1
|
Huang R, Liu Z, Pan Y, Ma Z, Wang H, Wan B, Li J, Chang J. Mechanistic insight into the neurodevelopmental toxicity of the novel pesticide pyrifluquinazon (PFQ) and its major metabolite in early-life stage zebrafish. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 366:125469. [PMID: 39643230 DOI: 10.1016/j.envpol.2024.125469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 11/24/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024]
Abstract
Pyrifluquinazon (PFQ), a novel insecticide containing a heptafluoroisopropyl moiety, has seen increasing use. However, limited research has been conducted on the toxicological effects and mechanisms of PFQ in aquatic organisms. To investigate the toxicity and underlying mechanisms of PFQ and its primary metabolite dPFQ in aquatic organisms, morphological, behavioral, hormonal, multi-omics analyses, and molecular docking studies were conducted on zebrafish larvae after exposure. The results showed that both PFQ and dPFQ induced developmental abnormalities, behavioral impairment, hormonal disruptions, and alterations in neurologically related metabolites and gene expression in early-stage zebrafish. Notably, delayed retinal vascular development was observed, which is also likely linked to the neurodevelopmental toxicity. Subsequently, identification and relative quantification of PFQ metabolites suggested that its toxicity might be primarily attributed to dPFQ. Finally, an Adverse Outcome Pathway (AOP) was proposed, initiating with the binding of dPFQ to the TRPV4 protein and ultimately leading to neurodevelopmental toxicity. This study delineated the neurodevelopmental toxicity of PFQ and its toxicological mechanisms in zebrafish, emphasizing the hazards posed by pesticide metabolites to non-target organisms and highlighting inherent limitations of extrapolating in vitro toxicity experiments.
Collapse
Affiliation(s)
- Rui Huang
- Laboratory for Chemical Environmental Risk Assessment, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Shuangqing RD 18, Beijing, 100085, China; University of Chinese Academy of Sciences, Yuquan RD 19 a, Beijing, 100049, China
| | - Zijun Liu
- Laboratory for Chemical Environmental Risk Assessment, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Shuangqing RD 18, Beijing, 100085, China
| | - Yunrui Pan
- Laboratory for Chemical Environmental Risk Assessment, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Shuangqing RD 18, Beijing, 100085, China; University of Chinese Academy of Sciences, Yuquan RD 19 a, Beijing, 100049, China
| | - Zheng Ma
- Laboratory for Chemical Environmental Risk Assessment, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Shuangqing RD 18, Beijing, 100085, China; University of Chinese Academy of Sciences, Yuquan RD 19 a, Beijing, 100049, China
| | - Huili Wang
- Laboratory for Chemical Environmental Risk Assessment, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Shuangqing RD 18, Beijing, 100085, China
| | - Bin Wan
- Laboratory for Chemical Environmental Risk Assessment, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Shuangqing RD 18, Beijing, 100085, China; University of Chinese Academy of Sciences, Yuquan RD 19 a, Beijing, 100049, China
| | - Jianzhong Li
- Laboratory for Chemical Environmental Risk Assessment, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Shuangqing RD 18, Beijing, 100085, China
| | - Jing Chang
- Laboratory for Chemical Environmental Risk Assessment, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Shuangqing RD 18, Beijing, 100085, China.
| |
Collapse
|
2
|
Lötsch J, Gasimli K, Malkusch S, Hahnefeld L, Angioni C, Schreiber Y, Trautmann S, Wedel S, Thomas D, Ferreiros Bouzas N, Brandts CH, Schnappauf B, Solbach C, Geisslinger G, Sisignano M. Machine learning and biological validation identify sphingolipids as potential mediators of paclitaxel-induced neuropathy in cancer patients. eLife 2024; 13:RP91941. [PMID: 39347767 PMCID: PMC11444680 DOI: 10.7554/elife.91941] [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: 10/01/2024] Open
Abstract
Background Chemotherapy-induced peripheral neuropathy (CIPN) is a serious therapy-limiting side effect of commonly used anticancer drugs. Previous studies suggest that lipids may play a role in CIPN. Therefore, the present study aimed to identify the particular types of lipids that are regulated as a consequence of paclitaxel administration and may be associated with the occurrence of post-therapeutic neuropathy. Methods High-resolution mass spectrometry lipidomics was applied to quantify d=255 different lipid mediators in the blood of n=31 patients drawn before and after paclitaxel therapy for breast cancer treatment. A variety of supervised statistical and machine-learning methods was applied to identify lipids that were regulated during paclitaxel therapy or differed among patients with and without post-therapeutic neuropathy. Results Twenty-seven lipids were identified that carried relevant information to train machine learning algorithms to identify, in new cases, whether a blood sample was drawn before or after paclitaxel therapy with a median balanced accuracy of up to 90%. One of the top hits, sphinganine-1-phosphate (SA1P), was found to induce calcium transients in sensory neurons via the transient receptor potential vanilloid 1 (TRPV1) channel and sphingosine-1-phosphate receptors.SA1P also showed different blood concentrations between patients with and without neuropathy. Conclusions Present findings suggest a role for sphinganine-1-phosphate in paclitaxel-induced biological changes associated with neuropathic side effects. The identified SA1P, through its receptors, may provide a potential drug target for co-therapy with paclitaxel to reduce one of its major and therapy-limiting side effects. Funding This work was supported by the Deutsche Forschungsgemeinschaft (German Research Foundation, DFG, Grants SFB1039 A09 and Z01) and by the Fraunhofer Foundation Project: Neuropathic Pain as well as the Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD). This work was also supported by the Leistungszentrum Innovative Therapeutics (TheraNova) funded by the Fraunhofer Society and the Hessian Ministry of Science and Arts. Jörn Lötsch was supported by the Deutsche Forschungsgemeinschaft (DFG LO 612/16-1).
Collapse
Affiliation(s)
- Jörn Lötsch
- Institute of Clinical Pharmacology, Goethe - University, Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Frankfurt, Germany
| | - Khayal Gasimli
- Goethe University, Department of Gynecology and Obstetrics, Frankfurt, Germany
| | - Sebastian Malkusch
- Institute of Clinical Pharmacology, Goethe - University, Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Frankfurt, Germany
| | - Lisa Hahnefeld
- Institute of Clinical Pharmacology, Goethe - University, Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Frankfurt, Germany
| | - Carlo Angioni
- Institute of Clinical Pharmacology, Goethe - University, Frankfurt, Germany
| | - Yannick Schreiber
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Frankfurt, Germany
| | - Sandra Trautmann
- Institute of Clinical Pharmacology, Goethe - University, Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Frankfurt, Germany
| | - Saskia Wedel
- Institute of Clinical Pharmacology, Goethe - University, Frankfurt, Germany
| | - Dominique Thomas
- Institute of Clinical Pharmacology, Goethe - University, Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Frankfurt, Germany
| | - Nerea Ferreiros Bouzas
- Institute of Clinical Pharmacology, Goethe - University, Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Frankfurt, Germany
| | - Christian H Brandts
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Goethe University, University Cancer Center Frankfurt (UCT), Goethe University Hospital, Frankfurt, Germany
| | | | - Christine Solbach
- Goethe University, Department of Gynecology and Obstetrics, Frankfurt, Germany
| | - Gerd Geisslinger
- Institute of Clinical Pharmacology, Goethe - University, Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Frankfurt, Germany
| | - Marco Sisignano
- Institute of Clinical Pharmacology, Goethe - University, Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Frankfurt, Germany
| |
Collapse
|
3
|
Suito T, Tominaga M. Functional relationship between peripheral thermosensation and behavioral thermoregulation. Front Neural Circuits 2024; 18:1435757. [PMID: 39045140 PMCID: PMC11263211 DOI: 10.3389/fncir.2024.1435757] [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: 05/21/2024] [Accepted: 06/27/2024] [Indexed: 07/25/2024] Open
Abstract
Thermoregulation is a fundamental mechanism for maintaining homeostasis in living organisms because temperature affects essentially all biochemical and physiological processes. Effector responses to internal and external temperature cues are critical for achieving effective thermoregulation by controlling heat production and dissipation. Thermoregulation can be classified as physiological, which is observed primarily in higher organisms (homeotherms), and behavioral, which manifests as crucial physiological functions that are conserved across many species. Neuronal pathways for physiological thermoregulation are well-characterized, but those associated with behavioral regulation remain unclear. Thermoreceptors, including Transient Receptor Potential (TRP) channels, play pivotal roles in thermoregulation. Mammals have 11 thermosensitive TRP channels, the functions for which have been elucidated through behavioral studies using knockout mice. Behavioral thermoregulation is also observed in ectotherms such as the fruit fly, Drosophila melanogaster. Studies of Drosophila thermoregulation helped elucidate significant roles for thermoreceptors as well as regulatory actions of membrane lipids in modulating the activity of both thermosensitive TRP channels and thermoregulation. This review provides an overview of thermosensitive TRP channel functions in behavioral thermoregulation based on results of studies involving mice or Drosophila melanogaster.
Collapse
Affiliation(s)
- Takuto Suito
- Division of Cell Signaling, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan
- Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, Okazaki, Japan
| | - Makoto Tominaga
- Division of Cell Signaling, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan
- Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, Okazaki, Japan
- Nagoya Advanced Research and Development Center, Nagoya City University, Nagoya, Japan
| |
Collapse
|
4
|
Rezzani R, Favero G, Gianò M, Pinto D, Labanca M, van Noorden CJ, Rinaldi F. Transient Receptor Potential Channels in the Healthy and Diseased Blood-Brain Barrier. J Histochem Cytochem 2024; 72:199-231. [PMID: 38590114 PMCID: PMC11020746 DOI: 10.1369/00221554241246032] [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: 09/19/2023] [Accepted: 03/18/2024] [Indexed: 04/10/2024] Open
Abstract
The large family of transient receptor potential (TRP) channels are integral membrane proteins that function as environmental sensors and act as ion channels after activation by mechanical (touch), physical (heat, pain), and chemical stimuli (pungent compounds such as capsaicin). Most TRP channels are localized in the plasma membrane of cells but some of them are localized in membranes of organelles and function as intracellular Ca2+-ion channels. TRP channels are involved in neurological disorders but their precise role(s) and relevance in these disorders are not clear. Endothelial cells of the blood-brain barrier (BBB) express TRP channels such as TRP vanilloid 1-4 and are involved in thermal detection by regulating BBB permeability. In neurological disorders, TRP channels in the BBB are responsible for edema formation in the brain. Therefore, drug design to modulate locally activity of TRP channels in the BBB is a hot topic. Today, the application of TRP channel antagonists against neurological disorders is still limited.
Collapse
Affiliation(s)
- Rita Rezzani
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Interdipartimental University Center of Research Adaption and Regeneration of Tissues and Organs - ARTO, University of Brescia, Brescia, Italy
- Italian Society for the Study of Orofacial Pain (Società Italiana Studio Dolore Orofacciale - SISDO), Brescia, Italy
| | - Gaia Favero
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Interdipartimental University Center of Research Adaption and Regeneration of Tissues and Organs - ARTO, University of Brescia, Brescia, Italy
| | - Marzia Gianò
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Daniela Pinto
- Human Microbiome Advanced Project Institute, Milan, Italy
| | - Mauro Labanca
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Italian Society for the Study of Orofacial Pain (Società Italiana Studio Dolore Orofacciale - SISDO), Brescia, Italy
| | - Cornelis J.F. van Noorden
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia
| | - Fabio Rinaldi
- Human Microbiome Advanced Project Institute, Milan, Italy
| |
Collapse
|
5
|
Zhang Y, Lin G, Xue N, Wang Y, Du T, Liu H, Xiong W, Shang W, Wu H, Song L. Differential outcomes of high-fat diet on age-related rescaling of cochlear frequency place coding. FASEB J 2023; 37:e23167. [PMID: 37651093 DOI: 10.1096/fj.202300457rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 08/01/2023] [Accepted: 08/14/2023] [Indexed: 09/01/2023]
Abstract
Auditory frequency coding is place-specific, which depends on the mechanical coupling of the basilar membrane-outer hair cell (OHC)-tectorial membrane network. Prestin-based OHC electromotility improves cochlear frequency selectivity and sensitivity. Cochlear amplification determines the frequency coding wherein discrete sound frequencies find a 'best' place along the cochlear length. Loss of OHC is the leading cause of age-related hearing loss (ARHL) and is the most common cause of sensorineural hearing loss and compromised speech perception. Lipid interaction with Prestin impacts OHC function. It has been established that high-fat diet (HFD) is associated with ARHL. To determine whether genetic background and metabolism preserve cochlear frequency place coding, we examined the effect of HFD in C57BL/6J (B6) and CBA/CaJ (CBA) on ARHL.We found a significant rescuing effect on ARHL in aged B6 HFD cohort. Prestin levels and cell sizes were better maintained in the experimental B6-HFD group. We also found that distortion product otoacoustic emission (DPOAE) group delay measurement was preserved, which suggested stable frequency place coding. In contrast, the response to HFD in the CBA cohort was modest with no appreciable benefit to hearing threshold. Notably, group delay was shortened with age along with the control. In addition, the frequency dependent OHC nonlinear capacitance gradient was most pronounced at young age but decreased with age. Cochlear RNA-seq analysis revealed differential TRPV1 expression and lipid homeostasis. Activation of TRPV1 and downregulation of arachidonic acid led to downregulation of inflammatory response in B6 HFD, which protects the cochlea from ARHL. The genetic background and metabolic state-derived changes in OHC morphology and function collectively contribute to a redefined cochlear frequency place coding and improved age-related pitch perception.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Otolaryngology, Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Guotong Lin
- Department of Otolaryngology, Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Na Xue
- Department of Otolaryngology, Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Yi Wang
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Tingting Du
- Department of Otolaryngology, Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Huihui Liu
- Department of Otolaryngology, Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Wei Xiong
- Chinese Institute for Brain Research, Beijing, China
| | - Wei Shang
- Navy Clinical Medical School, Anhui Medical University, Hefei, China
- In Vitro Fertility (IVF) Center Department of Obstetrics and Gynecology, the Sixth Medical Center of PLA General Hospital, Beijing, China
- Department of Obstetrics and Gynecology, Chinese PLA General Hospital, Beijing, China
| | - Hao Wu
- Department of Otolaryngology, Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Lei Song
- Department of Otolaryngology, Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| |
Collapse
|
6
|
Zhang M, Ma Y, Ye X, Zhang N, Pan L, Wang B. TRP (transient receptor potential) ion channel family: structures, biological functions and therapeutic interventions for diseases. Signal Transduct Target Ther 2023; 8:261. [PMID: 37402746 DOI: 10.1038/s41392-023-01464-x] [Citation(s) in RCA: 59] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 03/26/2023] [Accepted: 04/25/2023] [Indexed: 07/06/2023] Open
Abstract
Transient receptor potential (TRP) channels are sensors for a variety of cellular and environmental signals. Mammals express a total of 28 different TRP channel proteins, which can be divided into seven subfamilies based on amino acid sequence homology: TRPA (Ankyrin), TRPC (Canonical), TRPM (Melastatin), TRPML (Mucolipin), TRPN (NO-mechano-potential, NOMP), TRPP (Polycystin), TRPV (Vanilloid). They are a class of ion channels found in numerous tissues and cell types and are permeable to a wide range of cations such as Ca2+, Mg2+, Na+, K+, and others. TRP channels are responsible for various sensory responses including heat, cold, pain, stress, vision and taste and can be activated by a number of stimuli. Their predominantly location on the cell surface, their interaction with numerous physiological signaling pathways, and the unique crystal structure of TRP channels make TRPs attractive drug targets and implicate them in the treatment of a wide range of diseases. Here, we review the history of TRP channel discovery, summarize the structures and functions of the TRP ion channel family, and highlight the current understanding of the role of TRP channels in the pathogenesis of human disease. Most importantly, we describe TRP channel-related drug discovery, therapeutic interventions for diseases and the limitations of targeting TRP channels in potential clinical applications.
