1
|
Park KD, Son JY, Kim HK, Kim YM, Ju JS, Jo MJ, Park MK, Lee MK, Ahn DK. Differential Regulation of Intracisternally Injected Angiotensin II-Induced Mechanical Allodynia and Thermal Hyperalgesia in Rats. Biomedicines 2023; 11:3279. [PMID: 38137500 PMCID: PMC10741042 DOI: 10.3390/biomedicines11123279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 12/01/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023] Open
Abstract
The present study examined the underlying mechanisms of mechanical allodynia and thermal hyperalgesia induced by the intracisternal injection of angiotensin (Ang) II. Intracisternal Ang II injection decreased the air puff threshold and head withdrawal latency. To determine the operative receptors for each distinct type of pain behavior, we intracisternally injected Ang II receptor antagonists 2 h after Ang II injection. Losartan, an Ang II type 1 receptor (AT1R) antagonist, alleviated mechanical allodynia. Conversely, PD123319, an Ang II type 1 receptor (AT2R) antagonist, blocked only thermal hyperalgesia. Immunofluorescence analyses revealed the co-localization of AT1R with the astrocyte marker GFAP in the trigeminal subnucleus caudalis and co-localization of AT2R with CGRP-positive neurons in the trigeminal ganglion. Intracisternal pretreatment with minocycline, a microglial inhibitor, did not affect Ang II-induced mechanical allodynia, whereas L-α-aminoadipate, an astrocyte inhibitor, significantly inhibited Ang II-induced mechanical allodynia. Furthermore, subcutaneous pretreatment with botulinum toxin type A significantly alleviated Ang II-induced thermal hyperalgesia, but not Ang II-induced mechanical allodynia. These results indicate that central Ang II-induced nociception is differentially regulated by AT1R and AT2R. Thus, distinct therapeutic targets must be regulated to overcome pain symptoms caused by multiple underlying mechanisms.
Collapse
Affiliation(s)
- Ki-Don Park
- Department of Oral Physiology, School of Dentistry, Kyungpook National University, Daegu 41940, Republic of Korea; (K.-D.P.); (J.-Y.S.); (H.-K.K.); (Y.-M.K.); (J.-S.J.); (M.-J.J.)
| | - Jo-Young Son
- Department of Oral Physiology, School of Dentistry, Kyungpook National University, Daegu 41940, Republic of Korea; (K.-D.P.); (J.-Y.S.); (H.-K.K.); (Y.-M.K.); (J.-S.J.); (M.-J.J.)
| | - Hak-Kyun Kim
- Department of Oral Physiology, School of Dentistry, Kyungpook National University, Daegu 41940, Republic of Korea; (K.-D.P.); (J.-Y.S.); (H.-K.K.); (Y.-M.K.); (J.-S.J.); (M.-J.J.)
| | - Yu-Mi Kim
- Department of Oral Physiology, School of Dentistry, Kyungpook National University, Daegu 41940, Republic of Korea; (K.-D.P.); (J.-Y.S.); (H.-K.K.); (Y.-M.K.); (J.-S.J.); (M.-J.J.)
| | - Jin-Sook Ju
- Department of Oral Physiology, School of Dentistry, Kyungpook National University, Daegu 41940, Republic of Korea; (K.-D.P.); (J.-Y.S.); (H.-K.K.); (Y.-M.K.); (J.-S.J.); (M.-J.J.)
| | - Min-Jeong Jo
- Department of Oral Physiology, School of Dentistry, Kyungpook National University, Daegu 41940, Republic of Korea; (K.-D.P.); (J.-Y.S.); (H.-K.K.); (Y.-M.K.); (J.-S.J.); (M.-J.J.)
| | - Min-Kyoung Park
- Department of Dental Hygiene, Kyung-Woon University, Gumi 39160, Republic of Korea;
| | - Min-Kyung Lee
- Department of Dental Hygiene, Dong-Eui University, Busan 47340, Republic of Korea;
| | - Dong-Kuk Ahn
- Department of Oral Physiology, School of Dentistry, Kyungpook National University, Daegu 41940, Republic of Korea; (K.-D.P.); (J.-Y.S.); (H.-K.K.); (Y.-M.K.); (J.-S.J.); (M.-J.J.)
| |
Collapse
|
2
|
Villapol S, Janatpour ZC, Affram KO, Symes AJ. The Renin Angiotensin System as a Therapeutic Target in Traumatic Brain Injury. Neurotherapeutics 2023; 20:1565-1591. [PMID: 37759139 PMCID: PMC10684482 DOI: 10.1007/s13311-023-01435-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2023] [Indexed: 09/29/2023] Open
Abstract
Traumatic brain injury (TBI) is a major public health problem, with limited pharmacological options available beyond symptomatic relief. The renin angiotensin system (RAS) is primarily known as a systemic endocrine regulatory system, with major roles controlling blood pressure and fluid homeostasis. Drugs that target the RAS are used to treat hypertension, heart failure and kidney disorders. They have now been used chronically by millions of people and have a favorable safety profile. In addition to the systemic RAS, it is now appreciated that many different organ systems, including the brain, have their own local RAS. The major ligand of the classic RAS, Angiotensin II (Ang II) acts predominantly through the Ang II Type 1 receptor (AT1R), leading to vasoconstriction, inflammation, and heightened oxidative stress. These processes can exacerbate brain injuries. Ang II receptor blockers (ARBs) are AT1R antagonists. They have been shown in several preclinical studies to enhance recovery from TBI in rodents through improvements in molecular, cellular and behavioral correlates of injury. ARBs are now under consideration for clinical trials in TBI. Several different RAS peptides that signal through receptors distinct from the AT1R, are also potential therapeutic targets for TBI. The counter regulatory RAS pathway has actions that oppose those stimulated by AT1R signaling. This alternative pathway has many beneficial effects on cells in the central nervous system, bringing about vasodilation, and having anti-inflammatory and anti-oxidative stress actions. Stimulation of this pathway also has potential therapeutic value for the treatment of TBI. This comprehensive review will provide an overview of the various components of the RAS, with a focus on their direct relevance to TBI pathology. It will explore different therapeutic agents that modulate this system and assess their potential efficacy in treating TBI patients.
Collapse
Affiliation(s)
- Sonia Villapol
- Department of Neurosurgery, Houston Methodist Hospital, Houston, TX, USA
| | - Zachary C Janatpour
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Kwame O Affram
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Aviva J Symes
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
| |
Collapse
|
3
|
Gómez-Dabó L, Melgarejo-Martínez L, Caronna E, Pozo-Rosich P. Headache in COVID-19 and Long COVID: to Know Facts for Clinical Practice. Curr Neurol Neurosci Rep 2023; 23:551-560. [PMID: 37665495 DOI: 10.1007/s11910-023-01296-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/21/2023] [Indexed: 09/05/2023]
Abstract
PURPOSE OF REVIEW Headache is one of the most frequent symptoms of the acute and post-acute phase of COVID-19. Specific epidemiology, clinical features, risk factors, pathophysiology, and treatment have been reported in these two scenarios. With this narrative review of the literature, we aim to provide updated knowledge on headache in the COVID-19 setting and give clinicians a practical approach on this topic to guide them in their clinical practice. RECENT FINDINGS Headache mechanisms in COVID-19 are still poorly understood. Strong evidence is also lacking on how to best treat and manage these patients, especially those with persistent and disabling headache after COVID-19. Data are also scarce on the characteristics of headache in COVID-19 caused by the new SARS-CoV-2 (Omicron) variants and how these may influence the acute and persistent symptoms of COVID-19. Patients with pre-existing primary headache disorders remain a particularly concerning population due to their biological predisposition in suffering from headaches and the potential risk of worsening in the setting of SARS-CoV-2 infection. Although there is an exponential growth of scientific evidence, studies are often controversial and focused on the first wave of the pandemic, making COVID-19 headache still a challenging matter for clinicians. New research is therefore needed.
Collapse
Affiliation(s)
- Laura Gómez-Dabó
- Neurology Department, Hospital Universitari Vall d'Hebron, Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Laura Melgarejo-Martínez
- Neurology Department, Hospital Universitari Vall d'Hebron, Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Edoardo Caronna
- Headache Unit, Neurology Department, Hospital Universitari Vall d'Hebron, Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain.
- Headache and Neurological Pain Research Group, Vall d'Hebron Research Institute, Department of Medicine, Universitat Autònoma de Barcelona, Ps. Vall d'Hebron 119-129, 08035, Barcelona, Spain.
| | - Patricia Pozo-Rosich
- Headache Unit, Neurology Department, Hospital Universitari Vall d'Hebron, Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
- Headache and Neurological Pain Research Group, Vall d'Hebron Research Institute, Department of Medicine, Universitat Autònoma de Barcelona, Ps. Vall d'Hebron 119-129, 08035, Barcelona, Spain
| |
Collapse
|
4
|
Abstract
Headache disorders can produce recurrent, incapacitating pain. Migraine and cluster headache are notable for their ability to produce significant disability. The anatomy and physiology of headache disorders is fundamental to evolving treatment approaches and research priorities. Key concepts in headache mechanisms include activation and sensitization of trigeminovascular, brainstem, thalamic, and hypothalamic neurons; modulation of cortical brain regions; and activation of descending pain circuits. This review will examine the relevant anatomy of the trigeminal, brainstem, subcortical, and cortical brain regions and concepts related to the pathophysiology of migraine and cluster headache disorders.
