1
|
Bukovics P, Lőrinczy D. Deconvolution Analysis of G and F-Actin Unfolding: Insights into the Thermal Stability and Structural Modifications Induced by PACAP. Int J Mol Sci 2025; 26:3336. [PMID: 40244223 PMCID: PMC11989792 DOI: 10.3390/ijms26073336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 03/31/2025] [Accepted: 04/02/2025] [Indexed: 04/18/2025] Open
Abstract
Actin, a key component of the cytoskeleton, undergoes significant structural and thermal changes in response to various regulatory factors, including the neuropeptide pituitary adenylate cyclase-activating polypeptide (PACAP). In this study, we applied deconvolution analysis to previously obtained differential scanning calorimetry (DSC) data to resolve overlapping thermal transitions in G- and F-actin unfolding. Our findings reveal that PACAP38 and PACAP6-38 significantly alter actin stability, increasing structural cooperativity in G-actin while reducing monomer-monomer interactions in F-actin. These thermodynamic changes suggest a potential role for PACAP in modulating actin polymerization and depolymerization dynamics, contributing to cytoskeletal remodeling.
Collapse
Affiliation(s)
- Péter Bukovics
- Department of Biophysics, Medical School, University of Pécs, Szigeti Str. 12, H-7624 Pécs, Hungary;
| | | |
Collapse
|
2
|
Hornung E, Robbins S, Srivastava A, Achanta S, Chen J, Cheng ZJ, Schwaber J, Vadigepalli R. Neuromodulatory co-expression in cardiac vagal motor neurons of the dorsal motor nucleus of the vagus. iScience 2024; 27:110549. [PMID: 39171288 PMCID: PMC11338141 DOI: 10.1016/j.isci.2024.110549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 05/31/2024] [Accepted: 07/16/2024] [Indexed: 08/23/2024] Open
Abstract
Vagal innervation is well known to be crucial to the maintenance of cardiac health, and to protect and recover the heart from injury. Only recently has this role been shown to depend on the activity of the underappreciated dorsal motor nucleus of the vagus (DMV). By combining neural tracing, transcriptomics, and anatomical mapping in male and female Sprague-Dawley rats, we characterize cardiac-specific neuronal phenotypes in the DMV. We find that the DMV cardiac-projecting neurons differentially express pituitary adenylate cyclase-activating polypeptide (PACAP), cocaine- and amphetamine-regulated transcript (CART), and synucleins, as well as evidence that they participate in neuromodulatory co-expression involving catecholamines. The significance of these findings is enhanced by previous knowledge of the role of PACAP at the heart and of the other neuromodulators in peripheral vagal targets.
Collapse
Affiliation(s)
- Eden Hornung
- Daniel Baugh Institute of Functional Genomics/Computational Biology, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Shaina Robbins
- Daniel Baugh Institute of Functional Genomics/Computational Biology, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Ankita Srivastava
- Daniel Baugh Institute of Functional Genomics/Computational Biology, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Sirisha Achanta
- Daniel Baugh Institute of Functional Genomics/Computational Biology, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Jin Chen
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, BMS Building 20, Room 230, 4110 Libra Drive, Orlando, FL 32816, USA
| | - Zixi Jack Cheng
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, BMS Building 20, Room 230, 4110 Libra Drive, Orlando, FL 32816, USA
| | - James Schwaber
- Daniel Baugh Institute of Functional Genomics/Computational Biology, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Rajanikanth Vadigepalli
- Daniel Baugh Institute of Functional Genomics/Computational Biology, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
3
|
Puri S, Kenyon BM, Hamrah P. Immunomodulatory Role of Neuropeptides in the Cornea. Biomedicines 2022; 10:1985. [PMID: 36009532 PMCID: PMC9406019 DOI: 10.3390/biomedicines10081985] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/11/2022] [Accepted: 08/12/2022] [Indexed: 12/21/2022] Open
Abstract
The transparency of the cornea along with its dense sensory innervation and resident leukocyte populations make it an ideal tissue to study interactions between the nervous and immune systems. The cornea is the most densely innervated tissue of the body and possesses both immune and vascular privilege, in part due to its unique repertoire of resident immune cells. Corneal nerves produce various neuropeptides that have a wide range of functions on immune cells. As research in this area expands, further insights are made into the role of neuropeptides and their immunomodulatory functions in the healthy and diseased cornea. Much remains to be known regarding the details of neuropeptide signaling and how it contributes to pathophysiology, which is likely due to complex interactions among neuropeptides, receptor isoform-specific signaling events, and the inflammatory microenvironment in disease. However, progress in this area has led to an increase in studies that have begun modulating neuropeptide activity for the treatment of corneal diseases with promising results, necessitating the need for a comprehensive review of the literature. This review focuses on the role of neuropeptides in maintaining the homeostasis of the ocular surface, alterations in disease settings, and the possible therapeutic potential of targeting these systems.
Collapse
Affiliation(s)
- Sudan Puri
- Center for Translational Ocular Immunology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA 02111, USA
- Department of Ophthalmology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Brendan M. Kenyon
- Center for Translational Ocular Immunology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA 02111, USA
- Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University, Boston, MA 02111, USA
| | - Pedram Hamrah
- Center for Translational Ocular Immunology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA 02111, USA
- Department of Ophthalmology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA 02111, USA
- Program in Neuroscience, Graduate School of Biomedical Sciences, Tufts University, Boston, MA 02111, USA
- Departments of Immunology and Neuroscience, Tufts University School of Medicine, Boston, MA 02111, USA
- Cornea Service, Tufts New England Eye Center, Boston, MA 02111, USA
| |
Collapse
|
4
|
Lizot G, Pasqualin C, Tissot A, Pagès S, Faivre JF, Chatelier A. Molecular and functional characterization of the mouse intracardiac nervous system. Heart Rhythm 2022; 19:1352-1362. [PMID: 35447308 DOI: 10.1016/j.hrthm.2022.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 04/11/2022] [Accepted: 04/13/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND The intracardiac nervous system (ICNS) refers to clusters of neurons, located within the heart, which participate to the neuronal regulation of cardiac functions and which are involved in the initiation of cardiac arrhythmias. Therefore, deciphering its role in cardiac physiology and physiopathology is mandatory. OBJECTIVE The aim of this study is to provide a phenotypic, electrophysiological and pharmacological characterization of the mouse ICNS, which is still poorly characterized. METHODS Global cardiac innervation and phenotypic diversity were investigated using immunohistochemistry on cleared murine heart and on tissue sections. Patch clamp technique was used for electrophysiological and pharmacological characterization of isolated mouse intracardiac neurons. RESULTS We have identified the expression of seven distinct neuronal markers within mouse ICNS, thus proving the neurochemical diversity of this network. Of note, it was the first time that the existence of neurons expressing the calcium binding protein calbindin, the neuropeptide Y (NPY) and the cocain and amphetamine regulated transcript (CART) peptide, was described in the mouse. Electrophysiological studies also revealed the existence of four different neuronal populations based on their electrical behavior. Finally, we showed that these neurons can be modulated by several neuromodulators. CONCLUSION This study showed that mouse ICNS presents a molecular and functional complexity similar to other species, and is therefore a suitable model to decipher the role of individual neuronal subtypes regarding the modulation of cardiac function and the initiation of cardiac arrhythmias.
Collapse
Affiliation(s)
| | - Côme Pasqualin
- PReTI laboratory, UR 24184, University of Poitiers, France
| | - Audrey Tissot
- Wyss Center for Bio and Neuroengineering, Campus Biotech, Geneva, Switzerland
| | - Stephane Pagès
- Wyss Center for Bio and Neuroengineering, Campus Biotech, Geneva, Switzerland
| | | | | |
Collapse
|
5
|
May V, Johnson GC, Hammack SE, Braas KM, Parsons RL. PAC1 Receptor Internalization and Endosomal MEK/ERK Activation Is Essential for PACAP-Mediated Neuronal Excitability. J Mol Neurosci 2021; 71:1536-1542. [PMID: 33675454 PMCID: PMC8450765 DOI: 10.1007/s12031-021-01821-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 02/22/2021] [Indexed: 12/15/2022]
Abstract
Pituitary adenylate cyclase activating polypeptide (PACAP, Adcyap1) activation of PAC1 receptors (Adcyap1r1) can significantly increase the excitability of diverse neurons through differential mechanisms. For guinea pig cardiac neurons, the modulation of excitability can be mediated in part by PAC1 receptor plasma membrane G protein-dependent activation of adenylyl cyclase and downstream signaling cascades. By contrast, PAC1 receptor-mediated excitability of hippocampal dentate gyrus granule cells appears independent of membrane-delimited AC/cAMP/PKA and PLC/PKC signaling. For both neuronal types, there is mechanistic convergence demonstrating that endosomal PAC1 receptor signaling has prominent roles. In these models, neuronal exposure to Pitstop2 to inhibit β-arrestin/clathrin-mediated PAC1 receptor internalization eliminates PACAP modulation of excitability. β-arrestin is a scaffold for a number of effectors especially MEK/ERK and notably, paradigms that inhibit PAC1 receptor endosome formation and ERK signaling also blunt the PACAP-induced increase in excitability. Detailed PAC1 receptor internalization and endosomal ERK signaling mechanisms have been confirmed in HEK PAC1R-EGFP cells and shown to be long lasting which appear to recapitulate the sustained electrophysiological responses. Thus, PAC1 receptor internalization/endosomal recruitment efficiently and efficaciously activates MEK/ERK signaling and appears to represent a singular and critical common denominator in regulating neuronal excitability by PACAP.
Collapse
Affiliation(s)
- Victor May
- Departmental of Neurological Sciences, University of Vermont College of Medicine, Burlington, VT, USA.
| | - Gregory C Johnson
- Department of Psychological Science, University of Vermont, Burlington, VT, USA
| | - Sayamwong E Hammack
- Department of Psychological Science, University of Vermont, Burlington, VT, USA
| | - Karen M Braas
- Departmental of Neurological Sciences, University of Vermont College of Medicine, Burlington, VT, USA
| | - Rodney L Parsons
- Departmental of Neurological Sciences, University of Vermont College of Medicine, Burlington, VT, USA
| |
Collapse
|
6
|
Tedoldi A, Argent L, Montgomery JM. The role of the tripartite synapse in the heart: how glial cells may contribute to the physiology and pathophysiology of the intracardiac nervous system. Am J Physiol Cell Physiol 2020; 320:C1-C14. [PMID: 33085497 DOI: 10.1152/ajpcell.00363.2020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
One of the major roles of the intracardiac nervous system (ICNS) is to act as the final site of signal integration for efferent information destined for the myocardium to enable local control of heart rate and rhythm. Multiple subtypes of neurons exist in the ICNS where they are organized into clusters termed ganglionated plexi (GP). The majority of cells in the ICNS are actually glial cells; however, despite this, ICNS glial cells have received little attention to date. In the central nervous system, where glial cell function has been widely studied, glia are no longer viewed simply as supportive cells but rather have been shown to play an active role in modulating neuronal excitability and synaptic plasticity. Pioneering studies have demonstrated that in addition to glia within the brain stem, glial cells within multiple autonomic ganglia in the peripheral nervous system, including the ICNS, can also act to modulate cardiovascular function. Clinically, patients with atrial fibrillation (AF) undergoing catheter ablation show high plasma levels of S100B, a protein produced by cardiac glial cells, correlated with decreased AF recurrence. Interestingly, S100B also alters GP neuron excitability and neurite outgrowth in the ICNS. These studies highlight the importance of understanding how glial cells can affect the heart by modulating GP neuron activity or synaptic inputs. Here, we review studies investigating glia both in the central and peripheral nervous systems to discuss the potential role of glia in controlling cardiac function in health and disease, paying particular attention to the glial cells of the ICNS.
