1
|
Pedrazzi JFC, Silva-Amaral D, Issy AC, Gomes FV, Crippa JA, Guimarães FS, Del Bel E. Cannabidiol attenuates prepulse inhibition disruption by facilitating TRPV1 and 5-HT1A receptor-mediated neurotransmission. Pharmacol Biochem Behav 2024; 245:173879. [PMID: 39305939 DOI: 10.1016/j.pbb.2024.173879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/05/2024] [Accepted: 09/10/2024] [Indexed: 12/07/2024]
Abstract
Individuals with schizophrenia (SCZ) often present sensorimotor gating impairments that can be investigated by the prepulse inhibition test (PPI). PPI disruption can be mimicked experimentally with psychostimulants such as amphetamine and attenuated/reversed by antipsychotics. Cannabidiol (CBD), the main non-psychotomimetic component of the Cannabis sativa plant, produces antipsychotic-like effects in clinical and preclinical studies. CBD can interact with many pharmacological targets, but the mechanisms involved in its antipsychotic activity are unclear. Using amphetamine-induced PPI disruption in mice, we investigated the involvement of four CBD potential pharmacological targets (CB1, CB2 TRPV1, and 5-HT1A receptors) in its antipsychotic properties. CBD effects were blocked by the TRPV1 antagonist capsazepine and, to a greater extent, by the 5-HT1A receptor antagonist WAY100635. No effect was observed with the CB1 (AM251) or CB2 (AM630) receptor antagonists. These results corroborate findings showing the antipsychotic effects of CBD in the PPI model and indicate that they involve the participation of TRPV1 and 5-HT1A receptors.
Collapse
Affiliation(s)
- João F C Pedrazzi
- Department of Neurosciences and Behavioral Sciences, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil.
| | - Danyelle Silva-Amaral
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Ana C Issy
- Department of Basic and Oral Biology, School of Dentistry of Ribeirão Preto, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Felipe V Gomes
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - José A Crippa
- Department of Neurosciences and Behavioral Sciences, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Francisco S Guimarães
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Elaine Del Bel
- Department of Neurosciences and Behavioral Sciences, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil; Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil; Department of Basic and Oral Biology, School of Dentistry of Ribeirão Preto, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| |
Collapse
|
2
|
Briânis RC, Andreotti JP, Moreira FA, Iglesias LP. Interplay between endocannabinoid and endovanilloid mechanisms in fear conditioning. Acta Neuropsychiatr 2024; 36:255-264. [PMID: 37982167 DOI: 10.1017/neu.2023.54] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2023]
Abstract
OBJECTIVE The transient receptor potential cation channel, subfamily V (vanilloid), member 1 (TRPV1) mediates pain perception to thermal and chemical stimuli in peripheral neurons. The cannabinoid receptor type 1 (CB1), on the other hand, promotes analgesia in both the periphery and the brain. TRPV1 and CB1 have also been implicated in learned fear, which involves the association of a previously neutral stimulus with an aversive event. In this review, we elaborate on the interplay between CB1 receptors and TRPV1 channels in learned fear processing. METHODS We conducted a PubMed search for a narrative review on endocannabinoid and endovanilloid mechanisms on fear conditioning. RESULTS TRPV1 and CB1 receptors are activated by a common endogenous agonist, arachidonoyl ethanolamide (anandamide), Moreover, they are expressed in common neuroanatomical structures and recruit converging cellular pathways, acting in concert to modulate fear learning. However, evidence suggests that TRPV1 exerts a facilitatory role, whereas CB1 restrains fear responses. CONCLUSION TRPV1 and CB1 seem to mediate protective and aversive roles of anandamide, respectively. However, more research is needed to achieve a better understanding of how these receptors interact to modulate fear learning.
Collapse
Affiliation(s)
- Rayssa C Briânis
- Department of Pharmacology, Institute of Biological Sciences; Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Julia P Andreotti
- Department of Pharmacology, Institute of Biological Sciences; Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Fabrício A Moreira
- Department of Pharmacology, Institute of Biological Sciences; Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Lia P Iglesias
- Department of Pharmacology, Institute of Biological Sciences; Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| |
Collapse
|
3
|
Gao N, Li M, Wang W, Liu Z, Guo Y. The dual role of TRPV1 in peripheral neuropathic pain: pain switches caused by its sensitization or desensitization. Front Mol Neurosci 2024; 17:1400118. [PMID: 39315294 PMCID: PMC11417043 DOI: 10.3389/fnmol.2024.1400118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 08/12/2024] [Indexed: 09/25/2024] Open
Abstract
The transient receptor potential vanilloid 1 (TRPV1) channel plays a dual role in peripheral neuropathic pain (NeuP) by acting as a "pain switch" through its sensitization and desensitization. Hyperalgesia, commonly resulting from tissue injury or inflammation, involves the sensitization of TRPV1 channels, which modulates sensory transmission from primary afferent nociceptors to spinal dorsal horn neurons. In chemotherapy-induced peripheral neuropathy (CIPN), TRPV1 is implicated in neuropathic pain mechanisms due to its interaction with ion channels, neurotransmitter signaling, and oxidative stress. Sensitization of TRPV1 in dorsal root ganglion neurons contributes to CIPN development, and inhibition of TRPV1 channels can reduce chemotherapy-induced mechanical hypersensitivity. In diabetic peripheral neuropathy (DPN), TRPV1 is involved in pain modulation through pathways including reactive oxygen species and cytokine production. TRPV1's interaction with TRPA1 channels further influences chronic pain onset and progression. Therapeutically, capsaicin, a TRPV1 agonist, can induce analgesia through receptor desensitization, while TRPV1 antagonists and siRNA targeting TRPV1 show promise in preclinical studies. Cannabinoid modulation of TRPV1 provides another potential pathway for alleviating neuropathic pain. This review summarizes recent preclinical research on TRPV1 in association with peripheral NeuP.
Collapse
Affiliation(s)
- Ning Gao
- Department of Acupuncture and Moxibustion, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Meng Li
- Department of Gastroenterology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Weiming Wang
- Department of Acupuncture and Moxibustion, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhen Liu
- Department of Gastroenterology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yufeng Guo
- Department of Acupuncture and Moxibustion, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
4
|
Socała K, Jakubiec M, Abram M, Mlost J, Starowicz K, Kamiński RM, Ciepiela K, Andres-Mach M, Zagaja M, Metcalf CS, Zawadzki P, Wlaź P, Kamiński K. TRPV1 channel in the pathophysiology of epilepsy and its potential as a molecular target for the development of new antiseizure drug candidates. Prog Neurobiol 2024; 240:102634. [PMID: 38834133 DOI: 10.1016/j.pneurobio.2024.102634] [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: 10/25/2023] [Revised: 04/26/2024] [Accepted: 05/28/2024] [Indexed: 06/06/2024]
Abstract
Identification of transient receptor potential cation channel, subfamily V member 1 (TRPV1), also known as capsaicin receptor, in 1997 was a milestone achievement in the research on temperature sensation and pain signalling. Very soon after it became evident that TRPV1 is implicated in a wide array of physiological processes in different peripheral tissues, as well as in the central nervous system, and thereby could be involved in the pathophysiology of numerous diseases. Increasing evidence suggests that modulation of TRPV1 may also affect seizure susceptibility and epilepsy. This channel is localized in brain regions associated with seizures and epilepsy, and its overexpression was found both in animal models of seizures and in brain samples from epileptic patients. Moreover, modulation of TRPV1 on non-neuronal cells (microglia, astrocytes, and/or peripheral immune cells) may have an impact on the neuroinflammatory processes that play a role in epilepsy and epileptogenesis. In this paper, we provide a comprehensive and critical overview of currently available data on TRPV1 as a possible molecular target for epilepsy management, trying to identify research gaps and future directions. Overall, several converging lines of evidence implicate TRPV1 channel as a potentially attractive target in epilepsy research but more studies are needed to exploit the possible role of TRPV1 in seizures/epilepsy and to evaluate the value of TRPV1 ligands as candidates for new antiseizure drugs.
Collapse
Affiliation(s)
- Katarzyna Socała
- Department of Animal Physiology and Pharmacology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19, Lublin PL 20-033, Poland.
| | - Marcin Jakubiec
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Cracow PL 30-688, Poland
| | - Michał Abram
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Cracow PL 30-688, Poland
| | - Jakub Mlost
- Department of Neurochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, Cracow PL 31-343, Poland
| | - Katarzyna Starowicz
- Department of Neurochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, Cracow PL 31-343, Poland
| | - Rafał M Kamiński
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Cracow PL 30-688, Poland
| | - Katarzyna Ciepiela
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Cracow PL 30-688, Poland; Selvita S.A., Bobrzyńskiego 14, Cracow PL 30-348, Poland
| | - Marta Andres-Mach
- Department of Experimental Pharmacology, Institute of Rural Health, Jaczewskiego 2, Lublin PL 20-090, Poland
| | - Mirosław Zagaja
- Department of Experimental Pharmacology, Institute of Rural Health, Jaczewskiego 2, Lublin PL 20-090, Poland
| | - Cameron S Metcalf
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT 84112, USA
| | - Przemysław Zawadzki
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Cracow PL 30-688, Poland
| | - Piotr Wlaź
- Department of Animal Physiology and Pharmacology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19, Lublin PL 20-033, Poland
| | - Krzysztof Kamiński
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Cracow PL 30-688, Poland
| |
Collapse
|
5
|
Dos Santos Pereira M, Dias de Abreu GH, Vanderlei LCA, Raisman-Vozari R, Guimarães FS, Lu HC, Michel PP, Del Bel E. 4'-fluorocannabidiol associated with capsazepine restrains L-DOPA-induced dyskinesia in hemiparkinsonian mice: Contribution of anti-inflammatory and anti-glutamatergic mechanisms. Neuropharmacology 2024; 251:109926. [PMID: 38554815 PMCID: PMC11988267 DOI: 10.1016/j.neuropharm.2024.109926] [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: 01/10/2024] [Revised: 03/05/2024] [Accepted: 03/15/2024] [Indexed: 04/02/2024]
Abstract
We tested the efficacy of 4'-fluorocannabidiol (4'-F-CBD), a semisynthetic cannabidiol derivative, and HU-910, a cannabinoid receptor 2 (CB2) agonist in resolving l-DOPA-induced dyskinesia (LID). Specifically, we were interested in studying whether these compounds could restrain striatal inflammatory responses and rescue glutamatergic disturbances characteristic of the dyskinetic state. C57BL/6 mice were rendered hemiparkinsonian by unilateral striatal lesioning with 6-OHDA. Abnormal involuntary movements were then induced by repeated i.p. injections of l-DOPA + benserazide. After LID was installed, the effects of a 3-day treatment with 4'-F-CBD or HU-910 in combination or not with the TRPV1 antagonist capsazepine (CPZ) or CB2 agonists HU-308 and JWH015 were assessed. Immunostaining was conducted to investigate the impacts of 4'-F-CBD and HU-910 (with CPZ) on inflammation and glutamatergic synapses. Our results showed that the combination of 4'-F-CBD + CPZ, but not when administered alone, decreased LID. Neither HU-910 alone nor HU-910+CPZ were effective. The CB2 agonists HU-308 and JWH015 were also ineffective in decreasing LID. Both combination treatments efficiently reduced microglial and astrocyte activation in the dorsal striatum of dyskinetic mice. However, only 4'-F-CBD + CPZ normalized the density of glutamate vesicular transporter-1 (vGluT1) puncta colocalized with the postsynaptic density marker PSD95. These findings suggest that 4'-F-CBD + CPZ normalizes dysregulated cortico-striatal glutamatergic inputs, which could be involved in their anti-dyskinetic effects. Although it is not possible to rule out the involvement of anti-inflammatory mechanisms, the decrease in striatal neuroinflammation markers by 4'-F-CBD and HU-910 without an associated reduction in LID indicates that they are insufficient per se to prevent LID manifestations.
Collapse
Affiliation(s)
- Maurício Dos Santos Pereira
- Department of Basic and Oral Biology, FORP, Campus USP, University of São Paulo, Ribeirão Preto, Brazil; Paris Brain Institute, Inserm, CNRS, Sorbonne Université, Paris, France.
| | - Gabriel Henrique Dias de Abreu
- Department of Psychological and Brain Sciences, Program in Neuroscience, Gill Center for Bimolecular Sciences, Indiana University, Bloomington, United States.
| | | | | | | | - Hui-Chen Lu
- Department of Psychological and Brain Sciences, Program in Neuroscience, Gill Center for Bimolecular Sciences, Indiana University, Bloomington, United States.
| | | | - Elaine Del Bel
- Department of Basic and Oral Biology, FORP, Campus USP, University of São Paulo, Ribeirão Preto, Brazil.
| |
Collapse
|
6
|
Bazan HA, Bhattacharjee S, Reid MM, Jun B, Polk C, Strain M, St Pierre LA, Desai N, Daly PW, Cucinello-Ragland JA, Edwards S, Recio J, Alvarez-Builla J, Cai JJ, Bazan NG. Transcriptomic signature, bioactivity and safety of a non-hepatotoxic analgesic generating AM404 in the midbrain PAG region. Sci Rep 2024; 14:11103. [PMID: 38750093 PMCID: PMC11096368 DOI: 10.1038/s41598-024-61791-z] [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: 03/04/2024] [Accepted: 05/09/2024] [Indexed: 05/18/2024] Open
Abstract
Safe and effective pain management is a critical healthcare and societal need. The potential for acute liver injury from paracetamol (ApAP) overdose; nephrotoxicity and gastrointestinal damage from chronic non-steroidal anti-inflammatory drug (NSAID) use; and opioids' addiction are unresolved challenges. We developed SRP-001, a non-opioid and non-hepatotoxic small molecule that, unlike ApAP, does not produce the hepatotoxic metabolite N-acetyl-p-benzoquinone-imine (NAPQI) and preserves hepatic tight junction integrity at high doses. CD-1 mice exposed to SRP-001 showed no mortality, unlike a 70% mortality observed with increasing equimolar doses of ApAP within 72 h. SRP-001 and ApAP have comparable antinociceptive effects, including the complete Freund's adjuvant-induced inflammatory von Frey model. Both induce analgesia via N-arachidonoylphenolamine (AM404) formation in the midbrain periaqueductal grey (PAG) nociception region, with SRP-001 generating higher amounts of AM404 than ApAP. Single-cell transcriptomics of PAG uncovered that SRP-001 and ApAP also share modulation of pain-related gene expression and cell signaling pathways/networks, including endocannabinoid signaling, genes pertaining to mechanical nociception, and fatty acid amide hydrolase (FAAH). Both regulate the expression of key genes encoding FAAH, 2-arachidonoylglycerol (2-AG), cannabinoid receptor 1 (CNR1), CNR2, transient receptor potential vanilloid type 4 (TRPV4), and voltage-gated Ca2+ channel. Phase 1 trial (NCT05484414) (02/08/2022) demonstrates SRP-001's safety, tolerability, and favorable pharmacokinetics, including a half-life from 4.9 to 9.8 h. Given its non-hepatotoxicity and clinically validated analgesic mechanisms, SRP-001 offers a promising alternative to ApAP, NSAIDs, and opioids for safer pain treatment.
