1
|
Tanaka M, Szabó Á, Körtési T, Szok D, Tajti J, Vécsei L. From CGRP to PACAP, VIP, and Beyond: Unraveling the Next Chapters in Migraine Treatment. Cells 2023; 12:2649. [PMID: 37998384 PMCID: PMC10670698 DOI: 10.3390/cells12222649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 11/25/2023] Open
Abstract
Migraine is a neurovascular disorder that can be debilitating for individuals and society. Current research focuses on finding effective analgesics and management strategies for migraines by targeting specific receptors and neuropeptides. Nonetheless, newly approved calcitonin gene-related peptide (CGRP) monoclonal antibodies (mAbs) have a 50% responder rate ranging from 27 to 71.0%, whereas CGRP receptor inhibitors have a 50% responder rate ranging from 56 to 71%. To address the need for novel therapeutic targets, researchers are exploring the potential of another secretin family peptide, pituitary adenylate cyclase-activating polypeptide (PACAP), as a ground-breaking treatment avenue for migraine. Preclinical models have revealed how PACAP affects the trigeminal system, which is implicated in headache disorders. Clinical studies have demonstrated the significance of PACAP in migraine pathophysiology; however, a few clinical trials remain inconclusive: the pituitary adenylate cyclase-activating peptide 1 receptor mAb, AMG 301 showed no benefit for migraine prevention, while the PACAP ligand mAb, Lu AG09222 significantly reduced the number of monthly migraine days over placebo in a phase 2 clinical trial. Meanwhile, another secretin family peptide vasoactive intestinal peptide (VIP) is gaining interest as a potential new target. In light of recent advances in PACAP research, we emphasize the potential of PACAP as a promising target for migraine treatment, highlighting the significance of exploring PACAP as a member of the antimigraine armamentarium, especially for patients who do not respond to or contraindicated to anti-CGRP therapies. By updating our knowledge of PACAP and its unique contribution to migraine pathophysiology, we can pave the way for reinforcing PACAP and other secretin peptides, including VIP, as a novel treatment option for migraines.
Collapse
Affiliation(s)
- Masaru Tanaka
- HUN-REN-SZTE Neuroscience Research Group, Hungarian Research Network, University of Szeged (HUN-REN-SZTE), Danube Neuroscience Research Laboratory, Tisza Lajos krt. 113, H-6725 Szeged, Hungary;
| | - Ágnes Szabó
- Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary; (Á.S.); (D.S.); (J.T.)
- Doctoral School of Clinical Medicine, University of Szeged, Korányi fasor 6, H-6720 Szeged, Hungary
| | - Tamás Körtési
- HUN-REN-SZTE Neuroscience Research Group, Hungarian Research Network, University of Szeged (HUN-REN-SZTE), Danube Neuroscience Research Laboratory, Tisza Lajos krt. 113, H-6725 Szeged, Hungary;
- Faculty of Health Sciences and Social Studies, University of Szeged, Temesvári krt. 31, H-6726 Szeged, Hungary;
- Preventive Health Sciences Research Group, Incubation Competence Centre of the Centre of Excellence for Interdisciplinary Research, Development and Innovation of the University of Szeged, H-6720 Szeged, Hungary
| | - Délia Szok
- Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary; (Á.S.); (D.S.); (J.T.)
| | - János Tajti
- Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary; (Á.S.); (D.S.); (J.T.)
| | - László Vécsei
- HUN-REN-SZTE Neuroscience Research Group, Hungarian Research Network, University of Szeged (HUN-REN-SZTE), Danube Neuroscience Research Laboratory, Tisza Lajos krt. 113, H-6725 Szeged, Hungary;
- Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary; (Á.S.); (D.S.); (J.T.)
| |
Collapse
|
2
|
Li C, Shen W, Xu Z, Li C, Liu Q, Pang Y, Li J, Wang X, Wang Z, Feng S. The discovery of the new mechanism: Celastrol improves spinal cord injury by increasing cAMP through VIP-ADCYAP1R1-GNAS pathway. Biomed Pharmacother 2023; 165:115250. [PMID: 37531781 DOI: 10.1016/j.biopha.2023.115250] [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: 05/12/2023] [Revised: 07/25/2023] [Accepted: 07/27/2023] [Indexed: 08/04/2023] Open
Abstract
Spinal cord injury (SCI) is a debilitating condition that results in significant impairment of motor function and sensation. Despite the ongoing efforts to develop effective treatments, there are currently very limited options available for patients with SCI. Celastrol, a natural anti-inflammatory compound extracted from Tripterygium wilfordii, has been shown to exhibit anti-inflammatory and anti-apoptotic properties. In this study, we aimed to explore the therapeutic potential of celastrol for SCI and elucidate the underlying molecular mechanisms involved. We found that local tissue often experiences a significant decrease in cAMP content and occurrs apoptosis after SCI. However, the treatment of celastrol could promote the production of cAMP by up-regulating the VIP-ADCYAP1R1-GNAS pathway. This could effectively inhibit the phosphorylation of JNK and prevent apoptosis, ultimately improving the exercise ability after SCI. Together, our results reveal celastrol may be a promising therapeutic agent for the treatment of SCI.
Collapse
Affiliation(s)
- Chuanhao Li
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin 300052, China
| | - Wenyuan Shen
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin 300052, China; Spine Surgery Department of the Second Hospital, Cheeloo College of Medicine, Shandong University, 247 Beiyuan Street, Jinan, Shandong 250033, China
| | - Zhengyu Xu
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin 300052, China
| | - Chao Li
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin 300052, China
| | - Quan Liu
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin 300052, China
| | - Yilin Pang
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin 300052, China
| | - Junjin Li
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin 300052, China
| | - Xiaoyu Wang
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin 300052, China
| | - Zhishuo Wang
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin 300052, China
| | - Shiqing Feng
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin 300052, China; Spine Surgery Department of the Second Hospital, Cheeloo College of Medicine, Shandong University, 247 Beiyuan Street, Jinan, Shandong 250033, China; Orthopedic Research Center of Shandong University &Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250063, China.
| |
Collapse
|
3
|
Zhang Z, Li Q, Huang Y, Xu Z, Chen X, Jiang B, Huang Y, Jian J. Vasoactive Intestinal Peptide (VIP) Protects Nile Tilapia ( Oreochromis niloticus) against Streptococcus agalatiae Infection. Int J Mol Sci 2022; 23:ijms232314895. [PMID: 36499231 PMCID: PMC9738603 DOI: 10.3390/ijms232314895] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/24/2022] [Accepted: 11/25/2022] [Indexed: 11/29/2022] Open
Abstract
Vasoactive intestinal peptide (VIP), a member of secretin/glucagon family, is involved in a variety of biological activities such as gut motility, immune responses, and carcinogenesis. In this study, the VIP precursor gene (On-VIP) and its receptor gene VIPR1 (On-VIPR1) were identified from Nile tilapia (Oreochromis niloticus), and the functions of On-VIP in the immunomodulation of Nile tilapia against bacterial infection were investigated and characterized. On-VIP and On-VIPR1 contain a 450 bp and a 1326 bp open reading frame encoding deduced protein of 149 and 441 amino acids, respectively. Simultaneously, the transcript of both On-VIP and On-VIPR1 were highly expressed in the intestine and sharply induced by Streptococcus agalatiae. Moreover, the positive signals of On-VIP and On-VIPR1 were detected in the longitudinal muscle layer and mucosal epithelium of intestine, respectively. Furthermore, both in vitro and in vivo experiments indicated several immune functions of On-VIP, including reduction of P65, P38, MyD88, STAT3, and AP1, upregulation of CREB and CBP, and suppression of inflammation. Additionally, in vivo experiments proved that On-VIP could protect Nile tilapia from bacterial infection and promote apoptosis and pyroptosis. These data lay a theoretical basis for further understanding of the mechanism of VIP guarding bony fish against bacterial infection.
