1
|
Ali S, Naz Awan A, Batool S, Aslam S, Naseer A. Comprehensive drug-like assessment of pyridine carbothioamide analogs: from molecular modeling to in-vivo evaluation. Future Med Chem 2025:1-11. [PMID: 39743753 DOI: 10.1080/17568919.2024.2444864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 12/11/2024] [Indexed: 01/04/2025] Open
Abstract
AIM To evaluate the anti-inflammatory potential of novel class of chemical compounds designed by the linkage of carbothioamide moiety with pyridine. MATERIALS & METHODS In silico analysis was conducted using molecular docking followed by an in vitro cytotoxicity assay and evaluation of anti-inflammatory activity. Subsequently, in vivo performance was determined using the Complete Freund's Adjuvant-induced inflammatory model, employing macroscopic, histopathological, and protein expression analyses. RESULTS Molecular interaction studies revealed that compound R2 displayed the most favorable binding mode with human nitric oxide synthase, cyclooxygenase-1, and cycloxygenase-2. All compounds exhibit dose-dependent cytotoxicity. Notably, compound R4 was safer at higher concentration, whereas compound R2 was comparatively toxic. The in vitro anti-inflammatory activity demonstrated half maximal inhibitory concentration (IC50) values ranging from 10.25 ± 0.0 to 23.15 ± 4.24 µM, with compound R6 exhibiting the lowest IC50 value and compound R3 showing the highest. The in vivo results corroborated the anti-inflammatory effects, with a significant reduction in paw size (p < 0.001). Among the tested compounds, compound R4 exhibited the most potent anti-inflammatory activity, whereas R2 exhibited the least potency. CONCLUSION The study highlights the promise of discovering new anti-inflammatory drugs containing pyridine moiety with proven potency, efficacy, and reduced side effects.
Collapse
Affiliation(s)
- Sana Ali
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Karachi, Karachi, Pakistan
| | - Asia Naz Awan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Karachi, Karachi, Pakistan
| | - Sehrish Batool
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Karachi, Karachi, Pakistan
| | - Shazmeen Aslam
- Department of Biotechnology, University of Karachi, Karachi, Pakistan
| | - Ayesha Naseer
- Isra College of Pharmacy, Isra University, Hyderabad, Pakistan
| |
Collapse
|
2
|
Fusco A, Perrone M, Ricciardi F, Morace AM, Bonsale R, Teweldemedhin MM, Di Martino E, Limongelli R, Papa A, Maione S, Guida F, Luongo L. Combining Acmella oleracea and Boswellia serrata extracts: a novel pharmacological approach in inflammatory vestibulodynia. Front Pharmacol 2024; 15:1508107. [PMID: 39691401 PMCID: PMC11650173 DOI: 10.3389/fphar.2024.1508107] [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: 10/08/2024] [Accepted: 11/18/2024] [Indexed: 12/19/2024] Open
Abstract
Vulvodynia is a chronic pain condition that affects the vulvar area, often resulting in significant discomfort and a reduced quality of life. Current treatments for vulvodynia are limited, and there is a need for more effective therapeutic options. Acmella oleracea, known for its spilanthol content, and Boswellia serrata, rich in boswellic acids, have been explored for their potential analgesic properties in pain management. In this study, vulvodynia-like symptoms were induced in female mice using Complete Freund's adjuvant (CFA). After the induction of symptoms, the mice were treated with a combination of Acmella oleracea and Boswellia serrata extracts (AO + BS). Behavioral pain assessments were conducted to monitor the effects of the treatment. Additionally, biochemical and functional evaluations were performed to measure spinal microgliosis and neuronal overexcitation. The combination of Acmella oleracea and Boswellia serrata (AO + BS) resulted in a significant reduction of vulvar hypersensitivity in mice. Besides alleviating pain, AO + BS therapy also reduced spinal microgliosis and neuronal overexcitation in mice with vulvodynia. The findings suggest that the AO + BS combination has the potential to alleviate vulvodynia associated pain through mechanisms involving the reduction of spinal microgliosis and neuronal overexcitation. These results point to the therapeutic promise of these plant extracts for chronic pain conditions like vulvodynia. The combination of Acmella oleracea and Boswellia serrata shows potential as a treatment for vulvodynia. However, further studies are needed to explore the underlying mechanisms and to optimize the dosage for clinical use.