Collapse
Affiliation(s)
- Hernan A Bazan
- Section of Vascular/Endovascular Surgery, Department of Surgery, Ochsner Clinic, New Orleans, LA, 70118, USA.
| | - Surjyadipta Bhattacharjee
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, 70112, USA
| | - Madigan M Reid
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, 70112, USA
| | - Bokkyoo Jun
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, 70112, USA
| | - Connor Polk
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, 70112, USA
| | - Madeleine Strain
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, 70112, USA
| | - Linsey A St Pierre
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, 70112, USA
| | - Neehar Desai
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, 70112, USA
| | - Patrick W Daly
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, 70112, USA
| | - Jessica A Cucinello-Ragland
- Department of Physiology, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, 70112, USA
| | - Scott Edwards
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, 70112, USA
- Department of Physiology, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, 70112, USA
| | - Javier Recio
- Department of Organic Chemistry and IQAR, University of Alcala, 28805, Alcala de Henares, Madrid, Spain
| | - Julio Alvarez-Builla
- Department of Organic Chemistry and IQAR, University of Alcala, 28805, Alcala de Henares, Madrid, Spain
| | - James J Cai
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX, 77843, USA
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, 77843, USA
| | - Nicolas G Bazan
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, 70112, USA.
| |
Collapse
|
7
|
Karimi SA, Zahra FT, Martin LJ. IUPHAR review: Navigating the role of preclinical models in pain research. Pharmacol Res 2024; 200:107073. [PMID: 38232910 DOI: 10.1016/j.phrs.2024.107073] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/09/2024] [Accepted: 01/11/2024] [Indexed: 01/19/2024]
Abstract
Chronic pain is a complex and challenging medical condition that affects millions of people worldwide. Understanding the underlying mechanisms of chronic pain is a key goal of preclinical pain research so that more effective treatment strategies can be developed. In this review, we explore nociception, pain, and the multifaceted factors that lead to chronic pain by focusing on preclinical models. We provide a detailed look into inflammatory and neuropathic pain models and discuss the most used animal models for studying the mechanisms behind these conditions. Additionally, we emphasize the vital role of these preclinical models in developing new pain-relief drugs, focusing on biologics and the therapeutic potential of NMDA and cannabinoid receptor antagonists. We also discuss the challenges of TRPV1 modulation for pain treatment, the clinical failures of neurokinin (NK)- 1 receptor antagonists, and the partial success story of Ziconotide to provide valuable lessons for preclinical pain models. Finally, we highlight the overall success and limitations of current treatments for chronic pain while providing critical insights into the development of more effective therapies to alleviate the burden of chronic pain.
Collapse
Affiliation(s)
- Seyed Asaad Karimi
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada
| | - Fatama Tuz Zahra
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Loren J Martin
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada; Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada.
| |
Collapse
|
8
|
Xue Y, Mo S, Li Y, Cao Y, Xu X, Xie Q. Dissecting neural circuits from rostral ventromedial medulla to spinal trigeminal nucleus bidirectionally modulating craniofacial mechanical sensitivity. Prog Neurobiol 2024; 232:102561. [PMID: 38142769 DOI: 10.1016/j.pneurobio.2023.102561] [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: 08/27/2023] [Revised: 11/24/2023] [Accepted: 12/15/2023] [Indexed: 12/26/2023]
Abstract
Chronic craniofacial pain is intractable and its mechanisms remain unclarified. The rostral ventromedial medulla (RVM) plays a crucial role in descending pain facilitation and inhibition. It is unclear how the descending circuits from the RVM to spinal trigeminal nucleus (Sp5) are organized to bidirectionally modulate craniofacial nociception. We used viral tracing, in vivo optogenetics, calcium signaling recording, and chemogenetic manipulations to investigate the structure and function of RVM-Sp5 circuits. We found that most RVM neurons projecting to Sp5 were GABAergic or glutamatergic and facilitated or inhibited craniofacial nociception, respectively. Both GABAergic interneurons and glutamatergic projection neurons in Sp5 received RVM inputs: the former were antinociceptive, whereas the latter were pronociceptive. Furthermore, we demonstrated activation of both GABAergic and glutamatergic Sp5 neurons receiving RVM inputs in inflammation- or dysfunction-induced masseter hyperalgesia. Activating GABAergic Sp5 neurons or inhibiting glutamatergic Sp5 neurons that receive RVM projections reversed masseter hyperalgesia. Our study identifies specific cell types and projections of RVM-Sp5 circuits involved in facilitating or inhibiting craniofacial nociception respectively. Selective manipulation of RVM-Sp5 circuits can be used as potential treatment strategy to relieve chronic craniofacial muscle pain.
Collapse
Affiliation(s)
- Yang Xue
- Department of Prosthodontics, Center for Oral and Jaw Functional Diagnosis, Treatment and Research, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing 100081, PR China
| | - Siyi Mo
- Department of Prosthodontics, Center for Oral and Jaw Functional Diagnosis, Treatment and Research, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing 100081, PR China
| | - Yuan Li
- Department of Prosthodontics, Center for Oral and Jaw Functional Diagnosis, Treatment and Research, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing 100081, PR China
| | - Ye Cao
- Department of Prosthodontics, Center for Oral and Jaw Functional Diagnosis, Treatment and Research, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing 100081, PR China.
| | - Xiaoxiang Xu
- Department of Prosthodontics, Center for Oral and Jaw Functional Diagnosis, Treatment and Research, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing 100081, PR China.
| | - Qiufei Xie
- Department of Prosthodontics, Center for Oral and Jaw Functional Diagnosis, Treatment and Research, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing 100081, PR China.
| |
Collapse
|
9
|
Peel J, John K, Page J, Jeffries O, Heffernan SM, Tallent J, Waldron M. Topical application of isolated menthol and combined menthol-capsaicin creams: Exercise tolerance, thermal perception, pain, attentional focus and thermoregulation in the heat. Eur J Sport Sci 2023; 23:2038-2048. [PMID: 37161852 DOI: 10.1080/17461391.2023.2211966] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
We determined the effects of topically applied (i) isolated menthol cream, (ii) menthol and capsaicin co-application or (iii) placebo cream on exercise tolerance, thermal perception, pain, attentional focus and thermoregulation during exercise in the heat. Ten participants cycled at 70% maximal power output until exhaustion in 35°C and 20% relative humidity after application of (i) 5% isolated menthol, (ii) 5% menthol and 0.025% capsaicin co-application or (iii) placebo cream. Thermo-physiological responses were measured during exercise, with attentional focus and pain determined post-exercise on a 0-to-10 scale. Across the three conditions, time to exhaustion was 13.4 ± 4.8 min, mean ± SD infrared tympanic and skin temperature was 37.2 ± 0.6°C and 35.1 ± 1.2°C, respectively, and heart rate was 152 ± 47 bpm, with no changes between conditions (p > 0.05). Perceived exertion was lower in the isolated menthol vs. all other conditions (p < 0.05, ηp2 = 0.44). Thermal sensation was higher in menthol-capsaicin co-application vs. isolated menthol (p < 0.05, d = 1.1), while sweat rate was higher for capsaicin and menthol co-application compared to menthol (p < 0.05, d = 0.85). The median and interquartile range scores for pain were lower (p < 0.05) in the menthol condition (8, 7-8) compared to both menthol and capsaicin (10, 9-10) and placebo (9, 9-10), which was coupled with a greater distraction (p < 0.05) in the menthol condition (9, 7-10) compared to placebo (6, 5-7). Despite no performance effects for any topical cream application condition, these data reiterate the advantageous perceptual and analgesic role of menthol application and demonstrate no advantage of co-application with capsaicin.HighlightsTopical application of isolated menthol cream to cold-sensitive areas of the body during exhaustive exercise in the heat, elicited reduced perception of pain and enhanced sensation of cooling.While this reduction in generally unpleasant feelings (i.e. pain and heat) were coupled with lower RPE scores in the menthol condition and could be considered beneficial, there was no apparent ergogenic effect in an exercise tolerance test.Co-application of capsaicin and menthol appeared to inhibit the positive sensory effects elicited by menthol.Isolated menthol can induce changes in cognitive processes related to pain and exertion, while also reducing thermal sensation; however, the decision to use menthol creams must be balanced with the limited performance or thermoregulatory effects reported herein during exercise in hot environments.
Collapse
Affiliation(s)
- Jenny Peel
- A-STEM Centre, Faculty of Science & Engineering, Swansea University, Swansea, UK
| | - Kevin John
- A-STEM Centre, Faculty of Science & Engineering, Swansea University, Swansea, UK
| | - Joe Page
- A-STEM Centre, Faculty of Science & Engineering, Swansea University, Swansea, UK
| | - Owen Jeffries
- School of Biomedical, Nutritional and Sport Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Shane M Heffernan
- A-STEM Centre, Faculty of Science & Engineering, Swansea University, Swansea, UK
| | - Jamie Tallent
- School of Sport, Rehabilitation, and Exercise Sciences, University of Essex, Colchester, UK
- Department of Physiotherapy, Faculty of Medicine, Nursing and Health Sciences, School of Primary and Allied Health Care, Monash University, Colchester, Australia
| | - Mark Waldron
- A-STEM Centre, Faculty of Science & Engineering, Swansea University, Swansea, UK
- Welsh Institute of Performance Science, Swansea University, Swansea, UK
- School of Health and Behavioural Sciences, University of the Sunshine Coast, Queensland, Australia
| |
Collapse
|
10
|
Bai X, Zhang K, Ou C, Nie B, Zhang J, Huang Y, Zhang Y, Huang J, Ouyang H, Cao M, Huang W. Selective activation of AKAP150/TRPV1 in ventrolateral periaqueductal gray GABAergic neurons facilitates conditioned place aversion in male mice. Commun Biol 2023; 6:742. [PMID: 37460788 PMCID: PMC10352381 DOI: 10.1038/s42003-023-05106-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 07/06/2023] [Indexed: 07/20/2023] Open
Abstract
Aversion refers to feelings of strong dislike or avoidance toward particular stimuli or situations. Aversion can be caused by pain stimuli and has a long-term negative impact on physical and mental health. Aversion can also be caused by drug abuse withdrawal, resulting in people with substance use disorder to relapse. However, the mechanisms underlying aversion remain unclear. The ventrolateral periaqueductal gray (vlPAG) is considered to play a key role in aversive behavior. Our study showed that inhibition of vlPAG GABAergic neurons significantly attenuated the conditioned place aversion (CPA) induced by hindpaw pain pinch or naloxone-precipitated morphine withdrawal. However, activating or inhibiting glutamatergic neurons, or activating GABAergic neurons cannot affect or alter CPA response. AKAP150 protein expression and phosphorylated TRPV1 (p-TRPV1) were significantly upregulated in these two CPA models. In AKAP150flox/flox mice and C57/B6J wild-type mice, cell-type-selective inhibition of AKAP150 in GABAergic neurons in the vlPAG attenuated aversion. However, downregulating AKAP150 in glutamatergic neurons did not attenuate aversion. Knockdown of AKAP150 in GABAergic neurons effectively reversed the p-TRPV1 upregulation in these two CPA models utilized in our study. Collectively, inhibition of the AKAP150/p-TRPV1 pathway in GABAergic neurons in the vlPAG may be considered a potential therapeutic target for the CPA response.
Collapse
Affiliation(s)
- Xiaohui Bai
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Anesthesiology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation. Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Kun Zhang
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Chaopeng Ou
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Bilin Nie
- Department of Anesthesiology, Guangdong Women and Children Hospital, Guangzhou, China
| | - Jianxing Zhang
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yongtian Huang
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yingjun Zhang
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Jingxiu Huang
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Handong Ouyang
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
| | - Minghui Cao
- Department of Anesthesiology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation. Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Wan Huang
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
| |
Collapse
|
11
|
Hamilton LJ, Pattabiraman M, Zhong HA, Walker M, Vaughn H, Chandra S. Curcumin Stereoisomer, Cis-Trans Curcumin, as a Novel Ligand to A 1 and A 3 Adenosine Receptors. Pharmaceuticals (Basel) 2023; 16:917. [PMID: 37513829 PMCID: PMC10385834 DOI: 10.3390/ph16070917] [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: 03/23/2023] [Revised: 06/05/2023] [Accepted: 06/16/2023] [Indexed: 07/30/2023] Open
Abstract
Adenosine receptors (ARs) are being explored to generate non-opioid pain therapeutics. Vanilloid compounds, curcumin, capsaicin, and vanillin possess antinociceptive properties through their interactions with the transient receptor potential channel family. However, their binding with adenosine receptors has not been well studied. The hypothesis in this study was that a vanilloid compound, cis-trans curcumin (CTCUR), binds to each of the two Gi-linked AR subtypes (A1AR and A3AR). CTCUR was synthesized from curcumin (CUR) using the cavitand-mediated photoisomerization technique. The cell lines transfected with the specific receptor (A1AR or A3AR) were treated with CTCUR or CUR and the binding was analyzed using competitive assays, confocal microscopy, and docking. The binding assays and molecular docking indicated that CTCUR had Ki values of 306 nM (A1AR) and 400 nM (A3AR). These values suggest that CTCUR is selective for Gi-linked ARs (A1AR or A3AR) over Gs-linked ARs (A2AAR or A2BAR), based on our previous published research. In addition, the docking showed that CTCUR binds to the toggle switch domain of ARs. Curcumin (CUR) did not exhibit binding at any of these receptors. In summary, CTCUR and other modifications of CUR can be developed as novel therapeutic ligands for the Gi-linked ARs (A1AR and A3AR) involved with pain and cancer.
Collapse
Affiliation(s)
- Luke J Hamilton
- Department of Biology, University of Nebraska at Kearney, Kearney, NE 68849, USA
| | - Mahesh Pattabiraman
- Department of Chemistry, University of Nebraska at Kearney, Kearney, NE 68849, USA
| | - Haizhen A Zhong
- Department of Chemistry, University of Nebraska at Omaha, Omaha, NE 68182, USA
| | - Michaela Walker
- Department of Biology, University of Nebraska at Kearney, Kearney, NE 68849, USA
| | - Hilary Vaughn
- Department of Biology, University of Nebraska at Kearney, Kearney, NE 68849, USA
| | - Surabhi Chandra
- Department of Biology, University of Nebraska at Kearney, Kearney, NE 68849, USA
| |
Collapse
|
12
|
Peng B, Jiao Y, Zhang Y, Li S, Chen S, Xu S, Gao P, Fan Y, Yu W. Bulbospinal nociceptive ON and OFF cells related neural circuits and transmitters. Front Pharmacol 2023; 14:1159753. [PMID: 37153792 PMCID: PMC10157642 DOI: 10.3389/fphar.2023.1159753] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 04/10/2023] [Indexed: 05/10/2023] Open
Abstract
The rostral ventromedial medulla (RVM) is a bulbospinal nuclei in the descending pain modulation system, and directly affects spinal nociceptive transmission through pronociceptive ON cells and antinociceptive OFF cells in this area. The functional status of ON and OFF neurons play a pivotal role in pain chronification. As distinct pain modulative information converges in the RVM and affects ON and OFF cell excitability, neural circuits and transmitters correlated to RVM need to be defined for an in-depth understanding of central-mediated pain sensitivity. In this review, neural circuits including the role of the periaqueductal gray, locus coeruleus, parabrachial complex, hypothalamus, amygdala input to the RVM, and RVM output to the spinal dorsal horn are discussed. Meanwhile, the role of neurotransmitters is concluded, including serotonin, opioids, amino acids, cannabinoids, TRPV1, substance P and cholecystokinin, and their dynamic impact on both ON and OFF cell activities in modulating pain transmission. Via clarifying potential specific receptors of ON and OFF cells, more targeted therapies can be raised to generate pain relief for patients who suffer from chronic pain.
