51
|
Sappington RM, Sidorova T, Ward NJ, Chakravarthy R, Ho KW, Calkins DJ. Activation of transient receptor potential vanilloid-1 (TRPV1) influences how retinal ganglion cell neurons respond to pressure-related stress. Channels (Austin) 2016; 9:102-13. [PMID: 25713995 DOI: 10.1080/19336950.2015.1009272] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Our recent studies implicate the transient receptor potential vanilloid-1 (TRPV1) channel as a mediator of retinal ganglion cell (RGC) function and survival. With elevated pressure in the eye, TRPV1 increases in RGCs, supporting enhanced excitability, while Trpv1 -/- accelerates RGC degeneration in mice. Here we find TRPV1 localized in monkey and human RGCs, similar to rodents. Expression increases in RGCs exposed to acute changes in pressure. In retinal explants, contrary to our animal studies, both Trpv1 -/- and pharmacological antagonism of the channel prevented pressure-induced RGC apoptosis, as did chelation of extracellular Ca(2+). Finally, while TRPV1 and TRPV4 co-localize in some RGC bodies and form a protein complex in the retina, expression of their mRNA is inversely related with increasing ocular pressure. We propose that TRPV1 activation by pressure-related insult in the eye initiates changes in expression that contribute to a Ca(2+)-dependent adaptive response to maintain excitatory signaling in RGCs.
Collapse
Affiliation(s)
- Rebecca M Sappington
- a The Vanderbilt Eye Institute and Vanderbilt Brain Institute ; Vanderbilt University School of Medicine ; Nashville , TN USA
| | | | | | | | | | | |
Collapse
|
52
|
Pérez-Faginas P, Teresa Aranda M, Torre-Martínez RDL, Quirce S, Fernández-Carvajal A, Ferrer-Montiel A, González-Muñiz R. New transient receptor potential TRPV1, TRPM8 and TRPA1 channel antagonists from a single linear β,γ-diamino ester scaffold. RSC Adv 2016. [DOI: 10.1039/c5ra25709c] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Incorporation of minor changes in the structure of a single β,γ-diaminoester linear scaffold resulted in selective hits for TRPV1, TRPM8 and TRPA1 blockade, as well as some dual antagonists.
Collapse
Affiliation(s)
| | | | - Roberto de la Torre-Martínez
- Instituto de Biología Molecular y Celular
- Universidad Miguel Hernández
- Avenida de la Universidad s/n
- 03202 Elche
- Spain
| | - Susana Quirce
- Instituto de Biología Molecular y Celular
- Universidad Miguel Hernández
- Avenida de la Universidad s/n
- 03202 Elche
- Spain
| | - Asia Fernández-Carvajal
- Instituto de Biología Molecular y Celular
- Universidad Miguel Hernández
- Avenida de la Universidad s/n
- 03202 Elche
- Spain
| | - Antonio Ferrer-Montiel
- Instituto de Biología Molecular y Celular
- Universidad Miguel Hernández
- Avenida de la Universidad s/n
- 03202 Elche
- Spain
| | | |
Collapse
|
53
|
Bertin S, Raz E. Transient Receptor Potential (TRP) channels in T cells. Semin Immunopathol 2015; 38:309-19. [PMID: 26468011 DOI: 10.1007/s00281-015-0535-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 10/01/2015] [Indexed: 12/16/2022]
Abstract
The transient receptor potential (TRP) family of ion channels is widely expressed in many cell types and plays various physiological roles. Growing evidence suggests that certain TRP channels are functionally expressed in the immune system. Indeed, an increasing number of reports have demonstrated the functional expression of several TRP channels in innate and adaptive immune cells and have highlighted their critical role in the activation and function of these cells. However, very few reviews have been entirely dedicated to this subject. Here, we will summarize the recent findings with regards to TRP channel expression in T cells and discuss their emerging role as regulators of T cell activation and functions. Moreover, these studies suggest that beyond their pharmaceutical interest in pain management, certain TRP channels may represent potential novel therapeutic targets for various immune-related diseases.
Collapse
Affiliation(s)
- Samuel Bertin
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0663, USA.
| | - Eyal Raz
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0663, USA
| |
Collapse
|
54
|
|
55
|
Sousa-Valente J, Andreou AP, Urban L, Nagy I. Transient receptor potential ion channels in primary sensory neurons as targets for novel analgesics. Br J Pharmacol 2014; 171:2508-27. [PMID: 24283624 DOI: 10.1111/bph.12532] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 11/11/2013] [Accepted: 11/20/2013] [Indexed: 12/12/2022] Open
Abstract
The last decade has witnessed an explosion in novel findings relating to the molecules involved in mediating the sensation of pain in humans. Transient receptor potential (TRP) ion channels emerged as the greatest group of molecules involved in the transduction of various physical stimuli into neuronal signals in primary sensory neurons, as well as, in the development of pain. Here, we review the role of TRP ion channels in primary sensory neurons in the development of pain associated with peripheral pathologies and possible strategies to translate preclinical data into the development of effective new analgesics. Based on available evidence, we argue that nociception-related TRP channels on primary sensory neurons provide highly valuable targets for the development of novel analgesics and that, in order to reduce possible undesirable side effects, novel analgesics should prevent the translocation from the cytoplasm to the cell membrane and the sensitization of the channels rather than blocking the channel pore or binding sites for exogenous or endogenous activators.
Collapse
Affiliation(s)
- J Sousa-Valente
- Anaesthetics, Pain Medicine and Intensive Care Section, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK
| | | | | | | |
Collapse
|
56
|
Lin YJ, Lin RL, Ruan T, Khosravi M, Lee LY. A synergistic effect of simultaneous TRPA1 and TRPV1 activations on vagal pulmonary C-fiber afferents. J Appl Physiol (1985) 2014; 118:273-81. [PMID: 25414245 DOI: 10.1152/japplphysiol.00805.2014] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Transient receptor potential ankyrin type 1 (TRPA1) and vanilloid type 1 (TRPV1) receptors are coexpressed in vagal pulmonary C-fiber sensory nerves. Because both these receptors are sensitive to a number of endogenous inflammatory mediators, it is conceivable that they can be activated simultaneously during airway inflammation. This study aimed to determine whether there is an interaction between these two polymodal transducers upon simultaneous activation, and how it modulates the activity of vagal pulmonary C-fiber sensory nerves. In anesthetized, spontaneously breathing rats, the reflex-mediated apneic response to intravenous injection of a combined dose of allyl isothiocyanate (AITC, a TRPA1 activator) and capsaicin (Cap, a TRPV1 activator) was ∼202% greater than the mathematical sum of the responses to AITC and Cap when they were administered individually. Similar results were also observed in anesthetized mice. In addition, the synergistic effect was clearly demonstrated when the afferent activity of single vagal pulmonary C-fiber afferents were recorded in anesthetized, artificially ventilated rats; C-fiber responses to AITC, Cap and AITC + Cap (in combination) were 0.6 ± 0.1, 0.8 ± 0.1, and 4.8 ± 0.6 impulses/s (n = 24), respectively. This synergism was absent when either AITC or Cap was replaced by other chemical activators of pulmonary C-fiber afferents. The pronounced potentiating effect was further demonstrated in isolated vagal pulmonary sensory neurons using the Ca(2+) imaging technique. In summary, this study showed a distinct positive interaction between TRPA1 and TRPV1 when they were activated simultaneously in pulmonary C-fiber sensory nerves.