Collapse
Affiliation(s)
- Zhiqiang Zhang
- Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, College of Fishery, Guangdong Ocean University, Zhanjiang 524088, China
| | - Qi Li
- Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, College of Fishery, Guangdong Ocean University, Zhanjiang 524088, China
| | - Yongxiong Huang
- Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, College of Fishery, Guangdong Ocean University, Zhanjiang 524088, China
| | - Zhou Xu
- Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, College of Fishery, Guangdong Ocean University, Zhanjiang 524088, China
| | - Xinjin Chen
- Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, College of Fishery, Guangdong Ocean University, Zhanjiang 524088, China
| | - Baijian Jiang
- Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, College of Fishery, Guangdong Ocean University, Zhanjiang 524088, China
| | - Yu Huang
- Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, College of Fishery, Guangdong Ocean University, Zhanjiang 524088, China
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, China
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen 327005, China
| | - Jichang Jian
- Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, College of Fishery, Guangdong Ocean University, Zhanjiang 524088, China
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, China
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen 327005, China
- Correspondence:
| |
Collapse
|
4
|
Lu J, Piper SJ, Zhao P, Miller LJ, Wootten D, Sexton PM. Targeting VIP and PACAP Receptor Signaling: New Insights into Designing Drugs for the PACAP Subfamily of Receptors. Int J Mol Sci 2022; 23:8069. [PMID: 35897648 PMCID: PMC9331257 DOI: 10.3390/ijms23158069] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/18/2022] [Accepted: 07/20/2022] [Indexed: 12/16/2022] Open
Abstract
Pituitary Adenylate Cyclase-Activating Peptide (PACAP) and Vasoactive Intestinal Peptide (VIP) are neuropeptides involved in a diverse array of physiological and pathological processes through activating the PACAP subfamily of class B1 G protein-coupled receptors (GPCRs): VIP receptor 1 (VPAC1R), VIP receptor 2 (VPAC2R), and PACAP type I receptor (PAC1R). VIP and PACAP share nearly 70% amino acid sequence identity, while their receptors PAC1R, VPAC1R, and VPAC2R share 60% homology in the transmembrane regions of the receptor. PACAP binds with high affinity to all three receptors, while VIP binds with high affinity to VPAC1R and VPAC2R, and has a thousand-fold lower affinity for PAC1R compared to PACAP. Due to the wide distribution of VIP and PACAP receptors in the body, potential therapeutic applications of drugs targeting these receptors, as well as expected undesired side effects, are numerous. Designing selective therapeutics targeting these receptors remains challenging due to their structural similarities. This review discusses recent discoveries on the molecular mechanisms involved in the selectivity and signaling of the PACAP subfamily of receptors, and future considerations for therapeutic targeting.
Collapse
Affiliation(s)
- Jessica Lu
- Drug Discovery Biology, Australian Research Council Centre for Cryo-Electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia; (J.L.); (S.J.P.); (P.Z.)
| | - Sarah J. Piper
- Drug Discovery Biology, Australian Research Council Centre for Cryo-Electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia; (J.L.); (S.J.P.); (P.Z.)
| | - Peishen Zhao
- Drug Discovery Biology, Australian Research Council Centre for Cryo-Electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia; (J.L.); (S.J.P.); (P.Z.)
| | - Laurence J. Miller
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, AZ 85259, USA;
| | - Denise Wootten
- Drug Discovery Biology, Australian Research Council Centre for Cryo-Electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia; (J.L.); (S.J.P.); (P.Z.)
| | - Patrick M. Sexton
- Drug Discovery Biology, Australian Research Council Centre for Cryo-Electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia; (J.L.); (S.J.P.); (P.Z.)
| |
Collapse
|
5
|
Pellesi L, Al-Karagholi MAM, De Icco R, Chaudhry BA, Lopez CL, Snellman J, Hannibal J, Amin FM, Ashina M. Plasma Levels of CGRP During a 2-h Infusion of VIP in Healthy Volunteers and Patients With Migraine: An Exploratory Study. Front Neurol 2022; 13:871176. [PMID: 35432170 PMCID: PMC9011105 DOI: 10.3389/fneur.2022.871176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 02/24/2022] [Indexed: 11/16/2022] Open
Abstract
Introduction The activation of perivascular fibers and the consequent release of vasoactive peptides, including the vasoactive intestinal polypeptide (VIP), play a role in migraine pathogenesis. A 2-h infusion of VIP provoked migraine, but the mechanisms remain unknown. We investigated whether 2-h infusion of VIP caused alterations in plasma levels of the calcitonin gene-related peptide (CGRP) and whether any changes might be related to the induced migraine attacks. Materials and Methods We enrolled individuals with episodic migraine without aura and healthy participants to randomly receive a 2-h infusion of either VIP (8 pmol/kg/min) or placebo (sterile saline) in two randomized, placebo-controlled crossover trials. We collected clinical data and measured plasma levels of VIP and CGRP at fixed time points: at baseline (T0) and every 30 min until 180 min (T180) after the start of the infusion. Results Blood samples were collected from patients with migraine (n = 19) and healthy individuals (n = 12). During VIP infusion, mixed effects analysis revealed a significant increase in plasma CGRP (p = 0.027) at T30 (vs. T180, adjusted p-value = 0.039) and T60 (vs. T180, adjusted p-value = 0.027) in patients with migraine. We found no increase in plasma CGRP during VIP-induced migraine attacks (p = 0.219). In healthy individuals, there was no increase in plasma CGRP during VIP (p = 0.205) or placebo (p = 0.428) days. Discussion Plasma CGRP was elevated in patients with migraine during a prolonged infusion of VIP, but these alterations were not associated with VIP-induced migraine attacks. Given the exploratory design of our study, further investigations are needed to clarify the role of CGRP in VIP-induced migraine. Clinical Trial Registration ClinicalTrials.gov, identifier: NCT03989817 and NCT04260035.
Collapse
Affiliation(s)
- Lanfranco Pellesi
- Danish Headache Center, Department of Neurology, Rigshospitalet Glostrup, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mohammad Al-Mahdi Al-Karagholi
- Danish Headache Center, Department of Neurology, Rigshospitalet Glostrup, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Roberto De Icco
- Headache Science & Neurorehabilitation Center, Istituto Neurologico Nazionale a Carattere Scientifico (IRCCS) Mondino Foundation, Pavia, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Basit Ali Chaudhry
- Danish Headache Center, Department of Neurology, Rigshospitalet Glostrup, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | - Jens Hannibal
- Department of Clinical Biochemistry, Bispebjerg Frederiksberg Hospital, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Faisal Mohammad Amin
- Danish Headache Center, Department of Neurology, Rigshospitalet Glostrup, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Neurorehabilitation and Traumatic Brain Injury, Rigshospitalet, Copenhagen, Denmark
| | - Messoud Ashina
- Danish Headache Center, Department of Neurology, Rigshospitalet Glostrup, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- *Correspondence: Messoud Ashina
| |
Collapse
|
6
|
Solés-Tarrés I, Cabezas-Llobet N, Lefranc B, Leprince J, Alberch J, Vaudry D, Xifró X. Pituitary Adenylate Cyclase-Activating Polypeptide (PACAP) Protects Striatal Cells and Improves Motor Function in Huntington’s Disease Models: Role of PAC1 Receptor. Front Pharmacol 2022; 12:797541. [PMID: 35153755 PMCID: PMC8832515 DOI: 10.3389/fphar.2021.797541] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 12/31/2021] [Indexed: 12/21/2022] Open
Abstract
Huntington’s disease (HD) is a hereditary neurodegenerative disorder caused by the expression of mutant huntingtin (mHtt). One of the main features of HD is the degeneration of the striatum that leads to motor discoordination. Pituitary adenylate cyclase-activating polypeptide (PACAP) is a neuropeptide that acts through three receptors named PAC1R, VPAC1R, and VPAC2R. In the present study, we first investigated the effect of PACAP on STHdhQ7/Q7 and STHdhQ111/Q111 cells that express wild-type Htt with 7 and mHtt with 111 glutamines, respectively. Then we explored the capacity of PACAP to rescue motor symptoms in the R6/1, a murine model of HD. We found that PACAP treatment (10–7 M) for 24 h protects STHdhQ111/Q111 cells from mHtt-induced apoptosis. This effect is associated with an increase in PAC1R transcription, phosphorylation of ERK and Akt, and an increase of intracellular c-fos, egr1, CBP, and BDNF protein content. Moreover, the use of pharmacological inhibitors revealed that activation of ERK and Akt mediates these antiapoptotic and neurotrophic effects of PACAP. To find out PAC1R implication, we treated STHdh cells with vasoactive intestinal peptide (VIP), which exhibits equal affinity for VPAC1R and VPAC2R, but lower affinity for PAC1R, in contrast to PACAP which has same affinity for the three receptors. VIP reduced cleaved caspase-3 protein level, without promoting the expression of c-fos, egr1, CBP, and the neurotrophin BDNF. We next measured the protein level of PACAP receptors in the striatum and cortex of R6/1 mice. We observed a specific reduction of PAC1R at the onset of motor symptoms. Importantly, the intranasal administration of PACAP to R6/1 animals restored the motor function and increased the striatal levels of PAC1R, CBP, and BDNF. In conclusion, PACAP exerts antiapoptotic and neurotrophic effects in striatal neurons mainly through PAC1R. This effect in HD striatum allows the recovery of motor function and point out PAC1R as a therapeutic target for treatment of HD.