Collapse
Affiliation(s)
- Bingxue Peng
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| | - Yingfu Jiao
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| | - Yunchun Zhang
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| | - Shian Li
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| | - Sihan Chen
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| | - Saihong Xu
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| | - Po Gao
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| | - Yinghui Fan
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
- *Correspondence: Yinghui Fan, ; Weifeng Yu,
| | - Weifeng Yu
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
- *Correspondence: Yinghui Fan, ; Weifeng Yu,
| |
Collapse
|
13
|
Manna SSS. Dual effects of anandamide in the antiepileptic activity of diazepam in pentylenetetrazole-induced seizures in mice. Behav Pharmacol 2022; 33:527-541. [PMID: 36094027 DOI: 10.1097/fbp.0000000000000700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The prototype endocannabinoid, anandamide activates both CB 1 and transient receptor potential vanilloid type 1 channels (TRPV1) receptor at different concentrations. At high concentrations, anandamide-mediated TRPV1 effects are opposite to its effects at low concentrations via CB 1 receptor. Thus, synaptic concentrations of anandamide govern the neuronal activity and consequently might affect the response of a drug. This study was undertaken to investigate the influence of high and low doses of anandamide on the anticonvulsant action of diazepam on the subcutaneous dose of pentylenetetrazole (PTZ) in Swiss mice weighing 20-25 g. Results revealed that intracerebroventricular administration of capsazepine (a TRPV1 antagonist: 1, 10, or 100 µg/mouse) and the low doses (10 µg/mouse) of anandamide, AM404 (anandamide transport inhibitor), or URB597 (fatty acid amide hydrolase inhibitor) augmented the anticonvulsant effect of diazepam. Conversely, higher dose of anandamide, AM404, URB597 (100 µg/mouse) as well as capsaicin (a TRPV1 agonist: 1, 10, or 100 µg/mouse) attenuated the protective effect of diazepam against PTZ-induced seizures. Thus, this study demonstrates that the effects of diazepam may be augmented by activating CB 1 receptors or dampened via TRPV1 receptors. The findings of the present study can be extrapolated to understand the use of TRPV1 blockers alone or in combination of benzodiazepines in the treatment of benzodiazepines-refractory status epilepticus, a condition associated with maladaptive trafficking of synaptic gamma-aminobutyric acid and glutamate receptors. However, potential clinical applications are needed to further support such preclinical studies.
Collapse
|
14
|
Gambino G, Gallo D, Covelo A, Ferraro G, Sardo P, Giglia G. TRPV1 channels in nitric oxide-mediated signalling: insight on excitatory transmission in rat CA1 pyramidal neurons. Free Radic Biol Med 2022; 191:128-136. [PMID: 36029909 DOI: 10.1016/j.freeradbiomed.2022.08.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 08/13/2022] [Accepted: 08/15/2022] [Indexed: 11/18/2022]
Abstract
Nitric oxide (NO) is a fascinating signalling molecule implicated in a plethora of biological functions, especially at the synaptic level. Exploring neurotransmission in the hippocampus could be instrumental in the individuation of putative targets for nitric-oxide mediated neuromodulation, especially in terms of the potential repercussions on fundamental processes i.e. synaptic plasticity and excitability-related phenomena. Among these targets, endovanilloid signalling constitutes an object of study since Transient Receptors Vanilloid type 1 (TRPV1) channels possess a NO-sensitive gate modulating its activation. Also, NO has been referred to as a mediator for numerous endocannabinoid effects. Notwithstanding, the linkage between TRPV1 and NO systems in neuromodulation still remains elusive. To this end, we aim at investigating the involvement of TRPV1 in nitric oxide-mediated influence on hippocampal processes. Electrophysiological whole-cell recordings in CA1 pyramidal neurons were applied to evaluate excitatory neurotransmission in rat brain slices. Indeed, miniature excitatory postsynaptic currents (mEPSCs) were analysed upon pharmacological manipulation of TRPV1 and NO signalling pathways. In detail, only the administration of the specific TRPV1 exogenous agonist - capsaicin - reduced the frequency and amplitude of mEPSC similarly to the inhibitor of neuronal nitric oxide synthase (nNOS), 7-nitroindazole (7NI). In contrast, capsazepine, TRPV1 antagonist, does not influence excitatory transmission. The combined TRPV1 activation and nNOS blockade confirm the presence of a putative common mechanism. When we administered the endovanilloid-endocannabinoid ligand, i.e. anandamide, we unveiled a potentiation of neurotransmission that was selectively reverted by 7NI. Our data suggest that nitric oxide influences TRPV1 hippocampal signalling since these channels are not constitutively active, but can be "on-demand" activated to modulate excitation in CA1 pyramidal neurons, and that this effect is linked to nitric oxide production.
Collapse
Affiliation(s)
- Giuditta Gambino
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, Italy.
| | - Daniele Gallo
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, Italy
| | - Ana Covelo
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1215 NeuroCentre Magendie, Bordeaux, France
| | - Giuseppe Ferraro
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, Italy
| | - Pierangelo Sardo
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, Italy
| | - Giuseppe Giglia
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BIND), University of Palermo, Italy
| |
Collapse
|
15
|
Lyubashina OA, Sivachenko IB, Panteleev SS. Supraspinal Mechanisms of Intestinal Hypersensitivity. Cell Mol Neurobiol 2022; 42:389-417. [PMID: 33030712 PMCID: PMC11441296 DOI: 10.1007/s10571-020-00967-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 09/22/2020] [Indexed: 12/13/2022]
Abstract
Gut inflammation or injury causes intestinal hypersensitivity (IHS) and hyperalgesia, which can persist after the initiating pathology resolves, are often referred to somatic regions and exacerbated by psychological stress, anxiety or depression, suggesting the involvement of both the spinal cord and the brain. The supraspinal mechanisms of IHS remain to be fully elucidated, however, over the last decades the series of intestinal pathology-associated neuroplastic changes in the brain has been revealed, being potentially responsible for the phenomenon. This paper reviews current clinical and experimental data, including the authors' own findings, on these functional, structural, and neurochemical/molecular changes within cortical, subcortical and brainstem regions processing and modulating sensory signals from the gut. As concluded in the review, IHS can develop and maintain due to the bowel inflammation/injury-induced persistent hyperexcitability of viscerosensory brainstem and thalamic nuclei and sensitization of hypothalamic, amygdala, hippocampal, anterior insular, and anterior cingulate cortical areas implicated in the neuroendocrine, emotional and cognitive modulation of visceral sensation and pain. An additional contribution may come from the pathology-triggered dysfunction of the brainstem structures inhibiting nociception. The mechanism underlying IHS-associated regional hyperexcitability is enhanced NMDA-, AMPA- and group I metabotropic receptor-mediated glutamatergic neurotransmission in association with altered neuropeptide Y, corticotropin-releasing factor, and cannabinoid 1 receptor signaling. These alterations are at least partially mediated by brain microglia and local production of cytokines, especially tumor necrosis factor α. Studying the IHS-related brain neuroplasticity in greater depth may enable the development of new therapeutic approaches against chronic abdominal pain in inflammatory bowel disease.
Collapse
Affiliation(s)
- Olga A Lyubashina
- Laboratory of Cortico-Visceral Physiology, Pavlov Institute of Physiology, Russian Academy of Sciences, 6 Nab. Makarova, Saint Petersburg, 199034, Russia.
| | - Ivan B Sivachenko
- Laboratory of Cortico-Visceral Physiology, Pavlov Institute of Physiology, Russian Academy of Sciences, 6 Nab. Makarova, Saint Petersburg, 199034, Russia
| | - Sergey S Panteleev
- Laboratory of Cortico-Visceral Physiology, Pavlov Institute of Physiology, Russian Academy of Sciences, 6 Nab. Makarova, Saint Petersburg, 199034, Russia
| |
Collapse
|
16
|
Lazarini-Lopes W, Silva-Cardoso GK, Leite-Panissi CRA, Garcia-Cairasco N. Increased TRPV1 Channels and FosB Protein Expression Are Associated with Chronic Epileptic Seizures and Anxiogenic-like Behaviors in a Preclinical Model of Temporal Lobe Epilepsy. Biomedicines 2022; 10:biomedicines10020416. [PMID: 35203625 PMCID: PMC8962263 DOI: 10.3390/biomedicines10020416] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 01/15/2022] [Accepted: 01/20/2022] [Indexed: 02/04/2023] Open
Abstract
Epilepsies are neurological disorders characterized by chronic seizures and their related neuropsychiatric comorbidities, such as anxiety. The Transient Receptor Potential Vanilloid type-1 (TRPV1) channel has been implicated in the modulation of seizures and anxiety-like behaviors in preclinical models. Here, we investigated the impact of chronic epileptic seizures in anxiety-like behavior and TRPV1 channels expression in a genetic model of epilepsy, the Wistar Audiogenic Rat (WAR) strain. WARs were submitted to audiogenic kindling (AK), a preclinical model of temporal lobe epilepsy (TLE) and behavioral tests were performed in the open-field (OF), and light-dark box (LDB) tests 24 h after AK. WARs displayed increased anxiety-like behavior and TRPV1R expression in the hippocampal CA1 area and basolateral amygdala nucleus (BLA) when compared to control Wistar rats. Chronic seizures increased anxiety-like behaviors and TRPV1 and FosB expression in limbic and brainstem structures involved with epilepsy and anxiety comorbidity, such as the hippocampus, superior colliculus, and periaqueductal gray matter. Therefore, these results highlight previously unrecognized alterations in TRPV1 expression in brain structures involved with TLE and anxiogenic-like behaviors in a genetic model of epilepsy, the WAR strain, supporting an important role of TRPV1 in the modulation of neurological disorders and associated neuropsychiatric comorbidities.
Collapse
Affiliation(s)
- Willian Lazarini-Lopes
- Neuroscience and Behavioral Sciences Department, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto 14049-900, Brazil;
| | - Gleice Kelli Silva-Cardoso
- Psychology Department, Faculty of Philosophy, Science, and Letters, University of São Paulo, Ribeirão Preto 14040-901, Brazil; (G.K.S.-C.); (C.R.A.L.-P.)
| | - Christie Ramos Andrade Leite-Panissi
- Psychology Department, Faculty of Philosophy, Science, and Letters, University of São Paulo, Ribeirão Preto 14040-901, Brazil; (G.K.S.-C.); (C.R.A.L.-P.)
| | - Norberto Garcia-Cairasco
- Neuroscience and Behavioral Sciences Department, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto 14049-900, Brazil;
- Physiology Department, Ribeirão Preto School of Medicine and Neuroscience and Behavioral Sciences Department, University of São Paulo, Ribeirão Preto 14049-900, Brazil
- Correspondence:
| |
Collapse
|
17
|
Impaired visceral pain-related functions of the midbrain periaqueductal gray in rats with colitis. Brain Res Bull 2022; 182:12-25. [DOI: 10.1016/j.brainresbull.2022.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 01/12/2022] [Accepted: 02/03/2022] [Indexed: 11/18/2022]
|
18
|
Wang X, Liao Q, Chen H, Gong G, Siu SWI, Chen Q, Kam H, Ung COL, Cheung KK, Rádis-Baptista G, Wong CTT, Lee SMY. Toxic Peptide From Palythoa caribaeorum Acting on the TRPV1 Channel Prevents Pentylenetetrazol-Induced Epilepsy in Zebrafish Larvae. Front Pharmacol 2021; 12:763089. [PMID: 34925021 PMCID: PMC8672801 DOI: 10.3389/fphar.2021.763089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/08/2021] [Indexed: 11/25/2022] Open
Abstract
PcActx peptide, identified from the transcriptome of zoantharian Palythoa caribaeorum, was clustered into the phylogeny of analgesic polypeptides from sea anemone Heteractis crispa (known as APHC peptides). APHC peptides were considered as inhibitors of transient receptor potential cation channel subfamily V member 1 (TRPV1). TRPV1 is a calcium-permeable channel expressed in epileptic brain areas, serving as a potential target for preventing epileptic seizures. Through in silico and in vitro analysis, PcActx peptide was shown to be a potential TRPV1 channel blocker. In vivo studies showed that the linear and oxidized PcActx peptides caused concentration-dependent increases in mortality of zebrafish larvae. However, monotreatment with PcActx peptides below the maximum tolerated doses (MTD) did not affect locomotor behavior. Moreover, PcActx peptides (both linear and oxidized forms) could effectively reverse pentylenetetrazol (PTZ)-induced seizure-related behavior in zebrafish larvae and prevent overexpression of c-fos and npas4a at the mRNA level. The excessive production of ROS induced by PTZ was markedly attenuated by both linear and oxidized PcActx peptides. It was also verified that the oxidized PcActx peptide was more effective than the linear one. In particular, oxidized PcActx peptide notably modulated the mRNA expression of genes involved in calcium signaling and γ-aminobutyric acid (GABA)ergic-glutamatergic signaling, including calb1, calb2, gabra1, grm1, gria1b, grin2b, gat1, slc1a2b, gad1b, and glsa. Taken together, PcActx peptide, as a novel neuroactive peptide, exhibits prominent anti-epileptic activity, probably through modulating calcium signaling and GABAergic-glutamatergic signaling, and is a promising candidate for epilepsy management.
Collapse
Affiliation(s)
- Xiufen Wang
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Qiwen Liao
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macau, China.,School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, China
| | - Hanbin Chen
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Guiyi Gong
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Shirley Weng In Siu
- Department of Computer and Information Science, Faculty of Science and Technology, University of Macau, Macau, China
| | - Qian Chen
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Hiotong Kam
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Carolina Oi Lam Ung
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Kwok-Kuen Cheung
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong, China
| | - Gandhi Rádis-Baptista
- Laboratory of Biochemistry and Biotechnology, Institute for Marine Sciences, Federal University of Ceará, Fortaleza, Brazil
| | - Clarence Tsun Ting Wong
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China
| | - Simon Ming-Yuen Lee
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macau, China
| |
Collapse
|
19
|
Hamilton LJ, Walker M, Pattabiraman M, Zhong HA, Luedtke B, Chandra S. Novel curcumin analog (cis-trans curcumin) as ligand to adenosine receptors A 2A and A 2B: potential for therapeutics. Pharmacol Res 2021; 165:105410. [PMID: 33401004 PMCID: PMC7979524 DOI: 10.1016/j.phrs.2020.105410] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/22/2020] [Accepted: 12/23/2020] [Indexed: 02/06/2023]
Abstract
All four of the adenosine receptor (AR) subtypes mediate pain and have been targeted by pharmacologists to generate new therapeutics for chronic pain. The vanilloid phytochemicals, which include curcumin, capsaicin, and gingerol, have been shown to alleviate pain. However, there is little to no literature on the interaction of vanilloid phytochemicals with ARs. In this study, photochemical methods were used to generate a novel isomer of curcumin (cis-trans curcumin or CTCUR), and the interactions of both curcumin and CTCUR with the two Gs-linked AR subtypes were studied. Competitive binding assays, docking analysis, and confocal fluorescence microscopy were performed to measure binding affinity; cell survival assays were used to measure toxicity; and cAMP assays were performed to measure receptor activation. Competitive binding results indicated that CTCUR binds to both AR A2A and AR A2B with Ki values of 5 μM and 7 μM, respectively, which is consistent with our docking results. Fluorescence microscopy data also shows binding for A2B and A2A. Cell survival results show that CTCUR and CUR are nontoxic at the tested concentrations in these cell lines. Overall, our results suggest that vanilloid phytochemicals may be slightly modified to increase interaction with Gs-ARs, and thereby can be further explored to provide a novel class of non-opioid antinociceptives.