Collapse
Affiliation(s)
- Antimo Fusco
- Department of Experimental Medicine, Division of Pharmacology, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Michela Perrone
- Department of Experimental Medicine, Division of Pharmacology, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Federica Ricciardi
- Department of Experimental Medicine, Division of Pharmacology, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Andrea Maria Morace
- Department of Experimental Medicine, Division of Pharmacology, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Roozbe Bonsale
- Department of Experimental Medicine, Division of Pharmacology, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Milena Melake Teweldemedhin
- Department of Experimental Medicine, Division of Pharmacology, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Emanuele Di Martino
- Department of Experimental Medicine, Division of Pharmacology, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Rebecca Limongelli
- Department of Experimental Medicine, Division of Pharmacology, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Alfonso Papa
- Department of Pain Management—AO “Ospedale dei Colli”–Monaldi Hospital, Naples, Italy
| | - Sabatino Maione
- Department of Experimental Medicine, Division of Pharmacology, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Francesca Guida
- Department of Experimental Medicine, Division of Pharmacology, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Livio Luongo
- Department of Experimental Medicine, Division of Pharmacology, University of Campania “Luigi Vanvitelli”, Naples, Italy
| |
Collapse
|
3
|
Liu G, Jia D, Li W, Huang Z, Shan R, Huang C. Trifluoro-Icaritin Ameliorates Neuroinflammation Against Complete Freund's Adjuvant-Induced Microglial Activation by Improving CB2 Receptor-Mediated IL-10/β-endorphin Signaling in the Spinal Cord of Rats. J Neuroimmune Pharmacol 2024; 19:53. [PMID: 39387998 DOI: 10.1007/s11481-024-10152-8] [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: 08/12/2023] [Accepted: 09/30/2024] [Indexed: 10/15/2024]
Abstract
The underlying pathogenesis of chronic inflammatory pain is greatly complex, but the relevant therapies are still unavailable. Development of effective candidates for chronic inflammatory pain is highly urgent. We previously identified that trifluoro-icaritin (ICTF) exhibited a significant therapeutic activity against complete Freund's adjuvant (CFA)-induced chronic inflammatory pain, however, the precise mechanisms remain elusive. Here, the paw withdrawal threshold (PWT), paw withdrawal latency (PWL), and CatWalk gait analysis were used to determine the pain-related behaviors. The expression and co-localization of pain-related signaling molecules were detected by Western blot and immunofluorescence staining. Our results demonstrated that ICTF (3.0 mg/kg, i.p.) effectively attenuated mechanical allodynia, thermal hyperalgesia and improved motor dysfunction induced by CFA, and the molecular docking displayed that CB2 receptor may be the therapeutic target of ICTF. Furthermore, ICTF not only up-regulated the levels of CB2 receptor, IL-10, β-endorphin and CD206, but also reduced the expression of P2Y12 receptor, NLRP3, ASC, Caspase-1, IL-1β, CD11b, and iNOS in the spinal cord of CFA rats. Additionally, the immunofluorescence staining from the spinal cord showed that ICTF significantly increased the co-expression between the microglial marker Iba-1 and CB2 receptor, IL-10, β-endorphin, respectively, but markedly decreased the co-localization between Iba-1 and P2Y12 receptor. Conversely, intrathecal administration of CB2 receptor antagonist AM630 dramatically reversed the inhibitory effects of ICTF on CFA-induced chronic inflammatory pain, leading to a promotion of pain hypersensitivity, abnormal gait parameters, microglial activation, and up-regulation of P2Y12 receptor and NLRP3 inflammasome, as well as the inhibition of CB2 receptor and IL-10/β-endorphin cascade. Taken together, these findings highlighted that ICTF alleviated CFA-induced neuroinflammation by enhancing CB2 receptor-mediated IL-10/β-endorphin signaling and suppressing microglial activation in the spinal cord, and uncovered that CB2 receptor may be exploited as a novel and promising target for ICTF treatment of chronic inflammatory pain.
Collapse
Grants
- NO. 2021B614 Science and Technology Project of Administration of Chinese Medicine, Jiangxi Province, China
- NO. HX202207 Horizontal Project of Gannan Medical University, Jiangxi Province, China
- No.ZD201904 University-level Key Project of Gannan Medical University, Jiangxi Province, China
- No. 20204469 Health Commission General Science and Technology Program, Jiangxi Province, China
- No. 31160213 National Natural Science Foundation of China
- No. 20142BCBC22008 Talent Project of Department of Scientific and Technology, Jiangxi Province, China
Collapse
Affiliation(s)
- Guangsen Liu
- First Clinical Medical College, Gannan Medical University, Ganzhou, 341000, P. R. China
| | - Dandan Jia
- Department of Physiology, School of Basic Medicine Sciences, Gannan Medical University, Ganzhou, 341000, P. R. China
| | - Weiwei Li
- School of Public Health and Health management, Gannan Medical University, Ganzhou, 341000, P. R. China
| | - Zhihua Huang
- Department of Physiology, School of Basic Medicine Sciences, Gannan Medical University, Ganzhou, 341000, P. R. China
| | - Reai Shan
- First Clinical Medical College, Gannan Medical University, Ganzhou, 341000, P. R. China.
- Department of Anesthesiology, First Affiliated Hospital of Gannan Medical University, Jiangxi, 341000, P. R. China.
- Ganzhou Key Laboratory of Anesthesiology, Ganzhou, 341000, P. R. China.
| | - Cheng Huang
- Department of Physiology, School of Basic Medicine Sciences, Gannan Medical University, Ganzhou, 341000, P. R. China.
- School of Public Health and Health management, Gannan Medical University, Ganzhou, 341000, P. R. China.
| |
Collapse
|
4
|
Mohos V, Harmat M, Kun J, Aczél T, Zsidó BZ, Kitka T, Farkas S, Pintér E, Helyes Z. Topiramate inhibits adjuvant-induced chronic orofacial inflammatory allodynia in the rat. Front Pharmacol 2024; 15:1461355. [PMID: 39221150 PMCID: PMC11361966 DOI: 10.3389/fphar.2024.1461355] [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: 07/08/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
Chronic orofacial pain disorders are common debilitating conditions, affecting the trigeminal system. Its underlying pathophysiological mechanisms are still unclear and the therapy is often unsatisfactory, therefore, preclinical models are crucial to identify the key mediators and novel treatment options. Complete Freund's adjuvant (CFA)-induced orofacial inflammatory allodynia/hyperalgesia is commonly used in rodents, but it has not been validated with currently used drugs. Here we tested the effects of the adjuvant analgesic/antiepileptic voltage-gated Na+ channel blocker complex mechanism of action topiramate in comparison with the gold standard antimigraine serotonin 5-HT1B/D receptor agonist sumatriptan in this model. CFA was injected subcutaneously into the right whisker pad of male Sprague-Dawley rats (250-300 g), then mechanonociceptive threshold values were investigated with von Frey filaments (3, 5, and 7 days after CFA injection). Effects of topiramate (30 mg/kg per os) and sumatriptan (1 mg/kg subcutaneous) on the adjuvant-induced chronic inflammatory orofacial allodynia were investigated 60, 120, and 180 min after the treatments each day. To determine the optimal concentration for drug effect analysis, we tested the effects of two different CFA-concentrations (1 and 0.5 mg/mL) on mechanonociceptive thresholds. Both concentrations of CFA induced a chronic orofacial allodynia in 60% of all rats. Although, higher CFA concentration induced greater allodynia, much more stable threshold reduction was observed with the lower CFA concentration: on day 3 the thresholds decreased from 18.30 g to approximately 11 g (low) and 5 g (high), respectively, however a slight increase was observed in the case of higher CFA concentration (on days 5, 7, and 11). In all investigation days, topiramate showed significant anti-allodynic effect comparing the pre and post drug dose and comparing the vehicle treated to the drug treated groups. Sumatriptan also caused a significant threshold increase compared to pre dose thresholds (day 3) and also showed a slight anti-allodynic effect compared to the vehicle-treated group (day 3 and 5). In the present study CFA-induced chronic orofacial allodynia was reversed by topiramate in rats validating the model with the adjuvant analgesic. Other than establishing a validated orofacial pain-related syndrome model in rats, new ways are opened for the repurposing of topiramate.