Collapse
Affiliation(s)
- Miao Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- The Center for Microbes, Development and Health; Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yueming Ma
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xianglu Ye
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ning Zhang
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Lei Pan
- The Center for Microbes, Development and Health; Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, China.
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Bing Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
- Center for Pharmaceutics Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai, 201203, China.
| |
Collapse
|
7
|
Wedel S, Hahnefeld L, Schreiber Y, Namendorf C, Heymann T, Uhr M, Schmidt MV, de Bruin N, Hausch F, Thomas D, Geisslinger G, Sisignano M. SAFit2 ameliorates paclitaxel-induced neuropathic pain by reducing spinal gliosis and elevating pro-resolving lipid mediators. J Neuroinflammation 2023; 20:149. [PMID: 37355700 DOI: 10.1186/s12974-023-02835-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 06/16/2023] [Indexed: 06/26/2023] Open
Abstract
BACKGROUND Chemotherapy-induced neuropathic pain (CIPN) describes a pathological pain state that occurs dose-dependently as a side effect and can limit or even impede an effective cancer therapy. Unfortunately, current treatment possibilities for CIPN are remarkably confined and mostly inadequate as CIPN therapeutics themselves consist of low effectiveness and may induce severe side effects, pointing out CIPN as pathological entity with an emerging need for novel treatment targets. Here, we investigated whether the novel and highly specific FKBP51 inhibitor SAFit2 reduces paclitaxel-induced neuropathic pain. METHODS In this study, we used a well-established multiple low-dose paclitaxel model to investigate analgesic and anti-inflammatory properties of SAFit2. For this purpose, the behavior of the mice was recorded over 14 days and the mouse tissue was then analyzed using biochemical methods. RESULTS Here, we show that SAFit2 is capable to reduce paclitaxel-induced mechanical hypersensitivity in mice. In addition, we detected that SAFit2 shifts lipid levels in nervous tissue toward an anti-inflammatory and pro-resolving lipid profile that counteracts peripheral sensitization after paclitaxel treatment. Furthermore, SAFit2 reduced the activation of astrocytes and microglia in the spinal cord as well as the levels of pain-mediating chemokines. Its treatment also increased anti-inflammatory cytokines levels in neuronal tissues, ultimately leading to a resolution of neuroinflammation. CONCLUSIONS In summary, SAFit2 shows antihyperalgesic properties as it ameliorates paclitaxel-induced neuropathic pain by reducing peripheral sensitization and resolving neuroinflammation. Therefore, we consider SAFit2 as a potential novel drug candidate for the treatment of paclitaxel-induced neuropathic pain.
Collapse
Affiliation(s)
- Saskia Wedel
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt/ZAFES, University Hospital, Goethe-University, 60590, Frankfurt am Main, Germany
| | - Lisa Hahnefeld
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt/ZAFES, University Hospital, Goethe-University, 60590, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, 60596, Frankfurt am Main, Germany
| | - Yannick Schreiber
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, 60596, Frankfurt am Main, Germany
| | - Christian Namendorf
- Core Unit Analytics and Mass Spectrometry, Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Tim Heymann
- Department of Biochemistry, Technical University of Darmstadt, 64287, Darmstadt, Germany
| | - Manfred Uhr
- Core Unit Analytics and Mass Spectrometry, Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Mathias V Schmidt
- Core Unit Analytics and Mass Spectrometry, Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Natasja de Bruin
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, 60596, Frankfurt am Main, Germany
| | - Felix Hausch
- Department of Biochemistry, Technical University of Darmstadt, 64287, Darmstadt, Germany
| | - Dominique Thomas
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt/ZAFES, University Hospital, Goethe-University, 60590, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, 60596, Frankfurt am Main, Germany
| | - Gerd Geisslinger
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt/ZAFES, University Hospital, Goethe-University, 60590, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, 60596, Frankfurt am Main, Germany
| | - Marco Sisignano
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt/ZAFES, University Hospital, Goethe-University, 60590, Frankfurt am Main, Germany.
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, 60596, Frankfurt am Main, Germany.
| |
Collapse
|
8
|
The FKBP51 Inhibitor SAFit2 Restores the Pain-Relieving C16 Dihydroceramide after Nerve Injury. Int J Mol Sci 2022; 23:ijms232214274. [PMID: 36430751 PMCID: PMC9695264 DOI: 10.3390/ijms232214274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/14/2022] [Accepted: 11/16/2022] [Indexed: 11/19/2022] Open
Abstract
Neuropathic pain is a pathological pain state with a broad symptom scope that affects patients after nerve injuries, but it can also arise after infections or exposure to toxic substances. Current treatment possibilities are still limited because of the low efficacy and severe adverse effects of available therapeutics, highlighting an emerging need for novel analgesics and for a detailed understanding of the pathophysiological alterations in the onset and maintenance of neuropathic pain. Here, we show that the novel and highly specific FKBP51 inhibitor SAFit2 restores lipid signaling and metabolism in nervous tissue after nerve injury. More specifically, we identify that SAFit2 restores the levels of the C16 dihydroceramide, which significantly reduces the sensitization of the pain-mediating TRPV1 channel and subsequently the secretion of the pro-inflammatory neuropeptide CGRP in primary sensory neurons. Furthermore, we show that the C16 dihydroceramide is capable of reducing acute thermal hypersensitivity in a capsaicin mouse model. In conclusion, we report for the first time the C16 dihydroceramide as a novel and crucial lipid mediator in the context of neuropathic pain as it has analgesic properties, contributing to the pain-relieving properties of SAFit2.
Collapse
|
9
|
Differences in plasma lipoprotein profiles between patients with chronic peripheral neuropathic pain and healthy controls: an exploratory pilot study. Pain Rep 2022; 7:e1036. [PMID: 36203648 PMCID: PMC9529241 DOI: 10.1097/pr9.0000000000001036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 06/20/2022] [Accepted: 07/17/2022] [Indexed: 11/26/2022] Open
Abstract
Supplemental Digital Content is Available in the Text. Lipoprotein profiles were significantly different between patients with chronic peripheral neuropathic pain and healthy controls, indicative of ongoing systemic low-grade inflammation among the patients. Introduction: Little is still known about the underlying mechanisms that drive and maintain neuropathic pain (NeuP). Recently, lipids have been implicated as endogenous proalgesic ligands affecting onset and maintenance of pain; however, in the case of NeuP, the relationship is largely unexplored. Objectives: The aim of this study was to investigate the lipoprotein profile in patients with chronic peripheral NeuP compared with healthy controls. Methods: The concentrations of 112 lipoprotein fractions in plasma from patients with NeuP (n = 16) and healthy controls (n = 13) were analyzed using proton nuclear magnetic resonance spectroscopy. A multiplex immunoassay based on an electrochemiluminescent detection method was used to measure the concentration of 71 cytokines in plasma from patients with NeuP (n = 10) and healthy controls (n = 11). Multivariate data analysis was used to identify patterns of protein intercorrelations and proteins significant for group discrimination. Results: We found 23 lipoproteins that were significantly upregulated in patients with NeuP compared with healthy controls. When the influence of cytokines was included in a regression model, 30 proteins (8 cytokines and 22 lipoprotein fractions) were significantly upregulated or downregulated in patients with NeuP. Both conditions presented lipoprotein profiles consistent with inflammation. Body mass index did not affect lipoprotein profiles in either group. No relationship between age and lipoprotein pattern was found in NeuP, but a significant relationship was found in healthy controls. Conclusion: Patients with NeuP presented a lipoprotein profile consistent with systemic low-grade inflammation, like that seen in autoimmune, cardiometabolic, and neuroprogressive diseases. These preliminary results emphasize the importance of chronic low-grade inflammation in NeuP.
Collapse
|
10
|
Yamamoto S, Hashidate-Yoshida T, Shimizu T, Shindou H. Profiling of fatty acid metabolism in the dorsal root ganglion after peripheral nerve injury. FRONTIERS IN PAIN RESEARCH 2022; 3:948689. [PMID: 35965594 PMCID: PMC9372306 DOI: 10.3389/fpain.2022.948689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 07/06/2022] [Indexed: 12/04/2022] Open
Abstract
Peripheral nerve injury (PNI) induces neuronal hyperexcitability, which underlies neuropathic pain. The emergence of RNA sequencing technologies has enabled profiling of transcriptional changes in pathological conditions. However, these approaches do not provide information regarding metabolites such as lipids that are not directly encoded by genes. Fatty acids (FAs) are some of the essential lipids in mammalian organisms and are mainly stored as membrane phospholipids. In response to various biological stimuli, FAs are rapidly released and converted into several mediators, such as eicosanoids and docosanoids. FAs themselves or their metabolites play important roles in physiology and pathology. In this study, using a comprehensive lipidomic analysis of FA metabolites, 152 species were measured in the dorsal root ganglia of mice at multiple time points after PNI. We found that PNI increased the ω-6 FA metabolites produced by cyclooxygenases but not those produced by lipoxygenases or cytochrome P450 enzymes in the dorsal root ganglia. In contrast, ω-3 FA metabolites biosynthesized by any enzyme transiently increased after nerve injury. Overall, these findings provide a new resource and valuable insights into PNI pathologies, including pain and nerve regeneration.
Collapse
Affiliation(s)
- Shota Yamamoto
- Department of Lipid Life Science, National Center for Global Health and Medicine, Tokyo, Japan
- *Correspondence: Shota Yamamoto
| | | | - Takao Shimizu
- Institute of Microbial Chemistry, Tokyo, Japan
- Department of Lipid Signaling, National Center for Global Health and Medicine, Tokyo, Japan
| | - Hideo Shindou
- Department of Lipid Life Science, National Center for Global Health and Medicine, Tokyo, Japan
- Department of Medical Lipid Science, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Hideo Shindou
| |
Collapse
|
11
|
Oxidized linoleic acid metabolites maintain mechanical and thermal hypersensitivity during sub-chronic inflammatory pain. Biochem Pharmacol 2022; 198:114953. [PMID: 35149052 DOI: 10.1016/j.bcp.2022.114953] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 02/04/2022] [Accepted: 02/04/2022] [Indexed: 12/13/2022]
Abstract
Inflammatory pain serves as a protective defense mechanism which becomes pathological when it turns into chronic inflammatory pain. This transition is mediated by a variety of peripheral mediators that sensitize nociceptors and increase pain perception in sensory neurons. Besides cytokines, chemokines and growth factors, accumulating evidence shows that oxidized lipids, such as eicosanoids and oxidized linoleic acid metabolites, contribute to this sensitization process. Most notably, the oxidized linoleic acid metabolite and partial TRPV1 agonist 9-HODE (hydroxyoctadecadienoic acid) was shown to be involved in this sensitization process. However, it is still unknown how some of the oxidized linoleic acid metabolites are synthesized in the inflammatory environment and in which phase of inflammation they become relevant. Here we show that the concentrations of oxidized linoleic acid metabolites, especially 9-HODE and 13-HODE, are significantly increased in inflamed paw tissue and the corresponding dorsal root ganglia in the sub-chronic phase of inflammation. Surprisingly, classical inflammatory lipid markers, such as prostaglandins were at basal levels in this phase of inflammation. Moreover, we revealed the cell type specific synthesis pathways of oxidized linoleic acid metabolites in primary macrophages, primary neutrophils and dorsal root ganglia. Finally, we show that blocking the most elevated metabolites 9-HODE and 13-HODE at the site of inflammation in the sub-chronic phase of inflammation, leads to a significant relief of mechanical and thermal hypersensitivity in vivo. In summary, these data offer an approach to specifically target oxidized linoleic acid metabolites in the transition of acute inflammatory pain to chronic inflammatory pain.
Collapse
|
12
|
Cheng WWL, Arcario MJ, Petroff JT. Druggable Lipid Binding Sites in Pentameric Ligand-Gated Ion Channels and Transient Receptor Potential Channels. Front Physiol 2022; 12:798102. [PMID: 35069257 PMCID: PMC8777383 DOI: 10.3389/fphys.2021.798102] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 12/02/2021] [Indexed: 12/17/2022] Open
Abstract
Lipids modulate the function of many ion channels, possibly through direct lipid-protein interactions. The recent outpouring of ion channel structures by cryo-EM has revealed many lipid binding sites. Whether these sites mediate lipid modulation of ion channel function is not firmly established in most cases. However, it is intriguing that many of these lipid binding sites are also known sites for other allosteric modulators or drugs, supporting the notion that lipids act as endogenous allosteric modulators through these sites. Here, we review such lipid-drug binding sites, focusing on pentameric ligand-gated ion channels and transient receptor potential channels. Notable examples include sites for phospholipids and sterols that are shared by anesthetics and vanilloids. We discuss some implications of lipid binding at these sites including the possibility that lipids can alter drug potency or that understanding protein-lipid interactions can guide drug design. Structures are only the first step toward understanding the mechanism of lipid modulation at these sites. Looking forward, we identify knowledge gaps in the field and approaches to address them. These include defining the effects of lipids on channel function in reconstituted systems using asymmetric membranes and measuring lipid binding affinities at specific sites using native mass spectrometry, fluorescence binding assays, and computational approaches.