Collapse
Affiliation(s)
- Angelo Tedoldi
- Department of Physiology, University of Auckland, Auckland, New Zealand.,Manaaki Mānawa Centre for Heart Research, University of Auckland, Auckland, New Zealand
| | - Liam Argent
- Department of Physiology, University of Auckland, Auckland, New Zealand.,Manaaki Mānawa Centre for Heart Research, University of Auckland, Auckland, New Zealand
| | - Johanna M Montgomery
- Department of Physiology, University of Auckland, Auckland, New Zealand.,Manaaki Mānawa Centre for Heart Research, University of Auckland, Auckland, New Zealand
| |
Collapse
|
7
|
Intrabladder PAC1 Receptor Antagonist, PACAP(6-38), Reduces Urinary Bladder Frequency and Pelvic Sensitivity in Mice Exposed to Repeated Variate Stress (RVS). J Mol Neurosci 2020; 71:1575-1588. [PMID: 32613552 DOI: 10.1007/s12031-020-01649-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 06/22/2020] [Indexed: 12/18/2022]
Abstract
Stress causes symptom exacerbation in functional disorders of the urinary bladder. However, the potential mediators and underlying mechanisms of stress effects on micturition reflex function are unknown. We have characterized PACAP (Adcyap1) and PAC1 receptor (Adcyap1r1) signaling in stress-induced urinary bladder dysfunction in mice. We determined PACAP and PAC1 transcripts and protein expressions in the urinary bladder and lumbosacral dorsal root ganglia (DRG) and spinal cord in repeated variate stress (RVS) or control mouse (handling only) groups. RVS in mice significantly (p ≤ 0.01) increased serum corticosterone and urinary bladder NGF content and decreased weight gain. PACAP and PAC1 mRNA and protein were differentially regulated in lower urinary tract tissues with changes observed in lumbosacral DRG and spinal cord but not in urinary bladder. RVS exposure in mice significantly (p ≤ 0.01) increased (2.5-fold) voiding frequency as determined using conscious cystometry. Intrabladder administration of the PAC1 receptor antagonist, PACAP(6-38) (300 nM), significantly (p ≤ 0.01) increased infused volume (1.5-2.7-fold) to elicit a micturition event and increased the intercontraction interval (i.e., decreased voiding frequency) in mice exposed to RVS and in control mice, but changes were smaller in magnitude in control mice. We also evaluated the effect of PAC1 blockade at the level of the urinary bladder on pelvic sensitivity in RVS or control mouse groups using von Frey filament testing. Intrabladder administration of PACAP(6-38) (300 nM) significantly (p ≤ 0.01) reduced pelvic sensitivity following RVS. PACAP/receptor signaling in the CNS and PNS contributes to increased voiding frequency and pelvic sensitivity following RVS and may represent a potential target for therapeutic intervention.
Collapse
|
8
|
Johnson GC, Parsons RL, May V, Hammack SE. Pituitary adenylate cyclase-activating polypeptide-induced PAC1 receptor internalization and recruitment of MEK/ERK signaling enhance excitability of dentate gyrus granule cells. Am J Physiol Cell Physiol 2020; 318:C870-C878. [PMID: 32186931 DOI: 10.1152/ajpcell.00065.2020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Pituitary adenylate cyclase activating polypeptide (PACAP; ADCYAP1) is a pleiotropic neuropeptide widely distributed in both the peripheral and central nervous systems. PACAP and its specific cognate PAC1 receptor (ADCYAP1R1) play critical roles in the homeostatic maintenance of multiple physiological and behavioral systems. Notably, maladaptations in the PACAPergic system have been associated with several psychopathologies related to fear and anxiety. PAC1 receptor transcripts are highly expressed in granule cells of the dentate gyrus (DG). Here, we examined the direct effects of PACAP on DG granule cells in brain slices using whole cell patch recordings in current clamp mode. PACAP significantly increased the intrinsic excitability of DG granule cells via PAC1 receptor activation. This increased excitability was not mediated by adenylyl cyclase/cAMP or phospholipase C/PKC activation, but instead via activation of an extracellular signal-regulated kinase (ERK) signaling pathway initiated through PAC1 receptor endocytosis/endosomal signaling. PACAP failed to increase excitability in DG granule cells pretreated with the persistent sodium current blocker riluzole, suggesting that the observed PACAP effects required this component of the inward sodium current.
Collapse
Affiliation(s)
- Gregory C Johnson
- Department of Psychological Science, University of Vermont, Burlington, Vermont
| | - Rodney L Parsons
- Department of Neurological Sciences, University of Vermont, Burlington, Vermont
| | - Victor May
- Department of Neurological Sciences, University of Vermont, Burlington, Vermont
| | - Sayamwong E Hammack
- Department of Psychological Science, University of Vermont, Burlington, Vermont
| |
Collapse
|
9
|
Parsons RL, May V. PACAP-Induced PAC1 Receptor Internalization and Recruitment of Endosomal Signaling Regulate Cardiac Neuron Excitability. J Mol Neurosci 2019; 68:340-347. [PMID: 30054797 PMCID: PMC6348136 DOI: 10.1007/s12031-018-1127-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 07/18/2018] [Indexed: 11/27/2022]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP, Adcyap1) activation of PAC1 receptors (Adcyap1r1) significantly increases excitability of guinea pig cardiac neurons. This modulation of excitability is mediated in part by plasma membrane G protein-dependent activation of adenylyl cyclase and downstream signaling cascades, as well as by endosomal signaling mechanisms. PACAP/PAC1 receptor-mediated activation of plasma membrane adenylyl cyclase (AC) and the resulting increase in cellular cAMP enhances a hyperpolarization-induced nonselective cationic current Ih, which contributes to the PACAP-induced increase in cardiac neuron excitability. Further, PACAP-mediated AC/cAMP/PKA downstream signaling also appears to enhance cardiac neuron IT to facilitate the excitatory responses. PACAP activation of PAC1 receptors rapidly stimulates receptor internalization, and reducing ambient temperature or treatments with the clathrin inhibitor Pitstop2 or the dynamin I/II inhibitor dynasore to block endocytic events can suppress PACAP-enhanced neuronal excitability. Thus, endocytosis inhibitors essentially eliminate PACAP-enhanced excitability suggesting that endosomal platforms represent a primary signaling mechanism. Endosomal signaling is associated canonically with ERK activation and in accord, PACAP-enhanced cardiac neuron excitability is reduced by MEK inhibitor pretreatments. PACAP activation of MEK/ERK signaling can enhance currents through voltage-dependent Nav1.7 channels. Hence, PACAP-induced PAC1 receptor internalization/endosomal signaling, recruitment of MEK/ERK signaling, and modulation of Nav1.7 are implicated as key mechanisms contributing to the PACAP-enhanced neuronal excitability. PACAP/PAC1 receptor-mediated endosomal ERK signaling in central circuits can play key roles in development of chronic pain and anxiety-related responses; thus, PAC1 endosomal signaling likely participates in a variety of homeostatic responses within neuronal circuits in the CNS.
Collapse
Affiliation(s)
- Rodney L Parsons
- Departmental of Neurological Sciences, Robert Larner College of Medicine, University of Vermont, Burlington, VT, USA.
| | - Victor May
- Departmental of Neurological Sciences, Robert Larner College of Medicine, University of Vermont, Burlington, VT, USA
| |
Collapse
|
10
|
Arichi S, Sasaki-Hamada S, Kadoya Y, Ogata M, Ishibashi H. Excitatory effect of bradykinin on intrinsic neurons of the rat heart. Neuropeptides 2019; 75:65-74. [PMID: 31047706 DOI: 10.1016/j.npep.2019.04.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 04/07/2019] [Accepted: 04/23/2019] [Indexed: 01/16/2023]
Abstract
The heart receives sympathetic and parasympathetic innervation through the intrinsic cardiac nervous system. Although bradykinin (BK) has negative inotropic and chronotropic properties of cardiac contraction, the direct effect of BK on the intrinsic neural network of the heart is still unclear. In the present study, the effect of BK on the intracardiac ganglion neurons isolated from rats was investigated using the perforated patch-clamp technique. Under current-clamp conditions, application of 0.1 μM BK depolarized the membrane, accompanied by repetitive firing of action potentials. When BK was applied repeatedly, the second responses were considerably less intense than the first application. The BK action was fully inhibited by the B2 receptor antagonist Hoe-140, but not by the B1 receptor antagonist des-Arg9-[Leu8]-BK. The BK response was mimicked by the B2 agonist [Hyp3]-BK. The BK-induced depolarization was inhibited by the phospholipase C inhibitor U-73122. BK evoked inward currents under voltage-clamp conditions at a holding potential of -60 mV. Removal of extracellular Ca2+ markedly increased the BK-induced currents, suggesting an involvement of Ca2+-permeable non-selective cation channels. The muscarinic agonist oxotremorine-M (OxoM) also elicited the extracellular Ca2+-sensitive cationic currents. The OxoM response did not exhibit rundown with repeated agonist application. The amplitude of current evoked by 1 μM OxoM was comparable to that induced by 0.1 μM BK. Co-application of 0.1 μM BK and 1 μM OxoM elicited the current whose peak amplitude was almost the same as that elicited by OxoM alone, suggesting that BK and OxoM activate same cation channels. BK also reduced the amplitude of M-current, while the M-current inhibitor XE-991 affected neither resting membrane potential nor the BK-induced depolarization. From these results, we suggest that BK regulates excitability of intrinsic cardiac neurons by both an activation of non-selective cation channels and an inhibition of M-type K+ channels through B2 receptors.
Collapse
Affiliation(s)
- Shiho Arichi
- Department of Physiology, School of Allied Health Sciences, Kitasato University, Sagamihara 252-0373, Japan; Department of Brain Science, Kitasato University Graduate School of Medical Sciences, Sagamihara 252-0373, Japan
| | - Sachie Sasaki-Hamada
- Department of Physiology, School of Allied Health Sciences, Kitasato University, Sagamihara 252-0373, Japan
| | - Yuichi Kadoya
- Department of Anatomical Science, School of Allied Health Sciences, Kitasato University, Sagamihara 252-0373, Japan
| | - Masanori Ogata
- Department of Physiology, School of Allied Health Sciences, Kitasato University, Sagamihara 252-0373, Japan; Department of Brain Science, Kitasato University Graduate School of Medical Sciences, Sagamihara 252-0373, Japan
| | - Hitoshi Ishibashi
- Department of Physiology, School of Allied Health Sciences, Kitasato University, Sagamihara 252-0373, Japan; Department of Brain Science, Kitasato University Graduate School of Medical Sciences, Sagamihara 252-0373, Japan.
| |
Collapse
|
11
|
Ojala J, Tooke K, Hsiang H, Girard BM, May V, Vizzard MA. PACAP/PAC1 Expression and Function in Micturition Pathways. J Mol Neurosci 2018; 68:357-367. [PMID: 30259317 DOI: 10.1007/s12031-018-1170-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 09/13/2018] [Indexed: 12/29/2022]
Abstract
Neural injury, inflammation, or diseases commonly and adversely affect micturition reflex function that is organized by neural circuits in the CNS and PNS. One neuropeptide receptor system, pituitary adenylate cyclase-activating polypeptide (PACAP; Adcyap1), and its cognate receptor, PAC1 (Adcyap1r1), have tissue-specific distributions in the lower urinary tract. PACAP and associated receptors are expressed in the LUT and exhibit changes in expression, distribution, and function in preclinical animal models of bladder pain syndrome (BPS)/interstitial cystitis (IC), a chronic, visceral pain syndrome characterized by pain, and LUT dysfunction. Blockade of the PACAP/PAC1 receptor system reduces voiding frequency and somatic (e.g., hindpaw, pelvic) sensitivity in preclinical animal models and a transgenic mouse model that mirrors some clinical symptoms of BPS/IC. The PACAP/receptor system in micturition pathways may represent a potential target for therapeutic intervention to reduce LUT dysfunction following urinary bladder inflammation.
Collapse
Affiliation(s)
- Jacqueline Ojala
- Department of Neurological Sciences, The Robert Larner, M.D. College of Medicine at The University of Vermont, Given Building, D405A, Burlington, VT, 05405, USA
| | - Katharine Tooke
- Department of Neurological Sciences, The Robert Larner, M.D. College of Medicine at The University of Vermont, Given Building, D405A, Burlington, VT, 05405, USA
| | - Harrison Hsiang
- Department of Neurological Sciences, The Robert Larner, M.D. College of Medicine at The University of Vermont, Given Building, D405A, Burlington, VT, 05405, USA
| | - Beatrice M Girard
- Department of Neurological Sciences, The Robert Larner, M.D. College of Medicine at The University of Vermont, Given Building, D405A, Burlington, VT, 05405, USA
| | - Victor May
- Department of Neurological Sciences, The Robert Larner, M.D. College of Medicine at The University of Vermont, Given Building, D405A, Burlington, VT, 05405, USA
| | - Margaret A Vizzard
- Department of Neurological Sciences, The Robert Larner, M.D. College of Medicine at The University of Vermont, Given Building, D405A, Burlington, VT, 05405, USA.
| |
Collapse
|
12
|
Pituitary Adenylate Cyclase-Activating Peptide (PACAP) Signaling and the Dark Side of Addiction. J Mol Neurosci 2018; 68:453-464. [PMID: 30074172 DOI: 10.1007/s12031-018-1147-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 07/27/2018] [Indexed: 12/15/2022]
Abstract
While addiction to drugs of abuse represents a significant health problem worldwide, the behavioral and neural mechanisms that underlie addiction and relapse are largely unclear. The concept of the dark side of addiction, developed and explored by George Koob and colleagues, describes a systematic decrease in reward-related processing following drug self-administration and subsequent recruitment of anti-reward (i.e., stress) systems. Indeed, the activation of central nervous system (CNS) stress-response systems by drugs of abuse is contributory not only to mood and anxiety-related disorders but critical to both the maintenance of addiction and relapse following abstinence. In both human and animal studies, compounds that activate the bed nucleus of the stria terminalis (BNST) have roles in stress-related behaviors and addiction processes. The activation of pituitary adenylate cyclase-activating peptide (PACAP) systems in the BNST mediates many consequences of chronic stressor exposure that may engage in part downstream corticotropin-releasing hormone (CRH) signaling. Similar to footshock stress, the BNST administration of PACAP or the PAC1 receptor-specific agonist maxadilan can facilitate relapse following extinction of cocaine-seeking behavior. Further, in the same paradigm, the footshock-induced relapse could be attenuated following BNST pretreatment with PAC1 receptor antagonist PACAP6-38, implicating PACAP systems as critical components underlying stress-induced reinstatement. In congruence with previous work, the PAC1 receptor internalization and endosomal MEK/ERK signaling appear contributory mechanisms to the addiction processes. The studies offer new insights and approaches to addiction and relapse therapeutics.
