1
|
Song Y, Xu Z, Zhang L, Gao L. Sigma-1 Receptor Modulates CFA-Induced Inflammatory Pain via Sodium Channels in Small DRG Neurons. Biomolecules 2025; 15:73. [PMID: 39858467 PMCID: PMC11764217 DOI: 10.3390/biom15010073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/03/2025] [Accepted: 01/05/2025] [Indexed: 01/30/2025] Open
Abstract
The sigma-1 receptor (Sig-1R) has emerged as a significant target in the realm of pain management and has been the subject of extensive research. Nonetheless, its specific function in inflammatory pain within dorsal root ganglion (DRG) neurons remains inadequately elucidated. This study utilized whole-cell patch clamp techniques, single-cell real-time PCR, and immunohistochemistry to examine the influence of Sig-1R on inflammatory pain induced by complete Freund's adjuvant (CFA) in a rat model. Our results revealed several key findings: (1) The expression of Sig-1R was found to be upregulated during the progression of inflammatory pain, with a notable translocation from the cytoplasm to the membrane; (2) Inhibition of peripheral Sig-1R using S1RA resulted in a reduction of CFA-induced allodynia; (3) Activation of Sig-1R through PRE-084 led to a decrease in the fast sodium current in isolated DRG neurons from CFA-treated rats, which was associated with a diminished action potential (AP) peak and maximum depolarizing rate (MDR), as well as an increased rheobase; (4) Furthermore, PRE-084 was observed to enhance the slow component of the sodium current, resulting in hyperpolarization of the threshold potential and an increase in AP firing frequency, alongside an elevation in the mRNA expression of the slow sodium channel Nav1.9 in CFA-treated rats. In conclusion, our findings suggest that the modulation of sodium channels by Sig-1R in DRG neurons plays a significant role in the mechanisms underlying inflammatory pain.
Collapse
Affiliation(s)
- Yuanlong Song
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Rd., Wuhan 430030, China; (Y.S.); (Z.X.); (L.Z.)
- Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, 13 Hangkong Rd., Wuhan 430030, China
| | - Zifen Xu
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Rd., Wuhan 430030, China; (Y.S.); (Z.X.); (L.Z.)
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Liangpin Zhang
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Rd., Wuhan 430030, China; (Y.S.); (Z.X.); (L.Z.)
- Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, 13 Hangkong Rd., Wuhan 430030, China
| | - Linlin Gao
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Rd., Wuhan 430030, China; (Y.S.); (Z.X.); (L.Z.)
- Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, 13 Hangkong Rd., Wuhan 430030, China
| |
Collapse
|
2
|
Li J, Zhou Z, Wu Y, Zhao J, Duan H, Peng Y, Wang X, Fan Z, Yin L, Li M, Liu F, Yang Y, Du L, Li J, Zhong H, Hou W, Zhang F, Ma H, Zhang X. Heat acclimation defense against exertional heat stroke by improving the function of preoptic TRPV1 neurons. Theranostics 2025; 15:1376-1398. [PMID: 39816678 PMCID: PMC11729562 DOI: 10.7150/thno.101422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 11/19/2024] [Indexed: 01/18/2025] Open
Abstract
Rationale: Record-breaking heatwaves caused by greenhouse effects lead to multiple hyperthermia disorders, the most serious of which is exertional heat stroke (EHS) with the mortality reaching 60 %. Repeat exercise with heat exposure, termed heat acclimation (HA), protects against EHS by fine-tuning feedback control of body temperature (Tb), the mechanism of which is opaque. This study aimed to explore the molecular and neural circuit mechanisms of the HA training against EHS. Methods: Male C57BL/6 mice (6-8 weeks) and male TRPV1-Cre mice (6-8 weeks) were used in our experiments. The EHS model with or without HA training were established for this study. RNA sequencing, qPCR, immunoblot, immunofluorescent assays, calcium imaging, optogenetic/ chemical genetic intervention, virus tracing, patch clamp, and other methods were employed to investigate the molecular mechanism and neural circuit by which HA training improves the function of the medial preoptic area (mPOA) neurons. Furthermore, a novel exosome-based strategy targeting the central nervous system to deliver irisin, a protective peptide generated by HA, was established to protect against EHS. Results: HA-related neurons in the mPOA expressing transient receptor potential vanilloid-1 (TRPV1) were identified as a population whose activation reduces Tb; inversely, dysfunction of these neurons contributes to hyperthermia and EHS. mPOATRPV1 neurons facilitate vasodilation and reduce adipose tissue thermogenesis, which is associated with their inhibitory projection to the raphe pallidus nucleus (RPa) and dorsal medial hypothalamus (DMH) neurons, respectively. Furthermore, HA improves the function of preoptic heat-sensitive neurons by enhancing TRPV1 expression, and Trpv1 ablation reverses the HA-induced heat tolerance. A central nervous system-targeted exosome strategy to deliver irisin, a protective peptide generated by HA, can promote preoptic TRPV1 expression and exert similar protective effects against EHS. Conclusions: Preoptic TRPV1 neurons could be enhanced by HA, actively contributing to heat defense through the mPOA"DMH/RPa circuit during EHS, which results in the suppression of adipose tissue thermogenesis and facilitation of vasodilatation. A delivery strategy of exosomes engineered with RVG-Lamp2b-Irisin significantly improves the function of mPOATRPV1 neurons, providing a promising preventive strategy for EHS in the future.
Collapse
Affiliation(s)
- Jing Li
- Department of Critical Care Medicine and Department of Anaesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China, 710032
| | - Ziqing Zhou
- Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, China, 100071
| | - You Wu
- Department of Critical Care Medicine and Department of Anaesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China, 710032
| | - Jianshuai Zhao
- Department of Critical Care Medicine and Department of Anaesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China, 710032
| | - Haokai Duan
- Department of Microbiology, School of Basic Medical Sciences, Fourth Military Medical University, Xi'an, Shaanxi, China, 710032
| | - Yuliang Peng
- Department of Critical Care Medicine and Department of Anaesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China, 710032
| | - Xiaoke Wang
- Department of Microbiology, School of Basic Medical Sciences, Fourth Military Medical University, Xi'an, Shaanxi, China, 710032
| | - Zhongmin Fan
- Department of Critical Care Medicine and Department of Anaesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China, 710032
| | - Lu Yin
- Department of Critical Care Medicine and Department of Anaesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China, 710032
| | - Mengyun Li
- Department of Microbiology, School of Basic Medical Sciences, Fourth Military Medical University, Xi'an, Shaanxi, China, 710032
| | - Fuhong Liu
- Department of Critical Care Medicine and Department of Anaesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China, 710032
| | - Yongheng Yang
- Department of Critical Care Medicine and Department of Anaesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China, 710032
| | - Lixia Du
- Department of Critical Care Medicine and Department of Anaesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China, 710032
| | - Jin Li
- Department of Critical Care Medicine and Department of Anaesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China, 710032
| | - Haixing Zhong
- Department of Critical Care Medicine and Department of Anaesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China, 710032
| | - Wugang Hou
- Department of Critical Care Medicine and Department of Anaesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China, 710032
| | - Fanglin Zhang
- Department of Microbiology, School of Basic Medical Sciences, Fourth Military Medical University, Xi'an, Shaanxi, China, 710032
| | - Hongwei Ma
- Department of Critical Care Medicine and Department of Anaesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China, 710032
- Department of Microbiology, School of Basic Medical Sciences, Fourth Military Medical University, Xi'an, Shaanxi, China, 710032
| | - Xijing Zhang
- Department of Critical Care Medicine and Department of Anaesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China, 710032
| |
Collapse
|
3
|
Forrester EA, Benítez-Angeles M, Redford KE, Rosenbaum T, Abbott GW, Barrese V, Dora K, Albert AP, Dannesboe J, Salles-Crawley I, Jepps TA, Greenwood IA. Crucial role for sensory nerves and Na/H exchanger inhibition in dapagliflozin- and empagliflozin-induced arterial relaxation. Cardiovasc Res 2024; 120:1811-1824. [PMID: 39056245 PMCID: PMC11587556 DOI: 10.1093/cvr/cvae156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 06/06/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
AIMS Sodium/glucose transporter 2 (SGLT2 or SLC5A2) inhibitors lower blood glucose and are also approved treatments for heart failure independent of raised glucose. Various studies have showed that SGLT2 inhibitors relax arteries, but the underlying mechanisms are poorly understood and responses variable across arterial beds. We speculated that SGLT2 inhibitor-mediated arterial relaxation is dependent upon calcitonin gene-related peptide (CGRP) released from sensory nerves independent of glucose transport. METHODS AND RESULTS The functional effects of SGLT1 and 2 inhibitors (mizagliflozin, dapagliflozin, and empagliflozin) and the sodium/hydrogen exchanger 1 (NHE1) blocker cariporide were determined on pre-contracted resistance arteries (mesenteric and cardiac septal arteries) as well as main renal conduit arteries from male Wistar rats using wire myography. SGLT2, CGRP, TRPV1, and NHE1 expression was determined by western blot and immunohistochemistry. Kv7.4/5/KCNE4 and TRPV1 currents were measured in the presence and absence of dapagliflozin and empagliflozin. All SGLT inhibitors (1-100 µM) and cariporide (30 µM) relaxed mesenteric arteries but had negligible effect on renal or septal arteries. Immunohistochemistry with TRPV1 and CGRP antibodies revealed a dense innervation of sensory nerves in mesenteric arteries that were absent in renal and septal arteries. Consistent with a greater sensory nerve component, the TRPV1 agonist capsaicin relaxed mesenteric arteries more effectively than renal or septal arteries. In mesenteric arteries, relaxations to dapagliflozin, empagliflozin, and cariporide were attenuated by the CGRP receptor antagonist BIBN-4096, depletion of sensory nerves with capsaicin, and blockade of TRPV1 or Kv7 channels. Neither dapagliflozin nor empagliflozin activated heterologously expressed TRPV1 channels or Kv7 channels directly. Sensory nerves also expressed NHE1 but not SGLT2 and cariporide pre-application as well as knockdown of NHE1 by translation stop morpholinos prevented the relaxant response to SGLT2 inhibitors. CONCLUSION SGLT2 inhibitors relax mesenteric arteries by promoting the release of CGRP from sensory nerves in a NHE1-dependent manner.
Collapse
Affiliation(s)
- Elizabeth A Forrester
- Vascular Biology Section, Molecular & Clinical Sciences Research Institute, St George’s University, Cranmer Terrace, London SW17 ORE, UK
| | - Miguel Benítez-Angeles
- I Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Kaitlyn E Redford
- Bioelectricity Lab, Department of Physiology & Biophysics, School of Medicine, University of California, Irvine, USA
| | - Tamara Rosenbaum
- I Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Geoffrey W Abbott
- Bioelectricity Lab, Department of Physiology & Biophysics, School of Medicine, University of California, Irvine, USA
| | - Vincenzo Barrese
- Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples Federico II, Naples, Italy
| | - Kim Dora
- Department of Pharmacology, Oxford University, Oxford, UK
| | - Anthony P Albert
- Vascular Biology Section, Molecular & Clinical Sciences Research Institute, St George’s University, Cranmer Terrace, London SW17 ORE, UK
| | - Johs Dannesboe
- Biomedical Sciences, Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Isabelle Salles-Crawley
- Vascular Biology Section, Molecular & Clinical Sciences Research Institute, St George’s University, Cranmer Terrace, London SW17 ORE, UK
| | - Thomas A Jepps
- Biomedical Sciences, Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Iain A Greenwood
- Vascular Biology Section, Molecular & Clinical Sciences Research Institute, St George’s University, Cranmer Terrace, London SW17 ORE, UK
| |
Collapse
|
4
|
Liu C, Chen IS, Barri M, Murrell-Lagnado R, Kubo Y. Structural determinants of M2R involved in inhibition by Sigma-1R. J Biol Chem 2024; 300:108006. [PMID: 39551139 DOI: 10.1016/j.jbc.2024.108006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 10/31/2024] [Accepted: 11/07/2024] [Indexed: 11/19/2024] Open
Abstract
Sigma-1 receptor (S1R) is a multimodal chaperone protein that is implicated in various pathophysiological conditions including drug addiction, Alzheimer's disease, and amyotrophic lateral sclerosis (ALS). S1R interacts with various ion channels and receptors on the endoplasmic reticulum or plasma membrane (PM). It has been reported that S1R colocalizes with the M2-muscarinic acetylcholine receptor (M2R) on the soma of motoneurons, although a functional interaction between these two proteins has not been established. Here, we investigated the regulation of M2R signaling by S1R using electrophysiological recordings of GIRK currents in HEK293T cells. We observed that S1R strongly inhibited M2R-mediated activation of GIRK1/2, but the disease mutant linked to ALS, S1R E102Q, did not. The inhibitory effect of S1R was selective for M2R and wasn't seen when S1R was co-expressed with other Gi/o coupled receptors including M4R. Chimeric and mutant receptors of M2R and M4R were generated and analyzed, and this highlighted Ala401 in the transmembrane 6 domain (TM6) of M2R and Glu172 as well as Glu175 in the extracellular loop 2 regions of M2R, as essential for the inhibition by S1R. Co-immunoprecipitation confirmed the physical interaction between M2R and S1R. Immunocytochemical labeling of M2R and S1R expressed in HeLa cells, HEK293T cells, and cultured hippocampal neurons, showed clear PM expression of M2R throughout the cell which was decreased by coexpression with S1R but was still apparent. Taken together, our results show that S1R interacts with M2R to reduce both its PM expression and function, and this involves TM6 and the extracellular loop 2.
Collapse
Affiliation(s)
- Chang Liu
- Division of Biophysics and Neurobiology, Department of Molecular and Cellular Physiology, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan; Program of Physiological Sciences, Field of Life Science, Department of Advanced Studies, SOKENDAI (The Graduate University for Advanced Studies), Hayama, Japan.
| | - I-Shan Chen
- Division of Biophysics and Neurobiology, Department of Molecular and Cellular Physiology, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan; Program of Physiological Sciences, Field of Life Science, Department of Advanced Studies, SOKENDAI (The Graduate University for Advanced Studies), Hayama, Japan; Department of Pharmacology, Faculty of Medicine, Wakayama Medical University, Wakayama, Japan
| | - Muruj Barri
- School of Life Sciences, University of Sussex, Brighton, UK
| | | | - Yoshihiro Kubo
- Division of Biophysics and Neurobiology, Department of Molecular and Cellular Physiology, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan; Program of Physiological Sciences, Field of Life Science, Department of Advanced Studies, SOKENDAI (The Graduate University for Advanced Studies), Hayama, Japan.
| |
Collapse
|
5
|
Barr J, Walz A, Restaino AC, Amit M, Barclay SM, Vichaya EG, Spanos WC, Dantzer R, Talbot S, Vermeer PD. Tumor-infiltrating nerves functionally alter brain circuits and modulate behavior in a mouse model of head-and-neck cancer. eLife 2024; 13:RP97916. [PMID: 39302290 DOI: 10.7554/elife.97916] [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] [Indexed: 09/22/2024] Open
Abstract
Cancer patients often experience changes in mental health, prompting an exploration into whether nerves infiltrating tumors contribute to these alterations by impacting brain functions. Using a mouse model for head and neck cancer and neuronal tracing, we show that tumor-infiltrating nerves connect to distinct brain areas. The activation of this neuronal circuitry altered behaviors (decreased nest-building, increased latency to eat a cookie, and reduced wheel running). Tumor-infiltrating nociceptor neurons exhibited heightened calcium activity and brain regions receiving these neural projections showed elevated Fos as well as increased calcium responses compared to non-tumor-bearing counterparts. The genetic elimination of nociceptor neurons decreased brain Fos expression and mitigated the behavioral alterations induced by the presence of the tumor. While analgesic treatment restored nesting and cookie test behaviors, it did not fully restore voluntary wheel running indicating that pain is not the exclusive driver of such behavioral shifts. Unraveling the interaction between the tumor, infiltrating nerves, and the brain is pivotal to developing targeted interventions to alleviate the mental health burdens associated with cancer.