Collapse
Affiliation(s)
- Wayland W L Cheng
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, United States
| | - Mark J Arcario
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, United States
| | - John T Petroff
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, United States
| |
Collapse
|
13
|
|
14
|
Hamers A, Primus CP, Whitear C, Kumar NA, Masucci M, Montalvo Moreira SA, Rathod K, Chen J, Bubb K, Colas R, Khambata RS, Dalli J, Ahluwalia A. 20-HETE is a pivotal endogenous ligand for TRPV1-mediated neurogenic inflammation in the skin. Br J Pharmacol 2021; 179:1450-1469. [PMID: 34755897 DOI: 10.1111/bph.15726] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 10/11/2021] [Accepted: 10/26/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Transient receptor potential cation channel subfamily V member 1 (TRPV1) is localised to sensory C-fibres and its opening leads to membrane depolarization, resulting in neuropeptide release and neurogenic inflammation. However, the identity of the endogenous activator of TRPV1 in this setting is unknown. The arachidonic acid (AA) metabolites 12-hydroperoxyeicosatetraenoyl acid (12-HpETE) and 20-hydroxyeicosatetraenoic acid (20-HETE) have emerged as potential endogenous activators of TRPV1 however, whether these lipids underlie TRPV1-mediated neurogenic inflammation remains unknown. EXPERIMENTAL APPROACH we analysed human cantharidin-induced blister samples and inflammatory responses in TRPV1 transgenic mice. KEY RESULTS In a human cantharidin-blister model the potent TRPV1 activators 20-HETE but not 12-HETE (stable metabolite of 12-HpETE) correlated with AA levels. Similarly, in mice levels of 20-HETE (but not 12-HETE) and AA were strongly positively correlated within the inflammatory milieu. Furthermore, LPS-induced oedema formation and neutrophil recruitment were substantially and significantly attenuated by pharmacological block or genetic deletion of TRPV1 channels, inhibition of 20-HETE formation or SP receptor neurokinin 1 (NK1 ) blockade. LPS treatment also increased cytochrome-P450 ώ-hydroxylase gene expression, the enzyme responsible for 20-HETE production. CONCLUSIONS AND IMPLICATIONS Taken together, our findings suggest that endogenously generated 20-HETE activates TRPV1 causing C-fibre activation and consequent oedema formation. These findings identify a novel pathway that may be useful in the therapeutics of diseases/conditions characterized by a prominent neurogenic inflammation, as in several skin diseases.
Collapse
Affiliation(s)
- Alexander Hamers
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London
| | - Christopher P Primus
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London
| | - Charlotte Whitear
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London
| | - Nitin Ajit Kumar
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London
| | - Michael Masucci
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London
| | - Shanik A Montalvo Moreira
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London
| | - Krishnaraj Rathod
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London
| | - Jianmin Chen
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London
| | - Kristen Bubb
- Monash Cardiovascular Research Centre, Monash University, Melbourne, Australia
| | - Romain Colas
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London
| | - Rayomand S Khambata
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London
| | - Jesmond Dalli
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London
| | - Amrita Ahluwalia
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London
| |
Collapse
|
15
|
Yan Q, Tang J, Zhang X, Wu L, Xu Y, Wang L. Does Transient Receptor Potential Vanilloid Type 1 Alleviate or Aggravate Pathological Myocardial Hypertrophy? Front Pharmacol 2021; 12:681286. [PMID: 34040539 PMCID: PMC8143375 DOI: 10.3389/fphar.2021.681286] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 04/27/2021] [Indexed: 12/21/2022] Open
Abstract
Transient receptor potential vanilloid type 1 (TRPV1) is a non-selective cation channel, which is involved in the endogenous stress adaptation mechanism for protection of the heart as well as the occurrence and development of some heart diseases. Although the effect of activation of the TRPV1 channel on different types of non-neural cells in the heart remains unclear, most data show that stimulation of sensory nerves expressing TRPV1 or stimulation/overexpression of the TRPV1 channel has a beneficial role in heart disease. Some studies have proven that TRPV1 has an important relationship with pathological myocardial hypertrophy, but the specific mechanism and effect are not clear. In order to help researchers better understand the relationship between TRPV1 and pathological myocardial hypertrophy, this paper aims to summarize the effect of TRPV1 and the related mechanism in the occurrence and development of pathological myocardial hypertrophy from the following three points of view: 1) role of TRPV1 in alleviation of pathological myocardial hypertrophy; 2) role of TRPV1 in aggravation of pathological myocardial hypertrophy; and 3) the point of view of our team of researchers. It is expected that new therapies can provide potential targets for pathological myocardial hypertrophy.
Collapse
Affiliation(s)
- Qiqi Yan
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China.,Department of Cardiovascular Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Jun Tang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China.,Department of Cardiovascular Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Xin Zhang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China.,Department of Cardiovascular Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Liuyang Wu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China.,Department of Cardiovascular Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Yunyi Xu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China.,Department of Cardiovascular Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Lihong Wang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China.,Department of Cardiovascular Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
16
|
Manchanda M, Leishman E, Sangani K, Alamri A, Bradshaw HB. Activation of TRPV1 by Capsaicin or Heat Drives Changes in 2-Acyl Glycerols and N-Acyl Ethanolamines in a Time, Dose, and Temperature Dependent Manner. Front Cell Dev Biol 2021; 9:611952. [PMID: 33937226 PMCID: PMC8085603 DOI: 10.3389/fcell.2021.611952] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 02/24/2021] [Indexed: 12/25/2022] Open
Abstract
Endocannabinoids (eCBs) and transient receptor potential (TRP) channels are associated with thermoregulation; however, there are many gaps in the understanding of how these signaling systems work together in responding to changes in temperature. TRPV1, a calcium-permeable ion channel, is activated by capsaicin, elevated temperature, the eCB Anandamide, and over 15 additional endogenous lipids. There is also evidence for signaling crosstalk between TRPV1 and the eCB receptor, CB1. We recently found that activation of TRPV1-HEK cells by capsaicin increases the production of the eCB, 2-arachidonoyl glycerol (2-AG), suggesting a molecular link between these receptors. Here, we tested the hypothesis that TRPV1 activation by capsaicin drives regulation of a wider-range of lipid signaling molecules and is time and dose-dependent. We also tested the hypothesis that changes in temperature that drive changes in calcium mobilization in TRPV1-HEK will likewise drive similar changes in lipid signaling molecule regulation. Lipid analysis was conducted by partial purification of methanolic extracts on C18 solid phase extraction columns followed by HPLC/MS/MS. Capsaicin increased the release of 2-acyl glycerols (2-AG, 2-linoleoyl glycerol, 2-oleoyl glycerol), in a concentration- and time-dependent manner, whereas levels of N-acyl ethanolamines (NAEs), including Anandamide, were significantly decreased. Analogous changes in 2-acyl glycerols and NAEs were measured upon ramping the temperature from 37 to 45°C. In contrast, opposite effects were measured when analyzing lipids after they were maintained at 27°C and then quickly ramped to 37°C, wherein 2-acyl glycerol levels decreased and NAEs increased. These results provide further evidence that the eCB system and TRPV1 have integrated signaling functions that are associated with the molecular response to temperature variation.
Collapse
Affiliation(s)
- Meera Manchanda
- Bradshaw Laboratory of Lipid Neuroscience, Indiana University Bloomington, Bloomington, IN, United States
| | - Emma Leishman
- Bradshaw Laboratory of Lipid Neuroscience, Indiana University Bloomington, Bloomington, IN, United States
| | - Kishan Sangani
- Bradshaw Laboratory of Lipid Neuroscience, Indiana University Bloomington, Bloomington, IN, United States
| | - Ali Alamri
- Bradshaw Laboratory of Lipid Neuroscience, Indiana University Bloomington, Bloomington, IN, United States
| | - Heather B Bradshaw
- Bradshaw Laboratory of Lipid Neuroscience, Indiana University Bloomington, Bloomington, IN, United States
| |
Collapse
|
17
|
Ren J, Cheng Y, Wen X, Liu P, Zhao F, Xin F, Wang M, Huang H, Wang W. BK Ca channel participates in insulin-induced lipid deposition in adipocytes by increasing intracellular calcium. J Cell Physiol 2021; 236:5818-5831. [PMID: 33432604 DOI: 10.1002/jcp.30266] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 12/22/2020] [Accepted: 12/24/2020] [Indexed: 12/21/2022]
Abstract
Storing energy in the form of triglyceride (TG) is one of the basic functions of adipose tissue. Large-conductance calcium-activated potassium channels (BKCa channels) are expressed in adipose tissue and adipocyte-specific BKCa deficiency resists obesity in mice, but the role of BKCa channels in lipid deposition and the underlying mechanisms have not been elucidated. In the present study, we generated BKCa knockout (KO) rats and performed a transcriptome analysis of adipose tissue. We found that the phosphoinositide 3-kinase (PI3K)-protein kinase B (Akt) signaling pathway, which is important for lipid deposition, exhibited the most notable reduction among various signaling pathways in BKCa KO rats compared to wild-type rats. Insulin-induced TG deposition, glucose uptake, and Akt (Ser473) phosphorylation were significantly reduced in cultured adipocytes differentiated from adipose-derived stem cells of BKCa KO rats. Furthermore, we found that the insulin-induced increase of intracellular calcium resulting from extracellular calcium influx was significantly impaired in BKCa KO adipocytes. Finally, insulin activated BKCa currents through PI3K, which was independent of Akt and intracellular calcium. The results of this study suggested that BKCa channels participate in the insulin signaling pathway and promote TG deposition by increasing extracellular calcium influx in adipocytes.
Collapse
Affiliation(s)
- Jie Ren
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yuan Cheng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xinxin Wen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Ping Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Beijing Lab for Cardiovascular Precision Medicine, Beijing, China
| | - Feng Zhao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Fang Xin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Beijing Lab for Cardiovascular Precision Medicine, Beijing, China
| | - Meili Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing, China
| | - Haixia Huang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Beijing Lab for Cardiovascular Precision Medicine, Beijing, China.,Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing, China
| | - Wei Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Beijing Lab for Cardiovascular Precision Medicine, Beijing, China
| |
Collapse
|
18
|
Nagpal R, Mishra SK, Deep G, Yadav H. Role of TRP Channels in Shaping the Gut Microbiome. Pathogens 2020; 9:pathogens9090753. [PMID: 32947778 PMCID: PMC7559121 DOI: 10.3390/pathogens9090753] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 08/29/2020] [Accepted: 09/09/2020] [Indexed: 12/13/2022] Open
Abstract
Transient receptor potential (TRP) channel family proteins are sensors for pain, which sense a variety of thermal and noxious chemicals. Sensory neurons innervating the gut abundantly express TRPA1 and TRPV1 channels and are in close proximity of gut microbes. Emerging evidence indicates a bi-directional gut–brain cross-talk in several entero-neuronal pathologies; however, the direct evidence of TRP channels interacting with gut microbial populations is lacking. Herein, we examine whether and how the knockout (KO) of TRPA1 and TRPV1 channels individually or combined TRPA1/V1 double-knockout (dKO) impacts the gut microbiome in mice. We detect distinct microbiome clusters among the three KO mouse models versus wild-type (WT) mice. All three TRP-KO models have reduced microbial diversity, harbor higher abundance of Bacteroidetes, and a reduced proportion of Firmicutes. Specifically distinct arrays in the KO models are determined mainly by S24-7, Bacteroidaceae, Clostridiales, Prevotellaceae, Helicobacteriaceae, Rikenellaceae, and Ruminococcaceae. A1KO mice have lower Prevotella, Desulfovibrio, Bacteroides, Helicobacter and higher Rikenellaceae and Tenericutes; V1KO mice demonstrate higher Ruminococcaceae, Lachnospiraceae, Ruminococcus, Desulfovibrio and Mucispirillum; and A1V1dKO mice exhibit higher Bacteroidetes, Bacteroides and S24-7 and lower Firmicutes, Ruminococcaceae, Oscillospira, Lactobacillus and Sutterella abundance. Furthermore, the abundance of taxa involved in biosynthesis of lipids and primary and secondary bile acids is higher while that of fatty acid biosynthesis-associated taxa is lower in all KO groups. To our knowledge, this is the first study demonstrating distinct gut microbiome signatures in TRPA1, V1 and dKO models and should facilitate prospective studies exploring novel diagnostic/ therapeutic modalities regarding the pathophysiology of TRP channel proteins.
Collapse
Affiliation(s)
- Ravinder Nagpal
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA;
| | - Santosh Kumar Mishra
- Department of Molecular Biomedical Sciences, NC State Veterinary Medicine, Raleigh, NC 27606, USA;
| | - Gagan Deep
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA;
| | - Hariom Yadav
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA;
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
- Correspondence: ; Tel.: +1-336-713-5049
| |
Collapse
|
19
|
The Lipid Receptor G2A (GPR132) Mediates Macrophage Migration in Nerve Injury-Induced Neuropathic Pain. Cells 2020; 9:cells9071740. [PMID: 32708184 PMCID: PMC7409160 DOI: 10.3390/cells9071740] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/15/2020] [Accepted: 07/18/2020] [Indexed: 12/17/2022] Open
Abstract
Nerve injury-induced neuropathic pain is difficult to treat and mechanistically characterized by strong neuroimmune interactions, involving signaling lipids that act via specific G-protein coupled receptors. Here, we investigated the role of the signaling lipid receptor G2A (GPR132) in nerve injury-induced neuropathic pain using the robust spared nerve injury (SNI) mouse model. We found that the concentrations of the G2A agonist 9-HODE (9-Hydroxyoctadecadienoic acid) are strongly increased at the site of nerve injury during neuropathic pain. Moreover, G2A-deficient mice show a strong reduction of mechanical hypersensitivity after nerve injury. This phenotype is accompanied by a massive reduction of invading macrophages and neutrophils in G2A-deficient mice and a strongly reduced release of the proalgesic mediators TNFα, IL-6 and VEGF at the site of injury. Using a global proteome analysis to identify the underlying signaling pathways, we found that G2A activation in macrophages initiates MyD88-PI3K-AKT signaling and transient MMP9 release to trigger cytoskeleton remodeling and migration. We conclude that G2A-deficiency reduces inflammatory responses by decreasing the number of immune cells and the release of proinflammatory cytokines and growth factors at the site of nerve injury. Inhibiting the G2A receptor after nerve injury may reduce immune cell-mediated peripheral sensitization and may thus ameliorate neuropathic pain.
