat 169: 209‐236, 1990.
 620. Tompkins JD , Ardell JL , Hoover DB , Parsons RL . Neurally released pituitary adenylate cyclase‐activating polypeptide enhances guinea pig intrinsic cardiac neurone excitability. J Physiol 582: 87‐93, 2007.
 621. Tsien RW , Giles W , Greengard P . Cyclic AMP mediates the effects of adrenaline on cardiac purkinje fibres. Nat: New Biol 240: 181‐183, 1972.
 622. Tsigelny IF , Kouznetsova VL , Biswas N , Mahata SK , O'Connor DT . Development of a pharmacophore model for the catecholamine release‐inhibitory peptide catestatin: Virtual screening and functional testing identify novel small molecule therapeutics of hypertension. Bioorg Med Chem 21: 5855‐5869, 2013.
 623. Tsujimoto A , Nishikawa T . Further evidence for nicotinic and muscarinic receptors and their interaction in dog adrenal medulla. Eur J Pharmacol 34: 337‐344, 1975.
 624. Twitchell WA , Rane SG . Opioid peptide modulation of Ca(2+)‐dependent K+ and voltage‐activated Ca2+ currents in bovine adrenal chromaffin cells. Neuron 10: 701‐709, 1993.
 625. Uceda G , Artalejo AR , de la Fuente MT , Lopez MG , Albillos A , Michelena P , Garcia AG , Montiel C . Modulation by L‐type Ca2+ channels and apamin‐sensitive K+ channels of muscarinic responses in cat chromaffin cells. Am J Physiol 266: C1432‐C1439, 1994.
 626. Uceda G , Artalejo AR , Lopez MG , Abad F , Neher E , Garcia AG . Ca(2+)‐activated K+ channels modulate muscarinic secretion in cat chromaffin cells. J Physiol 454: 213‐230, 1992.
 627. Ulate G , Scott SR , Gonzalez J , Gilabert JA , Artalejo AR . Extracellular ATP regulates exocytosis in inhibiting multiple Ca(2+) channel types in bovine chromaffin cells. Pflug Archiv: Eur J Physiol 439: 304‐314, 2000.
 628. Unsicker K , Habura‐Fluh O , Zwarg U . Different types of small granule‐containing cells and neurons in the guinea‐pig adrenal medulla. Cell Tissue Res 189: 109‐130, 1978.
 629. Uvnäs B , Aborg C‐H . The ability of ATP‐free granule material from bovine adrenal medulla to bind inorganic cations and biogenic amines. Acta Physiol Scand 99: 476‐483, 1977.
 630. van Kempen GT , vanderLeest HT , van den Berg RJ , Eilers P , Westerink RH . Three distinct modes of exocytosis revealed by amperometry in neuroendocrine cells. Biophys J 100: 968‐977, 2011.
 631. Vandael DH , Marcantoni A , Cav1 CE . 3 Channels as key regulators of neuron‐like firings and catecholamine release in chromaffin cells. Curr Mol Pharmacol 8: 149‐161, 2015.
 632. Vandael DH , Marcantoni A , Mahapatra S , Caro A , Ruth P , Zuccotti A , Knipper M , Carbone E . Ca(v)1.3 and BK channels for timing and regulating cell firing. Mol Neurobiol 42: 185‐198, 2010.
 633. Vandael DH , Ottaviani MM , Legros C , Lefort C , Guerineau NC , Allio A , Carabelli V , Carbone E . Reduced availability of voltage‐gated sodium channels by depolarization or blockade by tetrodotoxin boosts burst firing and catecholamine release in mouse chromaffin cellslabilit. J Physiol 593: 905‐927, 2015.
 634. Vandael DHF , Mahapatra S , Calorio C , Marcantoni A , Carbone E . Cav1.3 and Cav1.2 channels of adrenal chromaffin cells: Emerging views on cAMP/cGMP‐mediated phosphorylation and role in pacemaking. Biochim Biophys Acta‐Biomembr 1828: 1608‐1618, 2013.
 635. Vandael DHF , Zuccotti A , Striessnig J , Carbone E . Ca(V)1.3‐driven SK channel activation regulates pacemaking and spike frequency adaptation in mouse chromaffin cells. J Neurosci 32: 16345‐16359, 2012.
 636. Vestring S , Fernandez‐Morales JC , Mendez‐Lopez I , CM D , AM GD , Padin JF , GG A . Tight mitochondrial control of calcium and exocytotic signals in chromaffin cells at embryonic life. Pflug Archiv: Eur J Physiol 467: 2589‐2601, 2015.