Collapse
Affiliation(s)
- Yu-Jung Lin
- Department of Physiology, University of Kentucky Medical Center, Lexington, Kentucky
| | - Ruei-Lung Lin
- Department of Physiology, University of Kentucky Medical Center, Lexington, Kentucky
| | - Ting Ruan
- School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan; and
| | - Mehdi Khosravi
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Kentucky Medical Center, Lexington, Kentucky
| | - Lu-Yuan Lee
- Department of Physiology, University of Kentucky Medical Center, Lexington, Kentucky;
| |
Collapse
|
57
|
Abstract
![]()
To
date, 28 mammalian transient receptor potential (TRP) channels
have been cloned and characterized. They are grouped into six subfamilies
on the basis of their amino acid sequence homology: TRP Ankyrin (TRPA),
TRP Canonical (TRPC), TRP Melastatin (TRPM), TRP Mucolipin (TRPML),
TRP Polycystin (TRPP), and TRP Vanilloid (TRPV). Most of the TRP channels
are nonselective cation channels expressed on the cell membrane and
exhibit variable permeability ratios for Ca2+ versus Na+. They mediate sensory functions (such as vision, nociception,
taste transduction, temperature sensation, and pheromone signaling)
and homeostatic functions (such as divalent cation flux, hormone release,
and osmoregulation). Significant progress has been made in our understanding
of the specific roles of these TRP channels and their activation mechanisms.
In this Review, the emphasis will be on the activation of TRP channels
by phytochemicals that are claimed to exert health benefits. Recent
findings complement the anecdotal evidence that some of these phytochemicals
have specific receptors and the activation of which is responsible
for the physiological effects. Now, the targets for these phytochemicals
are being unveiled; a specific hypothesis can be proposed and tested
experimentally to infer a scientific validity of the claims of the
health benefits. The broader and pressing issues that have to be addressed
are related to the quantities of the active ingredients in a given
preparation, their bioavailability, metabolism, adverse effects, excretion,
and systemic versus local effects.
Collapse
Affiliation(s)
- Louis S. Premkumar
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, Illinois 62702, United States
| |
Collapse
|
58
|
Zhang Y, Qin W, Qian Z, Liu X, Wang H, Gong S, Sun YG, Snutch TP, Jiang X, Tao J. Peripheral pain is enhanced by insulin-like growth factor 1 through a G protein-mediated stimulation of T-type calcium channels. Sci Signal 2014; 7:ra94. [DOI: 10.1126/scisignal.2005283] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
59
|
Ferrandiz-Huertas C, Mathivanan S, Wolf CJ, Devesa I, Ferrer-Montiel A. Trafficking of ThermoTRP Channels. MEMBRANES 2014; 4:525-64. [PMID: 25257900 PMCID: PMC4194048 DOI: 10.3390/membranes4030525] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 07/11/2014] [Accepted: 08/08/2014] [Indexed: 12/19/2022]
Abstract
ThermoTRP channels (thermoTRPs) define a subfamily of the transient receptor potential (TRP) channels that are activated by changes in the environmental temperature, from noxious cold to injurious heat. Acting as integrators of several stimuli and signalling pathways, dysfunction of these channels contributes to several pathological states. The surface expression of thermoTRPs is controlled by both, the constitutive and regulated vesicular trafficking. Modulation of receptor surface density during pathological processes is nowadays considered as an interesting therapeutic approach for management of diseases, such as chronic pain, in which an increased trafficking is associated with the pathological state. This review will focus on the recent advances trafficking of the thermoTRP channels, TRPV1, TRPV2, TRPV4, TRPM3, TRPM8 and TRPA1, into/from the plasma membrane. Particularly, regulated membrane insertion of thermoTRPs channels contributes to a fine tuning of final channel activity, and indeed, it has resulted in the development of novel therapeutic approaches with successful clinical results such as disruption of SNARE-dependent exocytosis by botulinum toxin or botulinomimetic peptides.
Collapse
Affiliation(s)
| | - Sakthikumar Mathivanan
- Instituto de Biología Molecular y Celular, Universidad Miguel Hernández, Alicante 03202, Spain.
| | - Christoph Jakob Wolf
- Instituto de Biología Molecular y Celular, Universidad Miguel Hernández, Alicante 03202, Spain.
| | - Isabel Devesa
- Instituto de Biología Molecular y Celular, Universidad Miguel Hernández, Alicante 03202, Spain.
| | - Antonio Ferrer-Montiel
- Instituto de Biología Molecular y Celular, Universidad Miguel Hernández, Alicante 03202, Spain.
| |
Collapse
|
60
|
Dunn TN, Adams SH. Relations between metabolic homeostasis, diet, and peripheral afferent neuron biology. Adv Nutr 2014; 5:386-93. [PMID: 25022988 PMCID: PMC4085187 DOI: 10.3945/an.113.005439] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
It is well established that food intake behavior and energy balance are regulated by crosstalk between peripheral organ systems and the central nervous system (CNS), for instance, through the actions of peripherally derived leptin on hindbrain and hypothalamic loci. Diet- or obesity-associated disturbances in metabolic and hormonal signals to the CNS can perturb metabolic homeostasis bodywide. Although interrelations between metabolic status and diet with CNS biology are well characterized, afferent networks (those sending information to the CNS from the periphery) have received far less attention. It is increasingly appreciated that afferent neurons in adipose tissue, the intestines, liver, and other tissues are important controllers of energy balance and feeding behavior. Disruption in their signaling may have consequences for cardiovascular, pancreatic, adipose, and immune function. This review discusses the diverse ways that afferent neurons participate in metabolic homeostasis and highlights how changes in their function associate with dysmetabolic states, such as obesity and insulin resistance.
Collapse
Affiliation(s)
- Tamara N. Dunn
- Graduate Group in Nutritional Biology and Department of Nutrition, University of California, Davis, CA; and
| | - Sean H. Adams
- Graduate Group in Nutritional Biology and Department of Nutrition, University of California, Davis, CA; and,Obesity and Metabolism Research Unit, USDA–Agricultural Research Service Western Human Nutrition Research Center, Davis, CA,To whom correspondence should be addressed. E-mail:
| |
Collapse
|
61
|
Nilius B, Szallasi A. Transient receptor potential channels as drug targets: from the science of basic research to the art of medicine. Pharmacol Rev 2014; 66:676-814. [PMID: 24951385 DOI: 10.1124/pr.113.008268] [Citation(s) in RCA: 377] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2025] Open
Abstract
The large Trp gene family encodes transient receptor potential (TRP) proteins that form novel cation-selective ion channels. In mammals, 28 Trp channel genes have been identified. TRP proteins exhibit diverse permeation and gating properties and are involved in a plethora of physiologic functions with a strong impact on cellular sensing and signaling pathways. Indeed, mutations in human genes encoding TRP channels, the so-called "TRP channelopathies," are responsible for a number of hereditary diseases that affect the musculoskeletal, cardiovascular, genitourinary, and nervous systems. This review gives an overview of the functional properties of mammalian TRP channels, describes their roles in acquired and hereditary diseases, and discusses their potential as drug targets for therapeutic intervention.
Collapse
Affiliation(s)
- Bernd Nilius
- KU Leuven, Department of Cellular and Molecular Medicine, Laboratory of Ion Channel Research, Campus Gasthuisberg, Leuven, Belgium (B.N.); and Department of Pathology, Monmouth Medical Center, Long Branch, New Jersey (A.S.)
| | - Arpad Szallasi
- KU Leuven, Department of Cellular and Molecular Medicine, Laboratory of Ion Channel Research, Campus Gasthuisberg, Leuven, Belgium (B.N.); and Department of Pathology, Monmouth Medical Center, Long Branch, New Jersey (A.S.)
| |
Collapse
|
62
|
Premkumar LS, Pabbidi RM. Diabetic peripheral neuropathy: role of reactive oxygen and nitrogen species. Cell Biochem Biophys 2014; 67:373-83. [PMID: 23722999 DOI: 10.1007/s12013-013-9609-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The prevalence of diabetes has reached epidemic proportions. There are two forms of diabetes: type 1 diabetes mellitus is due to auto-immune-mediated destruction of pancreatic β-cells resulting in absolute insulin deficiency and type 2 diabetes mellitus is due to reduced insulin secretion and or insulin resistance. Both forms of diabetes are characterized by chronic hyperglycemia, leading to the development of diabetic peripheral neuropathy (DPN) and microvascular pathology. DPN is characterized by enhanced or reduced thermal, chemical, and mechanical pain sensitivities. In the long-term, DPN results in peripheral nerve damage and accounts for a substantial number of non-traumatic lower-limb amputations. This review will address the mechanisms, especially the role of reactive oxygen and nitrogen species in the development and progression of DPN.