Collapse
Affiliation(s)
- Jeffrey Barr
- Sanford Research, Cancer Biology and Immunotherapies Group, Sioux Falls, Sioux Falls, United States
| | - Austin Walz
- Sanford Research, Cancer Biology and Immunotherapies Group, Sioux Falls, Sioux Falls, United States
| | - Anthony C Restaino
- Sanford Research, Cancer Biology and Immunotherapies Group, Sioux Falls, Sioux Falls, United States
- University of South Dakota, Sanford School of Medicine, Vermillion, United States
| | - Moran Amit
- University of Texas, MD Anderson Cancer Center, Houston, United States
| | - Sarah M Barclay
- Sanford Research, Cancer Biology and Immunotherapies Group, Sioux Falls, Sioux Falls, United States
| | - Elisabeth G Vichaya
- Baylor University, Department of Psychology and Neuroscience, Waco, United States
| | - William C Spanos
- Sanford Research, Cancer Biology and Immunotherapies Group, Sioux Falls, Sioux Falls, United States
- University of South Dakota, Sanford School of Medicine, Vermillion, United States
| | - Robert Dantzer
- University of Texas, MD Anderson Cancer Center, Houston, United States
| | - Sebastien Talbot
- Queen's University, Department of Biomedical and Molecular Sciences, Kingston, Canada
| | - Paola D Vermeer
- Sanford Research, Cancer Biology and Immunotherapies Group, Sioux Falls, Sioux Falls, United States
- University of South Dakota, Sanford School of Medicine, Vermillion, United States
| |
Collapse
|
6
|
Gao N, Li M, Wang W, Liu Z, Guo Y. The dual role of TRPV1 in peripheral neuropathic pain: pain switches caused by its sensitization or desensitization. Front Mol Neurosci 2024; 17:1400118. [PMID: 39315294 PMCID: PMC11417043 DOI: 10.3389/fnmol.2024.1400118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 08/12/2024] [Indexed: 09/25/2024] Open
Abstract
The transient receptor potential vanilloid 1 (TRPV1) channel plays a dual role in peripheral neuropathic pain (NeuP) by acting as a "pain switch" through its sensitization and desensitization. Hyperalgesia, commonly resulting from tissue injury or inflammation, involves the sensitization of TRPV1 channels, which modulates sensory transmission from primary afferent nociceptors to spinal dorsal horn neurons. In chemotherapy-induced peripheral neuropathy (CIPN), TRPV1 is implicated in neuropathic pain mechanisms due to its interaction with ion channels, neurotransmitter signaling, and oxidative stress. Sensitization of TRPV1 in dorsal root ganglion neurons contributes to CIPN development, and inhibition of TRPV1 channels can reduce chemotherapy-induced mechanical hypersensitivity. In diabetic peripheral neuropathy (DPN), TRPV1 is involved in pain modulation through pathways including reactive oxygen species and cytokine production. TRPV1's interaction with TRPA1 channels further influences chronic pain onset and progression. Therapeutically, capsaicin, a TRPV1 agonist, can induce analgesia through receptor desensitization, while TRPV1 antagonists and siRNA targeting TRPV1 show promise in preclinical studies. Cannabinoid modulation of TRPV1 provides another potential pathway for alleviating neuropathic pain. This review summarizes recent preclinical research on TRPV1 in association with peripheral NeuP.
Collapse
Affiliation(s)
- Ning Gao
- Department of Acupuncture and Moxibustion, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Meng Li
- Department of Gastroenterology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Weiming Wang
- Department of Acupuncture and Moxibustion, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhen Liu
- Department of Gastroenterology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yufeng Guo
- Department of Acupuncture and Moxibustion, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
7
|
Davis MP. Novel drug treatments for pain in advanced cancer and serious illness: a focus on neuropathic pain and chemotherapy-induced peripheral neuropathy. Palliat Care Soc Pract 2024; 18:26323524241266603. [PMID: 39086469 PMCID: PMC11289827 DOI: 10.1177/26323524241266603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 06/19/2024] [Indexed: 08/02/2024] Open
Abstract
Drugs that are commercially available but have novel mechanisms of action should be explored as analgesics. This review will discuss haloperidol, miragabalin, palmitoylethanolamide (PEA), and clonidine as adjuvant analgesics or analgesics. Haloperidol is a sigma-1 receptor antagonist. Under stress and neuropathic injury, sigma-1 receptors act as a chaperone protein, which downmodulates opioid receptor activities and opens several ion channels. Clinically, there is only low-grade evidence that haloperidol improves pain when combined with morphine, methadone, or tramadol in patients who have cancer, pain from fibrosis, radiation necrosis, or neuropathic pain. Miragabalin is a gabapentinoid approved for the treatment of neuropathic pain in Japan since 2019. In randomized trials, patients with diabetic neuropathy have responded to miragabalin. Its long binding half-life on the calcium channel subunit may provide an advantage over other gabapentinoids. PEA belongs to a group of endogenous bioactive lipids called ALIAmides (autocoid local injury antagonist amides), which have a sense role in modulating numerous biological processes in particular non-neuronal neuroinflammatory responses to neuropathic injury and systemic inflammation. Multiple randomized trials and meta-analyses have demonstrated PEA's effectiveness in reducing pain severity arising from diverse pain phenotypes. Clonidine is an alpha2 adrenoceptor agonist and an imidazoline2 receptor agonist, which is U.S. Federal Drug Administration approved for attention deficit hyperactivity disorder in children, Tourette's syndrome, adjunctive therapy for cancer-related pain, and hypertension. Clonidine activation at alpha2 adrenoceptors causes downstream activation of inhibitory G-proteins (Gi/Go), which inhibits cyclic Adenosine monophosphate (AMP) production and hyperpolarizes neuron membranes, thus reducing allodynia. Intravenous clonidine has been used in terminally ill patients with poorly controlled symptoms, in particular pain and agitation.
Collapse
Affiliation(s)
- Mellar P. Davis
- Geisinger Commonwealth School of Medicine, 100 North Academy Avenue, Danville, PA 17822, USA
| |
Collapse
|
8
|
Zhu A, Song S, Pei L, Huang Y. Supportive care of female hormones in brain health: what and how? Front Pharmacol 2024; 15:1403969. [PMID: 39114348 PMCID: PMC11303335 DOI: 10.3389/fphar.2024.1403969] [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: 03/20/2024] [Accepted: 07/03/2024] [Indexed: 08/10/2024] Open
Abstract
Female hormones, functioning as neuroactive steroids, are utilized beyond menopausal hormone therapy. The rapid onset of allopregnanolone analogs, such as brexanolone and zuranolone, in treating depression, and the effectiveness of megestrol acetate in addressing appetite and weight gain, prompted the Food and Drug Administration to authorize the use of progesterone for treating postpartum depression and cancer-related cachexia. Progesterone has also been found to alleviate neuropathic pain in animal studies. These off-label applications offer a promising option for patients with advanced cancer who often experience various mood disorders such as depression, persistent pain, social isolation, and physical complications like cachexia. These patients have shown low tolerance to opioids and mood-regulating medications. However, the potential risks and uncertainties associated with hormone therapy treatment modalities can be daunting for both patients and medical professionals. This review aims to offer a comprehensive understanding of the non-reproductive functions and mechanisms of female hormones in brain health.
Collapse
Affiliation(s)
| | | | - Lijian Pei
- Department of Anesthesiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Yuguang Huang
- Department of Anesthesiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
9
|
Barr J, Walz A, Restaino AC, Amit M, Barclay SM, Vichaya EG, Spanos WC, Dantzer R, Talbot S, Vermeer PD. Tumor-infiltrating nerves functionally alter brain circuits and modulate behavior in a male mouse model of head-and-neck cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.18.562990. [PMID: 37905135 PMCID: PMC10614955 DOI: 10.1101/2023.10.18.562990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Cancer patients often experience changes in mental health, prompting an exploration into whether nerves infiltrating tumors contribute to these alterations by impacting brain functions. Using a male mouse model for head and neck cancer, we utilized neuronal tracing techniques and show that tumor-infiltrating nerves indeed connect to distinct brain areas via the ipsilateral trigeminal ganglion. The activation of this neuronal circuitry led to behavioral alterations represented by decreased nest-building, increased latency to eat a cookie, and reduced wheel running. Tumor-infiltrating nociceptor neurons exhibited heightened activity, as indicated by increased calcium mobilization. Correspondingly, the specific brain regions receiving these neural projections showed elevated cFos and delta FosB expression in tumor-bearing mice, alongside markedly intensified calcium responses compared to non-tumor-bearing counterparts. The genetic elimination of nociceptor neurons in tumor-bearing mice led to decreased brain Fos expression and mitigated the behavioral alterations induced by the presence of the tumor. While analgesic treatment successfully restored behaviors involving oral movements to normalcy in tumor-bearing mice, it did not have a similar therapeutic effect on voluntary wheel running. This discrepancy points towards an intricate relationship, where pain is not the exclusive driver of such behavioral shifts. Unraveling the interaction between the tumor, infiltrating nerves, and the brain is pivotal to developing targeted interventions to alleviate the mental health burdens associated with cancer.
Collapse
|
10
|
MO X, CHEN Y, YIN Q, CHEN H, BAN Q, LI J, CHEN S, YAO J. Transient receptor potential vanilloid 1 involved in the analgesic effects of total flavonoids extracted from Longxuejie (). J TRADIT CHIN MED 2024; 44:437-447. [PMID: 38767627 PMCID: PMC11077159 DOI: 10.19852/j.cnki.jtcm.20240423.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 07/06/2023] [Indexed: 05/22/2024]
Abstract
OBJECTIVE To evaluate the analgesic effects of total flavonoids of Longxuejie (Resina Dracaenae Cochinchinensis) (TFDB) and explore the possible analgesic mechanism associated with transient receptor potential vanilloid 1 (TRPV1). METHODS Whole-cell patch clamp technique was used to observe the effects of TFDB on capsaicin-induced TRPV1 currents. Rat experiments in vivo were used to observe the analgesic effects of TFDB. Western blot and immunofluorescence experiments were used to test the change of TRPV1 expression in DRG neurons induced by TFDB. RESULTS Results showed that TFDB inhibited capsaicin-induced TRPV1 receptor currents in acutely isolated dorsal root ganglion (DRG) neurons of rats and the half inhibitory concentration was (16.7 ± 1.6) mg/L. TFDB (2-20 mg/kg) showed analgesic activity in the phase Ⅱ of formalin test and (0.02-2 mg per paw) reduced capsaicin-induced licking times of rats. TFDB (20 mg/kg) was fully efficacious on complete Freund's adjuvant (CFA)-induced inflammatory thermal hyperalgesia and capsaicin could weaken the analgesic effects. The level of TRPV1 expressions of DRG neurons was also decreased in TFDB-treated CFA-inflammatory pain rats. CONCLUSION All these results indicated that the analgesic effect of TFDB may contribute to their modulations on both function and expression of TRPV1 channels in DRG neurons.
Collapse
Affiliation(s)
- Xiaoqiang MO
- 1 Department of Stomatology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, China
- 2 College of General Practitioners, Youjiang Medical University for Nationalities, Baise 533000, China
| | - Yating CHEN
- 3 College of Biomedical Engineering, South-Central Minzu University, Key Laboratory of Cognitive Science of State Ethnic Affairs Commission, Hubei Key Laboratory of Medical Information Analysis and Tumor Diagnosis and Treatment, Wuhan 430074, China
| | - Qian YIN
- 3 College of Biomedical Engineering, South-Central Minzu University, Key Laboratory of Cognitive Science of State Ethnic Affairs Commission, Hubei Key Laboratory of Medical Information Analysis and Tumor Diagnosis and Treatment, Wuhan 430074, China
| | - Haibo CHEN
- 1 Department of Stomatology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, China
| | - Qiang BAN
- 1 Department of Stomatology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, China
| | - Jun LI
- 4 College of Pharmacy, South-Central Minzu University, Wuhan 430074, China
| | - Su CHEN
- 3 College of Biomedical Engineering, South-Central Minzu University, Key Laboratory of Cognitive Science of State Ethnic Affairs Commission, Hubei Key Laboratory of Medical Information Analysis and Tumor Diagnosis and Treatment, Wuhan 430074, China
| | - Jinguang YAO
- 1 Department of Stomatology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, China
| |
Collapse
|
11
|
Cheng XL, Ruan YL, Dai JY, Fan HZ, Ling JY, Chen J, Lu WG, Gao XJ, Cao P. 8-shogaol derived from dietary ginger alleviated acute and inflammatory pain by targeting TRPV1. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155500. [PMID: 38484627 DOI: 10.1016/j.phymed.2024.155500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 01/24/2024] [Accepted: 02/26/2024] [Indexed: 05/01/2024]
Abstract
Ginger, a well-known spice plant, has been used widely in medicinal preparations for pain relief. However, little is known about its analgesic components and the underlying mechanism. Here, we ascertained, the efficacy of ginger ingredient 8-Shogaol (8S), on inflammatory pain and tolerance induced by morphine, and probed the role of TRPV1 in its analgesic action using genetic and electrophysiology approaches. Results showed that 8S effectively reduced nociceptive behaviors of mice elicited by chemical stimuli, noxious heat as well as inflammation, and antagonized morphine analgesic tolerance independent on opioid receptor function. Genetic deletion of TRPV1 significantly abolished 8S' analgesia action. Further calcium imaging and patch-clamp recording showed that 8S could specifically activate TRPV1 in TRPV1-expressing HEK293T cells and dorsal root ganglion (DRG) neurons. The increase of [Ca2+]i in DRG was primarily mediated through TRPV1. Mutational and computation studies revealed the key binding sites for the interactions between 8S and TRPV1 included Leu515, Leu670, Ile573, Phe587, Tyr511, and Phe591. Further studies showed that TRPV1 activation evoked by 8S resulted in channel desensitization both in vitro and in vivo, as may be attributed to TRPV1 degradation or TRPV1 withdrawal from the cell surface. Collectively, this work provides the first evidence for the attractive analgesia of 8S in inflammatory pain and morphine analgesic tolerance mediated by targeting pain-sensing TRPV1 channel. 8S from dietary ginger has potential as a candidate drug for the treatment of inflammatory pain.