 637. Vicente S , Gonzalez MP , Oset‐Gasque MJ . Neuronal nitric oxide synthase modulates basal catecholamine secretion in bovine chromaffin cells. J Neurosci Res 69: 327‐340, 2002.
 638. Vijayaragavan K , O'Leary ME , Chahine M . Gating properties of Na(v)1.7 and Na(v)1.8 peripheral nerve sodium channels. J Neurosci 21: 7909‐7918, 2001.
 639. Villalobos C , Nunez L , Chamero P , Alonso MT , Garcia‐Sancho J . Mitochondrial [Ca(2+)] oscillations driven by local high [Ca(2+)] domains generated by spontaneous electric activity. J Biol Chem 276: 40293‐40297, 2001.
 640. Villalobos C , Nunez L , Montero M , Garcia AG , Alonso MT , Chamero P , Alvarez J , Garcia‐Sancho J . Redistribution of Ca2+ among cytosol and organella during stimulation of bovine chromaffin cells. FASEB J 16: 343‐353, 2002.
 641. Villanueva J , Torres V , Torregrosa‐Hetland CJ , Garcia‐Martinez V , Lopez‐Font I , Viniegra S , Gutierrez LM . F‐actin‐myosin II inhibitors affect chromaffin granule plasma membrane distance and fusion kinetics by retraction of the cytoskeletal cortex. J Mol Neurosci 48: 328‐338, 2012.
 642. Vitale ML , Rodriguez Del Castillo A , Tchakarov L , Trifaro JM . Cortical filamentous actin disassembly and scinderin redistribution during chromaffin cell stimulation precede exocytosis, a phenomenon not exhibited by gelsolin. J Cell Biol 113: 1057‐1067, 1991.
 643. Voets T . Dissection of three Ca2+‐dependent steps leading to secretion in chromaffin cells from mouse adrenal slices. Neuron 28: 537‐545, 2000.
 644. Voets T , Neher E , Moser T . Mechanisms underlying phasic and sustained secretion in chromaffin cells from mouse adrenal slices. Neuron 23: 607‐615, 1999.
 645. Vollmer RR , Balcita JJ , Sved AF , Edwards DJ . Adrenal epinephrine and norepinephrine release to hypoglycemia measured by microdialysis in conscious rats. Am J Physiol 273: R1758‐R1763, 1997.
 646. Vollmer RR , Baruchin A , Kolibal‐Pegher SS , Corey SP , Stricker EM , Kaplan BB . Selective activation of norepinephrine‐ and epinephrine‐secreting chromaffin cells in rat adrenal medulla. Am J Physiol 263: R716‐R721, 1992.
 647. Von Euler US . Pathophysiological aspects of catecholamine production. Clin Chem 18: 1445‐1448, 1972.
 648. Von Euler US , Folkow B . The effect of stimulation of autonomic areas in the cerebral cortex upon the adrenaline and noradrenaline secretion from the adrenal gland in the cat. Acta Physiol Scand 42: 313‐320, 1958.
 649. von Grafenstein H , Borges R , Knight DE . The effect of botulinum toxin type D on the triggered and constitutive exocytosis/endocytosis cycles in cultures of bovine adrenal medullary cells. FEBS Lett 298: 118‐122, 1992.
 650. von Grafenstein H , Knight DE . Membrane recapture and early triggered secretion from the newly formed endocytotic compartment in bovine chromaffin cells. J Physiol 453: 15‐31, 1992.
 651. von Lewinski F , Keller BU . Ca2+, mitochondria and selective motoneuron vulnerability: Implications for ALS. Trends Neurosci 28: 494‐500, 2005.
 652. von Ruden L , Neher E . A Ca‐dependent early step in the release of catecholamines from adrenal chromaffin cells. Science 262: 1061‐1065, 1993.
 653. Vonsattel JP , DiFiglia M . Huntington disease. J Neuropathol Exp Neurol 57: 369‐384, 1998.
 654. Wada A , Wanke E , Gullo F , Schiavon E . Voltage‐dependent Na(v)1.7 sodium channels: Multiple roles in adrenal chromaffin cells and peripheral nervous system. Acta Physiol (Oxford, England) 192: 221‐231, 2008.
 655. Wada A , Yanagita T , Yokoo H , Kobayashi H . Regulation of cell surface expression of voltage‐dependent Nav1.7 sodium channels: mRNA stability and posttranscriptional control in adrenal chromaffin cells. Front Biosci 9: 1954‐1966, 2004.