Collapse
Affiliation(s)
- Irene Solés-Tarrés
- New Therapeutic Targets Group, Department of Medical Science, Faculty of Medicine, University of Girona, Girona, Spain
| | - Núria Cabezas-Llobet
- New Therapeutic Targets Group, Department of Medical Science, Faculty of Medicine, University of Girona, Girona, Spain
| | - Benjamin Lefranc
- Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Neuropeptides, Neuronal Death and Cell Plasticity Team, UNIROUEN, Inserm, Normandie University, Rouen, France
- Regional Cell Imaging Platform of Normandy (PRIMACEN), UNIROUEN, Normandie University, Rouen, France
| | - Jérôme Leprince
- Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Neuropeptides, Neuronal Death and Cell Plasticity Team, UNIROUEN, Inserm, Normandie University, Rouen, France
- Regional Cell Imaging Platform of Normandy (PRIMACEN), UNIROUEN, Normandie University, Rouen, France
| | - Jordi Alberch
- Departament de Biomedicina, Institut de Neurociències, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain
- Institut D’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - David Vaudry
- Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Neuropeptides, Neuronal Death and Cell Plasticity Team, UNIROUEN, Inserm, Normandie University, Rouen, France
- Regional Cell Imaging Platform of Normandy (PRIMACEN), UNIROUEN, Normandie University, Rouen, France
| | - Xavier Xifró
- New Therapeutic Targets Group, Department of Medical Science, Faculty of Medicine, University of Girona, Girona, Spain
- *Correspondence: Xavier Xifró,
| |
Collapse
|
7
|
Apostol CR, Bernard K, Tanguturi P, Molnar G, Bartlett MJ, Szabò L, Liu C, Ortiz JB, Saber M, Giordano KR, Green TRF, Melvin J, Morrison HW, Madhavan L, Rowe RK, Streicher JM, Heien ML, Falk T, Polt R. Design and Synthesis of Brain Penetrant Glycopeptide Analogues of PACAP With Neuroprotective Potential for Traumatic Brain Injury and Parkinsonism. FRONTIERS IN DRUG DISCOVERY 2022; 1. [PMID: 35237767 PMCID: PMC8887546 DOI: 10.3389/fddsv.2021.818003] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
There is an unmet clinical need for curative therapies to treat neurodegenerative disorders. Most mainstay treatments currently on the market only alleviate specific symptoms and do not reverse disease progression. The Pituitary adenylate cyclase-activating polypeptide (PACAP), an endogenous neuropeptide hormone, has been extensively studied as a potential regenerative therapeutic. PACAP is widely distributed in the central nervous system (CNS) and exerts its neuroprotective and neurotrophic effects via the related Class B GPCRs PAC1, VPAC1, and VPAC2, at which the hormone shows roughly equal activity. Vasoactive intestinal peptide (VIP) also activates these receptors, and this close analogue of PACAP has also shown to promote neuronal survival in various animal models of acute and progressive neurodegenerative diseases. However, PACAP's poor pharmacokinetic profile (non-linear PK/PD), and more importantly its limited blood-brain barrier (BBB) permeability has hampered development of this peptide as a therapeutic. We have demonstrated that glycosylation of PACAP and related peptides promotes penetration of the BBB and improves PK properties while retaining efficacy and potency in the low nanomolar range at its target receptors. Furthermore, judicious structure-activity relationship (SAR) studies revealed key motifs that can be modulated to afford compounds with diverse selectivity profiles. Most importantly, we have demonstrated that select PACAP glycopeptide analogues (2LS80Mel and 2LS98Lac) exert potent neuroprotective effects and anti-inflammatory activity in animal models of traumatic brain injury and in a mild-toxin lesion model of Parkinson's disease, highlighting glycosylation as a viable strategy for converting endogenous peptides into robust and efficacious drug candidates.
Collapse
Affiliation(s)
- Christopher R Apostol
- Department of Chemistry and Biochemistry, BIO5, The University of Arizona, Tucson, AZ, United States
| | - Kelsey Bernard
- Graduate Interdisciplinary Program in Physiological Sciences, The University of Arizona, Tucson, AZ, United States
| | | | - Gabriella Molnar
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ, United States
| | - Mitchell J Bartlett
- Department of Neurology, College of Medicine, The University of Arizona, Tucson, AZ, United States
| | - Lajos Szabò
- Department of Chemistry and Biochemistry, BIO5, The University of Arizona, Tucson, AZ, United States
| | - Chenxi Liu
- Department of Chemistry and Biochemistry, BIO5, The University of Arizona, Tucson, AZ, United States
| | - J Bryce Ortiz
- Barrow Neurological Institute at Phoenix Children's Hospital, The University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States.,Department of Child Health, The University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States.,Phoenix Veteran Affairs Health Care System, Phoenix, AZ, United States
| | - Maha Saber
- Barrow Neurological Institute at Phoenix Children's Hospital, The University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States.,Department of Child Health, The University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States
| | - Katherine R Giordano
- Barrow Neurological Institute at Phoenix Children's Hospital, The University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States.,Department of Child Health, The University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States.,Phoenix Veteran Affairs Health Care System, Phoenix, AZ, United States
| | - Tabitha R F Green
- Department of Child Health, The University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States
| | - James Melvin
- Department of Child Health, The University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States.,Department of Biological Sciences, University of Bath, Bath, United Kingdom
| | - Helena W Morrison
- College of Nursing, University of Arizona, Tucson, AZ, United States
| | - Lalitha Madhavan
- Graduate Interdisciplinary Program in Physiological Sciences, The University of Arizona, Tucson, AZ, United States.,Department of Neurology, College of Medicine, The University of Arizona, Tucson, AZ, United States
| | - Rachel K Rowe
- Barrow Neurological Institute at Phoenix Children's Hospital, The University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States.,Department of Child Health, The University of Arizona College of Medicine-Phoenix, Phoenix, AZ, United States.,Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States
| | - John M Streicher
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ, United States
| | - Michael L Heien
- Department of Chemistry and Biochemistry, BIO5, The University of Arizona, Tucson, AZ, United States
| | - Torsten Falk
- Graduate Interdisciplinary Program in Physiological Sciences, The University of Arizona, Tucson, AZ, United States.,Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ, United States.,Department of Neurology, College of Medicine, The University of Arizona, Tucson, AZ, United States
| | - Robin Polt
- Department of Chemistry and Biochemistry, BIO5, The University of Arizona, Tucson, AZ, United States
| |
Collapse
|
8
|
Apostol CR, Tanguturi P, Szabò LZ, Varela D, Gilmartin T, Streicher JM, Polt R. Synthesis and In Vitro Characterization of Glycopeptide Drug Candidates Related to PACAP 1-23. Molecules 2021; 26:4932. [PMID: 34443519 PMCID: PMC8401035 DOI: 10.3390/molecules26164932] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/10/2021] [Accepted: 08/11/2021] [Indexed: 01/05/2023] Open
Abstract
The search for efficacious treatment of neurodegenerative and progressive neuroinflammatory diseases continues, as current therapies are unable to halt or reverse disease progression. PACAP represents one potential therapeutic that provides neuroprotection effects on neurons, and also modulates inflammatory responses and circulation within the brain. However, PACAP is a relatively long peptide hormone that is not trivial to synthesize. Based on previous observations that the shortened isoform PACAP1-23 is capable of inducing neuroprotection in vitro, we were inspired to synthesize shortened glycopeptide analogues of PACAP1-23. Herein, we report the synthesis and in vitro characterization of glycosylated PACAP1-23 analogues that interact strongly with the PAC1 and VPAC1 receptors, while showing reduced activity at the VPAC2 receptor.
Collapse
Affiliation(s)
- Christopher R. Apostol
- Department of Chemistry and Biochemistry, BIO5, The University of Arizona, 1306 E. University Blvd, Tucson, AZ 85721, USA; (C.R.A.); (L.Z.S.)
| | - Parthasaradhireddy Tanguturi
- Department of Pharmacology, College of Medicine, The University of Arizona, 1501 N. Campbell Ave, Tucson, AZ 85724, USA; (P.T.); (J.M.S.)
| | - Lajos Z. Szabò
- Department of Chemistry and Biochemistry, BIO5, The University of Arizona, 1306 E. University Blvd, Tucson, AZ 85721, USA; (C.R.A.); (L.Z.S.)
| | - Daniel Varela
- Facultat de Quìmica Tarragona, Universitat Rovera I Virgili, 43007 Barcelona, Spain; (D.V.); (T.G.)
| | - Thiago Gilmartin
- Facultat de Quìmica Tarragona, Universitat Rovera I Virgili, 43007 Barcelona, Spain; (D.V.); (T.G.)
| | - John M. Streicher
- Department of Pharmacology, College of Medicine, The University of Arizona, 1501 N. Campbell Ave, Tucson, AZ 85724, USA; (P.T.); (J.M.S.)
| | - Robin Polt
- Department of Chemistry and Biochemistry, BIO5, The University of Arizona, 1306 E. University Blvd, Tucson, AZ 85721, USA; (C.R.A.); (L.Z.S.)
| |
Collapse
|
9
|
Zhu X, Zhang K, He L, Liao F, Ren Y, Jan YK. Spectral analysis of blood flow oscillations to assess the plantar skin blood flow regulation in response to preconditioning local vibrations. Biorheology 2021; 58:39-49. [PMID: 33896803 DOI: 10.3233/bir-201011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Local vibration has shown promise in improving skin blood flow and wound healing. However, the underlying mechanism of local vibration as a preconditioning intervention to alter plantar skin blood flow after walking is unclear. OBJECTIVE The objective was to use wavelet analysis of skin blood flow oscillations to investigate the effect of preconditioning local vibration on plantar tissues after walking. METHODS A double-blind, repeated measures design was tested in 10 healthy participants. The protocol included 10-min baseline, 10-min local vibrations (100 Hz or sham), 10-min walking, and 10-min recovery periods. Skin blood flow was measured over the first metatarsal head of the right foot during the baseline and recovery periods. Wavelet amplitudes after walking were expressed as the ratio of the wavelet amplitude before walking. RESULTS The results showed the significant difference in the metabolic (vibration 10.06 ± 1.97, sham 5.78 ± 1.53, p < 0.01) and neurogenic (vibration 7.45 ± 1.54, sham 4.78 ± 1.22, p < 0.01) controls. There were no significant differences in the myogenic, respiratory and cardiac controls between the preconditioning local vibration and sham conditions. CONCLUSIONS Our results showed that preconditioning local vibration altered the normalization rates of plantar skin blood flow after walking by stimulating the metabolic and neurogenic controls.