Collapse
Affiliation(s)
- Xiao-Lan Cheng
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 100 Hongshan Road, Nanjing 210028, China; School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yong-Lan Ruan
- Department of Neurology, Changzhou Hospital Affiliated to Nanjing University of Chinese Medicine, Changzhou, 213003, China
| | - Jing-Ya Dai
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 100 Hongshan Road, Nanjing 210028, China; Wanbei Health Vocational College, Suzhou, Anhui, 234000, China
| | - Hai-Zhen Fan
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 100 Hongshan Road, Nanjing 210028, China
| | - Jin-Ying Ling
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 100 Hongshan Road, Nanjing 210028, China
| | - Jiao Chen
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 100 Hongshan Road, Nanjing 210028, China
| | - Wu-Guang Lu
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 100 Hongshan Road, Nanjing 210028, China
| | - Xue-Jiao Gao
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 100 Hongshan Road, Nanjing 210028, China.
| | - Peng Cao
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 100 Hongshan Road, Nanjing 210028, China; The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, 100 Minjiang Road, Quzhou, Zhejiang 324000, China.
| |
Collapse
|
12
|
Zheng H, Kim M, Kim C, Kim Y, Cho PS, Lim JY, Lee H, Yun HI, Choi J, Hwang SW. GnRH peripherally modulates nociceptor functions, exacerbating mechanical pain. Front Mol Neurosci 2024; 17:1160435. [PMID: 38783903 PMCID: PMC11111891 DOI: 10.3389/fnmol.2024.1160435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 04/22/2024] [Indexed: 05/25/2024] Open
Abstract
The function of peripheral nociceptors, the neurons that relay pain signals to the brain, are frequently tuned by local and systemic modulator substances. In this context, neurohormonal effects are emerging as an important modulatory mechanism, but many aspects remain to be elucidated. Here we report that gonadotropin-releasing hormone (GnRH), a brain-specific neurohormone, can aggravate pain by acting on nociceptors in mice. GnRH and GnRHR, the receptor for GnRH, are expressed in a nociceptor subpopulation. Administration of GnRH and its analogue, localized for selectively affecting the peripheral neurons, deteriorated mechanical pain, which was reproducible in neuropathic conditions. Nociceptor function was promoted by GnRH treatment in vitro, which appears to involve specific sensory transient receptor potential ion channels. These data suggest that peripheral GnRH can positively modulate nociceptor activities in its receptor-specific manner, contributing to pain exacerbation. Our study indicates that GnRH plays an important role in neurohormonal pain modulation via a peripheral mechanism.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Sun Wook Hwang
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
13
|
Shen B, Yoon D, Castillo J, Biswal S. A Practical Guide to Sigma-1 Receptor Positron Emission Tomography/Magnetic Resonance Imaging: A New Clinical Molecular Imaging Method to Identify Peripheral Pain Generators in Patients with Chronic Pain. Semin Musculoskelet Radiol 2023; 27:601-617. [PMID: 37935207 PMCID: PMC10629991 DOI: 10.1055/s-0043-1775744] [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] [Indexed: 11/09/2023]
Abstract
Accurately identifying the peripheral pain generator in patients with chronic pain remains a major challenge for modern medicine. Millions of patients around the world suffer endlessly from difficult-to-manage debilitating pain because of very limited diagnostic tests and a paucity of pain therapies. To help these patients, we have developed a novel clinical molecular imaging approach, and, in its early stages, it has been shown to accurately identify the exact site of pain generation using an imaging biomarker for the sigma-1 receptor and positron emission tomography/magnetic resonance imaging. We hope the description of the work in this article can help others begin their own pain imaging programs at their respective institutions.
Collapse
Affiliation(s)
- Bin Shen
- Cyclotron Radiochemistry Facility, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California
| | - Daehyun Yoon
- Department of Radiology, University of California San Francisco School of Medicine, San Francisco, California
| | - Jessa Castillo
- Radiochemistry Facility, University of California San Francisco School of Medicine, San Francisco, California
| | - Sandip Biswal
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| |
Collapse
|
14
|
Ruiz-Cantero MC, Cortés-Montero E, Jain A, Montilla-García Á, Bravo-Caparrós I, Shim J, Sánchez-Blázquez P, Woolf CJ, Baeyens JM, Cobos EJ. The sigma-1 receptor curtails endogenous opioid analgesia during sensitization of TRPV1 nociceptors. Br J Pharmacol 2023; 180:1148-1167. [PMID: 36478100 DOI: 10.1111/bph.16003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 08/18/2022] [Accepted: 08/21/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND PURPOSE Peripheral sensitization contributes to pathological pain. While prostaglandin E2 (PGE2) and nerve growth factor (NGF) sensitize peptidergic C-nociceptors (TRPV1+), glial cell line-derived neurotrophic factor (GDNF) sensitizes non-peptidergic C-neurons (IB4+). The sigma-1 receptor (sigma-1R) is a Ca2+ -sensing chaperone known to modulate opoid analgesia. This receptor binds both to TRPV1 and the μ opioid receptor, although the functional repercussions of these physical interactions in peripheral sensitization are unknown. EXPERIMENTAL APPROACH We tested the effects of sigma-1 antagonism on PGE2-, NGF-, and GDNF-induced mechanical and heat hyperalgesia in mice. We used immunohistochemistry to determine the presence of endomorphin-2, an endogenous μ receptor agonist, on dorsal root ganglion (DRG) neurons. Recombinant proteins were used to study the interactions between sigma-1R, μ- receptor, and TRPV1. We used calcium imaging to study the effects of sigma-1 antagonism on PGE2-induced sensitization of TRPV1+ nociceptors. KEY RESULTS Sigma1 antagonists reversed PGE2- and NGF-induced hyperalgesia but not GDNF-induced hyperalgesia. Endomorphin-2 was detected on TRPV1+ but not on IB4+ neurons. Peripheral opioid receptor antagonism by naloxone methiodide or administration of an anti-endomorphin-2 antibody to a sensitized paw reversed the antihyperalgesia induced by sigma-1 antagonists. Sigma-1 antagonism transfers sigma-1R from TRPV1 to μ receptors, suggesting that sigma-1R participate in TRPV1-μ receptor crosstalk. Moreover, sigma-1 antagonism reversed, in a naloxone-sensitive manner, PGE2-induced sensitization of DRG neurons to the calcium flux elicited by capsaicin, the prototypic TRPV1 agonist. CONCLUSION AND IMPLICATIONS Sigma-1 antagonism harnesses endogenous opioids produced by TRPV1+ neurons to reduce hyperalgesia by increasing μ receptor activity.
Collapse
Affiliation(s)
- M Carmen Ruiz-Cantero
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, Spain.,Institute of Neuroscience, Biomedical Research Center, University of Granada, Granada, Spain.,Biosanitary Research Institute ibs. GRANADA, Granada, Spain
| | - Elsa Cortés-Montero
- Department of Translational Neurosciences, Neuropharmacology, Cajal Institute, CSIC, Madrid, Spain
| | - Aakanksha Jain
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA.,F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
| | - Ángeles Montilla-García
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, Spain.,Institute of Neuroscience, Biomedical Research Center, University of Granada, Granada, Spain
| | - Inmaculada Bravo-Caparrós
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, Spain.,Institute of Neuroscience, Biomedical Research Center, University of Granada, Granada, Spain
| | - Jaehoon Shim
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA.,F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
| | - Pilar Sánchez-Blázquez
- Department of Translational Neurosciences, Neuropharmacology, Cajal Institute, CSIC, Madrid, Spain
| | - Clifford J Woolf
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA.,F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
| | - José M Baeyens
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, Spain.,Institute of Neuroscience, Biomedical Research Center, University of Granada, Granada, Spain.,Biosanitary Research Institute ibs. GRANADA, Granada, Spain
| | - Enrique J Cobos
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, Spain.,Institute of Neuroscience, Biomedical Research Center, University of Granada, Granada, Spain.,Biosanitary Research Institute ibs. GRANADA, Granada, Spain.,Teófilo Hernando Institute for Drug Discovery, Madrid, Spain
| |
Collapse
|
15
|
Pergolizzi J, Varrassi G, Coleman M, Breve F, Christo DK, Christo PJ, Moussa C. The Sigma Enigma: A Narrative Review of Sigma Receptors. Cureus 2023; 15:e35756. [PMID: 37020478 PMCID: PMC10069457 DOI: 10.7759/cureus.35756] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 03/04/2023] [Indexed: 03/07/2023] Open
Abstract
The sigma-1 and sigma-2 receptors were first discovered in the 1960s and were thought to be a form of opioid receptors initially. Over time, more was gradually learned about these receptors, which are actually protein chaperones, and many of their unique or unusual properties can contribute to a range of important new therapeutic applications. These sigma receptors translocate in the body and regulate calcium homeostasis and mitochondrial bioenergetics and they also have neuroprotective effects. The ligands to which these sigma receptors respond are several and dissimilar, including neurosteroids, neuroleptics, and cocaine. There is controversy as to their endogenous ligands. Sigma receptors are also involved in the complex processes of cholesterol homeostasis and protein folding. While previous work on this topic has been limited, research has been conducted in multiple disease states, such as addiction, aging. Alzheimer's disease, cancer, psychiatric disorders, pain and neuropathic pain, Parkinson's disease, and others. There is currently increasing interest in sigma-1 and sigma-2 receptors as they provide potential therapeutic targets for many disease indications.
Collapse
|
16
|
Ferreyra S, González S. Therapeutic potential of progesterone in spinal cord injury-induced neuropathic pain: At the crossroads between neuroinflammation and N-methyl-D-aspartate receptor. J Neuroendocrinol 2023; 35:e13181. [PMID: 35924434 DOI: 10.1111/jne.13181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/13/2022] [Accepted: 06/19/2022] [Indexed: 10/17/2022]
Abstract
In recent decades, an area of active research has supported the notion that progesterone promotes a wide range of remarkable protective actions in experimental models of nervous system trauma or disease, and has also provided a strong basis for considering this steroid as a promising molecule for modulating the complex maladaptive changes that lead to neuropathic pain, especially after spinal cord injury. In this review, we intend to give the readers a brief appraisal of the main mechanisms underlying the increased excitability of the spinal circuit in the pain pathway after trauma, with particular emphasis on those mediated by the activation of resident glial cells, the subsequent release of proinflammatory cytokines and their impact on N-methyl-D-aspartate receptor function. We then summarize the available preclinical data pointing to progesterone as a valuable repurposing molecule for blocking critical cellular and molecular events that occur in the dorsal horn of the injured spinal cord and are related to the development of chronic pain. Since the treatment and management of neuropathic pain after spinal injury remains challenging, the potential therapeutic value of progesterone opens new traslational perspectives to prevent central pain.
Collapse
Affiliation(s)
- Sol Ferreyra
- Instituto de Biología y Medicina Experimental, Laboratorio de Nocicepción y Dolor Neuropático, CONICET, Buenos Aires, Argentina
| | - Susana González
- Instituto de Biología y Medicina Experimental, Laboratorio de Nocicepción y Dolor Neuropático, CONICET, Buenos Aires, Argentina
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Buenos Aires, Argentina
| |
Collapse
|
17
|
Petroianu GA, Aloum L, Adem A. Neuropathic pain: Mechanisms and therapeutic strategies. Front Cell Dev Biol 2023; 11:1072629. [PMID: 36727110 PMCID: PMC9884983 DOI: 10.3389/fcell.2023.1072629] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 01/06/2023] [Indexed: 01/18/2023] Open
Abstract
The physiopathology and neurotransmission of pain are of an owe inspiring complexity. Our ability to satisfactorily suppress neuropathic or other forms of chronic pain is limited. The number of pharmacodynamically distinct and clinically available medications is low and the successes achieved modest. Pain Medicine practitioners are confronted with the ethical dichotomy imposed by Hippocrates: On one hand the mandate of primum non nocere, on the other hand, the promise of heavenly joys if successful divinum est opus sedare dolorem. We briefly summarize the concepts associated with nociceptive pain from nociceptive input (afferents from periphery), modulatory output [descending noradrenergic (NE) and serotoninergic (5-HT) fibers] to local control. The local control is comprised of the "inflammatory soup" at the site of pain origin and synaptic relay stations, with an ATP-rich environment promoting inflammation and nociception while an adenosine-rich environment having the opposite effect. Subsequently, we address the transition from nociceptor pain to neuropathic pain (independent of nociceptor activation) and the process of sensitization and pain chronification (transient pain progressing into persistent pain). Having sketched a model of pain perception and processing we attempt to identify the sites and modes of action of clinically available drugs used in chronic pain treatment, focusing on adjuvant (co-analgesic) medication.