Collapse
Affiliation(s)
- Andrea M Harriott
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Yulia Orlova
- Department of Neurology, University of Florida, Gainesville, Florida
| |
Collapse
|
5
|
Togha M, Hashemi SM, Yamani N, Martami F, Salami Z. A Review on Headaches Due to COVID-19 Infection. Front Neurol 2022; 13:942956. [PMID: 35911910 PMCID: PMC9327440 DOI: 10.3389/fneur.2022.942956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
Since December 2019, the time when the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was spotted, numerous review studies have been published on COVID-19 and its neuro invasion. A growing number of studies have reported headaches as a common neurological manifestation of COVID-19. Although several hypotheses have been proposed regarding the association between headache and the coronavirus, no solid evidence has been presented for the mechanism and features of headache in COVID-19. Headache also is a common complaint with the omicron variant of the virus. COVID-19 vaccination also is a cause of new-onset headaches or aggravation of the previous headache in migraine or tension headache sufferers. In this review study, the types of headaches reported in previous studies and their possible pathogenic mechanisms are outlined. To accomplish this objective, various types of headaches are classified and their patterns are discussed according to ICHD-3 diagnostic criteria, including, headaches attributed to systemic viral infection, viral meningitis or encephalitis, non-infectious inflammatory intracranial disease, hypoxia and/or hypercapnia, cranial or cervical vascular disorder, increased cerebrospinal fluid (CSF) pressure, refractive error, external-compression headache, and cough headache. Then, their pathogeneses are categorized into three main categories, direct trigeminal involvement, vascular invasion, and inflammatory mediators. Furthermore, persistent headache after recovery and the predictors of intensity is further investigated. Post-vaccination headache is also discussed in this review.
Collapse
Affiliation(s)
- Mansoureh Togha
- Headache Department, Iranian Center of Neurological Researches, Institute of Neuroscience, Tehran University of Medical Sciences, Tehran, Iran
- Headache Department, Neurology Ward, Sina Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyedeh Melika Hashemi
- Headache Department, Iranian Center of Neurological Researches, Institute of Neuroscience, Tehran University of Medical Sciences, Tehran, Iran
| | - Nooshin Yamani
- Neurology Department, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Fahimeh Martami
- Headache Department, Iranian Center of Neurological Researches, Institute of Neuroscience, Tehran University of Medical Sciences, Tehran, Iran
| | - Zhale Salami
- Headache Department, Iranian Center of Neurological Researches, Institute of Neuroscience, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Szczepanska-Sadowska E, Wsol A, Cudnoch-Jedrzejewska A, Czarzasta K, Żera T. Multiple Aspects of Inappropriate Action of Renin-Angiotensin, Vasopressin, and Oxytocin Systems in Neuropsychiatric and Neurodegenerative Diseases. J Clin Med 2022; 11:908. [PMID: 35207180 PMCID: PMC8877782 DOI: 10.3390/jcm11040908] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/04/2022] [Accepted: 02/05/2022] [Indexed: 02/04/2023] Open
Abstract
The cardiovascular system and the central nervous system (CNS) closely cooperate in the regulation of primary vital functions. The autonomic nervous system and several compounds known as cardiovascular factors, especially those targeting the renin-angiotensin system (RAS), the vasopressin system (VPS), and the oxytocin system (OTS), are also efficient modulators of several other processes in the CNS. The components of the RAS, VPS, and OTS, regulating pain, emotions, learning, memory, and other cognitive processes, are present in the neurons, glial cells, and blood vessels of the CNS. Increasing evidence shows that the combined function of the RAS, VPS, and OTS is altered in neuropsychiatric/neurodegenerative diseases, and in particular in patients with depression, Alzheimer's disease, Parkinson's disease, autism, and schizophrenia. The altered function of the RAS may also contribute to CNS disorders in COVID-19. In this review, we present evidence that there are multiple causes for altered combined function of the RAS, VPS, and OTS in psychiatric and neurodegenerative disorders, such as genetic predispositions and the engagement of the RAS, VAS, and OTS in the processes underlying emotions, memory, and cognition. The neuroactive pharmaceuticals interfering with the synthesis or the action of angiotensins, vasopressin, and oxytocin can improve or worsen the effectiveness of treatment for neuropsychiatric/neurodegenerative diseases. Better knowledge of the multiple actions of the RAS, VPS, and OTS may facilitate programming the most efficient treatment for patients suffering from the comorbidity of neuropsychiatric/neurodegenerative and cardiovascular diseases.
Collapse
Affiliation(s)
- Ewa Szczepanska-Sadowska
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland; (A.W.); (A.C.-J.); (K.C.); (T.Ż.)
| | | | | | | | | |
Collapse
|
7
|
Király K, Karádi DÁ, Zádor F, Mohammadzadeh A, Galambos AR, Balogh M, Riba P, Tábi T, Zádori ZS, Szökő É, Fürst S, Al-Khrasani M. Shedding Light on the Pharmacological Interactions between μ-Opioid Analgesics and Angiotensin Receptor Modulators: A New Option for Treating Chronic Pain. Molecules 2021; 26:6168. [PMID: 34684749 PMCID: PMC8537077 DOI: 10.3390/molecules26206168] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/05/2021] [Accepted: 10/08/2021] [Indexed: 12/20/2022] Open
Abstract
The current protocols for neuropathic pain management include µ-opioid receptor (MOR) analgesics alongside other drugs; however, there is debate on the effectiveness of opioids. Nevertheless, dose escalation is required to maintain their analgesia, which, in turn, contributes to a further increase in opioid side effects. Finding novel approaches to effectively control chronic pain, particularly neuropathic pain, is a great challenge clinically. Literature data related to pain transmission reveal that angiotensin and its receptors (the AT1R, AT2R, and MAS receptors) could affect the nociception both in the periphery and CNS. The MOR and angiotensin receptors or drugs interacting with these receptors have been independently investigated in relation to analgesia. However, the interaction between the MOR and angiotensin receptors has not been excessively studied in chronic pain, particularly neuropathy. This review aims to shed light on existing literature information in relation to the analgesic action of AT1R and AT2R or MASR ligands in neuropathic pain conditions. Finally, based on literature data, we can hypothesize that combining MOR agonists with AT1R or AT2R antagonists might improve analgesia.
Collapse
MESH Headings
- Analgesics/pharmacology
- Analgesics, Opioid/pharmacology
- Animals
- Chronic Pain/drug therapy
- Humans
- Neuralgia/drug therapy
- Nociception/drug effects
- Pain Management/methods
- Proto-Oncogene Mas
- Receptors, Angiotensin/drug effects
- Receptors, Angiotensin/metabolism
- Receptors, Opioid/agonists
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/drug effects
- Receptors, Opioid, mu/metabolism
Collapse
Affiliation(s)
- Kornél Király
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, P.O. Box 370, H-1445 Budapest, Hungary; (D.Á.K.); (F.Z.); (A.M.); (A.R.G.); (M.B.); (P.R.); (Z.S.Z.); (S.F.)
| | - Dávid Á. Karádi
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, P.O. Box 370, H-1445 Budapest, Hungary; (D.Á.K.); (F.Z.); (A.M.); (A.R.G.); (M.B.); (P.R.); (Z.S.Z.); (S.F.)
| | - Ferenc Zádor
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, P.O. Box 370, H-1445 Budapest, Hungary; (D.Á.K.); (F.Z.); (A.M.); (A.R.G.); (M.B.); (P.R.); (Z.S.Z.); (S.F.)
- Department of Pharmacodynamics, Faculty of Pharmacy, Semmelweis University, Nagyvárad tér 4, H-1089 Budapest, Hungary; (T.T.); (É.S.)
| | - Amir Mohammadzadeh
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, P.O. Box 370, H-1445 Budapest, Hungary; (D.Á.K.); (F.Z.); (A.M.); (A.R.G.); (M.B.); (P.R.); (Z.S.Z.); (S.F.)
| | - Anna Rita Galambos
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, P.O. Box 370, H-1445 Budapest, Hungary; (D.Á.K.); (F.Z.); (A.M.); (A.R.G.); (M.B.); (P.R.); (Z.S.Z.); (S.F.)
| | - Mihály Balogh
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, P.O. Box 370, H-1445 Budapest, Hungary; (D.Á.K.); (F.Z.); (A.M.); (A.R.G.); (M.B.); (P.R.); (Z.S.Z.); (S.F.)
| | - Pál Riba
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, P.O. Box 370, H-1445 Budapest, Hungary; (D.Á.K.); (F.Z.); (A.M.); (A.R.G.); (M.B.); (P.R.); (Z.S.Z.); (S.F.)
| | - Tamás Tábi
- Department of Pharmacodynamics, Faculty of Pharmacy, Semmelweis University, Nagyvárad tér 4, H-1089 Budapest, Hungary; (T.T.); (É.S.)
| | - Zoltán S. Zádori
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, P.O. Box 370, H-1445 Budapest, Hungary; (D.Á.K.); (F.Z.); (A.M.); (A.R.G.); (M.B.); (P.R.); (Z.S.Z.); (S.F.)
| | - Éva Szökő
- Department of Pharmacodynamics, Faculty of Pharmacy, Semmelweis University, Nagyvárad tér 4, H-1089 Budapest, Hungary; (T.T.); (É.S.)
| | - Susanna Fürst
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, P.O. Box 370, H-1445 Budapest, Hungary; (D.Á.K.); (F.Z.); (A.M.); (A.R.G.); (M.B.); (P.R.); (Z.S.Z.); (S.F.)
| | - Mahmoud Al-Khrasani
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, P.O. Box 370, H-1445 Budapest, Hungary; (D.Á.K.); (F.Z.); (A.M.); (A.R.G.); (M.B.); (P.R.); (Z.S.Z.); (S.F.)
| |
Collapse
|
8
|
Abstract
Growing evidence implicates the renin-angiotensin system (RAS) in multiple facets of neuropathic pain (NP). This narrative review focuses primarily on the major bioactive RAS peptide, Angiotensin II (Ang II), and its receptors, namely type 1 (AT1R) and type 2 (AT2R). Both receptors are involved in the development of NP and represent potential therapeutic targets. We first discuss the potential role of Ang II receptors in modulation of NP in the central nervous system. Ang II receptor expression is widespread in circuits associated with the perception and modulation of pain, but more studies are required to fully characterize receptor distribution, downstream signaling, and therapeutic potential of targeting the central nervous system RAS in NP. We then describe the peripheral neuronal and nonneuronal distribution of the RAS, and its contribution to NP. Other RAS modulators (such as Ang (1-7)) are briefly reviewed as well. AT1R antagonists are analgesic across different pain models, including NP. Several studies show neuronal protection and outgrowth downstream of AT2R activation, which may lead to the use of AT2R agonists in NP. However, blockade of AT2R results in analgesia. Furthermore, expression of the RAS in the immune system and a growing appreciation of neuroimmune crosstalk in NP add another layer of complexity and therapeutic potential of targeting this pathway. A growing number of human studies also hint at the analgesic potential of targeting Ang II signaling. Altogether, Ang II receptor signaling represents a promising, far-reaching, and novel strategy to treat NP.