Collapse
Affiliation(s)
- Louis S Premkumar
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA,
| | | |
Collapse
|
63
|
Straub RH. TRPV1, TRPA1, and TRPM8 channels in inflammation, energy redirection, and water retention: role in chronic inflammatory diseases with an evolutionary perspective. J Mol Med (Berl) 2014; 92:925-37. [DOI: 10.1007/s00109-014-1175-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 05/17/2014] [Accepted: 05/20/2014] [Indexed: 01/04/2023]
|
64
|
Brito R, Sheth S, Mukherjea D, Rybak LP, Ramkumar V. TRPV1: A Potential Drug Target for Treating Various Diseases. Cells 2014; 3:517-45. [PMID: 24861977 PMCID: PMC4092862 DOI: 10.3390/cells3020517] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 05/14/2014] [Accepted: 05/15/2014] [Indexed: 12/11/2022] Open
Abstract
Transient receptor potential vanilloid 1 (TRPV1) is an ion channel present on sensory neurons which is activated by heat, protons, capsaicin and a variety of endogenous lipids termed endovanilloids. As such, TRPV1 serves as a multimodal sensor of noxious stimuli which could trigger counteractive measures to avoid pain and injury. Activation of TRPV1 has been linked to chronic inflammatory pain conditions and peripheral neuropathy, as observed in diabetes. Expression of TRPV1 is also observed in non-neuronal sites such as the epithelium of bladder and lungs and in hair cells of the cochlea. At these sites, activation of TRPV1 has been implicated in the pathophysiology of diseases such as cystitis, asthma and hearing loss. Therefore, drugs which could modulate TRPV1 channel activity could be useful for the treatment of conditions ranging from chronic pain to hearing loss. This review describes the roles of TRPV1 in the normal physiology and pathophysiology of selected organs of the body and highlights how drugs targeting this channel could be important clinically.
Collapse
Affiliation(s)
- Rafael Brito
- Department of Pharmacology and Neuroscience, Southern Illinois University School of Medicine, Springfield, IL 62702, USA.
| | - Sandeep Sheth
- Department of Pharmacology and Neuroscience, Southern Illinois University School of Medicine, Springfield, IL 62702, USA.
| | - Debashree Mukherjea
- Department of Surgery (Otoloryngalogy), Southern Illinois University School of Medicine, Springfield, IL 62702, USA.
| | - Leonard P Rybak
- Department of Pharmacology and Neuroscience, Southern Illinois University School of Medicine, Springfield, IL 62702, USA.
| | - Vickram Ramkumar
- Department of Pharmacology and Neuroscience, Southern Illinois University School of Medicine, Springfield, IL 62702, USA.
| |
Collapse
|
65
|
Absence of transient receptor potential vanilloid-1 accelerates stress-induced axonopathy in the optic projection. J Neurosci 2014; 34:3161-70. [PMID: 24573275 DOI: 10.1523/jneurosci.4089-13.2014] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
How neurons respond to stress in degenerative disease is of fundamental importance for identifying mechanisms of progression and new therapeutic targets. Members of the transient receptor potential (TRP) family of cation-selective ion channels are candidates for mediating stress signals, since different subunits transduce a variety of stimuli relevant in both normal and pathogenic physiology. We addressed this possibility for the TRP vanilloid-1 (TRPV1) subunit by comparing how the optic projection of Trpv1(-/-) mice and age-matched C57 controls responds to stress from elevated ocular pressure, the critical stressor in the most common optic neuropathy, glaucoma. Over a 5 week period of elevated pressure induced by microbead occlusion of ocular fluid, Trpv1(-/-) accelerated both degradation of axonal transport from retinal ganglion cells to the superior colliculus and degeneration of the axons themselves in the optic nerve. Ganglion cell body loss, which is normally later in progression, occurred in nasal sectors of Trpv1(-/-) but not C57 retina. Pharmacological antagonism of TRPV1 in rats similarly accelerated ganglion cell axonopathy. Elevated ocular pressure resulted in differences in spontaneous firing rate and action potential threshold current in Trpv1(-/-) ganglion cells compared with C57. In the absence of elevated pressure, ganglion cells in the two strains had similar firing patterns. Based on these data, we propose that TRPV1 may help neurons respond to disease-relevant stressors by enhancing activity necessary for axonal signaling.
Collapse
|
66
|
Ma W, Quirion R. Targeting cell surface trafficking of pain-facilitating receptors to treat chronic pain conditions. Expert Opin Ther Targets 2014; 18:459-72. [DOI: 10.1517/14728222.2014.887683] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
67
|
Amaya F, Izumi Y, Matsuda M, Sasaki M. Tissue injury and related mediators of pain exacerbation. Curr Neuropharmacol 2014; 11:592-7. [PMID: 24396335 PMCID: PMC3849785 DOI: 10.2174/1570159x11311060003] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Revised: 06/13/2013] [Accepted: 07/12/2013] [Indexed: 01/03/2023] Open
Abstract
Tissue injury and inflammation result in release of various mediators that promote ongoing pain or pain hypersensitivity against mechanical, thermal and chemical stimuli. Pro-nociceptive mediators activate primary afferent neurons directly or indirectly to enhance nociceptive signal transmission to the central nervous system. Excitation of primary afferents by peripherally originating mediators, so-called “peripheral sensitization”, is a hallmark of tissue injury-related pain. Many kinds of pro-nociceptive mediators, including ATP, glutamate, kinins, cytokines and tropic factors, synthesized at the damaged tissue, contribute to the development of peripheral sensitization. In the present review we will discuss the molecular mechanisms of peripheral sensitization following tissue injury.
Collapse
Affiliation(s)
- Fumimasa Amaya
- Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kajiicho 465, Kamigyo-Ku, Kyoto 602-8566, Japan
| | - Yuta Izumi
- Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kajiicho 465, Kamigyo-Ku, Kyoto 602-8566, Japan
| | - Megumi Matsuda
- Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kajiicho 465, Kamigyo-Ku, Kyoto 602-8566, Japan
| | - Mika Sasaki
- Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kajiicho 465, Kamigyo-Ku, Kyoto 602-8566, Japan
| |
Collapse
|
68
|
Verge VMK, Andreassen CS, Arnason TG, Andersen H. Mechanisms of disease: role of neurotrophins in diabetes and diabetic neuropathy. HANDBOOK OF CLINICAL NEUROLOGY 2014; 126:443-60. [PMID: 25410238 DOI: 10.1016/b978-0-444-53480-4.00032-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Neuropathy is an insidious and devastating consequence of diabetes. Early studies provided a strong rationale for deficient neurotrophin support in the pathogenesis of diabetic neuropathy in a number of critical tissues and organs. It has now been over a decade since the first failed human neurotrophin supplementation clinical trials, but mounting evidence still implicates these trophic factors in diabetic neuropathy. Since then, tremendous advances have been made in our understanding of the complexities of neurotrophin signaling and processing and how the diabetic milieu might impact this. This in turn changes both our perception of how the altered trophic environment contributes to the etiology of diabetic neuropathy and the design of future neurotrophin therapeutic interventions. This chapter summarizes some of these findings and attempts to integrate neurotrophin actions on the nervous system with an increasing appreciation of their role in the regulation of metabolic processes in diabetes that impact the diabetic neuropathic state.
