1
|
Meng K, Zhao Z, Gao Y, Wu K, Liu W, Wang X, Zheng Y, Zhao W, Wang B. The synergistic effects of anoikis-related genes and EMT-related genes in the prognostic prediction of Wilms tumor. Front Mol Biosci 2024; 11:1469775. [PMID: 39351154 PMCID: PMC11439783 DOI: 10.3389/fmolb.2024.1469775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 09/03/2024] [Indexed: 10/04/2024] Open
Abstract
Wilms tumor (WT) is the most common type of malignant abdominal tumor in children; it exhibits a high degree of malignancy, grow rapidly, and is prone to metastasis. This study aimed to construct a prognosis model based on anoikis-related genes (ARGs) and epithelial-mesenchymal transition (EMT)-related genes (ERGs) for WT patients; we assessed the characteristics of the tumor microenvironment and treatment efficacy, as well as identifying potential therapeutic targets. To this end, we downloaded transcriptome sequencing data and clinical data for WT and normal renal cortices and used R to construct and validate the prognostic model based on ARGs and ERGs. Additionally, we performed clinical feature analysis, nomogram construction, mutation analysis, drug sensitivity analysis, Connectivity Map (cMAP) analysis, functional enrichment analysis, and immune infiltration analysis. Finally, we screened the hub gene using the STRING database and validated it via experiments. In this way, we constructed a model with good accuracy and robustness, which was composed of seven anoikis- and EMT-related genes. Paclitaxel and mesna were selected as potential chemotherapeutic drugs and adjuvant chemotherapeutic drugs for the WT high-risk group by using the Genomics of Drug Sensitivity in Cancer (GDSC) and cMAP compound libraries, respectively. We proved the existence of a strong correlation between invasive immune cells and prognostic genes and risk scores. Next, we selected NTRK2 as the hub gene, and in vitro experiments confirmed that its inhibition can significantly inhibit the proliferation and migration of tumor cells and promote late apoptosis. In summary, we screened out the potential biomarkers and chemotherapeutic drugs that can improve the prognosis of patients with WT.
Collapse
Affiliation(s)
- Kexin Meng
- Department of Medical Ultrasound, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Medical and Health Key Laboratory of Abdominal Medical Imaging, Jinan, China
| | - Zerui Zhao
- Department of Clinical Pharmacy, Clinical Trial Center, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Yaqing Gao
- Department of Medical Ultrasound, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Medical and Health Key Laboratory of Abdominal Medical Imaging, Jinan, China
| | - Keliang Wu
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Wei Liu
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xiaoqing Wang
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yi Zheng
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Wei Zhao
- Department of Clinical Pharmacy, Clinical Trial Center, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Bei Wang
- Department of Medical Ultrasound, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Medical and Health Key Laboratory of Abdominal Medical Imaging, Jinan, China
| |
Collapse
|
2
|
Li L, Yao H, Mo R, Xu L, Chen P, Chen Y, Hu JJ, Xie W, Song XJ. Blocking proteinase-activated receptor 2 signaling relieves pain, suppresses nerve sprouting, improves tissue repair, and enhances analgesic effect of B vitamins in rats with Achilles tendon injury. Pain 2024; 165:2055-2067. [PMID: 38598349 DOI: 10.1097/j.pain.0000000000003229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 02/02/2024] [Indexed: 04/12/2024]
Abstract
ABSTRACT Tendon injury produces intractable pain and disability in movement, but the medications for analgesia and restoring functional integrity of tendon are still limited. In this study, we report that proteinase-activated receptor 2 (PAR2) activation in dorsal root ganglion (DRG) neurons contributes to chronic pain and tendon histopathological changes produced by Achilles tendon partial transection injury (TTI). Tendon partial transection injury increases the expression of PAR2 protein in both somata of DRG neurons and their peripheral terminals within the injured Achilles tendon. Activation of PAR2 promotes the primary sensory neuron plasticity by activating downstream cAMP-PKA pathway, phosphorylation of PKC, CaMKII, and CREB. Blocking PAR2 signaling by PAR2 small-interference RNA or antagonistic peptide PIP delays the onset of TTI-induced pain, reverses the ongoing pain, as well as inhibits sensory nerve sprouting, and promotes structural remodeling of the injured tendon. Vitamin B complex (VBC), containing thiamine (B1), pyridoxine (B6), and cyanocobalamin (B12), is effective to ameliorate TTI-induced pain, inhibit ectopic nerve sprouting, and accelerate tendon repair, through suppressing PAR2 activation. These findings reveal a critical role of PAR2 signaling in the development of chronic pain and histopathological alterations of injured tendon following Achilles tendon injury. This study suggests that the pharmaceuticals targeting PAR2, such as VBC, may be an effective approach for the treatment of tendon injury-induced pain and promoting tendon repair.
Collapse
Affiliation(s)
- Lihui Li
- Department of Medical Neuroscience, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- SUSTech Center for Pain Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Hongyu Yao
- Department of Medical Neuroscience, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- SUSTech Center for Pain Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Rufan Mo
- SUSTech Center for Pain Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Lihong Xu
- Department of Medical Neuroscience, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Peng Chen
- Department of Medical Neuroscience, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- SUSTech Center for Pain Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Yuchen Chen
- Department of Medical Neuroscience, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Jiang-Jian Hu
- Department of Medical Neuroscience, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- SUSTech Center for Pain Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Wei Xie
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, The School of Life Science and Technology, Southeast University, Nanjing, China
| | - Xue-Jun Song
- Department of Medical Neuroscience, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- SUSTech Center for Pain Medicine, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
3
|
Reches G, Piran R. Par2-mediated responses in inflammation and regeneration: choosing between repair and damage. Inflamm Regen 2024; 44:26. [PMID: 38816842 PMCID: PMC11138036 DOI: 10.1186/s41232-024-00338-1] [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: 01/14/2024] [Accepted: 05/19/2024] [Indexed: 06/01/2024] Open
Abstract
The protease activated receptor 2 (Par2) plays a pivotal role in various damage models, influencing injury, proliferation, inflammation, and regeneration. Despite extensive studies, its binary roles- EITHER aggravating injury or promoting recovery-make a conclusive translational decision on its modulation strategy elusive. Analyzing two liver regeneration models, autoimmune hepatitis and direct hepatic damage, we discovered Par2's outcome depends on the injury's nature. In immune-mediated injury, Par2 exacerbates damage, while in direct tissue injury, it promotes regeneration. Subsequently, we evaluated the clinical significance of this finding by investigating Par2's expression in the context of autoimmune diabetes. We found that the absence of Par2 in all lymphocytes provided full protection against the autoimmune destruction of insulin-producing β-cells in mice, whereas the introduction of a β-cell-specific Par2 null mutation accelerated the onset of autoimmune diabetes. This pattern led us to hypothesize whether these observations are universal. A comprehensive review of recent Par2 publications across tissues and systems confirms the claim drafted above: Par2's initial activation in the immune system aggravates inflammation, hindering recovery, whereas its primary activation in the damaged tissue fosters regeneration. As a membrane-anchored receptor, Par2 emerges as an attractive drug target. Our findings highlight a crucial translational modulation strategy in regenerative medicine based on injury type.
Collapse
Affiliation(s)
- Gal Reches
- The Azrieli Faculty of Medicine, Bar-Ilan University, 8 Henrietta Szold St, Safed, Israel
| | - Ron Piran
- The Azrieli Faculty of Medicine, Bar-Ilan University, 8 Henrietta Szold St, Safed, Israel.
| |
Collapse
|
4
|
Merighi A. Brain-Derived Neurotrophic Factor, Nociception, and Pain. Biomolecules 2024; 14:539. [PMID: 38785946 PMCID: PMC11118093 DOI: 10.3390/biom14050539] [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: 02/08/2024] [Revised: 04/26/2024] [Accepted: 04/26/2024] [Indexed: 05/25/2024] Open
Abstract
This article examines the involvement of the brain-derived neurotrophic factor (BDNF) in the control of nociception and pain. BDNF, a neurotrophin known for its essential role in neuronal survival and plasticity, has garnered significant attention for its potential implications as a modulator of synaptic transmission. This comprehensive review aims to provide insights into the multifaceted interactions between BDNF and pain pathways, encompassing both physiological and pathological pain conditions. I delve into the molecular mechanisms underlying BDNF's involvement in pain processing and discuss potential therapeutic applications of BDNF and its mimetics in managing pain. Furthermore, I highlight recent advancements and challenges in translating BDNF-related research into clinical practice.
Collapse
Affiliation(s)
- Adalberto Merighi
- Department of Veterinary Sciences, University of Turin, 10095 Turin, Italy
| |
Collapse
|
5
|
Altier C. Breaking Down the Pain Pathway: Bacterial Proteases Activate Nociceptors to Cause Pain. Cell Mol Gastroenterol Hepatol 2024; 18:101337. [PMID: 38583482 PMCID: PMC11440130 DOI: 10.1016/j.jcmgh.2024.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 03/18/2024] [Indexed: 04/09/2024]
Affiliation(s)
- Christophe Altier
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Inflammation Research Network-Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
6
|
Li L, Li ZE, Mo YL, Li WY, Li HJ, Yan GH, Qin XZ, Piao LH. Molecular and cellular pruritus mechanisms in the host skin. Exp Mol Pathol 2024; 136:104889. [PMID: 38316203 DOI: 10.1016/j.yexmp.2024.104889] [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: 03/29/2023] [Revised: 12/28/2023] [Accepted: 01/31/2024] [Indexed: 02/07/2024]
Abstract
Pruritus, also known as itching, is a complex sensation that involves the activation of specific physiological and cellular receptors. The skin is innervated with sensory nerves as well as some receptors for various sensations, and its immune system has prominent neurological connections. Sensory neurons have a considerable impact on the sensation of itching. However, immune cells also play a role in this process, as they release pruritogens. Disruption of the dermal barrier activates an immune response, initiating a series of chemical, physical, and cellular reactions. These reactions involve various cell types, including keratinocytes, as well as immune cells involved in innate and adaptive immunity. Collective activation of these immune responses confers protection against potential pathogens. Thus, understanding the molecular and cellular mechanisms that contribute to pruritus in host skin is crucial for the advancement of effective treatment approaches. This review provides a comprehensive analysis of the present knowledge concerning the molecular and cellular mechanisms underlying itching signaling in the skin. Additionally, this review explored the integration of these mechanisms with the broader context of itch mediators and the expression of their receptors in the skin.
Collapse
Affiliation(s)
- Li Li
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji 133002, China; Department of Anatomy, Yanbian University Medical College, Yanji 133002, China
| | - Zhi-En Li
- Clinical Medicine, Yanbian University Medical College, Yanji 133002, China
| | - Yun-Li Mo
- Clinical Medicine, Yanbian University Medical College, Yanji 133002, China
| | - Wan-Yao Li
- Clinical Medicine, Yanbian University Medical College, Yanji 133002, China
| | - Hui-Jing Li
- Clinical Medicine, Yanbian University Medical College, Yanji 133002, China
| | - Guang-Hai Yan
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji 133002, China; Department of Anatomy, Yanbian University Medical College, Yanji 133002, China
| | - Xiang-Zheng Qin
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji 133002, China; Department of Anatomy, Yanbian University Medical College, Yanji 133002, China.
| | - Li-Hua Piao
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji 133002, China; Department of Anatomy, Yanbian University Medical College, Yanji 133002, China.
| |
Collapse
|
7
|
Inyang KE, Evans CM, Heussner M, Petroff M, Reimers M, Vermeer PD, Tykocki N, Folger JK, Laumet G. HPV+ head and neck cancer-derived small extracellular vesicles communicate with TRPV1+ neurons to mediate cancer pain. Pain 2024; 165:608-620. [PMID: 37678566 PMCID: PMC10915104 DOI: 10.1097/j.pain.0000000000003045] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 06/15/2023] [Indexed: 09/09/2023]
Abstract
ABSTRACT Severe pain is often experienced by patients with head and neck cancer and is associated with a poor prognosis. Despite its frequency and severity, current treatments fail to adequately control cancer-associated pain because of our lack of mechanistic understanding. Although recent works have shed some light of the biology underlying pain in HPV-negative oral cancers, the mechanisms mediating pain in HPV+ cancers remain unknown. Cancer-derived small extracellular vesicles (cancer-sEVs) are well positioned to function as mediators of communication between cancer cells and neurons. Inhibition of cancer-sEV release attenuated pain in tumor-bearing mice. Injection of purified cancer-sEVs is sufficient to induce pain hypersensitivity in naive mice that is prevented by QX-314 treatment and in Trpv1-/- mice. Cancer-sEVs triggered calcium influx in nociceptors, and inhibition or ablation of nociceptors protects against cancer pain. Interrogation of published sequencing data of human sensory neurons exposed to human cancer-sEVs suggested a stimulation of protein translation in neurons. Induction of translation by cancer-sEVs was validated in our mouse model, and its inhibition alleviated cancer pain in mice. In summary, our work reveals that HPV+ head and neck squamous cell carcinoma-derived sEVs alter TRPV1+ neurons by promoting nascent translation to mediate cancer pain and identified several promising therapeutic targets to interfere with this pathway.
Collapse
Affiliation(s)
| | - Christine M. Evans
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Matthew Heussner
- Department of Physiology, Michigan State University, East Lansing, MI, USA
- College of Osteopathic Medicine, Michigan State University, East Lansing, MI
| | - Margaret Petroff
- Department of Pathology Michigan State University College of Veterinary Medicine, East Lansing, MI
| | - Mark Reimers
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Paola D. Vermeer
- Cancer Biology and Immunotherapies Group, Sanford Research, Sioux Falls, South Dakota
| | - Nathan Tykocki
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI
| | - Joseph K. Folger
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Geoffroy Laumet
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
8
|
Qian Z, Zhang M, Lu T, Yu J, Yin S, Wang H, Wang J. Propolis alleviates ulcerative colitis injury by inhibiting the protein kinase C - transient receptor potential cation channel subfamily V member 1 - calcitonin gene-related peptide/substance P (PKC-TRPV1-CGRP/SP) signaling axis. PLoS One 2024; 19:e0294169. [PMID: 38206948 PMCID: PMC10783729 DOI: 10.1371/journal.pone.0294169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 10/27/2023] [Indexed: 01/13/2024] Open
Abstract
This study investigated the protective effect of water-soluble propolis (WSP) on colonic tissues in ulcerative colitis (UC) and the role of the protein kinase C - transient receptor potential cation channel subfamily V member 1 - calcitonin gene-related peptide/substance P (PKC-TRPV1-CGRP/SP) signaling pathway. Male SD rats were divided into a control group, a UC model group, various WSP groups (Low-WSP, Medium-WSP, and High-WSP) with UC, and a salazosulfapyridine (SASP) positive control group with UC. After UC was established, the WSP and SASP groups were treated with WSP or SASP, respectively, for 7 d. Each day, body weight measurements were obtained, and the disease activity index (DAI) was recorded by observing fecal characteristics and blood in the stool. After the experiment, hematoxylin and eosin (HE) colonic tissue staining was performed to observe pathological changes, western blotting and immunohistochemistry were performed to detect PKC, TRPV1, CGRP, and SP expression in colonic tissues, and laser confocal microscopy was performed to observe the fluorescence colocalization of PKC/TRPV1, TRPV1/CGRP, and TRPV1/SP. HE staining showed significant colonic tissue structure disruption and inflammatory infiltration in the UC group. Western blotting and immunohistochemistry showed that the expression of PKC, TRPV1, CGRP, and SP in the colonic tissues of the UC group increased significantly compared with that of the control group. Compared with the UC group, the expression of PKC, TRPV1, CGRP, and SP in colonic tissues was significantly reduced in the High-WSP, Medium-WSP, and SASP groups. Immunofluorescence showed the colocalized expression of PKC/TRPV1, TRPV1/CGRP, and TRPV1/SP proteins in the colon tissue of the UC group was significantly reduced after WSP and SASP interventions compared with that of the control group. The results suggest that the mechanism of UC alleviation by propolis may inhibit the PKC-TRPV1-CGRP/SP signaling pathway and the release of inflammatory mediators, thus alleviating inflammation.