Collapse
|
13
|
PACAP38-Mediated Bladder Afferent Nerve Activity Hyperexcitability and Ca 2+ Activity in Urothelial Cells from Mice. J Mol Neurosci 2018; 68:348-356. [PMID: 30022438 DOI: 10.1007/s12031-018-1119-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Accepted: 07/10/2018] [Indexed: 12/11/2022]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP; Adcyap1) and its cognate PAC1 receptor (Adcyap1r1) have tissue-specific distributions in the lower urinary tract (LUT). The afferent limb of the micturition reflex is often compromised following bladder injury, disease, and inflammatory conditions. We have previously demonstrated that PACAP signaling contributes to increased voiding frequency and decreased bladder capacity with cystitis. Thus, the present studies investigated the sensory components (e.g., urothelial cells, bladder afferent nerves) of the urinary bladder that may underlie the pathophysiology of aberrant PACAP activation. We utilized bladder-pelvic nerve preparations and urothelial sheet preparations to characterize PACAP-induced bladder afferent nerve discharge with distention and PACAP-induced Ca2+ activity, respectively. We determined that PACAP38 (100 nM) significantly (p ≤ 0.01) increased bladder afferent nerve activity with distention that was blocked with a PAC1/VPAC2 receptor antagonist PACAP6-38 (300 nM). PACAP38 (100 nM) also increased Ca2+ activity in urothelial cells over that observed in control preparations. Taken together, these results establish a role for PACAP signaling in bladder sensory components (e.g., urothelial cells, bladder afferent nerves) that may ultimately facilitate increased voiding frequency.
Collapse
|
14
|
Ashton JL, Burton RAB, Bub G, Smaill BH, Montgomery JM. Synaptic Plasticity in Cardiac Innervation and Its Potential Role in Atrial Fibrillation. Front Physiol 2018; 9:240. [PMID: 29615932 PMCID: PMC5869186 DOI: 10.3389/fphys.2018.00240] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 03/06/2018] [Indexed: 12/30/2022] Open
Abstract
Synaptic plasticity is defined as the ability of synapses to change their strength of transmission. Plasticity of synaptic connections in the brain is a major focus of neuroscience research, as it is the primary mechanism underpinning learning and memory. Beyond the brain however, plasticity in peripheral neurons is less well understood, particularly in the neurons innervating the heart. The atria receive rich innervation from the autonomic branch of the peripheral nervous system. Sympathetic neurons are clustered in stellate and cervical ganglia alongside the spinal cord and extend fibers to the heart directly innervating the myocardium. These neurons are major drivers of hyperactive sympathetic activity observed in heart disease, ventricular arrhythmias, and sudden cardiac death. Both pre- and postsynaptic changes have been observed to occur at synapses formed by sympathetic ganglion neurons, suggesting that plasticity at sympathetic neuro-cardiac synapses is a major contributor to arrhythmias. Less is known about the plasticity in parasympathetic neurons located in clusters on the heart surface. These neuronal clusters, termed ganglionated plexi, or “little brains,” can independently modulate neural control of the heart and stimulation that enhances their excitability can induce arrhythmia such as atrial fibrillation. The ability of these neurons to alter parasympathetic activity suggests that plasticity may indeed occur at the synapses formed on and by ganglionated plexi neurons. Such changes may not only fine-tune autonomic innervation of the heart, but could also be a source of maladaptive plasticity during atrial fibrillation.
Collapse
Affiliation(s)
- Jesse L Ashton
- Department of Physiology, University of Auckland, Auckland, New Zealand
| | | | - Gil Bub
- Department of Physiology, McGill University, Montreal, QC, Canada
| | - Bruce H Smaill
- Department of Physiology, University of Auckland, Auckland, New Zealand.,Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | | |
Collapse
|
15
|
Girard BM, Tooke K, Vizzard MA. PACAP/Receptor System in Urinary Bladder Dysfunction and Pelvic Pain Following Urinary Bladder Inflammation or Stress. Front Syst Neurosci 2017; 11:90. [PMID: 29255407 PMCID: PMC5722809 DOI: 10.3389/fnsys.2017.00090] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 11/16/2017] [Indexed: 12/11/2022] Open
Abstract
Complex organization of CNS and PNS pathways is necessary for the coordinated and reciprocal functions of the urinary bladder, urethra and urethral sphincters. Injury, inflammation, psychogenic stress or diseases that affect these nerve pathways and target organs can produce lower urinary tract (LUT) dysfunction. Numerous neuropeptide/receptor systems are expressed in the neural pathways of the LUT and non-neural components of the LUT (e.g., urothelium) also express peptides. One such neuropeptide receptor system, pituitary adenylate cyclase-activating polypeptide (PACAP; Adcyap1) and its cognate receptor, PAC1 (Adcyap1r1), have tissue-specific distributions in the LUT. Mice with a genetic deletion of PACAP exhibit bladder dysfunction and altered somatic sensation. PACAP and associated receptors are expressed in the LUT and exhibit neuroplastic changes with neural injury, inflammation, and diseases of the LUT as well as psychogenic stress. Blockade of the PACAP/PAC1 receptor system reduces voiding frequency in preclinical animal models and transgenic mouse models that mirror some clinical symptoms of bladder dysfunction. A change in the balance of the expression and resulting function of the PACAP/receptor system in CNS and PNS bladder reflex pathways may underlie LUT dysfunction including symptoms of urinary urgency, increased voiding frequency, and visceral pain. The PACAP/receptor system in micturition pathways may represent a potential target for therapeutic intervention to reduce LUT dysfunction.
Collapse
Affiliation(s)
| | | | - Margaret A. Vizzard
- Department of Neurological Sciences, Larner College of Medicine, The University of Vermont, Burlington, VT, United States
| |
Collapse
|
16
|
Pituitary adenylate cyclase activating polypeptide induces long-term, transcription-dependent plasticity and remodeling at autonomic synapses. Mol Cell Neurosci 2017; 85:170-182. [DOI: 10.1016/j.mcn.2017.10.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 09/19/2017] [Accepted: 10/06/2017] [Indexed: 12/28/2022] Open
|
17
|
Tompkins JD, Clason TA, Buttolph TR, Girard BM, Linden AK, Hardwick JC, Merriam LA, May V, Parsons RL. Src family kinase inhibitors blunt PACAP-induced PAC1 receptor endocytosis, phosphorylation of ERK, and the increase in cardiac neuron excitability. Am J Physiol Cell Physiol 2017; 314:C233-C241. [PMID: 29141923 DOI: 10.1152/ajpcell.00223.2017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Pituitary adenylate cyclase activating polypeptide (PACAP, Adcyap1) activation of PAC1 receptors ( Adcyap1r1) significantly increases excitability of guinea pig cardiac neurons. This modulation of excitability is mediated in part by plasma membrane G protein-dependent activation of adenylyl cyclase and downstream signaling cascades. However, additional mechanisms responsible for the enhanced excitability are activated following internalization of the PAC1 receptor and endosomal signaling. Src family kinases play critical roles mediating endocytosis of many trophic factor and G protein-coupled receptors. The present study investigated whether Src family kinases also support the PACAP-induced PAC1 receptor internalization, phosphorylation of ERK, and enhanced neuronal excitability. Using human embryonic kidney cells stably expressing a green fluorescent protein-tagged PAC1 receptor, treatment with the Src family kinase inhibitor PP2 (10 µM) markedly reduced the PACAP-induced PAC1 receptor internalization, and in parallel, both PP2 and Src inhibitor 1 (Src-1, 2 µM) reduced ERK activation determined by Western blot analysis. In contrast, Src family kinase inhibitors did not eliminate a PACAP-induced rise in global calcium generated by inositol (1,4,5)-trisphosphate-induced release of calcium from endoplasmic reticulum stores. From confocal analysis of phosphorylated ERK immunostaining, PP2 treatment significantly attenuated PACAP activation of ERK in neurons within cardiac ganglia whole mount preparations. Intracellular recordings demonstrated that PP2 also significantly blunted a PACAP-induced increase in cardiac neuron excitability. These studies demonstrate Src-related kinase activity in PAC1 receptor internalization, activation of MEK/ERK signaling, and regulation of neuronal excitability. The present results provide further support for the importance of PAC1 receptor endosomal signaling as a key mechanism regulating cellular function.
Collapse
Affiliation(s)
- John D Tompkins
- Department of Medicine (Cardiology), David Geffen School of Medicine, University of California , Los Angeles, California
| | - Todd A Clason
- Department of Neurological Sciences, Robert Larner MD College of Medicine, University of Vermont , Burlington, Vermont
| | - Thomas R Buttolph
- Department of Neurological Sciences, Robert Larner MD College of Medicine, University of Vermont , Burlington, Vermont
| | - Beatrice M Girard
- Department of Neurological Sciences, Robert Larner MD College of Medicine, University of Vermont , Burlington, Vermont
| | - Anne K Linden
- Department of Neurological Sciences, Robert Larner MD College of Medicine, University of Vermont , Burlington, Vermont
| | | | - Laura A Merriam
- Department of Neurological Sciences, Robert Larner MD College of Medicine, University of Vermont , Burlington, Vermont
| | - Victor May
- Department of Neurological Sciences, Robert Larner MD College of Medicine, University of Vermont , Burlington, Vermont
| | - Rodney L Parsons
- Department of Neurological Sciences, Robert Larner MD College of Medicine, University of Vermont , Burlington, Vermont
| |
Collapse
|
18
|
Hardwick JC, Clason TA, Tompkins JD, Girard BM, Baran CN, Merriam LA, May V, Parsons RL. Recruitment of endosomal signaling mediates the forskolin modulation of guinea pig cardiac neuron excitability. Am J Physiol Cell Physiol 2017; 313:C219-C227. [PMID: 28592413 DOI: 10.1152/ajpcell.00094.2017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 05/31/2017] [Accepted: 06/01/2017] [Indexed: 12/18/2022]
Abstract
Forskolin, a selective activator of adenylyl cyclase (AC), commonly is used to establish actions of G protein-coupled receptors (GPCRs) that are initiated primarily through activation of AC/cAMP signaling pathways. In the present study, forskolin was used to evaluate the potential role of AC/cAMP, which is a major signaling mechanism for the pituitary adenylate cyclase-activating polypeptide (PACAP)-selective PAC1 receptor, in the regulation of guinea pig cardiac neuronal excitability. Forskolin (5-10 µM) increases excitability in ~60% of the cardiac neurons. The forskolin-mediated increase in excitability was considered related to cAMP regulation of a cyclic nucleotide gated channel or via protein kinase A (PKA)/ERK signaling, mechanisms that have been linked to PAC1 receptor activation. However, unlike PACAP mechanisms, forskolin enhancement of excitability was not significantly reduced by treatment with cesium to block currents through hyperpolarization-activated nonselective cation channels (Ih) or by treatment with PD98059 to block MEK/ERK signaling. In contrast, treatment with the clathrin inhibitor Pitstop2 or the dynamin inhibitor dynasore eliminated the forskolin-induced increase in excitability; treatments with the inactive Pitstop analog or PP2 treatment to inhibit Src-mediated endocytosis mechanisms were ineffective. The PKA inhibitor KT5702 significantly suppressed the forskolin-induced change in excitability; further, KT5702 and Pitstop2 reduced the forskolin-stimulated MEK/ERK activation in cardiac neurons. Collectively, the present results suggest that forskolin activation of AC/cAMP/PKA signaling leads to the recruitment of clathrin/dynamin-dependent endosomal transduction cascades, including MEK/ERK signaling, and that endosomal signaling is the critical mechanism underlying the forskolin-induced increase in cardiac neuron excitability.