Collapse
Affiliation(s)
- Violetta Mohos
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
| | - Máté Harmat
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
| | - Jozsef Kun
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
- Hungarian Centre for Genomics and Bioinformatics, Szentágothai Research Centre, University of Pécs, Pécs, Hungary
- National Laboratory for Drug Research and Development, Budapest, Hungary
| | - Tímea Aczél
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
| | - Balázs Zoltán Zsidó
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
| | - Tamás Kitka
- Uzsoki Cardiovascular Center Ltd., Budapest, Hungary
| | - Sándor Farkas
- Uzsoki Cardiovascular Center Ltd., Budapest, Hungary
| | - Erika Pintér
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
- National Laboratory for Drug Research and Development, Budapest, Hungary
- Hungarian Research Network, PTE HUN-REN Chronic Pain Research Group, Budapest, Hungary
- PharmInVivo Ltd., Pécs, Hungary
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary
- National Laboratory for Drug Research and Development, Budapest, Hungary
- Hungarian Research Network, PTE HUN-REN Chronic Pain Research Group, Budapest, Hungary
- PharmInVivo Ltd., Pécs, Hungary
| |
Collapse
|
5
|
Ciszek-Lenda M, Nowak B, Majka G, Suski M, Walczewska M, Fedor A, Golińska E, Górska S, Gamian A, Olszanecki R, Strus M, Marcinkiewicz J. Saccharomyces cerevisiae β-glucan improves the response of trained macrophages to severe P. aeruginosa infections. Inflamm Res 2024; 73:1283-1297. [PMID: 38850343 PMCID: PMC11282130 DOI: 10.1007/s00011-024-01898-1] [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: 01/12/2024] [Revised: 05/15/2024] [Accepted: 05/27/2024] [Indexed: 06/10/2024] Open
Abstract
OBJECTIVE P. AERUGINOSA: (PA), the major pathogen of lung cystic fibrosis (CF), polarizes macrophages into hyperinflammatory tissue damaging phenotype. The main aim of this study was to verify whether training of macrophages with β-glucan might improve their response to P. aeruginosa infections. METHODS To perform this task C57BL/6 mice sensitive to infections with P. aeruginosa were used. Peritoneal macrophages were trained with Saccharomyces cerevisiae β-glucan and exposed to PA57, the strong biofilm-forming bacterial strain isolated from the patient with severe lung CF. The release of cytokines and the expression of macrophage phenotypic markers were measured. A quantitative proteomic approach was used for the characterization of proteome-wide changes in macrophages. The effect of in vivo β-glucan-trained macrophages in the air pouch model of PA57 infection was investigated. In all experiments the effect of trained and naïve macrophages was compared. RESULTS Trained macrophages acquired a specific phenotype with mixed pro-inflammatory and pro-resolution characteristics, however they retained anti-bacterial properties. Most importantly, transfer of trained macrophages into infected air pouches markedly ameliorated the course of infection. PA57 bacterial growth and formation of biofilm were significantly suppressed. The level of serum amyloid A (SAA), a systemic inflammation biomarker, was reduced. CONCLUSIONS Training of murine macrophages with S. cerevisiae β-glucan improved macrophage defense properties along with inhibition of secretion of some detrimental inflammatory agents. We suggest that training of macrophages with such β-glucans might be a new therapeutic strategy in P. aeruginosa biofilm infections, including CF, to promote eradication of pathogens and resolution of inflammation.
Collapse
Affiliation(s)
- Marta Ciszek-Lenda
- Department of Immunology, Faculty of Medicine, Jagiellonian University Medical College, Czysta 18, Krakow, 31-121, Poland
| | - Bernadeta Nowak
- Department of Immunology, Faculty of Medicine, Jagiellonian University Medical College, Czysta 18, Krakow, 31-121, Poland
| | - Grzegorz Majka
- Department of Immunology, Faculty of Medicine, Jagiellonian University Medical College, Czysta 18, Krakow, 31-121, Poland.