Collapse
Affiliation(s)
- Zhen Qian
- School of Clinical Medicine, Wannan Medical College, Wuhu, Anhui province, China
| | - Mengjie Zhang
- Graduate School, Wannan Medical College, Wuhu, Anhui province, China
| | - Taiyu Lu
- School of Clinical Medicine, Wannan Medical College, Wuhu, Anhui province, China
| | - Jiayi Yu
- School of Clinical Medicine, Wannan Medical College, Wuhu, Anhui province, China
| | - Siyuan Yin
- School of Medical Imageology, Wannan Medical College, Wuhu, Anhui province, China
| | - Haihua Wang
- Department of Physiology, School of Basic Medical Sciences, Wannan Medical College, Wuhu, Anhui province, China
| | - Jing Wang
- Department of Physiology, School of Basic Medical Sciences, Wannan Medical College, Wuhu, Anhui province, China
| |
Collapse
|
9
|
Kume M, Ahmad A, DeFea KA, Vagner J, Dussor G, Boitano S, Price TJ. Protease-Activated Receptor 2 (PAR2) Expressed in Sensory Neurons Contributes to Signs of Pain and Neuropathy in Paclitaxel Treated Mice. THE JOURNAL OF PAIN 2023; 24:1980-1993. [PMID: 37315729 PMCID: PMC10615692 DOI: 10.1016/j.jpain.2023.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 05/26/2023] [Accepted: 06/07/2023] [Indexed: 06/16/2023]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a common, dose-limiting side effect of cancer therapy. Protease-activated receptor 2 (PAR2) is implicated in a variety of pathologies, including CIPN. In this study, we demonstrate the role of PAR2 expressed in sensory neurons in a paclitaxel (PTX)-induced model of CIPN in mice. PAR2 knockout/wildtype (WT) mice and mice with PAR2 ablated in sensory neurons were treated with PTX administered via intraperitoneal injection. In vivo behavioral studies were done in mice using von Frey filaments and the Mouse Grimace Scale. We then examined immunohistochemical staining of dorsal root ganglion (DRG) and hind paw skin samples from CIPN mice to measure satellite cell gliosis and intra-epidermal nerve fiber (IENF) density. The pharmacological reversal of CIPN pain was tested with the PAR2 antagonist C781. Mechanical allodynia caused by PTX treatment was alleviated in PAR2 knockout mice of both sexes. In the PAR2 sensory neuronal conditional knockout (cKO) mice, both mechanical allodynia and facial grimacing were attenuated in mice of both sexes. In the DRG of the PTX-treated PAR2 cKO mice, satellite glial cell activation was reduced compared to control mice. IENF density analysis of the skin showed that the PTX-treated control mice had a reduction in nerve fiber density while the PAR2 cKO mice had a comparable skin innervation as the vehicle-treated animals. Similar results were seen with satellite cell gliosis in the DRG, where gliosis induced by PTX was absent in PAR cKO mice. Finally, C781 was able to transiently reverse established PTX-evoked mechanical allodynia. PERSPECTIVE: Our work demonstrates that PAR2 expressed in sensory neurons plays a key role in PTX-induced mechanical allodynia, spontaneous pain, and signs of neuropathy, suggesting PAR2 as a possible therapeutic target in multiple aspects of PTX CIPN.
Collapse
Affiliation(s)
- Moeno Kume
- University of Texas at Dallas, Department of Neuroscience and Center for Advanced Pain Studies
| | - Ayesha Ahmad
- University of Texas at Dallas, Department of Neuroscience and Center for Advanced Pain Studies
| | | | | | - Gregory Dussor
- University of Texas at Dallas, Department of Neuroscience and Center for Advanced Pain Studies
| | - Scott Boitano
- University of Arizona Bio5 Research Institute
- University of Arizona Heath Sciences, Asthma and Airway Disease Research Center
- University of Arizona Heath Sciences, Department of Physiology
| | - Theodore J. Price
- University of Texas at Dallas, Department of Neuroscience and Center for Advanced Pain Studies
| |
Collapse
|
10
|
Mason BN, Hassler SN, DeFea K, Boitano S, Vagner J, Price TJ, Dussor G. PAR2 activation in the dura causes acute behavioral responses and priming to glyceryl trinitrate in a mouse migraine model. J Headache Pain 2023; 24:42. [PMID: 37072694 PMCID: PMC10114383 DOI: 10.1186/s10194-023-01574-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 04/03/2023] [Indexed: 04/20/2023] Open
Abstract
BACKGROUND Migraine is a severely debilitating disorder that affects millions of people worldwide. Studies have indicated that activation of protease-activated receptor-2 (PAR2) in the dura mater causes headache responses in preclinical models. It is also well known that vasodilators such as nitric oxide (NO) donors can trigger migraine attacks in migraine patients but not controls. In the current study we examined whether activation of PAR2 in the dura causes priming to the NO donor glyceryl trinitrate (GTN). METHODS A preclinical behavioral model of migraine was used where stimuli (PAR2 agonists: 2at-LIGRL-NH2 (2AT) or neutrophil elastase (NE); and IL-6) were applied to the mouse dura through an injection made at the intersection of the lamdoidal and sagittal sutures on the skull. Following dural injection, periorbital von Frey thresholds and facial grimace responses were measured until their return to baseline. GTN was then given by intraperitoneal injection and periorbital hypersensitivity and facial grimace responses observed until they returned to baseline. RESULTS We found that application of the selective PAR2 agonist 2at-LIGRL-NH2 (2AT) onto the dura causes headache-related behavioral responses in WT but not PAR2-/- mice with no differences between sexes. Additionally, dural PAR2 activation with 2AT caused priming to GTN (1 mg/kg) at 14 days after primary dural stimulation. PAR2-/- mice showed no priming to GTN. We also tested behavioral responses to the endogenous protease neutrophil elastase, which can cleave and activate PAR2. Dural neutrophil elastase caused both acute responses and priming to GTN in WT but not PAR2-/- mice. Finally, we show that dural IL-6 causes acute responses and priming to GTN that is identical in WT and PAR2-/- mice, indicating that IL-6 does not act through PAR2 in this model. CONCLUSIONS These results indicate that PAR2 activation in the meninges can cause acute headache behavioral responses and priming to an NO donor, and support further exploration of PAR2 as a novel therapeutic target for migraine.
Collapse
Affiliation(s)
- Bianca N Mason
- Department of Neuroscience, University of Texas at Dallas, 800 West Campbell Rd, Richardson, TX, 75080, USA
- Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Shayne N Hassler
- Department of Neuroscience, University of Texas at Dallas, 800 West Campbell Rd, Richardson, TX, 75080, USA
| | | | - Scott Boitano
- Department of Physiology, University of Arizona, Tucson, 85724, USA
| | - Josef Vagner
- Bio5 Institute, University of Arizona, Tucson, AZ, 85724, USA
| | - Theodore J Price
- Department of Neuroscience, University of Texas at Dallas, 800 West Campbell Rd, Richardson, TX, 75080, USA
- Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Greg Dussor
- Department of Neuroscience, University of Texas at Dallas, 800 West Campbell Rd, Richardson, TX, 75080, USA.
- Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, 75080, USA.
| |
Collapse
|
11
|
Kume M, Ahmad A, Shiers S, Burton MD, DeFea KA, Vagner J, Dussor G, Boitano S, Price TJ. C781, a β-Arrestin Biased Antagonist at Protease-Activated Receptor-2 (PAR2), Displays in vivo Efficacy Against Protease-Induced Pain in Mice. THE JOURNAL OF PAIN 2023; 24:605-616. [PMID: 36417966 PMCID: PMC10079573 DOI: 10.1016/j.jpain.2022.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/02/2022] [Accepted: 11/09/2022] [Indexed: 11/21/2022]
Abstract
Given the limited options and often harmful side effects of current analgesics and the suffering caused by the opioid crisis, new classes of pain therapeutics are needed. Protease-activated receptors (PARs), particularly PAR2, are implicated in a variety of pathologies, including pain. Since the discovery of the role of PAR2 in pain, development of potent and specific antagonists has been slow. In this study, we describe the in vivo characterization of a novel small molecule/peptidomimetic hybrid compound, C781, as a β-arrestin-biased PAR2 antagonist. In vivo behavioral studies were done in mice using von Frey filaments and the Mouse Grimace Scale. Pharmacokinetic studies were done to assess pharmacokinetic/pharmacodynamic relationship in vivo. We used both prevention and reversal paradigms with protease treatment to determine whether C781 could attenuate protease-evoked pain. C781 effectively prevented and reversed mechanical and spontaneous nociceptive behaviors in response to small molecule PAR2 agonists, mast cell activators, and neutrophil elastase. The ED50 of C781 (intraperitoneal dosing) for inhibition of PAR2 agonist (20.9 ng 2-AT)-evoked nociception was 6.3 mg/kg. C781 was not efficacious in the carrageenan inflammation model. Pharmacokinetic studies indicated limited long-term systemic bioavailability for C781 suggesting that optimizing pharmacokinetic properties could improve in vivo efficacy. Our work demonstrates in vivo efficacy of a biased PAR2 antagonist that selectively inhibits β-arrestin/MAPK signaling downstream of PAR2. Given the importance of this signaling pathway in PAR2-evoked nociception, C781 exemplifies a key pharmacophore for PAR2 that can be optimized for clinical development. PERSPECTIVE: Our work provides evidence that PAR2 antagonists that only block certain aspects of signaling by the receptor can be effective for blocking protease-evoked pain in mice. This is important because it creates a rationale for developing safer PAR2-targeting approaches for pain treatment.
Collapse
Affiliation(s)
- Moeno Kume
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, Texas
| | - Ayesha Ahmad
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, Texas
| | - Stephanie Shiers
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, Texas
| | - Michael D Burton
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, Texas
| | | | - Josef Vagner
- University of Arizona Bio5 Institute, Tucson, Arizona
| | - Gregory Dussor
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, Texas
| | - Scott Boitano
- University of Arizona Bio5 Institute, Tucson, Arizona; Asthma and Airway Disease Research Center, University of Arizona Heath Sciences, Tucson, Arizona; Department of Physiology, University of Arizona Heath Sciences, Tucson, Arizona
| | - Theodore J Price
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, Texas.
| |
Collapse
|
12
|
Peach CJ, Edgington-Mitchell LE, Bunnett NW, Schmidt BL. Protease-activated receptors in health and disease. Physiol Rev 2023; 103:717-785. [PMID: 35901239 PMCID: PMC9662810 DOI: 10.1152/physrev.00044.2021] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 07/08/2022] [Accepted: 07/10/2022] [Indexed: 11/22/2022] Open
Abstract
Proteases are signaling molecules that specifically control cellular functions by cleaving protease-activated receptors (PARs). The four known PARs are members of the large family of G protein-coupled receptors. These transmembrane receptors control most physiological and pathological processes and are the target of a large proportion of therapeutic drugs. Signaling proteases include enzymes from the circulation; from immune, inflammatory epithelial, and cancer cells; as well as from commensal and pathogenic bacteria. Advances in our understanding of the structure and function of PARs provide insights into how diverse proteases activate these receptors to regulate physiological and pathological processes in most tissues and organ systems. The realization that proteases and PARs are key mediators of disease, coupled with advances in understanding the atomic level structure of PARs and their mechanisms of signaling in subcellular microdomains, has spurred the development of antagonists, some of which have advanced to the clinic. Herein we review the discovery, structure, and function of this receptor system, highlight the contribution of PARs to homeostatic control, and discuss the potential of PAR antagonists for the treatment of major diseases.
Collapse
Affiliation(s)
- Chloe J Peach
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York
- Department of Neuroscience and Physiology and Neuroscience Institute, Grossman School of Medicine, New York University, New York, New York
| | - Laura E Edgington-Mitchell
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
- Bluestone Center for Clinical Research, Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, New York, New York
| | - Nigel W Bunnett
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York
- Department of Neuroscience and Physiology and Neuroscience Institute, Grossman School of Medicine, New York University, New York, New York
| | - Brian L Schmidt
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York
- Bluestone Center for Clinical Research, Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, New York, New York
| |
Collapse
|
13
|
赵 佳, 杨 荷, 王 招, 朱 海, 谢 敏. [ANA- 12 inhibits spinal inflammation and alleviates acute and chronic pain in rats by targeted blocking of BDNF/TrkB signaling]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2022; 42:232-237. [PMID: 35365447 PMCID: PMC8983361 DOI: 10.12122/j.issn.1673-4254.2022.02.09] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Indexed: 01/24/2023]
Abstract
OBJECTIVE To investigate the inhibitory effect of ANA-12 that blocks brain-derived neurotrophic factor (BDNF)/ tropomyosin receptor kinase B (TrkB) signaling on inflammatory pain in rats and explore the underlying mechanism. METHODS Forty-two adult SD rats were randomized into BDNF-induced acute pain group (n=24) and CFA-induced chronic pain group. The former group were randomly divided into 4 subgroups, including a control group, ANA-12 treatment group, BDNF treatment group, and BDNF+ANA-12 treatment group; the latter group were subgrouped into control group, CFA treatment group (CFA) and CFA + ANA-12 treatment group. The effects of ANA-12 treatment on pain behaviors of the rats with BDNF-induced acute pain and CFA-induced chronic inflammatory pain were observed. Western blotting was used to examine TrkB signaling and expressions of microglia marker protein Iba1 and TNF-α in the spinal cord of the rats. RESULTS BDNF injection into the subarachnoid space significantly increased the number of spontaneous paw withdrawal of the rats (P < 0.05), which was obviously reduced by ANA-12 treatment (P < 0.05). The rats with intraplantar injection of CFA, showed significantly increased ipsilateral mechanical stimulation sensitivity (P < 0.05), and ANA-12 treatment obviously increased the ipsilateral foot withdrawal threshold (P < 0.05). Treatment with either BDNF or CFA significantly increased the phosphorylation level of TrkB (Y705) in the spinal cord of the rats (P < 0.05), which was significantly lowered by ANA-12 treatment (P < 0.05). Treatment with BDNF and CFA both significantly up-regulated the expressions of Iba1 and TNF-α in the spinal cord (P < 0.05), but ANA-12 significantly reduced their expression levels (P < 0.05). CONCLUSION ANA-12 can reduce spinal cord inflammation and relieve acute and chronic pain in rats by targeted blocking of BDNF/TrkB signaling.