Collapse
|
9
|
Mutiawati E, Syahrul S, Fahriani M, Fajar JK, Mamada SS, Maliga HA, Samsu N, Ilmawan M, Purnamasari Y, Asmiragani AA, Ichsan I, Emran TB, Rabaan AA, Masyeni S, Nainu F, Harapan H. Global prevalence and pathogenesis of headache in COVID-19: A systematic review and meta-analysis. F1000Res 2020; 9:1316. [PMID: 33953911 PMCID: PMC8063523 DOI: 10.12688/f1000research.27334.1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/01/2021] [Indexed: 09/01/2023] Open
Abstract
Background: This study was conducted to determine the prevalence of headache in coronavirus disease 2019 (COVID-19) and to assess its association as a predictor for COVID-19. This study also aimed to discuss the possible pathogenesis of headache in COVID-19. Methods: Available articles from PubMed, Scopus, and Web of Science were searched as of September 2 nd, 2020. Data on characteristics of the study, headache and COVID-19 were extracted following the PRISMA guidelines. Biases were assessed using the Newcastle-Ottawa scale. The cumulative prevalence of headache was calculated for the general population (i.e. adults and children). The pooled odd ratio (OR) with 95% confidence intervals (95%CI) was calculated using the Z test to assess the association between headache and the presence of COVID-19 cases. Results: We included 104,751 COVID-19 cases from 78 eligible studies to calculate the global prevalence of headache in COVID-19 and 17 studies were included to calculate the association of headache and COVID-19. The cumulative prevalence of headache in COVID-19 was 25.2% (26,464 out of 104,751 cases). Headache was found to be more prevalent, approximately by two-fold, in COVID-19 patients than in non-COVID-19 patients (other respiratory viral infections), OR: 1.73; 95% CI: 1.94, 2.5 with p=0.04. Conclusion: Headache is common among COVID-19 patients and seems to be more common in COVID-19 patients compared to those with the non-COVID-19 viral infection. No definitive mechanisms on how headache emerges in COVID-19 patients but several possible hypotheses have been proposed. However, extensive studies are warranted to elucidate the mechanisms. PROSPERO registration: CRD42020210332 (28/09/2020).
Collapse
Affiliation(s)
- Endang Mutiawati
- Department of Neurology, Universitas Syiah Kuala, Banda Aceh, Aceh, 23111, Indonesia
- Department of Neurology, Dr. Zainoel Abidin Hospital, Banda Aceh, Aceh, 23126, Indonesia
| | - Syahrul Syahrul
- Department of Neurology, Universitas Syiah Kuala, Banda Aceh, Aceh, 23111, Indonesia
- Department of Neurology, Dr. Zainoel Abidin Hospital, Banda Aceh, Aceh, 23126, Indonesia
| | - Marhami Fahriani
- Medical Research Unit, School of Medicine, Universitas Syiah Kuala, Banda Aceh, 23111, Indonesia
| | - Jonny Karunia Fajar
- Medical Research Unit, School of Medicine, Universitas Syiah Kuala, Banda Aceh, 23111, Indonesia
- Brawijaya Internal Medicine Research Center, Department of Internal Medicine, Faculty of Medicine, Universitas Brawijaya, Malang, East Java, 65145, Indonesia
| | - Sukamto S. Mamada
- Faculty of Pharmacy, Hasanuddin University, Makassar, South Sulawesi, 90245, Indonesia
| | | | - Nur Samsu
- Brawijaya Internal Medicine Research Center, Department of Internal Medicine, Faculty of Medicine, Universitas Brawijaya, Malang, East Java, 65145, Indonesia
| | - Muhammad Ilmawan
- Faculty of Medicine, Universitas Brawijaya, Malang, East Java, 65117, Indonesia
| | - Yeni Purnamasari
- Faculty of Medicine, Universitas Brawijaya, Malang, East Java, 65117, Indonesia
| | | | - Ichsan Ichsan
- Medical Research Unit, School of Medicine, Universitas Syiah Kuala, Banda Aceh, 23111, Indonesia
- Department of Microbiology, School of Medicine, Universitas Syiah Kuala, Banda Aceh, Aceh, 23111, Indonesia
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, 4381, Bangladesh
| | - Ali A. Rabaan
- Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran, 31311, Saudi Arabia
| | - Sri Masyeni
- Department of Internal Medicine, Faculty of Medicine and Health Sciences, Universitas Warmadewa, Denpasar, Bali, 80235, Indonesia
- Department of Internal Medicine, Sanjiwani Hospital, Denpasar, Bali, 80235, Indonesia
| | - Firzan Nainu
- Faculty of Pharmacy, Hasanuddin University, Makassar, South Sulawesi, 90245, Indonesia
| | - Harapan Harapan
- Medical Research Unit, School of Medicine, Universitas Syiah Kuala, Banda Aceh, 23111, Indonesia
- Department of Microbiology, School of Medicine, Universitas Syiah Kuala, Banda Aceh, Aceh, 23111, Indonesia
- Tropical Disease Centre, School of Medicine, Universitas Syiah Kuala, Banda Aceh, Aceh, 23111, Indonesia
| |
Collapse
|
10
|
Mutiawati E, Syahrul S, Fahriani M, Fajar JK, Mamada SS, Maliga HA, Samsu N, Ilmawan M, Purnamasari Y, Asmiragani AA, Ichsan I, Emran TB, Rabaan AA, Masyeni S, Nainu F, Harapan H. Global prevalence and pathogenesis of headache in COVID-19: A systematic review and meta-analysis. F1000Res 2020; 9:1316. [PMID: 33953911 PMCID: PMC8063523 DOI: 10.12688/f1000research.27334.2] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/01/2021] [Indexed: 01/19/2023] Open
Abstract
Background: This study was conducted to determine the prevalence of headache in coronavirus disease 2019 (COVID-19) and to assess its association as a predictor for COVID-19. This study also aimed to discuss the possible pathogenesis of headache in COVID-19. Methods: Available articles from PubMed, Scopus, and Web of Science were searched as of September 2 nd, 2020. Data on characteristics of the study, headache and COVID-19 were extracted following the PRISMA guidelines. Biases were assessed using the Newcastle-Ottawa scale. The cumulative prevalence of headache was calculated for the general population (i.e. adults and children). The pooled odd ratio (OR) with 95% confidence intervals (95%CI) was calculated using the Z test to assess the association between headache and the presence of COVID-19 cases. Results: We included 104,751 COVID-19 cases from 78 eligible studies to calculate the global prevalence of headache in COVID-19 and 17 studies were included to calculate the association of headache and COVID-19. The cumulative prevalence of headache in COVID-19 was 25.2% (26,464 out of 104,751 cases). Headache was found to be more prevalent, approximately by two-fold, in COVID-19 patients than in non-COVID-19 patients (other respiratory viral infections), OR: 1.73; 95% CI: 1.94, 2.5 with p=0.04. Conclusion: Headache is common among COVID-19 patients and seems to be more common in COVID-19 patients compared to those with the non-COVID-19 viral infection. No definitive mechanisms on how headache emerges in COVID-19 patients but several possible hypotheses have been proposed. However, extensive studies are warranted to elucidate the mechanisms. PROSPERO registration: CRD42020210332 (28/09/2020).