Collapse
Affiliation(s)
- Luke J Hamilton
- Department of Biology, University of Nebraska-Kearney, United States
| | - Michaela Walker
- Department of Biology, University of Nebraska-Kearney, United States
| | | | - Haizhen A Zhong
- Department of Chemistry, University of Nebraska-Omaha, United States
| | - Brandon Luedtke
- Department of Biology, University of Nebraska-Kearney, United States
| | - Surabhi Chandra
- Department of Biology, University of Nebraska-Kearney, United States.
| |
Collapse
|
20
|
CB1-cannabinoid-, TRPV1-vanilloid- and NMDA-glutamatergic-receptor-signalling systems interact in the prelimbic cerebral cortex to control neuropathic pain symptoms. Brain Res Bull 2020; 165:118-128. [DOI: 10.1016/j.brainresbull.2020.09.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 08/20/2020] [Accepted: 09/16/2020] [Indexed: 12/28/2022]
|
21
|
Uliana DL, Antero LS, Borges-Assis AB, Rosa J, Vila-Verde C, Lisboa SF, Resstel LB. Differential modulation of the contextual conditioned emotional response by CB1 and TRPV1 receptors in the ventromedial prefrontal cortex: Possible involvement of NMDA/nitric oxide-related mechanisms. J Psychopharmacol 2020; 34:1043-1055. [PMID: 32638638 DOI: 10.1177/0269881120928201] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Blockade of cannabinoid CB1 or vanilloid TRPV1 receptors in the ventromedial prefrontal cortex of rats respectively increases or decreases the conditioned emotional response during re-exposure to a context previously paired with footshocks. Although these mechanisms are unknown, they may involve local modulation of glutamatergic and nitrergic signaling. AIM We investigated whether these mechanisms are involved in the reported effects of CB1 and TRPV1 modulation in the ventromedial prefrontal cortex. METHODS Freezing behavior and autonomic parameters were recorded during the conditioned response expression. RESULTS The CB1 receptors antagonist NIDA, or the TRPV1 agonist capsaicin (CPS) in the ventromedial prefrontal cortex increased the conditioned emotional response expression, and these effects were prevented by TRPV1 and CB1 antagonism, respectively. The increased conditioned emotional response evoked by NIDA and CPS were prevented by an NMDA antagonist or a neuronal nitric oxide synthase inhibitor. A nitric oxide scavenger or a soluble guanylate cyclase inhibitor prevented only the NIDA effects and the CPS effect was prevented by a non-selective antioxidant drug, as nitric oxide can also induce reactive oxygen species production. CONCLUSION Our results suggest that CB1 and TRPV1 receptors in the ventromedial prefrontal cortex differently modulate the expression of conditioned emotional response through glutamatergic and nitrergic mechanisms, although different pathways may be involved.
Collapse
Affiliation(s)
- Daniela L Uliana
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, Pittsburgh, USA.,Department of Pharmacology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirao Preto, São Paulo, Brazil
| | - Leandro S Antero
- Department of Pharmacology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirao Preto, São Paulo, Brazil
| | - Anna B Borges-Assis
- Department of Pharmacology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirao Preto, São Paulo, Brazil
| | - Jessica Rosa
- Department of Pharmacology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirao Preto, São Paulo, Brazil
| | - Carla Vila-Verde
- Department of Pharmacology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirao Preto, São Paulo, Brazil
| | - Sabrina F Lisboa
- Department of Pharmacology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirao Preto, São Paulo, Brazil.,Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirao Preto, São Paulo, Brazil.,National Institute of Science and Technology for Translational Medicine, Brazilian National Council for Scientific and Technological Development, Brasília, Brazil
| | - Leonardo Bm Resstel
- Department of Pharmacology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirao Preto, São Paulo, Brazil.,National Institute of Science and Technology for Translational Medicine, Brazilian National Council for Scientific and Technological Development, Brasília, Brazil
| |
Collapse
|
22
|
Stensson N, Grimby-Ekman A. Altered relationship between anandamide and glutamate in circulation after 30 min of arm cycling: A comparison of chronic pain subject with healthy controls. Mol Pain 2020; 15:1744806919898360. [PMID: 31838922 PMCID: PMC6964246 DOI: 10.1177/1744806919898360] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The insufficient knowledge of biochemical mechanisms behind the emergence and
maintenance of chronic musculoskeletal pain conditions constrains the
development of diagnostic and therapeutic tools for clinical use. However,
physical activity and exercise may improve pain severity and physical function
during chronic pain conditions. Nevertheless, the biochemical consequences of
physical activity and exercise in chronic pain need to be elucidated to increase
the precision of this therapeutic tool in chronic pain treatment. The
endocannabinoid system has been suggested to play an important role in
exercise-induced reward and pain inhibition. Moreover, glutamatergic signalling
has been suggested as an important factor for sensation and transmission of
pain. In addition, a link has been established between the endocannabinoid
system and glutamatergic pathways. This study examines the effect of dynamic
load arm cycling (30 min) on levels of lipid mediators related to the
endocannabinoid system and glutamate in plasma of chronic pain subjects and
pain-free controls. Pain assessments and plasma levels of
arachidonoylethanolamide (anandamide), 2-aracidonoylglycerol,
oleoylethanolamide, palmitoylethanolamide, stearoylethanolamide and glutamate
from 21 subjects with chronic neck pain (chronic pain group) and 11 healthy
controls were analysed pre and post intervention of dynamic load arm cycling.
Pain intensity was significantly different between groups pre and post exercise.
Post exercise, anandamide levels were significantly decreased in health controls
but not in the chronic pain group. A strong positive correlation existed between
anandamide and glutamate in the control group post exercise but not in the
chronic pain group. Moreover, the glutamate/anandamide ratio increased
significantly in the control group and differed significantly with the chronic
pain group post exercise. The altered relationship between anandamide and
glutamate after the intervention in the chronic pain group might reflect
alterations in the endocannabinoid-glutamate mechanistic links in the chronic
pain group compared to the pain-free control group.
Collapse
Affiliation(s)
- Niclas Stensson
- Pain and Rehabilitation Centre, Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Anna Grimby-Ekman
- Health Metrics, Department of Medicine, School of Public Health and Community Medicine, Gothenburg University, Gothenburg, Sweden
| |
Collapse
|
23
|
Barrière DA, Boumezbeur F, Dalmann R, Cadeddu R, Richard D, Pinguet J, Daulhac L, Sarret P, Whittingstall K, Keller M, Mériaux S, Eschalier A, Mallet C. Paracetamol is a centrally acting analgesic using mechanisms located in the periaqueductal grey. Br J Pharmacol 2020; 177:1773-1792. [PMID: 31734950 DOI: 10.1111/bph.14934] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 10/01/2019] [Accepted: 10/24/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND AND PURPOSE We previously demonstrated that paracetamol has to be metabolised in the brain by fatty acid amide hydrolase enzyme into AM404 (N-(4-hydroxyphenyl)-5Z,8Z,11Z,14Z-eicosatetraenamide) to activate CB1 receptors and TRPV1 channels, which mediate its analgesic effect. However, the brain mechanisms supporting paracetamol-induced analgesia remain unknown. EXPERIMENTAL APPROACH The effects of paracetamol on brain function in Sprague-Dawley rats were determined by functional MRI. Levels of neurotransmitters in the periaqueductal grey (PAG) were measured using in vivo 1 H-NMR and microdialysis. Analgesic effects of paracetamol were assessed by behavioural tests and challenged with different inhibitors, administered systemically or microinjected in the PAG. KEY RESULTS Paracetamol decreased the connectivity of major brain structures involved in pain processing (insula, somatosensory cortex, amygdala, hypothalamus, and the PAG). This effect was particularly prominent in the PAG, where paracetamol, after conversion to AM404, (a) modulated neuronal activity and functional connectivity, (b) promoted GABA and glutamate release, and (c) activated a TRPV1 channel-mGlu5 receptor-PLC-DAGL-CB1 receptor signalling cascade to exert its analgesic effects. CONCLUSIONS AND IMPLICATIONS The elucidation of the mechanism of action of paracetamol as an analgesic paves the way for pharmacological innovations to improve the pharmacopoeia of analgesic agents.
Collapse
Affiliation(s)
- David André Barrière
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Basics and Clinical Pharmacology of Pain, Clermont-Ferrand, France.,Analgesia Institute, Faculty of Medicine, Clermont-Ferrand, France.,NeuroSpin, CEA, Université Paris-Saclay, Gif-sur-Yvette, France
| | | | - Romain Dalmann
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Basics and Clinical Pharmacology of Pain, Clermont-Ferrand, France.,Analgesia Institute, Faculty of Medicine, Clermont-Ferrand, France
| | - Roberto Cadeddu
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Basics and Clinical Pharmacology of Pain, Clermont-Ferrand, France.,Analgesia Institute, Faculty of Medicine, Clermont-Ferrand, France
| | - Damien Richard
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Basics and Clinical Pharmacology of Pain, Clermont-Ferrand, France.,Analgesia Institute, Faculty of Medicine, Clermont-Ferrand, France
| | - Jérémy Pinguet
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Basics and Clinical Pharmacology of Pain, Clermont-Ferrand, France.,Analgesia Institute, Faculty of Medicine, Clermont-Ferrand, France
| | - Laurence Daulhac
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Basics and Clinical Pharmacology of Pain, Clermont-Ferrand, France.,Analgesia Institute, Faculty of Medicine, Clermont-Ferrand, France
| | - Philippe Sarret
- Département de Physiologie et Biophysique/Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Kevin Whittingstall
- Département de Radiologie Diagnostique, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Matthieu Keller
- UMR Physiologie de la Reproduction et des Comportements, INRA/CNRS/Université de Tours/IFCE, Nouzilly, France
| | | | - Alain Eschalier
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Basics and Clinical Pharmacology of Pain, Clermont-Ferrand, France.,Analgesia Institute, Faculty of Medicine, Clermont-Ferrand, France
| | - Christophe Mallet
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Basics and Clinical Pharmacology of Pain, Clermont-Ferrand, France.,Analgesia Institute, Faculty of Medicine, Clermont-Ferrand, France
| |
Collapse
|
24
|
Han QW, Yuan YH, Chen NH. The therapeutic role of cannabinoid receptors and its agonists or antagonists in Parkinson's disease. Prog Neuropsychopharmacol Biol Psychiatry 2020; 96:109745. [PMID: 31442553 DOI: 10.1016/j.pnpbp.2019.109745] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 08/19/2019] [Accepted: 08/19/2019] [Indexed: 12/16/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease and its characteristic is the progressive degeneration of dopaminergic neurons within the substantia nigra (SN) of the midbrain. There is hardly any clinically proven efficient therapeutics for its cure in several recent preclinical advances proposed to treat PD. Recent studies have found that the endocannabinoid signaling system in particular the comprised two receptors, CB1 and CB2 receptors, has a significant regulatory function in basal ganglia and is involved in the pathogenesis of PD. Therefore, adding new insights into the biochemical interactions between cannabinoids and other signaling pathways may help develop new pharmacological strategies. Factors of the endocannabinoid system (ECS) are abundantly expressed in the neural circuits of basal ganglia, where they interact interactively with glutamatergic, γ-aminobutyric acid-ergic (GABAergic), and dopaminergic signaling systems. Although preclinical studies on PD are promising, the use of cannabinoids at the clinical level has not been thoroughly studied. In this review, we evaluated the available evidence and reviewed the involvement of ECS in etiologies, symptoms and treatments related to PD. Since CB1 and CB2 receptors are the two main receptors of endocannabinoids, we primarily put the focus on the therapeutic role of CB1 and CB2 receptors in PD. We will try to determine future research clues that will help understand the potential therapeutic benefits of the ECS in the treatment of PD, aiming to open up new strategies and ideas for the treatment of PD.
Collapse
Affiliation(s)
- Qi-Wen Han
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica& Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yu-He Yuan
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica& Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Nai-Hong Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica& Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
25
|
Gambino G, Rizzo V, Giglia G, Ferraro G, Sardo P. Cannabinoids, TRPV and nitric oxide: the three ring circus of neuronal excitability. Brain Struct Funct 2019; 225:1-15. [PMID: 31792694 DOI: 10.1007/s00429-019-01992-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 11/22/2019] [Indexed: 12/13/2022]
Abstract
Endocannabinoid system is considered a relevant player in the regulation of neuronal excitability, since it contributes to maintaining the balance of the synaptic ionic milieu. Perturbations to bioelectric conductances have been implicated in the pathophysiological processes leading to hyperexcitability and epileptic seizures. Cannabinoid influence on neurosignalling is exerted on classic receptor-mediated mechanisms or on further molecular targets. Among these, transient receptor potential vanilloid (TRPV) are ionic channels modulated by cannabinoids that are involved in the transduction of a plethora of stimuli and trigger fundamental downstream pathways in the post-synaptic site. In this review, we aim at providing a brief summary of the most recent data about the cross-talk between cannabinoid system and TRPV channels, drawing attention on their role on neuronal hyperexcitability. Then, we aim to unveil a plausible point of interaction between these neural signalling systems taking into consideration nitric oxide, a gaseous molecule inducing profound modifications to neural performances. From this novel perspective, we struggle to propose innovative cellular mechanisms in the regulation of hyperexcitability phenomena, with the goal of exploring plausible CB-related mechanisms underpinning epileptic seizures.