Collapse
Affiliation(s)
| | - Todd A Clason
- Department of Neurological Sciences, Robert Larner MD College of Medicine, University of Vermont, Burlington, Vermont
| | - John D Tompkins
- Department of Medicine (Cardiology), David Geffen School of Medicine, University of California, Los Angeles, California; and
| | - Beatrice M Girard
- Department of Neurological Sciences, Robert Larner MD College of Medicine, University of Vermont, Burlington, Vermont
| | - Caitlin N Baran
- Department of Medicine, Robert Larner MD College of Medicine, University of Vermont, Burlington, Vermont
| | - Laura A Merriam
- Department of Neurological Sciences, Robert Larner MD College of Medicine, University of Vermont, Burlington, Vermont
| | - Victor May
- Department of Neurological Sciences, Robert Larner MD College of Medicine, University of Vermont, Burlington, Vermont
| | - Rodney L Parsons
- Department of Neurological Sciences, Robert Larner MD College of Medicine, University of Vermont, Burlington, Vermont;
| |
Collapse
|
19
|
Mikhailov N, V. Mamontov O, A. Kamshilin A, Giniatullin R. Parasympathetic Cholinergic and Neuropeptide Mechanisms of Migraine. Anesth Pain Med 2017; 7:e42210. [PMID: 28920040 PMCID: PMC5554415 DOI: 10.5812/aapm.42210] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 10/19/2016] [Accepted: 11/21/2016] [Indexed: 12/24/2022] Open
Abstract
CONTEXT Migraine mechanisms remain largely uncovered for various reasons including a very high complexity of the neurophysiological mechanisms implicated in this disorder and a plethora of endogenous biologically active compounds involved in the pathological process. The functional role of parasympathetic innervation of meninges and cholinergic mechanisms of migraine are among little explored issues despite multiple evidence indirectly indicating the role of acetylcholine (ACh) and its analogues in migraine and other types of headache. In the current short review, we discuss morphological, functional, and clinical issues related to the role of ACh and its analogues such as carbachol and nicotine in this most common neurological disorder. EVIDENCE ACQUISITION In the present work, studies published from 1953 to 2016 were investigated. Literature was searched with following keywords: acetylcholine (ACh), carbachol, nicotine, parasympathetic, mast cells, vasoactive intestinal polypeptide (VIP), and pituitary adenylate cyclase-activating polypeptide (PACAP). RESULTS Parasympathetic fibers originated from SPG and trigeminal nerves can interact at the level of meninges which is considered to be the origin site of migraine pain. Here, in dura mater, ACh, VIP, and PACAP released by parasympathetic afferents can both affect mast cells provoking its degranulation and additional release of neurotransmitters, or they can directly affect trigeminal nerves inducing nociception. CONCLUSIONS In summary, cholinergic mechanisms in migraine and other types of headache remain little elucidated and future studies should clarify the role of parasympathetic nerves and molecular mechanisms of cholinergic modulation within the nociceptive system.
Collapse
Affiliation(s)
- Nikita Mikhailov
- Department of Neurobiology, University of Eastern Finland, 70210 Kuopio, Finland
| | - Oleg V. Mamontov
- Department of Circulation Physiology, Federal Almazov North-West Medical Research Centre, St. Petersburg, 197341, Russia
- Department of Computer Photonics and Videomatics, ITMO University, St. Petersburg, 197101, Russia
| | - Alexei A. Kamshilin
- Department of Computer Photonics and Videomatics, ITMO University, St. Petersburg, 197101, Russia
| | - Rashid Giniatullin
- Department of Neurobiology, University of Eastern Finland, 70210 Kuopio, Finland
- Department of Computer Photonics and Videomatics, ITMO University, St. Petersburg, 197101, Russia
- Laboratory of Neurobiology, Kazan Federal University, Kazan, Russia
| |
Collapse
|
20
|
May V, Parsons RL. G Protein-Coupled Receptor Endosomal Signaling and Regulation of Neuronal Excitability and Stress Responses: Signaling Options and Lessons From the PAC1 Receptor. J Cell Physiol 2016; 232:698-706. [DOI: 10.1002/jcp.25615] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 09/22/2016] [Indexed: 02/03/2023]
Affiliation(s)
- Victor May
- Department of Neurological Sciences; University of Vermont College of Medicine; Burlington Vermont
| | - Rodney L. Parsons
- Department of Neurological Sciences; University of Vermont College of Medicine; Burlington Vermont
| |
Collapse
|
21
|
Tompkins JD, Clason TA, Hardwick JC, Girard BM, Merriam LA, May V, Parsons RL. Activation of MEK/ERK signaling contributes to the PACAP-induced increase in guinea pig cardiac neuron excitability. Am J Physiol Cell Physiol 2016; 311:C643-C651. [PMID: 27488668 DOI: 10.1152/ajpcell.00164.2016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 08/01/2016] [Indexed: 01/12/2023]
Abstract
Pituitary adenylate cyclase (PAC)-activating polypeptide (PACAP) peptides (Adcyap1) signaling at the selective PAC1 receptor (Adcyap1r1) participate in multiple homeostatic and stress-related responses, yet the cellular mechanisms underlying PACAP actions remain to be completely elucidated. PACAP/PAC1 receptor signaling increases excitability of neurons within the guinea pig cardiac ganglia, and as these neurons are readily accessible, this neuronal system is particularly amenable to study of PACAP modulation of ionic conductances. The present study investigated how PACAP activation of MEK/ERK signaling contributed to the peptide-induced increase in cardiac neuron excitability. Treatment with the MEK inhibitor PD 98059 blocked PACAP-stimulated phosphorylated ERK and, in parallel, suppressed the increase in cardiac neuron excitability. However, PD 98059 did not blunt the ability of PACAP to enhance two inward ionic currents, one flowing through hyperpolarization-activated nonselective cationic channels (Ih) and another flowing through low-voltage-activated calcium channels (IT), which support the peptide-induced increase in excitability. Thus a PACAP- and MEK/ERK-sensitive, voltage-dependent conductance(s), in addition to Ih and IT, modulates neuronal excitability. Despite prior work implicating PACAP downregulation of the KV4.2 potassium channel in modulation of excitability in other cells, treatment with the KV4.2 current blocker 4-aminopyridine did not replicate the PACAP-induced increase in excitability in cardiac neurons. However, cardiac neurons express the ERK target, the NaV1.7 sodium channel, and treatment with the selective NaV1.7 channel inhibitor PF-04856264 decreased the PACAP modulation of excitability. From these results, PACAP/PAC1 activation of MEK/ERK signaling may phosphorylate the NaV1.7 channel, enhancing sodium currents near the threshold, an action contributing to repetitive firing of the cardiac neurons exposed to PACAP.
Collapse
Affiliation(s)
- John D Tompkins
- Department of Medicine (Cardiology), David Geffen School of Medicine, University of California, Los Angeles, California
| | - Todd A Clason
- Department of Neurological Sciences, College of Medicine, University of Vermont, Burlington, Vermont; and
| | | | - Beatrice M Girard
- Department of Neurological Sciences, College of Medicine, University of Vermont, Burlington, Vermont; and
| | - Laura A Merriam
- Department of Neurological Sciences, College of Medicine, University of Vermont, Burlington, Vermont; and
| | - Victor May
- Department of Neurological Sciences, College of Medicine, University of Vermont, Burlington, Vermont; and
| | - Rodney L Parsons
- Department of Neurological Sciences, College of Medicine, University of Vermont, Burlington, Vermont; and
| |
Collapse
|
22
|
Activation of MEK/ERK Signaling by PACAP in Guinea Pig Cardiac Neurons. J Mol Neurosci 2016; 59:309-16. [PMID: 27194157 DOI: 10.1007/s12031-016-0766-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 04/29/2016] [Indexed: 10/21/2022]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) signaling can increase guinea pig cardiac neuron excitability in part through extracellular signal-regulated kinase (ERK) activation. The present study examined the PACAP receptors and signaling cascades that stimulate guinea pig cardiac neuron ERK signaling using confocal microscopy to quantify PACAP-induced neuronal phosphorylated ERK (pERK) immunoreactivity. PACAP and maxadilan, but not vasoactive intestinal polypeptide (VIP), increased cardiac neuron pERK, implicating primary roles for PACAP-selective PAC1 receptor (Adcyap1r1) signaling rather than VPAC receptors (Vipr1 and Vipr2) in the generation of cardiac neuron pERK. The adenylyl cyclase (AC) activator forskolin, but not the protein kinase C (PKC) activator phorbol myristate acetate (PMA), increased pERK. Also, Bim1 did not blunt PACAP activation of pERK. Together, the results suggest PAC1 receptor signal transduction via Gs/adenylyl cyclase (AC)/cAMP rather than Gq/phospholipase C (PLC) generated neuronal pERK. Activator and inhibitor studies suggested that the PACAP-mediated pERK activation was PKA-dependent rather than an exchange protein directly activated by a cAMP (EPAC), PKA-independent mechanism. The PACAP-induced pERK was inhibited by the clathrin inhibitor Pitstop2 to block receptor internalization and endosomal signaling. We propose that the PACAP-mediated MEK/ERK activation in cardiac neurons involves both AC/cAMP/PKA signaling and PAC1 receptor internalization/activation of signaling endosomes.
Collapse
|
23
|
Girard BM, Malley SE, Mathews MM, May V, Vizzard MA. Intravesical PAC1 Receptor Antagonist, PACAP(6-38), Reduces Urinary Bladder Frequency and Pelvic Sensitivity in NGF-OE Mice. J Mol Neurosci 2016; 59:290-9. [PMID: 27146136 DOI: 10.1007/s12031-016-0764-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 04/27/2016] [Indexed: 12/18/2022]
Abstract
Chronic NGF overexpression (OE) in the urothelium, achieved through the use of a highly urothelium-specific uroplakin II promoter, stimulates neuronal sprouting in the urinary bladder, produces increased voiding frequency and non-voiding contractions, and referred somatic sensitivity. Additional NGF-mediated pleiotropic changes might contribute to increased voiding frequency and pelvic hypersensitivity in NGF-OE mice such as neuropeptide/receptor systems including PACAP(Adcyap1) and PAC1 receptor (Adcyap1r1). Given the presence of PAC1-immunoreactive fibers and the expression of PAC1 receptor expression in bladder tissues, and PACAP-facilitated detrusor contraction, whether PACAP/receptor signaling contributes to increased voiding frequency and somatic sensitivity was evaluated in NGF-OE mice. Intravesical administration of the PAC1 receptor antagonist, PACAP(6-38) (300 nM), significantly (p ≤ 0.01) increased intercontraction interval (2.0-fold) and void volume (2.5-fold) in NGF-OE mice. Intravesical instillation of PACAP(6-38) also decreased baseline bladder pressure in NGF-OE mice. PACAP(6-38) had no effects on bladder function in WT mice. Intravesical administration of PACAP(6-38) (300 nM) significantly (p ≤ 0.01) reduced pelvic sensitivity in NGF-OE mice but was without effect in WT mice. PACAP/receptor signaling contributes to the increased voiding frequency and pelvic sensitivity observed in NGF-OE mice.