Collapse
|
18
|
Keylani K, Arbab Mojeni F, Khalaji A, Rasouli A, Aminzade D, Karimi MA, Sanaye PM, Khajevand N, Nemayandeh N, Poudineh M, Azizabadi Farahani M, Esfandiari MA, Haghshoar S, Kheirandish A, Amouei E, Abdi A, Azizinezhad A, Khani A, Deravi N. Endoplasmic reticulum as a target in cardiovascular diseases: Is there a role for flavonoids? Front Pharmacol 2023; 13:1027633. [PMID: 36703744 PMCID: PMC9871646 DOI: 10.3389/fphar.2022.1027633] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 12/19/2022] [Indexed: 01/11/2023] Open
Abstract
Flavonoids are found in natural health products and plant-based foods. The flavonoid molecules contain a 15-carbon skeleton with the particular structural construction of subclasses. The most flavonoid's critical subclasses with improved health properties are the catechins or flavonols (e.g., epigallocatechin 3-gallate from green tea), the flavones (e.g., apigenin from celery), the flavanones (e.g., naringenin from citrus), the flavanols (e.g., quercetin glycosides from berries, onion, and apples), the isoflavones (e.g., genistein from soya beans) and the anthocyanins (e.g., cyanidin-3-O-glucoside from berries). Scientific data conclusively demonstrates that frequent intake of efficient amounts of dietary flavonoids decreases chronic inflammation and the chance of oxidative stress expressing the pathogenesis of human diseases like cardiovascular diseases (CVDs). The endoplasmic reticulum (ER) is a critical organelle that plays a role in protein folding, post-transcriptional conversion, and transportation, which plays a critical part in maintaining cell homeostasis. Various stimuli can lead to the creation of unfolded or misfolded proteins in the endoplasmic reticulum and then arise in endoplasmic reticulum stress. Constant endoplasmic reticulum stress triggers unfolded protein response (UPR), which ultimately causes apoptosis. Research has shown that endoplasmic reticulum stress plays a critical part in the pathogenesis of several cardiovascular diseases, including diabetic cardiomyopathy, ischemic heart disease, heart failure, aortic aneurysm, and hypertension. Endoplasmic reticulum stress could be one of the crucial points in treating multiple cardiovascular diseases. In this review, we summarized findings on flavonoids' effects on the endoplasmic reticulum and their role in the prevention and treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Kimia Keylani
- School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fariba Arbab Mojeni
- Student Research Committee, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | | | - Asma Rasouli
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Dlnya Aminzade
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Amin Karimi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Nazanin Khajevand
- Student Research Committee, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Nasrin Nemayandeh
- Drug and Food Control Department, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | - Mohammad Ali Esfandiari
- Student Research Committee, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Sepehr Haghshoar
- Faculty of Pharmacy, Cyprus International University, Nicosia, Cyprus
| | - Ali Kheirandish
- Student Research Committee, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Erfan Amouei
- Research Center for Prevention of Cardiovascular Disease, Institute of Endocrinology and Metabolism, Iran University of Medical Science, Tehran, Iran
| | - Amir Abdi
- Student Research Committee, School of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Arash Azizinezhad
- Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Afshin Khani
- Department of Cardiovascular Disease, Cardiovascular Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Niloofar Deravi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
19
|
Acharya TK, Kumar A, Kumar S, Goswami C. TRPV4 interacts with MFN2 and facilitates endoplasmic reticulum-mitochondrial contact points for Ca2+-buffering. Life Sci 2022; 310:121112. [DOI: 10.1016/j.lfs.2022.121112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 10/10/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022]
|
20
|
Luan Y, Jin Y, Zhang P, Li H, Yang Y. Mitochondria-associated endoplasmic reticulum membranes and cardiac hypertrophy: Molecular mechanisms and therapeutic targets. Front Cardiovasc Med 2022; 9:1015722. [PMID: 36337896 PMCID: PMC9630933 DOI: 10.3389/fcvm.2022.1015722] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 09/29/2022] [Indexed: 09/13/2023] Open
Abstract
Cardiac hypertrophy has been shown to compensate for cardiac performance and improve ventricular wall tension as well as oxygen consumption. This compensatory response results in several heart diseases, which include ischemia disease, hypertension, heart failure, and valvular disease. Although the pathogenesis of cardiac hypertrophy remains complicated, previous data show that dysfunction of the mitochondria and endoplasmic reticulum (ER) mediates the progression of cardiac hypertrophy. The interaction between the mitochondria and ER is mediated by mitochondria-associated ER membranes (MAMs), which play an important role in the pathology of cardiac hypertrophy. The function of MAMs has mainly been associated with calcium transfer, lipid synthesis, autophagy, and reactive oxygen species (ROS). In this review, we discuss key MAMs-associated proteins and their functions in cardiovascular system and define their roles in the progression of cardiac hypertrophy. In addition, we demonstrate that MAMs is a potential therapeutic target in the treatment of cardiac hypertrophy.
Collapse
Affiliation(s)
- Yi Luan
- Clinical Systems Biology Research Laboratories, Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yage Jin
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Pengjie Zhang
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongqiang Li
- Department of Thyroid Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yang Yang
- Clinical Systems Biology Research Laboratories, Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
21
|
Sex-related differences in oxaliplatin-induced changes in the expression of transient receptor potential channels and their contribution to cold hypersensitivity. Neurosci Lett 2022; 788:136863. [PMID: 36067900 DOI: 10.1016/j.neulet.2022.136863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/29/2022] [Accepted: 08/31/2022] [Indexed: 11/23/2022]
Abstract
Transient receptor potential (TRP) channels are involved in the development of oxaliplatin-induced neuropathic pain, a frequent and debilitating side effect of cancer therapy. Here we explored whether oxaliplatin-induced changes in the expression of TRP channels, as well as the development of pain-related behaviours, differed between male and female animals. Adult rats were injected with oxaliplatin or saline and mechanical and cold allodynia were evaluated using Von Frey and Choi Tests. The mRNA levels of TRPV1, TRPM8 and TRPA1 were assessed in lumbar ganglia and spinal cord by using real time RT-PCR. Oxaliplatin administration induced mechanical and cold hypersensitivity and allodynia in both sexes, with more severe responses to cold stimulation detected in females. Oxaliplatin also induced a significant increase in the expression of TRPV1, TRPM8 and TRPA1 in lumbar dorsal root ganglia. Interestingly, while TRPV1 and TRPA1 upregulation showed no sex difference, the increase in TRPM8 mRNA levels was more pronounced in female ganglia, correlating with the increased sensitivity to innocuous cold stimuli observed in females. TRPV1 and TRPM8 were also found to be upregulated in the spinal cord of animals of both sexes. Our results reveal previously undescribed changes in the expression of TRP channels occurring in peripheral ganglia and spinal cord of both male and female oxaliplatin-treated animals, with some of these changes exhibiting sex-related differences that could underlie the development of sex-specific patterns of pain-related behaviours.
Collapse
|
22
|
Unveil the pain of endometriosis: from the perspective of the nervous system. Expert Rev Mol Med 2022; 24:e36. [PMID: 36059111 DOI: 10.1017/erm.2022.26] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Endometriosis is a chronic inflammatory disease with pelvic pain and uncharacteristic accompanying symptoms. Endometriosis-associated pain often persists despite treatment of the disease, thus it brings a deleterious impact on their personal lives as well as imposing a substantial economic burden on them. At present, mechanisms underlie endometriosis-associated pain including inflammatory reaction, injury, aberrant blood vessels and the morphological and functional anomaly of the peripheral and central nervous systems. The nerve endings are influenced by the physical and chemical factors surrounding the lesion, via afferent nerve to the posterior root of the spinal nerve, then to the specific cerebral cortex involved in nociception. However, our understanding of the aetiology and mechanism of this complex pain process caused by endometriosis remains incomplete. Identifying the pathogenesis of endometriosis is crucial to disease management, offering proper treatment, and helping patients to seek novel targets for the maintenance and contributors of chronic pain. The main aim of this review is to focus on every possible mechanism of pain related to endometriosis in both peripheral and central nervous systems, and to present related mechanisms of action from the interaction between peripheral lesions and nerves to the changes in transmission of pain, resulting in hyperalgesia and the corresponding alterations in cerebral cortex and brain metabolism.
Collapse
|
23
|
Turnaturi R, Chiechio S, Pasquinucci L, Spoto S, Costanzo G, Dichiara M, Piana S, Grasso M, Amata E, Marrazzo A, Parenti C. Novel N-normetazocine Derivatives with Opioid Agonist/Sigma-1 Receptor Antagonist Profile as Potential Analgesics in Inflammatory Pain. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27165135. [PMID: 36014375 PMCID: PMC9413390 DOI: 10.3390/molecules27165135] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/04/2022] [Accepted: 08/07/2022] [Indexed: 11/30/2022]
Abstract
Although opioids and nonsteroidal anti-inflammatory drugs (NSAIDs) are the most common drugs used in persistent pain treatment; they have shown many side effects. The development of new analgesics endowed with mu opioid receptor/delta opioid receptor (MOR/DOR) activity represents a promising alternative to MOR-selective compounds. Moreover, new mechanisms, such as sigma-1 receptor (σ1R) antagonism, could be an opioid adjuvant strategy. The in vitro σ1R and σ2R profiles of previous synthesized MOR/DOR agonists (−)-2R/S-LP2 (1), (−)-2R-LP2 (2), and (−)-2S-LP2 (3) were assayed. To investigate the pivotal role of N-normetazocine stereochemistry, we also synthesized the (+)-2R/S-LP2 (7), (+)-2R-LP2 (8), and (+)-2S-LP2 (9) compounds. (−)-2R/S-LP2 (1), (−)-2R-LP2 (2), and (−)-2S-LP2 (3) compounds have Ki values for σ1R ranging between 112.72 and 182.81 nM, showing a multitarget opioid/σ1R profile. Instead, (+)-2R/S-LP2 (7), (+)-2R-LP2 (8), and (+)-2S-LP2 (9) isomers displayed a nanomolar affinity for σ1R, with significative selectivity vs. σ2R and opioid receptors. All isomers were evaluated using an in vivo formalin test. (−)-2S-LP2, at 0.7 mg/kg i.p., showed a significative and naloxone-reversed analgesic effect. The σ1R selective compound (+)-2R/S-LP2 (7), at 5.0 mg/kg i.p., decreased the second phase of the formalin test, showing an antagonist σ1R profile. The multitarget or single target profile of assayed N-normetazocine derivatives could represent a promising pharmacological strategy to enhance opioid potency and/or increase the safety margin.
Collapse
Affiliation(s)
- Rita Turnaturi
- Department of Drug and Health Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Santina Chiechio
- Department of Drug and Health Sciences, Section of Pharmacology and Toxicology, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
- Oasi Research Institute—IRCCS, 94018 Troina, Italy
| | - Lorella Pasquinucci
- Department of Drug and Health Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
- Correspondence: ; Tel.: +39-095-738-4273
| | - Salvatore Spoto
- Department of Drug and Health Sciences, Section of Pharmacology and Toxicology, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Giuliana Costanzo
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via Santa Sofia, 97, 95123 Catania, Italy
| | - Maria Dichiara
- Department of Drug and Health Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Silvia Piana
- Department of Drug and Health Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | | | - Emanuele Amata
- Department of Drug and Health Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Agostino Marrazzo
- Department of Drug and Health Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Carmela Parenti
- Department of Drug and Health Sciences, Section of Pharmacology and Toxicology, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| |
Collapse
|
24
|
Song X, Luan M, Zhang W, Zhang R, Xue L, Luan Y. Moderate-Intensity Ultrasound-Triggered On-Demand Analgesia Nanoplatforms for Postoperative Pain Management. Int J Nanomedicine 2022; 17:3177-3189. [PMID: 35909815 PMCID: PMC9329681 DOI: 10.2147/ijn.s367190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 07/07/2022] [Indexed: 11/23/2022] Open
Abstract
Introduction The restricted duration is a fundamental drawback of traditional local anesthetics during postoperative pain from a single injection. Therefore, an injectable local anesthetic that produces repeatable on-demand nerve blocks would be ideal. Methods We offer ultrasound-triggered on-demand analgesia consisting of dendritic mesoporous silica nanoparticles (DMSN) carried with ultrasound-sensitive perfluoropentane (PFP) and levobupivacaine (DMSN-bupi-PFP) to achieve repeatable and customizable on-demand local anesthetics. Results The vaporization of liquid PFP was triggered by ultrasound irradiation to produce a gas environment. Subsequently, the enhanced cavitation effect could improve the release of levobupivacaine to achieve pain relief under a moderate-intensity ultrasound irradiation. DMSN-bupi-PFP demonstrated a controlled-release pattern and showed a reinforced ultrasonic sensitivity compared to levobupivacaine loaded DMSN (DMSN-bupi). The sustained release of levobupivacaine produced continuous analgesia of more than 9 hours in a model of incision pain, approximately 3 times longer than a single free levobupivacaine injection (3 hours). The external ultrasound irradiation can trigger the release of levobupivacaine repeatedly, resulting in on-demand analgesia. In addition, DMSN-bupi-PFP nanoplatforms for ultrasound-enabled analgesia showed low neurotoxicity and good biocompatibility in vitro and in vivo. Conclusion This DMSN-bupi-PFP nanoplatform can be used in pain management by providing long-lasting and on-demand pain alleviation with the help of moderate-intensity ultrasound.
Collapse
Affiliation(s)
- Xinye Song
- Department of Anesthesiology, the First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People's Republic of China
| | - Mengxiao Luan
- Department of Biomedical Engineering, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Weiyi Zhang
- Department of Anesthesiology, the First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People's Republic of China
| | - Ruizheng Zhang
- Department of Anesthesiology, the First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People's Republic of China
| | - Li Xue
- Department of Anesthesiology, the First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People's Republic of China
| | - Yong Luan
- Department of Anesthesiology, the First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People's Republic of China
| |
Collapse
|
25
|
Marcotti A, Fernández-Trillo J, González A, Vizcaíno-Escoto M, Ros-Arlanzón P, Romero L, Vela JM, Gomis A, Viana F, de la Peña E. TRPA1 modulation by Sigma-1 receptor prevents oxaliplatin-induced painful peripheral neuropathy. Brain 2022; 146:475-491. [PMID: 35871491 PMCID: PMC9924907 DOI: 10.1093/brain/awac273] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 05/30/2022] [Accepted: 07/03/2022] [Indexed: 11/13/2022] Open
Abstract
Chemotherapy-induced peripheral neuropathy is a frequent, disabling side effect of anticancer drugs. Oxaliplatin, a platinum compound used in the treatment of advanced colorectal cancer, often leads to a form of chemotherapy-induced peripheral neuropathy characterized by mechanical and cold hypersensitivity. Current therapies for chemotherapy-induced peripheral neuropathy are ineffective, often leading to the cessation of treatment. Transient receptor potential ankyrin 1 (TRPA1) is a polymodal, non-selective cation-permeable channel expressed in nociceptors, activated by physical stimuli and cellular stress products. TRPA1 has been linked to the establishment of chemotherapy-induced peripheral neuropathy and other painful neuropathic conditions. Sigma-1 receptor is an endoplasmic reticulum chaperone known to modulate the function of many ion channels and receptors. Sigma-1 receptor antagonist, a highly selective antagonist of Sigma-1 receptor, has shown effectiveness in a phase II clinical trial for oxaliplatin chemotherapy-induced peripheral neuropathy. However, the mechanisms involved in the beneficial effects of Sigma-1 receptor antagonist are little understood. We combined biochemical and biophysical (i.e. intermolecular Förster resonance energy transfer) techniques to demonstrate the interaction between Sigma-1 receptor and human TRPA1. Pharmacological antagonism of Sigma-1R impaired the formation of this molecular complex and the trafficking of functional TRPA1 to the plasma membrane. Using patch-clamp electrophysiological recordings we found that antagonists of Sigma-1 receptor, including Sigma-1 receptor antagonist, exert a marked inhibition on plasma membrane expression and function of human TRPA1 channels. In TRPA1-expressing mouse sensory neurons, Sigma-1 receptor antagonists reduced inward currents and the firing of actions potentials in response to TRPA1 agonists. Finally, in a mouse experimental model of oxaliplatin neuropathy, systemic treatment with a Sigma-1 receptor antagonists prevented the development of painful symptoms by a mechanism involving TRPA1. In summary, the modulation of TRPA1 channels by Sigma-1 receptor antagonists suggests a new strategy for the prevention and treatment of chemotherapy-induced peripheral neuropathy and could inform the development of novel therapeutics for neuropathic pain.