Collapse
Affiliation(s)
- Giuditta Gambino
- Department of Experimental Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), Sezione di Fisiologia Umana G. Pagano, University of Palermo, Corso Tukory 129, Palermo, Italy.
| | - Valerio Rizzo
- Department of Experimental Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), Sezione di Fisiologia Umana G. Pagano, University of Palermo, Corso Tukory 129, Palermo, Italy
| | - Giuseppe Giglia
- Department of Experimental Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), Sezione di Fisiologia Umana G. Pagano, University of Palermo, Corso Tukory 129, Palermo, Italy
| | - Giuseppe Ferraro
- Department of Experimental Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), Sezione di Fisiologia Umana G. Pagano, University of Palermo, Corso Tukory 129, Palermo, Italy
| | - Pierangelo Sardo
- Department of Experimental Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), Sezione di Fisiologia Umana G. Pagano, University of Palermo, Corso Tukory 129, Palermo, Italy
| |
Collapse
|
26
|
Kynurenines and the Endocannabinoid System in Schizophrenia: Common Points and Potential Interactions. Molecules 2019; 24:molecules24203709. [PMID: 31619006 PMCID: PMC6832375 DOI: 10.3390/molecules24203709] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 10/09/2019] [Accepted: 10/14/2019] [Indexed: 12/15/2022] Open
Abstract
Schizophrenia, which affects around 1% of the world’s population, has been described as a complex set of symptoms triggered by multiple factors. However, the exact background mechanisms remain to be explored, whereas therapeutic agents with excellent effectivity and safety profiles have yet to be developed. Kynurenines and the endocannabinoid system (ECS) play significant roles in both the development and manifestation of schizophrenia, which have been extensively studied and reviewed previously. Accordingly, kynurenines and the ECS share multiple features and mechanisms in schizophrenia, which have yet to be reviewed. Thus, the present study focuses on the main common points and potential interactions between kynurenines and the ECS in schizophrenia, which include (i) the regulation of glutamatergic/dopaminergic/γ-aminobutyric acidergic neurotransmission, (ii) their presence in astrocytes, and (iii) their role in inflammatory mechanisms. Additionally, promising pharmaceutical approaches involving the kynurenine pathway and the ECS will be reviewed herein.
Collapse
|
27
|
Wang L, Shen J, Cai XT, Tao WW, Wan YD, Li DL, Tan XX, Wang Y. Ventrolateral Periaqueductal Gray Matter Neurochemical Lesion Facilitates Epileptogenesis and Enhances Pain Sensitivity in Epileptic Rats. Neuroscience 2019; 411:105-118. [PMID: 31158436 DOI: 10.1016/j.neuroscience.2019.05.027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 05/13/2019] [Accepted: 05/15/2019] [Indexed: 12/31/2022]
Abstract
The ventrolateral periaqueductal gray matter (vlPAG) plays a critical role in the pathogenesis of migraine and few studies have shown that vlPAG might be involved in the pathophysiology of epilepsy. But its roles in epileptogenesis and comorbid relationship between migraine and epilepsy have never been reported. In this study, the impairments of vlPAG neuronal network during spontaneous recurrent seizure (SRS) development after status epilepticus (SE) were investigated, and the pain sensitivity as well as the SRS investigated after neurochemical lesion to vlPAG to determine the role of vlPAG in epileptogenesis and in migraine comorbidity with epilepsy. Neuronal loss and alterations of excitatory and inhibitory neural transmission within vlPAG accompanied the development of epileptogenesis induced by SE. On the other hand, neurochemical lesion to vlPAG enhanced frequency and duration of spontaneous seizure event and frequency of epileptiform inter-ictal spike discharges in electroencephalography (EEG), but decreased pain threshold in epileptic rats. This indicates an involvement of the pain regulating structure, vlPAG, in the pathogenesis of epilepsy. This may imply that vlPAG network alterations could be a possible underlying mechanism of the interactive comorbid relationship between epilepsy and migraine.
Collapse
Affiliation(s)
- Lei Wang
- Department of Neurology, Epilepsy and Headache Group, the First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei 230022, China
| | - Jie Shen
- Department of Neurology, Epilepsy and Headache Group, the First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei 230022, China
| | - Xin-Ting Cai
- Department of Neurology, Epilepsy and Headache Group, the First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei 230022, China
| | - Wei-Wei Tao
- Department of Neurology, Epilepsy and Headache Group, the First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei 230022, China
| | - Ya-Di Wan
- Department of Neurology, Epilepsy and Headache Group, the First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei 230022, China
| | - Dong-Lin Li
- Department of Neurology, Epilepsy and Headache Group, the First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei 230022, China
| | - Xiu-Xiu Tan
- Department of Neurology, Epilepsy and Headache Group, the First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei 230022, China
| | - Yu Wang
- Department of Neurology, Epilepsy and Headache Group, the First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei 230022, China; Department of Neurology, the Fourth Affiliated Hospital of Anhui Medical University, Huaihai Avenue 100, Hefei 230000, China.
| |
Collapse
|
28
|
Lisboa SF, Vila-Verde C, Rosa J, Uliana DL, Stern CAJ, Bertoglio LJ, Resstel LB, Guimaraes FS. Tempering aversive/traumatic memories with cannabinoids: a review of evidence from animal and human studies. Psychopharmacology (Berl) 2019; 236:201-226. [PMID: 30604182 DOI: 10.1007/s00213-018-5127-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 11/14/2018] [Indexed: 01/08/2023]
Abstract
RATIONALE Aversive learning and memory are essential to cope with dangerous and stressful stimuli present in an ever-changing environment. When this process is dysfunctional, however, it is associated with posttraumatic stress disorder (PTSD). The endocannabinoid (eCB) system has been implicated in synaptic plasticity associated with physiological and pathological aversive learning and memory. OBJECTIVE AND METHODS The objective of this study was to review and discuss evidence on how and where in the brain genetic or pharmacological interventions targeting the eCB system would attenuate aversive/traumatic memories through extinction facilitation in laboratory animals and humans. The effect size of the experimental intervention under investigation was also calculated. RESULTS Currently available data indicate that direct or indirect activation of cannabinoid type-1 (CB1) receptor facilitates the extinction of aversive/traumatic memories. Activating CB1 receptors around the formation of aversive/traumatic memories or their reminders can potentiate their subsequent extinction. In most cases, the effect size has been large (Cohen's d ≥ 1.0). The brain areas responsible for the abovementioned effects include the medial prefrontal cortex, amygdala, and/or hippocampus. The potential role of cannabinoid type-2 (CB2) receptors in extinction learning is now under investigation. CONCLUSION Drugs augmenting the brain eCB activity can temper the impact of aversive/traumatic experiences by diverse mechanisms depending on the moment of their administration. Considering the pivotal role the extinction process plays in PTSD, the therapeutic potential of these drugs is evident. The sparse number of clinical trials testing these compounds in stress-related disorders is a gap in the literature that needs to be addressed.
Collapse
Affiliation(s)
- Sabrina F Lisboa
- Department of Pharmacology, Medical School of Ribeirão Preto, University of São Paulo (FMRP/USP), Av Bandeirantes 3900, Monte Alegre, 14049900, Ribeirão Preto, São Paulo, Brazil. .,Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), Medical School of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil.
| | - C Vila-Verde
- Department of Pharmacology, Medical School of Ribeirão Preto, University of São Paulo (FMRP/USP), Av Bandeirantes 3900, Monte Alegre, 14049900, Ribeirão Preto, São Paulo, Brazil.,Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), Medical School of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - J Rosa
- Department of Pharmacology, Medical School of Ribeirão Preto, University of São Paulo (FMRP/USP), Av Bandeirantes 3900, Monte Alegre, 14049900, Ribeirão Preto, São Paulo, Brazil.,Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), Medical School of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - D L Uliana
- Department of Pharmacology, Medical School of Ribeirão Preto, University of São Paulo (FMRP/USP), Av Bandeirantes 3900, Monte Alegre, 14049900, Ribeirão Preto, São Paulo, Brazil.,Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), Medical School of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - C A J Stern
- Department of Pharmacology, Federal University of Parana, Curitiba, PR, Brazil
| | - L J Bertoglio
- Department of Pharmacology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - L B Resstel
- Department of Pharmacology, Medical School of Ribeirão Preto, University of São Paulo (FMRP/USP), Av Bandeirantes 3900, Monte Alegre, 14049900, Ribeirão Preto, São Paulo, Brazil.,Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), Medical School of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - F S Guimaraes
- Department of Pharmacology, Medical School of Ribeirão Preto, University of São Paulo (FMRP/USP), Av Bandeirantes 3900, Monte Alegre, 14049900, Ribeirão Preto, São Paulo, Brazil.,Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), Medical School of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| |
Collapse
|
29
|
Reynoso-Moreno I, Chicca A, Flores-Soto ME, Viveros-Paredes JM, Gertsch J. The Endocannabinoid Reuptake Inhibitor WOBE437 Is Orally Bioavailable and Exerts Indirect Polypharmacological Effects via Different Endocannabinoid Receptors. Front Mol Neurosci 2018; 11:180. [PMID: 29910713 PMCID: PMC5992379 DOI: 10.3389/fnmol.2018.00180] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 05/09/2018] [Indexed: 12/22/2022] Open
Abstract
Different anandamide (AEA) transport inhibitors show antinociceptive and antiinflammatory effects in vivo, but due to their concomitant inhibition of fatty acid amide hydrolase (FAAH) and overall poor bioavailability, they cannot be used unequivocally to study the particular role of endocannabinoid (EC) transport in pathophysiological conditions in vivo. Here, the potent and selective endocannabinoid reuptake inhibitor WOBE437, which inhibits AEA and 2-arachidonoylglycerol (2-AG) transport, was tested for its oral bioavailability to the brain. WOBE437 is assumed to locally increase EC levels in tissues in which facilitated EC reuptake intermediates subsequent hydrolysis. Given the marked polypharmacology of ECs, we hypothesized to see differential effects on distinct EC receptors in animal models of acute and chronic pain/inflammation. In C57BL6/J male mice, WOBE437 was orally bioavailable with an estimated tmax value of ≤20 min in plasma (Cmax ∼ 2000 pmol/mL after 50 mg/kg, p.o.) and brain (Cmax ∼ 500 pmol/g after 50 mg/kg, p.o.). WOBE437 was cleared from the brain after approximately 180 min. In addition, in BALB/c male mice, acute oral administration of WOBE437 (50 mg/kg) exhibited similar brain concentrations after 60 min and inhibited analgesia in the hot plate test in a cannabinoid CB1 receptor-dependent manner, without inducing catalepsy or affecting locomotion. WOBE437 significantly elevated AEA in the somatosensory cortex, while showing dose-dependent biphasic effects on 2-AG levels in plasma but no significant changes in N-acylethanolamines other than AEA in any of the tissues. In order to explore the presumed polypharmacology mediated via elevated EC levels, we tested this EC reuptake inhibitor in complete Freud's adjuvant induced monoarthritis in BALB/c mice as a model of chronic inflammation. Repetitive doses of WOBE437 (10 mg/kg, i.p.) attenuated allodynia and edema via cannabinoid CB2, CB1, and PPARγ receptors. The allodynia inhibition of WOBE437 treatment for 3 days was fully reversed by antagonists of any of the receptors. In the single dose treatment the CB2 and TRPV1 antagonists significantly blocked the effect of WOBE437. Overall, our results show the broad utility of WOBE437 for animal experimentation for both p.o. and i.p. administrations. Furthermore, the data indicate the possible involvement of EC reuptake/transport in pathophysiological processes related to pain and inflammation.
Collapse
Affiliation(s)
- Inés Reynoso-Moreno
- Institute of Biochemistry and Molecular Medicine, National Centre of Competence in Research TransCure, University of Bern, Bern, Switzerland.,Laboratorio de Investigación y Desarrollo Farmacéutico, Departamento de Farmacología, Centro Universitario de Ciencias Exactas e Ingenierías, Universidad de Guadalajara, Guadalajara, Mexico
| | - Andrea Chicca
- Institute of Biochemistry and Molecular Medicine, National Centre of Competence in Research TransCure, University of Bern, Bern, Switzerland
| | - Mario E Flores-Soto
- Laboratorio de Neurobiología Celular y Molecular, División de Neurociencias, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Segura Social, Guadalajara, Mexico
| | - Juan M Viveros-Paredes
- Laboratorio de Investigación y Desarrollo Farmacéutico, Departamento de Farmacología, Centro Universitario de Ciencias Exactas e Ingenierías, Universidad de Guadalajara, Guadalajara, Mexico
| | - Jürg Gertsch
- Institute of Biochemistry and Molecular Medicine, National Centre of Competence in Research TransCure, University of Bern, Bern, Switzerland
| |
Collapse
|
30
|
Fatahi Z, Reisi Z, Rainer G, Haghparast A, Khani A. Cannabinoids induce apathetic and impulsive patterns of choice through CB1 receptors and TRPV1 channels. Neuropharmacology 2018; 133:75-84. [DOI: 10.1016/j.neuropharm.2018.01.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 12/25/2017] [Accepted: 01/15/2018] [Indexed: 01/29/2023]
|
31
|
Molecular Understanding of the Activation of CB1 and Blockade of TRPV1 Receptors: Implications for Novel Treatment Strategies in Osteoarthritis. Int J Mol Sci 2018; 19:ijms19020342. [PMID: 29364174 PMCID: PMC5855564 DOI: 10.3390/ijms19020342] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 01/18/2018] [Accepted: 01/19/2018] [Indexed: 12/20/2022] Open
Abstract
Osteoarthritis (OA) is a joint disease in which cartilage degenerates as a result of mechanical and biochemical changes. The main OA symptom is chronic pain involving both peripheral and central mechanisms of nociceptive processing. Our previous studies have implicated the benefits of dual- over single-acting compounds interacting with the endocannabinoid system (ECS) in OA treatment. In the present study, we focused on the specific molecular alterations associated with pharmacological treatment. OA was induced in Wistar rats by intra-articular injection of 3 mg of monoiodoacetate (MIA). Single target compounds (URB597, an FAAH inhibitor, and SB366791, a TRPV1 antagonist) and a dual-acting compound OMDM198 (FAAH inhibitor/TRPV1 antagonist) were used in the present study. At day 21 post-MIA injection, rats were sacrificed 1 h after i.p. treatment, and changes in mRNA expression were evaluated in the lumbar spinal cord by RT-qPCR. Following MIA administration, we observed 2-4-fold increase in mRNA expression of targeted receptors (Cnr1, Cnr2, and Trpv1), endocannabinoid degradation enzymes (Faah, Ptgs2, and Alox12), and TRPV1 sensitizing kinases (Mapk3, Mapk14, Prkcg, and Prkaca). OMDM198 treatment reversed some of the MIA effects on the spinal cord towards intact levels (Alox12, Mapk14, and Prkcg). Apparent regulation of ECS and TRPV1 in response to pharmacological intervention is a strong justification for novel ECS-based multi-target drug treatment in OA.