Collapse
Affiliation(s)
- Endang Mutiawati
- Department of Neurology, Universitas Syiah Kuala, Banda Aceh, Aceh, 23111, Indonesia
- Department of Neurology, Dr. Zainoel Abidin Hospital, Banda Aceh, Aceh, 23126, Indonesia
| | - Syahrul Syahrul
- Department of Neurology, Universitas Syiah Kuala, Banda Aceh, Aceh, 23111, Indonesia
- Department of Neurology, Dr. Zainoel Abidin Hospital, Banda Aceh, Aceh, 23126, Indonesia
| | - Marhami Fahriani
- Medical Research Unit, School of Medicine, Universitas Syiah Kuala, Banda Aceh, 23111, Indonesia
| | - Jonny Karunia Fajar
- Medical Research Unit, School of Medicine, Universitas Syiah Kuala, Banda Aceh, 23111, Indonesia
- Brawijaya Internal Medicine Research Center, Department of Internal Medicine, Faculty of Medicine, Universitas Brawijaya, Malang, East Java, 65145, Indonesia
| | - Sukamto S. Mamada
- Faculty of Pharmacy, Hasanuddin University, Makassar, South Sulawesi, 90245, Indonesia
| | | | - Nur Samsu
- Brawijaya Internal Medicine Research Center, Department of Internal Medicine, Faculty of Medicine, Universitas Brawijaya, Malang, East Java, 65145, Indonesia
| | - Muhammad Ilmawan
- Faculty of Medicine, Universitas Brawijaya, Malang, East Java, 65117, Indonesia
| | - Yeni Purnamasari
- Faculty of Medicine, Universitas Brawijaya, Malang, East Java, 65117, Indonesia
| | | | - Ichsan Ichsan
- Medical Research Unit, School of Medicine, Universitas Syiah Kuala, Banda Aceh, 23111, Indonesia
- Department of Microbiology, School of Medicine, Universitas Syiah Kuala, Banda Aceh, Aceh, 23111, Indonesia
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, 4381, Bangladesh
| | - Ali A. Rabaan
- Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran, 31311, Saudi Arabia
| | - Sri Masyeni
- Department of Internal Medicine, Faculty of Medicine and Health Sciences, Universitas Warmadewa, Denpasar, Bali, 80235, Indonesia
- Department of Internal Medicine, Sanjiwani Hospital, Denpasar, Bali, 80235, Indonesia
| | - Firzan Nainu
- Faculty of Pharmacy, Hasanuddin University, Makassar, South Sulawesi, 90245, Indonesia
| | - Harapan Harapan
- Medical Research Unit, School of Medicine, Universitas Syiah Kuala, Banda Aceh, 23111, Indonesia
- Department of Microbiology, School of Medicine, Universitas Syiah Kuala, Banda Aceh, Aceh, 23111, Indonesia
- Tropical Disease Centre, School of Medicine, Universitas Syiah Kuala, Banda Aceh, Aceh, 23111, Indonesia
| |
Collapse
|
11
|
Bolay H, Özge A, Uludüz D, Baykan B. Are Migraine Patients at Increased Risk for Symptomatic Coronavirus Disease 2019 Due to Shared Comorbidities? Headache 2020; 60:2508-2521. [PMID: 33124044 DOI: 10.1111/head.13998] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 09/24/2020] [Accepted: 10/02/2020] [Indexed: 01/08/2023]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has rapidly transformed the whole world and forced us to look through comorbid diseases and risk factors from a different perspective. COVID-19 shows some inherent risk factors like cardiovascular comorbidities independent from age, gender, and geographic location. One of the most peculiar features of the COVID-19 pandemic is that severe acute respiratory syndrome coronavirus 2 respiratory infections disproportionately impact patients with hypertension, diabetes, and other cardiovascular comorbidities rather than those with allergic respiratory diseases and immune-compromised conditions. Migraine is a complex neuro-vasculo-inflammatory disorder that is also packed frequently with certain medical conditions including vascular disorders, hypertension, allergic diseases such as asthma and systemic inflammatory disorders. Accordingly, 2 different questions arise during the pandemic: (1) Do share comorbidities of cardiovascular diseases and hypertension increase the risk of symptomatic COVID-19 for migraine patients? (2) Do comorbid allergic and atopic diseases, including asthma act as opposite influencers alongside with female gender? This paper focuses on the co-existence of comorbidities of COVID-19, in comparison with migraine, based on a wide clinical dataset and available reports. Discussed mechanisms include potential strategic roles of angiotensin-converting enzyme 2, angiotensin-II, and nucleotide oligomerization domain-like receptor family, pyrin domain containing 3 inflammasome, playing remarkable parts in the pathogenesis of COVID-19 and migraine. There are also some clues about the importance of endothelial and pericyte dysfunction and neuroinflammation in COVID-19 infection, related to complications and survival of the patients. The large epidemiological studies as well as basic research, focusing on migraine patients with COVID-19 will clarify these vital questions during the upcoming periods.
Collapse
Affiliation(s)
- Hayrunnisa Bolay
- Department of Neurology and Algology, Medical Faculty, Gazi University, Ankara, Turkey
| | - Aynur Özge
- Department of Neurology and Algology, Medical Faculty, Mersin University, Mersin, Turkey
| | - Derya Uludüz
- Department of Neurology and Algology, Cerrahpasa Medical Faculty, Istanbul University, Istanbul, Turkey
| | - Betül Baykan
- Department of Neurology, Headache Center, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| |
Collapse
|
12
|
Szczepanska-Sadowska E, Cudnoch-Jedrzejewska A, Sadowski B. Differential role of specific cardiovascular neuropeptides in pain regulation: Relevance to cardiovascular diseases. Neuropeptides 2020; 81:102046. [PMID: 32284215 DOI: 10.1016/j.npep.2020.102046] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 03/27/2020] [Accepted: 03/29/2020] [Indexed: 12/11/2022]
Abstract
In many instances, the perception of pain is disproportionate to the strength of the algesic stimulus. Excessive or inadequate pain sensation is frequently observed in cardiovascular diseases, especially in coronary ischemia. The mechanisms responsible for individual differences in the perception of cardiovascular pain are not well recognized. Cardiovascular disorders may provoke pain in multiple ways engaging molecules released locally in the heart due to tissue ischemia, inflammation or cellular stress, and through neurogenic and endocrine mechanisms brought into action by hemodynamic disturbances. Cardiovascular neuropeptides, namely angiotensin II (Ang II), angiotensin-(1-7) [Ang-(1-7)], vasopressin, oxytocin, and orexins belong to this group. Although participation of these peptides in the regulation of circulation and pain has been firmly established, their mutual interaction in the regulation of pain in cardiovascular diseases has not been profoundly analyzed. In the present review we discuss the regulation of the release, and mechanisms of the central and systemic actions of these peptides on the cardiovascular system in the context of their central and peripheral nociceptive (Ang II) and antinociceptive [Ang-(1-7), vasopressin, oxytocin, orexins] properties. We also consider the possibility that they may play a significant role in the modulation of pain in cardiovascular diseases. The rationale for focusing attention on these very compounds was based on the following premises (1) cardiovascular disturbances influence the release of these peptides (2) they regulate vascular tone and cardiac function and can influence the intensity of ischemia - the factor initiating pain signals in the cardiovascular system, (3) they differentially modulate nociception through peripheral and central mechanisms, and their effect strongly depends on specific receptors and site of action. Accordingly, an altered release of these peptides and/or pharmacological blockade of their receptors may have a significant but different impact on individual sensation of pain and comfort of an individual patient.
Collapse
Affiliation(s)
- Ewa Szczepanska-Sadowska
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1b, Warsaw, Poland.
| | - Agnieszka Cudnoch-Jedrzejewska
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1b, Warsaw, Poland
| | - Bogdan Sadowski
- School of Engineering and Health, Bitwy Warszawskiej 1920 r. 18, Warsaw, Poland
| |
Collapse
|
13
|
Bolay H, Gül A, Baykan B. COVID-19 is a Real Headache! Headache 2020; 60:1415-1421. [PMID: 32412101 PMCID: PMC7272895 DOI: 10.1111/head.13856] [Citation(s) in RCA: 154] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 05/01/2020] [Accepted: 05/02/2020] [Indexed: 01/08/2023]
Abstract
After the emergence of a novel coronavirus named SARS‐CoV‐2, coronavirus disease 2019 (COVID‐19) was initially characterized by fever, sore throat, cough, and dyspnea, mainly manifestations of respiratory system. However, other manifestations such as headache, abdominal pain, diarrhea, loss of taste and smell were added to the clinical spectrum, during the course of the COVID‐19 pandemic. The reports on the neurological findings are increasing rapidly and headache seems to be the leader on the symptom list. Headache was reported in 11%‐34% of the hospitalized COVID‐19 patients, but clinical features of these headaches were totally missing in available publications. According to our initial experience, significant features of headache presentation in the symptomatic COVID‐19 patients were new‐onset, moderate‐severe, bilateral headache with pulsating or pressing quality in the temporoparietal, forehead or periorbital region. The most striking features of the headache were sudden to gradual onset and poor response to common analgesics, or high relapse rate, that was limited to the active phase of the COVID‐19. Symptomatic COVID‐19 patients, around 6%‐10%, also reported headache as a presenting symptom. The possible pathophysiological mechanisms of headache include activation of peripheral trigeminal nerve endings by the SARS‐CoV‐2 directly or through the vasculopathy and/or increased circulating pro‐inflammatory cytokines and hypoxia. We concluded that as a common non‐respiratory symptom of COVID‐19, headache should not be overlooked, and its characteristics should be recorded with scrutiny.