Collapse
Affiliation(s)
- Aida Marcotti
- Present address: Instituto de Farmacología Experimental de Córdoba (IFEC) – CONICET, Haya de la Torre y Medina Allende, Ciudad Universitaria, Córdoba 5000, Argentina
| | | | - Alejandro González
- Present address: Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Marta Vizcaíno-Escoto
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, 03550 San Juan de Alicante, Spain
| | - Pablo Ros-Arlanzón
- Present address: Institute for Health and Biomedical Research (ISABIAL), 03550 San Juan de Alicante, Spain
| | - Luz Romero
- WeLab Barcelona, Parc Científic de Barcelona, 08028 Barcelona, Spain
| | - José Miguel Vela
- WeLab Barcelona, Parc Científic de Barcelona, 08028 Barcelona, Spain
| | - Ana Gomis
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, 03550 San Juan de Alicante, Spain
| | - Félix Viana
- Correspondence may also be addressed to: Felix Viana E-mail:
| | - Elvira de la Peña
- Correspondence to: Elvira de la Peña Instituto de Neurociencias de Alicante Universidad Miguel Hernández-CSIC 03550 San Juan de Alicante, Spain E-mail:
| |
Collapse
|
26
|
Lv Y, Cheng L, Peng F. Compositions and Functions of Mitochondria-Associated Endoplasmic Reticulum Membranes and Their Contribution to Cardioprotection by Exercise Preconditioning. Front Physiol 2022; 13:910452. [PMID: 35733995 PMCID: PMC9207531 DOI: 10.3389/fphys.2022.910452] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 05/11/2022] [Indexed: 11/13/2022] Open
Abstract
Mitochondria-associated endoplasmic reticulum membranes (MAMs) are important components of intracellular signaling and contribute to the regulation of intracellular Ca2+/lipid homeostasis, mitochondrial dynamics, autophagy/mitophagy, apoptosis, and inflammation. Multiple studies have shown that proteins located on MAMs mediate cardioprotection. Exercise preconditioning (EP) has been shown to protect the myocardium from adverse stimuli, but these mechanisms are still being explored. Recently, a growing body of evidence points to MAMs, suggesting that exercise or EP may be involved in cardioprotection by modulating proteins on MAMs and subsequently affecting MAMs. In this review, we summarize the latest findings on MAMs, analyzing the structure and function of MAMs and the role of MAM-related proteins in cardioprotection. We focused on the possible mechanisms by which exercise or EP can modulate the involvement of MAMs in cardioprotection. We found that EP may affect MAMs by regulating changes in MFN2, MFN1, AMPK, FUNDC1, BECN1, VDAC1, GRP75, IP3R, CYPD, GSK3β, AKT, NLRP3, GRP78, and LC3, thus playing a cardioprotective role. We also provided direction for future studies that may be of interest so that more in-depth studies can be conducted to elucidate the relationship between EP and cardioprotection.
Collapse
|
27
|
Li YE, Sowers JR, Hetz C, Ren J. Cell death regulation by MAMs: from molecular mechanisms to therapeutic implications in cardiovascular diseases. Cell Death Dis 2022; 13:504. [PMID: 35624099 PMCID: PMC9142581 DOI: 10.1038/s41419-022-04942-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 05/04/2022] [Accepted: 05/12/2022] [Indexed: 12/13/2022]
Abstract
The endoplasmic reticulum (ER) and mitochondria are interconnected intracellular organelles with vital roles in the regulation of cell signaling and function. While the ER participates in a number of biological processes including lipid biosynthesis, Ca2+ storage and protein folding and processing, mitochondria are highly dynamic organelles governing ATP synthesis, free radical production, innate immunity and apoptosis. Interplay between the ER and mitochondria plays a crucial role in regulating energy metabolism and cell fate control under stress. The mitochondria-associated membranes (MAMs) denote physical contact sites between ER and mitochondria that mediate bidirectional communications between the two organelles. Although Ca2+ transport from ER to mitochondria is vital for mitochondrial homeostasis and energy metabolism, unrestrained Ca2+ transfer may result in mitochondrial Ca2+ overload, mitochondrial damage and cell death. Here we summarize the roles of MAMs in cell physiology and its impact in pathological conditions with a focus on cardiovascular disease. The possibility of manipulating ER-mitochondria contacts as potential therapeutic approaches is also discussed.
Collapse
Affiliation(s)
- Yiran E Li
- Department of Cardiology, Zhongshan Hospital, Fudan University; Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
| | - James R Sowers
- Diabetes and Cardiovascular Center, University of Missouri School of Medicine, Columbia, MO, 65212, USA
| | - Claudio Hetz
- Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
- FONDAP Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
- Buck Institute for Research in Aging, Novato, CA, 94945, USA
| | - Jun Ren
- Department of Cardiology, Zhongshan Hospital, Fudan University; Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China.
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
28
|
Li L, Chen C, Xiang Q, Fan S, Xiao T, Chen Y, Zheng D. Transient Receptor Potential Cation Channel Subfamily V Member 1 Expression Promotes Chemoresistance in Non-Small-Cell Lung Cancer. Front Oncol 2022; 12:773654. [PMID: 35402237 PMCID: PMC8990814 DOI: 10.3389/fonc.2022.773654] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 03/02/2022] [Indexed: 12/23/2022] Open
Abstract
Approximately 85% of lung cancer cases are non-small-cell lung cancer (NSCLC). Chemoresistance is a leading cause of chemotherapy failure in NSCLC treatment. Transient receptor potential cation channel subfamily V, member 1 (TRPV1), a non-selective cation channel, plays multiple roles in tumorigenesis and tumor development, including tumor cell proliferation, death, and metastasis as well as the response to therapy. In this study, we found TRPV1 expression was increased in NSCLC. TRPV1 overexpression induced cisplatin (DDP) and fluorouracil (5-FU) resistance in A549 cells independent of its channel function. TRPV1 expression was upregulated in A549-DDP/5-FU resistant cells, and DDP/5-FU sensitivity was restored by TRPV1 knockdown. TRPV1 overexpression mediated DDP and 5-FU resistance by upregulation of ABCA5 drug transporter gene expression, thereby increasing drug efflux, enhancing homologous recombination (HR) DNA repair pathway to alleviate apoptosis and activating IL-8 signaling to promote cell survival. These findings demonstrate an essential role of TRPV1 in chemoresistance in NSCLC and implicate TRPV1 as a potential chemotherapeutic target.
Collapse
Affiliation(s)
- Li Li
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Shenzhen University International Cancer Center, Department of Cell Biology and Genetics, School of Medicine, Shenzhen University, Shenzhen, China
| | - Cheng Chen
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Shenzhen University International Cancer Center, Department of Cell Biology and Genetics, School of Medicine, Shenzhen University, Shenzhen, China
| | - Qin Xiang
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Shenzhen University International Cancer Center, Department of Cell Biology and Genetics, School of Medicine, Shenzhen University, Shenzhen, China
| | - Songqing Fan
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Tian Xiao
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Shenzhen University International Cancer Center, Department of Cell Biology and Genetics, School of Medicine, Shenzhen University, Shenzhen, China
| | - Yangchao Chen
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Duo Zheng
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Shenzhen University International Cancer Center, Department of Cell Biology and Genetics, School of Medicine, Shenzhen University, Shenzhen, China
- *Correspondence: Duo Zheng,
| |
Collapse
|
29
|
Zieglgänsberger W, Brenneisen R, Berthele A, Wotjak CT, Bandelow B, Tölle TR, Lutz B. Chronic Pain and the Endocannabinoid System: Smart Lipids - A Novel Therapeutic Option? Med Cannabis Cannabinoids 2022; 5:61-75. [PMID: 35702403 PMCID: PMC9149512 DOI: 10.1159/000522432] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 01/21/2022] [Indexed: 08/05/2023] Open
Abstract
The development of a high-end cannabinoid-based therapy is the result of intense translational research, aiming to convert recent discoveries in the laboratory into better treatments for patients. Novel compounds and new regimes for drug treatment are emerging. Given that previously unreported signaling mechanisms for cannabinoids have been uncovered, clinical studies detailing their high therapeutic potential are mandatory. The advent of novel genomic, optogenetic, and viral tracing and imaging techniques will help to further detail therapeutically relevant functional and structural features. An evolutionarily highly conserved group of neuromodulatory lipids, their receptors, and anabolic and catabolic enzymes are involved in a remarkable variety of physiological and pathological processes and has been termed the endocannabinoid system (ECS). A large body of data has emerged in recent years, pointing to a crucial role of this system in the regulation of the behavioral domains of acquired fear, anxiety, and stress-coping. Besides neurons, also glia cells and components of the immune system can differentially fine-tune patterns of neuronal activity. Dysregulation of ECS signaling can lead to a lowering of stress resilience and increased incidence of psychiatric disorders. Chronic pain may be understood as a disease process evoked by fear-conditioned nociceptive input and appears as the dark side of neuronal plasticity. By taking a toll on every part of your life, this abnormal persistent memory of an aversive state can be more damaging than its initial experience. All strategies for the treatment of chronic pain conditions must consider stress-related comorbid conditions since cognitive factors such as beliefs, expectations, and prior experience (memory of pain) are key modulators of the perception of pain. The anxiolytic and anti-stress effects of medical cannabinoids can substantially modulate the efficacy and tolerability of therapeutic interventions and will help to pave the way to a successful multimodal therapy. Why some individuals are more susceptible to the effects of stress remains to be uncovered. The development of personalized prevention or treatment strategies for anxiety and depression related to chronic pain must also consider gender differences. An emotional basis of chronic pain opens a new horizon of opportunities for developing treatment strategies beyond the repeated sole use of acutely acting analgesics. A phase I trial to determine the pharmacokinetics, psychotropic effects, and safety profile of a novel nanoparticle-based cannabinoid spray for oromucosal delivery highlights a remarkable innovation in galenic technology and urges clinical studies further detailing the huge therapeutic potential of medical cannabis (Lorenzl et al.; this issue).
Collapse
Affiliation(s)
| | | | | | | | - Borwin Bandelow
- Department of Psychiatry and Psychotherapy, University Medical Center, Göttingen, Germany
| | | | - Beat Lutz
- Institute of Physiological Chemistry, University Medical Center Mainz, Mainz, Germany
| |
Collapse
|
30
|
Inferiority complex: why do sensory ion channels multimerize? Biochem Soc Trans 2022; 50:213-222. [PMID: 35166323 PMCID: PMC9022975 DOI: 10.1042/bst20211002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 11/17/2022]
Abstract
Peripheral somatosensory nerves are equipped with versatile molecular sensors which respond to acute changes in the physical environment. Most of these sensors are ion channels that, when activated, depolarize the sensory nerve terminal causing it to generate action potentials, which is the first step in generation of most somatic sensations, including pain. The activation and inactivation of sensory ion channels is tightly regulated and modulated by a variety of mechanisms. Amongst such mechanisms is the regulation of sensory ion channel activity via direct molecular interactions with other proteins in multi-protein complexes at the plasma membrane of sensory nerve terminals. In this brief review, we will consider several examples of such complexes formed around a prototypic sensory receptor, transient receptor potential vanilloid type 1 (TRPV1). We will also discuss some inherent conceptual difficulties arising from the multitude of reported complexes.
Collapse
|
31
|
Rabiner EA, Smith K, Bennett C, Rizzo G, Lewis Y, Mundin G, Dooner H, Oksche A. Pharmacokinetics and brain σ1 receptor occupancy of MR309, a selective σ1 receptor antagonist. Br J Clin Pharmacol 2022; 88:1644-1654. [PMID: 34156715 DOI: 10.1111/bcp.14952] [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: 02/16/2021] [Revised: 06/02/2021] [Accepted: 06/09/2021] [Indexed: 11/30/2022] Open
Abstract
AIMS Preclinical studies of MR309, a selective sigma-1 receptor (σ1R) antagonist, support a potential role in treating neuropathic pain. We report 2 studies that provide insight into the pharmacokinetics (PK) and brain σ1R binding of MR309. METHODS Steady-state PK of MR309 (400 mg once daily and 200 mg twice-daily [BID] for 10 days; EudraCT 2015-001818-99 [PK study]) and the relationship between MR309 plasma exposure and brain σ1R occupancy (EudraCT 2017-000670-11 [positron emission tomography study]) were investigated in healthy volunteers. Positron emission tomography using the σ1R ligand [11 C]SA4503 was conducted at baseline, and 2 and 8 hours after a single dose of MR309 (200-800 mg). The relationship between brain σ1R occupancy and MR309 exposure was explored using data-driven model fitting. RESULTS MR309 was well tolerated, brain σ1R occupancy ranged between 30.5 and 74.9% following single-dose MR309 (n = 7). MR309 BID provided a plasma PK profile with less fluctuation than once daily dosing (n = 16). MR309 200 mg BID yielded average steady state plasma concentrations between 2000 and 4000 ng/mL in the PK study, which corresponded to an estimated brain σ1R occupancy of 59-74%. CONCLUSION MR309 200 mg BID dose was below the 75% σ1R occupancy threshold expected to elicit maximal antinociceptive effect as observed in neuropathic pain models. Further investigations of MR309 for neuropathic pain will require higher brain σ1R occupancy, and establish the optimal dose by elucidating the clinical impact of a broad range of brain σ1R occupancy across different neuropathic pain indications.
Collapse
Affiliation(s)
- Eugenii A Rabiner
- Invicro(former Imanova Ltd), A Konica Minolta Company, London, UK
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, UK
| | | | | | - Gaia Rizzo
- Invicro(former Imanova Ltd), A Konica Minolta Company, London, UK
- Division of Brain Sciences, Department of Medicine, Imperial College, London, UK
| | - Yvonne Lewis
- Invicro(former Imanova Ltd), A Konica Minolta Company, London, UK
| | | | | | - Alexander Oksche
- Mundipharma Research Limited, Cambridge, UK
- Institut für medizinische und pharmazeutische Prüfungsfragen, Mainz, Germany
- Rudolf-Buchheim Institute of Pharmacology, Giessen, Germany
| |
Collapse
|
32
|
Hornung RS, Raut NGR, Cantu DJ, Lockhart LM, Averitt DL. Sigma-1 receptors and progesterone metabolizing enzymes in nociceptive sensory neurons of the female rat trigeminal ganglia: A neural substrate for the antinociceptive actions of progesterone. Mol Pain 2022; 18:17448069211069255. [PMID: 35040378 PMCID: PMC8777333 DOI: 10.1177/17448069211069255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Orofacial pain disorders are predominately experienced by women. Progesterone, a major ovarian hormone, is neuroprotective and antinociceptive. We recently reported that progesterone attenuates estrogen-exacerbated orofacial pain behaviors, yet it remains unclear what anatomical substrate underlies progesterone's activity in the trigeminal system. Progesterone has been reported to exert protective effects through actions at intracellular progesterone receptors (iPR), membrane-progesterone receptors (mPR), or sigma 1 receptors (Sig-1R). Of these, the iPR and Sig-1R have been reported to have a role in pain. Progesterone can also have antinociceptive effects through its metabolite, allopregnanolone. Two enzymes, 5α-reductase and 3α-hydroxysteroid dehydrogenase (3α-HSD), are required for the metabolism of progesterone to allopregnanolone. Both progesterone and allopregnanolone rapidly attenuate pain sensitivity, implicating action of either progesterone at Sig-1R and/or conversion to allopregnanolone which targets GABAA receptors. In the present study, we investigated whether Sig-1 Rs are expressed in nociceptors within the trigeminal ganglia of cycling female rats and whether the two enzymes required for progesterone metabolism to allopregnanolone, 5α-reductase and 3α-hydroxysteroid dehydrogenase, are also present. Adult female rats from each stage of the estrous cycle were rapidly decapitated and the trigeminal ganglia collected. Trigeminal ganglia were processed by either fluorescent immunochemistry or western blotting to for visualization and quantification of Sig-1R, 5α-reductase, and 3α-hydroxysteroid dehydrogenase. Here we report that Sig-1Rs and both enzymes involved in progesterone metabolism are highly expressed in a variety of nociceptive sensory neuron populations in the female rat trigeminal ganglia at similar levels across the four stages of the estrous cycle. These data indicate that trigeminal sensory neurons are an anatomical substrate for the reported antinociceptive activity of progesterone via Sig-1R and/or conversion to allopregnanolone.