Collapse
|
32
|
Peng X, Studholme K, Kanjiya MP, Luk J, Bogdan D, Elmes MW, Carbonetti G, Tong S, Gary Teng YH, Rizzo RC, Li H, Deutsch DG, Ojima I, Rebecchi MJ, Puopolo M, Kaczocha M. Fatty-acid-binding protein inhibition produces analgesic effects through peripheral and central mechanisms. Mol Pain 2017; 13:1744806917697007. [PMID: 28326944 PMCID: PMC5407663 DOI: 10.1177/1744806917697007] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Background Fatty-acid-binding proteins (FABPs) are intracellular carriers for endocannabinoids, N-acylethanolamines, and related lipids. Previous work indicates that systemically administered FABP5 inhibitors produce analgesia in models of inflammatory pain. It is currently not known whether FABP inhibitors exert their effects through peripheral or central mechanisms. Here, we examined FABP5 distribution in dorsal root ganglia and spinal cord and examined the analgesic effects of peripherally and centrally administered FABP5 inhibitors. Results Immunofluorescence revealed robust expression of FABP5 in lumbar dorsal root ganglia. FABP5 was distributed in peptidergic calcitonin gene-related peptide-expressing dorsal root ganglia and non-peptidergic isolectin B4-expressing dorsal root ganglia. In addition, the majority of dorsal root ganglia expressing FABP5 also expressed transient receptor potential vanilloid 1 (TRPV1) and peripherin, a marker of nociceptive fibers. Intraplantar administration of FABP5 inhibitors reduced thermal and mechanical hyperalgesia in the complete Freund’s adjuvant model of chronic inflammatory pain. In contrast to its robust expression in dorsal root ganglia, FABP5 was sparsely distributed in the lumbar spinal cord and intrathecal administration of FABP inhibitor did not confer analgesic effects. Administration of FABP inhibitor via the intracerebroventricular (i.c.v.) route reduced thermal hyperalgesia. Antagonists of peroxisome proliferator-activated receptor alpha blocked the analgesic effects of peripherally and i.c.v. administered FABP inhibitor while antagonism of cannabinoid receptor 1 blocked the effects of peripheral FABP inhibition and a TRPV1 antagonist blocked the effects of i.c.v. administered inhibitor. Although FABP5 and TRPV1 were co-expressed in the periaqueductal gray region of the brain, which is known to modulate pain, knockdown of FABP5 in the periaqueductal gray using adeno-associated viruses and pharmacological FABP5 inhibition did not produce analgesic effects. Conclusions This study demonstrates that FABP5 is highly expressed in nociceptive dorsal root ganglia neurons and FABP inhibitors exert peripheral and supraspinal analgesic effects. This indicates that peripherally restricted FABP inhibitors may serve as a new class of analgesic and anti-inflammatory agents.
Collapse
Affiliation(s)
- Xiaoxue Peng
- 1 Department of Anesthesiology, Stony Brook University, Stony Brook, NY, USA
| | - Keith Studholme
- 1 Department of Anesthesiology, Stony Brook University, Stony Brook, NY, USA
| | - Martha P Kanjiya
- 1 Department of Anesthesiology, Stony Brook University, Stony Brook, NY, USA
| | - Jennifer Luk
- 1 Department of Anesthesiology, Stony Brook University, Stony Brook, NY, USA
| | - Diane Bogdan
- 1 Department of Anesthesiology, Stony Brook University, Stony Brook, NY, USA
| | - Matthew W Elmes
- 2 Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA
| | - Gregory Carbonetti
- 2 Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA
| | - Simon Tong
- 3 Department of Chemistry, Stony Brook University, Stony Brook, NY, USA.,4 Institute of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook, NY, USA
| | - Yu-Han Gary Teng
- 3 Department of Chemistry, Stony Brook University, Stony Brook, NY, USA.,4 Institute of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook, NY, USA
| | - Robert C Rizzo
- 4 Institute of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook, NY, USA.,5 Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY, USA
| | - Huilin Li
- 2 Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA.,4 Institute of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook, NY, USA
| | - Dale G Deutsch
- 2 Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA.,4 Institute of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook, NY, USA
| | - Iwao Ojima
- 3 Department of Chemistry, Stony Brook University, Stony Brook, NY, USA.,4 Institute of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook, NY, USA
| | - Mario J Rebecchi
- 1 Department of Anesthesiology, Stony Brook University, Stony Brook, NY, USA
| | - Michelino Puopolo
- 1 Department of Anesthesiology, Stony Brook University, Stony Brook, NY, USA
| | - Martin Kaczocha
- 1 Department of Anesthesiology, Stony Brook University, Stony Brook, NY, USA.,2 Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA.,4 Institute of Chemical Biology and Drug Discovery, Stony Brook University, Stony Brook, NY, USA
| |
Collapse
|
33
|
Mascarenhas DC, Gomes KS, Sorregotti T, Nunes-de-Souza RL. Blockade of Cannabinoid CB1 Receptors in the Dorsal Periaqueductal Gray Unmasks the Antinociceptive Effect of Local Injections of Anandamide in Mice. Front Pharmacol 2017; 8:695. [PMID: 29046638 PMCID: PMC5632997 DOI: 10.3389/fphar.2017.00695] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 09/19/2017] [Indexed: 12/12/2022] Open
Abstract
Divergent results in pain management account for the growing number of studies aiming at elucidating the pharmacology of the endocannabinoid/endovanilloid anandamide (AEA) within several pain-related brain structures. For instance, the stimulation of both Transient Receptor Potential Vanilloid type 1 (TRPV1) and Cannabinoid type 1 (CB1) receptors led to paradoxical effects on nociception. Here, we attempted to propose a clear and reproducible methodology to achieve the antinociceptive effect of exogenous AEA within the dorsal periaqueductal gray (dPAG) of mice exposed to the tail-flick test. Accordingly, male Swiss mice received intra-dPAG injection of AEA (CB1/TRPV1 agonist), capsaicin (TRPV1 agonist), WIN (CB1 agonist), AM251 (CB1 antagonist), and 6-iodonordihydrocapsaicin (6-IODO) (TRPV1 selective antagonist) and their nociceptive response was assessed with the tail-flick test. In order to assess AEA effects on nociception specifically at vanilloid or cannabinoid (CB) substrates into the dPAG, mice underwent an intrinsically inactive dose of AM251 or 6-IODO followed by local AEA injections and were subjected to the same test. While intra-dPAG AEA did not change acute pain, local injections of capsaicin or WIN induced a marked TRPV1- and CB1-dependent antinociceptive effect, respectively. Regarding the role of AEA specifically at CB/vanilloid substrates, while the blockade of TRPV1 did not change the lack of effects of intra-dPAG AEA on nociception, local pre-treatment of AM251, a CB1 antagonist, led to a clear AEA-induced antinociception. It seems that the exogenous AEA-induced antinociception is unmasked when it selectively binds to vanilloid substrates, which might be useful to address acute pain in basic and perhaps clinical trials.
Collapse
Affiliation(s)
- Diego C Mascarenhas
- Joint Graduate Program in Physiological Sciences, Federal University of São Carlos and São Paulo State University, São Carlos, Brazil.,Laboratory of Neuropsychopharmacology, School of Pharmaceutical Sciences, São Paulo State University, Araraquara, Brazil
| | - Karina S Gomes
- Laboratory of Neuropsychopharmacology, School of Pharmaceutical Sciences, São Paulo State University, Araraquara, Brazil
| | - Tatiani Sorregotti
- Joint Graduate Program in Physiological Sciences, Federal University of São Carlos and São Paulo State University, São Carlos, Brazil.,Laboratory of Neuropsychopharmacology, School of Pharmaceutical Sciences, São Paulo State University, Araraquara, Brazil
| | - Ricardo L Nunes-de-Souza
- Joint Graduate Program in Physiological Sciences, Federal University of São Carlos and São Paulo State University, São Carlos, Brazil.,Laboratory of Neuropsychopharmacology, School of Pharmaceutical Sciences, São Paulo State University, Araraquara, Brazil
| |
Collapse
|
34
|
Supraspinal-selective TRPV1 desensitization induced by intracerebroventricular treatment with resiniferatoxin. Sci Rep 2017; 7:12452. [PMID: 28963471 PMCID: PMC5622082 DOI: 10.1038/s41598-017-12717-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 09/18/2017] [Indexed: 11/22/2022] Open
Abstract
The transient receptor potential vanilloid type 1 (TRPV1) is a thermosensitive cation channel that triggers heat pain in the periphery. Long-term desensitization of TRPV1, which can be induced by excess amounts of agonists, has been a method for investigating the physiological relevance of TRPV1-containing neuronal circuits, and desensitization induced by various routes of administration, including systemic, intrathecal and intraganglionic, has been demonstrated in rodents. In the present study, we examined the effect of intracerebroventricular (i.c.v.) treatment with an ultrapotent TRPV1 agonist, resiniferatoxin (RTX), on nociception and the analgesic effect of acetaminophen, which is known to mediate the activation of central TRPV1. I.c.v. administration of RTX a week before the test did not affect the licking/biting response to intraplantar injection of RTX (RTX test), suggesting that such i.c.v. treatment spares the function of TRPV1 at the hindpaw. Mice that had been i.c.v.-administered RTX also exhibited normal nociceptive responses in the formalin test and the tail pressure test, but acetaminophen failed to induce analgesia in those mice in any of the tests. These results suggest that i.c.v. administration of RTX leads to brain-selective TRPV1 desensitization in mice.
Collapse
|
35
|
Martins I, Tavares I. Reticular Formation and Pain: The Past and the Future. Front Neuroanat 2017; 11:51. [PMID: 28725185 PMCID: PMC5497058 DOI: 10.3389/fnana.2017.00051] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 06/19/2017] [Indexed: 01/10/2023] Open
Abstract
The involvement of the reticular formation (RF) in the transmission and modulation of nociceptive information has been extensively studied. The brainstem RF contains several areas which are targeted by spinal cord afferents conveying nociceptive input. The arrival of nociceptive input to the RF may trigger alert reactions which generate a protective/defense reaction to pain. RF neurons located at the medulla oblongata and targeted by ascending nociceptive information are also involved in the control of vital functions that can be affected by pain, namely cardiovascular control. The RF contains centers that belong to the pain modulatory system, namely areas involved in bidirectional balance (decrease or enhancement) of pain responses. It is currently accepted that the imbalance of pain modulation towards pain facilitation accounts for chronic pain. The medullary RF has the peculiarity of harboring areas involved in bidirectional pain control namely by the existence of specific neuronal populations involved in antinociceptive or pronociceptive behavioral responses, namely at the rostroventromedial medulla (RVM) and the caudal ventrolateral medulla (VLM). Furthermore the dorsal reticular nucleus (also known as subnucleus reticularis dorsalis; DRt) may enhance nociceptive responses, through a reverberative circuit established with spinal lamina I neurons and inhibit wide-dynamic range (WDR) neurons of the deep dorsal horn. The components of the triad RVM-VLM-DRt are reciprocally connected and represent a key gateway for top-down pain modulation. The RVM-VLM-DRt triad also represents the neurobiological substrate for the emotional and cognitive modulation of pain, through pathways that involve the periaqueductal gray (PAG)-RVM connection. Collectively, we propose that the RVM-VLM-DRt triad represents a key component of the “dynamic pain connectome” with special features to provide integrated and rapid responses in situations which are life-threatening and involve pain. The new available techniques in neurobiological studies both in animal and human studies are producing new and fascinating data which allow to understand the complex role of the RF in pain modulation and its integration with several body functions and also how the RF accounts for chronic pain.
Collapse
Affiliation(s)
- Isabel Martins
- Departamento de Biomedicina, Faculdade de Medicina do PortoPorto, Portugal.,Unidade de Biologia Experimental, Faculdade de Medicina do Porto, Universidade do PortoPorto, Portugal.,Instituto de Biologia Celular e Molecular (IBMC), Universidade do PortoPorto, Portugal.,Instituto de Investigação e Inovação em Saúde, Universidade do Porto (I3S)Porto, Portugal
| | - Isaura Tavares
- Departamento de Biomedicina, Faculdade de Medicina do PortoPorto, Portugal.,Unidade de Biologia Experimental, Faculdade de Medicina do Porto, Universidade do PortoPorto, Portugal.,Instituto de Biologia Celular e Molecular (IBMC), Universidade do PortoPorto, Portugal.,Instituto de Investigação e Inovação em Saúde, Universidade do Porto (I3S)Porto, Portugal
| |
Collapse
|
36
|
Starowicz K, Finn DP. Cannabinoids and Pain: Sites and Mechanisms of Action. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2017; 80:437-475. [PMID: 28826543 DOI: 10.1016/bs.apha.2017.05.003] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The endocannabinoid system, consisting of the cannabinoid1 receptor (CB1R) and cannabinoid2 receptor (CB2R), endogenous cannabinoid ligands (endocannabinoids), and metabolizing enzymes, is present throughout the pain pathways. Endocannabinoids, phytocannabinoids, and synthetic cannabinoid receptor agonists have antinociceptive effects in animal models of acute, inflammatory, and neuropathic pain. CB1R and CB2R located at peripheral, spinal, or supraspinal sites are important targets mediating these antinociceptive effects. The mechanisms underlying the analgesic effects of cannabinoids likely include inhibition of presynaptic neurotransmitter and neuropeptide release, modulation of postsynaptic neuronal excitability, activation of the descending inhibitory pain pathway, and reductions in neuroinflammatory signaling. Strategies to dissociate the psychoactive effects of cannabinoids from their analgesic effects have focused on peripherally restricted CB1R agonists, CB2R agonists, inhibitors of endocannabinoid catabolism or uptake, and modulation of other non-CB1R/non-CB2R targets of cannabinoids including TRPV1, GPR55, and PPARs. The large body of preclinical evidence in support of cannabinoids as potential analgesic agents is supported by clinical studies demonstrating their efficacy across a variety of pain disorders.
Collapse
Affiliation(s)
- Katarzyna Starowicz
- Institute of Pharmacology, Polish Academy of Sciences, Laboratory of Pain Pathophysiology, Krakow, Poland
| | - David P Finn
- Pharmacology and Therapeutics, School of Medicine, Galway Neuroscience Centre and Centre for Pain Research, NCBES, National University of Ireland, Galway, Ireland.
| |
Collapse
|
37
|
Gobira PH, Lima IV, Batista LA, de Oliveira AC, Resstel LB, Wotjak CT, Aguiar DC, Moreira FA. N-arachidonoyl-serotonin, a dual FAAH and TRPV1 blocker, inhibits the retrieval of contextual fear memory: Role of the cannabinoid CB1 receptor in the dorsal hippocampus. J Psychopharmacol 2017; 31:750-756. [PMID: 28583049 DOI: 10.1177/0269881117691567] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Anandamide, an endocannabinoid, inhibits aversive responses by activating the CB1 cannabinoid receptor. At high concentrations, however, anandamide may exert pro-aversive activities mediated by the transient receptor potential vanilloid type-1 channel (TRPV1). Accordingly, N-arachidonoyl-serotonin (AA-5-HT), a dual blocker of the anandamide-hydrolysing enzyme fatty acid amide hydrolase (FAAH) and the TRPV1 channel, induces anxiolytic-like effects. Here we tested the hypothesis that AA-5-HT inhibits the expression of contextual fear conditioning by facilitating CB1 receptor signalling in the dorsal hippocampus of mice. Intraperitoneal injection of AA-5-HT (0.1, 0.3, 1 mg/kg) inhibited the retrieval of contextual fear memory (freezing response). The effect of AA-5-HT (0.3 mg/kg) was prevented by systemic injection of the CB1 receptor antagonist, AM251 (1.0 mg/kg), and mimicked by simultaneous FAAH inhibition (URB597, 0.3 mg/kg) and TRPV1 blockage (SB366791, 1 mg/kg). Injection of AA-5-HT (0.125, 0.25, 0.5 nmol) into the dorsal hippocampus also reduced freezing. Finally, the effect of systemic AA-5-HT (0.3 mg/kg) was prevented by intra-hippocampal injection of AM251 (1 nmol). In conclusion, dual FAAH and TRPV1 blockage inhibits contextual fear memory by facilitating anandamide-induced CB1 receptor activation in the dorsal hippocampus. This approach may lead to new pharmacological treatments for traumatic memories and related psychiatric disorders.