Collapse
Affiliation(s)
- Hayrunnisa Bolay
- Department of Neurology and Algology, Gazi University, Ankara, Turkey
| | - Ahmet Gül
- Istanbul Faculty of Medicine, Department of Rheumatology, Istanbul University, Istanbul, Turkey
| | - Betül Baykan
- Istanbul Faculty of Medicine, Department of Neurology, Headache Center, Istanbul University, Istanbul, Turkey
| |
Collapse
|
14
|
Cross-talk signaling in the trigeminal ganglion: role of neuropeptides and other mediators. J Neural Transm (Vienna) 2020; 127:431-444. [PMID: 32088764 PMCID: PMC7148261 DOI: 10.1007/s00702-020-02161-7] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 02/12/2020] [Indexed: 11/08/2022]
Abstract
The trigeminal ganglion with its three trigeminal nerve tracts consists mainly of clusters of sensory neurons with their peripheral and central processes. Most neurons are surrounded by satellite glial cells and the axons are wrapped by myelinating and non-myelinating Schwann cells. Trigeminal neurons express various neuropeptides, most notably, calcitonin gene-related peptide (CGRP), substance P, and pituitary adenylate cyclase-activating polypeptide (PACAP). Two types of CGRP receptors are expressed in neurons and satellite glia. A variety of other signal molecules like ATP, nitric oxide, cytokines, and neurotrophic factors are released from trigeminal ganglion neurons and signal to neighboring neurons or satellite glial cells, which can signal back to neurons with same or other mediators. This potential cross-talk of signals involves intracellular mechanisms, including gene expression, that can modulate mediators of sensory information, such as neuropeptides, receptors, and neurotrophic factors. From the ganglia cell bodies, which are outside the blood–brain barrier, the mediators are further distributed to peripheral sites and/or to the spinal trigeminal nucleus in the brainstem, where they can affect neural transmission. A major question is how the sensory neurons in the trigeminal ganglion differ from those in the dorsal root ganglion. Despite their functional overlap, there are distinct differences in their ontogeny, gene expression, signaling pathways, and responses to anti-migraine drugs. Consequently, drugs that modulate cross-talk in the trigeminal ganglion can modulate both peripheral and central sensitization, which may potentially be distinct from sensitization mediated in the dorsal root ganglion.
Collapse
|
15
|
Bardsley EN, Neely OC, Paterson DJ. Angiotensin peptide synthesis and cyclic nucleotide modulation in sympathetic stellate ganglia. J Mol Cell Cardiol 2020; 138:234-243. [PMID: 31836539 PMCID: PMC7049903 DOI: 10.1016/j.yjmcc.2019.11.157] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/25/2019] [Accepted: 11/26/2019] [Indexed: 12/12/2022]
Abstract
Chronically elevated angiotensin II is a widely-established contributor to hypertension and heart failure via its action on the kidneys and vasculature. It also augments the activity of peripheral sympathetic nerves through activation of presynaptic angiotensin II receptors, thus contributing to sympathetic over-activity. Although some cells can synthesise angiotensin II locally, it is not known if this machinery is present in neurons closely coupled to the heart. Using a combination of RNA sequencing and quantitative real-time polymerase chain reaction, we demonstrate evidence for a renin-angiotensin synthesis pathway within human and rat sympathetic stellate ganglia, where significant alterations were observed in the spontaneously hypertensive rat stellate ganglia compared with Wistar stellates. We also used Förster Resonance Energy Transfer to demonstrate that administration of angiotensin II and angiotensin 1-7 peptides significantly elevate cyclic guanosine monophosphate in the rat stellate ganglia. Whether the release of angiotensin peptides from the sympathetic stellate ganglia alters neurotransmission and/or exacerbates cardiac dysfunction in states associated with sympathetic over activity remains to be established.
Collapse
Affiliation(s)
- Emma N Bardsley
- Wellcome Trust OXION Initiative in Ion Channels and Disease, Oxford, UK; Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK; British Heart Foundation, Centre of Research Excellence, UK.
| | - Oliver C Neely
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK; British Heart Foundation, Centre of Research Excellence, UK
| | - David J Paterson
- Wellcome Trust OXION Initiative in Ion Channels and Disease, Oxford, UK; Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK; British Heart Foundation, Centre of Research Excellence, UK.
| |
Collapse
|
16
|
Messlinger K, Russo AF. Current understanding of trigeminal ganglion structure and function in headache. Cephalalgia 2018; 39:1661-1674. [PMID: 29989427 DOI: 10.1177/0333102418786261] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION The trigeminal ganglion is unique among the somatosensory ganglia regarding its topography, structure, composition and possibly some functional properties of its cellular components. Being mainly responsible for the sensory innervation of the anterior regions of the head, it is a major target for headache research. One intriguing question is if the trigeminal ganglion is merely a transition site for sensory information from the periphery to the central nervous system, or if intracellular modulatory mechanisms and intercellular signaling are capable of controlling sensory information relevant for the pathophysiology of headaches. METHODS An online search based on PubMed was made using the keyword "trigeminal ganglion" in combination with "anatomy", "headache", "migraine", "neuropeptides", "receptors" and "signaling". From the relevant literature, further references were selected in view of their relevance for headache mechanisms. The essential information was organized based on location and cell types of the trigeminal ganglion, neuropeptides, receptors for signaling molecules, signaling mechanisms, and their possible relevance for headache generation. RESULTS The trigeminal ganglion consists of clusters of sensory neurons and their peripheral and central axon processes, which are arranged according to the three trigeminal partitions V1-V3. The neurons are surrounded by satellite glial cells, the axons by Schwann cells. In addition, macrophage-like cells can be found in the trigeminal ganglion. Neurons express various neuropeptides, among which calcitonin gene-related peptide is the most prominent in terms of its prevalence and its role in primary headaches. The classical calcitonin gene-related peptide receptors are expressed in non-calcitonin gene-related peptide neurons and satellite glial cells, although the possibility of a second calcitonin gene-related peptide receptor in calcitonin gene-related peptide neurons remains to be investigated. A variety of other signal molecules like adenosine triphosphate, nitric oxide, cytokines, and neurotrophic factors are released from trigeminal ganglion cells and may act at receptors on adjacent neurons or satellite glial cells. CONCLUSIONS The trigeminal ganglion may act as an integrative organ. The morphological and functional arrangement of trigeminal ganglion cells suggests that intercellular and possibly also autocrine signaling mechanisms interact with intracellular mechanisms, including gene expression, to modulate sensory information. Receptors and neurotrophic factors delivered to the periphery or the trigeminal brainstem can contribute to peripheral and central sensitization, as in the case of primary headaches. The trigeminal ganglion as a target of drug action outside the blood-brain barrier should therefore be taken into account.
Collapse
Affiliation(s)
- Karl Messlinger
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Andrew F Russo
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, USA.,Iowa VA Health Care System, Iowa City, IA, USA
| |
Collapse
|
17
|
Bohlender JM, Nussberger J, Tevaearai H, Imboden H. Angiotensinergic Innervation of the Human Right Atrium: Implications for Cardiac Reflexes. Am J Hypertens 2018; 31:188-196. [PMID: 28985343 PMCID: PMC5861579 DOI: 10.1093/ajh/hpx163] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Accepted: 09/03/2017] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The right atrium is densely innervated and provides sensory input to important cardiocirculatory reflexes controlling cardiac output and blood pressure. Its angiotensin (Ang) II-expressing innervation may release Ang II as a neuropeptide cotransmitter to modulate reflexes but has not yet been characterized. METHODS Intraoperative surgical biopsies from human right atria (n = 7) were immunocytologically stained for Ang II, tyrosine hydroxylase (TH), and synaptophysin (SYN). Tissue angiotensins were extracted and quantified by radioimmunoassay. RESULTS Angiotensinergic fibers were frequent in epicardial nerves and around vessels with variable TH co-localization (none to >50%/bundle). Fibers were also widely distributed between cardiomyocytes and in the endocardium where they were typically nonvaricose, TH/SYN-negative and usually accompanied by varicose catecholaminergic fibers. In the endocardium, some showed large varicosities and were partially TH or SYN-positive. A few endocardial regions showed scattered nonvaricose Ang fibers ending directly between endothelial cells. Occasional clusters of thin varicose terminals co-localizing SYN or TH were located underneath, or protruded into, the endothelium. Endocardial density of Ang and TH-positive fibers was 30-300 vs. 200-450/mm2. Atrial Ang II, III, and I concentrations were 67, 16, and 5 fmol/g (median) while Ang IV and V were mostly undetectable. CONCLUSIONS The human right atrium harbors an abundant angiotensinergic innervation and a novel potential source of atrial Ang II. Most peripheral fibers were noncatecholaminergic afferents or preterminal vagal efferents and a minority was presumably sympathetic. Neuronal Ang II release from these fibers may modulate cardiac and circulatory reflexes independently from plasma and tissue Ang II sources.
Collapse
Affiliation(s)
- Jürgen M Bohlender
- Division of Clinical Pharmacology, Department of General Internal Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Jürg Nussberger
- Department of Medicine, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Hendrik Tevaearai
- Department of Cardiovascular Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Hans Imboden
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| |
Collapse
|
18
|
Bohlender JM, Nussberger J, Birkhäuser F, Grouzmann E, Thalmann GN, Imboden H. Resetting of renal tissular renin-angiotensin and bradykinin-kallikrein systems after unilateral kidney denervation in rats. Histochem Cell Biol 2017; 147:585-593. [PMID: 28220244 DOI: 10.1007/s00418-017-1543-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2017] [Indexed: 01/11/2023]
Abstract
The renal tissular renin-angiotensin and bradykinin-kallikrein systems control kidney function together with the renal sympathetic innervation but their interaction is still unclear. To further elucidate this relationship, we investigated these systems in rats 6 days after left kidney denervation (DNX, n = 8) compared to sham-operated controls (CTR, n = 8). Plasma renin concentration was unchanged in DNX vs. CTR (p = NS). Kidney bradykinin (BK) and angiotensin (Ang) I and II concentrations decreased bilaterally in DNX vs. CTR rats (~20 to 40%, p < 0.05) together with Ang IV and V concentrations that were extremely low (p = NS). Renin, Ang III and dopamine concentrations decreased by ~25 to 50% and norepinephrine concentrations by 99% in DNX kidneys (p < 0.05) but were unaltered in opposite kidneys. Ang II/I and KA were comparable in DNX, contralateral and CTR kidneys. Ang III/II increased in right vs. DNX or CTR kidneys (40-50%, p < 0.05). Ang II was mainly located in tubular epithelium by immunocytological staining and its cellular distribution was unaffected by DNX. Moreover, the angiotensinergic and catecholaminergic innervation of right kidneys was unchanged vs. CTR. We found an important dependency of tissular Ang and BK levels on the renal innervation that may contribute to the resetting of kidney function after DNX. The DNX-induced peptide changes were not readily explained by kidney KA, renin or plasma Ang I generation. However, tissular peptide metabolism and compartmentalization may have played a central role. The mechanisms behind the concentration changes remain unclear and deserve further clarification.