Collapse
Affiliation(s)
| | | | - Daisy J Cantu
- Division of Biology, School of the Sciences, Texas Woman’s
University, Denton, TX, USA
| | - Lauren M Lockhart
- Division of Biology, School of the Sciences, Texas Woman’s
University, Denton, TX, USA
| | - Dayna L Averitt
- Division of Biology, School of the Sciences, Texas Woman’s
University, Denton, TX, USA
| |
Collapse
|
33
|
López-Estévez S, Gris G, de la Puente B, Carceller A, Martínez V. Intestinal inflammation-associated hypersensitivity is attenuated in a DSS model of colitis in Sigma-1 knockout C57BL/6 mice. Biomed Pharmacother 2021; 143:112126. [PMID: 34474349 DOI: 10.1016/j.biopha.2021.112126] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/20/2021] [Accepted: 08/24/2021] [Indexed: 02/07/2023] Open
Abstract
Sigma-1 receptors (σ1R) have been implicated in several pain pathways. We assessed the implication of σ1Rs in the development of intestinal inflammation and inflammation-associated referred hypersensitivity in a model of colitis in σ1R knockout (KO) mice. Colitis was induced with dextran sulfate sodium (DSS) in wild type (WT) and σ1R KO mice. The development of referred mechanical hypersensitivity (von Frey test) was assessed. Colonic and spinal changes in expression of immune- and sensory-related markers were also investigated (RT-qPCR/Western blot). Absence of σ1Rs had little impact in colitis generation and progression, although during the chronic phase a reduction in edema and a down-regulation of iNOS gene expression was observed. In σ1R KO mice, inflammation-associated hypersensitivity was significantly attenuated (paw) or completely prevented (abdomen). During colitis, in WT mice, changes in the colonic expression of nociceptive markers were observed during the acute and chronic phases of inflammation. Although σ1R KO mice showed similar regulation in the acute phase, an attenuated response was observed during the chronic phase of colitis. These differences were especially relevant for CB2 and TRPV1 receptors, which could play an important role in σ1-mediated regulation of sensitivity. No changes were detected on ERK phosphorylation at the level of the lumbosacral spinal cord. In summary, intestinal inflammation-associated referred hyperalgesia was reduced (paw) or absent (abdomen) in σ1R KO mice, thus confirming an important role for σ1R in the development of colitis-associated hypersensitivity. These results identify σ1Rs as a possible therapeutic target for the treatment of hypersensitivity associated to intestinal inflammation.
Collapse
Affiliation(s)
- Sergio López-Estévez
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain; Neuroscience Institute, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
| | - Georgia Gris
- Department of Pharmacology, Drug Discovery & Preclinical Development, ESTEVE, 08028 Barcelona, Spain
| | - Beatriz de la Puente
- Department of Pharmacology, Drug Discovery & Preclinical Development, ESTEVE, 08028 Barcelona, Spain
| | - Alicia Carceller
- Department of Pharmacology, Drug Discovery & Preclinical Development, ESTEVE, 08028 Barcelona, Spain
| | - Vicente Martínez
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain; Neuroscience Institute, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
34
|
Uchida Y, Izumizaki M. Effect of menstrual cycle and female hormones on TRP and TREK channels in modifying thermosensitivity and physiological functions in women. J Therm Biol 2021; 100:103029. [PMID: 34503776 DOI: 10.1016/j.jtherbio.2021.103029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 05/10/2021] [Accepted: 05/11/2021] [Indexed: 12/15/2022]
Abstract
Thermoregulation is crucial for human survival at various ambient temperatures. Transient receptor potential (TRP) and TWIK-related K+ (TREK) channels expressed in sensory neurons play a role in peripheral thermosensitivity for temperature detection. In addition, these channels have various physiological roles in the skeletal, nervous, immune, vascular, digestive, and urinary systems. In women, the female hormones estradiol (E2) and progesterone (P4), which fluctuate during the menstrual cycle, affect various physiological functions, such as thermoregulation in hot and cold environments. The present review describes the effect of female hormones on TRP and TREK channels and related physiological functions. The P4 decreased thermosensitivity via TRPV1. E2 facilitates temporomandibular joint disease (TRPV1), breast cancer (TRPM8), and calcium absorption in the digestive system (TRPV5 and TRPV6), inhibits the facilitation of vasoconstriction (TRPM3), nerve inflammation (TRPM4), sweetness sensitivity (TRPM5), and menstrual disorders (TRPC1), and prevents insulin resistance (TRPC5) via each channel. P4 inhibits vasoconstriction (TRPM3), sweetness sensitivity (TRPM5), ciliary motility in the lungs (TRPV4), menstrual disorder (TRPC1), and immunity (TRPC3), and facilitates breast cancer (TRPV6) via each channel as indicated. The effects of female hormones on TREK channels and physiological functions are still under investigation. In summary, female hormones influence physiological functions via some TRP channels; however, the literature is not comprehensive and future studies are needed, especially those related to thermoregulation in women.
Collapse
Affiliation(s)
- Yuki Uchida
- Department of Physiology, Showa University School of Medicine, Tokyo, Japan.
| | - Masahiko Izumizaki
- Department of Physiology, Showa University School of Medicine, Tokyo, Japan
| |
Collapse
|
35
|
Luan Y, Luan Y, Yuan RX, Feng Q, Chen X, Yang Y. Structure and Function of Mitochondria-Associated Endoplasmic Reticulum Membranes (MAMs) and Their Role in Cardiovascular Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:4578809. [PMID: 34336092 PMCID: PMC8289621 DOI: 10.1155/2021/4578809] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 06/30/2021] [Indexed: 12/12/2022]
Abstract
Abnormal function of suborganelles such as mitochondria and endoplasmic reticulum often leads to abnormal function of cardiomyocytes or vascular endothelial cells and cardiovascular disease (CVD). Mitochondria-associated membrane (MAM) is involved in several important cellular functions. Increasing evidence shows that MAM is involved in the pathogenesis of CVD. MAM mediates multiple cellular processes, including calcium homeostasis regulation, lipid metabolism, unfolded protein response, ROS, mitochondrial dynamics, autophagy, apoptosis, and inflammation, which are key risk factors for CVD. In this review, we discuss the structure of MAM and MAM-associated proteins, their role in CVD progression, and the potential use of MAM as the therapeutic targets for CVD treatment.
Collapse
Affiliation(s)
- Yi Luan
- Department of Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Ying Luan
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Rui-Xia Yuan
- Department of Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Qi Feng
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou 450052, China
| | - Xing Chen
- Department of Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yang Yang
- Department of Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
36
|
Ramal-Sanchez M, Bernabò N, Valbonetti L, Cimini C, Taraschi A, Capacchietti G, Machado-Simoes J, Barboni B. Role and Modulation of TRPV1 in Mammalian Spermatozoa: An Updated Review. Int J Mol Sci 2021; 22:4306. [PMID: 33919147 PMCID: PMC8122410 DOI: 10.3390/ijms22094306] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/19/2021] [Accepted: 04/19/2021] [Indexed: 12/26/2022] Open
Abstract
Based on the abundance of scientific publications, the polymodal sensor TRPV1 is known as one of the most studied proteins within the TRP channel family. This receptor has been found in numerous cell types from different species as well as in spermatozoa. The present review is focused on analyzing the role played by this important channel in the post-ejaculatory life of spermatozoa, where it has been described to be involved in events such as capacitation, acrosome reaction, calcium trafficking, sperm migration, and fertilization. By performing an exhaustive bibliographic search, this review gathers, for the first time, all the modulators of the TRPV1 function that, to our knowledge, were described to date in different species and cell types. Moreover, all those modulators with a relationship with the reproductive process, either found in the female tract, seminal plasma, or spermatozoa, are presented here. Since the sperm migration through the female reproductive tract is one of the most intriguing and less understood events of the fertilization process, in the present work, chemotaxis, thermotaxis, and rheotaxis guiding mechanisms and their relationship with TRPV1 receptor are deeply analyzed, hypothesizing its (in)direct participation during the sperm migration. Last, TRPV1 is presented as a pharmacological target, with a special focus on humans and some pathologies in mammals strictly related to the male reproductive system.
Collapse
Affiliation(s)
- Marina Ramal-Sanchez
- Faculty of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy; (N.B.); (L.V.); (C.C.); (A.T.); (G.C.); (J.M.-S.); (B.B.)
| | - Nicola Bernabò
- Faculty of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy; (N.B.); (L.V.); (C.C.); (A.T.); (G.C.); (J.M.-S.); (B.B.)
- Institute of Biochemistry and Cell Biology (CNR-IBBC/EMMA/Infrafrontier/IMPC), National Research Council, Monterotondo Scalo, 00015 Rome, Italy
| | - Luca Valbonetti
- Faculty of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy; (N.B.); (L.V.); (C.C.); (A.T.); (G.C.); (J.M.-S.); (B.B.)
- Institute of Biochemistry and Cell Biology (CNR-IBBC/EMMA/Infrafrontier/IMPC), National Research Council, Monterotondo Scalo, 00015 Rome, Italy
| | - Costanza Cimini
- Faculty of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy; (N.B.); (L.V.); (C.C.); (A.T.); (G.C.); (J.M.-S.); (B.B.)
| | - Angela Taraschi
- Faculty of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy; (N.B.); (L.V.); (C.C.); (A.T.); (G.C.); (J.M.-S.); (B.B.)
- Istituto Zooprofilattico Sperimentale dell’Abruzzo e del Molise “G. Caporale”, Via Campo Boario 1, 64100 Teramo, Italy
| | - Giulia Capacchietti
- Faculty of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy; (N.B.); (L.V.); (C.C.); (A.T.); (G.C.); (J.M.-S.); (B.B.)
| | - Juliana Machado-Simoes
- Faculty of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy; (N.B.); (L.V.); (C.C.); (A.T.); (G.C.); (J.M.-S.); (B.B.)
| | - Barbara Barboni
- Faculty of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy; (N.B.); (L.V.); (C.C.); (A.T.); (G.C.); (J.M.-S.); (B.B.)
| |
Collapse
|
37
|
Xu Q, Sun L, Zhang W, Wu H, Jiao C, Xu L, Qian X, Yao H, Chen Q, Xu F, Fu F, Feng Y, Wang L, Chen X. A novel visceral pain model of uterine cervix inflammation in rat. Eur J Pharmacol 2021; 900:174080. [PMID: 33811839 DOI: 10.1016/j.ejphar.2021.174080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 03/21/2021] [Accepted: 03/26/2021] [Indexed: 10/21/2022]
Abstract
Treatment of visceral pain originating from the uterine cervix is a substantial clinical problem. The underlying mechanisms of such visceral pain remain unclear mainly due to a lack of reliable model. This study aimed to develop and evaluate the performance of a rat model of pain induced by uterine cervix inflammation. Rats were randomized to six groups according to the solution injected into the uterine cervix: normal saline, vehicle, capsaicin (0.3 mg, 0.6 mg, 0.9 mg), capsaicin 0.9 mg + morphine (n = 15 in each group). Spontaneous behaviors after cervical injection were recorded by a computerized video system and analyzed offline. An equation for calculating a novel pain score was derived from particular behaviors, based on Pearson's correlation analysis and regression analysis. c-Fos expression in the spinal cord was detected. The pain score and c-fos expression in the spinal cord were highest in the 0.9 mg capsaicin group and lowest in the normal saline and vehicle groups (P < 0.05). Intrathecal morphine significantly decreased the pain score (P < 0.05) and c-fos expression in the spinal cord (P < 0.05). Injection of capsaicin into the uterine cervix in rats could be a practical model of inflammatory cervical pain, which can be evaluated using our novel pain score. This model will provide further insight into the mechanism underlying visceral pain originating from the uterine cervix.
Collapse
Affiliation(s)
- Qi Xu
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lihong Sun
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenxin Zhang
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hui Wu
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Cuicui Jiao
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lili Xu
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaowei Qian
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Huaqi Yao
- Department of Anesthesia, Maternity and Child Care Hospital, Huzhou, China
| | - Qing Chen
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fang Xu
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Feng Fu
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ying Feng
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Luyang Wang
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xinzhong Chen
- Department of Anesthesia, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
38
|
Inserra A, De Gregorio D, Gobbi G. Psychedelics in Psychiatry: Neuroplastic, Immunomodulatory, and Neurotransmitter Mechanisms. Pharmacol Rev 2021; 73:202-277. [PMID: 33328244 DOI: 10.1124/pharmrev.120.000056] [Citation(s) in RCA: 118] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mounting evidence suggests safety and efficacy of psychedelic compounds as potential novel therapeutics in psychiatry. Ketamine has been approved by the Food and Drug Administration in a new class of antidepressants, and 3,4-methylenedioxymethamphetamine (MDMA) is undergoing phase III clinical trials for post-traumatic stress disorder. Psilocybin and lysergic acid diethylamide (LSD) are being investigated in several phase II and phase I clinical trials. Hence, the concept of psychedelics as therapeutics may be incorporated into modern society. Here, we discuss the main known neurobiological therapeutic mechanisms of psychedelics, which are thought to be mediated by the effects of these compounds on the serotonergic (via 5-HT2A and 5-HT1A receptors) and glutamatergic [via N-methyl-d-aspartate (NMDA) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors] systems. We focus on 1) neuroplasticity mediated by the modulation of mammalian target of rapamycin-, brain-derived neurotrophic factor-, and early growth response-related pathways; 2) immunomodulation via effects on the hypothalamic-pituitary-adrenal axis, nuclear factor ĸB, and cytokines such as tumor necrosis factor-α and interleukin 1, 6, and 10 production and release; and 3) modulation of serotonergic, dopaminergic, glutamatergic, GABAergic, and norepinephrinergic receptors, transporters, and turnover systems. We discuss arising concerns and ways to assess potential neurobiological changes, dependence, and immunosuppression. Although larger cohorts are required to corroborate preliminary findings, the results obtained so far are promising and represent a critical opportunity for improvement of pharmacotherapies in psychiatry, an area that has seen limited therapeutic advancement in the last 20 years. Studies are underway that are trying to decouple the psychedelic effects from the therapeutic effects of these compounds. SIGNIFICANCE STATEMENT: Psychedelic compounds are emerging as potential novel therapeutics in psychiatry. However, understanding of molecular mechanisms mediating improvement remains limited. This paper reviews the available evidence concerning the effects of psychedelic compounds on pathways that modulate neuroplasticity, immunity, and neurotransmitter systems. This work aims to be a reference for psychiatrists who may soon be faced with the possibility of prescribing psychedelic compounds as medications, helping them assess which compound(s) and regimen could be most useful for decreasing specific psychiatric symptoms.