Collapse
Affiliation(s)
- Pedro H Gobira
- 1 Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Isabel V Lima
- 1 Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Luara A Batista
- 1 Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Antônio C de Oliveira
- 1 Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Leonardo B Resstel
- 2 Department of Pharmacology, Medical School of Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | | | - Daniele C Aguiar
- 1 Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fabricio A Moreira
- 1 Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
38
|
Faraji N, Komaki A, Salehi I. Interaction Between the Cannabinoid and Vanilloid Systems on Anxiety in Male Rats. Basic Clin Neurosci 2017; 8:129-137. [PMID: 28539997 PMCID: PMC5440922 DOI: 10.18869/nirp.bcn.8.2.129] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Introduction: Previous studies have shown that the cannabinoid system is involved in anxiety. In addition, transient receptor potential vanilloid type-1 (TRPV1) channels are new targets for the development of anxiolytics. The present study investigated the possible interaction between the cannabinoid and vanilloid systems on anxiety-like behavior in rats. Methods: Four different groups of male Wistar rats received intraperitoneal (IP) injections of (1) vehicle (DMSO+saline), (2) cannabinoid receptor agonist WIN55212-2 (WIN) (1 mg/kg), (3) TRPV1 receptor antagonist capsazepine (CPZ) (5 mg/kg), or (4) combined WIN (1 mg/kg) and CPZ (5 mg/kg) treatment 30 minutes before testing in the elevated plus maze. Results: The results showed that compared to the control (vehicle), both WIN and CPZ increased the time spent and number of entries on the open arms. Co-administration of WIN and CPZ had a synergistic effect, i.e., the number of entries and time spent on the open arms was greater than that in the groups administered the two compounds alone. The total distance travelled by rats and total number of entries on to the arms did not significantly differ between groups. Conclusion: Acute neuropharmacological blockade of the TRPV1 receptor or stimulation of the CB1 receptor produced an anxiolytic effect. It seems that antagonism of the vanilloid system modulates cannabinoid gain that rises the anxiolytic effect. TRPV1 antagonism may amend generation of endocannabinoids, which in turn increases anxiolytic impact. These results suggest that two systems could act on or share a common signaling pathway affecting the expression of anxiety.
Collapse
Affiliation(s)
- Nafiseh Faraji
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.,Department of Biology, Hamadan Branch, Islamic Azad University, Hamadan, Iran
| | - Alireza Komaki
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Iraj Salehi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
39
|
Divergent Modulation of Nociception by Glutamatergic and GABAergic Neuronal Subpopulations in the Periaqueductal Gray. eNeuro 2017; 4:eN-NWR-0129-16. [PMID: 28374016 PMCID: PMC5370278 DOI: 10.1523/eneuro.0129-16.2017] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 03/01/2017] [Accepted: 03/03/2017] [Indexed: 12/27/2022] Open
Abstract
The ventrolateral periaqueductal gray (vlPAG) constitutes a major descending pain modulatory system and is a crucial site for opioid-induced analgesia. A number of previous studies have demonstrated that glutamate and GABA play critical opposing roles in nociceptive processing in the vlPAG. It has been suggested that glutamatergic neurotransmission exerts antinociceptive effects, whereas GABAergic neurotransmission exert pronociceptive effects on pain transmission, through descending pathways. The inability to exclusively manipulate subpopulations of neurons in the PAG has prevented direct testing of this hypothesis. Here, we demonstrate the different contributions of genetically defined glutamatergic and GABAergic vlPAG neurons in nociceptive processing by employing cell type-specific chemogenetic approaches in mice. Global chemogenetic manipulation of vlPAG neuronal activity suggests that vlPAG neural circuits exert tonic suppression of nociception, consistent with previous pharmacological and electrophysiological studies. However, selective modulation of GABAergic or glutamatergic neurons demonstrates an inverse regulation of nociceptive behaviors by these cell populations. Selective chemogenetic activation of glutamatergic neurons, or inhibition of GABAergic neurons, in vlPAG suppresses nociception. In contrast, inhibition of glutamatergic neurons, or activation of GABAergic neurons, in vlPAG facilitates nociception. Our findings provide direct experimental support for a model in which excitatory and inhibitory neurons in the PAG bidirectionally modulate nociception.
Collapse
|
40
|
Carletti F, Gambino G, Rizzo V, Ferraro G, Sardo P. Involvement of TRPV1 channels in the activity of the cannabinoid WIN 55,212-2 in an acute rat model of temporal lobe epilepsy. Epilepsy Res 2016; 122:56-65. [PMID: 26970948 DOI: 10.1016/j.eplepsyres.2016.02.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 01/18/2016] [Accepted: 02/09/2016] [Indexed: 10/21/2022]
Abstract
The exogenous cannabinoid agonist WIN 55,212-2, (R)-(+)-[2,3-dihydro-5-methyl-3-(4-morpholinylmethyl) pyrrolo[1,2,3-de]-1,4-benzoxazin-6-Yl]-1-naphthalenylmethanone (WIN), has revealed to play a role on modulating the hyperexcitability phenomena in the hippocampus. Cannabinoid-mediated mechanisms of neuroprotection have recently been found to imply the modulation of transient receptor potential vanilloid 1 (TRPV1), a cationic channel subfamily that regulate synaptic excitation. In our study, we assessed the influence of pharmacological manipulation of TRPV1 function, alone and on WIN antiepileptic activity, in the Maximal Dentate Activation (MDA) acute model of temporal lobe epilepsy. Our results showed that the TRPV1 agonist, capsaicin, increased epileptic outcomes; whilst antagonizing TRPV1 with capsazepine exerts a protective role on paroxysmal discharge. When capsaicin is co-administered with WIN effective dose of 10mg/kg is able to reduce its antiepileptic strength, especially on the triggering of MDA response. Accordingly, capsazepine at the protective dose of 2mg/kg managed to potentiate WIN antiepileptic effects, when co-treated. Moreover, WIN subeffective dose of 5mg/kg was turned into effective when capsazepine comes into play. This evidence suggests that systemic administration of TRPV1-active drugs influences electrically induced epilepsy, with a noticeable protective activity for capsazepine. Furthermore, results from the pharmacological interaction with WIN support an interplay between cannabinoid and TRPV1 signaling that could represent a promising approach for a future pharmacological strategy to challenge hyperexcitability-based diseases.
Collapse
Affiliation(s)
- Fabio Carletti
- Department of "Biomedicina Sperimentale e Neuroscienze Cliniche" (Bio.Ne.C.), "Sezione di Fisiologia umana G. Pagano", University of Palermo, Corso Tukory 129-90134 Palermo, Italy.
| | - Giuditta Gambino
- Department of "Biomedicina Sperimentale e Neuroscienze Cliniche" (Bio.Ne.C.), "Sezione di Fisiologia umana G. Pagano", University of Palermo, Corso Tukory 129-90134 Palermo, Italy.
| | - Valerio Rizzo
- Department of "Biomedicina Sperimentale e Neuroscienze Cliniche" (Bio.Ne.C.), "Sezione di Fisiologia umana G. Pagano", University of Palermo, Corso Tukory 129-90134 Palermo, Italy.
| | - Giuseppe Ferraro
- Department of "Biomedicina Sperimentale e Neuroscienze Cliniche" (Bio.Ne.C.), "Sezione di Fisiologia umana G. Pagano", University of Palermo, Corso Tukory 129-90134 Palermo, Italy.
| | - Pierangelo Sardo
- Department of "Biomedicina Sperimentale e Neuroscienze Cliniche" (Bio.Ne.C.), "Sezione di Fisiologia umana G. Pagano", University of Palermo, Corso Tukory 129-90134 Palermo, Italy.
| |
Collapse
|
41
|
Aghazadeh Tabrizi M, Baraldi PG, Baraldi S, Gessi S, Merighi S, Borea PA. Medicinal Chemistry, Pharmacology, and Clinical Implications of TRPV1 Receptor Antagonists. Med Res Rev 2016; 37:936-983. [PMID: 27976413 DOI: 10.1002/med.21427] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 10/24/2016] [Accepted: 11/01/2016] [Indexed: 12/28/2022]
Abstract
Transient receptor potential vanilloid 1 (TRPV1) is an ion channel expressed on sensory neurons triggering an influx of cations. TRPV1 receptors function as homotetramers responsive to heat, proinflammatory substances, lipoxygenase products, resiniferatoxin, endocannabinoids, protons, and peptide toxins. Its phosphorylation increases sensitivity to both chemical and thermal stimuli, while desensitization involves a calcium-dependent mechanism resulting in receptor dephosphorylation. TRPV1 functions as a sensor of noxious stimuli and may represent a target to avoid pain and injury. TRPV1 activation has been associated to chronic inflammatory pain and peripheral neuropathy. Its expression is also detected in nonneuronal areas such as bladder, lungs, and cochlea where TRPV1 activation is responsible for pathology development of cystitis, asthma, and hearing loss. This review offers a comprehensive overview about TRPV1 receptor in the pathophysiology of chronic pain, epilepsy, cough, bladder disorders, diabetes, obesity, and hearing loss, highlighting how drug development targeting this channel could have a clinical therapeutic potential. Furthermore, it summarizes the advances of medicinal chemistry research leading to the identification of highly selective TRPV1 antagonists and their analysis of structure-activity relationships (SARs) focusing on new strategies to target this channel.
Collapse
Affiliation(s)
- Mojgan Aghazadeh Tabrizi
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, 44121, Ferrara, Italy
| | - Pier Giovanni Baraldi
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, 44121, Ferrara, Italy
| | - Stefania Baraldi
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, 44121, Ferrara, Italy
| | - Stefania Gessi
- Section of Pharmacology, Department of Medical Sciences, University of Ferrara, 44121, Ferrara, Italy
| | - Stefania Merighi
- Section of Pharmacology, Department of Medical Sciences, University of Ferrara, 44121, Ferrara, Italy
| | - Pier Andrea Borea
- Section of Pharmacology, Department of Medical Sciences, University of Ferrara, 44121, Ferrara, Italy
| |
Collapse
|
42
|
Olmo IG, Ferreira-Vieira TH, Ribeiro FM. Dissecting the Signaling Pathways Involved in the Crosstalk between Metabotropic Glutamate 5 and Cannabinoid Type 1 Receptors. Mol Pharmacol 2016; 90:609-619. [PMID: 27338080 DOI: 10.1124/mol.116.104372] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 06/16/2016] [Indexed: 02/06/2023] Open
Abstract
The metabotropic glutamate 5 receptor and the cannabinoid type 1 receptor are G protein-coupled receptors that are widely expressed in the central nervous system. Metabotropic glutamate 5 receptors, present at the postsynaptic site, are coupled to Gαq/11 proteins and display an excitatory response upon activation, whereas the cannabinoid type 1 receptor, mainly present at presynaptic terminals, is coupled to the Gi/o protein and triggers an inhibitory response. Recent studies suggest that the glutamatergic and endocannabinoid systems exhibit a functional interaction to modulate several neural processes. In this review, we discuss possible mechanisms involved in this crosstalk and its relationship with physiologic and pathologic conditions, including nociception, addiction, and fragile X syndrome.
Collapse
Affiliation(s)
- Isabella G Olmo
- Department of Biochemistry and Immunology, Instituto de Ciencias Biologicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Talita H Ferreira-Vieira
- Department of Biochemistry and Immunology, Instituto de Ciencias Biologicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fabiola M Ribeiro
- Department of Biochemistry and Immunology, Instituto de Ciencias Biologicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
43
|
Pain modulation from the brain during diabetic neuropathy: Uncovering the role of the rostroventromedial medulla. Neurobiol Dis 2016; 96:346-356. [PMID: 27717882 DOI: 10.1016/j.nbd.2016.10.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 09/06/2016] [Accepted: 10/01/2016] [Indexed: 01/17/2023] Open
Abstract
Diabetic neuropathy has a profound impact in the quality of life of patients who frequently complain of pain. The mechanisms underlying diabetic neuropathic pain (DNP) are no longer ascribed only to damage of peripheral nerves. The effects of diabetes at the central nervous system are currently considered causes of DPN. Management of DNP may be achieved by antidepressants that act on serotonin (5-HT) uptake, namely specific serotonin reuptake inhibitors. The rostroventromedial medulla (RVM) is a key pain control center involved in descending pain modulation at the spinal cord through local release of 5-HT and plays a peculiar role in the balance of bidirectional control (i.e. inhibitory and facilitatory) from the brain to the spinal cord. This review discusses recently uncovered neurobiological mechanisms that mediate nociceptive modulation from the RVM during diabetes installation. In early phases of the disease, facilitation of pain modulation from the RVM prevails through a triplet of mechanisms which include increase in serotonin expression at the RVM and consequent rise of serotonin levels at the spinal cord and upregulation of local facilitatory 5HT3 receptors, enhancement of spontaneous activity of facilitatory RVM neurons and up-regulation of the expression of transient receptor potential vanilloid type 1 (TRPV1) receptor. With the progression of diabetes the alterations in the RVM increase dramatically, with oxidative stress and neuronal death associated to microglia-mediated inflammation. In a manner similar to other central areas, like the thalamus, the RVM is likely to be a "pain generator/amplifier" during diabetes, accounting to increase DNP. Early interventions in DNP prevention using strategies that simultaneously tackle the exacerbation of 5-HT3 spinal receptors and of microglial RVM activity, namely those that increase the levels of anti-inflammatory cytokines, should be considered in the future of DNP treatment.
Collapse
|
44
|
Ligresti A, De Petrocellis L, Di Marzo V. From Phytocannabinoids to Cannabinoid Receptors and Endocannabinoids: Pleiotropic Physiological and Pathological Roles Through Complex Pharmacology. Physiol Rev 2016; 96:1593-659. [DOI: 10.1152/physrev.00002.2016] [Citation(s) in RCA: 253] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Apart from having been used and misused for at least four millennia for, among others, recreational and medicinal purposes, the cannabis plant and its most peculiar chemical components, the plant cannabinoids (phytocannabinoids), have the merit to have led humanity to discover one of the most intriguing and pleiotropic endogenous signaling systems, the endocannabinoid system (ECS). This review article aims to describe and critically discuss, in the most comprehensive possible manner, the multifaceted aspects of 1) the pharmacology and potential impact on mammalian physiology of all major phytocannabinoids, and not only of the most famous one Δ9-tetrahydrocannabinol, and 2) the adaptive pro-homeostatic physiological, or maladaptive pathological, roles of the ECS in mammalian cells, tissues, and organs. In doing so, we have respected the chronological order of the milestones of the millennial route from medicinal/recreational cannabis to the ECS and beyond, as it is now clear that some of the early steps in this long path, which were originally neglected, are becoming important again. The emerging picture is rather complex, but still supports the belief that more important discoveries on human physiology, and new therapies, might come in the future from new knowledge in this field.