Collapse
Affiliation(s)
- Jürgen M Bohlender
- Institute for Cell Biology, University of Bern, Bern, Switzerland. .,Department of Nephrology, Hypertension and Clinical Pharmacology, Inselspital, Bern University Hospital, University of Bern, Freiburgstr. 4, 3010, Bern, Switzerland.
| | - Jürg Nussberger
- Department of Medicine, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Frédéric Birkhäuser
- Department of Urology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Eric Grouzmann
- Service of Biomedicine, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - George N Thalmann
- Department of Urology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Hans Imboden
- Institute for Cell Biology, University of Bern, Bern, Switzerland
| |
Collapse
|
19
|
Selective small molecule angiotensin II type 2 receptor antagonists for neuropathic pain: preclinical and clinical studies. Pain 2016; 157 Suppl 1:S33-S41. [PMID: 26785154 DOI: 10.1097/j.pain.0000000000000369] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Neuropathic pain affects up to 10% of the general population, but drug treatments recommended for the treatment of neuropathic pain are associated with modest efficacy and/or produce dose-limiting side effects. Hence, neuropathic pain is an unmet medical need. In the past 2 decades, research on the pathobiology of neuropathic pain has revealed many novel pain targets for use in analgesic drug discovery programs. However, these efforts have been largely unsuccessful as molecules that showed promising pain relief in rodent models of neuropathic pain generally failed to produce analgesia in early phase clinical trials in patients with neuropathic pain. One notable exception is the angiotensin II type 2 (AT2) receptor that has clinical validity on the basis of a successful double-blind, randomized, placebo-controlled, clinical trial of EMA401, a highly selective, orally active, peripherally restricted AT2 receptor antagonist in patients with postherpetic neuralgia. In this study, we review research to date on target validation, efficacy, and mode of action of small molecule AT2 receptor antagonists in rodent models of peripheral neuropathic pain and in cultured human sensory neurons, the preclinical pharmacokinetics of these compounds, and the outcome of the above clinical trial.
Collapse
|
20
|
Effect of angiotensin II on voltage-gated sodium currents in aortic baroreceptor neurons and arterial baroreflex sensitivity in heart failure rats. J Hypertens 2016; 33:1401-10. [PMID: 25827427 DOI: 10.1097/hjh.0000000000000563] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Impairment of arterial baroreflex sensitivity is associated with mortality in patients with chronic heart failure (CHF). Elevation of plasma angiotension II (Ang II) contributes to arterial baroreflex dysfunction in CHF. A reduced number of voltage-gated sodium (Nav) channels in aortic baroreceptor neurons are involved in CHF-blunted arterial baroreflex. METHOD In this study, we investigated acute effect of Ang II on Nav currents in the aortic baroreceptor neuron and on arterial baroreflex in sham and coronary artery ligation-induced CHF rats. RESULTS Using Ang II I radioimmunoassay, real-time reverse transcription-PCR and western blot, we found that Ang II levels, and mRNA and protein expression of angiotension II type 1 receptor in nodose ganglia from CHF rats were higher than that from sham rats. Local microinjection of Ang II (0.2 nmol) into the nodose ganglia decreased the arterial baroreflex sensitivity in sham rats, whereas losartan (1 nmol, an angiotension II type 1 receptor antagonist) improved the arterial baroreflex sensitivity in CHF rats. Data from patch-clamp recording showed that Ang II (100 nmol/l) acutely inhibited Nav currents in the aortic baroreceptor neurons from sham and CHF rats. In particular, inhibitory effect of Ang II on Nav currents in the aortic baroreceptor neurons was larger in CHF rats than that in sham rats. Losartan (1 μmol/l) totally abolished the inhibitory effect of Ang II on Nav currents in sham and CHF aortic baroreceptor neurons. CONCLUSION These results suggest that elevation of endogenous Ang II in the nodose ganglia contributes to impairment of the arterial baroreflex function in CHF rats through inhibiting Nav channels.
Collapse
|
21
|
Anand U, Yiangou Y, Sinisi M, Fox M, MacQuillan A, Quick T, Korchev YE, Bountra C, McCarthy T, Anand P. Mechanisms underlying clinical efficacy of Angiotensin II type 2 receptor (AT2R) antagonist EMA401 in neuropathic pain: clinical tissue and in vitro studies. Mol Pain 2015; 11:38. [PMID: 26111701 PMCID: PMC4482278 DOI: 10.1186/s12990-015-0038-x] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 06/11/2015] [Indexed: 12/20/2022] Open
Abstract
Background The clinical efficacy of the Angiotensin II (AngII) receptor AT2R antagonist EMA401, a novel peripherally-restricted analgesic, was reported recently in post-herpetic neuralgia. While previous studies have shown that AT2R is expressed by nociceptors in human DRG (hDRG), and that EMA401 inhibits capsaicin responses in cultured hDRG neurons, the expression and levels of its endogenous ligands AngII and AngIII in clinical neuropathic pain tissues, and their signalling pathways, require investigation. We have immunostained AngII, AT2R and the capsaicin receptor TRPV1 in control post-mortem and avulsion injured hDRG, control and injured human nerves, and in cultured hDRG neurons. AngII, AngIII, and Ang-(1-7) levels were quantified by ELISA. The in vitro effects of AngII, AT2R agonist C21, and Nerve growth factor (NGF) were measured on neurite lengths; AngII, NGF and EMA401 effects on expression of p38 and p42/44 MAPK were measured using quantitative immunofluorescence, and on capsaicin responses using calcium imaging. Results AngII immunostaining was observed in approximately 75% of small/medium diameter neurons in control (n = 5) and avulsion injured (n = 8) hDRG, but not large neurons i.e. similar to TRPV1. AngII was co-localised with AT2R and TRPV1 in hDRG and in vitro. AngII staining by image analysis showed no significant difference between control (n = 12) and injured (n = 13) human nerves. AngII levels by ELISA were also similar in control human nerves (4.09 ± 0.36 pmol/g, n = 31), injured nerves (3.99 ± 0.79 pmol/g, n = 7), and painful neuromas (3.43 ± 0.73 pmol/g, n = 12); AngIII and Ang-(1-7) levels were undetectable (<0.03 and 0.05 pmol/g respectively). Neurite lengths were significantly increased in the presence of NGF, AngII and C21 in cultured DRG neurons. AngII and, as expected, NGF significantly increased signal intensity of p38 and p42/44 MAPK, which was reversed by EMA401. AngII mediated sensitization of capsaicin responses was not observed in the presence of MAP kinase inhibitor PD98059, and the kinase inhibitor staurosporine. Conclusion The major AT2R ligand in human peripheral nerves is AngII, and its levels are maintained in injured nerves. EMA401 may act on paracrine/autocrine mechanisms at peripheral nerve terminals, or intracrine mechanisms, to reduce neuropathic pain signalling in AngII/NGF/TRPV1-convergent pathways.
Collapse
Affiliation(s)
- Uma Anand
- Peripheral Neuropathy Unit, Centre for Clinical Translation, Hammersmith Hospital, Imperial College London, Area A, Ground Floor, Du Cane Rd, London, W12 ONN, UK. .,Nanomedicine Research Laboratory, Division of Medicine, Hammersmith Hospital, Imperial College London, BN5 Commonwealth Building, London, W12 0NN, UK.
| | - Yiangos Yiangou
- Peripheral Neuropathy Unit, Centre for Clinical Translation, Hammersmith Hospital, Imperial College London, Area A, Ground Floor, Du Cane Rd, London, W12 ONN, UK.
| | - Marco Sinisi
- Peripheral Neuropathy Unit, Centre for Clinical Translation, Hammersmith Hospital, Imperial College London, Area A, Ground Floor, Du Cane Rd, London, W12 ONN, UK. .,Peripheral Nerve Injury Unit, Royal National Orthopaedic Hospital, Stanmore, Middlesex, HA7 4LP, UK.
| | - Michael Fox
- Peripheral Neuropathy Unit, Centre for Clinical Translation, Hammersmith Hospital, Imperial College London, Area A, Ground Floor, Du Cane Rd, London, W12 ONN, UK. .,Peripheral Nerve Injury Unit, Royal National Orthopaedic Hospital, Stanmore, Middlesex, HA7 4LP, UK.
| | - Anthony MacQuillan
- Peripheral Neuropathy Unit, Centre for Clinical Translation, Hammersmith Hospital, Imperial College London, Area A, Ground Floor, Du Cane Rd, London, W12 ONN, UK. .,Peripheral Nerve Injury Unit, Royal National Orthopaedic Hospital, Stanmore, Middlesex, HA7 4LP, UK.
| | - Tom Quick
- Peripheral Neuropathy Unit, Centre for Clinical Translation, Hammersmith Hospital, Imperial College London, Area A, Ground Floor, Du Cane Rd, London, W12 ONN, UK. .,Peripheral Nerve Injury Unit, Royal National Orthopaedic Hospital, Stanmore, Middlesex, HA7 4LP, UK.