 656. Wakade AR . Multiple transmitter control of catecholamine secretion in rat adrenal medulla. Adv Pharmacol (San Diego, CA) 42: 595‐598, 1998.
 657. Wakade AR . Noncholinergic transmitter(s) maintains secretion of catecholamines from rat adrenal medulla for several hours of continuous stimulation of splanchnic neurons. J Neurochem 50: 1302‐1308, 1988.
 658. Wakade AR . Studies on secretion of catecholamines evoked by acetylcholine or transmural stimulation of the rat adrenal gland. J Physiol 313: 463‐480, 1981.
 659. Wakade AR , Wakade TD , Malhotra RK . Restoration of catecholamine content of previously depleted adrenal medulla in vitro: Importance of synthesis in maintaining the catecholamine stores. J Neurochem 51: 820‐829, 1988.
 660. Wallace DJ , Chen C , Marley PD . Histamine promotes excitability in bovine adrenal chromaffin cells by inhibiting an M‐current. J Physiol 540: 921‐939, 2002.
 661. Wallner M , Meera P , Toro L . Molecular basis of fast inactivation in voltage and Ca2+‐activated K+ channels: A transmembrane beta‐subunit homolog. Proc Natl Acad Sci USA 96: 4137‐4142, 1999.
 662. Wang N , Kwan C , Gong X , de Chaves EP , Tse A , Tse FW . Influence of cholesterol on catecholamine release from the fusion pore of large dense core chromaffin granules. J Neurosci 30: 3904‐3911, 2010.
 663. Watanabe T , Masuo Y , Matsumoto H , Suzuki N , Ohtaki T , Masuda Y , Kitada C , Tsuda M , Fujino M . Pituitary adenylate cyclase activating polypeptide provokes cultured rat chromaffin cells to secrete adrenaline. Biochem Biophys Res Commun 182: 403‐411, 1992.
 664. Weihe E , Schafer MK , Erickson JD , Eiden LE . Localization of vesicular monoamine transporter isoforms (VMAT1 and VMAT2) to endocrine cells and neurons in rat. J Mol Neurosci 5: 149‐164, 1994.
 665. Weiss JL , Burgoyne RD . Voltage‐independent inhibition of P/Q‐type Ca2+ channels in adrenal chromaffin cells via a neuronal Ca2+ sensor‐1‐dependent pathway involves Src family tyrosine kinase. J Biol Chem 276: 44804‐44811, 2001.
 666. Wen PJ , Grenklo S , Arpino G , Tan X , Liao HS , Heureaux J , Peng SY , Chiang HC , Hamid E , Zhao WD , Shin W , Nareoja T , Evergren E , Jin Y , Karlsson R , Ebert SN , Jin A , Liu AP , Shupliakov O , Wu LG . Actin dynamics provides membrane tension to merge fusing vesicles into the plasma membrane. Nat Commun 7: 12604, 2016.
 667. Westerink RH , Ewing AG . The PC12 cell as model for neurosecretion. Acta Physiol (Oxford) 192: 273‐285, 2008.
 668. Wightman RM , Jankowski JA , Kennedy RT , Kawagoe KT , Schroeder TJ , Leszczyszyn DJ , Near JA , Diliberto EJ Jr. , Viveros OH . Temporally resolved catecholamine spikes correspond to single vesicle release from individual chromaffin cells. Proc Natl Acad Sci USA 88: 10754‐10758, 1991.
 669. Wightman RM , Schroeder TJ , Finnegan JM , Ciolkowski EL , Pihel K . Time‐course of release of catecholamines from individual vesicles during exocytosis at adrenal‐medullary cells. Biophys J 68: 383‐390, 1995.
 670. Wilson SP . Vasoactive intestinal peptide elevates cyclic AMP levels and potentiates secretion in bovine adrenal chromaffin cells. Neuropeptides 11: 17‐21, 1988.
 671. Winkler H . The adrenal chromaffin granule: A model for large dense core vesicles of endocrine and nervous tissue. J Anat 183 (Pt 2): 237‐252, 1993.
 672. Winkler H , Apps DK , Fischer‐Colbrie R . The molecular function of adrenal chromaffin granules: Established facts and unresolved topics. Neuroscience 18: 261‐290, 1986.
 673. Winkler H , Fischer‐Colbrie R . The chromogranins A and B: The first 25 years and future perspectives. Neuroscience 49: 497‐528, 1992.