Collapse
Affiliation(s)
- Xiaotong Zhu
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | - Keying Zhang
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | - Li He
- College of Physical Education and Sports, Beijing Normal University, Beijing, China
| | - Fuyuan Liao
- Department of Biomedical Engineering, Xi'an Technological University, Xi'an, China
| | - Yuanchun Ren
- College of Physical Education and Sports, Beijing Normal University, Beijing, China
| | - Yih-Kuen Jan
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Champaign, IL, USA.,Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, China
| |
Collapse
|
10
|
Zwanenburg PR, Backer SFM, Obdeijn MC, Lapid O, Gans SL, Boermeester MA. A Systematic Review and Meta-Analysis of the Pressure-Induced Vasodilation Phenomenon and Its Role in the Pathophysiology of Ulcers. Plast Reconstr Surg 2019; 144:669e-681e. [PMID: 31568315 DOI: 10.1097/prs.0000000000006090] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Physiologic studies show that tissue perfusion increases during moderate amounts of tissue compression. This is attributed to sensory nerves initiating a vasodilatory cascade referred to as pressure-induced vasodilation. METHODS PubMed, Embase, and the Cochrane Central Register of Controlled Trials were searched for studies investigating perfusion during pressure exposure longer than 10 minutes. Retrieved studies were assessed using the Office of Health Assessment and Translation Risk of Bias Rating Tool for Human and Animal Studies. Results were pooled with random effects models. The body of evidence was rated using the Office of Health Assessment and Translation approach. RESULTS Twenty-nine articles were included, of which 19 articles were included in meta-analyses. The evidence indicates that moderate amounts of tissue compression have the capacity to increase perfusion in healthy humans by 46 percent (95 percent CI, 30 to 62 percent). Using the Office of Health Assessment and Translation approach, the authors found a high level of confidence in the body of evidence. Pressure-induced vasodilation blockade was associated with increased pressure ulcer formation. Pressure-induced vasodilation was impaired by neuropathy and by the drugs diclofenac and amiloride. CONCLUSIONS This systematic review and meta-analysis indicates that healthy humans have the capacity to increase local perfusion in response to mechanical stress resulting from tissue compression. Because pressure-induced vasodilation is mediated by sensory nerves, pressure-induced vasodilation emphasizes the importance of sensory innervation for durable tissue integrity. Pressure-induced vasodilation impairment seems to provide a complementary explanation for the susceptibility of neuropathic tissues to pressure-induced lesions.
Collapse
Affiliation(s)
- Pieter R Zwanenburg
- From the Department of Surgery, Amsterdam Gastroenterology and Metabolism, Amsterdam Infection and Immunity, and the Department of Plastic, Reconstructive and Hand Surgery, Amsterdam University Medical Center, University of Amsterdam
| | - Sophia F M Backer
- From the Department of Surgery, Amsterdam Gastroenterology and Metabolism, Amsterdam Infection and Immunity, and the Department of Plastic, Reconstructive and Hand Surgery, Amsterdam University Medical Center, University of Amsterdam
| | - Miryam C Obdeijn
- From the Department of Surgery, Amsterdam Gastroenterology and Metabolism, Amsterdam Infection and Immunity, and the Department of Plastic, Reconstructive and Hand Surgery, Amsterdam University Medical Center, University of Amsterdam
| | - Oren Lapid
- From the Department of Surgery, Amsterdam Gastroenterology and Metabolism, Amsterdam Infection and Immunity, and the Department of Plastic, Reconstructive and Hand Surgery, Amsterdam University Medical Center, University of Amsterdam
| | - Sarah L Gans
- From the Department of Surgery, Amsterdam Gastroenterology and Metabolism, Amsterdam Infection and Immunity, and the Department of Plastic, Reconstructive and Hand Surgery, Amsterdam University Medical Center, University of Amsterdam
| | - Marja A Boermeester
- From the Department of Surgery, Amsterdam Gastroenterology and Metabolism, Amsterdam Infection and Immunity, and the Department of Plastic, Reconstructive and Hand Surgery, Amsterdam University Medical Center, University of Amsterdam
| |
Collapse
|
11
|
Fouchard M, Misery L, Le Garrec R, Sigaudo-Roussel D, Fromy B. Alteration of Pressure-Induced Vasodilation in Aging and Diabetes, a Neuro-Vascular Damage. Front Physiol 2019; 10:862. [PMID: 31333501 PMCID: PMC6616153 DOI: 10.3389/fphys.2019.00862] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 06/20/2019] [Indexed: 12/11/2022] Open
Abstract
Skin is constantly subjected to pressure at different levels. Pressure-induced vasodilation (PIV) is one of the response mechanisms to low pressure that maintains the homeostasis of the skin. PIV results from the interaction of primary afferent nerves and vascular endothelium of skin vessels. Thanks to this cutaneous neuro-vascular interaction, the cutaneous blood flow increase allows the maintenance of an optimal level of oxygenation and minimizes the lack of vascularization of the skin tissue under low pressure. It seems to be associated with the cutaneous protection mechanisms to prevent pressure ulcers. In some contexts, where microangiopathy and neuropathy can occur, such as aging and diabetes, PIV is impaired, leading to a dramatic early decrease in local skin blood flow when low pressure is applied. In aging, PIV alteration is due to endothelial dysfunction, essentially from an alteration of the nitric oxide pathway. In the inflamm-aging context, oxidative stress increases leading to endothelial cell and nerve damages. An age-related sensory neuropathy will exacerbate the alteration of PIV during the aging process. In diabetes, non-controlled hyperglycaemia leads to an increase in several pathological biochemical pathways that involve oxidative stress and can affect PIV. Sorbinil, alagebrium and alpha-lipoic acid are able individually to restore PIV through a possible oxidative stress reduction. Candesartan, an angiotensin II type 1 receptor blocker, is also able to restore PIV and prevent pressure ulcer formation. The possibility of preventing pressure ulcer associated to diabetes and/or aging with the restoration of PIV seems to be a promising research path.
Collapse
Affiliation(s)
- Maxime Fouchard
- LIEN, F-29200, University of Brest, Brest, France.,Department of Dermatology, University Hospital of Brest, Brest, France
| | - Laurent Misery
- LIEN, F-29200, University of Brest, Brest, France.,Department of Dermatology, University Hospital of Brest, Brest, France
| | | | | | | |
Collapse
|
12
|
Multiple nickel-sensitive targets elicit cardiac arrhythmia in isolated mouse hearts after pituitary adenylate cyclase-activating polypeptide-mediated chronotropy. Pharmacol Res 2016; 117:140-147. [PMID: 28007571 DOI: 10.1016/j.phrs.2016.12.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 10/24/2016] [Accepted: 12/16/2016] [Indexed: 12/11/2022]
Abstract
The pituitary adenylate cyclase-activating polypeptide (PACAP)-27 modulates various biological processes, from the cellular level to function specification. However, the cardiac actions of this neuropeptide are still under intense studies. Using control (+|+) and mice lacking (-|-) either R-type (Cav2.3) or T-type (Cav3.2) Ca2+ channels, we investigated the effects of PACAP-27 on cardiac activity of spontaneously beating isolated perfused hearts. Superfusion of PACAP-27 (20nM) caused a significant increase of baseline heart frequency in Cav2.3(+|+) (156.9±10.8 to 239.4±23.4 bpm; p<0.01) and Cav2.3(-|-) (190.3±26.4 to 270.5±25.8 bpm; p<0.05) hearts. For Cav3.2, the heart rate was significantly increased in Cav3.2(-|-) (133.1±8.5 bpm to 204.6±27.9 bpm; p<0.05) compared to Cav3.2(+|+) hearts (185.7±11.2 bpm to 209.3±22.7 bpm). While the P wave duration and QTc interval were significantly increased in Cav2.3(+|+) and Cav2.3(-|-) hearts following PACAP-27 superfusion, there was no effect in Cav3.2(+|+) and Cav3.2(-|-) hearts. The positive chronotropic effects observed in the four study groups, as well as the effect on P wave duration and QTc interval were abolished in the presence of Ni2+ (50μM) and PACAP-27 (20nM) in hearts from Cav2.3(+|+) and Cav2.3(-|-) mice. In addition to suppressing PACAP's response, Ni2+ also induced conduction disturbances in investigated hearts. In conclusion, the most Ni2+-sensitive Ca2+ channels (R- and T-type) may modulate the PACAP signaling cascade during cardiac excitation in isolated mouse hearts, albeit to a lesser extent than other Ni2+-sensitive targets.