Collapse
|
20
|
Doolen S, Keyes GS, Ramsden CE. Hydroxy-epoxide and keto-epoxide derivatives of linoleic acid activate trigeminal neurons. NEUROBIOLOGY OF PAIN 2020; 7:100046. [PMID: 32478201 PMCID: PMC7248286 DOI: 10.1016/j.ynpai.2020.100046] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 04/21/2020] [Accepted: 04/23/2020] [Indexed: 02/08/2023]
Abstract
11-hydroxy- and 11-keto-epoxide-LA derivatives elicit Ca2+ transients in trigeminal neuron subpopulations. 11H-12,13E-LA, 11 K-12,13E-LA, and 11H-9,10E-LA produce Ca2+ responses in higher proportions of neurons than linoleic acid or 9-HODE. 11-hydroxy-epoxide- and 11-keto-epoxide derivatives of linoleic acid potentially contribute to nociception.
Endogenous lipid mediators are proposed to contribute to headache and facial pain by activating trigeminal neurons (TN). We recently identified 11-hydroxy-epoxide- and 11-keto-epoxide derivatives of linoleic acid (LA) that are present in human skin and plasma and potentially contribute to nociception. Here we expand upon initial findings by examining the effects of 11-hydroxy- and 11-keto-epoxide-LA derivatives on TN activation in comparison to LA, the LA derivative [9-hydroxy-octadecadienoic acid (9-HODE)] and prostaglandin E2 (PGE2). 11-hydroxy- and 11-keto-epoxide-LA derivatives elicited Ca2+ transients in TN subpopulations. The proportion of neurons responding to test compounds (5 μM, 5 min) ranged from 16.2 ± 3.8 cells (11 K-9,10E-LA) to 34.1 ± 2.4 cells (11H-12,13E-LA). LA and 9-HODE (5 μM, 5 min) elicited responses in 11.6 ± 3.1% and 9.7 ± 3.4% of neurons, respectively. 11H-12,13E-LA, 11K-12,13E-LA, and 11H-9,10E-LA produced Ca2+ responses in significantly higher proportions of neurons compared to either LA or 9-HODE (F (6, 36) = 5.12, P = 0.0007). 11H-12,13E-LA and 11H-9,10E-LA increased proportions of responsive neurons in a concentration-dependent fashion, similar to PGE2. Most sensitive neurons responded to additional algesic agents (32.9% to capsaicin, 40.1% to PGE2, 58.0% to AITC), however 20.6% did not respond to any other agent. In summary, 11-hydroxy-epoxide derivatives of LA increase trigeminal neuron excitability, suggesting a potential role in headache or facial pain.
Collapse
Key Words
- 11-HEL, 11-hydroxy-epoxide-linoleic acid
- 11H-12,13E-LA, 11-hydroxy-12,13-trans-epoxy-(9Z)-octadecenoate
- 11H-9,10E-LA, 11-hydroxy-9,10-trans-epoxy-(12Z)-octadecenoate
- 9-HODE, 9-hydroxy-octadecadienoic acid
- CGRP, calcitonin gene related peptide
- DiHOMEs, dihydroxy-octadecenoic acids
- EpOMEs, epoxy-octadecenoic
- HODEs, octadecadienoic acids
- HpODEs, hydroperoxy-octadecadienoic acids
- Hyperalgesia
- LA, linoleic acid
- Linoleic acid
- Oxylipin
- PGE2, prostaglandin E2
- Pain
- Peroxidation
- TN, trigeminal neuron
- aCSF, artificial cerebrospinal fluid
Collapse
Affiliation(s)
- Suzanne Doolen
- Department of Physiology, University of Kentucky, 800 Rose Street, Lexington, KY 40536-0298, United States
| | - Gregory S Keyes
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health (NIH), Baltimore, MD 21224, USA
| | - Christopher E Ramsden
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health (NIH), Baltimore, MD 21224, USA.,Intramural Program of the National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20814, USA
| |
Collapse
|
21
|
Fontaine D, Figiel S, Félix R, Kouba S, Fromont G, Mahéo K, Potier-Cartereau M, Chantôme A, Vandier C. Roles of endogenous ether lipids and associated PUFAs in the regulation of ion channels and their relevance for disease. J Lipid Res 2020; 61:840-858. [PMID: 32265321 PMCID: PMC7269763 DOI: 10.1194/jlr.ra120000634] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/29/2020] [Indexed: 12/16/2022] Open
Abstract
Ether lipids (ELs) are lipids characterized by the presence of either an ether linkage (alkyl lipids) or a vinyl ether linkage [i.e., plasmalogens (Pls)] at the sn1 position of the glycerol backbone, and they are enriched in PUFAs at the sn2 position. In this review, we highlight that ELs have various biological functions, act as a reservoir for second messengers (such as PUFAs) and have roles in many diseases. Some of the biological effects of ELs may be associated with their ability to regulate ion channels that control excitation-contraction/secretion/mobility coupling and therefore cell physiology. These channels are embedded in lipid membranes, and lipids can regulate their activities directly or indirectly as second messengers or by incorporating into membranes. Interestingly, ELs and EL-derived PUFAs have been reported to play a key role in several pathologies, including neurological disorders, cardiovascular diseases, and cancers. Investigations leading to a better understanding of their mechanisms of action in pathologies have opened a new field in cancer research. In summary, newly identified lipid regulators of ion channels, such as ELs and PUFAs, may represent valuable targets to improve disease diagnosis and advance the development of new therapeutic strategies for managing a range of diseases and conditions.
Collapse
Affiliation(s)
- Delphine Fontaine
- Inserm N2C UMR1069, Université de Tours, F-37032 Tours CEDEX 1, France
| | - Sandy Figiel
- Inserm N2C UMR1069, Université de Tours, F-37032 Tours CEDEX 1, France
| | - Romain Félix
- Inserm N2C UMR1069, Université de Tours, F-37032 Tours CEDEX 1, France
| | - Sana Kouba
- Inserm N2C UMR1069, Université de Tours, F-37032 Tours CEDEX 1, France
| | - Gaëlle Fromont
- Inserm N2C UMR1069, Université de Tours, F-37032 Tours CEDEX 1, France; Department of Pathology, CHRU Bretonneau, F-37044 Tours CEDEX 9, France
| | - Karine Mahéo
- Inserm N2C UMR1069, Université de Tours, F-37032 Tours CEDEX 1, France; Faculté de Pharmacie, Université de Tours, F-37200 Tours, France
| | | | - Aurélie Chantôme
- Inserm N2C UMR1069, Université de Tours, F-37032 Tours CEDEX 1, France; Faculté de Pharmacie, Université de Tours, F-37200 Tours, France
| | - Christophe Vandier
- Inserm N2C UMR1069, Université de Tours, F-37032 Tours CEDEX 1, France. mailto:
| |
Collapse
|
22
|
Undem BJ, Sun H. Molecular/Ionic Basis of Vagal Bronchopulmonary C-Fiber Activation by Inflammatory Mediators. Physiology (Bethesda) 2020; 35:57-68. [PMID: 31799905 PMCID: PMC6985783 DOI: 10.1152/physiol.00014.2019] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 05/30/2019] [Accepted: 06/03/2019] [Indexed: 12/11/2022] Open
Abstract
Stimulation of bronchopulmonary vagal afferent C fibers by inflammatory mediators can lead to coughing, chest tightness, and changes in breathing pattern, as well as reflex bronchoconstriction and secretions. These responses serve a defensive function in healthy lungs but likely contribute to many of the signs and symptoms of inflammatory airway diseases. A better understanding of the mechanisms underlying the activation of bronchopulmonary C-fiber terminals may lead to novel therapeutics that would work in an additive or synergic manner with existing anti-inflammatory strategies.
Collapse
Affiliation(s)
| | - Hui Sun
- Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
23
|
Osthues T, Sisignano M. Oxidized Lipids in Persistent Pain States. Front Pharmacol 2019; 10:1147. [PMID: 31680947 PMCID: PMC6803483 DOI: 10.3389/fphar.2019.01147] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 09/05/2019] [Indexed: 12/13/2022] Open
Abstract
Chemotherapy, nerve injuries, or diseases like multiple sclerosis can cause pathophysiological processes of persistent and neuropathic pain. Thereby, the activation threshold of ion channels is reduced in peripheral sensory neurons to normally noxious stimuli like heat, cold, acid, or mechanical due to sensitization processes. This leads to enhanced neuronal activity, which can result in mechanical allodynia, cold allodynia, thermal hyperalgesia, spontaneous pain, and may initiate persistent and neuropathic pain. The treatment options for persistent and neuropathic pain patients are limited; for about 50% of them, current medication is not efficient due to severe side effects or low response to the treatment. Therefore, it is of special interest to find additional treatment strategies. One approach is the control of neuronal sensitization processes. Herein, signaling lipids are crucial mediators and play an important role during the onset and maintenance of pain. As preclinical studies demonstrate, lipids may act as endogenous ligands or may sensitize transient receptor potential (TRP)-channels. Likewise, they can cause enhanced activity of sensory neurons by mechanisms involving G-protein coupled receptors and activation of intracellular protein kinases. In this regard, oxidized metabolites of the essential fatty acid linoleic acid, 9- and 13-hydroxyoctadecadienoic acid (HODE), their dihydroxy-metabolites (DiHOMEs), as well as epoxides of linoleic acid (EpOMEs) and of arachidonic acid (EETs), as well as lysophospholipids, sphingolipids, and specialized pro-resolving mediators (SPMs) have been reported to play distinct roles in pain transmission or inhibition. Here, we discuss the underlying molecular mechanisms of the oxidized linoleic acid metabolites and eicosanoids. Furthermore, we critically evaluate their role as potential targets for the development of novel analgesics and for the treatment of persistent or neuropathic pain.
Collapse
Affiliation(s)
- Tabea Osthues
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Branch for Translational Medicine and Pharmacology TMP, Frankfurt, Germany
| | - Marco Sisignano
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt/ZAFES, University Hospital, Goethe-University, Frankfurt, Germany
| |
Collapse
|
24
|
Patil MJ, Sun H, Ru F, Meeker S, Undem BJ. Targeting C-fibers for peripheral acting anti-tussive drugs. Pulm Pharmacol Ther 2019; 56:15-19. [PMID: 30872160 DOI: 10.1016/j.pupt.2019.03.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 03/01/2019] [Accepted: 03/06/2019] [Indexed: 01/09/2023]
Abstract
Activation of vagal C-fibers is likely involved in some types of pathological coughing, especially coughing that is associated with airway inflammation. This is because stimulation of vagal C-fibers leads to strong urge to cough sensations, and because C-fiber terminals can be strongly activated by mediators associated with airway inflammation. The most direct manner in which a given mediator can activate a C-fiber terminal is through interacting with its receptor expressed in the terminal membrane. The agonist-receptor interaction then must lead to the opening (or potentially closing) of ion channels that lead to a membrane depolarization. This depolarization is referred to as a generator potential. If, and only if, the generator potential reaches the voltage necessary to activate voltage-gated sodium channels, action potentials are initiated and conducted to the central terminals within the CNS. Therefore, there are three target areas to block the inflammatory mediator induced activation of C-fiber terminals. First, at the level of the mediator-receptor interaction, secondly at the level of the generator potential, and third at the level of the voltage-gated sodium channels. Here we provide a brief overview of each of these therapeutic strategies.
Collapse
Affiliation(s)
- Mayur J Patil
- Department of Medicine, Johns Hopkins University School of Medicine, USA
| | - Hui Sun
- Department of Medicine, Johns Hopkins University School of Medicine, USA
| | - Fei Ru
- Department of Medicine, Johns Hopkins University School of Medicine, USA
| | - Sonya Meeker
- Department of Medicine, Johns Hopkins University School of Medicine, USA
| | - Bradley J Undem
- Department of Medicine, Johns Hopkins University School of Medicine, USA.
| |
Collapse
|
25
|
Miller JL, Blaszkiewicz M, Beaton C, Johnson CP, Waible S, Dubois AL, Klemmer A, Kiebish M, Townsend KL. A peroxidized omega-3-enriched polyunsaturated diet leads to adipose and metabolic dysfunction. J Nutr Biochem 2018; 64:50-60. [PMID: 30439568 DOI: 10.1016/j.jnutbio.2018.10.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 10/16/2018] [Accepted: 10/17/2018] [Indexed: 12/14/2022]
Abstract
Consumption of diets that differ in fat type and amount, and sequestration of various fatty acids to tissues and organs likely have effects on overall physiology and metabolic health. However, the contributions of dietary lipids to brain-adipose communication and adipose tissue function are poorly understood. We designed six custom diets that differed only in amount and type of dietary fat, with high or low levels of saturated fatty acids (SFA), omega-6 polyunsaturated fatty acids (n-6 PUFA) or omega-3 (n-3) PUFA. Mice fed the n-3 PUFA diet for 16 weeks displayed a striking reduction in weight gain accompanied by smaller adipose depots and improved glucose sensitivity. Reduced body weight occurred despite lowered energy expenditure and no difference in food intake. Despite the apparent beneficial effects to whole body physiology, we have demonstrated for the first time that a peroxidized n-3-enriched diet led to lipotoxicity of white adipose tissue, as evidenced by increased fibrosis, lipofuscin, reduced anti-inflammatory markers and loss of proper nerve supply. While healthful, n-3 fats are prone to peroxidation, and we observed peroxidated lipid metabolites in the adipose tissue of mice on these diets. Furthermore, using a lipidomics approach, we have observed that brain, white adipose tissue and brown adipose tissue accumulate lipid metabolites differently. The brain remained mostly shielded from changes in dietary fat type and amount, but differences in adipose lipid metabolites across these six diets may have affected metabolic function and brain-adipose communication, as observed in this study.