| | - Yuri E Korchev
- Nanomedicine Research Laboratory, Division of Medicine, Hammersmith Hospital, Imperial College London, BN5 Commonwealth Building, London, W12 0NN, UK.
| | - Chas Bountra
- University of Oxford Structural Genomics Consortium, Old Road, Campus Research Building, Roosevelt Drive, Headington, Oxford, OX3 7DQ, UK.
| | - Tom McCarthy
- Spinifex Pharmaceuticals Pty Ltd, Corporate One, Suite G5, 84 Hotham St, Preston, VIC, 3072, Australia.
| | - Praveen Anand
- Peripheral Neuropathy Unit, Centre for Clinical Translation, Hammersmith Hospital, Imperial College London, Area A, Ground Floor, Du Cane Rd, London, W12 ONN, UK.
| |
Collapse
|
22
|
Bohlender J, Nussberger J, Tevaearai H, Imboden H. [Autonomic angiotensinergic fibres in the human heart with an efferent sympathetic cophenotype]. Ann Cardiol Angeiol (Paris) 2015; 64:175-9. [PMID: 26049899 DOI: 10.1016/j.ancard.2015.04.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Accepted: 04/28/2015] [Indexed: 02/04/2023]
Abstract
AIM The autonomic innervation of the heart consists of sympathetic and parasympathetic nerve fibres, and fibres of the intrinsic ganglionated plexus with noradrenaline and acytylcholine as principal neurotransmitters. The fibres co-release neuropeptides to modulate intracardiac neurotransmission by specific presynaptic and postsynaptic receptors. The coexpression of angiotensin II in sympathetic fibres of the human heart and its role are not known so far. METHODS Autopsy specimens of human hearts were studied (n=3; ventricles). Using immunocytological methods, cryostat sections were stained by a murine monoclonal antibody (4B3) directed against angiotensin II and co-stained by polyclonal antibodies against tyrosine hydroxylase, a catecholaminergic marker. Visualisation of the antibodies was by confocal light microscopy or laser scanning microscopy. RESULTS Angiotensin II-positive autonomic fibres with and without a catecholaminergic cophenotype (hydroxylase-positive) were found in all parts of the human ventricles. In the epicardium, the fibres were grouped in larger bundles of up to 100 and more fibres. They followed the preformed anatomic septa and epicardial vessels towards the myocardium and endocardium where the bundles dissolved and the individual fibres spread between myocytes and within the endocardium. Generally, angiotensinergic fibres showed no synaptic enlargements or only a few if they were also catecholaminergic. The exclusively catechalominergic fibres were characterised by multiple beaded synapses. CONCLUSION The autonomic innervation of the human heart contains angiotensinergic fibres with a sympathetic efferent phenotype and exclusively angiotensinergic fibers representing probably afferents. Angiotensinergic neurotransmission may modulate intracardiac sympathetic and parasympathetic activity and thereby influence cardiac and circulatory function.
Collapse
Affiliation(s)
- J Bohlender
- Département de néphrologie, hypertension et pharmacologie clinique, Inselspital, hôpital universitaire, université de Berne, 10, Freiburgstrasse, 3010 Berne, Suisse; Institut de biologie cellulaire, université de Berne, 2, Baltzerstrasse, 3012 Berne, Suisse.
| | - J Nussberger
- Département de médecine, centre hospitalier universitaire vaudois (CHUV), 5, avenue Pierre-Decker, 1011 Lausanne, Suisse
| | - H Tevaearai
- Départment de chirurgie cardiovasculaire, Inselspital, hôpital universitaire, université de Berne, 10, Freiburgstrasse, 3010 Berne, Suisse
| | - H Imboden
- Institut de biologie cellulaire, université de Berne, 2, Baltzerstrasse, 3012 Berne, Suisse
| |
Collapse
|
23
|
Abstract
The renin-angiotensin system (RAS) is a major regulatory system controlling many different homeostatic mechanisms both within the brain and in the periphery. While it is primarily associated with blood pressure and salt/water regulation, increasing evidence points to the involvement of the RAS in both headache disorders specifically and pain regulation in general. Several publications have indicated that drugs blocking various elements of the renin-angiotensin system lead to a reduction in migraine. Additionally, interventions on different angiotensin peptides or their receptors have been shown to both reduce and increase pain in animal models. As such, modulation of the renin-angiotensin system is a promising approach to the treatment of headaches and other pain conditions.
Collapse
|
24
|
Ripa P, Ornello R, Pistoia F, Carolei A, Sacco S. The renin-angiotensin system: a possible contributor to migraine pathogenesis and prophylaxis. Expert Rev Neurother 2014; 14:1043-55. [PMID: 25115162 DOI: 10.1586/14737175.2014.946408] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The presence of a tissue-based renin-angiotensin system, independent of the systemic one, has been identified in several organs including the brain. Experimental models have suggested the involvement of the renin-angiotensin system in neurogenic inflammation, susceptibility to oxidative stress, endothelial dysfunction, and neuromodulation of nociceptive transmission, thus potentially contributing to the pathogenesis of migraine. Genetic factors that increase susceptibility to migraine may include angiotensin-converting enzyme polymorphism, although available data are controversial. Clinical studies have suggested that angiotensin-converting enzyme inhibitors and angiotensin receptor blockers may be effective in migraine prophylaxis. However, further research should clarify whether the postulated preventive effect is attributable to a pharmacological action over and above the antihypertensive effect and should test their tolerability in subjects with normal blood pressure values. In patients with contraindications or not responding to conventional prophylactic drugs and in patients with comorbid arterial hypertension, angiotensin-converting enzyme inhibitors and angiotensin receptor blockers may be used for migraine prophylaxis.
Collapse
Affiliation(s)
- Patrizia Ripa
- Department of Biotechnological and Applied Clinical Sciences, Division of Clinical Neurology, University of L'Aquila, via Vetoio, 67100 L'Aquila, Italy
| | | | | | | | | |
Collapse
|
25
|
Anand U, Facer P, Yiangou Y, Sinisi M, Fox M, McCarthy T, Bountra C, Korchev YE, Anand P. Angiotensin II type 2 receptor (AT2 R) localization and antagonist-mediated inhibition of capsaicin responses and neurite outgrowth in human and rat sensory neurons. Eur J Pain 2013; 17:1012-26. [PMID: 23255326 PMCID: PMC3748799 DOI: 10.1002/j.1532-2149.2012.00269.x] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/09/2012] [Indexed: 01/05/2023]
Abstract
BACKGROUND The angiotensin II (AngII) receptor subtype 2 (AT2 R) is expressed in sensory neurons and may play a role in nociception and neuronal regeneration. METHODS We used immunostaining with characterized antibodies to study the localization of AT2 R in cultured human and rat dorsal root ganglion (DRG) neurons and a range of human tissues. The effects of AngII and AT2 R antagonist EMA401 on capsaicin responses in cultured human and rat (DRG) neurons were measured with calcium imaging, on neurite length and density with Gap43 immunostaining, and on cyclic adenosine monophosphate (cAMP) expression using immunofluorescence. RESULTS AT2 R expression was localized in small-/medium-sized cultured neurons of human and rat DRG. Treatment with the AT2 R antagonist EMA401 resulted in dose-related functional inhibition of capsaicin responses (IC50 = 10 nmol/L), which was reversed by 8-bromo-cAMP, and reduced neurite length and density; AngII treatment significantly enhanced capsaicin responses, cAMP levels and neurite outgrowth. The AT1 R antagonist losartan had no effect on capsaicin responses. AT2 R was localized in sensory neurons of human DRG, and nerve fibres in peripheral nerves, skin, urinary bladder and bowel. A majority sub-population (60%) of small-/medium-diameter neuronal cells were immunopositive in both control post-mortem and avulsion-injured human DRG; some very small neurons appeared to be intensely immunoreactive, with TRPV1 co-localization. While AT2 R levels were reduced in human limb peripheral nerve segments proximal to injury, they were preserved in painful neuromas. CONCLUSIONS AT2 R antagonists could be particularly useful in the treatment of chronic pain and hypersensitivity associated with abnormal nerve sprouting.
Collapse
Affiliation(s)
- U Anand
- Peripheral Neuropathy Unit, Department of Clinical Neuroscience, Imperial College London, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Chakrabarty A, Liao Z, Smith PG. Angiotensin II receptor type 2 activation is required for cutaneous sensory hyperinnervation and hypersensitivity in a rat hind paw model of inflammatory pain. THE JOURNAL OF PAIN 2013; 14:1053-65. [PMID: 23726047 DOI: 10.1016/j.jpain.2013.04.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Revised: 03/19/2013] [Accepted: 04/02/2013] [Indexed: 11/18/2022]
Abstract
UNLABELLED Many pain syndromes are associated with abnormal proliferation of peripheral sensory fibers. We showed previously that angiotensin II, acting through its type 2 receptor (AT2), stimulates axon outgrowth by cultured dorsal root ganglion neurons. In this study, we assessed whether AT2 mediates nociceptor hyperinnervation in the rodent hind paw model of inflammatory pain. Plantar injection of complete Freund's adjuvant (CFA), but not saline, produced marked thermal and mechanical hypersensitivity through 7 days. This was accompanied by proliferation of dermal and epidermal PGP9.5-immunoreactive (ir) and calcitonin gene-related peptide-immunoreactive (CGRP-ir) axons, and dermal axons immunoreactive for GFRα2 but not tyrosine hydroxylase or neurofilament H. Continuous infusion of the AT2 antagonist PD123319 beginning with CFA injection completely prevented hyperinnervation as well as hypersensitivity over a 7-day period. A single PD123319 injection 7 days after CFA also reversed thermal hypersensitivity and partially reversed mechanical hypersensitivity 3 hours later, without affecting cutaneous innervation. Angiotensin II-synthesizing proteins renin and angiotensinogen were largely absent after saline but abundant in T cells and macrophages in CFA-injected paws with or without PD123319. Thus, emigrant cells at the site of inflammation apparently establish a renin-angiotensin system, and AT2 activation elicits nociceptor sprouting and heightened thermal and mechanical sensitivity. PERSPECTIVE Short-term AT2 activation is a potent contributor to thermal hypersensitivity, whereas long-term effects (such as hyperinnervation) also contribute to mechanical hypersensitivity. Pharmacologic blockade of AT2 signaling represents a potential therapeutic strategy aimed at biologic mechanisms underlying chronic inflammatory pain.