Collapse
Affiliation(s)
- Beatrice M Girard
- Department of Neurological Sciences, College of Medicine, University of Vermont, D405A Given Research Building, Burlington, VT, 05405, USA
| | - Susan E Malley
- Department of Neurological Sciences, College of Medicine, University of Vermont, D405A Given Research Building, Burlington, VT, 05405, USA
| | - Morgan M Mathews
- Department of Neurological Sciences, College of Medicine, University of Vermont, D405A Given Research Building, Burlington, VT, 05405, USA
| | - Victor May
- Department of Neurological Sciences, College of Medicine, University of Vermont, D405A Given Research Building, Burlington, VT, 05405, USA
| | - Margaret A Vizzard
- Department of Neurological Sciences, College of Medicine, University of Vermont, D405A Given Research Building, Burlington, VT, 05405, USA.
| |
Collapse
|
24
|
PACAP Modulates Distinct Neuronal Components to Induce Cell-Specific Plasticity at Central and Autonomic Synapses. CURRENT TOPICS IN NEUROTOXICITY 2016. [DOI: 10.1007/978-3-319-35135-3_7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
25
|
|
26
|
Tompkins JD, Merriam LA, Girard BM, May V, Parsons RL. Nickel suppresses the PACAP-induced increase in guinea pig cardiac neuron excitability. Am J Physiol Cell Physiol 2015; 308:C857-66. [PMID: 25810261 DOI: 10.1152/ajpcell.00403.2014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 03/18/2015] [Indexed: 10/23/2022]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is a potent intercellular signaling molecule involved in multiple homeostatic functions. PACAP/PAC1 receptor signaling increases excitability of neurons within the guinea pig cardiac ganglia, making them a unique system to establish mechanisms underlying PACAP modulation of neuronal function. Calcium influx is required for the PACAP-increased cardiac neuron excitability, although the pathway is unknown. This study tested whether PACAP enhancement of calcium influx through either T-type or R-type channels contributed to the modulation of excitability. Real-time quantitative polymerase chain reaction analyses indicated transcripts for Cav3.1, Cav3.2, and Cav3.3 T-type isoforms and R-type Cav2.3 in cardiac neurons. These neurons often exhibit a hyperpolarization-induced rebound depolarization that remains when cesium is present to block hyperpolarization-activated nonselective cationic currents (Ih). The T-type calcium channel inhibitors, nickel (Ni(2+)) or mibefradil, suppressed the rebound depolarization, and treatment with both drugs hyperpolarized cardiac neurons by 2-4 mV. Together, these results are consistent with the presence of functional T-type channels, potentially along with R-type channels, in these cardiac neurons. Fifty micromolar Ni(2+), a concentration that suppresses currents in both T-type and R-type channels, blunted the PACAP-initiated increase in excitability. Ni(2+) also blunted PACAP enhancement of the hyperpolarization-induced rebound depolarization and reversed the PACAP-mediated increase in excitability, after being initiated, in a subset of cells. Lastly, low voltage-activated currents, measured under perforated patch whole cell recording conditions and potentially flowing through T-type or R-type channels, were enhanced by PACAP. Together, our results suggest that a PACAP-enhanced, Ni(2+)-sensitive current contributes to PACAP-induced modulation of neuronal excitability.
Collapse
Affiliation(s)
- John D Tompkins
- Department of Neurological Sciences, College of Medicine, University of Vermont, Burlington, Vermont
| | - Laura A Merriam
- Department of Neurological Sciences, College of Medicine, University of Vermont, Burlington, Vermont
| | - Beatrice M Girard
- Department of Neurological Sciences, College of Medicine, University of Vermont, Burlington, Vermont
| | - Victor May
- Department of Neurological Sciences, College of Medicine, University of Vermont, Burlington, Vermont
| | - Rodney L Parsons
- Department of Neurological Sciences, College of Medicine, University of Vermont, Burlington, Vermont
| |
Collapse
|
27
|
Pituitary Adenylate Cyclase-Activating Polypeptide (PACAP) Signalling Enhances Osteogenesis in UMR-106 Cell Line. J Mol Neurosci 2014; 54:555-73. [DOI: 10.1007/s12031-014-0389-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 07/22/2014] [Indexed: 01/14/2023]
|
28
|
Hoover DB, Girard BM, Hoover JL, Parsons RL. PAC₁ receptors mediate positive chronotropic responses to PACAP-27 and VIP in isolated mouse atria. Eur J Pharmacol 2013; 713:25-30. [PMID: 23665113 DOI: 10.1016/j.ejphar.2013.04.037] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Revised: 04/02/2013] [Accepted: 04/26/2013] [Indexed: 11/26/2022]
Abstract
PACAP and VIP have prominent effects on cardiac function in several species, but little is known about their influence on the murine heart. Accordingly, we evaluated the expression of PACAP/VIP receptors in mouse heart and the response of isolated atria to peptide agonists. Quantitative PCR demonstrated that PAC₁, VPAC₁, and VPAC₂ receptor mRNAs are present throughout the mouse heart. Expression of all three receptor transcripts was low, PAC₁ being the lowest. No regional differences in expression were detected for individual receptor mRNAs after normalization to L32. Pharmacological effects of PACAP-27, VIP, and the selective PAC₁ agonist maxadilan were evaluated in isolated, spontaneously beating atria from C57BL/6 mice of either sex. Incremental additions of PACAP-27 at 1 min intervals caused a concentration-dependent tachycardia with a logEC₅₀=-9.08 ± 0.15 M (n=7) and a maximum of 96.3 ± 5.9% above baseline heart rate. VIP and maxadilan also caused tachycardia but their potencies were about two orders of magnitude less. Increasing the dosing interval to 5 min caused a leftward shift of the concentration-response curve to maxadilan but no changes in the curves for PACAP-27 or VIP. Under this condition, neither the potency nor the efficacy of maxadilan differed from those of PACAP-27. Neither PACAP-27 nor maxadilan caused tachyphylaxis, and maximal responses to maxadilan were maintained for at least 2 h. We conclude that all three VIP/PACAP family receptors are expressed by mouse cardiac tissue, but only PAC₁ receptors mediate positive chronotropic responses to PACAP-27 and VIP.
Collapse
Affiliation(s)
- Donald B Hoover
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, PO Box 70577, Johnson City, TN 37614, USA.
| | | | | | | |
Collapse
|
29
|
Effects of CYP-Induced Cystitis on PACAP/VIP and Receptor Expression in Micturition Pathways and Bladder Function in Mice with Overexpression of NGF in Urothelium. J Mol Neurosci 2012; 48:730-43. [DOI: 10.1007/s12031-012-9834-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Accepted: 06/05/2012] [Indexed: 12/22/2022]
|
30
|
Merriam LA, Roman CW, Baran CN, Girard BM, May V, Parsons RL. Pretreatment with nonselective cationic channel inhibitors blunts the PACAP-induced increase in guinea pig cardiac neuron excitability. J Mol Neurosci 2012; 48:721-9. [PMID: 22528456 DOI: 10.1007/s12031-012-9763-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Accepted: 03/26/2012] [Indexed: 11/29/2022]
Abstract
Calcium influx is required for the pituitary adenylyl cyclase activating polypeptide (PACAP)-induced increase in guinea pig cardiac neuron excitability, noted as a change from a phasic to multiple action potential firing pattern. Intracellular recordings indicated that pretreatment with the nonselective cationic channel inhibitors, 2-aminoethoxydiphenylborate (2-APB), 1-[β-[3-(4-methoxyphenyl)propoxy]-4-methoxyphenethyl]-1H-imidazole HCl (SKF 96365), and flufenamic acid (FFA) reduced the 20-nM PACAP-induced excitability increase. Additional experiments tested whether 2-APB, FFA, and SKF 96365 could suppress the increase in excitability by PACAP once it had developed. The increased action potential firing remained following application of 2-APB but was diminished by FFA. SKF 96365 transiently depressed the PACAP-induced excitability increase. A decrease and recovery of action potential amplitude paralleled the excitability shift. Since semiquantitative PCR indicated that cardiac neurons express TRPC subunit transcripts, we hypothesize that PACAP activates calcium-permeable, nonselective cationic channels, which possibly are members of the TRPC family. Our results are consistent with calcium influx being required for the initiation of the PACAP-induced increase in excitability, but suggest that it may not be required to sustain the peptide effect. The present results also demonstrate that nonselective cationic channel inhibitors could have other actions, which might contribute to the inhibition of the PACAP-induced excitability increase.
Collapse
Affiliation(s)
- Laura A Merriam
- Department of Anatomy and Neurobiology, College of Medicine, University of Vermont, Burlington, VT 05405, USA
| | | | | | | | | | | |
Collapse
|
31
|
Girasole AE, Palmer CP, Corrado SL, Marie Southerland E, Ardell JL, Hardwick JC. Angiotensin II potentiates adrenergic and muscarinic modulation of guinea pig intracardiac neurons. Am J Physiol Regul Integr Comp Physiol 2011; 301:R1391-9. [PMID: 21865545 DOI: 10.1152/ajpregu.00145.2011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The intrinsic cardiac plexus represents a major peripheral integration site for neuronal, hormonal, and locally produced neuromodulators controlling efferent neuronal output to the heart. This study examined the interdependence of norepinephrine, muscarinic agonists, and ANG II, to modulate intrinsic cardiac neuronal activity. Intracellular voltage recordings from whole-mount preparations of the guinea pig cardiac plexus were used to determine changes in active and passive electrical properties of individual intrinsic cardiac neurons. Application of either adrenergic or muscarinic agonists induced changes in neuronal resting membrane potentials, decreased afterhyperpolarization duration of single action potentials, and increased neuronal excitability. Adrenergic responses were inhibited by removal of extracellular calcium ions, while muscarinic responses were inhibited by application of TEA. The adrenergic responses were heterogeneous, responding to a variety of receptor-specific agonists (phenylephrine, clonidine, dobutamine, and terbutaline), although α-receptor agonists produced the most frequent responses. Application of ANG II alone produced a significant increase in excitability, while application of ANG II in combination with either adrenergic or muscarinic agonists produced a much larger potentiation of excitability. The ANG II-induced modulation of firing was blocked by the angiotensin type 2 (AT(2)) receptor inhibitor PD 123319 and was mimicked by the AT(2) receptor agonist CGP-42112A. AT(1) receptor blockade with telmasartin did not alter neuronal responses to ANG II. These data demonstrate that ANG II potentiates both muscarinically and adrenergically mediated activation of intrinsic cardiac neurons, doing so primarily via AT(2) receptor-dependent mechanisms. These neurohumoral interactions may be fundamental to regulation of neuronal excitability within the intrinsic cardiac nervous system.
Collapse
|
32
|
Njaine B, Martins RAP, Santiago MF, Linden R, Silveira MS. Pituitary adenylyl cyclase-activating polypeptide controls the proliferation of retinal progenitor cells through downregulation of cyclin D1. Eur J Neurosci 2010; 32:311-21. [PMID: 20646049 DOI: 10.1111/j.1460-9568.2010.07286.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
During retinal development, cell proliferation and exit from the cell cycle must be precisely regulated to ensure the generation of the appropriate numbers and proportions of the various retinal cell types. Previously, we showed that pituitary adenylyl cyclase-activating polypeptide (PACAP) exerts a neuroprotective effect in the developing retina of rats, through the cAMP-cAMP-dependent protein kinase (protein kinase A) (PKA) pathway. Here, we show that PACAP also regulates the proliferation of retinal progenitor cells. PACAP, PACAP-specific receptor (PAC1), and the receptors activated by both PACAP and vasoactive intestinal peptide (VIP), VPAC1 and VPAC2, are expressed during embryonic and postnatal development of the rat retina. Treatment of retinal explants with PACAP38 reduced the incorporation of [(3)H]thymidine as well as the number of 5-bromo-2'-deoxyuridine-positive and cyclin D1-positive cells. Pharmacological experiments indicated that PACAP triggers this antiproliferative effect through the activation of both PAC1 and VPACs, and the cAMP-PKA pathway. In addition, PACAP receptor activation decreased both cyclin D1 mRNA and protein content. Altogether, the data support the hypothesis that PACAP is a cell-extrinsic regulator with multiple roles during retinal development, including the regulation of proliferation in a subpopulation of retinal progenitor cells.
Collapse
Affiliation(s)
- Brian Njaine
- Instituto de Biofisica Carlos Chagas Filho-UFRJ, Edifício do Centro de Ciencias da Saude, Ilha do Fundão, Rio de Janeiro, Brazil
| | | | | | | | | |
Collapse
|
33
|
The role of PACAP in central cardiorespiratory regulation. Respir Physiol Neurobiol 2010; 174:65-75. [PMID: 20470908 DOI: 10.1016/j.resp.2010.05.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2010] [Revised: 05/03/2010] [Accepted: 05/03/2010] [Indexed: 11/22/2022]
Abstract
Pituitary adenylate cyclase activating polypeptide (PACAP) plays a role in almost every biological process from reproduction to hippocampal function. One area where a role for PACAP is not clearly delineated is central cardiorespiratory regulation. PACAP and its receptors (PAC1, VPAC1 and VPAC2) are present in cardiovascular areas of the ventral medulla and spinal cord and in the periphery. Central administration of PACAP generally increases arterial pressure. Knowledge about the role of PACAP in central cardiovascular regulation is growing, but even less is known about PACAP in central respiratory regulation. No specific data is currently available regarding the presence of PACAP or receptors in key respiratory centers, although it is known that neonatal PACAP knock-out mice die suddenly in a manner similar to sudden infant death syndrome (SIDS). Future studies in mature preparations investigating the role of PACAP in the physiology and integration of central cardiorespiratory reflexes are clearly essential for a full understanding of this important neuropeptide in breathing.