| | - Maciej Suski
- Department of Pharmacology, Faculty of Medicine, Jagiellonian University Medical College, Grzegorzecka 16, Krakow, 31-53, Poland
| | - Maria Walczewska
- Department of Immunology, Faculty of Medicine, Jagiellonian University Medical College, Czysta 18, Krakow, 31-121, Poland
| | - Angelika Fedor
- Department of Immunology, Faculty of Medicine, Jagiellonian University Medical College, Czysta 18, Krakow, 31-121, Poland
| | - Edyta Golińska
- Department of Microbiology, Faculty of Medicine, Jagiellonian University Medical College, Czysta 18, Krakow, 31-121, Poland
| | - Sabina Górska
- Hirszfeld Institute of Immunology and Experimental Therapy, Department of Microbiology, Laboratory of Microbiome Immunobiology, Polish Academy of Sciences, Weigla 12, Wroclaw, 53-114, Poland
| | - Andrzej Gamian
- Hirszfeld Institute of Immunology and Experimental Therapy, Department of Immunology of Infectious Diseases, Laboratory of Medical Microbiology, Polish Academy of Sciences, Weigla 12, Wroclaw, 53-114, Poland
| | - Rafał Olszanecki
- Department of Pharmacology, Faculty of Medicine, Jagiellonian University Medical College, Grzegorzecka 16, Krakow, 31-53, Poland
| | - Magdalena Strus
- Department of Microbiology, Faculty of Medicine, Jagiellonian University Medical College, Czysta 18, Krakow, 31-121, Poland
| | - Janusz Marcinkiewicz
- University of Agriculture, University Centre of Veterinary Medicine, Mickiewicza 24/28, Krakow, 30- 059, Poland
| |
Collapse
|
6
|
Boccella S, Perrone M, Fusco A, Bonsale R, Infantino R, Nuzzo S, Pecoraro G, Ricciardi F, Maria Morace A, Petrillo G, Leone I, Franzese M, de Novellis V, Guida F, Salvatore M, Maione S, Luongo L. Spinal neuronal activity and neuroinflammatory component in a mouse model of CFA-induced vestibulodynia. Brain Behav Immun 2024; 119:408-415. [PMID: 38636564 DOI: 10.1016/j.bbi.2024.04.012] [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] [Received: 12/14/2023] [Revised: 03/21/2024] [Accepted: 04/16/2024] [Indexed: 04/20/2024] Open
Abstract
Vestibulodynia is a complex pain disorder characterized by chronic discomfort in the vulvar region, often accompanied by tactile allodynia and spontaneous pain. In patients a depressive behaviour is also observed. In this study, we have used a model of vestibulodynia induced by complete Freund's adjuvant (CFA) focusing our investigation on the spinal cord neurons and microglia. We investigated tactile allodynia, spontaneous pain, and depressive-like behavior as key behavioral markers of vestibulodynia. In addition, we conducted in vivo electrophysiological recordings to provide, for the first time to our knowledge, the characterization of the spinal sacral neuronal activity in the L6-S1 dorsal horn of the spinal cord. Furthermore, we examined microglia activation in the L6-S1 dorsal horn using immunofluorescence, unveiling hypertrophic phenotypes indicative of neuroinflammation in the spinal cord. This represents a novel insight into the role of microglia in vestibulodynia pathology. To address the therapeutic aspect, we employed pharmacological interventions using GABApentin, amitriptyline, and PeaPol. Remarkably, all three drugs, also used in clinic, showed efficacy in alleviating tactile allodynia and depressive-like behavior. Concurrently, we also observed a normalization of the altered neuronal firing and a reduction of microglia hypertrophic phenotypes. In conclusion, our study provides a comprehensive understanding of the CFA-induced model of vestibulodynia, encompassing behavioral, neurophysiological and neuroinflammatory aspects. These data pave the way to investigate spinal cord first pain plasticity in vestibulodynia.
Collapse
Affiliation(s)
- Serena Boccella
- Department of Experimental Medicine, Division of Pharmacology, Università della Campania "L. Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - Michela Perrone
- Department of Experimental Medicine, Division of Pharmacology, Università della Campania "L. Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - Antimo Fusco
- Department of Experimental Medicine, Division of Pharmacology, Università della Campania "L. Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - Roozbe Bonsale
- Department of Experimental Medicine, Division of Pharmacology, Università della Campania "L. Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - Rosmara Infantino
- Department of Experimental Medicine, Division of Pharmacology, Università della Campania "L. Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - Silvia Nuzzo
- IRCCS Synlab SDN, Via Emanuele Gianturco 113, 80143 Naples, Italy
| | | | - Federica Ricciardi
- Department of Experimental Medicine, Division of Pharmacology, Università della Campania "L. Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - Andrea Maria Morace
- Department of Experimental Medicine, Division of Pharmacology, Università della Campania "L. Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - Gianluca Petrillo
- Department of Molecular Medicine and Medical Biotechnology, "Federico II" University of Naples, Via Tommaso de Amicis 95, 80131 Naples, Italy
| | - Ilaria Leone
- IRCCS Synlab SDN, Via Emanuele Gianturco 113, 80143 Naples, Italy
| | - Monica Franzese
- IRCCS Synlab SDN, Via Emanuele Gianturco 113, 80143 Naples, Italy
| | - Vito de Novellis
- Department of Experimental Medicine, Division of Pharmacology, Università della Campania "L. Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - Francesca Guida
- Department of Experimental Medicine, Division of Pharmacology, Università della Campania "L. Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - Marco Salvatore
- IRCCS Synlab SDN, Via Emanuele Gianturco 113, 80143 Naples, Italy
| | - Sabatino Maione
- Department of Experimental Medicine, Division of Pharmacology, Università della Campania "L. Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy.
| | - Livio Luongo
- Department of Experimental Medicine, Division of Pharmacology, Università della Campania "L. Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy.
| |
Collapse
|
7
|
Kudsi SQ, Viero FT, Pereira LG, Trevisan G. Involvement of the Transient Receptor Channels in Preclinical Models of Musculoskeletal Pain. Curr Neuropharmacol 2024; 22:72-87. [PMID: 37694792 PMCID: PMC10716882 DOI: 10.2174/1570159x21666230908094159] [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: 04/10/2023] [Revised: 08/14/2023] [Accepted: 08/16/2023] [Indexed: 09/12/2023] Open
Abstract
BACKGROUND Musculoskeletal pain is a condition that affects bones, muscles, and tendons and is present in various diseases and/or clinical conditions. This type of pain represents a growing problem with enormous socioeconomic impacts, highlighting the importance of developing treatments tailored to the patient's needs. TRP is a large family of non-selective cation channels involved in pain perception. Vanilloid (TRPV1 and TRPV4), ankyrin (TRPA1), and melastatin (TRPM8) are involved in physiological functions, including nociception, mediation of neuropeptide release, heat/cold sensing, and mechanical sensation. OBJECTIVE In this context, we provide an updated view of the most studied preclinical models of muscle hyperalgesia and the role of transient receptor potential (TRP) in these models. METHODS This review describes preclinical models of muscle hyperalgesia induced by intramuscular administration of algogenic substances and/or induction of muscle damage by physical exercise in the masseter, gastrocnemius, and tibial muscles. RESULTS The participation of TRPV1, TRPA1, and TRPV4 in different models of musculoskeletal pain was evaluated using pharmacological and genetic tools. All the studies detected the antinociceptive effect of respective antagonists or reduced nociception in knockout mice. CONCLUSION Hence, TRPV1, TRPV4, and TRPA1 blockers could potentially be utilized in the future for inducing analgesia in muscle hypersensitivity pathologies.