 674. Wolf K , Zarkua G , Chan SA , Sridhar A , Smith C . Spatial and activity‐dependent catecholamine release in rat adrenal medulla under native neuronal stimulation. Physiol Rep 4, 2016.
 675. Wu PC , Fann MJ , Kao LS . Characterization of Ca2+ signaling pathways in mouse adrenal medullary chromaffin cells. J Neurochem 112: 1210‐1222, 2010.
 676. Xia X‐M , Ding JP , Lingle CJ . Molecular basis for the inactivation of Ca2+‐ and voltage‐dependent BK channels in adrenal chromaffin cells and rat insulinoma tumor cells. J Neurosci 19: 5255‐5264, 1999.
 677. Xia XM , Ding JP , Lingle CJ . Inactivation of BK channels by the NH2 terminus of the beta2 auxiliary subunit: An essential role of a terminal peptide segment of three hydrophobic residues. J Gen Physiol 121: 125‐148, 2003.
 678. Xu T , Naraghi M , Kang H , Neher E . Kinetic studies of Ca2+ binding and Ca2+ clearance in the cytosol of adrenal chromaffin cells. Biophys J 73: 532‐545, 1997.
 679. Xu WF , Lipscombe D . Neuronal Ca(v)1.3 alpha(1) L‐type channels activate at relatively hyperpolarized membrane potentials and are incompletely inhibited by dihydropyridines. J Neurosci 21: 5944‐5951, 2001.
 680. Yamada‐Hanff J , Bean BP . Persistent sodium current drives conditional pacemaking in CA1 pyramidal neurons under muscarinic stimulation. J Neurosci: Off J Soc Neurosci 33: 15011‐15021, 2013.
 681. Yanagita T , Kobayashi H , Uezono Y , Yokoo H , Sugano T , Saitoh T , Minami S , Shiraishi S , Wada A . Destabilization of Na(v)1.7 sodium channel alpha‐subunit mRNA by constitutive phosphorylation of extracellular signal‐regulated kinase: Negative regulation of steady‐state level of cell surface functional sodium channels in adrenal chromaffin cells. Mol Pharmacol 63: 1125‐1136, 2003.
 682. Yang DM , Kao LS . Relative contribution of the Na(+)/Ca(2+) exchanger, mitochondria and endoplasmic reticulum in the regulation of cytosolic Ca(2+) and catecholamine secretion of bovine adrenal chromaffin cells. J Neurochem 76: 210‐216, 2001.
 683. Yoo SH . Secretory granules in inositol 1,4,5‐trisphosphate‐dependent Ca2+ signaling in the cytoplasm of neuroendocrine cells. FASEB J 24: 653‐664, 2010.
 684. Yoshizaki T . Participation of muscarinic receptors on splanchnic‐adrenal transmission in the rat. Jpn J Pharmacol 23: 813‐816, 1973.
 685. Zachowski A , Henry JP , Devaux PF . Control of transmembrane lipid asymmetry in chromaffin granules by an ATP‐dependent protein. Nature 340: 75‐76, 1989.
 686. Zamponi GW , Currie KP . Regulation of Ca(V)2 calcium channels by G protein coupled receptors. Biochim Biophys Acta 1828: 1629‐1643, 2013.
 687. Zerbes M , Bunn SJ , Powis DA . Histamine causes Ca2+ entry via both a store‐operated and a store‐independent pathway in bovine adrenal chromaffin cells. Cell Calcium 23: 379‐386, 1998.
 688. Zhang C , Wu B , Beglopoulos V , Wines‐Samuelson M , Zhang D , Dragatsis I , Sudhof TC , Shen J . Presenilins are essential for regulating neurotransmitter release. Nature 460: 632‐636, 2009.
 689. Zhou Z , Misler S . Action potential‐induced quantal secretion of catecholamines from rat adrenal chromaffin cells. J Biol Chem 270: 3498‐3505, 1995.
 690. Zhou Z , Misler S . Amperometric detection of stimulus‐induced quantal release of catecholamines from cultured superior cervical ganglion neurons. Proc Natl Acad Sci USA 92: 6938‐6942, 1995.
 691. Zhou Z , Neher E . Calcium permeability of nicotinic acetylcholine receptor channels in bovine adrenal chromaffin cells. Pflug Archiv: Eur J Physiol 425: 511‐517, 1993.
 692. Zhou Z , Neher E . Mobile and immobile calcium buffers in bovine adrenal chromaffin cells. J Physiol 469: 245‐273, 1993.