Collapse
|
13
|
Erdener SE, Dalkara T. Modelling headache and migraine and its pharmacological manipulation. Br J Pharmacol 2014; 171:4575-94. [PMID: 24611635 DOI: 10.1111/bph.12651] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 01/13/2014] [Accepted: 02/14/2014] [Indexed: 12/22/2022] Open
Abstract
Similarities between laboratory animals and humans in anatomy and physiology of the cephalic nociceptive pathways have allowed scientists to create successful models that have significantly contributed to our understanding of headache. They have also been instrumental in the development of novel anti-migraine drugs different from classical pain killers. Nevertheless, modelling the mechanisms underlying primary headache disorders like migraine has been challenging due to limitations in testing the postulated hypotheses in humans. Recent developments in imaging techniques have begun to fill this translational gap. The unambiguous demonstration of cortical spreading depolarization (CSD) during migraine aura in patients has reawakened interest in studying CSD in animals as a noxious brain event that can activate the trigeminovascular system. CSD-based models, including transgenics and optogenetics, may more realistically simulate pain generation in migraine, which is thought to originate within the brain. The realization that behavioural correlates of headache and migrainous symptoms like photophobia can be assessed quantitatively in laboratory animals, has created an opportunity to directly study the headache in intact animals without the confounding effects of anaesthetics. Headache and migraine-like episodes induced by administration of glyceryltrinitrate and CGRP to humans and parallel behavioural and biological changes observed in rodents create interesting possibilities for translational research. Not unexpectedly, species differences and model-specific observations have also led to controversies as well as disappointments in clinical trials, which, in return, has helped us improve the models and advance our understanding of headache. Here, we review commonly used headache and migraine models with an emphasis on recent developments.
Collapse
Affiliation(s)
- S E Erdener
- Department of Neurology, Faculty of Medicine, Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey
| | | |
Collapse
|
14
|
The selective PAC1 receptor agonist maxadilan inhibits neurogenic vasodilation and edema formation in the mouse skin. Neuropharmacology 2014; 85:538-47. [PMID: 24973707 DOI: 10.1016/j.neuropharm.2014.06.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 06/14/2014] [Accepted: 06/16/2014] [Indexed: 01/30/2023]
Abstract
We have earlier shown that PACAP-38 decreases neurogenic inflammation. However, there were no data on its receptorial mechanism and the involvement of its PAC1 and VPAC1/2 receptors (PAC1R, VPAC1/2R) in this inhibitory effect. Neurogenic inflammation in the mouse ear was induced by topical application of the Transient Receptor Potential Ankyrin 1 (TRPA1) receptor activator mustard oil (MO). Consequent neurogenic edema, vasodilation and plasma leakage were assessed by measuring ear thickness with engineer's micrometer, detecting tissue perfusion by laser Doppler scanning and Evans blue or indocyanine green extravasation by intravital videomicroscopy or fluorescence imaging, respectively. Myeloperoxidase activity, an indicator of neutrophil infiltration, was measured from the ear homogenates with spectrophotometry. The selective PAC1R agonist maxadilan, the VPAC1/2R agonist vasoactive intestinal polypeptide (VIP) or the vehicle were administered i.p. 15 min before MO. Substance P (SP) concentration of the ear was assessed by radioimmunoassay. Maxadilan significantly diminished MO-induced neurogenic edema, increase of vascular permeability and vasodilation. These inhibitory effects of maxadilan may be partially due to the decreased substance P (SP) levels. In contrast, inhibitory effect of VIP on ear swelling was moderate, without any effect on MO-induced plasma leakage or SP release, however, activation of VPAC1/2R inhibited the increased microcirculation caused by the early arteriolar vasodilation. Neither the PAC1R, nor the VPAC1/2R agonist influenced the MO-evoked increase in tissue myeloperoxidase activity. These results clearly show that PAC1R activation inhibits acute neurogenic arterial vasodilation and plasma protein leakage from the venules, while VPAC1/2R stimulation is only involved in the attenuation of vasodilation.
Collapse
|
15
|
Decorps J, Saumet JL, Sommer P, Sigaudo-Roussel D, Fromy B. Effect of ageing on tactile transduction processes. Ageing Res Rev 2014; 13:90-9. [PMID: 24373814 DOI: 10.1016/j.arr.2013.12.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Revised: 12/02/2013] [Accepted: 12/16/2013] [Indexed: 01/09/2023]
Abstract
With advancing age, a decline in the main sensory modalities including touch sensation and perception is well reported to occur. This review mainly outlines the peripheral components of touch perception highlighting ageing influences on morphological and functional features of cutaneous mechanical transducers and mechanosensitive ion channels, sensory innervation, neurotransmitters and even vascular system required to ensure efferent function of the afferent nerve fibres in the skin. This, in conjunction with effect of ageing on the skin per se and central nervous system, could explain the tactile deficit seen among the ageing population. We also discuss appropriate tools and experimental models available to study the age-related tactile decline.
Collapse
Affiliation(s)
- Johanna Decorps
- Laboratory of Tissue Biology and Therapeutic Engineering, Centre National de la Recherche Scientifique (CNRS) UMR 5305, Lyon, France; University of Lyon 1, UMR 5305, Lyon, France.
| | - Jean Louis Saumet
- Laboratory of Tissue Biology and Therapeutic Engineering, Centre National de la Recherche Scientifique (CNRS) UMR 5305, Lyon, France; University of Lyon 1, UMR 5305, Lyon, France.
| | - Pascal Sommer
- Laboratory of Tissue Biology and Therapeutic Engineering, Centre National de la Recherche Scientifique (CNRS) UMR 5305, Lyon, France; University of Lyon 1, UMR 5305, Lyon, France.
| | - Dominique Sigaudo-Roussel
- Laboratory of Tissue Biology and Therapeutic Engineering, Centre National de la Recherche Scientifique (CNRS) UMR 5305, Lyon, France; University of Lyon 1, UMR 5305, Lyon, France.
| | - Berengere Fromy
- Laboratory of Tissue Biology and Therapeutic Engineering, Centre National de la Recherche Scientifique (CNRS) UMR 5305, Lyon, France; University of Lyon 1, UMR 5305, Lyon, France.
| |
Collapse
|
16
|
Inglott MA, Lerner EA, Pilowsky PM, Farnham MMJ. Activation of PAC(1) and VPAC receptor subtypes elicits differential physiological responses from sympathetic preganglionic neurons in the anaesthetized rat. Br J Pharmacol 2013; 167:1089-98. [PMID: 22612450 DOI: 10.1111/j.1476-5381.2012.02045.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND AND PURPOSE Pituitary adenylate cyclase-activating polypeptide (PACAP) is an excitatory neuropeptide with central and peripheral cardiovascular actions. Intrathecal PACAP increases splanchnic sympathetic nerve activity and heart rate, but not mean arterial pressure (MAP). We hypothesize that the three PACAP receptors (PAC(1) , VPAC(1) and VPAC(2) ) have different actions in central cardiovascular control, and that their summed effect results in the lack of MAP response observed following intrathecal PACAP injection. EXPERIMENTAL APPROACH The effects of the PACAP receptors on baseline cardiovascular parameters were investigated using selective agonists and antagonists administered into the intrathecal space of urethane-anaesthetized, vagotomized and artificially ventilated male Sprague-Dawley rats. KEY RESULTS Selective activation of the PACAP receptors had different effects on MAP. When activated by maxadilan, PAC(1) receptors increased MAP. The VPAC receptors decreased MAP when both were activated with vasoactive intestinal polypeptide or when only VPAC(1) receptors were activated. The PAC(1) and VPAC(2) receptor antagonist PACAP(6-38) had no cardiovascular effects, suggesting that PACAP is not tonically released. CONCLUSIONS AND IMPLICATIONS PACAP neurotransmission was not responsible for the moment-to-moment tonic regulation of central cardiovascular control mechanisms. Nevertheless, PACAP release within the spinal cord may have pleiotropic effects on sympathetic outflow depending on the postsynaptic receptor type. PAC(1) and VPAC receptor subtypes produced opposing changes in blood pressure when activated by intrathecal PACAP-38 in the anaesthetized Sprague-Dawley rat, resulting in no net change in MAP.