Collapse
Affiliation(s)
- Sabrina Qader Kudsi
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), Avenida Roraima, 97105-900 Santa Maria (RS), Brazil
| | - Fernanda Tibolla Viero
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), Avenida Roraima, 97105-900 Santa Maria (RS), Brazil
| | - Leonardo Gomes Pereira
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), Avenida Roraima, 97105-900 Santa Maria (RS), Brazil
| | - Gabriela Trevisan
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), Avenida Roraima, 97105-900 Santa Maria (RS), Brazil
| |
Collapse
|
8
|
Li J, Peng S, Zhang Y, Ge J, Gao S, Zhu Y, Bai Y, Wu S, Huang J. Glutamatergic Neurons in the Zona Incerta Modulate Pain and Itch Behaviors in Mice. Mol Neurobiol 2023; 60:5866-5877. [PMID: 37354250 DOI: 10.1007/s12035-023-03431-7] [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: 02/16/2023] [Accepted: 06/05/2023] [Indexed: 06/26/2023]
Abstract
Emerging evidence suggest that parvalbumin neurons in zona incerta (ZI) modulate pain and itch behavior in opposite manners. However, the role of ZI glutamatergic neurons, a unique incertal neuronal subpopulation residing in the caudal division, in pain and itch modulation remains unknown. In the present study, by combining chemogenetic manipulation, fiber photometry, and behavioral tests, we proved that incertal glutamatergic neurons served as an endogenous negative diencephalic modulator for both pain and itch processing. We demonstrated that ZI vesicular glutamate transporter 2 (VGluT2) neurons exhibited increased calcium signal upon hindpaw withdrawal in response to experimental mechanical and thermal stimuli. Behavioral tests further showed that pharmacogenetic activation of this specific type of neurons reduced nocifensive withdrawal responses in both naïve and inflammatory pain mice. Similar neural activity and modulatory role of ZI VGluT2 neurons were also observed upon histaminergic and non-histaminergic acute itch stimuli. Together, our study would expedite our understandings of brain mechanisms underlying somatosensory processing and modulation, and supply a novel therapeutic target for the management of chronic pain and itch disorders.
Collapse
Affiliation(s)
- Jiaqi Li
- Department of Neurobiology, Basic Medical Science Academy, Fourth Military Medical University, Xi'an, 710032, China
| | - Shihao Peng
- Department of Neurobiology, Basic Medical Science Academy, Fourth Military Medical University, Xi'an, 710032, China
| | - Yiwen Zhang
- Department of Neurobiology, Basic Medical Science Academy, Fourth Military Medical University, Xi'an, 710032, China
| | - Junye Ge
- Department of Neurobiology, Basic Medical Science Academy, Fourth Military Medical University, Xi'an, 710032, China
| | - Shasha Gao
- Department of Neurobiology, Basic Medical Science Academy, Fourth Military Medical University, Xi'an, 710032, China
| | - Yuanyuan Zhu
- Department of Neurobiology, Basic Medical Science Academy, Fourth Military Medical University, Xi'an, 710032, China
| | - Yang Bai
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, 110015, China.
| | - Shengxi Wu
- Department of Neurobiology, Basic Medical Science Academy, Fourth Military Medical University, Xi'an, 710032, China.
| | - Jing Huang
- Department of Neurobiology, Basic Medical Science Academy, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
9
|
Kemenesi-Gedei PB, Csabafi KA, Kis G. Inflammatory Orofacial Pain Activates Peptidergic Neurons and Upregulates the Oxytocin Receptor Expression in Trigeminal Ganglion. Biomedicines 2023; 11:2419. [PMID: 37760859 PMCID: PMC10525584 DOI: 10.3390/biomedicines11092419] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 08/28/2023] [Accepted: 08/28/2023] [Indexed: 09/29/2023] Open
Abstract
The majority of orofacial pain is caused by musculoskeletal and neuropathological diseases related to inflammatory processes that lead even to transcriptional alterations in the trigeminal ganglion (TG) neurons. The hypothalamic nonapeptide oxytocin has been reported to modulate nociception via binding and activating its receptor in primary sensory neurons. The purpose of this study was to analyze the gene expression of the oxytocin receptor (OTR), c-Fos, an indicator of neuronal activity, and α-calcitonin gene-related peptide (αCGRP), a characteristic neurotransmitter of the peptidergic trigeminal primary afferents in an animal model of inflammation-induced orofacial pain. Carrageenan was unilaterally injected into the vibrissal pads of male and female adult Wistar rats. RT-qPCR was performed to analyze the levels of mRNA expression in TGs 24 h after injection. The gene expression analysis revealed higher fold changes regarding the c-Fos (mean ± S.E: ♀: 3.9 ± 0.19; ♂: 3.55 ± 0.18) and αCGRP (♀: 2.84 ± 0.13; ♂: 3.39 ± 0.47) expression levels of mRNA, and a moderate rise in the expression of the OTR mRNA (♀: 1.52 ± 0.07; ♂: 1.49 ± 0.07) was observed in comparison to both vehicle(saline)-treated and untreated controls. Our results furnish evidence for inflammation-induced activation of peptidergic neurons, and it is suggested that oxytocin modulates inflammation-induced nociception by enhancing their signaling capacity due to its elevated expression in the sensory ganglion cells, thus providing new therapies for orofacial pain relief that target the OTRs.