Collapse
Affiliation(s)
- James L Miller
- School of Biology and Ecology, University of Maine, Orono, ME
| | | | - Cordell Beaton
- School of Biology and Ecology, University of Maine, Orono, ME
| | - Cory P Johnson
- School of Biology and Ecology, University of Maine, Orono, ME
| | - Stephen Waible
- School of Biology and Ecology, University of Maine, Orono, ME
| | - Amanda L Dubois
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME
| | - Amanda Klemmer
- School of Biology and Ecology, University of Maine, Orono, ME
| | | | - Kristy L Townsend
- School of Biology and Ecology, University of Maine, Orono, ME; Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME.
| |
Collapse
|
26
|
Sarhan M, Land WG, Tonnus W, Hugo CP, Linkermann A. Origin and Consequences of Necroinflammation. Physiol Rev 2018; 98:727-780. [PMID: 29465288 DOI: 10.1152/physrev.00041.2016] [Citation(s) in RCA: 134] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
When cells undergo necrotic cell death in either physiological or pathophysiological settings in vivo, they release highly immunogenic intracellular molecules and organelles into the interstitium and thereby represent the strongest known trigger of the immune system. With our increasing understanding of necrosis as a regulated and genetically determined process (RN, regulated necrosis), necrosis and necroinflammation can be pharmacologically prevented. This review discusses our current knowledge about signaling pathways of necrotic cell death as the origin of necroinflammation. Multiple pathways of RN such as necroptosis, ferroptosis, and pyroptosis have been evolutionary conserved most likely because of their differences in immunogenicity. As the consequence of necrosis, however, all necrotic cells release damage associated molecular patterns (DAMPs) that have been extensively investigated over the last two decades. Analysis of necroinflammation allows characterizing specific signatures for each particular pathway of cell death. While all RN-pathways share the release of DAMPs in general, most of them actively regulate the immune system by the additional expression and/or maturation of either pro- or anti-inflammatory cytokines/chemokines. In addition, DAMPs have been demonstrated to modulate the process of regeneration. For the purpose of better understanding of necroinflammation, we introduce a novel classification of DAMPs in this review to help detect the relative contribution of each RN-pathway to certain physiological and pathophysiological conditions.
Collapse
Affiliation(s)
- Maysa Sarhan
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna , Vienna , Austria ; INSERM UMR_S 1109, Laboratory of Excellence Transplantex, University of Strasbourg , Strasbourg , France ; German Academy of Transplantation Medicine, Munich , Germany ; and Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden , Dresden , Germany
| | - Walter G Land
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna , Vienna , Austria ; INSERM UMR_S 1109, Laboratory of Excellence Transplantex, University of Strasbourg , Strasbourg , France ; German Academy of Transplantation Medicine, Munich , Germany ; and Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden , Dresden , Germany
| | - Wulf Tonnus
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna , Vienna , Austria ; INSERM UMR_S 1109, Laboratory of Excellence Transplantex, University of Strasbourg , Strasbourg , France ; German Academy of Transplantation Medicine, Munich , Germany ; and Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden , Dresden , Germany
| | - Christian P Hugo
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna , Vienna , Austria ; INSERM UMR_S 1109, Laboratory of Excellence Transplantex, University of Strasbourg , Strasbourg , France ; German Academy of Transplantation Medicine, Munich , Germany ; and Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden , Dresden , Germany
| | - Andreas Linkermann
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna , Vienna , Austria ; INSERM UMR_S 1109, Laboratory of Excellence Transplantex, University of Strasbourg , Strasbourg , France ; German Academy of Transplantation Medicine, Munich , Germany ; and Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden , Dresden , Germany
| |
Collapse
|
27
|
Tarighi N, Menger D, Pierre S, Kornstädt L, Thomas D, Ferreirós N, Nüsing RM, Geisslinger G, Scholich K. Thromboxane-Induced α-CGRP Release from Peripheral Neurons Is an Essential Positive Feedback Loop in Capsaicin-Induced Neurogenic Inflammation. J Invest Dermatol 2018; 139:656-664. [PMID: 30612974 DOI: 10.1016/j.jid.2018.10.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 10/17/2018] [Accepted: 10/22/2018] [Indexed: 11/30/2022]
Abstract
α-CGRP is synthesized by sensory nerves in the dermis and its release can cause vasodilation and local inflammation. Its vasorelaxant effects are based on the direct activation of smooth muscle and endothelial cells, as well as the activation of mast cells causing the release of vasoactive and proinflammatory mediators. Here, we show that in the capsaicin model for neurogenic inflammation, capsaicin-induced edema formation is mediated by α-CGRP and mast cells, but is absent in thromboxane receptor-deficient mice. Capsaicin treatment of mice induced a thromboxane synthesis, which was mediated by α-CGRP and mast cells. Fittingly, α-CGRP induced thromboxane synthesis in mast cells and the thromboxane receptor agonist I-BOP caused edema formation independently of mast cells, suggesting that mast cells are the source of thromboxane. Most importantly, I-BOP-induced edema formation was mediated by α-CGRP and I-BOP was able to stimulate through calcineurin the α-CGRP release from peripheral neurons. Likewise, the signaling pathway, including α-CGRP, thromboxane receptor, and mast cells, also mediated capsaicin-induced mechanical hypersensitivity, a common symptom of capsaicin treatment. Taken together, the thromboxane-induced α-CGRP release from neurons forms a positive feedback loop causing prolonged α-CGRP release and edema formation during capsaicin-induced neurogenic inflammation.
Collapse
Affiliation(s)
- Neda Tarighi
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt, University Hospital Frankfurt, Germany
| | - Dominic Menger
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt, University Hospital Frankfurt, Germany
| | - Sandra Pierre
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt, University Hospital Frankfurt, Germany
| | - Lisa Kornstädt
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt, University Hospital Frankfurt, Germany
| | - Dominique Thomas
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt, University Hospital Frankfurt, Germany
| | - Nerea Ferreirós
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt, University Hospital Frankfurt, Germany
| | - Rolf M Nüsing
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt, University Hospital Frankfurt, Germany
| | - Gerd Geisslinger
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt, University Hospital Frankfurt, Germany; Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine and Pharmacology, Frankfurt/Main, Germany
| | - Klaus Scholich
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt, University Hospital Frankfurt, Germany; Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine and Pharmacology, Frankfurt/Main, Germany.
| |
Collapse
|
28
|
Kern K, Schäfer SMG, Cohnen J, Pierre S, Osthues T, Tarighi N, Hohmann S, Ferreiros N, Brüne B, Weigert A, Geisslinger G, Sisignano M, Scholich K. The G2A Receptor Controls Polarization of Macrophage by Determining Their Localization Within the Inflamed Tissue. Front Immunol 2018; 9:2261. [PMID: 30327654 PMCID: PMC6174245 DOI: 10.3389/fimmu.2018.02261] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 09/11/2018] [Indexed: 12/17/2022] Open
Abstract
Macrophages are highly versatile cells, which acquire, depending on their microenvironment, pro- (M1-like), or antiinflammatory (M2-like) phenotypes. Here, we studied the role of the G-protein coupled receptor G2A (GPR132), in chemotactic migration and polarization of macrophages, using the zymosan-model of acute inflammation. G2A-deficient mice showed a reduced zymosan-induced thermal hyperalgesia, which was reversed after macrophage depletion. Fittingly, the number of M1-like macrophages was reduced in the inflamed tissue in G2A-deficient mice. However, G2A activation was not sufficient to promote M1-polarization in bone marrow-derived macrophages. While the number of monocyte-derived macrophages in the inflamed paw was not altered, G2A-deficient mice had less macrophages in the direct vicinity of the origin of inflammation, an area marked by the presence of zymosan, neutrophil accumulation and proinflammatory cytokines. Fittingly neutrophil efferocytosis was decreased in G2A-deficient mice and several lipids, which are released by neutrophils and promote G2A-mediated chemotaxis, were increased in the inflamed tissue. Taken together, G2A is necessary to position macrophages in the proinflammatory microenvironment surrounding the center of inflammation. In absence of G2A the macrophages are localized in an antiinflammatory microenvironment and macrophage polarization is shifted toward M2-like macrophages.
Collapse
Affiliation(s)
- Katharina Kern
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt, University Hospital Frankfurt, Frankfurt, Germany
| | - Stephan M G Schäfer
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt, University Hospital Frankfurt, Frankfurt, Germany
| | - Jennifer Cohnen
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt, University Hospital Frankfurt, Frankfurt, Germany
| | - Sandra Pierre
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt, University Hospital Frankfurt, Frankfurt, Germany
| | - Tabea Osthues
- Project Group Translational Medicine and Pharmacology, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Frankfurt, Germany
| | - Neda Tarighi
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt, University Hospital Frankfurt, Frankfurt, Germany
| | - Stefan Hohmann
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt, University Hospital Frankfurt, Frankfurt, Germany
| | - Nerea Ferreiros
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt, University Hospital Frankfurt, Frankfurt, Germany
| | - Bernhard Brüne
- Project Group Translational Medicine and Pharmacology, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Frankfurt, Germany.,Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany
| | - Andreas Weigert
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany
| | - Gerd Geisslinger
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt, University Hospital Frankfurt, Frankfurt, Germany.,Project Group Translational Medicine and Pharmacology, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Frankfurt, Germany
| | - Marco Sisignano
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt, University Hospital Frankfurt, Frankfurt, Germany
| | - Klaus Scholich
- Institute of Clinical Pharmacology, Pharmazentrum Frankfurt, University Hospital Frankfurt, Frankfurt, Germany.,Project Group Translational Medicine and Pharmacology, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Frankfurt, Germany
| |
Collapse
|
29
|
Alharthi N, Christensen P, Hourani W, Ortori C, Barrett DA, Bennett AJ, Chapman V, Alexander SPH. n-3 polyunsaturated N-acylethanolamines are CB 2 cannabinoid receptor-preferring endocannabinoids. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1863:1433-1440. [PMID: 30591150 DOI: 10.1016/j.bbalip.2018.08.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 07/18/2018] [Accepted: 08/04/2018] [Indexed: 12/16/2022]
Abstract
Anandamide, the first identified endogenous cannabinoid and TRPV1 agonist, is one of a series of endogenous N-acylethanolamines, NAEs. We have generated novel assays to quantify the levels of multiple NAEs in biological tissues and their rates of hydrolysis through fatty acid amide hydrolase. This range of NAEs was also tested in rapid response assays of CB1, CB2 cannabinoid and TRPV1 receptors. The data indicate that PEA, SEA and OEA are not endocannabinoids or endovanilloids, and that the higher endogenous levels of these metabolites compared to polyunsaturated analogues are a correlate of their slow rates of hydrolysis. The n-6 NAEs (AEA, docosatetraenoyl and docosapentaenoyl derivatives) activated both CB1 and CB2 receptors, as well as TRPV1 channels, suggesting them to be 'genuine' endocannabinoids and 'endovanilloids'. The n-3 NAEs (eicosapentaenoyl, docosapentaenoyl and docosahexaenoyl derivatives) activated CB2 receptors and some n-3 NAEs (docosapentaenoyl and docosahexaenoyl derivatives) also activated TRPV1 channels, but failed to activate the CB1 receptor. We hypothesise that the preferential activation of CB2 receptors by n-3 PUFA NAEs contributes, at least in some part, to their broad anti-inflammatory profile.
Collapse
Affiliation(s)
- Nahed Alharthi
- School of Life Sciences, University of Nottingham Medical School, Nottingham NG7 2UH, England, United Kingdom of Great Britain and Northern Ireland
| | - Peter Christensen
- School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, England, United Kingdom of Great Britain and Northern Ireland
| | - Wafa Hourani
- School of Life Sciences, University of Nottingham Medical School, Nottingham NG7 2UH, England, United Kingdom of Great Britain and Northern Ireland
| | - Catherine Ortori
- School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, England, United Kingdom of Great Britain and Northern Ireland
| | - David A Barrett
- School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, England, United Kingdom of Great Britain and Northern Ireland
| | - Andrew J Bennett
- School of Life Sciences, University of Nottingham Medical School, Nottingham NG7 2UH, England, United Kingdom of Great Britain and Northern Ireland
| | - Victoria Chapman
- School of Life Sciences, University of Nottingham Medical School, Nottingham NG7 2UH, England, United Kingdom of Great Britain and Northern Ireland
| | - Stephen P H Alexander
- School of Life Sciences, University of Nottingham Medical School, Nottingham NG7 2UH, England, United Kingdom of Great Britain and Northern Ireland.
| |
Collapse
|
30
|
Muallem S, Chung WY, Jha A, Ahuja M. Lipids at membrane contact sites: cell signaling and ion transport. EMBO Rep 2017; 18:1893-1904. [PMID: 29030479 DOI: 10.15252/embr.201744331] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 06/10/2017] [Accepted: 09/21/2017] [Indexed: 12/14/2022] Open
Abstract
Communication between organelles is essential to coordinate cellular functions and the cell's response to physiological and pathological stimuli. Organellar communication occurs at membrane contact sites (MCSs), where the endoplasmic reticulum (ER) membrane is tethered to cellular organelle membranes by specific tether proteins and where lipid transfer proteins and cell signaling proteins are located. MCSs have many cellular functions and are the sites of lipid and ion transfer between organelles and generation of second messengers. This review discusses several aspects of MCSs in the context of lipid transfer, formation of lipid domains, generation of Ca2+ and cAMP second messengers, and regulation of ion transporters by lipids.