 

Teaching Material

E. Carbone, R. Borges, L. E. Eiden, A. G. García, A. Hernández-Cruz. Chromaffin Cells of the Adrenal Medulla: Physiology, Pharmacology, and Disease. Compr Physiol 9: 2019, 1443-1502.

Didactic Synopsis

Major Teaching Points:

  • During stressful conflicts, chromaffin cells from the adrenal medulla release a surge of catecholamines (epinephrine and norepinephrine) into the bloodstream, which serves to prepare the different organs of the body for the fight-or-flight response, to escape from danger and survive.
  • The secretory response of chromaffin cells is triggered by a rapid elevation of the Ca2+ concentration in the cytosol, mainly contributed by Ca2+ entry via voltage-activated Ca2+ channels that open upon membrane depolarization as a consequence of the activation of postsynaptic cholinergic receptors by the neurotransmitter acetylcholine that is released from splanchnic nerve terminals (neurogenic control).
  • Chromaffin cells express a wide number of Na+, Ca2+, and K+ channels that are also able to generate spontaneous “neuronal-like” tonic and burst-firing patterns that can drive a sustained “nonneurogenic” release of catecholamines under different secretagogues stimulation.
  • The Ca2+ signal due to Ca2+ entry is subsequently modulated by Ca2+ uptake and release in mitochondria and endoplasmic reticulum. Intracellular Ca2+ controls granule movement to the plasma membrane, exocytosis and endocytosis, and couples cell activity to ATP generation.
  • Catecholamines, chromogranins, ATP, GABA, opioids, and other peptides are tightly packaged in secretory granules that release their contents by fusion with the plasmalemma. In this process (exocytosis) intervene several SNARE proteins that regulate the formation of a membrane fusion pore and its subsequent expansion to release the granule content.
  • Catecholamines are preferentially released from newly synthetized granules. Older granules may act as catecholamine donors for favoring their reload.
  • Secretagogues of the adrenal medulla stimulate both catecholamine/peptide synthesis and release via “stimulus-secretion-synthesis coupling.” Chromaffin cell excitability, Ca2+ channel currents, and the ensuing secretory responses are regulated further by several receptors for opioids, PACAP, ATP, GABA, and various other neurotransmitters.
  • Altered cell excitability, Ca2+ handling, and exocytosis defects have been reported in chromaffin cells from rodent models of diseases such as hypertension, Alzheimer’s disease, Huntington’s disease, autism, and amyotrophic lateral sclerosis, suggesting that the defective proteins of the disease also have an impact on chromaffin cell function.