Collapse
|
34
|
PACAP/VIP and receptor characterization in micturition pathways in mice with overexpression of NGF in urothelium. J Mol Neurosci 2010; 42:378-89. [PMID: 20449688 DOI: 10.1007/s12031-010-9384-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2010] [Accepted: 04/23/2010] [Indexed: 12/11/2022]
Abstract
Urothelium-specific overexpression of nerve growth factor (NGF) in the urinary bladder of transgenic mice stimulates neuronal sprouting or proliferation in the urinary bladder, produces urinary bladder hyperreflexia, and results in increased referred somatic hypersensitivity. Additional NGF-mediated changes might contribute to the urinary bladder hyperreflexia and pelvic hypersensitivity observed in these transgenic mice such as upregulation of neuropeptide/receptor systems. Chronic overexpression of NGF in the urothelium was achieved through the use of a highly urothelium-specific, uroplakin II promoter. In the present study, we examined pituitary adenylate cyclase activating polypeptide (PACAP), vasoactive intestinal polypeptide (VIP), and associated receptor (PAC1, VPAC1, VPAC2) transcripts or protein expression in urothelium and detrusor smooth muscle and lumbosacral dorsal root ganglia in NGF-overexpressing and littermate wildtype mice using real-time quantitative reverse transcription-polymerase chain reaction and immunohistochemical approaches. Results demonstrate upregulation of PAC1 receptor transcript and PAC1-immunoreactivity in urothelium of NGF-OE mice whereas PACAP transcript and PACAP-immunoreactivity were decreased in urothelium of NGF-OE mice. In contrast, VPAC1 receptor transcript was decreased in both urothelium and detrusor smooth muscle of NGF-OE mice. VIP transcript expression and immunostaining was not altered in urinary bladder of NGF-OE mice. Changes in PACAP, VIP, and associated receptor transcripts and protein expression in micturition pathways resemble some, but not all, changes observed after induction of urinary bladder inflammation known to involve NGF production.
Collapse
|
35
|
Mori H, Nakamachi T, Ohtaki H, Yofu S, Sato A, Endo K, Iso Y, Suzuki H, Takeyama Y, Shintani N, Hashimoto H, Baba A, Shioda S. Cardioprotective Effect of Endogenous Pituitary Adenylate Cyclase-Activating Polypeptide on Doxorubicin-Induced Cardiomyopathy in Mice. Circ J 2010; 74:1183-90. [DOI: 10.1253/circj.cj-09-1024] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Hiroyoshi Mori
- Division of Cardiology, Department of Internal Medicine, Showa University Fujigaoka Hospital
- Department of First Anatomy, Showa University School of Medicine
| | - Tomoya Nakamachi
- Department of First Anatomy, Showa University School of Medicine
- Center for Biotechnology, Showa University
| | - Hirokazu Ohtaki
- Department of First Anatomy, Showa University School of Medicine
| | - Sachiko Yofu
- Department of First Anatomy, Showa University School of Medicine
| | - Atsushi Sato
- Department of First Anatomy, Showa University School of Medicine
| | - Kimi Endo
- Department of First Anatomy, Showa University School of Medicine
| | - Yoshitaka Iso
- Division of Cardiology, Department of Internal Medicine, Showa University Fujigaoka Hospital
| | - Hiroshi Suzuki
- Division of Cardiology, Department of Internal Medicine, Showa University Fujigaoka Hospital
| | - Youichi Takeyama
- Division of Cardiology, Department of Internal Medicine, Showa University Fujigaoka Hospital
| | - Norihito Shintani
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University
| | - Hitoshi Hashimoto
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University
| | - Akemichi Baba
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University
| | - Seiji Shioda
- Department of First Anatomy, Showa University School of Medicine
| |
Collapse
|
36
|
Pugh PC, Jayakar SS, Margiotta JF. PACAP/PAC1R signaling modulates acetylcholine release at neuronal nicotinic synapses. Mol Cell Neurosci 2009; 43:244-57. [PMID: 19958833 DOI: 10.1016/j.mcn.2009.11.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2009] [Revised: 11/18/2009] [Accepted: 11/19/2009] [Indexed: 12/11/2022] Open
Abstract
Neuropeptides collaborate with conventional neurotransmitters to regulate synaptic output. Pituitary adenylate cyclase-activating polypeptide (PACAP) co-localizes with acetylcholine in presynaptic nerve terminals, is released by stimulation, and enhances nicotinic acetylcholine receptor- (nAChR-) mediated responses. Such findings implicate PACAP in modulating nicotinic neurotransmission, but relevant synaptic mechanisms have not been explored. We show here that PACAP acts via selective high-affinity G-protein coupled receptors (PAC(1)Rs) to enhance transmission at nicotinic synapses on parasympathetic ciliary ganglion (CG) neurons by rapidly and persistently increasing the frequency and amplitude of spontaneous, impulse-dependent nicotinic excitatory postsynaptic currents (sEPSCs). Of the canonical adenylate cyclase (AC) and phospholipase-C (PLC) transduction cascades stimulated by PACAP/PAC(1)R signaling, only AC-generated signals are critical for synaptic modulation since the increases in sEPSC frequency and amplitude were mimicked by 8-Bromo-cAMP, blocked by inhibiting AC or cAMP-dependent protein kinase (PKA), and unaffected by inhibiting PLC. Despite its ability to increase agonist-induced nAChR currents, PACAP failed to influence nAChR-mediated impulse-independent miniature EPSC amplitudes (quantal size). Instead, evoked transmission assays reveal that PACAP/PAC(1)R signaling increased quantal content, indicating that it modulates synaptic function by increasing vesicular ACh release from presynaptic terminals. Lastly, signals generated by the retrograde messenger, nitric oxide- (NO-) are critical for the synaptic modulation since the PACAP-induced increases in spontaneous EPSC frequency, amplitude and quantal content were mimicked by NO donor and absent after inhibiting NO synthase (NOS). These results indicate that PACAP/PAC(1)R activation recruits AC-dependent signaling that stimulates NOS to increase NO production and control presynaptic transmitter output at neuronal nicotinic synapses.
Collapse
Affiliation(s)
- Phyllis C Pugh
- University of Toledo College of Medicine, Department of Neurosciences, Toledo, OH 43614-5804, USA
| | | | | |
Collapse
|
37
|
Hoover DB, Tompkins JD, Parsons RL. Differential activation of guinea pig intrinsic cardiac neurons by the PAC1 agonists maxadilan and pituitary adenylate cyclase-activating polypeptide 27 (PACAP27). J Pharmacol Exp Ther 2009; 331:197-203. [PMID: 19602551 PMCID: PMC2766232 DOI: 10.1124/jpet.109.155747] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2009] [Accepted: 07/10/2009] [Indexed: 11/22/2022] Open
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) evokes tachycardia followed by a larger cholinergic bradycardia in isolated guinea pig hearts. We used the selective PAC1 receptor agonist maxadilan and vasoactive intestinal polypeptide (VIP) to test the hypothesis that PACAP27-evoked tachycardia and bradycardia are mediated by VPAC and PAC1 receptors, respectively. Chronotropic actions of these peptides were evaluated in isolated perfused hearts. Direct neuronal actions were determined by intracellular voltage recordings from cholinergic neurons in atrial ganglion whole mounts. Administration of 1 nmol of PACAP27 to isolated hearts evoked typical biphasic rate responses, whereas 1 nmol of maxadilan caused only a minor rate decrease. Desensitization with VIP eliminated the positive chronotropic effect of PACAP27 selectively. Local application of PACAP27 to cardiac neurons frequently evoked slow depolarization and caused prolonged increase of neuronal excitability. Maxadilan rarely affected membrane potential but consistently increased excitability. VIP had no effect on excitability and evoked depolarization in only a few neurons. Because maxadilan increased neuronal excitability but did not trigger action potentials as PACAP often does, we evaluated the interaction of maxadilan with substance P (SP) in isolated hearts. SP depolarizes cardiac neurons more consistently than PACAP, often triggers neuronal action potentials, and causes bradycardia but does not increase neuronal excitability. Maxadilan had a persistent effect to augment negative chronotropic responses to SP. These findings support our hypothesis that PACAP evokes tachycardia and bradycardia through VPAC and PAC1 receptors, respectively. They also suggest that maxadilan and PACAP27 differ in activating PAC1 receptors on cardiac neurons and/or stimulating downstream signaling mechanisms.
Collapse
Affiliation(s)
- Donald B Hoover
- Department of Pharmacology, East Tennessee State University, Johnson City, 37614, USA.
| | | | | |
Collapse
|
38
|
Vaudry D, Falluel-Morel A, Bourgault S, Basille M, Burel D, Wurtz O, Fournier A, Chow BKC, Hashimoto H, Galas L, Vaudry H. Pituitary adenylate cyclase-activating polypeptide and its receptors: 20 years after the discovery. Pharmacol Rev 2009; 61:283-357. [PMID: 19805477 DOI: 10.1124/pr.109.001370] [Citation(s) in RCA: 858] [Impact Index Per Article: 53.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2025] Open
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is a 38-amino acid C-terminally alpha-amidated peptide that was first isolated 20 years ago from an ovine hypothalamic extract on the basis of its ability to stimulate cAMP formation in anterior pituitary cells (Miyata et al., 1989. PACAP belongs to the vasoactive intestinal polypeptide (VIP)-secretin-growth hormone-releasing hormone-glucagon superfamily. The sequence of PACAP has been remarkably well conserved during evolution from protochordates to mammals, suggesting that PACAP is involved in the regulation of important biological functions. PACAP is widely distributed in the brain and peripheral organs, notably in the endocrine pancreas, gonads, respiratory and urogenital tracts. Characterization of the PACAP precursor has revealed the existence of a PACAP-related peptide, the activity of which remains unknown. Two types of PACAP binding sites have been characterized: type I binding sites exhibit a high affinity for PACAP and a much lower affinity for VIP, whereas type II binding sites have similar affinity for PACAP and VIP. Molecular cloning of PACAP receptors has shown the existence of three distinct receptor subtypes: the PACAP-specific PAC1-R, which is coupled to several transduction systems, and the PACAP/VIP-indifferent VPAC1-R and VPAC2-R, which are primarily coupled to adenylyl cyclase. PAC1-Rs are particularly abundant in the brain, the pituitary and the adrenal gland, whereas VPAC receptors are expressed mainly in lung, liver, and testis. The development of transgenic animal models and specific PACAP receptor ligands has strongly contributed to deciphering the various actions of PACAP. Consistent with the wide distribution of PACAP and its receptors, the peptide has now been shown to exert a large array of pharmacological effects and biological functions. The present report reviews the current knowledge concerning the pleiotropic actions of PACAP and discusses its possible use for future therapeutic applications.
Collapse
Affiliation(s)
- David Vaudry
- Institut National de la Santé et de la Recherche Médicale U413, European Institute for Peptide Research (Institut Fédératif de Recherches Multidisciplinaires sur les Peptides 23), Mont-Saint-Aignan, France.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Hardwick JC, Baran CN, Southerland EM, Ardell JL. Remodeling of the guinea pig intrinsic cardiac plexus with chronic pressure overload. Am J Physiol Regul Integr Comp Physiol 2009; 297:R859-66. [PMID: 19605763 DOI: 10.1152/ajpregu.00245.2009] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Chronic pressure overload (PO) is associated with cardiac hypertrophy and altered autonomic control of cardiac function, in which the latter may involve adaptations in central and/or peripheral cardiac neural control mechanisms. To evaluate the specific remodeling of the intrinsic cardiac nervous system following pressure overload, the descending thoracic aorta artery of the guinea pig was constricted approximately 20%, and the animals recovered for 9 wk. Thereafter, atrial neurons of the intrinsic cardiac plexus were isolated for electrophysiological and immunohistochemical analyses. Intracellular voltage recordings from intrinsic cardiac neurons demonstrated no significant changes in passive membrane properties or action potential depolarization compared with age-matched controls and sham-operated animals, but afterhyperpolarization duration was increased in PO animals. Neuronal excitability, as determined by the number of action potentials produced with depolarizing stimuli, was differentially increased in phasic neurons derived from PO animals in response to exogenously applied histamine compared with sham and age-matched controls. Conversely, pituitary adenylate cyclase-activating polypeptide-induced increases in intrinsic cardiac neuron evoked AP frequency were similar between control and PO animals. Immunohistochemical analysis demonstrated a twofold increase in the percentage of neurons immunoreactive for neuronal nitric oxide synthase in PO animals compared with control. The density of mast cells within the intrinsic cardiac plexus from PO animals was also increased twofold compared with preparations from control animals. These results indicate that congestive heart failure associated with chronic pressure overload induces a differential remodeling of intrinsic cardiac neurons and upregulation of neuronal responsiveness to specific neuromodulators.