Collapse
Affiliation(s)
- Alessia Ligresti
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche, Comprensorio Olivetti, Pozzuoli, Italy
| | - Luciano De Petrocellis
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche, Comprensorio Olivetti, Pozzuoli, Italy
| | - Vincenzo Di Marzo
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche, Comprensorio Olivetti, Pozzuoli, Italy
| |
Collapse
|
45
|
Malek N, Starowicz K. Dual-Acting Compounds Targeting Endocannabinoid and Endovanilloid Systems-A Novel Treatment Option for Chronic Pain Management. Front Pharmacol 2016; 7:257. [PMID: 27582708 PMCID: PMC4987369 DOI: 10.3389/fphar.2016.00257] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 08/02/2016] [Indexed: 12/17/2022] Open
Abstract
Compared with acute pain that arises suddenly in response to a specific injury and is usually treatable, chronic pain persists over time, and is often resistant to medical treatment. Because of the heterogeneity of chronic pain origins, satisfactory therapies for its treatment are lacking, leading to an urgent need for the development of new treatments. The leading approach in drug design is selective compounds, though they are often less effective and require chronic dosing with many side effects. Herein, we review novel approaches to drug design for the treatment of chronic pain represented by dual-acting compounds, which operate at more than one biological target. A number of studies suggest the involvement of the cannabinoid and vanilloid receptors in pain. Interestingly cannabinoid system is in interrelation with other systems that comprise lipid mediators: prostaglandins, produced by COX enzyme. Therefore, in the present review, we summarize the role of dual-acting molecules (FAAH/TRPV1 and FAAH/COX-2 inhibitors) that interact with endocannabinoid and endovanillinoid systems and act as analgesics by elevating the endogenously produced endocannabinoids and dampening the production of pro-inflammatory prostaglandins. The plasticity of the endocannabinoid system (ECS) and the ability of a single chemical entity to exert an activity on two receptor systems has been developed and extensively investigated. Here, we review up-to-date pharmacological studies on compounds interacting with FAAH enzyme together with TRPV1 receptor or COX-2 enzyme respectively. Multi-target pharmacological intervention for treating pain may lead to the development of original and efficient treatments.
Collapse
Affiliation(s)
- Natalia Malek
- Laboratory of Pain Pathophysiology, Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences Krakow, Poland
| | - Katarzyna Starowicz
- Laboratory of Pain Pathophysiology, Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences Krakow, Poland
| |
Collapse
|
46
|
Madasu MK, Okine BN, Olango WM, Rea K, Lenihan R, Roche M, Finn DP. Genotype-dependent responsivity to inflammatory pain: A role for TRPV1 in the periaqueductal grey. Pharmacol Res 2016; 113:44-54. [PMID: 27520401 DOI: 10.1016/j.phrs.2016.08.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 08/07/2016] [Accepted: 08/08/2016] [Indexed: 11/30/2022]
Abstract
Negative affective state has a significant impact on pain, and genetic background is an important moderating influence on this interaction. The Wistar-Kyoto (WKY) inbred rat strain exhibits a stress-hyperresponsive, anxiety/depressive-like phenotype and also displays a hyperalgesic response to noxious stimuli. Transient receptor potential subfamily V member 1 (TRPV1) within the midbrain periaqueductal grey (PAG) plays a key role in regulating both aversive and nociceptive behaviour. In the present study, we investigated the role of TRPV1 in the sub-columns of the PAG in formalin-evoked nociceptive behaviour in WKY versus Sprague-Dawley (SD) rats. TRPV1 mRNA expression was significantly lower in the dorsolateral (DL) PAG and higher in the lateral (L) PAG of WKY rats, compared with SD counterparts. There were no significant differences in TRPV1 mRNA expression in the ventrolateral (VL) PAG between the two strains. TRPV1 mRNA expression significantly decreased in the DLPAG and increased in the VLPAG of SD, but not WKY rats upon intra-plantar formalin administration. Intra-DLPAG administration of either the TRPV1 agonist capsaicin, or the TRPV1 antagonist 5'-Iodoresiniferatoxin (5'-IRTX), significantly increased formalin-evoked nociceptive behaviour in SD rats, but not in WKY rats. The effects of capsaicin were likely due to TRPV1 desensitisation, given their similarity to the effects of 5'-IRTX. Intra-VLPAG administration of capsaicin or 5'-IRTX reduced nociceptive behaviour in a moderate and transient manner in SD rats, and similar effects were seen with 5'-IRTX in WKY rats. Intra-LPAG administration of 5'-IRTX reduced nociceptive behaviour in a moderate and transient manner in SD rats, but not in WKY rats. These results indicate that modulation of inflammatory pain by TRPV1 in the PAG occurs in a sub-column-specific manner. The data also provide evidence for differences in the expression of TRPV1, and differences in the effects of pharmacological modulation of TRPV1 in specific PAG sub-columns, between WKY and SD rats, suggesting that TRPV1 expression and/or functionality in the PAG plays a role in hyper-responsivity to noxious stimuli in a genetic background prone to negative affect.
Collapse
Affiliation(s)
- Manish K Madasu
- Pharmacology and Therapeutics, School of Medicine, National University of Ireland Galway, University Road, Galway, Ireland; Galway Neuroscience Centre and Centre for Pain Research, NCBES, National University of Ireland Galway, University Road, Galway, Ireland
| | - Bright N Okine
- Pharmacology and Therapeutics, School of Medicine, National University of Ireland Galway, University Road, Galway, Ireland; Galway Neuroscience Centre and Centre for Pain Research, NCBES, National University of Ireland Galway, University Road, Galway, Ireland
| | - Weredeselam M Olango
- Pharmacology and Therapeutics, School of Medicine, National University of Ireland Galway, University Road, Galway, Ireland; Galway Neuroscience Centre and Centre for Pain Research, NCBES, National University of Ireland Galway, University Road, Galway, Ireland
| | - Kieran Rea
- Pharmacology and Therapeutics, School of Medicine, National University of Ireland Galway, University Road, Galway, Ireland; Galway Neuroscience Centre and Centre for Pain Research, NCBES, National University of Ireland Galway, University Road, Galway, Ireland
| | - Róisín Lenihan
- Pharmacology and Therapeutics, School of Medicine, National University of Ireland Galway, University Road, Galway, Ireland; Galway Neuroscience Centre and Centre for Pain Research, NCBES, National University of Ireland Galway, University Road, Galway, Ireland
| | - Michelle Roche
- Physiology, School of Medicine, National University of Ireland Galway, University Road, Galway, Ireland; Galway Neuroscience Centre and Centre for Pain Research, NCBES, National University of Ireland Galway, University Road, Galway, Ireland
| | - David P Finn
- Pharmacology and Therapeutics, School of Medicine, National University of Ireland Galway, University Road, Galway, Ireland; Galway Neuroscience Centre and Centre for Pain Research, NCBES, National University of Ireland Galway, University Road, Galway, Ireland.
| |
Collapse
|
47
|
Abstract
Epilepsy has 2-3% incidence worldwide. However, present antiepileptic drugs provide only partial control of seizures. Calcium ion accumulation in hippocampal neurons has long been known as a major contributor to the etiology of epilepsy. TRPV1 is a calcium-permeable channel and mediator of epilepsy in the hippocampus. TRPV1 is expressed in epileptic brain areas such as CA1 area and dentate gyrus of the hippocampus. Here the author reviews the patent literature on novel molecules targeting TRPV1 that are currently being investigated in the laboratory and are candidates for future clinical evaluation in the management of epilepsy. A limited number of recent reports have implicated TRPV1 in the induction or treatment of epilepsy suggesting that this may be new area for potential drugs targeting this debilitating disease. Thus activation of TRPV1 by oxidative stress, resiniferatoxin, cannabinoid receptor (CB1) activators (i.e. anandamide) or capsaicin induced epileptic effects, and these effects could be reduced by appropriate inhibitors, including capsazepine (CPZ), 5'-iodoresiniferatoxin (IRTX), resolvins, and CB1 antagonists. It has been also reported that CPZ and IRTX reduced spontaneous excitatory synaptic transmission through modulation of glutaminergic systems and desensitization of TRPV1 channels in the hippocampus of rats. Immunocytochemical studies indicated that TRPV1 channel expression increased in the hippocampus of mice and patients with temporal lobe epilepsy. Taken together, findings in the current literature support a role for calcium ion accumulation through TRPV1 channels in the etiology of epileptic seizures, indicating that inhibition of TRPV1 in the hippocampus may possibly be a novel target for prevention of epileptic seizures.
Collapse
Affiliation(s)
- Mustafa Nazıroğlu
- Director of Neuroscience Research Center, Suleyman Demirel University, TR-32260, Isparta, Turkey.
| |
Collapse
|
48
|
Fattori V, Hohmann MSN, Rossaneis AC, Pinho-Ribeiro FA, Verri WA. Capsaicin: Current Understanding of Its Mechanisms and Therapy of Pain and Other Pre-Clinical and Clinical Uses. Molecules 2016; 21:E844. [PMID: 27367653 PMCID: PMC6273101 DOI: 10.3390/molecules21070844] [Citation(s) in RCA: 249] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 04/27/2016] [Indexed: 02/06/2023] Open
Abstract
In this review, we discuss the importance of capsaicin to the current understanding of neuronal modulation of pain and explore the mechanisms of capsaicin-induced pain. We will focus on the analgesic effects of capsaicin and its clinical applicability in treating pain. Furthermore, we will draw attention to the rationale for other clinical therapeutic uses and implications of capsaicin in diseases such as obesity, diabetes, cardiovascular conditions, cancer, airway diseases, itch, gastric, and urological disorders.
Collapse
Affiliation(s)
- Victor Fattori
- Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Rodovia Celso Garcia Cid KM480 PR445, Caixa Postal 10.011, 86057-970 Londrina, Paraná, Brazil.
| | - Miriam S N Hohmann
- Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Rodovia Celso Garcia Cid KM480 PR445, Caixa Postal 10.011, 86057-970 Londrina, Paraná, Brazil.
| | - Ana C Rossaneis
- Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Rodovia Celso Garcia Cid KM480 PR445, Caixa Postal 10.011, 86057-970 Londrina, Paraná, Brazil.
| | - Felipe A Pinho-Ribeiro
- Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Rodovia Celso Garcia Cid KM480 PR445, Caixa Postal 10.011, 86057-970 Londrina, Paraná, Brazil.
| | - Waldiceu A Verri
- Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Rodovia Celso Garcia Cid KM480 PR445, Caixa Postal 10.011, 86057-970 Londrina, Paraná, Brazil.
| |
Collapse
|
49
|
Capsaicin, Nociception and Pain. Molecules 2016; 21:molecules21060797. [PMID: 27322240 PMCID: PMC6273518 DOI: 10.3390/molecules21060797] [Citation(s) in RCA: 136] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 06/06/2016] [Accepted: 06/14/2016] [Indexed: 12/13/2022] Open
Abstract
Capsaicin, the pungent ingredient of the hot chili pepper, is known to act on the transient receptor potential cation channel vanilloid subfamily member 1 (TRPV1). TRPV1 is involved in somatic and visceral peripheral inflammation, in the modulation of nociceptive inputs to spinal cord and brain stem centers, as well as the integration of diverse painful stimuli. In this review, we first describe the chemical and pharmacological properties of capsaicin and its derivatives in relation to their analgesic properties. We then consider the biochemical and functional characteristics of TRPV1, focusing on its distribution and biological effects within the somatosensory and viscerosensory nociceptive systems. Finally, we discuss the use of capsaicin as an agonist of TRPV1 to model acute inflammation in slices and other ex vivo preparations.
Collapse
|
50
|
Malek N, Kostrzewa M, Makuch W, Pajak A, Kucharczyk M, Piscitelli F, Przewlocka B, Di Marzo V, Starowicz K. The multiplicity of spinal AA-5-HT anti-nociceptive action in a rat model of neuropathic pain. Pharmacol Res 2016; 111:251-263. [PMID: 27326920 DOI: 10.1016/j.phrs.2016.06.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 05/25/2016] [Accepted: 06/11/2016] [Indexed: 11/27/2022]
Abstract
There is considerable evidence to support the role of anandamide (AEA), an endogenous ligand of cannabinoid receptors, in neuropathic pain modulation. AEA also produces effects mediated by other biological targets, of which the transient receptor potential vanilloid type 1 (TRPV1) has been the most investigated. Both, inhibition of AEA breakdown by fatty acid amide hydrolase (FAAH) and blockage of TRPV1 have been shown to produce anti-nociceptive effects. Recent research suggests the usefulness of dual-action compounds, which may afford greater anti-allodynic efficacy. Therefore, in the present study, we examined the effect of N-arachidonoyl-serotonin (AA-5-HT), a blocker of FAAH and TRPV1, in a rat model of neuropathic pain after intrathecal administration. We found that treatment with AA-5-HT increased the pain threshold to mechanical and thermal stimuli, with highest effect at the dose of 500nM, which was most strongly attenuated by AM-630, CB2 antagonist, administration. The single action blockers PF-3845 (1000nM, for FAAH) and I-RTX (1nM, for TRPV1) showed lower efficacy than AA-5-HT. Moreover AA-5-HT (500nM) elevated AEA and palmitoylethanolamide (PEA) levels. Among the possible targets of these mediators, only the mRNA levels of CB2, GPR18 and GPR55, which are believed to be novel cannabinoid receptors, were upregulated in the spinal cord and/or DRG of CCI rats. It was previously reported that AA-5-HT acts in CB1 and TRPV1-dependent manner after systemic administration, but here for the first time we show that AA-5-HT action at the spinal level involves CB2, with potential contributions from GRP18 and/or GPR55 receptors.
Collapse
Affiliation(s)
- Natalia Malek
- Laboratory of Pain Pathophysiology, Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Smetna 12 Street, 31-343 Krakow, Poland; Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Smetna 12 Street, 31-343 Krakow, Poland.
| | - Magdalena Kostrzewa
- Laboratory of Pain Pathophysiology, Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Smetna 12 Street, 31-343 Krakow, Poland; Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Smetna 12 Street, 31-343 Krakow, Poland.
| | - Wioletta Makuch
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Smetna 12 Street, 31-343 Krakow, Poland.
| | - Agnieszka Pajak
- Laboratory of Pain Pathophysiology, Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Smetna 12 Street, 31-343 Krakow, Poland; Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Smetna 12 Street, 31-343 Krakow, Poland.
| | - Mateusz Kucharczyk
- Laboratory of Pain Pathophysiology, Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Smetna 12 Street, 31-343 Krakow, Poland; Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Smetna 12 Street, 31-343 Krakow, Poland.
| | - Fabiana Piscitelli
- Endocannabinoid Research Group, Institute of Biomolecular ChemistryC.N.R., Via Campi Flegrei 34, Comprensorio Olivetti, 80078 Pozzuoli (NA), Italy.
| | - Barbara Przewlocka
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Smetna 12 Street, 31-343 Krakow, Poland.
| | - Vincenzo Di Marzo
- Endocannabinoid Research Group, Institute of Biomolecular ChemistryC.N.R., Via Campi Flegrei 34, Comprensorio Olivetti, 80078 Pozzuoli (NA), Italy.
| | - Katarzyna Starowicz
- Laboratory of Pain Pathophysiology, Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Smetna 12 Street, 31-343 Krakow, Poland; Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Smetna 12 Street, 31-343 Krakow, Poland.
| |
Collapse
|