Collapse
Affiliation(s)
- 佳佳 赵
- />湖北科技学院药学院,湖北 咸宁 437100School of Pharmacy, Hubei University of Science and Technology, Xianning 437100, China
| | - 荷雨 杨
- />湖北科技学院药学院,湖北 咸宁 437100School of Pharmacy, Hubei University of Science and Technology, Xianning 437100, China
| | - 招娣 王
- />湖北科技学院药学院,湖北 咸宁 437100School of Pharmacy, Hubei University of Science and Technology, Xianning 437100, China
| | - 海丽 朱
- />湖北科技学院药学院,湖北 咸宁 437100School of Pharmacy, Hubei University of Science and Technology, Xianning 437100, China
| | - 敏 谢
- />湖北科技学院药学院,湖北 咸宁 437100School of Pharmacy, Hubei University of Science and Technology, Xianning 437100, China
| |
Collapse
|
14
|
Guo ZB, Tang L, Wang LP, Wu HH, Huang CL, Zhan MX, Shi ZM, Yang CL, Chen GZ, Zou YQ, Yang F, Wu XZ. The analgesic effects of ulinastatin either as a single agent or in combination with sufentanil: A novel therapeutic potential for postoperative pain. Eur J Pharmacol 2021; 907:174267. [PMID: 34146590 DOI: 10.1016/j.ejphar.2021.174267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 06/07/2021] [Accepted: 06/14/2021] [Indexed: 01/11/2023]
Abstract
Ulinastatin is a broad-spectrum protease inhibitor widely used for the treatment of various inflammation-related diseases owing to its recognized excellent anti-inflammatory and cytoprotective properties. However, whether ulinastatin can relieve postoperative pain remains unclear. In this study, we evaluated the analgesic effects of ulinastatin administered either as a single agent or in combination with sufentanil in a validated preclinical rat model of postoperative pain induced by plantar incision. We found that incisional surgery on the hind paw of these rats induced sustained ipsilateral mechanical pain hypersensitivity that lasted for at least 10 days. A single intraperitoneal (i.p.) injection of ulinastatin prevented the development and reversed the maintenance of incision-induced mechanical pain hypersensitivity in a dose-dependent manner. However, ulinastatin had no effect on the baseline nociceptive threshold. Moreover, repeated i.p. injections of ulinastatin persistently attenuated incision-induced mechanical pain hypersensitivity and promoted recovery from the surgery. The rats did not develop any analgesic tolerance over the course of repeated injections of ulinastatin. A single i.p. injection of ulinastatin was also sufficient to inhibit the initiation and maintenance of incision-induced hyperalgesic priming when the rats were subsequently challenged with an ipsilateral intraplantar prostaglandin E2 injection. Furthermore, the combined administration of ulinastatin and sufentanil significantly enhanced the analgesic effect of sufentanil on postoperative pain, which involved mechanisms other than a direct influence on opioid receptors. These findings demonstrated that ulinastatin had a significant analgesic effect on postoperative pain and might be a novel pharmacotherapeutic agent for managing postoperative pain either alone or as an adjuvant.
Collapse
Affiliation(s)
- Zhi-Bin Guo
- Department of Anesthesiology and Perioperative Medicine, 900 Hospital of the Joint Logistic Support Force / Fuzong Clinical Medical College, Fujian Medical University, Fuzhou, 350025, Fujian, PR China; Department of Anesthesiology and Perioperative Medicine, Dongfang Hospital, Xiamen University, Fuzhou, 350025, Fujian, PR China
| | - Li Tang
- Department of Stomatology, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, PR China
| | - Li-Ping Wang
- Department of Anesthesiology and Perioperative Medicine, 900 Hospital of the Joint Logistic Support Force / Fuzong Clinical Medical College, Fujian Medical University, Fuzhou, 350025, Fujian, PR China; Department of Anesthesiology and Perioperative Medicine, Dongfang Hospital, Xiamen University, Fuzhou, 350025, Fujian, PR China
| | - Huang-Hui Wu
- Department of Anesthesiology and Perioperative Medicine, 900 Hospital of the Joint Logistic Support Force / Fuzong Clinical Medical College, Fujian Medical University, Fuzhou, 350025, Fujian, PR China
| | - Chang-Lu Huang
- Department of Anesthesiology and Perioperative Medicine, 900 Hospital of the Joint Logistic Support Force / Fuzong Clinical Medical College, Fujian Medical University, Fuzhou, 350025, Fujian, PR China
| | - Mei-Xiang Zhan
- Department of Anesthesiology and Perioperative Medicine, 900 Hospital of the Joint Logistic Support Force / Fuzong Clinical Medical College, Fujian Medical University, Fuzhou, 350025, Fujian, PR China
| | - Zhong-Mou Shi
- Department of Anesthesiology and Perioperative Medicine, 900 Hospital of the Joint Logistic Support Force / Fuzong Clinical Medical College, Fujian Medical University, Fuzhou, 350025, Fujian, PR China
| | - Chen-Long Yang
- Department of Anesthesiology and Perioperative Medicine, 900 Hospital of the Joint Logistic Support Force / Fuzong Clinical Medical College, Fujian Medical University, Fuzhou, 350025, Fujian, PR China
| | - Guo-Zhong Chen
- Department of Anesthesiology and Perioperative Medicine, 900 Hospital of the Joint Logistic Support Force / Fuzong Clinical Medical College, Fujian Medical University, Fuzhou, 350025, Fujian, PR China; Department of Anesthesiology and Perioperative Medicine, Dongfang Hospital, Xiamen University, Fuzhou, 350025, Fujian, PR China
| | - Yi-Qing Zou
- Department of Anesthesiology and Perioperative Medicine, 900 Hospital of the Joint Logistic Support Force / Fuzong Clinical Medical College, Fujian Medical University, Fuzhou, 350025, Fujian, PR China; Department of Anesthesiology and Perioperative Medicine, Dongfang Hospital, Xiamen University, Fuzhou, 350025, Fujian, PR China.
| | - Fei Yang
- Department of Anesthesiology and Perioperative Medicine, 900 Hospital of the Joint Logistic Support Force / Fuzong Clinical Medical College, Fujian Medical University, Fuzhou, 350025, Fujian, PR China; Department of Anesthesiology and Perioperative Medicine, Dongfang Hospital, Xiamen University, Fuzhou, 350025, Fujian, PR China; Pain Research Laboratory, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, Fujian, PR China.
| | - Xiao-Zhi Wu
- Department of Anesthesiology and Perioperative Medicine, 900 Hospital of the Joint Logistic Support Force / Fuzong Clinical Medical College, Fujian Medical University, Fuzhou, 350025, Fujian, PR China; Department of Anesthesiology and Perioperative Medicine, Dongfang Hospital, Xiamen University, Fuzhou, 350025, Fujian, PR China.
| |
Collapse
|
15
|
Mwirigi J, Kume M, Hassler SN, Ahmad A, Ray PR, Jiang C, Chamessian A, Mseeh N, Ludwig BP, Rivera BD, Nieman MT, Van de Ven T, Ji RR, Dussor G, Boitano S, Vagner J, Price TJ. A Role for Protease Activated Receptor Type 3 (PAR3) in Nociception Demonstrated Through Development of a Novel Peptide Agonist. THE JOURNAL OF PAIN 2021; 22:692-706. [PMID: 33429107 PMCID: PMC8197731 DOI: 10.1016/j.jpain.2020.12.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 12/02/2020] [Accepted: 12/20/2020] [Indexed: 02/07/2023]
Abstract
The protease activated receptor (PAR) family is a group of G-protein coupled receptors (GPCRs) activated by proteolytic cleavage of the extracellular domain. PARs are expressed in a variety of cell types with crucial roles in homeostasis, immune responses, inflammation, and pain. PAR3 is the least researched of the four PARs, with little known about its expression and function. We sought to better understand its potential function in the peripheral sensory nervous system. Mouse single-cell RNA sequencing data demonstrates that PAR3 is widely expressed in dorsal root ganglion (DRG) neurons. Co-expression of PAR3 mRNA with other PARs was identified in various DRG neuron subpopulations, consistent with its proposed role as a coreceptor of other PARs. We developed a lipid tethered PAR3 agonist, C660, that selectively activates PAR3 by eliciting a Ca2+ response in DRG and trigeminal neurons. In vivo, C660 induces mechanical hypersensitivity and facial grimacing in WT but not PAR3-/- mice. We characterized other nociceptive phenotypes in PAR3-/- mice and found a loss of hyperalgesic priming in response to IL-6, carrageenan, and a PAR2 agonist, suggesting that PAR3 contributes to long-lasting nociceptor plasticity in some contexts. To examine the potential role of PAR3 in regulating the activity of other PARs in sensory neurons, we administered PAR1, PAR2, and PAR4 agonists and assessed mechanical and affective pain behaviors in WT and PAR3-/- mice. We observed that the nociceptive effects of PAR1 agonists were potentiated in the absence of PAR3. Our findings suggest a complex role of PAR3 in the physiology and plasticity of nociceptors. PERSPECTIVE: We evaluated the role of PAR3, a G-protein coupled receptor, in nociception by developing a selective peptide agonist. Our findings suggest that PAR3 contributes to nociception in various contexts and plays a role in modulating the activity of other PARs.
Collapse
Affiliation(s)
- Juliet Mwirigi
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Richardson, Texas
| | - Moeno Kume
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Richardson, Texas
| | - Shayne N Hassler
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Richardson, Texas
| | - Ayesha Ahmad
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Richardson, Texas
| | - Pradipta R Ray
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Richardson, Texas
| | - Changyu Jiang
- Duke University School of Medicine, Department of Anesthesiology, Pharmacology, and Cancer Biology, Durham, North Carolina
| | - Alexander Chamessian
- Duke University School of Medicine, Department of Anesthesiology, Pharmacology, and Cancer Biology, Durham, North Carolina
| | - Nakleh Mseeh
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Richardson, Texas
| | - Breya P Ludwig
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Richardson, Texas
| | - Benjamin D Rivera
- Department of Physiology, University of Arizona, Asthma and Airway Disease Research Center, Tucson, Arizona
| | - Marvin T Nieman
- Case Western Reserve University School of Medicine, Department of Pharmacology, Cleveland, Ohio
| | - Thomas Van de Ven
- Duke University School of Medicine, Department of Anesthesiology, Pharmacology, and Cancer Biology, Durham, North Carolina
| | - Ru-Rong Ji
- Duke University School of Medicine, Department of Anesthesiology, Pharmacology, and Cancer Biology, Durham, North Carolina
| | - Gregory Dussor
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Richardson, Texas
| | - Scott Boitano
- Department of Physiology, University of Arizona, Asthma and Airway Disease Research Center, Tucson, Arizona
| | - Josef Vagner
- University of Arizona, Bio5 Research Institute, Tucson, Arizona
| | - Theodore J Price
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Richardson, Texas.
| |
Collapse
|
16
|
Whittaker AL, Liu Y, Barker TH. Methods Used and Application of the Mouse Grimace Scale in Biomedical Research 10 Years on: A Scoping Review. Animals (Basel) 2021; 11:673. [PMID: 33802463 PMCID: PMC7999303 DOI: 10.3390/ani11030673] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/25/2021] [Accepted: 02/25/2021] [Indexed: 12/11/2022] Open
Abstract
The Mouse Grimace Scale (MGS) was developed 10 years ago as a method for assessing pain through the characterisation of changes in five facial features or action units. The strength of the technique is that it is proposed to be a measure of spontaneous or non-evoked pain. The time is opportune to map all of the research into the MGS, with a particular focus on the methods used and the technique's utility across a range of mouse models. A comprehensive scoping review of the academic literature was performed. A total of 48 articles met our inclusion criteria and were included in this review. The MGS has been employed mainly in the evaluation of acute pain, particularly in the pain and neuroscience research fields. There has, however, been use of the technique in a wide range of fields, and based on limited study it does appear to have utility for pain assessment across a spectrum of animal models. Use of the method allows the detection of pain of a longer duration, up to a month post initial insult. There has been less use of the technique using real-time methods and this is an area in need of further research.
Collapse
Affiliation(s)
- Alexandra L. Whittaker
- School of Animal and Veterinary Sciences, Roseworthy Campus, The University of Adelaide, Roseworthy 5371, Australia;
| | - Yifan Liu
- School of Animal and Veterinary Sciences, Roseworthy Campus, The University of Adelaide, Roseworthy 5371, Australia;
| | - Timothy H. Barker
- JBI, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5005, Australia;
| |
Collapse
|
17
|
Kido K, Katagiri N, Kawana H, Sugino S, Yamauchi M, Masaki E. Nociceptive Sensitization by Activation of Protease-Activated Receptor 2 in a Rat Model of Incisional Pain. Brain Sci 2021; 11:brainsci11020144. [PMID: 33499207 PMCID: PMC7911446 DOI: 10.3390/brainsci11020144] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/13/2021] [Accepted: 01/20/2021] [Indexed: 12/20/2022] Open
Abstract
Postoperative pain and consequent inflammatory responses after tissue incision adversely affects many surgical patients due to complicated mechanisms. In this study, we examined whether activation of protease-activated receptor 2 (PAR-2), which is stimulated by tryptase from mast cells, elicits nociception and whether the PAR-2 antagonist could reduce incisional nociceptive responses in vivo and in vitro. The effects of a selective PAR-2 antagonist, N3-methylbutyryl-N-6-aminohexanoyl-piperazine (ENMD-1068), pretreatment on pain behaviors were assessed after plantar incision in rats. The effects of a PAR-2 agonist, SLIGRL-NH2, on nociception was assessed after the injection into the hind paw. Furthermore, the responses of C-mechanosensitive nociceptors to the PAR-2 agonist were observed using an in vitro skin–nerve preparation as well. Intraplantar injection of SLIGRL-NH2 elicited spontaneous nociceptive behavior and hyperalgesia. Local administration of ENMD-1068 suppressed guarding behaviors, mechanical and heat hyperalgesia only within the first few hours after incision. SLIGRL-NH2 caused ongoing activity in 47% of C-mechanonociceptors in vitro. This study suggests that PAR-2 may support early nociception after incision by direct or indirect sensitization of C-fibers in rats. Moreover, PAR-2 may play a regulatory role in the early period of postoperative pain together with other co-factors to that contribute to postoperative pain.
Collapse
Affiliation(s)
- Kanta Kido
- Department of Anesthesiology, Kanagawa Dental University Hospital, Yokosuka, Kanagawa 2388570, Japan;
- Correspondence:
| | - Norika Katagiri
- Department of Anesthesiology, Kanagawa Dental University Hospital, Yokosuka, Kanagawa 2388570, Japan;
| | - Hiromasa Kawana
- Department of Oral and Maxillofacial Implantology, Kanagawa Dental University Hospital, Yokosuka, Kanagawa 2388570, Japan;
| | - Shigekazu Sugino
- Department of Anesthesiology and Perioperative Medicine, Tohoku University School of Medicine, Sendai, Miyagi 9808575, Japan; (S.S.); (M.Y.)
| | - Masanori Yamauchi
- Department of Anesthesiology and Perioperative Medicine, Tohoku University School of Medicine, Sendai, Miyagi 9808575, Japan; (S.S.); (M.Y.)
| | - Eiji Masaki
- Department of Anesthesiology, International University of Health and Welfare Hospital, Nasushiobara, Tochigi 3292763, Japan;
| |
Collapse
|
18
|
Ferrini F, Salio C, Boggio EM, Merighi A. Interplay of BDNF and GDNF in the Mature Spinal Somatosensory System and Its Potential Therapeutic Relevance. Curr Neuropharmacol 2021; 19:1225-1245. [PMID: 33200712 PMCID: PMC8719296 DOI: 10.2174/1570159x18666201116143422] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 09/17/2020] [Accepted: 10/05/2020] [Indexed: 11/22/2022] Open
Abstract
The growth factors BDNF and GDNF are gaining more and more attention as modulators of synaptic transmission in the mature central nervous system (CNS). The two molecules undergo a regulated secretion in neurons and may be anterogradely transported to terminals where they can positively or negatively modulate fast synaptic transmission. There is today a wide consensus on the role of BDNF as a pro-nociceptive modulator, as the neurotrophin has an important part in the initiation and maintenance of inflammatory, chronic, and/or neuropathic pain at the peripheral and central level. At the spinal level, BDNF intervenes in the regulation of chloride equilibrium potential, decreases the excitatory synaptic drive to inhibitory neurons, with complex changes in GABAergic/glycinergic synaptic transmission, and increases excitatory transmission in the superficial dorsal horn. Differently from BDNF, the role of GDNF still remains to be unraveled in full. This review resumes the current literature on the interplay between BDNF and GDNF in the regulation of nociceptive neurotransmission in the superficial dorsal horn of the spinal cord. We will first discuss the circuitries involved in such a regulation, as well as the reciprocal interactions between the two factors in nociceptive pathways. The development of small molecules specifically targeting BDNF, GDNF and/or downstream effectors is opening new perspectives for investigating these neurotrophic factors as modulators of nociceptive transmission and chronic pain. Therefore, we will finally consider the molecules of (potential) pharmacological relevance for tackling normal and pathological pain.