Collapse
Affiliation(s)
- Péter Bátor Kemenesi-Gedei
- Department of Physiology, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary
| | - Krisztina Anna Csabafi
- Department of Pathophysiology, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary
| | - Gyöngyi Kis
- Department of Physiology, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, 6720 Szeged, Hungary
| |
Collapse
|
10
|
Ducza L, Gajtkó A, Hegedűs K, Bakk E, Kis G, Gaál B, Takács R, Szücs P, Matesz K, Holló K. Neuronal P2X4 receptor may contribute to peripheral inflammatory pain in rat spinal dorsal horn. Front Mol Neurosci 2023; 16:1115685. [PMID: 36969557 PMCID: PMC10033954 DOI: 10.3389/fnmol.2023.1115685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 02/14/2023] [Indexed: 03/11/2023] Open
Abstract
ObjectiveIntense inflammation may result in pain, which manifests as spinal central sensitization. There is growing evidence that purinergic signaling plays a pivotal role in the orchestration of pain processing. Over the last decade the ionotropic P2X purino receptor 4 (P2X4) got into spotlight in neuropathic disorders, however its precise spinal expression was scantily characterized during inflammatory pain. Thus, we intended to analyze the receptor distribution within spinal dorsal horn and lumbar dorsal root ganglia (DRG) of rats suffering in inflammatory pain induced by complete Freund adjuvant (CFA).MethodsCFA-induced peripheral inflammation was validated by mechanical and thermal behavioral tests. In order to ensure about the putative alteration of spinal P2X4 receptor gene expression qPCR reactions were designed, followed by immunoperoxidase and Western blot experiments to assess changes at a protein level. Colocalization of P2X4 with neuronal and glial markers was investigated by double immunofluorescent labelings, which were subsequently analyzed with IMARIS software. Transmission electronmicroscopy was applied to study the ultrastructural localization of the receptor. Concurrently, in lumbar DRG cells similar methodology has been carried out to complete our observations.ResultsThe figures of mechanical and thermal behavioral tests proved the establishment of CFA-induced inflammatory pain. We observed significant enhancement of P2X4 transcript level within the spinal dorsal horn 3 days upon CFA administration. Elevation of P2X4 immunoreactivity within Rexed lamina I-II of the spinal gray matter was synchronous with mRNA expression, and confirmed by protein blotting. According to IMARIS analysis the robust protein increase was mainly detected on primary afferent axonterminals and GFAP-labelled astrocyte membrane compartments, but not on postsynaptic dendrites was also validated ultrastructurally within the spinal dorsal horn. Furthermore, lumbar DRG analysis demonstrated that peptidergic and non-peptidergic nociceptive subsets of ganglia cells were also abundantly positive for P2X4 receptor in CFA model.ConclusionHere we provide novel evidence about involvement of neuronal and glial P2X4 receptor in the establishment of inflammatory pain.
Collapse
|
11
|
Temmermand R, Barrett JE, Fontana ACK. Glutamatergic systems in neuropathic pain and emerging non-opioid therapies. Pharmacol Res 2022; 185:106492. [PMID: 36228868 PMCID: PMC10413816 DOI: 10.1016/j.phrs.2022.106492] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/05/2022] [Accepted: 10/06/2022] [Indexed: 01/14/2023]
Abstract
Neuropathic pain, a disease of the somatosensory nervous system, afflicts many individuals and adequate management with current pharmacotherapies remains elusive. The glutamatergic system of neurons, receptors and transporters are intimately involved in pain but, to date, there have been few drugs developed that therapeutically modulate this system. Glutamate transporters, or excitatory amino acid transporters (EAATs), remove excess glutamate around pain transmitting neurons to decrease nociception suggesting that the modulation of glutamate transporters may represent a novel approach to the treatment of pain. This review highlights and summarizes (1) the physiology of the glutamatergic system in neuropathic pain, (2) the preclinical evidence for dysregulation of glutamate transport in animal pain models, and (3) emerging novel therapies that modulate glutamate transporters. Successful drug discovery requires continuous focus on basic and translational methods to fully elucidate the etiologies of this disease to enable the development of targeted therapies. Increasing the efficacy of astrocytic EAATs may serve as a new way to successfully treat those suffering from this devastating disease.
Collapse
Affiliation(s)
- Rhea Temmermand
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, USA
| | - James E Barrett
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Andréia C K Fontana
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, USA.
| |
Collapse
|
12
|
Patra PH, Tench B, Hitrec T, Holmes F, Drake R, Cerritelli S, Spanswick D, Pickering AE. Pro-Opiomelanocortin (POMC) neurons in the nucleus of the solitary tract mediate endorphinergic endogenous analgesia in mice. Pain 2022; 164:1051-1066. [PMID: 36448978 DOI: 10.1097/j.pain.0000000000002802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 09/27/2022] [Indexed: 12/02/2022]
Abstract
ABSTRACT The nucleus of the solitary tract (NTS) contains pro-opiomelanocortin (POMC) neurons which are one of the two major sources of β-endorphin in the brain. The functional role of these NTS POMC neurons in nociceptive and cardiorespiratory function is debated. We have shown that NTS POMC optogenetic activation produces bradycardia and transient apnoea in a working heart brainstem preparation and chemogenetic activation with an engineered ion channel (PSAM) produced opioidergic analgesia in vivo . To better define the role of the NTS POMC neurons in behaving animals, we adopted in vivo optogenetics (ChrimsonR) and excitatory/inhibitory chemogenetic DREADD (hM3Dq/hM4Di) strategies in POMC-Cre mice. We show that optogenetic activation of NTS POMC neurons produces time-locked, graded, transient bradycardia and bradypnoea in anaesthetised mice which is naloxone sensitive (1 mg/kg, i.p) suggesting a role of β-endorphin. Both optogenetic and chemogenetic activation of NTS POMC neurons produces sustained thermal analgesia in behaving mice which can be blocked by naloxone. It also produced analgesia in inflammatory pain (carrageenan) but not in a neuropathic pain model (tibial nerve transection). Inhibiting NTS POMC neurons does not produce any effect on basal nociception but inhibits stress-induced analgesia (unlike inhibition of arcuate POMC neurons). Activation of NTS POMC neuronal populations in conscious mice did not cause respiratory depression, anxiety or locomotor deficit (in open field) nor affective preference. These findings indicate that NTS POMC neurons play a key role in the generation of endorphinergic endogenous analgesia and can also regulate cardiorespiratory function.