Collapse
Affiliation(s)
- Antonio Inserra
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - Danilo De Gregorio
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - Gabriella Gobbi
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
39
|
Identifying Musculoskeletal Pain Generators Using Molecular Imaging. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00076-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
40
|
Andoh T, Suzuki K, Konno M, Tsuneyama K, Kuraishi Y. Pharmacological Characterization of a Novel Mouse Model of Cholestatic Pruritus. Biol Pharm Bull 2020; 43:1111-1117. [PMID: 32612073 DOI: 10.1248/bpb.b20-00097] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Patients with cholestatic liver diseases, such as primary biliary cirrhosis, usually suffer from pruritus. However, the pathogenesis of cholestatic pruritus is unclear, and there is no current effective treatment for it. In order to find a treatment for the condition, an appropriate mouse model should be developed. Therefore, here, we established a surgically-induced mouse model of cholestatic pruritus. The bile duct was ligated in order to block bile secretion from the anterior, right, and left lobes, with the exception of the caudate lobe. Serum levels of total bile acid increased after bile duct ligation (BDL). The spontaneous hind paw scratching was also increased in BDL mice. Spontaneous scratching was reduced in BDL mice by naloxone (µ-opioid receptor antagonist), U-50,488H (κ-opioid receptor agonist), and clonidine (α2-adrenoceptor agonist). Azelastine (H1 receptor antagonist with membrane-stabilizing activity) slightly reduced scratching. However, terfenadine (H1 receptor antagonist), methysergide (serotonin (5-HT)2 receptor antagonist), ondansetron (5-HT3 receptor antagonist), proteinase-activated receptor 2-neutralizing antibody, fluvoxamine (selective serotonin reuptake inhibitor), milnacipran (serotonin-noradrenalin reuptake inhibitor), and cyproheptadine (H1 and 5-HT2 receptor antagonist) did not affect scratching. These results suggested that partial obstruction of bile secretion in mice induced anti-histamine-resistant itching and that central opioid system is involved in cholestatic itching.
Collapse
Affiliation(s)
- Tsugunobu Andoh
- Department of Applied Pharmacology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama.,Department of Pathological Pharmacology, College of Pharmacy, Kinjo Gakuin University
| | - Kazunari Suzuki
- Department of Applied Pharmacology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama
| | - Mitsuhiro Konno
- Department of Applied Pharmacology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama
| | - Koichi Tsuneyama
- Department of Pathology and Laboratory Medicine, Institute of Health Biosciences, Tokushima University Graduate School
| | - Yasushi Kuraishi
- Department of Applied Pharmacology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama
| |
Collapse
|
41
|
Porter M, Xiao H, Maity S, Vail N, Smith SB, Topczewski JJ. Enhanced Affinity for 3-Amino-Chromane-Derived σ 1 Receptor Ligands. ACS OMEGA 2020; 5:32724-32737. [PMID: 33376910 PMCID: PMC7758967 DOI: 10.1021/acsomega.0c05117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 12/01/2020] [Indexed: 06/12/2023]
Abstract
The σ1 receptor is implicated in regulating a diverse range of physiology and is a target for developing therapies for cancer, pain management, neural degradation, and COVID-19. This report describes 36 phenethylamine-containing 3-amino-chromane ligands, which bind to σ1 with low nM affinities. The family consists of 18 distinct compounds and each enantiomer was independently assayed. Three compounds with the greatest affinity bind in the 2 nM K i range (∼8.7 pK i). Furthermore, ligands with the (3R,4R) absolute stereochemistry on the 3-amino-chromane core have a higher affinity and greater σ1 versus TMEM97 selectivity. The most promising ligands were assayed in 661W cells, which did not show significant protective effects.
Collapse
Affiliation(s)
- Matthew
R. Porter
- Department
of Chemistry, University of Minnesota Twin
Cities, Minneapolis, Minnesota 55455, United States
| | - Haiyan Xiao
- Department
of Cellular Biology and Anatomy, Medical
College of Georgia at Augusta University, Augusta, Georgia 30912, United States
- James
and Jean Culver Vision Discovery Institute, Augusta University, Augusta, Georgia 30912, United States
| | - Sanjay Maity
- Department
of Chemistry, University of Minnesota Twin
Cities, Minneapolis, Minnesota 55455, United States
| | - Nora Vail
- Department
of Chemistry, University of Minnesota Twin
Cities, Minneapolis, Minnesota 55455, United States
| | - Sylvia B. Smith
- Department
of Cellular Biology and Anatomy, Medical
College of Georgia at Augusta University, Augusta, Georgia 30912, United States
- James
and Jean Culver Vision Discovery Institute, Augusta University, Augusta, Georgia 30912, United States
- Department
of Ophthalmology, Medical College of Georgia
at Augusta University, Augusta, Georgia 30912, United States
| | - Joseph J. Topczewski
- Department
of Chemistry, University of Minnesota Twin
Cities, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
42
|
Juárez-Contreras R, Méndez-Reséndiz KA, Rosenbaum T, González-Ramírez R, Morales-Lázaro SL. TRPV1 Channel: A Noxious Signal Transducer That Affects Mitochondrial Function. Int J Mol Sci 2020; 21:ijms21238882. [PMID: 33255148 PMCID: PMC7734572 DOI: 10.3390/ijms21238882] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/24/2020] [Accepted: 10/31/2020] [Indexed: 12/19/2022] Open
Abstract
The Transient Receptor Vanilloid 1 (TRPV1) or capsaicin receptor is a nonselective cation channel, which is abundantly expressed in nociceptors. This channel is an important transducer of several noxious stimuli, having a pivotal role in pain development. Several TRPV1 studies have focused on understanding its structure and function, as well as on the identification of compounds that regulate its activity. The intracellular roles of these channels have also been explored, highlighting TRPV1′s actions in the homeostasis of Ca2+ in organelles such as the mitochondria. These studies have evidenced how the activation of TRPV1 affects mitochondrial functions and how this organelle can regulate TRPV1-mediated nociception. The close relationship between this channel and mitochondria has been determined in neuronal and non-neuronal cells, demonstrating that TRPV1 activation strongly impacts on cell physiology. This review focuses on describing experimental evidence showing that TRPV1 influences mitochondrial function.
Collapse
Affiliation(s)
- Rebeca Juárez-Contreras
- Department of Cognitive Neuroscience, Neurosciences Division, Institute of Cellular Physiology, National Autonomous University of Mexico, UNAM, Mexico City 04510, Mexico; (R.J.-C.); (K.A.M.-R.); (T.R.)
| | - Karina Angélica Méndez-Reséndiz
- Department of Cognitive Neuroscience, Neurosciences Division, Institute of Cellular Physiology, National Autonomous University of Mexico, UNAM, Mexico City 04510, Mexico; (R.J.-C.); (K.A.M.-R.); (T.R.)
| | - Tamara Rosenbaum
- Department of Cognitive Neuroscience, Neurosciences Division, Institute of Cellular Physiology, National Autonomous University of Mexico, UNAM, Mexico City 04510, Mexico; (R.J.-C.); (K.A.M.-R.); (T.R.)
| | - Ricardo González-Ramírez
- Department of Molecular Biology and Histocompatibility, “Dr. Manuel Gea González” General Hospital, Mexico City 14080, Mexico;
| | - Sara Luz Morales-Lázaro
- Department of Cognitive Neuroscience, Neurosciences Division, Institute of Cellular Physiology, National Autonomous University of Mexico, UNAM, Mexico City 04510, Mexico; (R.J.-C.); (K.A.M.-R.); (T.R.)
- Correspondence:
| |
Collapse
|
43
|
Sanjai Kumar P, Nayak TK, Mahish C, Sahoo SS, Radhakrishnan A, De S, Datey A, Sahu RP, Goswami C, Chattopadhyay S, Chattopadhyay S. Inhibition of transient receptor potential vanilloid 1 (TRPV1) channel regulates chikungunya virus infection in macrophages. Arch Virol 2020; 166:139-155. [PMID: 33125586 DOI: 10.1007/s00705-020-04852-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 09/08/2020] [Indexed: 11/29/2022]
Abstract
Chikungunya virus (CHIKV), a virus that induces pathogenic inflammatory host immune responses, is re-emerging worldwide, and there are currently no established antiviral control measures. Transient receptor potential vanilloid 1 (TRPV1), a non-selective Ca2+-permeable ion channel, has been found to regulate various host inflammatory responses including several viral infections. Immune responses to CHIKV infection in host macrophages have been reported recently. However, the possible involvement of TRPV1 during CHIKV infection in host macrophages has not been studied. Here, we investigated the possible role of TRPV1 in CHIKV infection of the macrophage cell line RAW 264.7. It was found that CHIKV infection upregulates TRPV1 expression in macrophages. To confirm this observation, the TRPV1-specific modulators 5'-iodoresiniferatoxin (5'-IRTX, a TRPV1 antagonist) and resiniferatoxin (RTX, a TRPV1 agonist) were used. Our results indicated that TRPV1 inhibition leads to a reduction in CHIKV infection, whereas TRPV1 activation significantly enhances CHIKV infection. Using a plaque assay and a time-of-addition assay, it was observed that functional modulation of TRPV1 affects the early stages of the viral lifecycle in RAW 264.7 cells. Moreover, CHIKV infection was found to induce of pNF-κB (p65) expression and nuclear localization. However, both activation and inhibition of TRPV1 were found to enhance the expression and nuclear localization of pNF-κB (p65) and production of pro-inflammatory TNF and IL-6 during CHIKV infection. In addition, it was demonstrated by Ca2+ imaging that TRPV1 regulates Ca2+ influx during CHIKV infection. Hence, the current findings highlight a potentially important regulatory role of TRPV1 during CHIKV infection in macrophages. This study might also have broad implications in the context of other viral infections as well.
Collapse
Affiliation(s)
- P Sanjai Kumar
- School of Biological Sciences, National Institute of Science Education & Research, Bhubaneswar, HBNI, Jatni, Khurda, Odisha, 752050, India
| | - Tapas K Nayak
- School of Biological Sciences, National Institute of Science Education & Research, Bhubaneswar, HBNI, Jatni, Khurda, Odisha, 752050, India.,Infectious Disease Biology, Institute of Life Sciences, (Autonomous Institute of Department of Biotechnology, Government of India), Nalco Square, Bhubaneswar, Odisha, 751023, India
| | - Chandan Mahish
- School of Biological Sciences, National Institute of Science Education & Research, Bhubaneswar, HBNI, Jatni, Khurda, Odisha, 752050, India
| | - Subhransu S Sahoo
- School of Biological Sciences, National Institute of Science Education & Research, Bhubaneswar, HBNI, Jatni, Khurda, Odisha, 752050, India
| | - Anukrishna Radhakrishnan
- School of Biological Sciences, National Institute of Science Education & Research, Bhubaneswar, HBNI, Jatni, Khurda, Odisha, 752050, India
| | - Saikat De
- Infectious Disease Biology, Institute of Life Sciences, (Autonomous Institute of Department of Biotechnology, Government of India), Nalco Square, Bhubaneswar, Odisha, 751023, India
| | - Ankita Datey
- Infectious Disease Biology, Institute of Life Sciences, (Autonomous Institute of Department of Biotechnology, Government of India), Nalco Square, Bhubaneswar, Odisha, 751023, India
| | - Ram P Sahu
- School of Biological Sciences, National Institute of Science Education & Research, Bhubaneswar, HBNI, Jatni, Khurda, Odisha, 752050, India
| | - Chandan Goswami
- School of Biological Sciences, National Institute of Science Education & Research, Bhubaneswar, HBNI, Jatni, Khurda, Odisha, 752050, India
| | - Soma Chattopadhyay
- Infectious Disease Biology, Institute of Life Sciences, (Autonomous Institute of Department of Biotechnology, Government of India), Nalco Square, Bhubaneswar, Odisha, 751023, India.
| | - Subhasis Chattopadhyay
- School of Biological Sciences, National Institute of Science Education & Research, Bhubaneswar, HBNI, Jatni, Khurda, Odisha, 752050, India.
| |
Collapse
|
44
|
Yoon D, Kogan F, Gold GE, Biswal S. Identifying Musculoskeletal Pain Generators Using Clinical PET. Semin Musculoskelet Radiol 2020; 24:441-450. [DOI: 10.1055/s-0040-1713607] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
AbstractIdentifying the source of a person's pain is a significant clinical challenge because the physical sensation of pain is believed to be subjective and difficult to quantify. The experience of pain is not only modulated by the individual's threshold to painful stimuli but also a product of the person's affective contributions, such as fear, anxiety, and previous experiences. Perhaps then to quantify pain is to examine the degree of nociception and pro-nociceptive inflammation, that is, the extent of cellular, chemical, and molecular changes that occur in pain-generating processes. Measuring changes in the local density of receptors, ion channels, mediators, and inflammatory/immune cells that are involved in the painful phenotype using targeted, highly sensitive, and specific positron emission tomography (PET) radiotracers is therefore a promising approach toward objectively identifying peripheral pain generators. Although several preclinical radiotracer candidates are being developed, a growing number of ongoing clinical PET imaging approaches can measure the degree of target concentration and thus serve as a readout for sites of pain generation. Further, when PET is combined with the spatial and contrast resolution afforded by magnetic resonance imaging, nuclear medicine physicians and radiologists can potentially identify pain drivers with greater accuracy and confidence. Clinical PET imaging approaches with fluorine-18 fluorodeoxyglucose, fluorine-18 sodium fluoride, and sigma-1 receptor PET radioligand and translocator protein radioligands to isolate the source of pain are described here.
Collapse
Affiliation(s)
- Daehyun Yoon
- Division of Musculoskeletal Radiology, Department of Radiology, Stanford University School of Medicine, Stanford, California
| | - Feliks Kogan
- Division of Musculoskeletal Radiology, Department of Radiology, Stanford University School of Medicine, Stanford, California
| | - Garry E. Gold
- Division of Musculoskeletal Radiology, Department of Radiology, Stanford University School of Medicine, Stanford, California
| | - Sandip Biswal
- Division of Musculoskeletal Radiology, Department of Radiology, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
45
|
Steroids and TRP Channels: A Close Relationship. Int J Mol Sci 2020; 21:ijms21113819. [PMID: 32471309 PMCID: PMC7325571 DOI: 10.3390/ijms21113819] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/02/2020] [Accepted: 05/04/2020] [Indexed: 02/07/2023] Open
Abstract
Transient receptor potential (TRP) channels are remarkable transmembrane protein complexes that are essential for the physiology of the tissues in which they are expressed. They function as non-selective cation channels allowing for the signal transduction of several chemical, physical and thermal stimuli and modifying cell function. These channels play pivotal roles in the nervous and reproductive systems, kidney, pancreas, lung, bone, intestine, among others. TRP channels are finely modulated by different mechanisms: regulation of their function and/or by control of their expression or cellular/subcellular localization. These mechanisms are subject to being affected by several endogenously-produced compounds, some of which are of a lipidic nature such as steroids. Fascinatingly, steroids and TRP channels closely interplay to modulate several physiological events. Certain TRP channels are affected by the typical genomic long-term effects of steroids but others are also targets for non-genomic actions of some steroids that act as direct ligands of these receptors, as will be reviewed here.