Collapse
Affiliation(s)
- Francesco Ferrini
- Department of Veterinary Sciences, University of Turin, Grugliasco, Italy
- Department of Psychiatry & Neuroscience, Université Laval, Québec, Canada
| | - Chiara Salio
- Department of Veterinary Sciences, University of Turin, Grugliasco, Italy
| | - Elena M. Boggio
- Department of Veterinary Sciences, University of Turin, Grugliasco, Italy
| | - Adalberto Merighi
- Department of Veterinary Sciences, University of Turin, Grugliasco, Italy
- National Institute of Neuroscience, Grugliasco, Italy
| |
Collapse
|
19
|
Garza Carbajal A, Ebersberger A, Thiel A, Ferrari L, Acuna J, Brosig S, Isensee J, Moeller K, Siobal M, Rose-John S, Levine J, Schaible HG, Hucho T. Oncostatin M induces hyperalgesic priming and amplifies signaling of cAMP to ERK by RapGEF2 and PKA. J Neurochem 2020; 157:1821-1837. [PMID: 32885411 DOI: 10.1111/jnc.15172] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 08/19/2020] [Accepted: 08/20/2020] [Indexed: 12/19/2022]
Abstract
Hyperalgesic priming is characterized by enhanced nociceptor sensitization by pronociceptive mediators, prototypically PGE2 . Priming has gained interest as a mechanism underlying the transition to chronic pain. Which stimuli induce priming and what cellular mechanisms are employed remains incompletely understood. In adult male rats, we present the cytokine Oncostatin M (OSM), a member of the IL-6 family, as an inducer of priming by a novel mechanism. We used a high content microscopy based approach to quantify the activation of endogenous PKA-II and ERK of thousands sensory neurons in culture. Incubation with OSM increased and prolonged ERK activation by agents that increase cAMP production such as PGE2 , forskolin, and cAMP analogs. These changes were specific to IB4/CaMKIIα positive neurons, required protein translation, and increased cAMP-to-ERK signaling. In both, control and OSM-treated neurons, cAMP/ERK signaling involved RapGEF2 and PKA but not Epac. Similar enhancement of cAMP-to-ERK signaling could be induced by GDNF, which acts mostly on IB4/CaMKIIα-positive neurons, but not by NGF, which acts mostly on IB4/CaMKIIα-negative neurons. In vitro, OSM pretreatment rendered baseline TTX-R currents ERK-dependent and switched forskolin-increased currents from partial to full ERK-dependence in small/medium sized neurons. In summary, priming induced by OSM uses a novel mechanism to enhance and prolong coupling of cAMP/PKA to ERK1/2 signaling without changing the overall pathway structure.
Collapse
Affiliation(s)
- Anibal Garza Carbajal
- Department of Anaesthesiology and Intensive Care Medicine, Translational Pain Research, University Hospital Cologne, University Cologne, Cologne, Germany
| | | | - Alina Thiel
- Department of Anaesthesiology and Intensive Care Medicine, Translational Pain Research, University Hospital Cologne, University Cologne, Cologne, Germany
| | - Luiz Ferrari
- Department of Oral and Maxillofacial Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Jeremy Acuna
- Department of Anaesthesiology and Intensive Care Medicine, Translational Pain Research, University Hospital Cologne, University Cologne, Cologne, Germany
| | - Stephanie Brosig
- Department of Anaesthesiology and Intensive Care Medicine, Translational Pain Research, University Hospital Cologne, University Cologne, Cologne, Germany
| | - Joerg Isensee
- Department of Anaesthesiology and Intensive Care Medicine, Translational Pain Research, University Hospital Cologne, University Cologne, Cologne, Germany
| | - Katharina Moeller
- Department of Anaesthesiology and Intensive Care Medicine, Translational Pain Research, University Hospital Cologne, University Cologne, Cologne, Germany
| | - Maike Siobal
- Department of Anaesthesiology and Intensive Care Medicine, Translational Pain Research, University Hospital Cologne, University Cologne, Cologne, Germany
| | | | - Jon Levine
- Department of Oral and Maxillofacial Surgery, University of California San Francisco, San Francisco, CA, USA
| | | | - Tim Hucho
- Department of Anaesthesiology and Intensive Care Medicine, Translational Pain Research, University Hospital Cologne, University Cologne, Cologne, Germany
| |
Collapse
|
20
|
Wangzhou A, McIlvried LA, Paige C, Barragan-Iglesias P, Shiers S, Ahmad A, Guzman CA, Dussor G, Ray PR, Gereau RW, Price TJ. Pharmacological target-focused transcriptomic analysis of native vs cultured human and mouse dorsal root ganglia. Pain 2020; 161:1497-1517. [PMID: 32197039 PMCID: PMC7305999 DOI: 10.1097/j.pain.0000000000001866] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Dorsal root ganglion (DRG) neurons detect sensory inputs and are crucial for pain processing. They are often studied in vitro as dissociated cell cultures with the assumption that this reasonably represents in vivo conditions. However, to the best of our knowledge, no study has directly compared genome-wide transcriptomes of DRG tissue in vivo versus in vitro or between laboratories and culturing protocols. Comparing RNA sequencing-based transcriptomes of native to cultured (4 days in vitro) human or mouse DRG, we found that the overall expression levels of many ion channels and G-protein-coupled receptors specifically expressed in neurons are markedly lower although still expressed in culture. This suggests that most pharmacological targets expressed in vivo are present under the condition of dissociated cell culture, but with changes in expression levels. The reduced relative expression for neuronal genes in human DRG cultures is likely accounted for by increased expression of genes in fibroblast-like and other proliferating cells, consistent with their mitotic status in these cultures. We found that the expression of a subset of genes typically expressed in neurons increased in human and mouse DRG cultures relative to the intact ganglion, including genes associated with nerve injury or inflammation in preclinical models such as BDNF, MMP9, GAL, and ATF3. We also found a striking upregulation of a number of inflammation-associated genes in DRG cultures, although many were different between mouse and human. Our findings suggest an injury-like phenotype in DRG cultures that has important implications for the use of this model system for pain drug discovery.
Collapse
Affiliation(s)
- Andi Wangzhou
- The University of Texas at Dallas, School of Behavioral and
Brain Sciences and Center for Advanced Pain Studies, 800 W Campbell Rd. Richardson,
TX, 75080, USA
| | - Lisa A. McIlvried
- Washington University Pain Center and Department of
Anesthesiology, Washington University School of Medicine
| | - Candler Paige
- The University of Texas at Dallas, School of Behavioral and
Brain Sciences and Center for Advanced Pain Studies, 800 W Campbell Rd. Richardson,
TX, 75080, USA
| | - Paulino Barragan-Iglesias
- The University of Texas at Dallas, School of Behavioral and
Brain Sciences and Center for Advanced Pain Studies, 800 W Campbell Rd. Richardson,
TX, 75080, USA
| | - Stephanie Shiers
- The University of Texas at Dallas, School of Behavioral and
Brain Sciences and Center for Advanced Pain Studies, 800 W Campbell Rd. Richardson,
TX, 75080, USA
| | - Ayesha Ahmad
- The University of Texas at Dallas, School of Behavioral and
Brain Sciences and Center for Advanced Pain Studies, 800 W Campbell Rd. Richardson,
TX, 75080, USA
| | - Carolyn A. Guzman
- The University of Texas at Dallas, School of Behavioral and
Brain Sciences and Center for Advanced Pain Studies, 800 W Campbell Rd. Richardson,
TX, 75080, USA
| | - Gregory Dussor
- The University of Texas at Dallas, School of Behavioral and
Brain Sciences and Center for Advanced Pain Studies, 800 W Campbell Rd. Richardson,
TX, 75080, USA
| | - Pradipta R. Ray
- The University of Texas at Dallas, School of Behavioral and
Brain Sciences and Center for Advanced Pain Studies, 800 W Campbell Rd. Richardson,
TX, 75080, USA
| | - Robert W. Gereau
- Washington University Pain Center and Department of
Anesthesiology, Washington University School of Medicine
| | - Theodore J. Price
- The University of Texas at Dallas, School of Behavioral and
Brain Sciences and Center for Advanced Pain Studies, 800 W Campbell Rd. Richardson,
TX, 75080, USA
| |
Collapse
|
21
|
Hassler SN, Kume M, Mwirigi JM, Ahmad A, Shiers S, Wangzhou A, Ray PR, Belugin SN, Naik DK, Burton MD, Vagner J, Boitano S, Akopian AN, Dussor G, Price TJ. The cellular basis of protease-activated receptor 2-evoked mechanical and affective pain. JCI Insight 2020; 5:137393. [PMID: 32352932 PMCID: PMC7308051 DOI: 10.1172/jci.insight.137393] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 04/24/2020] [Indexed: 12/31/2022] Open
Abstract
Protease-activated receptor 2 (PAR2) has long been implicated in inflammatory and visceral pain, but the cellular basis of PAR2-evoked pain has not been delineated. Although PAR2-evoked pain has been attributed to sensory neuron expression, RNA-sequencing experiments show ambiguous F2rl1 mRNA detection. Moreover, many pharmacological tools for PAR2 are nonspecific, acting also on the Mas-related GPCR family (Mrg) that are highly enriched in sensory neurons. We sought to clarify the cellular basis of PAR2-evoked pain. We developed a PAR2-conditional knockout mouse and specifically deleted PAR2 in all sensory neurons using the PirtCre mouse line. Our behavioral findings show that PAR2 agonist-evoked mechanical hyperalgesia and facial grimacing, but not thermal hyperalgesia, are dependent on PAR2 expression in sensory neurons that project to the hind paw in male and female mice. F2rl1 mRNA is expressed in a discrete population (~4%) of mostly small-diameter sensory neurons that coexpress the Nppb and IL31ra genes. This cell population has been implicated in itch, but our work shows that PAR2 activation in these cells causes clear pain-related behaviors from the skin. Our findings show that a discrete population of DRG sensory neurons mediate PAR2-evoked pain.
Collapse
Affiliation(s)
- Shayne N. Hassler
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, Dallas, Texas, USA
| | - Moeno Kume
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, Dallas, Texas, USA
| | - Juliet M. Mwirigi
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, Dallas, Texas, USA
| | - Ayesha Ahmad
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, Dallas, Texas, USA
| | - Stephanie Shiers
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, Dallas, Texas, USA
| | - Andi Wangzhou
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, Dallas, Texas, USA
| | - Pradipta R. Ray
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, Dallas, Texas, USA
| | - Serge N. Belugin
- Department of Endodontics, School of Dentistry, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Dhananjay K. Naik
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, Dallas, Texas, USA
| | - Michael D. Burton
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, Dallas, Texas, USA
| | | | - Scott Boitano
- BIO5 Research Institute and
- Department of Physiology, Asthma & Airway Disease Research Center, University of Arizona, Tucson, Arizona, USA
| | - Armen N. Akopian
- Department of Endodontics, School of Dentistry, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Gregory Dussor
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, Dallas, Texas, USA
| | - Theodore J. Price
- School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, University of Texas at Dallas, Dallas, Texas, USA
| |
Collapse
|
22
|
Long T, He W, Pan Q, Zhang S, Zhang D, Qin G, Chen L, Zhou J. Microglia P2X4R-BDNF signalling contributes to central sensitization in a recurrent nitroglycerin-induced chronic migraine model. J Headache Pain 2020; 21:4. [PMID: 31937253 PMCID: PMC6961410 DOI: 10.1186/s10194-019-1070-4] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 12/23/2019] [Indexed: 12/11/2022] Open
Abstract
Background According to our previous study, microglia P2X4 receptors (P2X4Rs) play a pivotal role in the central sensitization of chronic migraine (CM). However, the molecular mechanism that underlies the crosstalk between microglia P2X4Rs and neurons of the trigeminal nucleus caudalis (TNC) is not fully understood. Therefore, the aim of this study is to examine the exact P2X4Rs signalling pathway in the development of central sensitization in a CM animal model. Methods We used an animal model with recurrent intermittent administration of nitroglycerin (NTG), which closely mimics CM. NTG-induced basal mechanical and thermal hypersensitivity were evaluated using a von Frey filament test and an increasing-temperature hot plate apparatus (IITC). We detected P2X4Rs, brain-derived neurotrophic factor (BDNF) and phosphorylated p38 mitogen-activated protein kinase (p-p38-MAPK) expression profiles in the TNC. We investigated the effects of a P2X4R inhibitor (5-BDBD) and an agonist (IVM) on NTG-induced hyperalgesia and neurochemical changes as well as on the expression of p-p38-MAPK and BDNF. We also detected the effects of a tropomyosin-related kinase B (TrkB) inhibitor (ANA-12) on the CM animal model in vivo. Then, we evaluated the effect of 5-BDBD and SB203580 (a p38-MAPK inhibitors) on the release and synthesis of BDNF in BV2 microglia cells treated with 50 μM adenosine triphosphate (ATP). Results Chronic intermittent administration of NTG resulted in chronic mechanical and thermal hyperalgesia, accompanied by the upregulation of P2X4Rs and BDNF expression. 5-BDBD or ANA-12 prevented hyperalgesia induced by NTG, which was associated with a significant inhibition of the NTG-induced increase in phosphorylated extracellular regulated protein kinases (p-ERK) and calcitonin gene related peptide (CGRP) release in the TNC. Repeated administration of IVM produced sustained hyperalgesia and significantly increased the levels of p-ERK and CGRP release in the TNC. Activating P2X4Rs with ATP triggered BDNF release and increased BDNF synthesis in BV2 microglia, and these results were then reduced by 5-BDBD or SB203580. Conclusions Our results indicated that the P2X4R contributes to the central sensitization of CM by releasing BDNF and promoting TNC neuronal hyper-excitability. Blocking microglia P2X4R-BDNF signalling may have an effect on the prevention of migraine chronification.