Collapse
Affiliation(s)
- Jean C Hardwick
- Biology Dept., Ithaca College, 953 Danby Road, Ithaca, NY 14850, USA.
| | | | | | | |
Collapse
|
40
|
Tompkins JD, Lawrence YT, Parsons RL. Enhancement of Ih, but not inhibition of IM, is a key mechanism underlying the PACAP-induced increase in excitability of guinea pig intrinsic cardiac neurons. Am J Physiol Regul Integr Comp Physiol 2009; 297:R52-9. [PMID: 19403861 DOI: 10.1152/ajpregu.00039.2009] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) increases excitability of guinea pig cardiac neurons, an effect mediated by PACAP-selective PAC(1) receptors. In dissociated guinea pig cardiac neurons, PACAP causes a positive shift of the voltage dependence of activation of the hyperpolarization-activated nonselective cation current (I(h)). This observation suggested that an enhancement of I(h) contributed to the increase in excitability in neurons within whole-mount cardiac ganglia preparations. To evaluate the role of I(h) in the PACAP-induced increase in excitability, we compared the increase in action potentials generated by 10 nM PACAP in control neurons and in neurons treated with ZD7288 (10 or 100 muM) or CsCl (2 or 2.5 mM), drugs known to inhibit I(h). In control cells exposed to PACAP, 1-s depolarizing current pulses elicited multiple action potential firing in 79% of the neurons. In ZD7288- or CsCl-containing solutions, the 10 nM PACAP-induced increase in excitability was markedly suppressed, with 7% and 21% of the neurons generating multiple action potentials, respectively. Prior results indicated that PACAP initiates depolarization by activating an inward current, which is separate from its enhancement of I(h). Here, we show that a PACAP-induced depolarization was comparable in control neurons and neurons bathed in a CsCl-containing solution, an observation indicating that CsCl did not interfere with activation of the PAC(1) receptor by PACAP. Additional experiments indicated that pretreatment with the putative M current (I(M)) inhibitor 1 mM BaCl(2), but not 10 microM XE991, initiated multiple firing in a majority of neurons, with resting potentials maintained at approximately -60 mV. Furthermore, in Ba(2+)-treated cells, 10 nM PACAP increased the number of action potentials generated. Our results indicate that PACAP enhancement of I(h), rather than inhibition of I(M) and other 1 mM Ba(2+)-sensitive K(+) currents, is a key ionic mechanism contributing to the peptide-induced increase in excitability for neurons within whole-mount cardiac ganglia preparations.
Collapse
Affiliation(s)
- John D Tompkins
- Department of Anatomy and Neurobiology, College of Medicine, University of Vermont, Burlington, Vermont 05405, USA
| | | | | |
Collapse
|
41
|
Hardwick JC, Southerland EM, Ardell JL. Chronic myocardial infarction induces phenotypic and functional remodeling in the guinea pig cardiac plexus. Am J Physiol Regul Integr Comp Physiol 2008; 295:R1926-33. [PMID: 18832084 DOI: 10.1152/ajpregu.90306.2008] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Chronic myocardial infarction (CMI) is associated with remodeling of the ventricle and evokes adaption in the cardiac neurohumoral control systems. To evaluate the remodeling of the intrinsic cardiac nervous system following myocardial infarction, the dorsal descending coronary artery was ligated in the guinea pig heart and the animals were allowed to recover for 7-9 wk. Thereafter, atrial neurons of the intrinsic cardiac plexus were isolated for electrophysiological and immunohistochemical analyses. Intracellular voltage recordings from intrinsic cardiac neurons demonstrated no significant changes in passive membrane properties or action potential configuration compared with age-matched controls and sham-operated animals. The intrinsic cardiac neurons from chronic infarcted hearts did demonstrate an increase in evoked action potential (AP) frequency (as determined by the number of APs produced with depolarizing stimuli) and an increase in responses to exogenously applied histamine compared with sham and age-matched controls. Conversely, pituitary adenylate cyclase-activating polypeptide (PACAP)-induced increases in intrinsic cardiac neuron-evoked AP frequency were similar between control and CMI animals. Immunohistochemical analysis demonstrated a threefold increase in percentage of neurons immunoreactive for neuronal nitric oxide synthase (NOS) in CMI animals compared with control and the additional expression of inducible NOS by some neurons, which was not evident in control animals. Finally, the density of mast cells within the intrinsic cardiac plexus was increased threefold in preparations from CMI animals. These results indicate that CMI induces a differential remodeling of intrinsic cardiac neurons and functional upregulation of neuronal responsiveness to specific neuromodulators.
Collapse
Affiliation(s)
- Jean C Hardwick
- Department of Biology, Ithaca College, Ithaca, NY 14850, USA.
| | | | | |
Collapse
|
42
|
Yoshiyama M, de Groat WC. The role of vasoactive intestinal polypeptide and pituitary adenylate cyclase-activating polypeptide in the neural pathways controlling the lower urinary tract. J Mol Neurosci 2008; 36:227-40. [PMID: 18677446 DOI: 10.1007/s12031-008-9090-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2008] [Accepted: 04/21/2008] [Indexed: 12/18/2022]
Abstract
Vasoactive intestinal polypeptide (VIP) and pituitary adenylate cyclase-activating polypeptide (PACAP) are expressed in the neural pathways regulating the lower urinary tract. VIP-immunoreactivity (IR) is present in afferent and autonomic efferent neurons innervating the bladder and urethra, whereas PACAP-IR is present primarily in afferent neurons. Exogenously applied VIP relaxes bladder and urethral smooth muscle and excites parasympathetic neurons in bladder ganglia. PACAP relaxes bladder and urethral smooth muscle in some species (pig) but excites the smooth muscle in other species (mouse). Intrathecal administration of VIP in cats with an intact spinal cord suppresses reflex bladder activity, but intrathecal administration of VIP or PACAP in rats enhances bladder activity and suppresses urethral sphincter activity. PACAP has presynaptic facilitatory effects and direct excitatory effects on lumbosacral parasympathetic preganglionic neurons. Chronic spinal cord transection produces an expansion of VIP-IR (cats) and PACAP-IR (rats) in primary afferent axons in the lumbosacral spinal cord and unmasks spinal excitatory effects of VIP on bladder reflexes in cats. Intrathecal administration of PACAP6-38, a PAC1 receptor antagonist, reduces bladder hyperactivity in chronic spinal-cord-injured rats. These observations raise the possibility that VIP or PACAP have a role in the control of normal or abnormal voiding.
Collapse
|
43
|
Young BA, Girard BM, Parsons RL. Neurturin suppresses injury-induced neuronal activating transcription factor 3 expression in cultured guinea pig cardiac ganglia. J Comp Neurol 2008; 508:795-805. [PMID: 18393382 DOI: 10.1002/cne.21711] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cultured guinea pig atrial whole mounts containing the intrinsic cardiac ganglia were used as an in vitro model to investigate the induction of the stress/injury marker activating transcription factor 3 (ATF-3). ATF-3 expression was quantified by using immunocytochemical labeling and real-time PCR. In freshly isolated ganglia, no neuronal or Schwann cell nuclei exhibited ATF-3 immunoreactivity. In 2-hour cultures, the induction of ATF-3 expression was evident in many Schwann cell nuclei, whereas no neuronal nuclei were ATF-3 immunoreactive. Beginning at 4 hours, the percentage of neurons with ATF-3-immunoreactive nuclei increased progressively, and, by 48 hours in culture, approximately 95% of the cardiac neurons had ATF-3-immunoreactive nuclei. Neurturin significantly suppressed ATF-3 expression in 48-hour-cultured neurons without effect on ATF-3 expression in Schwann cell nuclei. Neuturin also could reverse neuronal ATF-3 expression after its induction. The suppression of ATF-3 induction by neurturin was mediated by activation of the phosphatidylinositol 3-kinase and mitogen-activated protein kinase pathways. Glial-derived neurotrophic factor (GDNF) also suppressed neuronal ATF-3 induction during culture. However, culture in serum-free media, presence of nerve growth factor, or addition of pituitary adenylate cyclase-activating polypeptide had no effect on ATF-3 induction in the 48-hour-cultured cardiac neurons. By 4 hours in culture, there was a significant increase in ATF-3 transcript levels, and neurturin partially suppressed ATF-3 transcript levels in 48-hour cultures. It is proposed that the loss of target-derived neurturin is a potential mechanism stimulating injury-induced expression of ATF-3 in cardiac neurons.
Collapse
Affiliation(s)
- Beth A Young
- Department of Anatomy and Neurobiology, University of Vermont College of Medicine, Burlington, Vermont 05405, USA
| | | | | |
Collapse
|
44
|
PACAP-mediated ATP release from rat urothelium and regulation of PACAP/VIP and receptor mRNA in micturition pathways after cyclophosphamide (CYP)-induced cystitis. J Mol Neurosci 2008; 36:310-20. [PMID: 18563302 DOI: 10.1007/s12031-008-9104-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2008] [Accepted: 05/09/2008] [Indexed: 12/29/2022]
Abstract
Pituitary adenylate cyclase-activating peptide (PACAP) peptides are expressed in micturition pathways, and PACAP expression is regulated by urinary bladder inflammation. Previous physiological studies have demonstrated roles for PACAP27 and PACAP38 in detrusor smooth muscle (DSM) contraction and a PAC1 receptor antagonist reduced cyclophosphamide (CYP)-induced bladder hyperreflexia. To gain insight into PACAP signaling in micturition and regulation with cystitis, receptor characterization by real-time quantitative polymerase chain reaction and physiological assays were performed. PACAP receptors were identified in tissues of rat micturition pathway, including DSM, urothelium (U), and dorsal root ganglia (DRG) after acute (4 h), intermediate (48 h) or chronic (8 days) CYP-induced cystitis. PAC1 messenger RNA expression significantly (p < or = 0.05) increased in U and DSM after 48 h and chronic CYP-induced cystitis after an initial decrease at 4 h. VPAC1 and VPAC2 transcripts increased in U and DSM after acute and intermediate CYP-induced cystitis followed by a decrease in VPAC2 expression with chronic cystitis. Application of PACAP27 (100 nM) to cultured urothelial cells evoked adenosine triphosphate (ATP) release that was blocked by the PAC1 specific antagonist, M65 (1 microM). PACAP38 (100 nM) also evoked ATP release from cultured urothelial cells, but ATP release was less than that observed with PACAP27. PACAP transcripts were increased in the U with intermediate and chronic cystitis, whereas vasoactive intestinal polypeptide (VIP) expression in both tissues was very low and showed no regulation with cystitis. Regulation of PACAP, galanin, and substance P transcripts expression was observed in lumbosacral DRG, but no regulation for VIP was observed. The current data demonstrate PACAP and PAC1 regulation in micturition pathways with inflammation and PACAP-mediated ATP release from urothelium.
Collapse
|
45
|
Tompkins JD, Parsons RL. Identification of intracellular signaling cascades mediating the PACAP-induced increase in guinea pig cardiac neuron excitability. J Mol Neurosci 2008; 36:292-8. [PMID: 18509761 DOI: 10.1007/s12031-008-9086-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2008] [Accepted: 04/14/2008] [Indexed: 11/25/2022]
Abstract
The pituitary adenylate cyclase-activating polypeptide (PACAP) increases excitability of guinea pig cardiac neurons, an effect mediated through activation of PAC1 receptors. The signaling cascades that couple activation of the PAC1 receptor to alterations in membrane ionic conductances responsible for the PACAP effect are unknown. Intracellular recordings were made from neurons in kinase inhibitor-treated cardiac ganglia preparations to determine which of the intracellular cascades activated by PAC1 receptor stimulation mediate the PACAP effect. In control cells, long depolarizing-current steps elicited one to three action potentials. In contrast, during the application of 10 nM PACAP, depolarizing-current pulses elicited multiple action potential firing (greater than or equal to five action potentials) in 79% of the neurons. Pretreatment with an adenylyl cyclase inhibitor, SQ 22536 (100 microM), suppressed the PACAP-induced increase in excitability, whereas the presence of U-73122 (10 microM), a potent phospholipase C (PLC) inhibitor, had no effect. Thus, the activation of adenylyl cyclase, but not PLC, was a critical step mediating the PACAP effect. Pretreatment with H-89 (1 microM), a protein kinase A inhibitor, and PD 98059 (50 microM), a MEK kinase inhibitor, also significantly blunted the PACAP-induced increase in excitability. Furthermore, treatment with forskolin (5 microM), an activator of adenylyl cyclase, or exposure to the cell-permeable cyclic adenosine monophosphate (cAMP) analogue, 8-bromo-cAMP (1 mM), partially recapitulated the effect of PACAP on excitability. We conclude that the activation of signaling cascades downstream of cAMP mediate the PACAP-induced increase in cardiac neuron excitability.