Collapse
Affiliation(s)
- Melissa A Inglott
- Australian School of Advanced Medicine, Macquarie University, Sydney, NSW, Australia
| | | | | | | |
Collapse
|
17
|
Pituitary adenylate cyclase-activating polypeptide induces postsynaptically expressed potentiation in the intra-amygdala circuit. J Neurosci 2013; 32:14165-77. [PMID: 23055486 DOI: 10.1523/jneurosci.1402-12.2012] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is a pleiotropic neuropeptide expressed in the brain, where it may act as a neuromodulator or neurotransmitter contributing to different behavioral processes and stress responses. PACAP is highly expressed in the amygdala, a subcortical brain area involved in both innate and learned fear, suggesting a role for PACAP-mediated signaling in fear-related behaviors. It remains unknown, however, whether and how PACAP affects neuronal and synaptic functions in the amygdala. In this study, we focused on neurons in the lateral division of the central nucleus (CeL), where PACAP-positive presynaptic terminals were predominantly found within the amygdala. In our experiments on rat brain slices, exogenous application of PACAP did not affect either resting membrane potential or membrane excitability of CeL neurons. PACAP enhanced, however, excitatory synaptic transmission in projections from the basolateral nucleus (BLA) to the CeL, while inhibitory transmission in the same pathway was unaffected. PACAP-induced potentiation of glutamatergic synaptic responses persisted after the washout of PACAP and was blocked by the VPAC1 receptor antagonist, suggesting that VPAC1 receptors might mediate synaptic effects of PACAP in the CeL. Moreover, potentiation of synaptic transmission by PACAP was dependent on postsynaptic activation of protein kinase A and calcium/calmodulin-dependent protein kinase II, as well as synaptic targeting of GluR1 subunit-containing AMPA receptors. Thus, PACAP may upregulate excitatory neurotransmission in the BLA-CeL pathway postsynaptically, consistent with the known roles of PACAP in control of fear-related behaviors.
Collapse
|
18
|
Kellogg DL, Zhao JL, Wu Y, Johnson JM. Nitric oxide and receptors for VIP and PACAP in cutaneous active vasodilation during heat stress in humans. J Appl Physiol (1985) 2012; 113:1512-8. [PMID: 22961270 DOI: 10.1152/japplphysiol.00859.2012] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
VPAC2 receptors sensitive to vasoactive intestinal polypeptide (VIP) and pituitary adenylyl cyclase activating polypeptide (PACAP), PAC1 receptors sensitive to PACAP, and nitric oxide (NO) generation by NO synthase (NOS) are all implicated in cutaneous active vasodilation (AVD) through incompletely defined mechanisms. We hypothesized that VPAC2/PAC1 receptor activation and NO are synergistic and interdependent in AVD and tested our hypothesis by examining the effects of VPAC2/PAC1 receptor blockade with and without NOS inhibition during heat stress. The VPAC2/PAC1 antagonist, pituitary adenylate cyclase activating peptide 6-38 (PACAP6-38) and the NOS inhibitor, N(G)-nitro-l-arginine methyl ester (l-NAME) were administered by intradermal microdialysis. PACAP6-38, l-NAME, a combination of PACAP6-38 and l-NAME, or Ringer's solution alone were perfused at four separate sites. Skin blood flow was monitored by laser-Doppler flowmetry at each site. Body temperature was controlled with water-perfused suits. Blood pressure was monitored by Finapres, and cutaneous vascular conductance (CVC) calculated (CVC = laser-Doppler flowmetry/mean arterial pressure). The protocol began with a 5- to 10-min baseline period without antagonist perfusion, followed by perfusion of PACAP6-38, l-NAME, or combined PACAP6-38 and l-NAME at the different sites in normothermia (45 min), followed by 3 min of whole body cooling. Whole body heating was then performed to induce heat stress and activate AVD. Finally, 58 mM sodium nitroprusside were perfused at all sites to effect maximal vasodilation for normalization of blood flow data. No significant differences in CVC (normalized to maximum) were found among Ringer's PACAP6-38, l-NAME, or combined antagonist sites during normothermia (P > 0.05 among sites) or cold stress (P > 0.05 among sites). CVC responses at all treated sites were attenuated during AVD (P < 0.05 vs. Ringer's). Attenuation was greater at l-NAME and combined PACAP6-38- and l-NAME-treated sites than at PACAP6-38 sites (P > 0.05). Because responses did not differ between l-NAME and combined treatment sites (P > 0.05), we conclude that VPAC2/PAC1 receptors require NO in series to effect AVD.
Collapse
Affiliation(s)
- Dean L Kellogg
- Geriatric Research, Education, and Clinical Center, Department of Veterans Affairs, South Texas Veterans Health Care System, Audie L. Murphy Memorial Veterans Hospital Division, San Antonio, TX 78229, USA.
| | | | | | | |
Collapse
|
19
|
Chronic sciatic nerve injury impairs the local cutaneous neurovascular interaction in rats. Pain 2012; 153:149-157. [DOI: 10.1016/j.pain.2011.10.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Revised: 09/23/2011] [Accepted: 10/03/2011] [Indexed: 12/15/2022]
|
20
|
Nishimoto M, Miyakawa H, Wada K, Furuta A. Activation of the VIP/VPAC2 system induces reactive astrocytosis associated with increased expression of glutamate transporters. Brain Res 2011; 1383:43-53. [DOI: 10.1016/j.brainres.2011.01.082] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2010] [Revised: 01/21/2011] [Accepted: 01/21/2011] [Indexed: 11/28/2022]
|
21
|
Doan ND, Bourgault S, Dejda A, Létourneau M, Detheux M, Vaudry D, Vaudry H, Chatenet D, Fournier A. Design and in vitro characterization of PAC1/VPAC1-selective agonists with potent neuroprotective effects. Biochem Pharmacol 2011; 81:552-61. [DOI: 10.1016/j.bcp.2010.11.015] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2010] [Revised: 11/19/2010] [Accepted: 11/19/2010] [Indexed: 01/25/2023]
|
22
|
Farnham MMJ, Inglott MA, Pilowsky PM. Intrathecal PACAP-38 causes increases in sympathetic nerve activity and heart rate but not blood pressure in the spontaneously hypertensive rat. Am J Physiol Heart Circ Physiol 2010; 300:H214-22. [PMID: 20952662 DOI: 10.1152/ajpheart.00662.2010] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The rostral ventrolateral medulla contains presympathetic neurons that project monosynaptically to sympathetic preganglionic neurons (SPN) in the spinal cord and are essential for the tonic and reflex control of the cardiovascular system. SPN directly innervate the adrenal medulla and, via postganglionic axons, affect the heart, kidneys, and blood vessels to alter sympathetic outflow and hence blood pressure. Over 80% of bulbospinal, catecholaminergic (C1) neurons contain pituitary adenylate cyclase-activating polypeptide (PACAP) mRNA. Activation of PACAP receptors with intrathecal infusion of PACAP-38 causes a robust, prolonged elevation in sympathetic tone. Given that a common feature of most forms of hypertension is elevated sympathetic tone, this study aimed to determine in the spontaneously hypertensive rat (SHR) and the Wistar Kyoto rat (normotensive control) 1) the proportion of C1 neurons containing PACAP mRNA and 2) responsiveness to intrathecal PACAP-38. We further investigated whether intrathecal infusion of the PACAP antagonist, PACAP(6-38), reduces the hypertension in the SHR. The principal findings are that 1) the proportion of PACAP mRNA-containing C1 neurons is not different between normotensive and hypertensive rats, 2) intrathecal PACAP-38 causes a strain-dependent, sustained sympathoexcitation and tachycardia with variable effects on mean arterial pressure in normotensive and hypertensive rats, and 3) PACAP(6-38) effectively attenuated the effects of intrathecal PACAP-38, but had no effect alone, on any baseline variables. This finding indicates that PACAP-38 is not tonically released in the spinal cord of rats. A role for PACAP in hypertension in conscious rats remains to be determined.