Didactic Legends

The following legends to the figures that appear throughout the article are written to be useful for teaching.

Figure 1 Adrenal chromaffin cells behave electrically like neurons. As central and peripheral neurons, chromaffin cells generate spontaneous “all-or-none” action potentials repeated at variable frequency. They either occur as “regular ”or “irregular” trains of single APs (tonic firing) or in rapid sequence followed by long periods of inactivity (burst firing).

Figure 2 Chromaffin cells can fire action potentials in “bursts.” This is the result of an equilibrium between inward (Na+ and Ca2+) and outward (K+) ions passing currents through the cell membrane. Among the inward components, Ca2+ currents are the most important. They sustain the burst up to the last AP, while Na+ currents inactivate quickly. Among the three outward current components (Kv, SK, and BK), the SK is the only K+ current increasing during each AP within the burst. When SK is sufficiently large, it ends the burst by generating the repolarization phase.

Figure 3 Chromaffin cells are sensitive to pH changes. When the extracellular pH lowers, CCs depolarize, increase their firing frequency, and switch their firing from tonic to bursts. This increases Ca2+ entry into the cell and stimulates catecholamine release. Our body uses this CC chemosensory response to compensate blood acidification and hyperkalemia during extreme muscle exercise.

Figure 4 Understanding the electrical properties of CCs. This requires test protocols to determine the ability of the cell to generate trains of APs. In response to square current stimulation pulses, CCs fire trains of APs that fatigue (adapt) with time. The same occurs in central and peripheral neurons. Under control condition mouse CCs, APs have high frequency (16 Hz) at the beginning of the stimulus and adapt to a lower frequency at the end (4 Hz). Blockade of SK channels with apamin (panel c) changes the steady-state adaptation to a frequency of about 8 Hz (red curve in b).

Figure 5 Control of CA secretion by autocrine inhibition. CCs have a potent autocrine feedback inhibition that involves Ca2+ channels. N- and P/Q-type Ca2+ currents activate slowly when cells are perfused with endogenous ATP (panel A), the opioid met-enkephalin (panel B), or a soluble mixture (SLV) containing EPI, NE, ATP, and opioids purified from the secretory granules (panel C). The same slow activation is observed when the perfusion with external solution is stopped (panel E) or when the recorded CC is in a cell cluster (F). In both cases, the unwashed materials released inhibit the Ca2+ currents autocrinally. The slow Ca2+ current activation can be speeded up when the cell is depolarized with a prepulse to +100 mV, which rescues the fast channel activation (B and F).

Figure 6 Opposing modulation by cAMP/PKA and cGMP/PKG pathways on L-type Ca2+ channels. The top half; ß-AR mediated upregulation of CaV1.2 and CaV1.3 Ca2+ channels through the following sequence: AC activation, cAMP production, PKA activation, CaV1 channel phosphorylation, and increase in open channel probability. The bottom half: NO mediated downregulation of L-type Ca2+ channels through the following sequence: soluble guanylate cyclase (sGC) activation, cGMP production, PKG activation, CaV1 channel phosphorylation in a different site, cause a decrease in open channel probability. The consequence is that a modulator acting on either one of the two pathways is potentially able to increase or decrease the L-type Ca2+ currents that are responsible of AP firing and secretory activity of adrenal CCs.