Collapse
|
46
|
Garami A, Shimansky YP, Rumbus Z, Vizin RCL, Farkas N, Hegyi J, Szakacs Z, Solymar M, Csenkey A, Chiche DA, Kapil R, Kyle DJ, Van Horn WD, Hegyi P, Romanovsky AA. Hyperthermia induced by transient receptor potential vanilloid-1 (TRPV1) antagonists in human clinical trials: Insights from mathematical modeling and meta-analysis. Pharmacol Ther 2020; 208:107474. [PMID: 31926897 DOI: 10.1016/j.pharmthera.2020.107474] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 12/23/2019] [Indexed: 02/06/2023]
Abstract
Antagonists of the transient receptor potential vanilloid-1 (TRPV1) channel alter body temperature (Tb) in laboratory animals and humans: most cause hyperthermia; some produce hypothermia; and yet others have no effect. TRPV1 can be activated by capsaicin (CAP), protons (low pH), and heat. First-generation (polymodal) TRPV1 antagonists potently block all three TRPV1 activation modes. Second-generation (mode-selective) TRPV1 antagonists potently block channel activation by CAP, but exert different effects (e.g., potentiation, no effect, or low-potency inhibition) in the proton mode, heat mode, or both. Based on our earlier studies in rats, only one mode of TRPV1 activation - by protons - is involved in thermoregulatory responses to TRPV1 antagonists. In rats, compounds that potently block, potentiate, or have no effect on proton activation cause hyperthermia, hypothermia, or no effect on Tb, respectively. A Tb response occurs when a TRPV1 antagonist blocks (in case of hyperthermia) or potentiates (hypothermia) the tonic TRPV1 activation by protons somewhere in the trunk, perhaps in muscles, and - via the acido-antithermogenic and acido-antivasoconstrictor reflexes - modulates thermogenesis and skin vasoconstriction. In this work, we used a mathematical model to analyze Tb data from human clinical trials of TRPV1 antagonists. The analysis suggests that, in humans, the hyperthermic effect depends on the antagonist's potency to block TRPV1 activation not only by protons, but also by heat, while the CAP activation mode is uninvolved. Whereas in rats TRPV1 drives thermoeffectors by mediating pH signals from the trunk, but not Tb signals, our analysis suggests that TRPV1 mediates both pH and thermal signals driving thermoregulation in humans. Hence, in humans (but not in rats), TRPV1 is likely to serve as a thermosensor of the thermoregulation system. We also conducted a meta-analysis of Tb data from human trials and found that polymodal TRPV1 antagonists (ABT-102, AZD1386, and V116517) increase Tb, whereas the mode-selective blocker NEO6860 does not. Several strategies of harnessing the thermoregulatory effects of TRPV1 antagonists in humans are discussed.
Collapse
Affiliation(s)
- Andras Garami
- Department of Thermophysiology, Institute for Translational Medicine, Medical School, University of Pecs, Pecs, Hungary.
| | - Yury P Shimansky
- Department of Neurobiology, Barrow Neurological Institute, Dignity Health, Phoenix, AZ, USA
| | - Zoltan Rumbus
- Department of Thermophysiology, Institute for Translational Medicine, Medical School, University of Pecs, Pecs, Hungary
| | - Robson C L Vizin
- Thermoregulation and Systemic Inflammation Laboratory (FeverLab), Trauma Research, St. Joseph's Hospital and Medical Center, Dignity Health, Phoenix, AZ, USA
| | - Nelli Farkas
- Institute for Translational Medicine, Medical School and Szentagothai Research Centre, University of Pecs, Pecs, Hungary
| | - Judit Hegyi
- Institute for Translational Medicine, Medical School and Szentagothai Research Centre, University of Pecs, Pecs, Hungary
| | - Zsolt Szakacs
- Institute for Translational Medicine, Medical School and Szentagothai Research Centre, University of Pecs, Pecs, Hungary
| | - Margit Solymar
- Department of Thermophysiology, Institute for Translational Medicine, Medical School, University of Pecs, Pecs, Hungary
| | - Alexandra Csenkey
- Department of Thermophysiology, Institute for Translational Medicine, Medical School, University of Pecs, Pecs, Hungary
| | | | | | | | - Wade D Van Horn
- School of Molecular Sciences, Arizona State University, Tempe, AZ, USA
| | - Peter Hegyi
- Institute for Translational Medicine, Medical School and Szentagothai Research Centre, University of Pecs, Pecs, Hungary; Department of Translational Medicine, First Department of Medicine, Medical School, University of Pecs, Pecs, Hungary
| | - Andrej A Romanovsky
- Thermoregulation and Systemic Inflammation Laboratory (FeverLab), Trauma Research, St. Joseph's Hospital and Medical Center, Dignity Health, Phoenix, AZ, USA; School of Molecular Sciences, Arizona State University, Tempe, AZ, USA; Zharko Pharma Inc., Olympia, WA, USA.
| |
Collapse
|
47
|
" Bridging the Gap" Everything that Could Have Been Avoided If We Had Applied Gender Medicine, Pharmacogenetics and Personalized Medicine in the Gender-Omics and Sex-Omics Era. Int J Mol Sci 2019; 21:ijms21010296. [PMID: 31906252 PMCID: PMC6982247 DOI: 10.3390/ijms21010296] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 12/21/2019] [Accepted: 12/30/2019] [Indexed: 02/06/2023] Open
Abstract
Gender medicine is the first step of personalized medicine and patient-centred care, an essential development to achieve the standard goal of a holistic approach to patients and diseases. By addressing the interrelation and integration of biological markers (i.e., sex) with indicators of psychological/cultural behaviour (i.e., gender), gender medicine represents the crucial assumption for achieving the personalized health-care required in the third millennium. However, ‘sex’ and ‘gender’ are often misused as synonyms, leading to frequent misunderstandings in those who are not deeply involved in the field. Overall, we have to face the evidence that biological, genetic, epigenetic, psycho-social, cultural, and environmental factors mutually interact in defining sex/gender differences, and at the same time in establishing potential unwanted sex/gender disparities. Prioritizing the role of sex/gender in physiological and pathological processes is crucial in terms of efficient prevention, clinical signs’ identification, prognosis definition, and therapy optimization. In this regard, the omics-approach has become a powerful tool to identify sex/gender-specific disease markers, with potential benefits also in terms of socio-psychological wellbeing for each individual, and cost-effectiveness for National Healthcare systems. “Being a male or being a female” is indeed important from a health point of view and it is no longer possible to avoid “sex and gender lens” when approaching patients. Accordingly, personalized healthcare must be based on evidence from targeted research studies aimed at understanding how sex and gender influence health across the entire life span. The rapid development of genetic tools in the molecular medicine approaches and their impact in healthcare is an example of highly specialized applications that have moved from specialists to primary care providers (e.g., pharmacogenetic and pharmacogenomic applications in routine medical practice). Gender medicine needs to follow the same path and become an established medical approach. To face the genetic, molecular and pharmacological bases of the existing sex/gender gap by means of omics approaches will pave the way to the discovery and identification of novel drug-targets/therapeutic protocols, personalized laboratory tests and diagnostic procedures (sex/gender-omics). In this scenario, the aim of the present review is not to simply resume the state-of-the-art in the field, rather an opportunity to gain insights into gender medicine, spanning from molecular up to social and psychological stances. The description and critical discussion of some key selected multidisciplinary topics considered as paradigmatic of sex/gender differences and sex/gender inequalities will allow to draft and design strategies useful to fill the existing gap and move forward.
Collapse
|
48
|
Noyer L, Lemonnier L, Mariot P, Gkika D. Partners in Crime: Towards New Ways of Targeting Calcium Channels. Int J Mol Sci 2019; 20:ijms20246344. [PMID: 31888223 PMCID: PMC6940757 DOI: 10.3390/ijms20246344] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 12/05/2019] [Accepted: 12/13/2019] [Indexed: 12/16/2022] Open
Abstract
The characterization of calcium channel interactome in the last decades opened a new way of perceiving ion channel function and regulation. Partner proteins of ion channels can now be considered as major components of the calcium homeostatic mechanisms, while the reinforcement or disruption of their interaction with the channel units now represents an attractive target in research and therapeutics. In this review we will focus on the targeting of calcium channel partner proteins in order to act on the channel activity, and on its consequences for cell and organism physiology. Given the recent advances in the partner proteins’ identification, characterization, as well as in the resolution of their interaction domain structures, we will develop the latest findings on the interacting proteins of the following channels: voltage-dependent calcium channels, transient receptor potential and ORAI channels, and inositol 1,4,5-trisphosphate receptor.
Collapse
Affiliation(s)
- Lucile Noyer
- Univ. Lille, Inserm, U1003-PHYCEL-Physiologie Cellulaire, F-59000 Lille, France; (L.N.); (L.L.); (P.M.)
- Laboratory of Excellence, Ion Channels Science and Therapeutics, Université de Lille, 59655 Villeneuve d’Ascq, France
| | - Loic Lemonnier
- Univ. Lille, Inserm, U1003-PHYCEL-Physiologie Cellulaire, F-59000 Lille, France; (L.N.); (L.L.); (P.M.)
- Laboratory of Excellence, Ion Channels Science and Therapeutics, Université de Lille, 59655 Villeneuve d’Ascq, France
| | - Pascal Mariot
- Univ. Lille, Inserm, U1003-PHYCEL-Physiologie Cellulaire, F-59000 Lille, France; (L.N.); (L.L.); (P.M.)
- Laboratory of Excellence, Ion Channels Science and Therapeutics, Université de Lille, 59655 Villeneuve d’Ascq, France
| | - Dimitra Gkika
- Univ. Lille, Inserm, U1003-PHYCEL-Physiologie Cellulaire, F-59000 Lille, France; (L.N.); (L.L.); (P.M.)
- Laboratory of Excellence, Ion Channels Science and Therapeutics, Université de Lille, 59655 Villeneuve d’Ascq, France
- Correspondence: ; Tél.: +33-(0)3-2043-6838
| |
Collapse
|
49
|
Wang X, Yang XL, Kong WL, Zeng ML, Shao L, Jiang GT, Cheng JJ, Kong S, He XH, Liu WH, Chen TX, Peng BW. TRPV1 translocated to astrocytic membrane to promote migration and inflammatory infiltration thus promotes epilepsy after hypoxic ischemia in immature brain. J Neuroinflammation 2019; 16:214. [PMID: 31722723 PMCID: PMC6852893 DOI: 10.1186/s12974-019-1618-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 10/16/2019] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Neonatal hypoxic-ischemic brain damage (HIBD), a leading cause of neonatal mortality, has intractable sequela such as epilepsy that seriously affected the life quality of HIBD survivors. We have previously shown that ion channel dysfunction in the central nervous system played an important role in the process of HIBD-induced epilepsy. Therefore, we continued to validate the underlying mechanisms of TRPV1 as a potential target for epilepsy. METHODS Neonatal hypoxic ischemia and oxygen-glucose deprivation (OGD) were used to simulate HIBD in vivo and in vitro. Primarily cultured astrocytes were used to assess the expression of TRPV1, glial fibrillary acidic protein (GFAP), cytoskeletal rearrangement, and inflammatory cytokines by using Western blot, q-PCR, and immunofluorescence. Furthermore, brain electrical activity in freely moving mice was recorded by electroencephalography (EEG). TRPV1 current and neuronal excitability were detected by whole-cell patch clamp. RESULTS Astrocytic TRPV1 translocated to the membrane after OGD. Mechanistically, astrocytic TRPV1 activation increased the inflow of Ca2+, which promoted G-actin polymerized to F-actin, thus promoted astrocyte migration after OGD. Moreover, astrocytic TRPV1 deficiency decreased the production and release of pro-inflammatory cytokines (TNF, IL-6, IL-1β, and iNOS) after OGD. It could also dramatically attenuate neuronal excitability after OGD and brain electrical activity in HIBD mice. Behavioral testing for seizures after HIBD revealed that TRPV1 knockout mice demonstrated prolonged onset latency, shortened duration, and decreased seizure severity when compared with wild-type mice. CONCLUSIONS Collectively, TRPV1 promoted astrocyte migration thus helped the infiltration of pro-inflammatory cytokines (TNF, IL-1β, IL-6, and iNOS) from astrocytes into the vicinity of neurons to promote epilepsy. Our study provides a strong rationale for astrocytic TRPV1 to be a therapeutic target for anti-epileptogenesis after HIBD.
Collapse
Affiliation(s)
- Xin Wang
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Xing-Liang Yang
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Wei-Lin Kong
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Meng-Liu Zeng
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Lin Shao
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Guang-Tong Jiang
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Jing-Jing Cheng
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Shuo Kong
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Xiao-Hua He
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Wan-Hong Liu
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Tao-Xiang Chen
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Bi-Wen Peng
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
50
|
Porter MR, Xiao H, Wang J, Smith SB, Topczewski JJ. 3-Amino-chromanes and Tetrahydroquinolines as Selective 5-HT 2B, 5-HT 7, or σ 1 Receptor Ligands. ACS Med Chem Lett 2019; 10:1436-1442. [PMID: 31620230 DOI: 10.1021/acsmedchemlett.9b00225] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 09/23/2019] [Indexed: 12/26/2022] Open
Abstract
The phenethylamine backbone is a privileged substructure found in a wide variety of G protein-coupled receptor (GPCR) ligands. This includes both endogenous neurotransmitters and active pharmaceutical agents. More than 20 structurally unique heterocyclic phenethylamine derivatives were broadly evaluated for GPCR affinity. Selective ligands for the 5-HT2B, 5-HT7, and σ1 receptors were identified, each with low nanomolar binding affinities. The σ1 receptor affinity was supported in a cellular assay that provided evidence for increased cell survival under oxidative stress.
Collapse
Affiliation(s)
- Matthew R. Porter
- Department of Chemistry, University of Minnesota Twin Cities, Minneapolis, Minnesota 55455, United States
| | - Haiyan Xiao
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia 30912, United States
- James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, Georgia 30912, United States
| | - Jing Wang
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia 30912, United States
- James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, Georgia 30912, United States
| | - Sylvia B. Smith
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia 30912, United States
- James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, Georgia 30912, United States
- Department of Ophthalmology, Medical College of Georgia at Augusta University, Augusta, Georgia 30912, United States
| | - Joseph J. Topczewski
- Department of Chemistry, University of Minnesota Twin Cities, Minneapolis, Minnesota 55455, United States
| |
Collapse
|