Collapse
Affiliation(s)
- Shmuel Muallem
- Epithelial Signaling and Transport Section, National Institute of Dental and Craniofacial Research, Bethesda, MD, USA
| | - Woo Young Chung
- Epithelial Signaling and Transport Section, National Institute of Dental and Craniofacial Research, Bethesda, MD, USA
| | - Archana Jha
- Epithelial Signaling and Transport Section, National Institute of Dental and Craniofacial Research, Bethesda, MD, USA
| | - Malini Ahuja
- Epithelial Signaling and Transport Section, National Institute of Dental and Craniofacial Research, Bethesda, MD, USA
| |
Collapse
|
31
|
Lawrence KM, Jones RC, Jackson TR, Baylie RL, Abbott B, Bruhn-Olszewska B, Board TN, Locke IC, Richardson SM, Townsend PA. Chondroprotection by urocortin involves blockade of the mechanosensitive ion channel Piezo1. Sci Rep 2017; 7:5147. [PMID: 28698554 PMCID: PMC5505992 DOI: 10.1038/s41598-017-04367-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 05/11/2017] [Indexed: 12/26/2022] Open
Abstract
Osteoarthritis (OA) is characterised by progressive destruction of articular cartilage and chondrocyte cell death. Here, we show the expression of the endogenous peptide urocortin1 (Ucn1) and two receptor subtypes, CRF-R1 and CRF-R2, in primary human articular chondrocytes (AC) and demonstrate its role as an autocrine/paracrine pro-survival factor. This effect could only be removed using the CRF-R1 selective antagonist CP-154526, suggesting Ucn1 acts through CRF-R1 when promoting chondrocyte survival. This cell death was characterised by an increase in p53 expression, and cleavage of caspase 9 and 3. Antagonism of CRF-R1 with CP-154526 caused an accumulation of intracellular calcium (Ca2+) over time and cell death. These effects could be prevented with the non-selective cation channel blocker Gadolinium (Gd3+). Therefore, opening of a non-selective cation channel causes cell death and Ucn1 maintains this channel in a closed conformation. This channel was identified to be the mechanosensitive channel Piezo1. We go on to determine that this channel inhibition by Ucn1 is mediated initially by an increase in cyclic adenosine monophosphate (cAMP) and a subsequent inactivation of phospholipase A2 (PLA2), whose metabolites are known to modulate ion channels. Knowledge of these novel pathways may present opportunities for interventions that could abrogate the progression of OA.
Collapse
Affiliation(s)
- K M Lawrence
- Division of Cancer Sciences, Manchester Cancer Research Centre, Manchester Academic Health Sciences Centre, The University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK.
| | - R C Jones
- Division of Cancer Sciences, Manchester Cancer Research Centre, Manchester Academic Health Sciences Centre, The University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK
| | - T R Jackson
- Division of Cancer Sciences, Manchester Cancer Research Centre, Manchester Academic Health Sciences Centre, The University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK
| | - R L Baylie
- Division of Cardiovascular Sciences, Manchester Academic Health Sciences Centre, University of Manchester, M13 9NT, Manchester, UK
| | - B Abbott
- Division of Cancer Sciences, Manchester Cancer Research Centre, Manchester Academic Health Sciences Centre, The University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK
| | - B Bruhn-Olszewska
- Division of Cancer Sciences, Manchester Cancer Research Centre, Manchester Academic Health Sciences Centre, The University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK
| | - T N Board
- The Center for Hip Surgery, Wrightington Hospital, Wigan, WN6 9EP, UK
| | - I C Locke
- Department of Biomedical Sciences, University of Westminster, London, W1W 6UW, UK
| | - S M Richardson
- Division of Cell Matrix Biology and Regenerative Medicine, Centre for Tissue Injury and Repair, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, M13 9PT, UK
| | - P A Townsend
- Division of Cancer Sciences, Manchester Cancer Research Centre, Manchester Academic Health Sciences Centre, The University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK
| |
Collapse
|
32
|
Claeson AS, Gouveia-Figueira S, Häggström J, Fowler CJ, Nording ML. Levels of oxylipins, endocannabinoids and related lipids in plasma before and after low-level exposure to acrolein in healthy individuals and individuals with chemical intolerance. Prostaglandins Leukot Essent Fatty Acids 2017; 121:60-67. [PMID: 28651699 DOI: 10.1016/j.plefa.2017.06.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 06/05/2017] [Accepted: 06/12/2017] [Indexed: 11/25/2022]
Abstract
Oxylipins and endocannabinoids play important biological roles, including effects upon inflammation. It is not known whether the circulating levels of these lipids are affected by inhalation of the environmental pollutant acrolein. In the present study, we have investigated the consequences of low-level exposure to acrolein on oxylipin, endocannabinoid and related lipid levels in the plasma of healthy individuals and individuals with chemical intolerance (CI), an affliction with a suggested inflammatory origin. Participants were exposed twice (60min) to heptane and a mixture of heptane and acrolein. Blood samples were collected before exposure, after and 24h post-exposure. There were no overt effects of acrolein exposure on the oxylipin lipidome or endocannibinoids detectable in the bloodstream at the time points investigated. No relationship between basal levels or levels after exposure to acrolein and CI could be identified. This implicates a minor role of inflammatory mediators on the systemic level in CI.
Collapse
Affiliation(s)
| | | | - Jenny Häggström
- Umeå School of Business and Economics, Department of Statistics, Umeå University, Sweden
| | | | | |
Collapse
|
33
|
Ciardo MG, Ferrer-Montiel A. Lipids as central modulators of sensory TRP channels. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1859:1615-1628. [PMID: 28432033 DOI: 10.1016/j.bbamem.2017.04.012] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 04/13/2017] [Accepted: 04/15/2017] [Indexed: 12/13/2022]
Abstract
The transient receptor potential (TRP) ion channel family is involved in a diversity of physiological processes including sensory and homeostatic functions, as well as muscle contraction and vasomotor control. Their dysfunction contributes to the etiology of several diseases, being validated as therapeutic targets. These ion channels may be activated by physical or chemical stimuli and their function is highly influenced by signaling molecules activated by extracellular signals. Notably, as integral membrane proteins, lipid molecules also modulate their membrane location and function either by direct interaction with the channel structure or by modulating the physico-chemical properties of the cellular membrane. This lipid-based modulatory effect is being considered an alternative and promising approach to regulate TRP channel dysfunction in diseases. Here, we review the current progress in this exciting field highlighting a complex channel regulation by a large diversity of lipid molecules and suggesting some diseases that may benefit from a membrane lipid therapy. This article is part of a Special Issue entitled: Membrane Lipid Therapy: Drugs Targeting Biomembranes edited by Pablo V. Escribá.
Collapse
Affiliation(s)
| | - Antonio Ferrer-Montiel
- Instituto de Biología Molecular y Celular, Universidad Miguel Hernández, Av. De la Universidad s/n, Elche, Spain.
| |
Collapse
|
34
|
Hohmann SW, Angioni C, Tunaru S, Lee S, Woolf CJ, Offermanns S, Geisslinger G, Scholich K, Sisignano M. The G2A receptor (GPR132) contributes to oxaliplatin-induced mechanical pain hypersensitivity. Sci Rep 2017; 7:446. [PMID: 28348394 PMCID: PMC5428564 DOI: 10.1038/s41598-017-00591-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 03/03/2017] [Indexed: 12/18/2022] Open
Abstract
Chemotherapy-induced peripheral neuropathic pain (CIPN) is a common and severe debilitating side effect of many widely used cytostatics. However, there is no approved pharmacological treatment for CIPN available. Among other substances, oxaliplatin causes CIPN in up to 80% of treated patients. Here, we report the involvement of the G-protein coupled receptor G2A (GPR132) in oxaliplatin-induced neuropathic pain in mice. We found that mice deficient in the G2A-receptor show decreased mechanical hypersensitivity after oxaliplatin treatment. Lipid ligands of G2A were found in increased concentrations in the sciatic nerve and dorsal root ganglia of oxaliplatin treated mice. Calcium imaging and patch-clamp experiments show that G2A activation sensitizes the ligand-gated ion channel TRPV1 in sensory neurons via activation of PKC. Based on these findings, we conclude that targeting G2A may be a promising approach to reduce oxaliplatin-induced TRPV1-sensitization and the hyperexcitability of sensory neurons and thereby to reduce pain in patients treated with this chemotherapeutic agent.
Collapse
Affiliation(s)
- Stephan W Hohmann
- Institute of Clinical Pharmacology, pharmazentrum frankfurt/ZAFES, University Hospital, Goethe-University, D-60590, Frankfurt am Main, Germany
| | - Carlo Angioni
- Institute of Clinical Pharmacology, pharmazentrum frankfurt/ZAFES, University Hospital, Goethe-University, D-60590, Frankfurt am Main, Germany
| | - Sorin Tunaru
- Department of Pharmacology, Max-Planck-Institute for Heart and Lung Research, 61231, Bad Nauheim, Germany
| | - Seungkyu Lee
- F. M. Kirby Neurobiology Center, Children's Hospital Boston, and Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Clifford J Woolf
- F. M. Kirby Neurobiology Center, Children's Hospital Boston, and Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Stefan Offermanns
- Department of Pharmacology, Max-Planck-Institute for Heart and Lung Research, 61231, Bad Nauheim, Germany
| | - Gerd Geisslinger
- Institute of Clinical Pharmacology, pharmazentrum frankfurt/ZAFES, University Hospital, Goethe-University, D-60590, Frankfurt am Main, Germany.,Fraunhofer Institute for Molecular Biology and Applied Ecology - Project Group Translational Medicine and Pharmacology (IME-TMP), Frankfurt am Main, Germany
| | - Klaus Scholich
- Institute of Clinical Pharmacology, pharmazentrum frankfurt/ZAFES, University Hospital, Goethe-University, D-60590, Frankfurt am Main, Germany
| | - Marco Sisignano
- Institute of Clinical Pharmacology, pharmazentrum frankfurt/ZAFES, University Hospital, Goethe-University, D-60590, Frankfurt am Main, Germany.
| |
Collapse
|
35
|
Zinn S, Sisignano M, Kern K, Pierre S, Tunaru S, Jordan H, Suo J, Treutlein EM, Angioni C, Ferreiros N, Leffler A, DeBruin N, Offermanns S, Geisslinger G, Scholich K. The leukotriene B4 receptors BLT1 and BLT2 form an antagonistic sensitizing system in peripheral sensory neurons. J Biol Chem 2017; 292:6123-6134. [PMID: 28242764 DOI: 10.1074/jbc.m116.769125] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 02/09/2017] [Indexed: 11/06/2022] Open
Abstract
Sensitization of the heat-activated ion channel transient receptor potential vanilloid 1 (TRPV1) through lipids is a fundamental mechanism during inflammation-induced peripheral sensitization. Leukotriene B4 is a proinflammatory lipid mediator whose role in peripheral nociceptive sensitization is not well understood to date. Two major G-protein-coupled receptors for leukotriene B4 have been identified: the high-affinity receptor BLT1 and the low-affinity receptor BLT2. Transcriptional screening for the expression G-protein-coupled receptors in murine dorsal root ganglia showed that both receptors were among the highest expressed in dorsal root ganglia. Calcium imaging revealed a sensitization of TRPV1-mediated calcium increases in a relative narrow concentration range for leukotriene B4 (100-200 nm). Selective antagonists and neurons from knock-out mice demonstrated a BLT1-dependent sensitization of TRPV1-mediated calcium increases. Accordingly, leukotriene B4-induced thermal hyperalgesia was mediated through BLT1 and TRPV1 as shown using the respective knock-out mice. Importantly, higher leukotriene B4 concentrations (>0.5 μm) and BLT2 agonists abolished sensitization of the TRPV1-mediated calcium increases. Also, BLT2 activation inhibited protein kinase C- and protein kinase A-mediated sensitization processes through the phosphatase calcineurin. Consequently, a selective BLT2-receptor agonist increased thermal and mechanical withdrawal thresholds during zymosan-induced inflammation. In accordance with these data, immunohistochemical analysis showed that both leukotriene B4 receptors were expressed in peripheral sensory neurons. Thus, the data show that the two leukotriene B4 receptors have opposing roles in the sensitization of peripheral sensory neurons forming a self-restricting system.
Collapse
Affiliation(s)
- Sebastian Zinn
- From the Institut für Klinische Pharmakologie, Pharmazentrum Frankfurt, Klinikum der Goethe-Universität Frankfurt, 60590 Frankfurt, Germany
| | - Marco Sisignano
- From the Institut für Klinische Pharmakologie, Pharmazentrum Frankfurt, Klinikum der Goethe-Universität Frankfurt, 60590 Frankfurt, Germany
| | - Katharina Kern
- From the Institut für Klinische Pharmakologie, Pharmazentrum Frankfurt, Klinikum der Goethe-Universität Frankfurt, 60590 Frankfurt, Germany
| | - Sandra Pierre
- From the Institut für Klinische Pharmakologie, Pharmazentrum Frankfurt, Klinikum der Goethe-Universität Frankfurt, 60590 Frankfurt, Germany
| | - Sorin Tunaru
- the Department of Pharmacology, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Holger Jordan
- the Fraunhofer Institute of Molecular Biology and Applied Ecology, Project Group Translational Medicine and Pharmacology, 60590 Frankfurt, Germany, and
| | - Jing Suo
- From the Institut für Klinische Pharmakologie, Pharmazentrum Frankfurt, Klinikum der Goethe-Universität Frankfurt, 60590 Frankfurt, Germany
| | - Elsa-Marie Treutlein
- From the Institut für Klinische Pharmakologie, Pharmazentrum Frankfurt, Klinikum der Goethe-Universität Frankfurt, 60590 Frankfurt, Germany
| | - Carlo Angioni
- From the Institut für Klinische Pharmakologie, Pharmazentrum Frankfurt, Klinikum der Goethe-Universität Frankfurt, 60590 Frankfurt, Germany
| | - Nerea Ferreiros
- From the Institut für Klinische Pharmakologie, Pharmazentrum Frankfurt, Klinikum der Goethe-Universität Frankfurt, 60590 Frankfurt, Germany
| | - Andreas Leffler
- the Department for Anaesthesiology and Critical Care Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Natasja DeBruin
- the Fraunhofer Institute of Molecular Biology and Applied Ecology, Project Group Translational Medicine and Pharmacology, 60590 Frankfurt, Germany, and
| | - Stefan Offermanns
- the Department of Pharmacology, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Gerd Geisslinger
- From the Institut für Klinische Pharmakologie, Pharmazentrum Frankfurt, Klinikum der Goethe-Universität Frankfurt, 60590 Frankfurt, Germany.,the Fraunhofer Institute of Molecular Biology and Applied Ecology, Project Group Translational Medicine and Pharmacology, 60590 Frankfurt, Germany, and
| | - Klaus Scholich
- From the Institut für Klinische Pharmakologie, Pharmazentrum Frankfurt, Klinikum der Goethe-Universität Frankfurt, 60590 Frankfurt, Germany,
| |
Collapse
|
36
|
Abstract
Among the clinically relevant pain conditions, pain in the musculoskeletal system is most frequent. This article reports extensive epidemiological data on musculoskeletal system pain in Germany and worldwide. Since back pain is most frequent, the diagnostics and therapeutic algorithms of acute, recurring, and chronic lower back pain in Germany will be particularly addressed. The importance of the physiologic-organic, the cognitive-emotional, the behavioral, and the social level to diagnostics and treatment will be discussed. We will also focus on osteoarthritic pain and address its epidemiology, clinical importance, and significance for the health care system. This article will list some reasons why the musculoskeletal system in particular is frequently the site of chronic pain. The authors believe that these reasons are to be sought in the complex structures of the musculoskeletal system; in the particular sensitivity of the deep somatic nociceptive system for long-term sensitization processes, as well as the ensuing nervous system reactions; and in the interactions between the nervous and immune systems. The article will give some insights into the research carried out on this topic in Germany.