Figure 7 Synergistic action of the cAMP/PKA and cGMP/PKG pathways on L-type Ca2+ channels. When the selective PKG inhibitor KT 5823 blocks the inhibitory effects of PKG and PKA is activated by forskolin (trace b in the left-bottom panel), a strong increase of the L-type Ca2+ current amplitude is induced, which drives burst firing and massive release of catecholamines. The opposite effect occurs when PKG is upregulated by the PKG activator 8-pCPT-cGMP and the PKA activity is reduced by the inhibitor H-89 blocks (trace b in the left-top panel).

Figure 8 Functional triads regulate local [Ca2+]i transients and exocytosis upon CCs stimulation. After cell depolarization by the neurotransmitter ACh, the voltage-dependent Ca2+ channels open (1). Ca2+ enters the cell, forming high-Ca2+ concentration microdomains (HCMD) of 10 µM or higher, near subplasmalemmal exocytotic sites (2). This high Ca2+ is required to trigger the fast exocytotic release of CAs (3). Ca2+ channels close, and HCMD quickly dissipates by mobile Ca2+ buffers, Ca2+ uptake by the ER Ca2+-ATPase (SERCA) (4) and Ca2+sequestration into the mitochondrial matrix by the Ca2+ uniporter (5). Inside both ER and mitochondria (Mito) matrix, [Ca2+] can reach about half a millimolar. Ca2+ can then be released back into the cytosol through the CICR mechanism via ryanodine receptors or lnsP3 receptors of the ER (6) or through the mitochondrial Na+/Ca2+ exchanger (7). Ca2+ diffusion redistributes Ca2+ at inner areas of the cell to generate low-Ca2+ microdomains (LCMD) of around half micromolar (8) that are required for the cytoskeleton-mediated Ca2+-dependent vesicle traffic and the refilling of RRP with new vesicles near subplasmalemmal exocytotic sites (9), securing new rounds of exocytosis.

Figure 9 The acidification is crucial for the accumulation of amines, ATP, and Ca2+ into the CG. A specific vesicular pump (V-ATPase) pumps H+ into the vesicle. To compensate the ?? gradient created by H+ pumping, Cl- channels open, allowing Cl- influx. Protons are used for accumulation by exchanging with catecholamines, Ca2+ or ATP. Dopamine is converted in NE inside the CG by the enzyme dopamine-?-hydroxylase (D???. In adrenergic cells, NE will be further transformed to EPI in the cytosol. Solutes are largely bound to the granule matrix, thus reducing the osmotic force caused by their elevated concentration.

Figure 10 Cycle of exocytosis and endocytosis of CGs. (A) For clarity, only one SNARE complex is shown, while some of the accessory proteins are omitted. 1: granule proteins are sorted and packaged into the Golgi apparatus. 2: granules are transported to the release sites. 3: actin filaments drive granules to specific tethering points. 4: “granule docking” to the membrane. 5: “priming,” both CG- and cell-membrane interact strongly by SNARE complexes, which promote the formation of the fusion pore. 6: the initial fusion pore allows limited exchange of water and solutes. 7: the extent of dilatation and duration of the O state regulate the amount of solutes released. The SNARE complex is disassembled. 8: partial exocytosis occurs when a dilated, but reversible, fusion pore allows the release of small molecules like peptides. 9: membrane recovery by endocytosis will either depend on clathrin and dynamin 2 or on a clathrin-independent mechanism mediated by dynamin 1. Granule rapidly acidifies. 10, 11: after clathrin disassembly, the granule travels deeper inside the cell toward either endosomes or lysosomes. Some of these steps are reversible (blue double head arrows). (B) Detail of the organization of the SNARE complex in the priming state.