Collapse
Affiliation(s)
- Melissa M J Farnham
- Australian School of Advanced Medicine, Macquarie University, Sydney, New South Wales, Australia
| | | | | |
Collapse
|
23
|
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: 1.6] [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
|
24
|
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: 1.5] [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
|
25
|
|
26
|
KASPAREK MS, FATIMA J, IQBAL CW, SARR MG. Effects of extrinsic denervation on innervation with VIP and substance P in circular muscle of rat jejunum. Neurogastroenterol Motil 2008; 20:808-17. [PMID: 18282172 PMCID: PMC2561231 DOI: 10.1111/j.1365-2982.2008.01083.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Extrinsic denervation contributes to enteric motor dysfunction after small bowel transplantation (SBT). Our aim was to determine changes in nonadrenergic, noncholinergic innervation with vasoactive intestinal polypeptide (VIP) and substance P (Sub P) in rat jejunal circular muscle after SBT. Muscle strips were studied in tissue chambers from six groups of rats (n > or = 6 per group): naïve controls (NC), animals 1 week after anaesthesia/sham celiotomy (SC-1), and 1 and 8 weeks after jejunal and ileal transection/reanastomosis (TA-1, TA-8) and after syngeneic, orthotopic SBT (SBT-1, SBT-8). Response to exogenous VIP and Sub P and their endogenous release during electrical field stimulation (EFS) were studied. Exogenous VIP and Sub P caused a dose-dependent inhibition and stimulation of mechanical activity in all groups respectively (P < 0.05). The responses to VIP and Sub P were decreased (compared to NC) in all groups at 1 and 8 weeks postoperatively. The VIP antagonist ([D-p-Cl-Phe(6),Leu(17)]-VIP) did not prevent the inhibition by exogenous VIP in any group, while the Sub P antagonist ([D-Pro(2),D-Trp(7,9)]-Sub P) prevented the effect of exogenous Sub P in NC, TA-8 and SBT-8 (P < 0.05). Responses to exogenous VIP were unaffected by the nitric oxide synthase inhibitor l-N(G)-nitro arginine and precontraction of muscle strips with Sub P. Endogenous release of VIP and Sub P during EFS was preserved after SBT. In circular muscle of rat jejunum, changes in neuromuscular transmission with VIP and Sub P during the first 8 weeks after SBT are not mediated by extrinsic denervation.
Collapse
Affiliation(s)
- M. S. KASPAREK
- Department of Surgery and Gastroenterology Research Unit (GU 10-01), Mayo Clinic, Rochester, MN, USA,Department of Surgery, Ludwig-Maximilian’s-University Munich, Munich, Germany
| | - J. FATIMA
- Department of Surgery and Gastroenterology Research Unit (GU 10-01), Mayo Clinic, Rochester, MN, USA
| | - C. W. IQBAL
- Department of Surgery and Gastroenterology Research Unit (GU 10-01), Mayo Clinic, Rochester, MN, USA
| | - M. G. SARR
- Department of Surgery and Gastroenterology Research Unit (GU 10-01), Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
27
|
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: 2.7] [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
|
28
|
KASPAREK MS, FATIMA J, IQBAL CW, DUENES JA, SARR MG. Effect of chronic, extrinsic denervation on functional NANC innervation with vasoactive intestinal polypeptide and substance P in longitudinal muscle of rat jejunum. Neurogastroenterol Motil 2008; 20:243-52. [PMID: 17971029 PMCID: PMC2561234 DOI: 10.1111/j.1365-2982.2007.01021.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Intestinal denervation contributes to enteric motor dysfunction after intestinal transplantation [small bowel transplantation (SBT)]. Our aim was to determine long-term effects of extrinsic denervation on functional non-adrenergic, non-cholinergic innervation with vasoactive intestinal polypeptide (VIP) and substance P. Contractile activity of jejunal longitudinal muscle from six age-matched, naïve control rats (NC) and eight rats 1 year after syngeneic SBT were studied in tissue chambers. Spontaneous contractile activity did not differ between groups. Exogenous VIP inhibited contractile activity dose-dependently in both groups, greater in NC than in SBT. The VIP antagonist ([D-p-Cl-Phe(6),Leu(17)]-VIP) and the nitric oxide synthase inhibitor l-N(G)-nitro arginine prevented inhibition by exogenous VIP and electrical field stimulation (EFS) in both groups. Exogenous substance P increased contractile activity dose-dependently, greater in NC than in SBT. The substance P antagonist ([D-Pro(2),D-Trp(7,9)]-substance P) inhibited effects of exogenous substance P and increased the EFS-induced inhibitory response. Immunohistofluorescence showed staining for tyrosine hydroxylase in the jejunoileum 1 year after SBT suggesting sympathetic reinnervation. In rat jejunal longitudinal muscle after chronic denervation, response to exogenous VIP and substance P is decreased, while endogenous release of both neurotransmitters is preserved. These alterations in excitatory and inhibitory pathways occur despite extrinsic reinnervation and might contribute to enteric motor dysfunction after SBT.
Collapse
Affiliation(s)
- M. S. KASPAREK
- Department of Surgery and Gastrointestinal Research Unit, Mayo Clinic College of Medicine, Rochester, MN, USA, Department of General Surgery, Eberhard-Karls-University, Tuebingen, Germany
| | - J. FATIMA
- Department of Surgery and Gastrointestinal Research Unit, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - C. W. IQBAL
- Department of Surgery and Gastrointestinal Research Unit, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - J. A. DUENES
- Department of Surgery and Gastrointestinal Research Unit, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - M. G. SARR
- Department of Surgery and Gastrointestinal Research Unit, Mayo Clinic College of Medicine, Rochester, MN, USA
| |
Collapse
|
29
|
Kasparek MS, Fatima J, Iqbal CW, Duenes JA, Sarr MG. Long-term effects of extrinsic denervation on VIP and substance P innervation in circular muscle of rat jejunum. J Gastrointest Surg 2007; 11:1339-50. [PMID: 17634766 DOI: 10.1007/s11605-007-0212-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2007] [Accepted: 06/10/2007] [Indexed: 01/31/2023]
Abstract
Intestinal denervation contributes to enteric motor dysfunction after small bowel transplantation (SBT). Our aim was to determine long-term effects of extrinsic denervation on function of nonadrenergic, noncholinergic innervation with substance P and vasoactive intestinal polypeptide (VIP). Contractile activity of jejunal circular muscle strips from six age-matched, naive control rats (NC) and eight rats 1 year after syngeneic SBT was studied in tissue chambers. Spontaneous contractile activity did not differ between groups. Exogenous VIP inhibited contractile activity dose-dependently to a comparable degree in both groups. The VIP antagonist ([D-p-Cl-Phe(6),Leu(17)]-VIP) and the nitric oxide synthase inhibitor L-NG-nitro-arginine did not affect VIP-induced inhibition but increased contractile activity during electrical field stimulation (EFS) in both groups. Exogenous substance P increased contractile activity dose-dependently, greater in NC than SBT. The substance P antagonist ([D-Pro(2),D-Trp(7,9)]-substance P) inhibited effects of exogenous substance P and decreased the excitatory EFS response. Immunohistofluorescence showed tyrosine hydroxylase staining after SBT indicating sympathetic reinnervation. In jejunal circular muscle after chronic denervation, response to exogenous substance P, but not VIP, is decreased, whereas endogenous release of both neurotransmitters is preserved. Alterations in balance of excitatory and inhibitory pathways occur despite extrinsic reinnervation and might contribute to enteric motor dysfunction after SBT.
Collapse
Affiliation(s)
- Michael S Kasparek
- Department of Surgery and Gastrointestinal Research Unit, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | | | | | | | | |
Collapse
|
30
|
Kasparek MS, Fatima J, Iqbal CW, Duenes JA, Sarr MG. Role of VIP and Substance P in NANC Innervation in the Longitudinal Smooth Muscle of the Rat Jejunum—Influence of Extrinsic Denervation. J Surg Res 2007; 141:22-30. [PMID: 17512547 DOI: 10.1016/j.jss.2007.01.021] [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: 01/08/2007] [Revised: 01/08/2007] [Accepted: 01/23/2007] [Indexed: 10/23/2022]
Abstract
BACKGROUND This study was designed to determine changes in nonadrenergic, noncholinergic (NANC) neurotransmission mediated by Vasoactive Intestinal Polypeptide (VIP) and Substance P after small bowel transplantation (SBT). MATERIALS AND METHODS Six groups of rats (n > or = 6 per group) were studied: naïve controls (NC); 1 wk after anesthesia/sham celiotomy (SC-1); 1 or 8 wk after jejunal and ileal transection/reanastomosis (TA-1, TA-8), or syngeneic, orthotopic SBT (SBT-1, SBT-8). Jejunal longitudinal muscle strips were studied under NANC-conditions for spontaneous contractile activity, response to exogenous VIP and Substance P, and electrical field stimulation (EFS). RESULTS Spontaneous activity did not differ between the six groups. VIP inhibited contractile activity in all groups 1 wk postoperatively (P < 0.05), which was prevented by the NO synthase inhibitor L-N(G)-nitro arginine (L-NNA). In contrast, VIP had no effect in the other groups. Precontraction with Substance P exposed an inhibitory effect of VIP in all groups (P < 0.05 each). Substance P increased contractile activity in all groups, but to a lesser extent in SBT-8 compared with NC, TA-8, and SBT-1 (P < 0.05). The inhibitory effect of EFS at 6 Hz was prevented by L-NNA in NC and TA-8; addition of the VIP antagonist ([D-p-Cl-Phe(6), Leu(17)]-VIP) increased contractile activity in NC, but not in TA-8 and SBT-8. The Substance P antagonist ([D-Pro(2), D-Trp(7,9)]-Substance P) decreased contractile activity during EFS at 50 Hz in NC and SBT-8. CONCLUSIONS SBT decreased response to exogenous Substance P, although release of endogenous Substance P (EFS) is preserved. Changes in VIP signaling are acute and reversible and not caused by effects of SBT.