Collapse
Affiliation(s)
- Valerie M K Verge
- Department of Anatomy and Cell Biology, University of Saskatchewan, Saskatoon, Canada; Cameco MS Neuroscience Research Center, University of Saskatchewan, Saskatoon City Hospital, Saskatoon, Canada.
| | - Christer S Andreassen
- Department of Otorhinolaryngology and Head and Neck Surgery, Aarhus University Hospital, Aarhus, Denmark
| | - Terra G Arnason
- Department of Anatomy and Cell Biology, University of Saskatchewan, Saskatoon, Canada; Department of Medicine, Division of Endocrinology and Metabolism, University of Saskatchewan, Saskatoon, Canada
| | - Henning Andersen
- Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
69
|
Wang H, Qin J, Gong S, Feng B, Zhang Y, Tao J. Insulin-like growth factor-1 receptor-mediated inhibition of A-type K(+) current induces sensory neuronal hyperexcitability through the phosphatidylinositol 3-kinase and extracellular signal-regulated kinase 1/2 pathways, independently of Akt. Endocrinology 2014; 155:168-79. [PMID: 24080365 DOI: 10.1210/en.2013-1559] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Although IGF-1 has been implicated in mediating hypersensitivity to pain, the underlying mechanisms remain unclear. We identified a novel functional of the IGF-1 receptor (IGF-1R) in regulating A-type K(+) currents (IA) as well as membrane excitability in small trigeminal ganglion neurons. Our results showed that IGF-1 reversibly decreased IA, whereas the sustained delayed rectifier K(+) current was unaffected. This IGF-1-induced IA decrease was associated with a hyperpolarizing shift in the voltage dependence of inactivation and was blocked by the IGF-1R antagonist PQ-401; an insulin receptor tyrosine kinase inhibitor had no such effect. An small interfering RNA targeting the IGF-1R, or pretreatment of neurons with specific phosphatidylinositol 3-kinase (PI3K) inhibitors abolished the IGF-1-induced IA decrease. Surprisingly, IGF-1-induced effects on IA were not regulated by Akt, a common downstream target of PI3K. The MAPK/ERK kinase inhibitor U0126, but not its inactive analog U0124, as well as the c-Raf-specific inhibitor GW5074, blocked the IGF-1-induced IA response. Analysis of phospho-ERK (p-ERK) showed that IGF-1 significantly activated ERK1/2 whereas p-JNK and p-p38 were unaffected. Moreover, the IGF-1-induced p-ERK1/2 increase was attenuated by PI3K and c-Raf inhibition, but not by Akt blockade. Functionally, we observed a significantly increased action potential firing rate induced by IGF-1; pretreatment with 4-aminopyridine abolished this effect. Taken together, our results indicate that IGF-1 attenuates IA through sequential activation of the PI3K- and c-Raf-dependent ERK1/2 signaling cascade. This occurred via the activation of IGF-1R and might contribute to neuronal hyperexcitability in small trigeminal ganglion neurons.
Collapse
Affiliation(s)
- Hua Wang
- Department of Neurobiology (H.W., S.G., J.T.), Medical College of Soochow University, Suzhou 215123, China; Institute of Neuroscience & Department of Neurology of the Second Affiliated Hospital (J.Q., S.G., Y.Z., J.T.), Soochow University, Suzhou 215004, China; and Department of Endocrinology (H.W., B.F.), East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | | | | | | | | | | |
Collapse
|
70
|
Paltser G, Liu XJ, Yantha J, Winer S, Tsui H, Wu P, Maezawa Y, Cahill LS, Laliberté CL, Ramagopalan SV, DeLuca GC, Sadovnick AD, Astsaturov I, Ebers GC, Henkelman RM, Salter MW, Dosch HM. TRPV1 gates tissue access and sustains pathogenicity in autoimmune encephalitis. Mol Med 2013; 19:149-59. [PMID: 23689362 DOI: 10.2119/molmed.2012.00329] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Accepted: 05/08/2013] [Indexed: 01/03/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic progressive, demyelinating condition whose therapeutic needs are unmet, and whose pathoetiology is elusive. We report that transient receptor potential vanilloid-1 (TRPV1) expressed in a major sensory neuron subset, controls severity and progression of experimental autoimmune encephalomyelitis (EAE) in mice and likely in primary progressive MS. TRPV1-/- B6 congenics are protected from EAE. Increased survival reflects reduced central nervous systems (CNS) infiltration, despite indistinguishable T cell autoreactivity and pathogenicity in the periphery of TRPV1-sufficient and -deficient mice. The TRPV1+ neurovascular complex defining the blood-CNS barriers promoted invasion of pathogenic lymphocytes without the contribution of TRPV1-dependent neuropeptides such as substance P. In MS patients, we found a selective risk-association of the missense rs877610 TRPV1 single nucleotide polymorphism (SNP) in primary progressive disease. Our findings indicate that TRPV1 is a critical disease modifier in EAE, and we identify a predictor of severe disease course and a novel target for MS therapy.
Collapse
Affiliation(s)
- Geoffrey Paltser
- Neuroscience and Mental Health Program, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
71
|
Min JW, Liu WH, He XH, Peng BW. Different types of toxins targeting TRPV1 in pain. Toxicon 2013; 71:66-75. [PMID: 23732125 DOI: 10.1016/j.toxicon.2013.05.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Revised: 05/14/2013] [Accepted: 05/22/2013] [Indexed: 02/06/2023]
Abstract
The transient receptor potential vanilloid 1(TRPV1) channels are members of the transient receptor potential (TRP) superfamily. Members of this family are expressed in primary sensory neurons and are best known for their role in nociception and sensory transmission. Multiple painful stimuli can activate these channels. In this review, we discussed the mechanisms of different types of venoms that target TRPV1, such as scorpion venom, botulinum neurotoxin, spider toxin, ciguatera fish poisoning (CFP) and neurotoxic shellfish poisoning (NSP). Some of these toxins activate TRPV1; however, some do not. Regardless of TRPV1 inhibition or activation, they occur through different pathways. For example, BoNT/A decreases TRPV1 expression levels by blocking TRPV1 trafficking to the plasma membrane, although the exact mechanism is still under debate. Vanillotoxins from tarantula (Psalmopoeus cambridgei) are proposed to activate TRPV1 via interaction with a region of TRPV1 that is homologous to voltage-dependent ion channels. Here, we offer a description of the present state of knowledge for this complex subject.
Collapse
Affiliation(s)
- Jia-Wei Min
- Department of Physiology, School of Basic Medical Sciences, Wuhan University, 185 Donghu Road, Wuhan, Hubei 430071, PR China
| | | | | | | |
Collapse
|
72
|
Sun J, Pu Y, Wang P, Chen S, Zhao Y, Liu C, Shang Q, Zhu Z, Liu D. TRPV1-mediated UCP2 upregulation ameliorates hyperglycemia-induced endothelial dysfunction. Cardiovasc Diabetol 2013; 12:69. [PMID: 23607427 PMCID: PMC3644255 DOI: 10.1186/1475-2840-12-69] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Accepted: 04/17/2013] [Indexed: 12/03/2022] Open
Abstract
Background Diabetic cardiovascular complications are characterised by oxidative stress-induced endothelial dysfunction. Uncoupling protein 2 (UCP2) is a regulator of mitochondrial reactive oxygen species (ROS) generation and can antagonise oxidative stress, but approaches that enhance the activity of UCP2 to inhibit ROS are scarce. Our previous studies show that activation of transient receptor potential vanilloid 1 (TRPV1) by capsaicin can prevent cardiometabolic disorders. In this study, we conducted experiments in vitro and in vivo to investigate the effect of capsaicin treatment on endothelial UCP2 and oxidative stress. We hypothesised that TRPV1 activation by capsaicin attenuates hyperglycemia-induced endothelial dysfunction through a UCP2-mediated antioxidant effect. Methods TRPV1-/-, UCP2 -/- and db/db mice, as well as matched wild type (WT) control mice, were included in this study. Some mice were subjected to dietary capsaicin for 14 weeks. Arteries isolated from mice and endothelial cells were cultured. Endothelial function was examined, and immunohistological and molecular analyses were performed. Results Under high-glucose conditions, TRPV1 expression and protein kinase A (PKA) phosphorylation were found to be decreased in the cultured endothelial cells, and the effects of high-glucose on these molecules were reversed by the administration of capsaicin. Furthermore, high-glucose exposure increased ROS production and reduced nitric oxide (NO) levels both in endothelial cells and in arteries that were evaluated respectively by dihydroethidium (DHE) and DAF-2 DA fluorescence. Capsaicin administration decreased the production of ROS, restored high-glucose-induced endothelial dysfunction through the activation of TRPV1 and acted in a UCP2-dependent manner in vivo. Administration of dietary capsaicin for 14 weeks increased the levels of PKA phosphorylation and UCP2 expression, ameliorated the vascular oxidative stress and increased NO levels observed in diabetic mice. Prolonged dietary administration of capsaicin promoted endothelium-dependent relaxation in diabetic mice. However, the beneficial effect of capsaicin on vasorelaxation was absent in the aortas of UCP2 -/- mice exposed to high-glucose levels. Conclusion TRPV1 activation by capsaicin might protect against hyperglycemia-induced endothelial dysfunction through a mechanism involving the PKA/UCP2 pathway.