Collapse
|
37
|
Kumar R, Hazan A, Geron M, Steinberg R, Livni L, Matzner H, Priel A. Activation of transient receptor potential vanilloid 1 by lipoxygenase metabolites depends on PKC phosphorylation. FASEB J 2016; 31:1238-1247. [PMID: 27986808 DOI: 10.1096/fj.201601132r] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Accepted: 12/06/2016] [Indexed: 11/11/2022]
Abstract
Peripheral neuronal activation by inflammatory mediators is a multifaceted physiological response that involves a multitude of regulated cellular functions. One key pathway that has been shown to be involved in inflammatory pain is Gq/GPCR, whose activation by inflammatory mediators is followed by the regulated response of the cation channel transient receptor potential vanilloid 1 (TRPV1). However, the mechanism that underlies TRPV1 activation downstream of the Gq/GPCR pathway has yet to be fully defined. In this study, we employ pharmacological and molecular biology tools to dissect this activation mechanism via perforated-patch recordings and calcium imaging of both neurons and a heterologous system. We showed that TRPV1 activity downstream of Gq/GPCR activation only produced a subdued current, which was noticeably different from the robust current that is typical of TRPV1 activation by exogenous stimuli. Moreover, we specifically demonstrated that 2 pathways downstream of Gq/GPCR signaling, namely endovanilloid production by lipoxygenases and channel phosphorylation by PKC, converge on TRPV1 to evoke a tightly regulated response. Of importance, we show that only when both pathways are acting on TRPV1 is the inflammatory-mediated response achieved. We propose that the requirement of multiple signaling events allows subdued TRPV1 activation to evoke regulated neuronal response during inflammation.-Kumar R., Hazan, A., Geron, M., Steinberg, R., Livni, L., Matzner, H., Priel, A. Activation of transient receptor potential vanilloid 1 by lipoxygenase metabolites depends on PKC phosphorylation.
Collapse
Affiliation(s)
- Rakesh Kumar
- Institute for Drug Research, School of Pharmacy, The Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Adina Hazan
- Institute for Drug Research, School of Pharmacy, The Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Matan Geron
- Institute for Drug Research, School of Pharmacy, The Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Rebbeca Steinberg
- Institute for Drug Research, School of Pharmacy, The Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Lital Livni
- Institute for Drug Research, School of Pharmacy, The Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Henry Matzner
- Institute for Drug Research, School of Pharmacy, The Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Avi Priel
- Institute for Drug Research, School of Pharmacy, The Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
38
|
Abstract
A response to environmental stress is critical to alleviate cellular injury and maintain cellular homeostasis. Eukaryotic initiation factor 2 (eIF2) is a key integrator of cellular stress responses and an important regulator of mRNA translation. Diverse stress signals lead to the phosphorylation of the α subunit of eIF2 (Ser51), resulting in inhibition of global protein synthesis while promoting expression of proteins that mediate cell adaptation to stress. Here we report that eIF2α is instrumental in the control of noxious heat sensation. Mice with decreased eIF2α phosphorylation (eIF2α+/S51A) exhibit reduced responses to noxious heat. Pharmacological attenuation of eIF2α phosphorylation decreases thermal, but not mechanical, pain sensitivity, whereas increasing eIF2α phosphorylation has the opposite effect on thermal nociception. The impact of eIF2α phosphorylation (p-eIF2α) on thermal thresholds is dependent on the transient receptor potential vanilloid 1. Moreover, we show that induction of eIF2α phosphorylation in primary sensory neurons in a chronic inflammation pain model contributes to thermal hypersensitivity. Our results demonstrate that the cellular stress response pathway, mediated via p-eIF2α, represents a mechanism that could be used to alleviate pathological heat sensation.
Collapse
|
39
|
Shapiro H, Singer P, Ariel A. Beyond the classic eicosanoids: Peripherally-acting oxygenated metabolites of polyunsaturated fatty acids mediate pain associated with tissue injury and inflammation. Prostaglandins Leukot Essent Fatty Acids 2016; 111:45-61. [PMID: 27067460 DOI: 10.1016/j.plefa.2016.03.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 02/25/2016] [Accepted: 03/01/2016] [Indexed: 02/06/2023]
Abstract
Pain is a complex sensation that may be protective or cause undue suffering and loss of function, depending on the circumstances. Peripheral nociceptor neurons (PNs) innervate most tissues, and express ion channels, nocisensors, which depolarize the cell in response to intense stimuli and numerous substances. Inflamed tissues manifest inflammatory hyperalgesia in which the threshold for pain and the response to painful stimuli are decreased and increased, respectively. Constituents of the inflammatory milieu sensitize PNs, thereby contributing to hyperalgesia. Polyunsaturated fatty acids undergo enzymatic and free radical-mediated oxygenation into an array of bioactive metabolites, oxygenated polyunsaturated fatty acids (oxy-PUFAs), including the classic eicosanoids. Oxy-PUFA production is enhanced during inflammation. Pioneering studies by Vane and colleagues from the early 1970s first implicated classic eicosanoids in the pain associated with inflammation. Here, we review the production and action of oxy-PUFAs that are not classic eicosanoids, but nevertheless are produced in injured/ inflamed tissues and activate or sensitize PNs. In general, oxy-PUFAs that sensitize PNs may do so directly, by activation of nocisensors, ion channels or GPCRs expressed on the surface of PNs, or indirectly, by increasing the production of inflammatory mediators that activate or sensitize PNs. We focus on oxy-PUFAs that act directly on PNs. Specifically, we discuss the role of arachidonic acid-derived 12S-HpETE, HNE, ONE, PGA2, iso-PGA2 and 15d-PGJ2, 5,6-and 8,9-EET, PGE2-G and 8R,15S-diHETE, as well as the linoleic acid-derived 9-and 13-HODE in inducing acute nocifensive behavior and/or inflammatory hyperalgesia in rodents. The nocisensors TRPV1, TRPV4 and TRPA1, and putative Gαs-type GPCRs are the PN targets of these oxy-PUFAs.
Collapse
Affiliation(s)
- Haim Shapiro
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, 199 Abba Khoushy Ave, Mount Carmel, Haifa 3498838, Israel.
| | - Pierre Singer
- Department of General Intensive Care, Institute for Nutrition Research, Rabin Medical Center, Sackler School of Medicine, Tel Aviv University, Petah Tikva 49100, Israel
| | - Amiram Ariel
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, 199 Abba Khoushy Ave, Mount Carmel, Haifa 3498838, Israel
| |
Collapse
|
40
|
Chen M, Xin J, Liu B, Luo L, Li J, Yin W, Li M. Mitogen-Activated Protein Kinase and Intracellular Polyamine Signaling Is Involved in TRPV1 Activation-Induced Cardiac Hypertrophy. J Am Heart Assoc 2016; 5:JAHA.116.003718. [PMID: 27473037 PMCID: PMC5015292 DOI: 10.1161/jaha.116.003718] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND The transient receptor potential vanilloid type 1 (TRPV1) is expressed in the cardiovascular system, and increased TRPV1 expression has been associated with cardiac hypertrophy. Nevertheless, the role of TRPV1 in the pathogenesis of cardiac hypertrophy and the underlying molecular mechanisms remain unclear. METHODS AND RESULTS In cultured cardiomyocytes, activation of TRPV1 increased cell size and elevated expression of atrial natriuretic peptide mRNA and intracellular calcium level, which was reversed by TRPV1 antagonist capsazepine. Increased expression of phosphorylated calmodulin-dependent protein kinase IIδ and mitogen-activated protein kinases were found in TRPV1 agonist capsaicin-treated cardiomyocytes. Selective inhibitor of calmodulin-dependent protein kinase IIδ decreased phosphorylation of extracellular signal-regulated kinases and p38. Capsaicin induced an increase in expression of ornithine decarboxylase protein, which is the key enzyme in polyamine biosynthesis in cardiomyocytes. Nevertheless, there was no obvious change of ornithine decarboxylase expression in TRPV1 knockdown cells after capsaicin treatment, and specific inhibitors of calmodulin-dependent protein kinase IIδ or p38 downregulated the capsaicin-induced expression of ornithine decarboxylase. Capsazepine alleviated the increase in cross-sectional area of cardiomyocytes and the ratio of heart weight to body weight and improved cardiac function, including left ventricular internal end-diastolic and -systolic dimensions and ejection fraction and fractional shortening percentages, in mice treated with transverse aorta constriction. Capsazepine also reduced expression of ornithine decarboxylase and cardiac polyamine levels. Transverse aorta constriction induced increases in phosphorylated calmodulin-dependent protein kinase IIδ and extracellular signal-regulated kinases, and p38 and Serca2a were attenuated by capsazepine treatment. CONCLUSIONS This study revealed that the mitogen-activated protein kinase signaling pathway and intracellular polyamines are essential for TRPV1 activation-induced cardiac hypertrophy.
Collapse
Affiliation(s)
- Mai Chen
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jiajia Xin
- Department of Blood Transfusion, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Baohui Liu
- Department of Cardiac Surgery, Binzhou Medical University Hospital, Binzhou, China
| | - Liyang Luo
- Department of Pharmacology, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Jiayi Li
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Wen Yin
- Department of Blood Transfusion, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Mingkai Li
- Department of Pharmacology, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
41
|
Hargreaves KM, Ruparel S. Role of Oxidized Lipids and TRP Channels in Orofacial Pain and Inflammation. J Dent Res 2016; 95:1117-23. [PMID: 27307050 DOI: 10.1177/0022034516653751] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Acute or chronic inflammation comprises a highly prevalent type of orofacial pain and is mediated by the generation of endogenous agonists that activate numerous receptors expressed on terminals of trigeminal (TG) nociceptive afferent neurons. One such studied receptor is transient receptor potential vanilloid subtype 1 (TRPV1). TRPV1 is a ligand-gated cation channel that is expressed on a major subclass of nociceptors and is found in many orofacial tissues, including dental pulp. Antagonists to TRPV1 reveal an important role for this channel in mediating hypersensitivity in preclinical models of inflammatory or neuropathic pain. Recent studies have demonstrated that endogenous TRPV1 agonists are generated by oxidation of omega-6 polyunsaturated fatty acids, including both linoleic acid and arachidonic acid. A major mechanism triggering the release of oxidative linoleic acid metabolites (OLAMs) and oxidative arachidonic acid metabolites (OAAMs) is the action of oxidative enzymes. Oxidative enzymes such as cytochrome P450 isozymes are rapidly upregulated in TG neurons after orofacial inflammation and increase the capacity of TG neurons to generate OLAMs. Cytochrome P450 isozymes are also increased in immune cells in irreversibly inflamed human dental pulp, and extracts of this tissue have significantly increased capacity to generate OLAMs. Together, these studies point to a novel pain mechanism involving the enzymatic generation of endogenous OLAM and OAAM agonists of TRPV1. This finding provides a rationale for an entirely new class of analgesics by inhibition of oxidative enzyme activity.
Collapse
Affiliation(s)
- K M Hargreaves
- Department of Endodontics, University of Texas Health Science Center at San Antonio, TX, USA
| | - S Ruparel
- Department of Endodontics, University of Texas Health Science Center at San Antonio, TX, USA
| |
Collapse
|
42
|
Hanson SM, Newstead S, Swartz KJ, Sansom MSP. Capsaicin interaction with TRPV1 channels in a lipid bilayer: molecular dynamics simulation. Biophys J 2016; 108:1425-1434. [PMID: 25809255 PMCID: PMC4375533 DOI: 10.1016/j.bpj.2015.02.013] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 01/29/2015] [Accepted: 02/03/2015] [Indexed: 12/18/2022] Open
Abstract
Transient receptor potential vanilloid subtype 1 (TRPV1) is a heat-sensitive ion channel also involved in pain sensation, and is the receptor for capsaicin, the active ingredient of hot chili peppers. The recent structures of TRPV1 revealed putative ligand density within the S1 to S4 voltage-sensor-like domain of the protein. However, questions remain regarding the dynamic role of the lipid bilayer in ligand binding to TRPV1. Molecular dynamics simulations were used to explore behavior of capsaicin in a 1-palmitoyl-2-oleoyl phosphatidylcholine bilayer and with the target S1–S4 transmembrane helices of TRPV1. Equilibrium simulations reveal a preferred interfacial localization for capsaicin. We also observed a capsaicin molecule flipping from the extracellular to the intracellular leaflet, and subsequently able to access the intracellular TRPV1 binding site. Calculation of the potential of mean force (i.e., free energy profile) of capsaicin along the bilayer normal confirms that it prefers an interfacial localization. The free energy profile indicates that there is a nontrivial but surmountable barrier to the flipping of capsaicin between opposing leaflets of the bilayer. Molecular dynamics of the S1–S4 transmembrane helices of the TRPV1 in a lipid bilayer confirm that Y511, known to be crucial to capsaicin binding, has a distribution along the bilayer normal similar to that of the aromatic group of capsaicin. Simulations were conducted of the TRPV1 S1–S4 transmembrane helices in the presence of capsaicin placed in the aqueous phase, in the lipid, or docked to the protein. No stable interaction between ligand and protein was seen for simulations initiated with capsaicin in the bilayer. However, interactions were seen between TRPV1 and capsaicin starting from the cytosolic aqueous phase, and capsaicin remained stable in the majority of simulations from the docked pose. We discuss the significance of capsaicin flipping from the extracellular to the intracellular leaflet and mechanisms of binding site access by capsaicin.