Collapse
Affiliation(s)
- John D Tompkins
- Department of Anatomy and Neurobiology, College of Medicine, University of Vermont, Burlington, VT 05405, USA.
| | | |
Collapse
|
46
|
Hernádi L, Pirger Z, Kiss T, Németh J, Mark L, Kiss P, Tamas A, Lubics A, Toth G, Shioda S, Reglodi D. The presence and distribution of pituitary adenylate cyclase activating polypeptide and its receptor in the snail Helix pomatia. Neuroscience 2008; 155:387-402. [PMID: 18590802 DOI: 10.1016/j.neuroscience.2008.05.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2008] [Revised: 05/06/2008] [Accepted: 05/06/2008] [Indexed: 12/17/2022]
Abstract
The aim of this study was to show the presence, distribution and function of the pituitary adenylate cyclase activating polypeptide (PACAP) and its receptors in the CNS and peripheral nervous system of the mollusk, Helix pomatia. PACAP-like and pituitary adenylate cyclase activating polypeptide receptor (PAC1-R)-like immunoreactivity was abundant both in the CNS and the peripheral nervous system of the snail. In addition several non-neuronal cells also revealed PACAP-like immunoreactivity. In inactive animals labeled cell bodies were mainly found and in the neuropile of active animals dense immunostained fiber system was additionally detected suggesting that expression of PACAP-like peptide was affected by the behavioral state of the animal. RIA measurements revealed the existence of both forms of PACAP in the CNS where the 27 amino acid form was found to be dominant. The concentration of PACAP27 was significantly higher in samples from active animals supporting the data obtained by immunohistochemistry. In Western blot experiments PACAP27 and PACAP38 antibodies specifically labeled protein band at 4.5 kDa both in rat and snail brain homogenates, and additionally an approximately 14 kDa band in snail. The 4.5 kDa protein corresponds to PACAP38 and the 14 kDa protein corresponds to the preproPACAP or to a PACAP-like peptide having larger molecular weight than mammalian PACAP38. In matrix-assisted laser desorption ionization time of flight (MALDI TOF) measurements fragments of PACAP38 were identified in brain samples suggesting the presence of a large molecular weight peptide in the snail. Applying antibodies developed against the PACAP receptor PAC1-R, immunopositive stained neurons and a dense network of fibers were identified in each of the ganglia. In electrophysiological experiments, extracellular application of PACAP27 and PACAP38 transiently depolarized or increased postsynaptic activity of neurons expressing PAC1-R. In several neurons PACAP elicited a long lasting hyperpolarization which was eliminated after 1.5 h continuous washing. Taken together, these results indicate that PACAP may have significant role in a wide range of basic physiological functions in snail.
Collapse
Affiliation(s)
- L Hernádi
- Department of Experimental Zoology, Balaton Limnological Research Institute, Hungarian Academy of Sciences, P.O. Box 35, H-8237 Tihany, Hungary
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Parsons RL, Tompkins JD, Merriam LA. Source and Action of Pituitary Adenylate Cyclase-Activating Polypeptide in Guinea Pig Intrinsic Cardiac Ganglia. Tzu Chi Med J 2008. [DOI: 10.1016/s1016-3190(08)60002-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
48
|
Tompkins JD, Ardell JL, Hoover DB, Parsons RL. Neurally released pituitary adenylate cyclase-activating polypeptide enhances guinea pig intrinsic cardiac neurone excitability. J Physiol 2007; 582:87-93. [PMID: 17495034 PMCID: PMC2075297 DOI: 10.1113/jphysiol.2007.134965] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Intracellular recordings were made in vitro from guinea-pig cardiac ganglia to determine whether endogenous neuropeptides such as pituitary adenylate cyclase-activating polypeptide (PACAP) or substance P released during tetanic neural stimulation modulate cardiac neurone excitability and/or contribute to slow excitatory postsynaptic potentials (sEPSPs). When nicotinic and muscarinic receptors were blocked by hexamethonium and atropine, 20 Hz stimulation for 10 s initiated a sEPSP in all innervated neurones. In 40% of the cells, excitability was enhanced after termination of the sEPSP. This suggested that non-cholinergic receptor-mediated mechanisms contributed to the sEPSP and modulated neuronal excitability. Exogenous PACAP and substance P initiated a slow depolarization in the neurones whereas neuronal excitability was only increased by PACAP. When ganglia were treated with the PAC1 antagonist PACAP6-38 (500 nM), the sEPSP evoked by 20 Hz stimulation was reduced by approximately 50% and an enhanced excitability occurred in only 10% of the cells. These observations suggested that PACAP released from preganglionic nerve terminals during tetanic stimulation enhanced neuronal excitability and evoked sEPSPs. After addition of 1 nM PACAP to the bath, 7 of 9 neurones exhibited a tonic firing pattern whereas in untreated preparations, the neurons had a phasic firing pattern. PACAP6-38 (500 nM) diminished the increase in excitability caused by 1 nM PACAP so that only 4 of 13 neurones exhibited a tonic firing pattern and the other 9 cells retained a phasic firing pattern. These findings indicate that PACAP can be released by tetanic neural stimulation in vitro and increase the excitability of intrinsic cardiac neurones. We hypothesize that in vivo PACAP released during preganglionic firing may modulate neurotransmission within the intrinsic cardiac ganglia.
Collapse
MESH Headings
- Animals
- Atropine/pharmacology
- Autonomic Fibers, Preganglionic/metabolism
- Electric Stimulation/methods
- Excitatory Postsynaptic Potentials
- Ganglia, Parasympathetic/cytology
- Ganglia, Parasympathetic/drug effects
- Ganglia, Parasympathetic/metabolism
- Guinea Pigs
- Heart/innervation
- Heart Atria/innervation
- Hexamethonium/pharmacology
- In Vitro Techniques
- Kinetics
- Muscarinic Antagonists/pharmacology
- Neurons/drug effects
- Neurons/metabolism
- Nicotinic Antagonists/pharmacology
- Peptide Fragments/pharmacology
- Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism
- Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology
- Receptors, Muscarinic/drug effects
- Receptors, Nicotinic/drug effects
- Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide, Type I/antagonists & inhibitors
- Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide, Type I/metabolism
- Substance P/pharmacology
- Synaptic Transmission/drug effects
Collapse
Affiliation(s)
- John D Tompkins
- Department of Anatomy and Neurobiology, College of Medicine, University of Vermont, Burlington, VT 05405, USA
| | | | | | | |
Collapse
|
49
|
Girard BM, Young BA, Buttolph TR, White SL, Parsons RL. Regulation of neuronal pituitary adenylate cyclase-activating polypeptide expression during culture of guinea-pig cardiac ganglia. Neuroscience 2007; 146:584-93. [PMID: 17367946 PMCID: PMC2048657 DOI: 10.1016/j.neuroscience.2007.02.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2006] [Revised: 01/17/2007] [Accepted: 02/01/2007] [Indexed: 10/23/2022]
Abstract
The trophic neuropeptide pituitary adenylate cyclase-activating polypeptide (PACAP) increases in many different neuron types following injury; a response postulated to support cell survival and regeneration. In acutely isolated cardiac ganglia, approximately 1% of the cardiac neurons exhibited PACAP immunoreactivity whereas after 72 h in culture, approximately 25% of the neurons were PACAP immunoreactive. In contrast, there was no increase in vasoactive intestinal polypeptide (VIP)-immunoreactive (IR) cells. Using a combination of immunocytochemical and molecular techniques, we have quantified PACAP expression, during explant culture of guinea-pig cardiac ganglia. Using real time polymerase chain reaction, PACAP transcript levels increased progressively up to 48 h in culture with no further increase after 72 h. PACAP transcript levels were reduced by neurturin at 48 h in culture but not after 24 or 72 h in culture. In addition, neurturin partially suppressed the percentage of PACAP-IR neurons after 72 h in culture, an effect mediated by activation of the phosphatidylinositol 3-kinase and mitogen-activated protein kinase signaling pathways. The addition of different known regulatory molecules, including ciliary neurotrophic factor (CNTF), interleukin-1 beta (Il-1beta), tumor necrosis factor-alpha (TNFalpha), fibroblast growth factor basic (bFGF), transforming growth factor-beta (TGF-beta) and nerve growth factor (NGF) did not increase the percentage of PACAP-IR neurons after 24 h in culture; a result indicating that the generation and secretion of these factors did not stimulate PACAP expression. The presence of 20 nM PACAP or 10 muM forskolin increased the percentage of PACAP-IR cardiac neurons in 24 h cultures, but not in 72 h cultures. Neither treatment enhanced the number of VIP-IR neurons. The addition of the PACAP selective receptor (PAC(1)) receptor antagonist, M65 (100 nM) suppressed the 20 nM PACAP-induced increase in percentage of PACAP-IR cells in 24 h cultures indicating the effect of PACAP was mediated through the PAC(1) receptor. However, 100 nM M65 had no effect on the percentage of PACAP-IR cells in either 24 or 48 h cultures not treated with exogenous PACAP, suggesting that endogenous release of PACAP likely did not contribute to the enhanced peptide expression. We postulate that the enhanced PACAP expression, which occurs in response to injury is facilitated in the explant cultured cardiac ganglia by the loss of a target-derived inhibitory factor, very likely neurturin. In intact tissues the presence of neurturin would normally suppress PACAP expression. Lastly, our results indicate that many common trophic factors do not enhance PACAP expression in the cultured cardiac neurons. However, the stimulatory role of an, as yet, unidentified factor cannot be excluded.
Collapse
Affiliation(s)
- B M Girard
- Department of Anatomy and Neurobiology, University of Vermont College of Medicine, Burlington, VT 05405, USA
| | | | | | | | | |
Collapse
|
50
|
Braas KM, May V, Zvara P, Nausch B, Kliment J, Dunleavy JD, Nelson MT, Vizzard MA. Role for pituitary adenylate cyclase activating polypeptide in cystitis-induced plasticity of micturition reflexes. Am J Physiol Regul Integr Comp Physiol 2006; 290:R951-62. [PMID: 16322346 PMCID: PMC1402357 DOI: 10.1152/ajpregu.00734.2005] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Pituitary adenylate cyclase activating polypeptide (PACAP) peptides are expressed and regulated in sensory afferents of the micturition pathway. Although these studies have implicated PACAP in bladder control, the physiological significance of these observations has not been firmly established. To clarify these issues, the roles of PACAP and PACAP signaling in micturition and cystitis were examined in receptor characterization and physiological assays. PACAP receptors were identified in various tissues of the micturition pathway, including bladder detrusor smooth muscle and urothelium. Bladder smooth muscle expressed heterogeneously PAC(1)null, PAC(1)HOP1, and VPAC(2) receptors; the urothelium was more restricted in expressing preferentially the PAC(1) receptor subtype only. Immunocytochemical studies for PAC(1) receptors were consistent with these tissue distributions. Furthermore, the addition of 50-100 nM PACAP27 or PACAP38 to isolated bladder strips elicited transient contractions and sustained increases in the amplitude of spontaneous phasic contractions. Treatment of the bladder strips with tetrodotoxin (1 muM) did not alter the spontaneous phasic contractions suggesting direct PACAP effects on bladder smooth muscle. PACAP also increased the amplitude of nerve-evoked contractions. By contrast, vasoactive intestinal polypeptide had no direct effects on bladder smooth muscle. In a rat cyclophosphamide (CYP)-induced cystitis paradigm, intrathecal or intravesical administration of PAC(1) receptor antagonist, PACAP6-38, reduced cystitis-induced bladder overactivity. In summary, these studies support roles for PACAP in micturition and suggest that inflammation-induced plasticity in PACAP expression in peripheral and central micturition pathways contribute to bladder dysfunction with cystitis.
Collapse
Affiliation(s)
- Karen M. Braas
- University of Vermont College of Medicine, Departments of Anatomy and Neurobiology
| | - Victor May
- University of Vermont College of Medicine, Departments of Anatomy and Neurobiology
- Pharmacology Burlington, VT 05405 USA
| | | | | | | | | | | | - Margaret A. Vizzard
- University of Vermont College of Medicine, Departments of Anatomy and Neurobiology
- Neurology and
| |
Collapse
|