Collapse
Affiliation(s)
- Ting Long
- Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Wei He
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1st Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Qi Pan
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1st Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Shanshan Zhang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1st Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Dunke Zhang
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guangcheng Qin
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lixue Chen
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiying Zhou
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1st Youyi Road, Yuzhong District, Chongqing, 400016, China.
| |
Collapse
|
23
|
Hoffman S, Aviv Cohen N, Carroll IM, Tulchinsky H, Borovok I, Dotan I, Maharshak N. Faecal Proteases from Pouchitis Patients Activate Protease Activating Receptor-2 to Disrupt the Epithelial Barrier. J Crohns Colitis 2019; 13:1558-1568. [PMID: 31056700 DOI: 10.1093/ecco-jcc/jjz086] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS The pathogenesis of pouch inflammation may involve epithelial barrier disruption. We investigated whether faecal proteolytic activity is increased during pouchitis and results in epithelial barrier dysfunction through protease activating receptor [PAR] activation, and assessed whether the intestinal microbiome may be the source of the proteases. METHODS Faecal samples were measured for protease activity using a fluorescein isothiocyanate [FITC]-casein florescence assay. Caco-2 cell monolayers were exposed to faecal supernatants to assess permeability to FITC-dextran. Tight junction protein integrity and PAR activation were assessed by immunoblot and immunofluorescence. A truncated PAR2 protein in Caco-2 cells was achieved by stable transfection using CRISPR/Cas9 plasmid. PAR2 activation in pouch biopsies was examined using antibodies directed to the N-terminus of the protein. Microbial composition was analysed based on 16S rRNA gene sequence analysis. RESULTS Ten pouchitis patients, six normal pouch [NP] patients and nine healthy controls [HC] were recruited. The pouchitis patients exhibited a 5.19- and 5.35-fold higher faecal protease [FP] activity [p ≤ 0.05] compared to the NP and HC participants, respectively. The genus Haemophilus was positively associated with FP activity [R = 0.718, false discovery rate < 0.1]. Faecal supernatants from pouchitis patients activated PAR2 on Caco-2 monolayers, disrupted tight junction proteins and increased epithelial permeability. PAR2 truncation in Caco-2 abrogated faecal protease-mediated permeability. Pouch biopsies obtained from pouchitis patients, but not from NP patients, displayed PAR2 activation. CONCLUSIONS Protease-producing bacteria may increase faecal proteolytic activity that results in pouch inflammation through disruption of tight junction proteins and increased epithelial permeability in a PAR2-dependent manner. This mechanism may initiate or propagate pouch inflammation.
Collapse
Affiliation(s)
- Sarit Hoffman
- The Research Center for Digestive Tract and Liver Diseases, Tel Aviv Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine Tel-Aviv University, Tel Aviv, Israel
| | - Nathaniel Aviv Cohen
- The Research Center for Digestive Tract and Liver Diseases, Tel Aviv Medical Center, Tel Aviv, Israel.,IBD Center, Tel Aviv Medical Center, Tel Aviv, Israel.,Department of Gastroenterology and Liver Diseases, Tel Aviv Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine Tel-Aviv University, Tel Aviv, Israel
| | - Ian M Carroll
- Center for Gastrointestinal Biology and Disease, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Hagit Tulchinsky
- Division of Surgery Colorectal Unit, Tel Aviv Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine Tel-Aviv University, Tel Aviv, Israel
| | - Ilya Borovok
- Department of Molecular and Microbiology and Biotechnology, Tel Aviv University, Tel Aviv, Israel
| | - Iris Dotan
- Division of Gastroenterology, Rabin Medical Center, Petah Tikva, Israel.,Sackler Faculty of Medicine Tel-Aviv University, Tel Aviv, Israel
| | - Nitsan Maharshak
- The Research Center for Digestive Tract and Liver Diseases, Tel Aviv Medical Center, Tel Aviv, Israel.,IBD Center, Tel Aviv Medical Center, Tel Aviv, Israel.,Department of Gastroenterology and Liver Diseases, Tel Aviv Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine Tel-Aviv University, Tel Aviv, Israel
| |
Collapse
|
24
|
Kang P, Li X, Liu Y, Shiers SI, Xiong H, Giannotta M, Dejana E, Price TJ, Randrianalisoa J, Nielsen SO, Qin Z. Transient Photoinactivation of Cell Membrane Protein Activity without Genetic Modification by Molecular Hyperthermia. ACS NANO 2019; 13:12487-12499. [PMID: 31613606 PMCID: PMC7096286 DOI: 10.1021/acsnano.9b01993] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Precise manipulation of protein activity in living systems has broad applications in biomedical sciences. However, it is challenging to use light to manipulate protein activity in living systems without genetic modification. Here, we report a technique to optically switch off protein activity in living cells with high spatiotemporal resolution, referred to as molecular hyperthermia (MH). MH is based on the nanoscale-confined heating of plasmonic gold nanoparticles by short laser pulses to unfold and photoinactivate targeted proteins of interest. First, we show that protease-activated receptor 2 (PAR2), a G-protein-coupled receptor and an important pathway that leads to pain sensitization, can be photoinactivated in situ by MH without compromising cell proliferation. PAR2 activity can be switched off in laser-targeted cells without affecting surrounding cells. Furthermore, we demonstrate the molecular specificity of MH by inactivating PAR2 while leaving other receptors intact. Second, we demonstrate that the photoinactivation of a tight junction protein in brain endothelial monolayers leads to a reversible blood-brain barrier opening in vitro. Lastly, the protein inactivation by MH is below the nanobubble generation threshold and thus is predominantly due to the nanoscale heating. MH is distinct from traditional hyperthermia (that induces global tissue heating) in both its time and length scales: nanoseconds versus seconds, nanometers versus millimeters. Our results demonstrate that MH enables selective and remote manipulation of protein activity and cellular behavior without genetic modification.
Collapse
Affiliation(s)
- Peiyuan Kang
- Department of Mechanical Engineering, University of Texas at Dallas, 800 West Campbell Rd., Richardson, Texas 75080, USA
| | - Xiaoqing Li
- Department of Bioengineering, University of Texas at Dallas, 800 West Campbell Rd., Richardson, Texas 75080, USA
| | - Yaning Liu
- Department of Mechanical Engineering, University of Texas at Dallas, 800 West Campbell Rd., Richardson, Texas 75080, USA
| | - Stephanie I. Shiers
- School of Behavioral and Brain Sciences, University of Texas at Dallas, 800 West Campbell Rd., Richardson, Texas 75080, USA
| | - Hejian Xiong
- Department of Mechanical Engineering, University of Texas at Dallas, 800 West Campbell Rd., Richardson, Texas 75080, USA
| | - Monica Giannotta
- Vascular Biology Laboratory, The FIRC Institute of Molecular Oncology (IFOM), 20139 Milan, Italy
| | - Elisabetta Dejana
- Vascular Biology Laboratory, The FIRC Institute of Molecular Oncology (IFOM), 20139 Milan, Italy
- Department of Immunology, Genetics and Pathology, University of Uppsala, 751 05 Uppsala, Sweden
| | - Theodore John Price
- School of Behavioral and Brain Sciences, University of Texas at Dallas, 800 West Campbell Rd., Richardson, Texas 75080, USA
| | - Jaona Randrianalisoa
- Institut de Thermique, Mécanique, Matériaux (ITheMM EA 7548), University of Reims Champagne-Ardenne, Reims Cedex 2, 51687 France
| | - Steven O. Nielsen
- Department of Chemistry and Biochemistry, University of Texas at Dallas, 800 West Campbell Rd., Richardson, Texas 75080, USA
| | - Zhenpeng Qin
- Department of Mechanical Engineering, University of Texas at Dallas, 800 West Campbell Rd., Richardson, Texas 75080, USA
- Department of Bioengineering, University of Texas at Dallas, 800 West Campbell Rd., Richardson, Texas 75080, USA
- Department of Surgery, University of Texas at Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, Texas 75390, USA
- Corresponding Author: . Phone: (972)883-4440
| |
Collapse
|
25
|
Sarkar D, Kang P, Nielsen SO, Qin Z. Non-Arrhenius Reaction-Diffusion Kinetics for Protein Inactivation over a Large Temperature Range. ACS NANO 2019; 13:8669-8679. [PMID: 31268674 PMCID: PMC7384293 DOI: 10.1021/acsnano.9b00068] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Understanding protein folding and unfolding has been a long-standing fundamental question and has important applications in manipulating protein activity in biological systems. Experimental investigations of protein unfolding have been predominately conducted by small temperature perturbations (e.g., temperature jump), while molecular simulations are limited to small time scales (microseconds) and high temperatures to observe unfolding. Thus, it remains unclear how fast a protein unfolds irreversibly and loses function (i.e., inactivation) across a large temperature range. In this work, using nanosecond pulsed heating of individual plasmonic nanoparticles to create precise localized heating, we examine the protein inactivation kinetics at extremely high temperatures. Connecting this with protein inactivation measurements at low temperatures, we observe that the kinetics of protein unfolding is less sensitive to temperature change at the higher temperatures, which significantly departs from the Arrhenius behavior extrapolated from low temperatures. To account for this effect, we propose a reaction-diffusion model that modifies the temperature-dependence of protein inactivation by introducing a diffusion limit. Analysis of the reaction-diffusion model provides general guidelines in the behavior of protein inactivation (reaction-limited, transition, diffusion-limited) across a large temperature range from physiological temperature to extremely high temperatures. We further demonstrate that the reaction-diffusion model is particularly useful for designing optimal operating conditions for protein photoinactivation. The experimentally validated reaction-diffusion kinetics of protein unfolding is an important step toward understanding protein-inactivation kinetics over a large temperature range. It has important applications including molecular hyperthermia and calls for future studies to examine this model for other protein molecules.
Collapse
Affiliation(s)
- Daipayan Sarkar
- Department of Mechanical Engineering, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Peiyuan Kang
- Department of Mechanical Engineering, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Steven O. Nielsen
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Zhenpeng Qin
- Department of Mechanical Engineering, The University of Texas at Dallas, Richardson, TX 75080, USA
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX 75080, USA
- Department of Surgery, The University of Texas at Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
26
|
Richards J, Tang S, Gunsch G, Sul P, Wiet M, Flanigan DC, Khan SN, Moore S, Walter B, Purmessur D. Mast Cell/Proteinase Activated Receptor 2 (PAR2) Mediated Interactions in the Pathogenesis of Discogenic Back Pain. Front Cell Neurosci 2019; 13:294. [PMID: 31333416 PMCID: PMC6625229 DOI: 10.3389/fncel.2019.00294] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 06/17/2019] [Indexed: 12/19/2022] Open
Abstract
Mast cells (MCs) are present in the painful degenerate human intervertebral disc (IVD) and are associated with disease pathogenesis. MCs release granules containing enzymatic and inflammatory factors in response to stimulants or allergens. The serine protease, tryptase, is unique to MCs and its activation of the G-protein coupled receptor, Protease Activated Receptor 2 (PAR2), induces inflammation and degradation in osteoarthritic cartilage. Our previously published work has demonstrated increased levels of MC marker tryptase in IVD samples from discogenic back pain patients compared to healthy control IVD samples including expression of chemotactic agents that may facilitate MC migration into the IVD. To further elucidate MCs’ role in the IVD and mechanisms underlying its effects, we investigated whether (1) human IVD cells can promote MC migration, (2) MC tryptase can mediate up-regulation of inflammatory/catabolic process in human IVD cells and tissue, and (3) the potential of PAR2 antagonist to function as a therapeutic drug in in vitro human and ex vivo bovine pilot models of disease. MC migration was quantitatively assessed using conditioned media from primary human IVD cells and MC migration examined through Matrigel. Exposure to soluble IVD factors significantly enhanced MC migration, suggesting IVD cells can recruit MCs. We also demonstrated significant upregulation of MC chemokine SCF and angiogenic factor VEGFA gene expression in human IVD cells in vitro in response to recombinant human tryptase, suggesting tryptase can enhance recruitment of MCs and promotion of angiogenesis into the usually avascular IVD. Furthermore, tryptase can degrade proteoglycans in IVD tissue as demonstrated by significant increases in glycosaminoglycans released into surrounding media. This can create a catabolic microenvironment compromising structural integrity and facilitating vascular migration usually inhibited by the anti-angiogenic IVD matrix. Finally, as a “proof of concept” study, we examined the therapeutic potential of PAR2 antagonist (PAR2A) on human IVD cells and bovine organ culture IVD model. While preliminary data shows promise and points toward structural restoration of the bovine IVD including down-regulation of VEGFA, effects of PAR2 antagonist on human IVD cells differ between gender and donors suggesting that further validation is required with larger cohorts of human specimens.
Collapse
Affiliation(s)
- Justin Richards
- College of Arts and Sciences, The Ohio State University, Columbus, OH, United States
| | - Shirley Tang
- College of Engineering, The Ohio State University, Columbus, OH, United States
| | - Gilian Gunsch
- College of Arts and Sciences, The Ohio State University, Columbus, OH, United States
| | - Pavel Sul
- College of Engineering, The Ohio State University, Columbus, OH, United States
| | - Matthew Wiet
- College of Engineering, The Ohio State University, Columbus, OH, United States
| | - David C Flanigan
- Department of Orthopaedics, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Safdar N Khan
- Department of Orthopaedics, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Sarah Moore
- College of Veterinary Medicine, The Ohio State University, Columbus, OH, United States
| | - Benjamin Walter
- College of Engineering, The Ohio State University, Columbus, OH, United States.,Department of Orthopaedics, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Devina Purmessur
- College of Engineering, The Ohio State University, Columbus, OH, United States.,Department of Orthopaedics, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
27
|
Intact mast cell content during mild head injury is required for development of latent pain sensitization: implications for mechanisms underlying post-traumatic headache. Pain 2019; 160:1050-1058. [PMID: 30624345 DOI: 10.1097/j.pain.0000000000001481] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Post-traumatic headache (PTH) is one of the most common, debilitating, and difficult symptoms to manage after a traumatic head injury. Although the mechanisms underlying PTH remain elusive, recent studies in rodent models suggest the potential involvement of calcitonin gene-related peptide (CGRP), a mediator of neurogenic inflammation, and the ensuing activation of meningeal mast cells (MCs), proalgesic resident immune cells that can lead to the activation of the headache pain pathway. Here, we investigated the relative contribution of MCs to the development of PTH-like pain behaviors in a model of mild closed-head injury (mCHI) in male rats. We initially tested the relative contribution of peripheral CGRP signaling to the activation of meningeal MCs after mCHI using a blocking anti-CGRP monoclonal antibody. We then used a prophylactic MC granule depletion approach to address the hypotheses that intact meningeal MC granule content is necessary for the development of PTH-related pain-like behaviors. The data suggest that after mCHI, ongoing activation of meningeal MCs is not mediated by peripheral CGRP signaling and does not contribute to the development of the mCHI-evoked cephalic mechanical pain hypersensitivity. Our data, however, also reveal that the development of latent sensitization, manifested as persistent hypersensitivity upon the recovery from mCHI-evoked acute cranial hyperalgesia to the headache trigger glyceryl trinitrate requires intact MC content during and immediately after mCHI. Collectively, our data implicate the acute activation of meningeal MCs as mediator of chronic pain hypersensitivity after a concussion or mCHI. Targeting MCs may be explored for early prophylactic treatment of PTH.
Collapse
|
28
|
Salzer I, Boehm S. Calcium-activated chloride channels: Potential targets for antinociceptive therapy. Int J Biochem Cell Biol 2019; 111:37-41. [PMID: 31005634 DOI: 10.1016/j.biocel.2019.04.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 04/13/2019] [Accepted: 04/17/2019] [Indexed: 01/24/2023]
Abstract
The molecular identity of calcium-activated chloride channels (CaCCs) was clarified only some ten years ago when it was linked to the family of "transmembrane proteins of unknown function 16″ (TMEM16). Since then, numerous studies have been conducted both to define their role in physiology and identify their biophysical functions. For the latter, the ultrastructural description of mouse TMEM16 A was a breakthrough. CaCCs were functionally described in a number of different tissues including first-order sensory neurons. The activating rise in intracellular calcium concentration can be caused by an influx of calcium through other calcium permeable ion channels. Calcium release from intracellular stores, mediated by G-protein coupled receptors, also leads to CaCC activation. Prominent inflammatory mediators like bradykinin or serotonin stimulate CaCCs via such a mechanism. The (patho) physiological function of these ion channels renders them promising targets for antinociceptive treatment.