Collapse
Affiliation(s)
- Pabitra Hriday Patra
- Anaesthesia, Pain & Critical Care Research, School of Physiology, Pharmacology and Neuroscience, University of Bristol, BS8 1TD, UK
| | - Becks Tench
- Anaesthesia, Pain & Critical Care Research, School of Physiology, Pharmacology and Neuroscience, University of Bristol, BS8 1TD, UK
| | - Timna Hitrec
- Anaesthesia, Pain & Critical Care Research, School of Physiology, Pharmacology and Neuroscience, University of Bristol, BS8 1TD, UK
| | - Fiona Holmes
- Anaesthesia, Pain & Critical Care Research, School of Physiology, Pharmacology and Neuroscience, University of Bristol, BS8 1TD, UK
| | - Robert Drake
- Anaesthesia, Pain & Critical Care Research, School of Physiology, Pharmacology and Neuroscience, University of Bristol, BS8 1TD, UK
| | - Serena Cerritelli
- Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - David Spanswick
- Neurosolutions, University of Warwick, Gibbet Hill Road, Coventry, West Midlands, CV4 7AL, UK
| | - Anthony Edward Pickering
- Anaesthesia, Pain & Critical Care Research, School of Physiology, Pharmacology and Neuroscience, University of Bristol, BS8 1TD, UK
| |
Collapse
|
13
|
Mattosinhos PDS, Sarandy MM, Novaes RD, Esposito D, Gonçalves RV. Anti-Inflammatory, Antioxidant, and Skin Regenerative Potential of Secondary Metabolites from Plants of the Brassicaceae Family: A Systematic Review of In Vitro and In Vivo Preclinical Evidence (Biological Activities Brassicaceae Skin Diseases). Antioxidants (Basel) 2022; 11:1346. [PMID: 35883837 PMCID: PMC9312357 DOI: 10.3390/antiox11071346] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/21/2022] [Accepted: 06/24/2022] [Indexed: 01/08/2023] Open
Abstract
The Brassicaceae family constitutes some of the most well-studied natural products in the world, due to their anti-inflammatory, anti-oxidative, and pro-regenerative properties as well as their ubiquitous distribution across the world. To evaluate the potential efficacy of the Brassicaceae family in the treatment of inflammatory skin disorders and wounds, based on preclinical evidence from in vivo and in vitro studies. This systematic review was performed according to the PRISMA guidelines, using a structured search on the PubMed-Medline, Scopus, and Web of Science platforms. The studies included were those that used murine models and in vitro studies to investigate the effect of Brassicaceae on skin disorders. Bias analysis and methodological quality assessments were examined through SYRCLE's RoB tool. Brassicaceae have shown positive impacts on inflammatory regulation of the skin, accelerating the wound healing process, and inhibiting the development of edema. The studies showed that the Brassicaceae family has antioxidant activity and effects on the modulation of cyclooxygenase 2 and the nuclear factor kappa β (NFκβ) pathway. The secondary metabolites present in Brassicas are polyphenols (68.75%; n = 11), terpenes/carotenoids (31.25%; n = 5), and glycosylates (25%; n = 4), which are responsible for their anti-inflammatory, healing, and antioxidant effects. In addition, the current evidence is reliable because the bias analysis showed a low risk of bias. Our review indicates that compounds derived from Brassicaceae present exceptional potential to treat inflammatory skin diseases and accelerate cutaneous wound healing. We hope that our critical analysis can help to expedite clinical research and to reduce methodological bias, thereby improving the quality of evidence in future research. The registration number on the Prospero platform is CRD42021262953.
Collapse
Affiliation(s)
| | - Mariáurea Matias Sarandy
- Department of General Biology, Federal University of Vicosa, Vicosa 36570-900, MG, Brazil; (P.d.S.M.); (M.M.S.)
| | - Rômulo Dias Novaes
- Department of Structural Biology, Federal University of Alfenas, Alfenas 37130-001, MG, Brazil;
| | - Debora Esposito
- Plants for Human Health Institute, North Carolina Research Campus, NC State University, 600 Laureate Way, Kannapolis, NC 28081, USA;
- Department of Animal Science, NC State University, 120 Broughton Drive, Raleigh, NC 27695, USA
| | - Reggiani Vilela Gonçalves
- Department of General Biology, Federal University of Vicosa, Vicosa 36570-900, MG, Brazil; (P.d.S.M.); (M.M.S.)