Collapse
Affiliation(s)
- Sonya M Hanson
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom; Computational Biology Program, Memorial Sloan-Kettering Cancer Center, New York, New York; Molecular Physiology and Biophysics Section, Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| | - Simon Newstead
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Kenton J Swartz
- Molecular Physiology and Biophysics Section, Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| | - Mark S P Sansom
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
43
|
Niederberger E, Kuner R, Geißlinger G. [Pharmacological aspects of pain research in Germany]. Schmerz 2015; 29:531-8. [PMID: 26294077 DOI: 10.1007/s00482-015-0042-0] [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: 11/25/2022]
Abstract
In spite of several approved analgesics, the therapy of pain still constitutes a challenge due to the fact that the drugs do not exert sufficient efficacy or are associated with severe side effects. Therefore, the development of new and improved painkillers is still of great importance. A number of highly qualified scientists in Germany are investigating signal transduction pathways in pain, effectivity of new drugs and the so far incompletely investigated mechanisms of well-known analgesics in preclinical and clinical studies. The highlights of pharmacological pain research in Germany are summarized in this article.
Collapse
Affiliation(s)
- E Niederberger
- pharmazentrum frankfurt/ZAFES, Institut für Klinische Pharmakologie, Klinikum der Goethe-Universität Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Deutschland
| | - R Kuner
- Pharmakologisches Institut, Universität Heidelberg, Im Neuenheimer Feld 584, 69120, Heidelberg, Deutschland
| | - G Geißlinger
- pharmazentrum frankfurt/ZAFES, Institut für Klinische Pharmakologie, Klinikum der Goethe-Universität Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Deutschland.
| |
Collapse
|
44
|
Gouveia-Figueira S, Nording ML, Gaida JE, Forsgren S, Alfredson H, Fowler CJ. Serum levels of oxylipins in achilles tendinopathy: an exploratory study. PLoS One 2015; 10:e0123114. [PMID: 25875933 PMCID: PMC4395257 DOI: 10.1371/journal.pone.0123114] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 02/27/2015] [Indexed: 12/11/2022] Open
Abstract
Background Linoleic acid-derived oxidation products are found in experimental pain models. However, little is known about the levels of such oxylipins in human pain. In consequence, in the present study, we have undertaken a lipidomic profiling of oxylipins in blood serum from patients with Achilles tendinopathy and controls. Methodology/Principal findings A total of 34 oxylipins were analysed in the serum samples. At a significance level of P<0.00147 (<0.05/34), two linoleic acid-derived oxylipins, 13-hydroxy-10E,12Z-octadecadienoic (13-HODE) and 12(13)-dihydroxy-9Z-octadecenoic acid (12,13-DiHOME) were present at significantly higher levels in the Achilles tendinopathy samples. This difference remained significant when the dataset was controlled for age, gender and body-mass index. In contrast, 0/21 of the arachidonic acid- and 0/4 of the dihomo-γ-linolenic acid, eicosapentaenoic acid or docosahenaenoic acid-derived oxylipins were higher in the patient samples at this level of significance. The area under the Receiver-Operator Characteristic (ROC) curve for 12,13-DiHOME was 0.91 (P<0.0001). Levels of four N-acylethanolamines were also analysed and found not to be significantly different between the controls and the patients at the level of P<0.0125 (<0.05/4). Conclusions/Significance It is concluded from this exploratory study that abnormal levels of linoleic acid-derived oxylipins are seen in blood serum from patients with Achilles tendinopathy. Given the ability of two of these, 9- and 13-HODE to activate transient receptor potential vanilloid 1, it is possible that these changes may contribute to the symptoms seen in Achilles tendinopathy.
Collapse
Affiliation(s)
- Sandra Gouveia-Figueira
- Department of Pharmacology and Clinical Neuroscience, Pharmacology Unit, Umeå University, Umeå, Sweden
- Department of Chemistry, Umeå University, Umeå, Sweden
| | | | - Jamie E. Gaida
- Discipline of Physiotherapy, University of Canberra, Bruce, ACT, Australia
- Department of Physiotherapy, Monash University, Melbourne, VIC, Australia
| | - Sture Forsgren
- Department of Integrative Medical Biology, Section for Anatomy, Umeå University, Umeå, Sweden
| | - Håkan Alfredson
- Department of Surgical and Perioperative Sciences, Sports Medicine Unit, Umeå University, Umeå, Sweden
| | - Christopher J. Fowler
- Department of Pharmacology and Clinical Neuroscience, Pharmacology Unit, Umeå University, Umeå, Sweden
- * E-mail:
| |
Collapse
|
45
|
Serhan CN, Chiang N, Dalli J. The resolution code of acute inflammation: Novel pro-resolving lipid mediators in resolution. Semin Immunol 2015; 27:200-15. [PMID: 25857211 DOI: 10.1016/j.smim.2015.03.004] [Citation(s) in RCA: 402] [Impact Index Per Article: 44.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 03/07/2015] [Accepted: 03/09/2015] [Indexed: 12/31/2022]
Abstract
Studies into the mechanisms in resolution of self-limited inflammation and acute reperfusion injury have uncovered a new genus of pro-resolving lipid mediators coined specialized pro-resolving mediators (SPM) including lipoxins, resolvins, protectins and maresins that are each temporally produced by resolving-exudates with distinct actions for return to homeostasis. SPM evoke potent anti-inflammatory and novel pro-resolving mechanisms as well as enhance microbial clearance. While born in inflammation-resolution, SPM are conserved structures with functions discovered in microbial defense, pain, organ protection and tissue regeneration, wound healing, cancer, reproduction, and neurobiology-cognition. This review covers these SPM mechanisms and other new omega-3 PUFA pathways that open their path for functions in resolution physiology.
Collapse
Affiliation(s)
- Charles N Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Harvard Institutes of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, United States.
| | - Nan Chiang
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Harvard Institutes of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, United States
| | - Jesmond Dalli
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Harvard Institutes of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, United States
| |
Collapse
|
46
|
Taberner FJ, Fernández-Ballester G, Fernández-Carvajal A, Ferrer-Montiel A. TRP channels interaction with lipids and its implications in disease. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1848:1818-27. [PMID: 25838124 DOI: 10.1016/j.bbamem.2015.03.022] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 03/21/2015] [Accepted: 03/23/2015] [Indexed: 01/21/2023]
Abstract
Transient receptor potential (TRP) proteins are a family of ion channels central for sensory signaling. These receptors and, in particular, those involved in thermal sensing are also involved in pain signaling. Noteworthy, thermosensory receptors are polymodal ion channels that respond to both physical and chemical stimuli, thus integrating different environmental clues. In addition, their activity is modulated by algesic agents and lipidergic substances that are primarily released in pathological states. Lipids and lipid-like molecules have been found that can directly activate some thermosensory channels or modulate their activity by either potentiating or inhibiting it. To date, more than 50 endogenous lipids that can regulate TRP channel activity in sensory neurons have been described, thus representing the majority of known endogenous TRP channel modulators. Lipid modulators of TRP channels comprise lipids from a variety of metabolic pathways, including metabolites of the cyclooxygenase, lipoxygenase and cytochrome-P450 pathways, phospholipids and lysophospholipids. Therefore, TRP-channels are able to integrate and interpret incoming signals from the different metabolic lipid pathways. Taken together, the large number of lipids that can activate, sensitize or inhibit neuronal TRP-channels highlights the pivotal role of these molecules in sensory biology as well as in pain transduction and perception. This article is part of a Special Issue entitled: Lipid-protein interactions. Guest Editors: Amitabha Chattopadhyay and Jean-Marie Ruysschaert.
Collapse
Affiliation(s)
- Francisco J Taberner
- Instituto de Biología Molecular y Celular, Universidad Miguel Hernández, Alicante, Spain
| | | | | | - Antonio Ferrer-Montiel
- Instituto de Biología Molecular y Celular, Universidad Miguel Hernández, Alicante, Spain.
| |
Collapse
|
47
|
Witschas K, Jobin ML, Korkut DN, Vladan MM, Salgado G, Lecomte S, Vlachova V, Alves ID. Interaction of a peptide derived from C-terminus of human TRPA1 channel with model membranes mimicking the inner leaflet of the plasma membrane. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1848:1147-56. [PMID: 25687973 DOI: 10.1016/j.bbamem.2015.02.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 01/21/2015] [Accepted: 02/04/2015] [Indexed: 11/29/2022]
Abstract
The transient receptor potential ankyrin 1 channel (TRPA1) belongs to the TRP cation channel superfamily that responds to a panoply of stimuli such as changes in temperature, calcium levels, reactive oxygen and nitrogen species and lipid mediators among others. The TRP superfamily has been implicated in diverse pathological states including neurodegenerative disorders, kidney diseases, inflammation, pain and cancer. The intracellular C-terminus is an important regulator of TRP channel activity. Studies with this and other TRP superfamily members have shown that the C-terminus association with lipid bilayer alters channel sensitivity and activation, especially interactions occurring through basic residues. Nevertheless, it is not yet clear how this process takes place and which regions in the C-terminus would be responsible for such membrane recognition. With that in mind, herein the first putative membrane interacting region of the C-terminus of human TRPA1, (corresponding to a 29 residue peptide, IAEVQKHASLKRIAMQVELHTSLEKKLPL) named H1 due to its potential helical character was chosen for studies of membrane interaction. The affinity of H1 to lipid membranes, H1 structural changes occurring upon this interaction as well as effects of this interaction in lipid organization and integrity were investigated using a biophysical approach. Lipid models systems composed of zwitterionic and anionic lipids, namely those present in the lipid membrane inner leaflet, where H1 is prone to interact, where used. The study reveals a strong interaction and affinity of H1 as well as peptide structuration especially with membranes containing anionic lipids. Moreover, the interactions and peptide structure adoption are headgroup specific.
Collapse
Affiliation(s)
- Katja Witschas
- CBMN-UMR 5248 CNRS, University of Bordeaux, IPB, Allée Geoffroy St. Hilaire, 33600 Pessac, France; Department of Cellular Neurophysiology, Institute of Physiology, Academy of Sciences of the Czech Republic, Videnska 1083, 142 20 Prague 4, Czech Republic
| | - Marie-Lise Jobin
- CBMN-UMR 5248 CNRS, University of Bordeaux, IPB, Allée Geoffroy St. Hilaire, 33600 Pessac, France
| | - Dursun Nizam Korkut
- INSERM, U869, ARNA Laboratory, University of Bordeaux, 2 rue Robert Escarpit, 33607 Pessac, France
| | - Maria Magdalena Vladan
- CBMN-UMR 5248 CNRS, University of Bordeaux, IPB, Allée Geoffroy St. Hilaire, 33600 Pessac, France
| | - Gilmar Salgado
- INSERM, U869, ARNA Laboratory, University of Bordeaux, 2 rue Robert Escarpit, 33607 Pessac, France
| | - Sophie Lecomte
- CBMN-UMR 5248 CNRS, University of Bordeaux, IPB, Allée Geoffroy St. Hilaire, 33600 Pessac, France
| | - Viktorie Vlachova
- Department of Cellular Neurophysiology, Institute of Physiology, Academy of Sciences of the Czech Republic, Videnska 1083, 142 20 Prague 4, Czech Republic
| | - Isabel D Alves
- CBMN-UMR 5248 CNRS, University of Bordeaux, IPB, Allée Geoffroy St. Hilaire, 33600 Pessac, France.
| |
Collapse
|
48
|
Yaksh TL. TRPV1 expression regulation… A further step in defining its biology: commentary for K. Zavala et al. "The anticancer antibiotic mithramycin-A inhibits TRPV1 expression in dorsal root ganglion neurons" [Neurosci. Lett. (2014) doi:10.1016/j.neulet.2014.01.021]. Neurosci Lett 2014; 578:209-10. [PMID: 24769421 DOI: 10.1016/j.neulet.2014.04.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 04/16/2014] [Indexed: 11/16/2022]
Affiliation(s)
- Tony L Yaksh
- Department of Anesthesiology, University of California, San Diego, United States.
| |
Collapse
|
49
|
Abstract
Current analgesics predominately modulate pain transduction and transmission in neurons and have limited success in controlling disease progression. Accumulating evidence suggests that neuroinflammation, which is characterized by infiltration of immune cells, activation of glial cells and production of inflammatory mediators in the peripheral and central nervous system, has an important role in the induction and maintenance of chronic pain. This Review focuses on emerging targets - such as chemokines, proteases and the WNT pathway - that promote spinal cord neuroinflammation and chronic pain. It also highlights the anti-inflammatory and pro-resolution lipid mediators that act on immune cells, glial cells and neurons to resolve neuroinflammation, synaptic plasticity and pain. Targeting excessive neuroinflammation could offer new therapeutic opportunities for chronic pain and related neurological and psychiatric disorders.
Collapse
|
50
|
Jafri MS. Mechanisms of Myofascial Pain. INTERNATIONAL SCHOLARLY RESEARCH NOTICES 2014; 2014:523924. [PMID: 25574501 PMCID: PMC4285362 DOI: 10.1155/2014/523924] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2014] [Revised: 06/08/2014] [Accepted: 06/10/2014] [Indexed: 01/17/2023]
Abstract
Myofascial pain syndrome is an important health problem. It affects a majority of the general population, impairs mobility, causes pain, and reduces the overall sense of well-being. Underlying this syndrome is the existence of painful taut bands of muscle that contain discrete, hypersensitive foci called myofascial trigger points. In spite of the significant impact on public health, a clear mechanistic understanding of the disorder does not exist. This is likely due to the complex nature of the disorder which involves the integration of cellular signaling, excitation-contraction coupling, neuromuscular inputs, local circulation, and energy metabolism. The difficulties are further exacerbated by the lack of an animal model for myofascial pain to test mechanistic hypothesis. In this review, current theories for myofascial pain are presented and their relative strengths and weaknesses are discussed. Based on new findings linking mechanoactivation of reactive oxygen species signaling to destabilized calcium signaling, we put forth a novel mechanistic hypothesis for the initiation and maintenance of myofascial trigger points. It is hoped that this lays a new foundation for understanding myofascial pain syndrome and how current therapies work, and gives key insights that will lead to the improvement of therapies for its treatment.
Collapse
Affiliation(s)
- M. Saleet Jafri
- Krasnow Institute for Advanced Study, George Mason University, 4400 University Drive, MNS 2A1, Fairfax, VA 22030, USA
| |
Collapse
|