Collapse
Affiliation(s)
- Isabella Salzer
- Division of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, 1090, Vienna, Austria.
| | - Stefan Boehm
- Division of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, 1090, Vienna, Austria
| |
Collapse
|
29
|
Li ZH, Cui D, Qiu CJ, Song XJ. Cyclic nucleotide signaling in sensory neuron hyperexcitability and chronic pain after nerve injury. NEUROBIOLOGY OF PAIN 2019; 6:100028. [PMID: 31223142 PMCID: PMC6565612 DOI: 10.1016/j.ynpai.2019.100028] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Revised: 03/06/2019] [Accepted: 03/07/2019] [Indexed: 11/08/2022]
Abstract
Activation of cAMP-PKA and cGMP-PKG pathways contributes to injury-induced sensory neuron hyperexcitability. Activation of cAMP and cGMP contributes to the development of bone cancer pain. PAR2 activation mediates injury-induced cAMP-dependent sensory neuron hyperexcitability.
The cyclic nucleotide signaling, including cAMP-PKA and cGMP-PKG pathways, has been well known to play critical roles in regulating cellular growth, metabolism and many other intracellular processes. In recent years, more and more studies have uncovered the roles of cAMP and cGMP in the nervous system. The cAMP and cGMP signaling mediates chronic pain induced by different forms of injury and stress. Here we summarize the roles of cAMP-PKA and cGMP-PKG signaling pathways in the pathogenesis of chronic pain after nerve injury. In addition, acute dissociation and chronic compression of the dorsal root ganglion (DRG) neurons, respectively, leads to neural hyperexcitability possibly through PAR2 activation-dependent activation of cAMP-PKA pathway. Clinically, radiotherapy can effectively alleviate bone cancer pain at least partly through inhibiting the cancer cell-induced activation of cAMP-PKA pathway. Roles of cyclic nucleotide signaling in neuropathic and inflammatory pain are also seen in many other animal models and are involved in many pro-nociceptive mechanisms including the activation of hyperpolarization-activated cyclic nucleotide (HCN)-modulated ion channels and the exchange proteins directly activated by cAMP (EPAC). Further understanding the roles of cAMP and cGMP signaling in the pathogenesis of chronic pain is theoretically significant and clinically valuable for treatment of chronic pain.
Collapse
Affiliation(s)
- Ze-Hua Li
- Department of Biology, SUSTech Center for Pain Medicine, and Medical School, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China.,Department of Anesthesiology and Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education of China), Peking University School of Oncology, Beijing Cancer Hospital & Institute, Beijing 100142, China
| | - Dong Cui
- Department of Biology, SUSTech Center for Pain Medicine, and Medical School, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China.,Department of Anesthesiology and Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education of China), Peking University School of Oncology, Beijing Cancer Hospital & Institute, Beijing 100142, China
| | - Cheng-Jie Qiu
- Department of Biology, SUSTech Center for Pain Medicine, and Medical School, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Xue-Jun Song
- Department of Biology, SUSTech Center for Pain Medicine, and Medical School, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China.,Department of Anesthesiology and Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education of China), Peking University School of Oncology, Beijing Cancer Hospital & Institute, Beijing 100142, China
| |
Collapse
|
30
|
Hassler SN, Ahmad FB, Burgos-Vega CC, Boitano S, Vagner J, Price TJ, Dussor G. Protease activated receptor 2 (PAR2) activation causes migraine-like pain behaviors in mice. Cephalalgia 2019; 39:111-122. [PMID: 29848111 PMCID: PMC6081257 DOI: 10.1177/0333102418779548] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
BACKGROUND Pain is the most debilitating symptom of migraine. The cause of migraine pain likely requires activation of meningeal nociceptors. Mast cell degranulation, with subsequent meningeal nociceptor activation, has been implicated in migraine pathophysiology. Degranulating mast cells release serine proteases that can cleave and activate protease activated receptors. The purpose of these studies was to investigate whether protease activated receptor 2 is a potential generator of nociceptive input from the meninges by using selective pharmacological agents and knockout mice. METHODS Ratiometric Ca++ imaging was performed on primary trigeminal and dural cell cultures after application of 2at-LIGRL-NH2, a specific protease activated receptor 2 agonist. Cutaneous hypersensitivity and facial grimace was measured in wild-type and protease activated receptor 2-/- mice after dural application of 2at-LIGRL-NH2 or compound 48-80, a mast cell degranulator. Behavioral experiments were also conducted in mice after dural application of 2at-LIGRL-NH2 (2AT) in the presence of either C391, a selective protease activated receptor 2 antagonist, or sumatriptan. RESULTS 2at-LIGRL-NH2 evoked Ca2+ signaling in mouse trigeminal neurons, dural fibroblasts and in meningeal afferents. Dural application of 2at-LIGRL-NH2 or 48-80 caused dose-dependent grimace behavior and mechanical allodynia that were attenuated by either local or systemic application of C391 as well as in protease activated receptor 2-/- mice. Nociceptive behavior after dural injection of 2at-LIGRL-NH2 was also attenuated by sumatriptan. CONCLUSIONS Functional protease activated receptor 2 receptors are expressed on both dural afferents and fibroblasts and activation of dural protease activated receptor 2 produces migraine-like behavioral responses. Protease activated receptor 2 may link resident immune cells to meningeal nociceptor activation, driving migraine-like pain and implicating protease activated receptor 2 as a therapeutic target for migraine in humans.
Collapse
Affiliation(s)
- Shayne N Hassler
- School of Brain and Behavioral Sciences, University of Texas at Dallas, TX, USA
| | - Fatima B Ahmad
- School of Brain and Behavioral Sciences, University of Texas at Dallas, TX, USA
| | | | - Scott Boitano
- Department of Physiology, University of Arizona, AZ, USA
- Arizona Respiratory Center, University of Arizona, AZ, USA
- Bio5 Institute, University of Arizona, AZ, USA
| | | | - Theodore J Price
- School of Brain and Behavioral Sciences, University of Texas at Dallas, TX, USA
| | - Gregory Dussor
- School of Brain and Behavioral Sciences, University of Texas at Dallas, TX, USA
| |
Collapse
|
31
|
Chamessian A, Young M, Qadri Y, Berta T, Ji RR, Van de Ven T. Transcriptional Profiling of Somatostatin Interneurons in the Spinal Dorsal Horn. Sci Rep 2018; 8:6809. [PMID: 29717160 PMCID: PMC5931607 DOI: 10.1038/s41598-018-25110-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 04/13/2018] [Indexed: 01/08/2023] Open
Abstract
The spinal dorsal horn (SDH) is comprised of distinct neuronal populations that process different somatosensory modalities. Somatostatin (SST)-expressing interneurons in the SDH have been implicated specifically in mediating mechanical pain. Identifying the transcriptomic profile of SST neurons could elucidate the unique genetic features of this population and enable selective analgesic targeting. To that end, we combined the Isolation of Nuclei Tagged in Specific Cell Types (INTACT) method and Fluorescence Activated Nuclei Sorting (FANS) to capture tagged SST nuclei in the SDH of adult male mice. Using RNA-sequencing (RNA-seq), we uncovered more than 13,000 genes. Differential gene expression analysis revealed more than 900 genes with at least 2-fold enrichment. In addition to many known dorsal horn genes, we identified and validated several novel transcripts from pharmacologically tractable functional classes: Carbonic Anhydrase 12 (Car12), Phosphodiesterase 11 A (Pde11a), and Protease-Activated Receptor 3 (F2rl2). In situ hybridization of these novel genes showed differential expression patterns in the SDH, demonstrating the presence of transcriptionally distinct subpopulations within the SST population. Overall, our findings provide new insights into the gene repertoire of SST dorsal horn neurons and reveal several novel targets for pharmacological modulation of this pain-mediating population and treatment of pathological pain.
Collapse
Affiliation(s)
- Alexander Chamessian
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, 27710, USA. .,Medical Scientist Training Program, Duke University School of Medicine, Durham, North Carolina, 27710, USA. .,Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, 27710, USA.
| | - Michael Young
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina, 27710, USA
| | - Yawar Qadri
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, 27710, USA
| | - Temugin Berta
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, Ohio, 45267, USA
| | - Ru-Rong Ji
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, 27710, USA.,Department of Neurobiology, Duke University Medical Center, Durham, North Carolina, 27710, USA
| | - Thomas Van de Ven
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, 27710, USA
| |
Collapse
|
32
|
Moy JK, Khoutorsky A, Asiedu MN, Dussor G, Price TJ. eIF4E Phosphorylation Influences Bdnf mRNA Translation in Mouse Dorsal Root Ganglion Neurons. Front Cell Neurosci 2018; 12:29. [PMID: 29467623 PMCID: PMC5808250 DOI: 10.3389/fncel.2018.00029] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Accepted: 01/23/2018] [Indexed: 01/19/2023] Open
Abstract
Plasticity in dorsal root ganglion (DRG) neurons that promotes pain requires activity-dependent mRNA translation. Protein synthesis inhibitors block the ability of many pain-promoting molecules to enhance excitability in DRG neurons and attenuate behavioral signs of pain plasticity. In line with this, we have recently shown that phosphorylation of the 5′ cap-binding protein, eIF4E, plays a pivotal role in plasticity of DRG nociceptors in models of hyperalgesic priming. However, mRNA targets of eIF4E phosphorylation have not been elucidated in the DRG. Brain-derived neurotrophic factor (BDNF) signaling from nociceptors in the DRG to spinal dorsal horn neurons is an important mediator of hyperalgesic priming. Regulatory mechanisms that promote pain plasticity via controlling BDNF expression that is involved in promoting pain plasticity have not been identified. We show that phosphorylation of eIF4E is paramount for Bdnf mRNA translation in the DRG. Bdnf mRNA translation is reduced in mice lacking eIF4E phosphorylation (eIF4ES209A) and pro-nociceptive factors fail to increase BDNF protein levels in the DRGs of these mice despite robust upregulation of Bdnf-201 mRNA levels. Importantly, bypassing the DRG by giving intrathecal injection of BDNF in eIF4ES209A mice creates a strong hyperalgesic priming response that is normally absent or reduced in these mice. We conclude that eIF4E phosphorylation-mediated translational control of BDNF expression is a key mechanism for nociceptor plasticity leading to hyperalgesic priming.
Collapse
Affiliation(s)
- Jamie K Moy
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, United States.,Department of Pharmacology, University of Arizona, Tucson, AZ, United States
| | - Arkady Khoutorsky
- Department of Anesthesia, McGill University, Montréal, QC, Canada.,Alan Edwards Centre for Research on Pain, McGill University, Montréal, QC, Canada
| | - Marina N Asiedu
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, United States.,Department of Pharmacology, University of Arizona, Tucson, AZ, United States
| | - Gregory Dussor
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, United States.,Department of Pharmacology, University of Arizona, Tucson, AZ, United States
| | - Theodore J Price
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, United States.,Department of Pharmacology, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
33
|
Domenichiello AF, Wilhite BC, Keyes GS, Ramsden CE. A dose response study of the effect of prostaglandin E2 on thermal nociceptive sensitivity. Prostaglandins Leukot Essent Fatty Acids 2017; 126:20-24. [PMID: 29031391 PMCID: PMC5679719 DOI: 10.1016/j.plefa.2017.08.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 08/27/2017] [Accepted: 08/28/2017] [Indexed: 12/31/2022]
Abstract
Inhibition of prostaglandin (PG) biosynthesis has been used to relieve pain for thousands of years. Today non-steroidal anti-inflammatory drugs (which largely inhibit PG synthesis) are widely used to treat pain. Four main types of PGs (PGD2, PGE2, PGF2 and PGI2) are synthesized from arachidonic acid during inflammation and have been demonstrated to impact nociception. PGE2 has been the most studied and utilized for its pain producing properties and has been demonstrated to increase hypersensitivity in rodent nociceptive behavioral models when applied centrally and/or peripherally. Surprisingly, there are no published reports that use withdrawal from radiant light beam (Hargreaves apparatus) to examine the dose response effect of peripherally applied PGE2 on thermal nociceptive hypersensitivity. To address this gap in the literature, we performed a dose response study examining the effect of PGE2 on thermal hypersensitivity (assessed using a Hargreaves apparatus) where rats were injected with 0.003-30μg of PGE2, intradermally into the hindpaw. Thermal hypersensitivity was assessed by measuring withdraw latency from a radiant light beam (Hargreaves test) and our primary objective was to determine the dose of PGE2 causing the most pronounced increase in thermal hypersensitivity (i.e. lowest withdraw latency). A secondary objective was to determine the minimum dose of PGE2 required to cause statistically significant decreases in thermal withdrawal latency as compared to rats injected with vehicle. We found that rats injected with the 30μg dose of PGE2 exhibited the most pronounced thermal nociceptive hypersensitivity though secondary analysis showed that rats injected with PGE2 doses of 0.03-30μg had lower withdrawal latencies as compared to rats injected with vehicle. This work fills an evidence gap and provides context to guide dose selection in future rodent pain behavior studies.
Collapse
Affiliation(s)
- Anthony F Domenichiello
- Lipid Mediators, Inflammation and Pain Unit, Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health (NIH), Baltimore, MD, United States.
| | - Breanne C Wilhite
- Section of Nutritional Neuroscience, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, NIH, United States
| | - Gregory S Keyes
- Lipid Mediators, Inflammation and Pain Unit, Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health (NIH), Baltimore, MD, United States
| | - Christopher E Ramsden
- Lipid Mediators, Inflammation and Pain Unit, Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health (NIH), Baltimore, MD, United States
| |
Collapse
|
34
|
Wu J, Liu TT, Zhou YM, Qiu CY, Ren P, Jiao M, Hu WP. Sensitization of ASIC3 by proteinase-activated receptor 2 signaling contributes to acidosis-induced nociception. J Neuroinflammation 2017; 14:150. [PMID: 28754162 PMCID: PMC5534107 DOI: 10.1186/s12974-017-0916-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 07/11/2017] [Indexed: 02/07/2023] Open
Abstract
Background Tissue acidosis and inflammatory mediators play critical roles in pain. Pro-inflammatory agents trypsin and tryptase cleave and activate proteinase-activated receptor 2 (PAR2) expressed on sensory nerves, which is involved in peripheral mechanisms of inflammation and pain. Extracellular acidosis activates acid-sensing ion channel 3 (ASIC3) to trigger pain sensation. Here, we show that a functional interaction of PAR2 and ASIC3 could contribute to acidosis-induced nociception. Methods Electrophysiological experiments were performed on both rat DRG neurons and Chinese hamster ovary (CHO) cells expressing ASIC3 and PAR2. Nociceptive behavior was induced by acetic acid in rats. Results PAR2-AP, PAR2-activating peptide, concentration-dependently increased the ASIC3 currents in CHO cells transfected with ASIC3 and PAR2. The proton concentration–response relationship was not changed, but that the maximal response increased 58.7 ± 3.8% after pretreatment of PAR2-AP. PAR2 mediated the potentiation of ASIC3 currents via an intracellular cascade. PAR2-AP potentiation of ASIC3 currents disappeared after inhibition of intracellular G protein, PLC, PKC, or PKA signaling. Moreover, PAR2 activation increased proton-evoked currents and spikes mediated by ASIC3 in rat dorsal root ganglion neurons. Finally, peripheral administration of PAR2-AP dose-dependently exacerbated acidosis-induced nocifensive behaviors in rats. Conclusions These results indicated that PAR2 signaling sensitized ASIC3, which may contribute to acidosis-induced nociception. These represent a novel peripheral mechanism underlying PAR2 involvement in hyperalgesia by sensitizing ASIC3 in primary sensory neurons.