Collapse
Affiliation(s)
- Jing Sun
- Center for Hypertension and Metabolic Diseases, Department of Hypertension and Endocrinology, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing 400042, China
| | | | | | | | | | | | | | | | | |
Collapse
|
73
|
Rios ERV, Rocha NFM, Carvalho AMR, Vasconcelos LF, Dias ML, de Sousa DP, de Sousa FCF, Fonteles MMDF. TRP and ASIC channels mediate the antinociceptive effect of citronellyl acetate. Chem Biol Interact 2013; 203:573-9. [PMID: 23562495 DOI: 10.1016/j.cbi.2013.03.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Revised: 03/03/2013] [Accepted: 03/16/2013] [Indexed: 01/28/2023]
Abstract
BACKGROUND Citronellyl acetate (CAT), a monoterpene product of the secondary metabolism of plants, has been shown in the literature to possess several different biological activities. However, no antinociceptive abilities have yet been discussed. Here, we used acute pain animal models to describe the antinociceptive action of CAT. METHODS The acetic acid-induced writhing test and the paw-licking test, in which paw licking was induced by glutamate and formalin, were performed to evaluate the antinociceptive action of CAT and to determine the involvement of PKC, PKA, TRPV1, TRPA1, TRPM8 and ASIC in its antinociceptive mechanism. To do so, we induced paw-linking using agonists. RESULTS CAT was administered intragastrically (25, 50, 75, 100 and 200 mg/kg), and the two higher doses caused antinociceptive effects in the acetic acid model; the highest dose reduced pain for 4h after it was administered (200 mg/kg). In the formalin test, two doses of CAT promoted antinociception in both the early and later phases of the test. The glutamate test showed that its receptors are involved in the antinociceptive mechanism of CAT. Pretreatment with CAT did not alter locomotor activity or motor coordination. In an investigation into the participation of TRP channels and ASICs in CAT's antinociceptive mechanism, we used capsaicin (2.2 μg/paw), cinnamaldehyde (10 mmol/paw), menthol (1.2 mmol/paw) and acidified saline (2% acetic acid, pH 1.98). The results showed that TRPV1, TRPM8 and ASIC, but not TRPA1, are involved in the antinociceptive mechanism. Finally, the involvement of PKC and PKA was also studied, and we showed that both play a role in the antinociceptive mechanism of CAT. CONCLUSION The results of this work contribute information regarding the antinociceptive properties of CAT on acute pain and show that, at least in part, TRPV1, TRPM8, ASIC, glutamate receptors, PKC and PKA participate in CAT's antinociceptive mechanism.
Collapse
|
74
|
Kaßmann M, Harteneck C, Zhu Z, Nürnberg B, Tepel M, Gollasch M. Transient receptor potential vanilloid 1 (TRPV1), TRPV4, and the kidney. Acta Physiol (Oxf) 2013; 207:546-64. [PMID: 23253200 DOI: 10.1111/apha.12051] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Revised: 11/15/2012] [Accepted: 12/11/2012] [Indexed: 12/13/2022]
Abstract
Recent preclinical data indicate that activators of transient receptor potential channels of the vanilloid receptor subtype 1 (TRPV1) may improve the outcome of ischaemic acute kidney injury (AKI). The underlying mechanisms are unclear, but may involve TRPV1 channels in dorsal root ganglion neurones that innervate the kidney. Recent data identified TRPV4, together with TRPV1, to serve as major calcium influx channels in endothelial cells. In these cells, gating of individual TRPV4 channels within a four-channel cluster provides elementary calcium influx (calcium sparklets) to open calcium-activated potassium channels and promote vasodilation. The TRPV receptors can also form heteromers that exhibit unique conductance and gating properties, further increasing their spatio-functional diversity. This review summarizes data on electrophysiological properties of TRPV1/4 and their modulation by endogenous channel agonists such as 20-HETE, phospholipase C and phosphatidylinositide 3-kinase (PI3 kinase). We review important roles of TRPV1 and TRPV4 in kidney physiology and renal ischaemia reperfusion injury; further studies are warranted to address renoprotective mechanism of vanilloid receptors in ischaemic AKI including the role of the capsaicin receptor TRPV1 in primary sensory nerves and/or endothelium. Particular attention should be paid to understand the kidneys' ability to respond to ischaemic stimuli after catheter-based renal denervation therapy in man, whereas the discovery of novel pharmacological TRPV modulators may be a successful strategy for better treatment of acute or chronic kidney failure.
Collapse
Affiliation(s)
- M. Kaßmann
- Charité University Medicine, Section Nephrology/Intensive Care, Campus Virchow, and Experimental and Clinical Research Center (ECRC); Berlin; Germany
| | - C. Harteneck
- Institut für Experimentelle & Klinische Pharmakologie & Toxikologie and Interfaculty Center of Pharmacogenomics and Pharmaceutical Research (ICePhA); Eberhard-Karls-Universität; Tübingen; Germany
| | - Z. Zhu
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases; Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension; Chongqing; China
| | - B. Nürnberg
- Institut für Experimentelle & Klinische Pharmakologie & Toxikologie and Interfaculty Center of Pharmacogenomics and Pharmaceutical Research (ICePhA); Eberhard-Karls-Universität; Tübingen; Germany
| | - M. Tepel
- Department of Nephrology, and University of Southern Denmark, Institute of Molecular Medicine, Cardiovascular and Renal Research, Institute of Clinical Research; Odense University Hospital; Odense; Denmark
| | - M. Gollasch
- Charité University Medicine, Section Nephrology/Intensive Care, Campus Virchow, and Experimental and Clinical Research Center (ECRC); Berlin; Germany
| |
Collapse
|
75
|
Cyclin-dependent kinase 5 controls TRPV1 membrane trafficking and the heat sensitivity of nociceptors through KIF13B. J Neurosci 2013; 32:14709-21. [PMID: 23077056 DOI: 10.1523/jneurosci.1634-12.2012] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The number of functional transient receptor potential vanilloid 1 (TRPV1) channels at the surface, especially at the peripheral terminals of primary sensory neurons, regulates heat sensitivity, and increased surface localization of TRPV1s contributes to heat hyperalgesia. However, the mechanisms for regulating TRPV1 surface localization are essentially unknown. Here, we show that cyclin-dependent kinase 5 (Cdk5), a new player in thermal pain sensation, positively regulates TRPV1 surface localization. Active Cdk5 was found to promote TRPV1 anterograde transport in vivo, suggesting a regulatory role of Cdk5 in TRPV1 membrane trafficking. TRPV1-containing vesicles bind to the forkhead-associated (FHA) domain of the KIF13B (kinesin-3 family member 13B) and are thus delivered to the cell surface. Overexpression of Cdk5 or its activator p35 promoted and inhibition of Cdk5 activity prevented the KIF13B-TRPV1 association, indicating that Cdk5 promotes TRPV1 anterograde transport by mediating the motor-cargo association. Cdk5 phosphorylates KIF13B at Thr-506, a residue located in the FHA domain. T506A mutation reduced the motor-cargo interaction and the cell-permeable TAT-T506 peptide, targeting to the Thr-506, decreased TRPV1 surface localization, demonstrating the essential role of Thr-506 phosphorylation in TRPV1 transport. Moreover, complete Freund's adjuvant (CFA) injection-induced activation of Cdk5 increased the anterograde transport of TRPV1s, contributing to the development and possibly the maintenance of heat hyperalgesia, whereas intrathecal delivery of the TAT-T506 peptide alleviated CFA-induced heat hyperalgesia in rats. Thus, Cdk5 regulation of TRPV1 membrane trafficking is a fundamental mechanism controlling the heat sensitivity of nociceptors, and moderate inhibition of Thr-506 phosphorylation during inflammation might be helpful for the treatment of inflammatory thermal pain.