Figure 11 Different pools of granules are involved in CA secretion. Even in the continuous presence of high concentrations of ACh, the secretion of CA for time unit in perfused cat adrenals progressively decays (A; gray bars). Something similar occurs when secretion is elicited by elevation of intracellular Ca2+ in permeabilized CCs (B). Exocytotic kinetics observed by cell capacitance measurements reveals at least two components, corresponding to the dynamic release of CGs pools (RRP and SRP; C). When perfused CCs are stimulated by pulses or agonist and CA secretion is continuously monitored by electrochemical detection, the release of CGs containing EGFP-labeled neuropeptide Y is found only during the first tree pulses, implying that newly produced CGs are the first to be released.

Figure 12 Signaling pathways of chromaffin cells. Acetylcholine (ACh) and PACAP are the transmitters controlling the release of catecholamines via splanchnic nerve stimulation in basal (ACh) and stimulated conditions (PACAP and ACh). These transmitters control not only the release of catecholamines and bioactive peptides from chromaffin cells, but also chromaffin cell gene expression that allows compensatory repletion of these molecules in chromaffin cells after they have been depleted by secretion. Both calcium and cAMP signaling control these processes (collectively called stimulus-secretion-synthesis coupling). In addition, circulating first messengers such as cytokines (IL-6 and TNF) and autocoids (such as histamine), through receptors on chromaffin cells, synergize with the splanchnic nerve secretagogues (e.g. PACAP) to amplify chromaffin cell secretion in response to inflammation. Stimulus-secretion-synthesis coupling also involves expression of connexins, molecules expressed on chromaffin cell membranes that further integrate, amplify, and coordinate whole-organ catecholamine secretion.

Figure 13 The adrenal medulla is an integrator of several inputs. CCs acts as a stress transducer and neuroimmunoinflammatory and cardiovascular regulator by integrating inputs from the first messengers mentioned in Figure 12, and providing secreted molecules that modulate the hormonal output of the adrenal cortex (e.g. glucocorticoids) to provide a mode of bidirectional communication to adjust secreted biomolecule activity across a broad range of organismic states, including stress, inflammation, and altered metabolic rate.

Figure 14 GABAA receptor can be excitatory or inhibitory in rat CCs. In immature neurons, GABA response is excitatory because NKCC1 is more active (A), while in mature neurons, GABA response is inhibitory because KCC2 is the dominant transporter (B). In ~44% of rat CCs, a GABAA agonist depolarizes and elevates [Ca2+]i (C), while it hyperpolarizes and causes a [Ca2+]i drop in ~26% of CCs (D). The scheme shows the anion-exchanger AE3 (pendrin), which accumulates Cl- in exchange for intracellular HCO3- and participates in Cl- transport into CCs. VDDC: voltage-dependent Ca2+ channel.

Figure 15 Ryanodine discloses an enhancement of CICR that underlies augmented CA secretion in CCs from hypertensive rats. Ryanodine, a potent blocker of RyRs, diminishes the amount of amperometric spikes elicited by a depolarizing pulse both in normotensive WKY (a) and hypertensive SHR CCs (b). The cumulative charge after RyR blockade is more significantly affected in SHR CCs (d) than in WKY CCs (c).Taken, with permission, from ref (566).

Figure 16 Chromaffin cells are sensitive to chronic and intermittent hypoxia.CCs respond to chronic hypoxia conditions by upregulating the usually unexpressed Cav3.2 (T-type) Ca2+ channels that regulate the release of neurotransmitters (EPI, NE, ATP, and opioids) at rather negative voltages (-50 mV; low threshold). This occurs through the activation of NOX2/NOX4, ROS, PLC, PKC, and the nuclear transcription factor HIF that leads to CACNA1H gene expression.

 


Related Articles:

Teaching Material

Contact Editor

Submit a note to the editor about this article by filling in the form below.

* Required Field

How to Cite

Emilio Carbone, Ricardo Borges, Lee E. Eiden, Antonio G. García, Arturo Hernández‐Cruz. Chromaffin Cells of the Adrenal Medulla: Physiology, Pharmacology, and Disease. Compr Physiol 2019, 9: 1443-1502. doi: 10.1002/cphy.c190003