- Plants for Human Health Institute, North Carolina Research Campus, NC State University, 600 Laureate Way, Kannapolis, NC 28081, USA;
| |
Collapse
|
14
|
Omura CM, Lüdtke DD, Horewicz VV, Fernandes PF, Galassi TDO, Salgado ASI, Palandi J, Baldança HDS, Bittencourt EB, Mack JM, Seim LA, Martins DF, Bobinski F. Decrease of IL-1β and TNF in the Spinal Cord Mediates Analgesia Produced by Ankle Joint Mobilization in Complete Freund Adjuvant-Induced Inflammation Mice Model. Front Physiol 2022; 12:816624. [PMID: 35095573 PMCID: PMC8795789 DOI: 10.3389/fphys.2021.816624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 12/24/2021] [Indexed: 01/26/2023] Open
Abstract
Objective This study aims to investigate the effects of ankle joint mobilization (AJM) on mechanical hyperalgesia and peripheral and central inflammatory biomarkers after intraplantar (i.pl.) Complete Freund’s Adjuvant (CFA)-induced inflammation. Methods Male Swiss mice were randomly assigned to 3 groups (n = 7): Saline/Sham, CFA/Sham, and CFA/AJM. Five AJM sessions were carried out at 6, 24, 48, 72, and 96 h after CFA injection. von Frey test was used to assess mechanical hyperalgesia. Tissues from paw skin, paw muscle and spinal cord were collected to measure pro-inflammatory (TNF, IL-1β) and anti-inflammatory cytokines (IL-4, IL-10, and TGF-β1) by ELISA. The macrophage phenotype at the inflammation site was evaluated by Western blotting assay using the Nitric Oxide Synthase 2 (NOS 2) and Arginase-1 immunocontent to identify M1 and M2 macrophages, respectively. Results Our results confirm a consistent analgesic effect of AJM following the second treatment session. AJM did not change cytokines levels at the inflammatory site, although it promoted a reduction in M2 macrophages. Also, there was a reduction in the levels of pro-inflammatory cytokines IL-1β and TNF in the spinal cord. Conclusion Taken together, the results confirm the anti-hyperalgesic effect of AJM and suggest a central neuroimmunomodulatory effect in a model of persistent inflammation targeting the pro-inflammatory cytokines IL-1β and TNF.
Collapse
Affiliation(s)
- Carlos Minoru Omura
- Experimental Neuroscience Laboratory (LaNEx), Graduate Program in Health Sciences, University of Southern Santa Catarina, Palhoça, Brazil
| | - Daniela Dero Lüdtke
- Experimental Neuroscience Laboratory (LaNEx), Graduate Program in Health Sciences, University of Southern Santa Catarina, Palhoça, Brazil
- Faculty of Physical Therapy, University of Southern Santa Catarina, Palhoça, Brazil
| | - Verônica Vargas Horewicz
- Experimental Neuroscience Laboratory (LaNEx), Graduate Program in Health Sciences, University of Southern Santa Catarina, Palhoça, Brazil
| | - Paula Franson Fernandes
- Experimental Neuroscience Laboratory (LaNEx), Graduate Program in Health Sciences, University of Southern Santa Catarina, Palhoça, Brazil
| | - Taynah de Oliveira Galassi
- Experimental Neuroscience Laboratory (LaNEx), Graduate Program in Health Sciences, University of Southern Santa Catarina, Palhoça, Brazil
| | | | - Juliete Palandi
- Experimental Neuroscience Laboratory (LaNEx), Graduate Program in Health Sciences, University of Southern Santa Catarina, Palhoça, Brazil
- Laboratory of Experimentation in Neuropathology (LEN), Graduate Program in Neuroscience, Department of Biochemistry, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Heloiza dos Santos Baldança
- Experimental Neuroscience Laboratory (LaNEx), Graduate Program in Health Sciences, University of Southern Santa Catarina, Palhoça, Brazil
- Faculty of Physical Therapy, University of Southern Santa Catarina, Palhoça, Brazil
| | | | - Josiel Mileno Mack
- Experimental Neuroscience Laboratory (LaNEx), Graduate Program in Health Sciences, University of Southern Santa Catarina, Palhoça, Brazil
- Graduate Program in Medical Sciences, Department of Medical Clinic, Federal University of Santa Catarina (UFSC), Florianopolis, Brazil
- Faculty of Medicine, University of Southern Santa Catarina, Palhoça, Brazil
| | - Lynsey A. Seim
- Department of Hospital Internal Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Daniel Fernandes Martins
- Experimental Neuroscience Laboratory (LaNEx), Graduate Program in Health Sciences, University of Southern Santa Catarina, Palhoça, Brazil
| | - Franciane Bobinski
- Experimental Neuroscience Laboratory (LaNEx), Graduate Program in Health Sciences, University of Southern Santa Catarina, Palhoça, Brazil
- *Correspondence: Franciane Bobinski,
| |
Collapse
|
15
|
Tmem160 contributes to the establishment of discrete nerve injury-induced pain behaviors in male mice. Cell Rep 2021; 37:110152. [PMID: 34936870 DOI: 10.1016/j.celrep.2021.110152] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 09/01/2021] [Accepted: 11/30/2021] [Indexed: 12/26/2022] Open
Abstract
Chronic pain is a prevalent medical problem, and its molecular basis remains poorly understood. Here, we demonstrate the significance of the transmembrane protein (Tmem) 160 for nerve injury-induced neuropathic pain. An extensive behavioral assessment suggests a pain modality- and entity-specific phenotype in male Tmem160 global knockout (KO) mice: delayed establishment of tactile hypersensitivity and alterations in self-grooming after nerve injury. In contrast, Tmem160 seems to be dispensable for other nerve injury-induced pain modalities, such as non-evoked and movement-evoked pain, and for other pain entities. Mechanistically, we show that global KO males exhibit dampened neuroimmune signaling and diminished TRPA1-mediated activity in cultured dorsal root ganglia. Neither these changes nor altered pain-related behaviors are observed in global KO female and male peripheral sensory neuron-specific KO mice. Our findings reveal Tmem160 as a sexually dimorphic factor contributing to the establishment, but not maintenance, of discrete nerve injury-induced pain behaviors in male mice.
Collapse
|