Collapse
|
76
|
Avelino A, Charrua A, Frias B, Cruz C, Boudes M, de Ridder D, Cruz F. Transient receptor potential channels in bladder function. Acta Physiol (Oxf) 2013; 207:110-22. [PMID: 23113869 DOI: 10.1111/apha.12021] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2012] [Revised: 01/27/2012] [Accepted: 09/10/2012] [Indexed: 01/17/2023]
Abstract
The transient receptor potential (TRP) superfamily of cationic ion channels includes proteins involved in the transduction of several physical and chemical stimuli to finely tune physiological functions. In the urinary bladder, they are highly expressed in, but not restricted to, primary afferent neurons. The urothelium and some interstitial cells also express several TRP channels. In this review, we describe the expression and the known roles of some members of TRP subfamilies, namely TRPV, TRPM and TRPA, in the urinary bladder. The therapeutic interest of modulating the activity of TRP channels to treat bladder dysfunctions is also discussed.
Collapse
Affiliation(s)
- A. Avelino
- Department of Experimental Biology; Faculty of Medicine of University of Porto; Porto; Portugal
| | | | | | | | | | - D. de Ridder
- Department of Molecular Cell Biology; Laboratory Ion Channel Research; KU Leuven; Leuven; Belgium
| | | |
Collapse
|
77
|
Reduction of TRPV1 expression in the trigeminal system by botulinum neurotoxin type-A. Neurobiol Dis 2012; 48:367-78. [DOI: 10.1016/j.nbd.2012.07.010] [Citation(s) in RCA: 97] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Revised: 06/30/2012] [Accepted: 07/09/2012] [Indexed: 01/30/2023] Open
|
78
|
Vay L, Gu C, McNaughton PA. Current perspectives on the modulation of thermo-TRP channels: new advances and therapeutic implications. Expert Rev Clin Pharmacol 2012; 3:687-704. [PMID: 22111750 DOI: 10.1586/ecp.10.41] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The thermo transient receptor potential (TRP) ion channels, a recently discovered family of ion channels activated by temperature, are expressed in primary sensory nerve terminals, where they provide information regarding thermal changes in the environment. Six thermo-TRPs have been characterized to date: TRPV1-4, which respond to different levels of warmth and heat, and TRPM8 and TRPA1, which respond to cool temperatures. We review the current state of knowledge of thermo-TRPs, and of the modulation of their thermal thresholds by a range of inflammatory mediators. Blockers of these channels are likely to have therapeutic uses as novel analgesics but may also cause unacceptable side effects. Controlling the modulation of thermo-TRPs by inflammatory mediators may be a useful alternative strategy in developing novel analgesics.
Collapse
Affiliation(s)
- Laura Vay
- Deptartment of Pharmacology, University of Cambridge, Tennis Court Rd, Cambridge, CB2 1PD, UK
| | | | | |
Collapse
|
79
|
Premkumar LS, Abooj M. TRP channels and analgesia. Life Sci 2012; 92:415-24. [PMID: 22910182 DOI: 10.1016/j.lfs.2012.08.010] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2012] [Revised: 07/25/2012] [Accepted: 08/02/2012] [Indexed: 01/07/2023]
Abstract
Since cloning and characterizing the first nociceptive ion channel Transient Receptor Potential (TRP) Vanilloid 1 (TRPV1), other TRP channels involved in nociception have been cloned and characterized, which include TRP Vanilloid 2 (TRPV2), TRP Vanilloid 3 (TRPV3), TRP Vanilloid 4 (TRPV4), TRP Ankyrin 1 (TRPA1) and TRP Melastatin 8 (TRPM8), more recently TRP Canonical 1, 5, 6 (TRPC1, 5, 6), TRP Melastatin 2 (TRPM2) and TRP Melastatin 3 (TRPM3). These channels are predominantly expressed in C and Aδ nociceptors and transmit noxious thermal, mechanical and chemical sensitivities. TRP channels are modulated by pro-inflammatory mediators, neuropeptides and cytokines. Significant advances have been made targeting these receptors either by antagonists or agonists to treat painful conditions. In this review, we will discuss TRP channels as targets for next generation analgesics and the side effects that may ensue as a result of blocking/activating these receptors, because they are also involved in physiological functions such as release of vasoactive neuropeptides and regulation of vascular tone, maintenance of the body temperature, gastrointestinal motility, urinary bladder control, etc.
Collapse
Affiliation(s)
- Louis S Premkumar
- Department of Pharmacology, Southern Illinois University School of Medicine Springfield, IL 62702, USA.
| | | |
Collapse
|
80
|
Zhao Y, Huang H, Jiang Y, Wei H, Liu P, Wang W, Niu W. Unusual localization and translocation of TRPV4 protein in cultured ventricular myocytes of the neonatal rat. Eur J Histochem 2012; 56:e32. [PMID: 23027348 PMCID: PMC3493978 DOI: 10.4081/ejh.2012.e32] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Revised: 05/25/2012] [Accepted: 05/30/2012] [Indexed: 12/02/2022] Open
Abstract
TRPV4 protein forms a Ca2+-permeable channel that is sensitive to osmotic and mechanical stimuli and responds to warm temperatures, and expresses widely in various kinds of tissues. As for cardiac myocytes, TRPV4 has been detected only at the mRNA level and there were few reports about subcel-lular localization of the protein. The purpose of the present study was to investigate the expression profile of TRPV4 protein in cultured neonatal rat ventricular myocytes. Using Western blots, immunofluorescence, confocal microscopy and immuno-electron microscopy, we have shown that TRPV4 protein was predominantly located in the nucleus of cultured neonatal myocytes. Furthermore, cardiac myocytes responded to hypotonic stimulation by translocating TRPV4 protein out of the nucleus. The significance and mechanism concerning the unusual distribution and translocation of TRPV4 protein in cardiac myocytes remain to be clarified.