Collapse
Affiliation(s)
- Michael S Kasparek
- Department of Surgery and Gastrointestinal Research Unit, Mayo Clinic, Rochester, Minnesota 55902, USA
| | | | | | | | | |
Collapse
|
31
|
Xie Y, Wolff DW, Lin MF, Tu Y. Vasoactive Intestinal Peptide Transactivates the Androgen Receptor through a Protein Kinase A-Dependent Extracellular Signal-Regulated Kinase Pathway in Prostate Cancer LNCaP Cells. Mol Pharmacol 2007; 72:73-85. [PMID: 17430995 DOI: 10.1124/mol.107.033894] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Acquisition of androgen independence by prostate cancer is the key problem of prostate cancer progression. Vasoactive intestinal peptide (VIP), a neuropeptide, may act as a survival factor for prostate cancer cells under androgen deprivation. However, the molecular mechanisms by which VIP promotes the androgen-independent growth of androgen-sensitive prostate cancer cells have not been addressed. We therefore investigated the biological effect and signal pathway of VIP in LNCaP cells, a prostate cancer cell line that requires androgens for growth. We showed that low nanomolar concentrations of VIP, acting through G(s)-protein-coupled VIP receptors, can induce LNCaP cell growth in the absence of androgen. Blockade of androgen-receptor (AR) in these cells by AR antagonist bicalutamide or by anti-AR small interfering RNA, inhibited the proliferative effect of VIP. In addition, VIP stimulated androgen-independent activation of AR with an EC(50) of 3.0 +/- 0.8 nM. We then investigated VIP-stimulated signaling events that may interact with the AR pathway in prostate cancer cells. VIP regulation of AR activation, mediated by VIP receptors, was protein kinase A (PKA)-dependent, and extracellular signal-regulated kinase 1/2 (ERK1/2) activation contributes to VIP-mediated AR activation. Furthermore, PKA-dependent Rap1 activation is required for both ERK1/2 activation and androgen-independent AR activation in LNCaP cells upon VIP stimulation. Finally, we showed that VIP-induced AR activation was also present in prostate cancer CWR22Rv1 and PC3 cells transfected with the wild-type AR. Altogether, we demonstrate that VIP acting through its G(s)-protein-coupled receptors can cause androgen-independent transactivation of AR through a PKA/Rap1/ERK1/2 pathway, thus promoting androgen-independent proliferation of androgen-sensitive prostate cancer cells.
Collapse
Affiliation(s)
- Yan Xie
- Department of Pharmacology, Creighton University School of Medicine, Omaha, NE 68178, USA
| | | | | | | |
Collapse
|
32
|
Fromy B, Sigaudo-Roussel D, Baron C, Roquelaure Y, Leftheriotis G, Saumet JL. Neuroendocrine pathway involvement in the loss of the cutaneous pressure-induced vasodilatation during acute pain in rats. J Physiol 2006; 579:247-54. [PMID: 17158176 PMCID: PMC2075372 DOI: 10.1113/jphysiol.2006.121426] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Pain is regarded as a risk factor in pressure ulcer development by contributing to immobility. Pressure-induced vasodilatation (PIV) is a mechanism whereby cutaneous blood flow increases in response to progressive locally applied pressure, thereby delaying the occurrence of ischaemia and appearing to be a protective response to local pressure. When the interaction between nervous and vascular systems is deregulated, PIV, which relies on both systems, is absent. We thus hypothesized that acute pain could alter PIV. This study investigated the effects on PIV of acute pain triggered by noxious heat (50 degrees C) applied to the tail of anaesthetized rats. To address the mechanisms underlying these effects, chronic sympathectomy was performed using guanethidine, and the plasma concentrations of pituitary adrenocorticotrophin (ACTH) and catecholamines were measured. Our results show that acute pain induces a loss of PIV associated with an increase of ACTH. Direct involvement of hypertensive effects and peripheral sympathetic nervous system are excluded in the loss of PIV, whereas the activation of brain structures that have descending inhibitory control cannot be excluded. A low dose of systemic morphine prevented this loss of PIV and maintained the ability of the cutaneous microcirculation to adapt to the applied pressure. The loss of a protective response to local pressure (PIV) induced by acute pain lends physiological support to the direct involvement of pain in pressure ulcer development. Therefore, an adequate evaluation and treatment of pain is crucial.
Collapse
Affiliation(s)
- Bérengère Fromy
- Integrative neuro-vascular biology, UMR CNRS 6214-INSERM 771, University of Angers, Rue Haute de Reculee, F-49045 Angers, France
| | | | | | | | | | | |
Collapse
|
33
|
Roosterman D, Goerge T, Schneider SW, Bunnett NW, Steinhoff M. Neuronal Control of Skin Function: The Skin as a Neuroimmunoendocrine Organ. Physiol Rev 2006; 86:1309-79. [PMID: 17015491 DOI: 10.1152/physrev.00026.2005] [Citation(s) in RCA: 416] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
This review focuses on the role of the peripheral nervous system in cutaneous biology and disease. During the last few years, a modern concept of an interactive network between cutaneous nerves, the neuroendocrine axis, and the immune system has been established. We learned that neurocutaneous interactions influence a variety of physiological and pathophysiological functions, including cell growth, immunity, inflammation, pruritus, and wound healing. This interaction is mediated by primary afferent as well as autonomic nerves, which release neuromediators and activate specific receptors on many target cells in the skin. A dense network of sensory nerves releases neuropeptides, thereby modulating inflammation, cell growth, and the immune responses in the skin. Neurotrophic factors, in addition to regulating nerve growth, participate in many properties of skin function. The skin expresses a variety of neurohormone receptors coupled to heterotrimeric G proteins that are tightly involved in skin homeostasis and inflammation. This neurohormone-receptor interaction is modulated by endopeptidases, which are able to terminate neuropeptide-induced inflammatory or immune responses. Neuronal proteinase-activated receptors or transient receptor potential ion channels are recently described receptors that may have been important in regulating neurogenic inflammation, pain, and pruritus. Together, a close multidirectional interaction between neuromediators, high-affinity receptors, and regulatory proteases is critically involved to maintain tissue integrity and regulate inflammatory responses in the skin. A deeper understanding of cutaneous neuroimmunoendocrinology may help to develop new strategies for the treatment of several skin diseases.
Collapse
|
34
|
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: 3.1] [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
|
35
|
Garry A, Sigaudo-Roussel D, Merzeau S, Dumont O, Saumet JL, Fromy B. Cellular mechanisms underlying cutaneous pressure-induced vasodilation: in vivo involvement of potassium channels. Am J Physiol Heart Circ Physiol 2005; 289:H174-80. [PMID: 15734881 DOI: 10.1152/ajpheart.01020.2004] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In the skin of humans and rodents, local pressure induces localized cutaneous vasodilation, which may be protective against pressure-induced microvascular dysfunction and lesion formation. Once activated by the local pressure application, capsaicin-sensitive nerve fibers release neuropeptides that act on the endothelium to synthesize and release nitric oxide (NO) and prostaglandins, leading to the development of the cutaneous pressure-induced vasodilation (PIV). The present study was undertaken to test in vivo the hypothesis that PIV is mediated or modulated by differential activation of K+ channels in anesthetized rats using pharmacological methods. Local pressure was applied at 11.1 Pa/s. Endothelium-independent and -dependent vasodilation were tested using iontophoretic delivery of sodium nitroprusside (SNP) and acetylcholine (ACh), respectively, and was correlated with PIV response. PIV was reduced after systemic administration of tetraethylammonium (a nonspecific K+ channel blocker), iberiotoxin [a specific large-conductance Ca2+-activated K+ (BKCa) channel blocker], and glibenclamide [a specific ATP-sensitive K+ (KATP) channel blocker], whereas PIV was unchanged by apamin (a specific small-conductance Ca2+-activated K+ channel blocker) and 4-aminopyridine (a specific voltage-sensitive K+ channel blocker). The responses to SNP and ACh were reduced by iberiotoxin but were unchanged by glibenclamide. We conclude that the cellular mechanism of PIV in skin involves BKCa and KATP channels. We suggest that the opening of BKCa and KATP channels contributes to the hyperpolarization of vascular smooth muscle cells to produce PIV development mainly via the NO and prostaglandin pathways, respectively.
Collapse
Affiliation(s)
- Ambroise Garry
- Laboratory of Physiology, UMR Centre National de la Recherche Scientifique 6188, School of Medicine, F-49045 Angers Cedex, France
| | | | | | | | | | | |
Collapse
|