Collapse
Affiliation(s)
- Anuradha Chakrabarty
- Institute for Neurological Discoveries, University of Kansas Medical Center, Kansas City, Kansas; Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas; Kansas Intellectual and Developmental Disabilities Research Center, University of Kansas Medical Center, Kansas City, Kansas
| | | | | |
Collapse
|
27
|
Bohlender J, Nussberger J, Imboden H. Angiotensinergic innervation of the kidney: present knowledge and its significance. Curr Hypertens Rep 2012. [PMID: 23197298 DOI: 10.1007/s11906-012-0322-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Intrarenal neurotransmission implies the co-release of neuropeptides at the neuro-effector junction with direct influence on parameters of kidney function. The presence of an angiotensin (Ang) II-containing phenotype in catecholaminergic postganglionic and sensory fibers of the kidney, based on immunocytological investigations, has only recently been reported. These angiotensinergic fibers display a distinct morphology and intrarenal distribution, suggesting anatomical and functional subspecialization linked to neuronal Ang II-expression. This review discusses the present knowledge concerning these fibers, and their significance for renal physiology and the pathogenesis of hypertension in light of established mechanisms. The data suggest a new role of Ang II as a co-transmitter stimulating renal target cells or modulating nerve traffic from or to the kidney. Neuronal Ang II is likely to be an independent source of intrarenal Ang II. Further physiological experimentation will have to explore the role of the angiotensinergic renal innervation and integrate it into existing concepts.
Collapse
Affiliation(s)
- Jürgen Bohlender
- Hôpital fribourgeois/CHUV and Institute of Cell Biology, University of Bern, Case postale, Fribourg, Switzerland.
| | | | | |
Collapse
|
28
|
Patil J, Stucki S, Nussberger J, Schaffner T, Gygax S, Bohlender J, Imboden H. Angiotensinergic and noradrenergic neurons in the rat and human heart. ACTA ACUST UNITED AC 2011; 167:31-41. [DOI: 10.1016/j.regpep.2010.11.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2010] [Revised: 10/28/2010] [Accepted: 11/25/2010] [Indexed: 11/29/2022]
|
29
|
Saavedra JM, Sánchez-Lemus E, Benicky J. Blockade of brain angiotensin II AT1 receptors ameliorates stress, anxiety, brain inflammation and ischemia: Therapeutic implications. Psychoneuroendocrinology 2011; 36:1-18. [PMID: 21035950 PMCID: PMC2998923 DOI: 10.1016/j.psyneuen.2010.10.001] [Citation(s) in RCA: 190] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2010] [Revised: 09/29/2010] [Accepted: 10/01/2010] [Indexed: 12/22/2022]
Abstract
Poor adaptation to stress, alterations in cerebrovascular function and excessive brain inflammation play critical roles in the pathophysiology of many psychiatric and neurological disorders such as major depression, schizophrenia, post traumatic stress disorder, Parkinson's and Alzheimer's diseases and traumatic brain injury. Treatment for these highly prevalent and devastating conditions is at present very limited and many times inefficient, and the search for novel therapeutic options is of major importance. Recently, attention has been focused on the role of a brain regulatory peptide, Angiotensin II, and in the translational value of the blockade of its physiological AT(1) receptors. In addition to its well-known cardiovascular effects, Angiotensin II, through AT(1) receptor stimulation, is a pleiotropic brain modulatory factor involved in the control of the reaction to stress, in the regulation of cerebrovascular flow and the response to inflammation. Excessive brain AT(1) receptor activity is associated with exaggerated sympathetic and hormonal response to stress, vulnerability to cerebrovascular ischemia and brain inflammation, processes leading to neuronal injury. In animal models, inhibition of brain AT(1) receptor activity with systemically administered Angiotensin II receptor blockers is neuroprotective; it reduces exaggerated stress responses and anxiety, prevents stress-induced gastric ulcerations, decreases vulnerability to ischemia and stroke, reverses chronic cerebrovascular inflammation, and reduces acute inflammatory responses produced by bacterial endotoxin. These effects protect neurons from injury and contribute to increase the lifespan. Angiotensin II receptor blockers are compounds with a good margin of safety widely used in the treatment of hypertension and their anti-inflammatory and vascular protective effects contribute to reduce renal and cardiovascular failure. Inhibition of brain AT(1) receptors in humans is also neuroprotective, reducing the incidence of stroke, improving cognition and decreasing the progression of Alzheimer's disease. Blockade of AT(1) receptors offers a novel and safe therapeutic approach for the treatment of illnesses of increasing prevalence and socioeconomic impact, such as mood disorders and neurodegenerative diseases of the brain.
Collapse
Affiliation(s)
- Juan M Saavedra
- Section on Pharmacology, Division of Intramural Research Programs, National Institute of Mental Health, National Institutes of Health, Department of Health and Human Services, 10 Center Drive, Building 10, Room 2D-57, Bethesda, MD 20892, USA.
| | | | | |
Collapse
|
30
|
Frenkel L, Dimant B, Portiansky EL, Imboden H, Maldonado H, Delorenzi A. Neuroanatomical distribution of angiotensin-II-like neuropeptide within the central nervous system of the crab Chasmagnathus; physiological changes triggered by water deprivation. Cell Tissue Res 2010; 341:181-95. [DOI: 10.1007/s00441-010-0990-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2009] [Accepted: 05/03/2010] [Indexed: 10/19/2022]
|
31
|
Patil J, Schwab A, Nussberger J, Schaffner T, Saavedra JM, Imboden H. Intraneuronal angiotensinergic system in rat and human dorsal root ganglia. ACTA ACUST UNITED AC 2010; 162:90-8. [PMID: 20346377 DOI: 10.1016/j.regpep.2010.03.004] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2009] [Revised: 01/29/2010] [Accepted: 03/16/2010] [Indexed: 11/30/2022]
Abstract
To elucidate the local formation of angiotensin II (Ang II) in the neurons of sensory dorsal root ganglia (DRG), we studied the expression of angiotensinogen (Ang-N)-, renin-, angiotensin converting enzyme (ACE)- and cathepsin D-mRNA, and the presence of protein renin, Ang II, Substance P and calcitonin gene-related peptide (CGRP) in the rat and human thoracic DRG. Quantitative real time PCR (qRT-PCR) studies revealed that rat DRG expressed substantial amounts of Ang-N- and ACE mRNA, while renin mRNA as well as the protein renin were untraceable. Cathepsin D-mRNA and cathepsin D-protein were detected in the rat DRG indicating the possibility of existence of pathways alternative to renin for Ang I formation. Angiotensin peptides were successfully detected with high performance liquid chromatography and radioimmunoassay in human DRG extracts. In situ hybridization in rat DRG confirmed additionally expression of Ang-N mRNA in the cytoplasm of numerous neurons. Intracellular Ang II staining could be shown in number of neurons and their processes in both the rat and human DRG. Interestingly we observed neuronal processes with angiotensinergic synapses en passant, colocalized with synaptophysin, within the DRG. In the DRG, we also identified by qRT-PCR, expression of Ang II receptor AT(1A) and AT(2)-mRNA while AT(1B)-mRNA was not traceable. In some neurons Substance P and CGRP were found colocalized with Ang II. The intracellular localization and colocalization of Ang II with Substance P and CGRP in the DRG neurons may indicate a participation and function of Ang II in the regulation of nociception. In conclusion, these results suggest that Ang II may be produced locally in the neurons of rat and human DRG and act as a neurotransmitter.
Collapse
Affiliation(s)
- Jaspal Patil
- Institute of Cell Biology, University of Bern, Baltzerstrasse 4, 3012 Bern, Switzerland
| | | | | | | | | | | |
Collapse
|
32
|
Abstract
After the triptans, a calcitonin gene-related peptide blocker (telcagepant) is the first acute medicine that has been developed primarily for treatment of acute migraine. Otherwise, the new drugs have been developed first for other purposes, like anticonvulsants, antihypertensives and antidepressants used for migraine prophylaxis. For acute attacks, a new way to administer a traditional drug like dihydroergotamine is under way, and documentation of efficacy in migraine has been gained for some commonly used painkillers and anti-inflammatory drugs, and for some herbal extracts. Based on insights into the basic pathophysiological mechanisms of the disorder, some drugs have been developed which seem promising in early phase II studies (NOS inhibitors and 5HT1F-receptor agonists). In the future, development and enhancements of existing medicines must be accompanied by increased efforts to develop truly new migraine drugs based on knowledge of the pathophysiology if one wishes to reduce substantially the great burden migraine poses on patients and society.
Collapse
Affiliation(s)
- Lars Jacob Stovner
- Norwegian National Headache Centre, Trondheim University Hospital, 7006 Trondheim, Norway.
| | | | | |
Collapse
|