Collapse
Affiliation(s)
- Y Zhao
- Department of Physiology and Pathophysiology, Capital Medical University, Beijing, China
| | | | | | | | | | | | | |
Collapse
|
81
|
Ponsati B, Carreño C, Curto-Reyes V, Valenzuela B, Duart MJ, Van den Nest W, Cauli O, Beltran B, Fernandez J, Borsini F, Caprioli A, Di Serio S, Veretchy M, Baamonde A, Menendez L, Barros F, de la Pena P, Borges R, Felipo V, Planells-Cases R, Ferrer-Montiel A. An inhibitor of neuronal exocytosis (DD04107) displays long-lasting in vivo activity against chronic inflammatory and neuropathic pain. J Pharmacol Exp Ther 2012; 341:634-45. [PMID: 22393248 DOI: 10.1124/jpet.111.190678] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Small peptides patterned after the N terminus of the synaptosomal protein of 25 kDa, a member of the protein complex implicated in Ca(2+)-dependent neuronal exocytosis, inhibit in vitro the release of neuromodulators involved in pain signaling, suggesting an in vivo analgesic activity. Here, we report that compound DD04107 (palmitoyl-EEMQRR-NH(2)), a 6-mer palmitoylated peptide that blocks the inflammatory recruitment of ion channels to the plasma membrane of nociceptors and the release of calcitonin gene-related peptide from primary sensory neurons, displays potent and long-lasting in vivo antihyperalgesia and antiallodynia in chronic models of inflammatory and neuropathic pain, such as the complete Freund's adjuvant, osteosarcoma, chemotherapy, and diabetic neuropathic models. Subcutaneous administration of the peptide produced a dose-dependent antihyperalgesic and antiallodynic activity that lasted ≥24 h. The compound showed a systemic distribution, characterized by a bicompartmental pharmacokinetic profile. Safety pharmacology studies indicated that the peptide is largely devoid of side effects and substantiated that the in vivo activity is not caused by locomotor impairment. Therefore, DD04107 is a potent and long-lasting antinociceptive compound that displays a safe pharmacological profile. These findings support the notion that neuronal exocytosis of receptors and neuronal algogens pivotally contribute to chronic inflammatory and neuropathic pain and imply a central role of peptidergic nociceptor sensitization to the pathogenesis of pain.
Collapse
|
82
|
Gao H, Miyata K, Bhaskaran MD, Derbenev AV, Zsombok A. Transient receptor potential vanilloid type 1-dependent regulation of liver-related neurons in the paraventricular nucleus of the hypothalamus diminished in the type 1 diabetic mouse. Diabetes 2012; 61:1381-90. [PMID: 22492526 PMCID: PMC3357291 DOI: 10.2337/db11-0820] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The paraventricular nucleus (PVN) of the hypothalamus controls the autonomic neural output to the liver, thereby participating in the regulation of hepatic glucose production (HGP); nevertheless, mechanisms controlling the activity of liver-related PVN neurons are not known. Transient receptor potential vanilloid type 1 (TRPV1) is involved in glucose homeostasis and colocalizes with liver-related PVN neurons; however, the functional role of TRPV1 regarding liver-related PVN neurons has to be elucidated. A retrograde viral tracer was used to identify liver-related neurons within the brain-liver circuit in control, type 1 diabetic, and insulin-treated mice. Our data indicate that TRPV1 regulates liver-related PVN neurons. This TRPV1-dependent excitation diminished in type 1 diabetic mice. In vivo and in vitro insulin restored TRPV1 activity in a phosphatidylinositol 3-kinase/protein kinase C-dependent manner and stimulated TRPV1 receptor trafficking to the plasma membrane. There was no difference in total TRPV1 protein expression; however, increased phosphorylation of TRPV1 receptors was observed in type 1 diabetic mice. Our data demonstrate that TRPV1 plays a pivotal role in the regulation of liver-related PVN neurons. Moreover, TRPV1-dependent excitation of liver-related PVN neurons diminishes in type 1 diabetes, thus indicating that the brain-liver autonomic circuitry is altered in type 1 diabetes and may contribute to the autonomic dysfunction of HGP.
Collapse
Affiliation(s)
- Hong Gao
- Department of Physiology, Tulane University, School of Medicine, New Orleans, Louisiana, USA.
| | | | | | | | | |
Collapse
|
83
|
Ho KW, Ward NJ, Calkins DJ. TRPV1: a stress response protein in the central nervous system. AMERICAN JOURNAL OF NEURODEGENERATIVE DISEASE 2012; 1:1-14. [PMID: 22737633 PMCID: PMC3560445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 04/15/2012] [Accepted: 04/21/2012] [Indexed: 06/01/2023]
Abstract
The transient receptor potential (TRP) family comprises a diverse group of cation channels that regulate a variety of intracellular signaling pathways. The TRPV1 (vanilloid 1) channel is best known for its role in nociception and sensory transmission. First studied in the dorsal root ganglia as the receptor for capsaicin, TRPV1 is now recognized to have a broader distribution and function within the central nervous system (CNS). Because it can be activated by a range of potentially noxious stimuli, TRPV1's polymodal nature and ability to interact with other receptor pathways make it a candidate for a stress response protein. As a result, TRPV1 is emerging as a key mediator of CNS function through modulation of both glial and neuronal activity. Growing evidence has suggested that TRPV1 can mediate a variety of pathways from glial reactivity and cytokine release to synaptic transmission and plasticity. This review highlights the increasing importance of TRPV1 as a regulator of CNS function in response to stress.
Collapse
Affiliation(s)
- Karen W Ho
- The Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN 37205, USA
| | | | | |
Collapse
|
84
|
Vay L, Gu C, McNaughton PA. The thermo-TRP ion channel family: properties and therapeutic implications. Br J Pharmacol 2012; 165:787-801. [PMID: 21797839 PMCID: PMC3312478 DOI: 10.1111/j.1476-5381.2011.01601.x] [Citation(s) in RCA: 215] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Revised: 04/13/2011] [Accepted: 05/01/2011] [Indexed: 01/09/2023] Open
Abstract
The thermo-transient receptor potentials (TRPs), a recently discovered family of ion channels activated by temperature, are expressed in primary sensory nerve terminals where they provide information about thermal changes in the environment. Six thermo-TRPs have been characterised to date: TRP vanilloid (TRPV) 1 and 2 are activated by painful levels of heat, TRPV3 and 4 respond to non-painful warmth, TRP melastatin 8 is activated by non-painful cool temperatures, while TRP ankyrin (TRPA) 1 is activated by painful cold. The thermal thresholds of many thermo-TRPs are known to be modulated by extracellular mediators, released by tissue damage or inflammation, such as bradykinin, PG and growth factors. There have been intensive efforts recently to develop antagonists of thermo-TRP channels, particularly of the noxious thermal sensors TRPV1 and TRPA1. Blockers of these channels are likely to have therapeutic uses as novel analgesics, but may also cause unacceptable side effects. Controlling the modulation of thermo-TRPs by inflammatory mediators may be a useful alternative strategy in developing novel analgesics.
Collapse
Affiliation(s)
- Laura Vay
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | | | | |
Collapse
|
85
|
New strategies to develop novel pain therapies: addressing thermoreceptors from different points of view. Pharmaceuticals (Basel) 2011; 5:16-48. [PMID: 24288041 PMCID: PMC3763626 DOI: 10.3390/ph5010016] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Revised: 12/16/2011] [Accepted: 12/21/2011] [Indexed: 01/23/2023] Open
Abstract
One approach to develop successful pain therapies is the modulation of dysfunctional ion channels that contribute to the detection of thermal, mechanical and chemical painful stimuli. These ion channels, known as thermoTRPs, promote the sensitization and activation of primary sensory neurons known as nociceptors. Pharmacological blockade and genetic deletion of thermoTRP have validated these channels as therapeutic targets for pain intervention. Several thermoTRP modulators have progressed towards clinical development, although most failed because of the appearance of unpredicted side effects. Thus, there is yet a need to develop novel channel modulators with improved therapeutic index. Here, we review the current state-of-the art and illustrate new pharmacological paradigms based on TRPV1 that include: (i) the identification of activity-dependent modulators of this thermoTRP channel; (ii) the design of allosteric modulators that interfere with protein-protein interaction involved in the functional coupling of stimulus sensing and gate opening; and (iii) the development of compounds that abrogate the inflammation-mediated increase of receptor expression in the neuronal surface. These new sites of action represent novel strategies to modulate pathologically active TRPV1, while minimizing an effect on the TRPV1 subpopulation involved in physiological and protective roles, thus increasing their potential therapeutic use.