Collapse
Affiliation(s)
- Jing Wu
- Research Center of Basic Medical Sciences, School of Basic Medical Sciences, Hubei University of Science and Technology, 88 Xianning Road, Xianning, 437100, Hubei, People's Republic of China
| | - Ting-Ting Liu
- Research Center of Basic Medical Sciences, School of Basic Medical Sciences, Hubei University of Science and Technology, 88 Xianning Road, Xianning, 437100, Hubei, People's Republic of China.,Department of Physiology, School of Basic Medical Sciences, Hubei University of Science and Technology, 88 Xianning Road, Xianning, 437100, Hubei, People's Republic of China
| | - Yi-Mei Zhou
- Research Center of Basic Medical Sciences, School of Basic Medical Sciences, Hubei University of Science and Technology, 88 Xianning Road, Xianning, 437100, Hubei, People's Republic of China
| | - Chun-Yu Qiu
- Department of Pharmacology, Hubei University of Science and Technology, 88 Xianning Road, Xianning, 437100, Hubei, People's Republic of China
| | - Ping Ren
- Department of Pharmacology, Hubei University of Science and Technology, 88 Xianning Road, Xianning, 437100, Hubei, People's Republic of China
| | - Ming Jiao
- Research Center of Basic Medical Sciences, School of Basic Medical Sciences, Hubei University of Science and Technology, 88 Xianning Road, Xianning, 437100, Hubei, People's Republic of China.
| | - Wang-Ping Hu
- Research Center of Basic Medical Sciences, School of Basic Medical Sciences, Hubei University of Science and Technology, 88 Xianning Road, Xianning, 437100, Hubei, People's Republic of China. .,Department of Physiology, School of Basic Medical Sciences, Hubei University of Science and Technology, 88 Xianning Road, Xianning, 437100, Hubei, People's Republic of China.
| |
Collapse
|
35
|
The MNK-eIF4E Signaling Axis Contributes to Injury-Induced Nociceptive Plasticity and the Development of Chronic Pain. J Neurosci 2017; 37:7481-7499. [PMID: 28674170 DOI: 10.1523/jneurosci.0220-17.2017] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 05/21/2017] [Accepted: 06/27/2017] [Indexed: 12/18/2022] Open
Abstract
Injury-induced sensitization of nociceptors contributes to pain states and the development of chronic pain. Inhibiting activity-dependent mRNA translation through mechanistic target of rapamycin and mitogen-activated protein kinase (MAPK) pathways blocks the development of nociceptor sensitization. These pathways convergently signal to the eukaryotic translation initiation factor (eIF) 4F complex to regulate the sensitization of nociceptors, but the details of this process are ill defined. Here we investigated the hypothesis that phosphorylation of the 5' cap-binding protein eIF4E by its specific kinase MAPK interacting kinases (MNKs) 1/2 is a key factor in nociceptor sensitization and the development of chronic pain. Phosphorylation of ser209 on eIF4E regulates the translation of a subset of mRNAs. We show that pronociceptive and inflammatory factors, such as nerve growth factor (NGF), interleukin-6 (IL-6), and carrageenan, produce decreased mechanical and thermal hypersensitivity, decreased affective pain behaviors, and strongly reduced hyperalgesic priming in mice lacking eIF4E phosphorylation (eIF4ES209A ). Tests were done in both sexes, and no sex differences were found. Moreover, in patch-clamp electrophysiology and Ca2+ imaging experiments on dorsal root ganglion neurons, NGF- and IL-6-induced increases in excitability were attenuated in neurons from eIF4ES209A mice. These effects were recapitulated in Mnk1/2-/- mice and with the MNK1/2 inhibitor cercosporamide. We also find that cold hypersensitivity induced by peripheral nerve injury is reduced in eIF4ES209A and Mnk1/2-/- mice and following cercosporamide treatment. Our findings demonstrate that the MNK1/2-eIF4E signaling axis is an important contributing factor to mechanisms of nociceptor plasticity and the development of chronic pain.SIGNIFICANCE STATEMENT Chronic pain is a debilitating disease affecting approximately one in three Americans. Chronic pain is thought to be driven by changes in the excitability of peripheral nociceptive neurons, but the precise mechanisms controlling these changes are not elucidated. Emerging evidence demonstrates that mRNA translation regulation pathways are key factors in changes in nociceptor excitability. Our work demonstrates that a single phosphorylation site on the 5' cap-binding protein eIF4E is a critical mechanism for changes in nociceptor excitability that drive the development of chronic pain. We reveal a new mechanistic target for the development of a chronic pain state and propose that targeting the upstream kinase, MAPK interacting kinase 1/2, could be used as a therapeutic approach for chronic pain.
Collapse
|
36
|
Neuroligin 2 regulates spinal GABAergic plasticity in hyperalgesic priming, a model of the transition from acute to chronic pain. Pain 2017; 157:1314-1324. [PMID: 26859820 DOI: 10.1097/j.pain.0000000000000513] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Plasticity in inhibitory receptors, neurotransmission, and networks is an important mechanism for nociceptive signal amplification in the spinal dorsal horn. We studied potential changes in GABAergic pharmacology and its underlying mechanisms in hyperalgesic priming, a model of the transition from acute to chronic pain. We find that while GABAA agonists and positive allosteric modulators reduce mechanical hypersensitivity to an acute insult, they fail to do so during the maintenance phase of hyperalgesic priming. In contrast, GABAA antagonism promotes antinociception and a reduction in facial grimacing after the transition to a chronic pain state. During the maintenance phase of hyperalgesic priming, we observed increased neuroligin (nlgn) 2 expression in the spinal dorsal horn. This protein increase was associated with an increase in nlgn2A splice variant mRNA, which promotes inhibitory synaptogenesis. Disruption of nlgn2 function with the peptide inhibitor, neurolide 2, produced mechanical hypersensitivity in naive mice but reversed hyperalgesic priming in mice previously exposed to brain-derived neurotrophic factor. Neurolide 2 treatment also reverses the change in polarity in GABAergic pharmacology observed in the maintenance of hyperalgesic priming. We propose that increased nlgn2 expression is associated with hyperalgesic priming where it promotes dysregulation of inhibitory networks. Our observations reveal new mechanisms involved in the spinal maintenance of a pain plasticity and further suggest that disinhibitory mechanisms are central features of neuroplasticity in the spinal dorsal horn.
Collapse
|
37
|
Shavit-Stein E, Artan-Furman A, Feingold E, Ben Shimon M, Itzekson-Hayosh Z, Chapman J, Vlachos A, Maggio N. Protease Activated Receptor 2 (PAR2) Induces Long-Term Depression in the Hippocampus through Transient Receptor Potential Vanilloid 4 (TRPV4). Front Mol Neurosci 2017; 10:42. [PMID: 28303089 PMCID: PMC5332813 DOI: 10.3389/fnmol.2017.00042] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 02/07/2017] [Indexed: 11/25/2022] Open
Abstract
Protease activated receptors (PARs) are involved in regulating synaptic transmission and plasticity in the brain. While it is well-accepted that PAR1 mediates long-term potentiation (LTP) of excitatory synaptic strength, the role of PAR2 in synaptic plasticity remains not well-understood. In this study, we assessed the role of PAR2-signaling in plasticity at hippocampal Schaffer collateral-CA1 synapses. Using field potential recordings, we report that PAR2-activation leads to long-term depression (LTD) of synaptic transmission through a protein kinase A -dependent, Transient Receptor Potential Vanilloid 4 -mediated mechanism, which requires the activation of N-methyl-D-aspartate receptors. These results demonstrate that the effects of PAR2 on synaptic plasticity are distinct from what is observed upon PAR1-activation. Thus, we propose that the activation of different classes of PARs, i.e., PAR1 and PAR2, may set the threshold of synaptic plasticity in the hippocampal network by balancing LTP and LTD.
Collapse
Affiliation(s)
- Efrat Shavit-Stein
- Department of Neurology, The Chaim Sheba Medical Center Tel HaShomer, Israel
| | - Avital Artan-Furman
- Department of Neurology, The Chaim Sheba Medical Center Tel HaShomer, Israel
| | - Ekaterina Feingold
- Department of Neurology, The Chaim Sheba Medical Center Tel HaShomer, Israel
| | - Marina Ben Shimon
- Department of Neurology, The Chaim Sheba Medical CenterTel HaShomer, Israel; Department of Neurology and Neurosurgery, Sackler Faculty of Medicine, Tel Aviv UniversityTel Aviv, Israel
| | | | - Joab Chapman
- Department of Neurology, The Chaim Sheba Medical CenterTel HaShomer, Israel; Department of Neurology and Neurosurgery, Sackler Faculty of Medicine, Tel Aviv UniversityTel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv UniversityTel Aviv, Israel
| | - Andreas Vlachos
- Institute of Anatomy II, Faculty of Medicine, Heinrich-Heine-University Duesseldorf, Germany
| | - Nicola Maggio
- Department of Neurology, The Chaim Sheba Medical CenterTel HaShomer, Israel; Department of Neurology and Neurosurgery, Sackler Faculty of Medicine, Tel Aviv UniversityTel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv UniversityTel Aviv, Israel; Talpiot Medical Leadership Program, The Chaim Sheba Medical CenterTel HaShomer, Israel
| |
Collapse
|
38
|
Meng J, Steinhoff M. Molecular mechanisms of pruritus. Curr Res Transl Med 2016; 64:203-206. [PMID: 27939459 DOI: 10.1016/j.retram.2016.08.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 08/16/2016] [Indexed: 02/08/2023]
Abstract
Pruritus is an unpleasant sensation that evokes the urgent desire to scratch. It is a symptom derived from many nervous system disorders that affects a large population of humans and is treated by a variety of pharmacological agents with variable access. Chronic itch is a huge unmet health problem which affect upward 20% of people worldwide. The mechanisms underlying the chronic pruritus are complex. Studies of the neurobiology, neurophysiology and cellular biology of itch have gradually been clarifying the mechanism of chronic itch both peripherally and centrally. The discussion has been focused on pruriceptive nerves and their receptors as well as the cytokines/chemokines that play major roles in itch induction. Though it is historically hypothesized that pain convey signal generated with the stimuli under high intensity, and itch transduces signal from the same nerves of pain but under low intensity, recently, with the identification of distinct itch specific sensory afferent fibers the theory has twisted the "intensity" to a existence of a complete separation of pain and itch pathways. This review helps to understand the unique properties of itch signaling pathways and their clinical importance of the itch perception and pruritic diseases.
Collapse
Affiliation(s)
- J Meng
- Department of Dermatology and UCD Charles Institute for Translational Dermatology, Dublin, Ireland.
| | - M Steinhoff
- Department of Dermatology and UCD Charles Institute for Translational Dermatology, Dublin, Ireland.
| |
Collapse
|
39
|
Farzaei MH, Bahramsoltani R, Abdollahi M, Rahimi R. The Role of Visceral Hypersensitivity in Irritable Bowel Syndrome: Pharmacological Targets and Novel Treatments. J Neurogastroenterol Motil 2016; 22:558-574. [PMID: 27431236 PMCID: PMC5056566 DOI: 10.5056/jnm16001] [Citation(s) in RCA: 123] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 03/26/2016] [Accepted: 04/17/2016] [Indexed: 12/13/2022] Open
Abstract
Irritable bowel syndrome (IBS) is the most common disorder referred to gastroenterologists and is characterized by altered bowel habits, abdominal pain, and bloating. Visceral hypersensitivity (VH) is a multifactorial process that may occur within the peripheral or central nervous systems and plays a principal role in the etiology of IBS symptoms. The pharmacological studies on selective drugs based on targeting specific ligands can provide novel therapies for modulation of persistent visceral hyperalgesia. The current paper reviews the cellular and molecular mechanisms underlying therapeutic targeting for providing future drugs to protect or treat visceroperception and pain sensitization in IBS patients. There are a wide range of mediators and receptors participating in visceral pain perception amongst which substances targeting afferent receptors are attractive sources of novel drugs. Novel therapeutic targets for the management of VH include compounds which alter gut-brain pathways and local neuroimmune pathways. Molecular mediators and receptors participating in pain perception and visceroperception include histamine-1 receptors, serotonin (5-hydrodytryptamine) receptors, transient receptor potential vanilloid type I, tachykinins ligands, opioid receptors, voltage-gated channels, tyrosine receptor kinase receptors, protease-activated receptors, adrenergic system ligands, cannabinoid receptors, sex hormones, and glutamate receptors which are discussed in the current review. Moreover, several plant-derived natural compounds with potential to alleviate VH in IBS have been highlighted. VH has an important role in the pathology and severity of complications in IBS. Therefore, managing VH can remarkably modulate the symptoms of IBS. More preclinical and clinical investigations are needed to provide efficacious and targeted medicines for the management of VH.
Collapse
Affiliation(s)
- Mohammad H Farzaei
- Pharmaceutical Sciences Research Center, Kermanshah University of Medical Sciences, Kermanshah,
Iran
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah,
Iran
| | - Roodabeh Bahramsoltani
- Faculty of Pharmacy and Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran,
Iran
| | - Mohammad Abdollahi
- Faculty of Pharmacy and Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran,
Iran
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran,
Iran
| | - Roja Rahimi
- Department of Traditional Pharmacy, School of Traditional Medicine, Tehran University of Medical Sciences, Tehran,
Iran
| |
Collapse
|
40
|
Mrozkova P, Spicarova D, Palecek J. Hypersensitivity Induced by Activation of Spinal Cord PAR2 Receptors Is Partially Mediated by TRPV1 Receptors. PLoS One 2016; 11:e0163991. [PMID: 27755539 PMCID: PMC5068818 DOI: 10.1371/journal.pone.0163991] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 09/19/2016] [Indexed: 12/23/2022] Open
Abstract
Protease-activated receptors 2 (PAR2) and transient receptor potential vanilloid 1 (TRPV1) receptors in the peripheral nerve endings are implicated in the development of increased sensitivity to mechanical and thermal stimuli, especially during inflammatory states. Both PAR2 and TRPV1 receptors are co-expressed in nociceptive dorsal root ganglion (DRG) neurons on their peripheral endings and also on presynaptic endings in the spinal cord dorsal horn. However, the modulation of nociceptive synaptic transmission in the superficial dorsal horn after activation of PAR2 and their functional coupling with TRPV1 is not clear. To investigate the role of spinal PAR2 activation on nociceptive modulation, intrathecal drug application was used in behavioural experiments and patch-clamp recordings of spontaneous, miniature and dorsal root stimulation-evoked excitatory postsynaptic currents (sEPSCs, mEPSCs, eEPSCs) were performed on superficial dorsal horn neurons in acute rat spinal cord slices. Intrathecal application of PAR2 activating peptide SLIGKV-NH2 induced thermal hyperalgesia, which was prevented by pretreatment with TRPV1 antagonist SB 366791 and was reduced by protein kinases inhibitor staurosporine. Patch-clamp experiments revealed robust decrease of mEPSC frequency (62.8 ± 4.9%), increase of sEPSC frequency (127.0 ± 5.9%) and eEPSC amplitude (126.9 ± 12.0%) in dorsal horn neurons after acute SLIGKV-NH2 application. All these EPSC changes, induced by PAR2 activation, were prevented by SB 366791 and staurosporine pretreatment. Our results demonstrate an important role of spinal PAR2 receptors in modulation of nociceptive transmission in the spinal cord dorsal horn at least partially mediated by activation of presynaptic TRPV1 receptors. The functional coupling between the PAR2 and TRPV1 receptors on the central branches of DRG neurons may be important especially during different pathological states when it may enhance pain perception.
Collapse
Affiliation(s)
- Petra Mrozkova
- Department of Functional Morphology, Institute of Physiology, The Czech Academy of Sciences, Prague, Czech Republic
| | - Diana Spicarova
- Department of Functional Morphology, Institute of Physiology, The Czech Academy of Sciences, Prague, Czech Republic
| | - Jiri Palecek
- Department of Functional Morphology, Institute of Physiology, The Czech Academy of Sciences, Prague, Czech Republic
- * E-mail:
| |
Collapse
|
41
|
Chodroff L, Bendele M, Valenzuela V, Henry M, Ruparel S. EXPRESS: BDNF Signaling Contributes to Oral Cancer Pain in a Preclinical Orthotopic Rodent Model. Mol Pain 2016; 12:12/0/1744806916666841. [PMID: 27590070 PMCID: PMC5015823 DOI: 10.1177/1744806916666841] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The majority of patients with oral cancer report intense pain that is only partially managed by current analgesics. Thus, there is a strong need to study mechanisms as well as develop novel analgesics for oral cancer pain. Current study employed an orthotopic tongue cancer model with molecular and non-reflexive behavioral assays to determine possible mechanisms of oral cancer pain. Human oral squamous cell carcinoma cells line, HSC2, was injected into the tongue of male athymic mice and tumor growth was observed by day 6. Immunohistological analyses revealed a well-differentiated tumor with a localized immune response and pronounced sensory and sympathetic innervation and vascularization. The tumor expressed TMPRSS2, a protein previously reported with oral squamous cell carcinoma. ATF3 expression in trigeminal ganglia was not altered by tumor growth. Molecular characterization of the model demonstrated altered expression of several pain-related genes, out of which up-regulation of BDNF was most striking. Moreover, BDNF protein expression in trigeminal ganglia neurons was increased and inhibition of BDNF signaling with a tyrosine kinase B antagonist, ANA-12, reversed pain-like behaviors induced by the oral tumor. Oral squamous cell carcinoma tumor growth was also associated with a reduction in feeding, mechanical hypersensitivity in the face, as well as spontaneous pain behaviors as measured by the conditioned place preference test, all of which were reversed by analgesics. Interestingly, injection of HSC2 into the hindpaw did not reproduce this spectrum of pain behaviors; nor did injection of a colonic cancer cell line into the tongue. Taken together, this orthotopic oral cancer pain model reproduces the spectrum of pain reported by oral cancer patients, including higher order cognitive changes, and demonstrates that BDNF signaling constitutes a novel mechanism by which oral squamous cell carcinoma induces pain. Identification of the key role of tyrosine kinase B signaling in oral cancer pain may serve as a novel target for drug development.
Collapse
Affiliation(s)
- Leah Chodroff
- University of Texas Health Science Center at San Antonio
| | - Michele Bendele
- University of Texas Health Science Center at San Antonio Cancer Therapy and Research Center
| | | | - Michael Henry
- University of Texas Health Science Center At San Antonio
| | - Shivani Ruparel
- University of Texas Health Science Center at San AntonioUniversity of Texas Health Science Center at San Antonio Cancer Therapy and Research CenterUniversity of Texas Health Science Center at San AntonioUniversity of Texas Health Science Center At San Antonio
| |
Collapse
|
42
|
Burma NE, Leduc-Pessah H, Fan CY, Trang T. Animal models of chronic pain: Advances and challenges for clinical translation. J Neurosci Res 2016; 95:1242-1256. [PMID: 27376591 DOI: 10.1002/jnr.23768] [Citation(s) in RCA: 119] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 04/25/2016] [Accepted: 04/29/2016] [Indexed: 12/17/2022]
Abstract
Chronic pain is a global problem that has reached epidemic proportions. An estimated 20% of adults suffer from pain, and another 10% are diagnosed with chronic pain each year (Goldberg and McGee, ). Despite the high prevalence of chronic pain (an estimated 1.5 billion people are afflicted worldwide), much remains to be understood about the underlying causes of this condition, and there is an urgent requirement for better pain therapies. The discovery of novel targets and the development of better analgesics rely on an assortment of preclinical animal models; however, there are major challenges to translating discoveries made in animal models to realized pain therapies in humans. This review discusses common animal models used to recapitulate clinical chronic pain conditions (such as neuropathic, inflammatory, and visceral pain) and the methods for assessing the sensory and affective components of pain in animals. We also discuss the advantages and limitations of modeling chronic pain in animals as well as highlighting strategies for improving the predictive validity of preclinical pain studies. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Nicole E Burma
- Departments of Comparative Biology and Experimental Medicine, and Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Heather Leduc-Pessah
- Departments of Comparative Biology and Experimental Medicine, and Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Churmy Y Fan
- Departments of Comparative Biology and Experimental Medicine, and Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Tuan Trang
- Departments of Comparative Biology and Experimental Medicine, and Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
43
|
Characterization and Functions of Protease-Activated Receptor 2 in Obesity, Diabetes, and Metabolic Syndrome: A Systematic Review. BIOMED RESEARCH INTERNATIONAL 2016; 2016:3130496. [PMID: 27006943 PMCID: PMC4781943 DOI: 10.1155/2016/3130496] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 01/26/2016] [Indexed: 12/16/2022]
Abstract
Proteinase-activated receptor 2 (PAR2) is a cell surface receptor activated by serine proteinases or specific synthetic compounds. Interest in PAR2 as a pharmaceutical target for various diseases is increasing. Here we asked two questions relevant to endothelial dysfunction and diabetes: How is PAR2 function affected in blood vessels? What role does PAR2 have in promoting obesity, diabetes, and/or metabolic syndrome, specifically via the endothelium and adipose tissues? We conducted a systematic review of the published literature in PubMed and Scopus (July 2015; search terms: par2, par-2, f2lr1, adipose, obesity, diabetes, and metabolic syndrome). Seven studies focused on PAR2 and vascular function. The obesity, diabetes, or metabolic syndrome animal models differed amongst studies, but each reported that PAR2-mediated vasodilator actions were preserved in the face of endothelial dysfunction. The remaining studies focused on nonvascular functions and provided evidence supporting the concept that PAR2 activation promoted obesity. Key studies showed that PAR2 activation regulated cellular metabolism, and PAR2 antagonists inhibited adipose gain and metabolic dysfunction in rats. We conclude that PAR2 antagonists for treatment of obesity indeed show early promise as a therapeutic strategy; however, endothelial-specific PAR2 functions, which may offset mechanisms that produce vascular dysfunction in diabetes, warrant additional study.
Collapse
|
44
|
Role of PAR2 in the Development of Lower Urinary Tract Dysfunction. J Urol 2016; 196:588-98. [PMID: 26860791 DOI: 10.1016/j.juro.2016.01.106] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2016] [Indexed: 02/06/2023]
Abstract
PURPOSE Lower urinary tract symptoms are a common finding in patients with chronic prostatitis/chronic pelvic pain syndrome. We previously reported that the mast cell-tryptase-PAR2 (protease activated receptor 2) axis has a critical role in the development of chronic pain in experimental autoimmune prostatitis, a mouse model of chronic prostatitis/chronic pelvic pain syndrome. Therefore, we examined whether PAR2 activation mediates lower urinary tract dysfunction. MATERIALS AND METHODS Functional cystometry was done in male B6 mice along with immunoblotting and immunohistochemistry for the expression of COL1A1 (collagen type I α I) and α-SMA (α-smooth muscle actin). Flow cytometry analysis was performed on single cell suspensions of the prostate, bladder, lymph nodes and spleen. RESULTS Experimental autoimmune prostatitis resulted in increased urinary voiding frequency and decreased bladder capacity 30 days after initiation. Concurrently, there was increased expression of COL1A1 and α-SMA in the prostates and bladders. In contrast, induction of experimental autoimmune prostatitis in PAR2 knockout mice did not result in altered urodynamics or increased markers of fibrosis in the prostate or the bladder. Single cell suspensions of the prostate, bladder, lymph nodes and spleen demonstrated that in the absence of PAR2 cellular inflammatory mechanisms were still initiated in experimental autoimmune prostatitis but PAR2 expression may be required to maintain chronic inflammation. Finally, antibody mediated PAR2 neutralization normalized urinary voiding frequency and bladder capacity, and attenuated chronic pelvic pain. CONCLUSIONS PAR2 activation in the prostate may contribute to the development of lower urinary tract dysfunction through proinflammatory as well as profibrotic pathways.
Collapse
|
45
|
Liu Y, Liu J, Li M, Dai S, Liang J, Ji W. The effect of kinin B1 receptor on chronic itching sensitization. Mol Pain 2015; 11:70. [PMID: 26576537 PMCID: PMC4650839 DOI: 10.1186/s12990-015-0070-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 10/13/2015] [Indexed: 01/15/2023] Open
Abstract
Background Altered kallikrein-related peptidase activity and bradykinin are associated with skin disorders in humans and mice under chronic inflammation conditions. The bradykinin B1 receptor (B1R), also known as one of the G-protein-coupled receptor family and usually absent in intact tissues and upregulated during tissue injury, is responsible for vasodilation, capillary permeability, nociceptor sensitization, and pain; it is indispensable for physiopathological progress in chronic inflammation conditions, but its roles and effectors in the itching sensation of the allergic contact dermatitis model are poorly defined. Results We focused on incurable itching in a diphenylcyclopropenone (DCP) chronic inflammation experimental model. Preventive treatment with the B1R antagonist R892 significantly suppressed spontaneous scratching, while the B2R selective antagonist did not. B1R expression in the skin tissues of this model was detected using a quantitative, real-time polymerase chain reaction, Western blotting, and immunohistochemistry; B1R mRNA and protein levels were increased compared with a sham-treated control group. A higher B1R IHC staining signal was observed in the keratinocytes in DCP-treated mice compared with a vehicle-treated group, so we studied the B1R function when superimposed on a protease-activated receptor 2 (PAR2) background, establishing B1R as a pivotal mediator of PAR2 function in HaCaT cell lines. Conclusion Our data provide evidence that B1R facilitates the chronic itching sensation related to keratinocytes in a DCP-treated chronic inflammation experimental model.
Collapse
Affiliation(s)
- Yuying Liu
- Postgraduate Institute, Southern Medical University, Guangzhou, 510015, People's Republic of China. .,Department of Anesthesiology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 96 DongChuan Road, Guangzhou, 510080, People's Republic of China.
| | - Jianhua Liu
- Postgraduate Institute, Southern Medical University, Guangzhou, 510015, People's Republic of China. .,Department of Anesthesiology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 96 DongChuan Road, Guangzhou, 510080, People's Republic of China.
| | - Mengran Li
- Postgraduate Institute, Southern Medical University, Guangzhou, 510015, People's Republic of China. .,Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 96 DongChun Road, Guangzhou, 510080, People's Republic of China.
| | - Sailin Dai
- Postgraduate Institute, Southern Medical University, Guangzhou, 510015, People's Republic of China. .,Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 96 DongChun Road, Guangzhou, 510080, People's Republic of China.
| | - Jiexian Liang
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 96 DongChun Road, Guangzhou, 510080, People's Republic of China.
| | - Wenjin Ji
- Department of Anesthesiology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 96 DongChuan Road, Guangzhou, 510080, People's Republic of China.
| |
Collapse
|
46
|
Boitano S, Hoffman J, Flynn AN, Asiedu MN, Tillu DV, Zhang Z, Sherwood CL, Rivas CM, DeFea KA, Vagner J, Price TJ. The novel PAR2 ligand C391 blocks multiple PAR2 signalling pathways in vitro and in vivo. Br J Pharmacol 2015; 172:4535-4545. [PMID: 26140338 DOI: 10.1111/bph.13238] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 06/18/2015] [Accepted: 06/28/2015] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND AND PURPOSE Proteinase-activated receptor-2 (PAR2) is a GPCR linked to diverse pathologies, including acute and chronic pain. PAR2 is one of the four PARs that are activated by proteolytic cleavage of the extracellular amino terminus, resulting in an exposed, tethered peptide agonist. Several peptide and peptidomimetic agonists, with high potency and efficacy, have been developed to probe the functions of PAR2, in vitro and in vivo. However, few similarly potent and effective antagonists have been described. EXPERIMENTAL APPROACH We modified the peptidomimetic PAR2 agonist, 2-furoyl-LIGRLO-NH2 , to create a novel PAR2 peptidomimetic ligand, C391. C391 was evaluated for PAR2 agonist/antagonist activity to PAR2 across Gq signalling pathways using the naturally expressing PAR2 cell line 16HBE14o-. For antagonist studies, a highly potent and specific peptidomimetic agonist (2-aminothiazo-4-yl-LIGRL-NH2 ) and proteinase agonist (trypsin) were used to activate PAR2. C391 was also evaluated in vivo for reduction of thermal hyperalgesia, mediated by mast cell degranulation, in mice. KEY RESULTS C391 is a potent and specific peptidomimetic antagonist, blocking multiple signalling pathways (Gq -dependent Ca2+ , MAPK) induced following peptidomimetic or proteinase activation of human PAR2. In a PAR2-dependent behavioural assay in mice, C391 dose-dependently (75 μg maximum effect) blocked the thermal hyperalgesia, mediated by mast cell degranulation. CONCLUSIONS AND IMPLICATIONS C391 is the first low MW antagonist to block both PAR2 Ca2+ and MAPK signalling pathways activated by peptidomimetics and/or proteinase activation. C391 represents a new molecular structure for PAR2 antagonism and can serve as a basis for further development for this important therapeutic target.
Collapse
Affiliation(s)
- Scott Boitano
- Arizona Respiratory Center and Department of Physiology, University of Arizona, Tucson, AZ, USA.,The BIO5 Collaborative Research Institute, University of Arizona, Tucson, AZ, USA
| | - Justin Hoffman
- Arizona Respiratory Center and Department of Physiology, University of Arizona, Tucson, AZ, USA.,The BIO5 Collaborative Research Institute, University of Arizona, Tucson, AZ, USA
| | - Andrea N Flynn
- Arizona Respiratory Center and Department of Physiology, University of Arizona, Tucson, AZ, USA.,The BIO5 Collaborative Research Institute, University of Arizona, Tucson, AZ, USA
| | - Marina N Asiedu
- Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - Dipti V Tillu
- Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - Zhenyu Zhang
- The BIO5 Collaborative Research Institute, University of Arizona, Tucson, AZ, USA
| | - Cara L Sherwood
- Arizona Respiratory Center and Department of Physiology, University of Arizona, Tucson, AZ, USA.,The BIO5 Collaborative Research Institute, University of Arizona, Tucson, AZ, USA
| | - Candy M Rivas
- Arizona Respiratory Center and Department of Physiology, University of Arizona, Tucson, AZ, USA.,The BIO5 Collaborative Research Institute, University of Arizona, Tucson, AZ, USA.,Graduate Interdisciplinary Program in Physiological Sciences, University of Arizona, Tucson, AZ, USA
| | - Kathryn A DeFea
- Biomedical Sciences Division, University of California, Riverside, CA, USA
| | - Josef Vagner
- The BIO5 Collaborative Research Institute, University of Arizona, Tucson, AZ, USA
| | - Theodore J Price
- The BIO5 Collaborative Research Institute, University of Arizona, Tucson, AZ, USA.,Department of Pharmacology, University of Arizona, Tucson, AZ, USA.,School of Behavioral and Brain Sciences, University of Texas at Dallas, Dallas, TX, USA
| |
Collapse
|