Collapse
|
86
|
Functional plasticity of central TRPV1 receptors in brainstem dorsal vagal complex circuits of streptozotocin-treated hyperglycemic mice. J Neurosci 2011; 31:14024-31. [PMID: 21957263 DOI: 10.1523/jneurosci.2081-11.2011] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Emerging data indicate that central neurons participate in diabetic processes by modulating autonomic output from neurons in the dorsal motor nucleus of the vagus (DMV). We tested the hypothesis that synaptic modulation by transient receptor potential vanilloid type 1 (TRPV1) receptors is reduced in the DMV in slices from a murine model of type 1 diabetes. The TRPV1 agonist capsaicin robustly enhanced glutamate release onto DMV neurons by acting at preterminal receptors in slices from intact mice, but failed to do so in slices from diabetic mice. TRPV1 receptor protein expression in the vagal complex was unaltered. Brief insulin preapplication restored TRPV1-dependent modulation of glutamate release in a PKC- and PI3K-dependent manner. The restorative effect of insulin was prevented by brefeldin A, suggesting that insulin induced TRPV1 receptor trafficking to the terminal membrane. Central vagal circuits critical to the autonomic regulation of metabolism undergo insulin-dependent synaptic plasticity involving TRPV1 receptor modulation in diabetic mice after several days of chronic hyperglycemia.
Collapse
|
87
|
Tsui H, Paltser G, Chan Y, Dorfman R, Dosch HM. 'Sensing' the link between type 1 and type 2 diabetes. Diabetes Metab Res Rev 2011; 27:913-8. [PMID: 22069284 DOI: 10.1002/dmrr.1279] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Obesity-associated insulin resistance is a core element of metabolic syndrome and type 2 diabetes (T2D). Notably, insulin resistance is also a feature of type 1 diabetes (T1D), where findings in the non-obese diabetic mouse model have implicated transient receptor potential vanilloid-1 (TRPV1+) sensory neurons in local islet inflammation and glucose metabolism. Here, we briefly review the role of TRPV1 in non-obese diabetic (NOD) T1D pathogenesis, highlighting commonalities that suggest TRPV1 may contribute to obesity and T2D as well. With the recently discovered importance of adipose infiltrating lymphocytes in the metabolic disturbances of obesity and T2D, sensory innervation of fat may thus play an analogous role to sensory neurons in the islet--modulating neuroendocrine homeostasis and inflammation. In such a scenario, TRPV1+ sensory nerves would provide the pathoaetiological link connecting the shared metabolic and immunologic features of type 1 diabetes and T2D.
Collapse
Affiliation(s)
- Hubert Tsui
- The Research Institute, Hospital For Sick Children, University of Toronto, Neuroscience and Mental Health Program, Toronto, ON M5G 1X8, Canada.
| | | | | | | | | |
Collapse
|
88
|
Richter D, Katz B, Oberacker T, Tzarfaty V, Belusic G, Minke B, Huber A. Translocation of the Drosophila transient receptor potential-like (TRPL) channel requires both the N- and C-terminal regions together with sustained Ca2+ entry. J Biol Chem 2011; 286:34234-43. [PMID: 21816824 PMCID: PMC3190804 DOI: 10.1074/jbc.m111.278564] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Revised: 07/29/2011] [Indexed: 11/06/2022] Open
Abstract
In Drosophila photoreceptors the transient receptor potential-like (TRPL), but not the TRP channels undergo light-dependent translocation between the rhabdomere and cell body. Here we studied which of the TRPL channel segments are essential for translocation and why the TRP channels are required for inducing TRPL translocation. We generated transgenic flies expressing chimeric TRP and TRPL proteins that formed functional light-activated channels. Translocation was induced only in chimera containing both the N- and C-terminal segments of TRPL. Using an inactive trp mutation and overexpressing the Na(+)/Ca(2+) exchanger revealed that the essential function of the TRP channels in TRPL translocation is to enhance Ca(2+)-influx. These results indicate that motifs present at both the N and C termini as well as sustained Ca(2+) entry are required for proper channel translocation.
Collapse
Affiliation(s)
- David Richter
- From the Department of Biosensorics, Institute of Physiology, University of Hohenheim, 70599 Stuttgart, Germany
| | - Ben Katz
- the Department of Medical Neurobiology and The Kühne Minerva Center for Studies of Visual Transduction, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel, and
| | - Tina Oberacker
- From the Department of Biosensorics, Institute of Physiology, University of Hohenheim, 70599 Stuttgart, Germany
| | - Vered Tzarfaty
- the Department of Medical Neurobiology and The Kühne Minerva Center for Studies of Visual Transduction, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel, and
| | - Gregor Belusic
- the Department of Biology, Biotechnical Faculty, University of Ljubljana 1000, Slovenia
| | - Baruch Minke
- the Department of Medical Neurobiology and The Kühne Minerva Center for Studies of Visual Transduction, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel, and
| | - Armin Huber
- From the Department of Biosensorics, Institute of Physiology, University of Hohenheim, 70599 Stuttgart, Germany
| |
Collapse
|
89
|
Fiorio Pla A, Avanzato D, Munaron L, Ambudkar IS. Ion channels and transporters in cancer. 6. Vascularizing the tumor: TRP channels as molecular targets. Am J Physiol Cell Physiol 2011; 302:C9-15. [PMID: 21832241 DOI: 10.1152/ajpcell.00280.2011] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Tumor vascularization is a critical process that determines tumor growth and metastasis. In the last decade new experimental evidence obtained from in vitro and in vivo studies have challenged the classical angiogenesis model forcing us to consider new scenarios for tumor neovascularization. In particular, the genetic stability of tumor-derived endothelial cells (TECs) has been recently questioned in several studies, which show that TECs, as well as pericytes, differ significantly from their normal counterparts at genetic and functional levels. In addition to such an epigenetic action of tumor microenvironment on endothelial cells (ECs) commitment, the distinct characteristics of TECs could be due to differences in their origin compared with preexisting differentiated ECs. Intracellular Ca(2+) signals are involved at different critical phases in the regulation of the complex process of angiogenesis and tumor progression. These signals are generated by a wide variety of intrinsic and extrinsic factors. Several key components of Ca(2+) signaling including Ca(2+) channels in the plasma membrane, endoplasmic reticulum, calcium pumps, and mitochondria contribute to the generation, amplitude, and frequency of these Ca(2+) change. In particular, several members of the transient receptor potential (TRP) family of calcium-permeable channels have profound effects on the function of ECs. Because of its multifaceted role in the control of cell function, proliferation, and motility, TRP channels have been suggested as a potential molecular target for control of tumor neovascularization. Since plasma membrane Ca(2+) channels are easily and directly accessible via the bloodstream, they are potential targets for a number of pharmacological and antibody-targeted therapeutic strategies, with specificity being the main limitation. In this review we discuss recent advances in understanding the role of Ca(2+) channels, with specific reference to TRP channels, in tumor vascularization process.
Collapse
|
90
|
Fiorio Pla A, Ong HL, Cheng KT, Brossa A, Bussolati B, Lockwich T, Paria B, Munaron L, Ambudkar IS. TRPV4 mediates tumor-derived endothelial cell migration via arachidonic acid-activated actin remodeling. Oncogene 2011; 31:200-12. [PMID: 21685934 DOI: 10.1038/onc.2011.231] [Citation(s) in RCA: 137] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Changes in intracellular calcium [Ca(2+)](i) levels control critical cytosolic and nuclear events that are involved in the initiation and progression of tumor angiogenesis in endothelial cells (ECs). Therefore, the mechanism(s) involved in agonist-induced Ca(2+)(i) signaling is a potentially important molecular target for controlling angiogenesis and tumor growth. Several studies have shown that blood vessels in tumors differ from normal vessels in their morphology, blood flow and permeability. We had previously reported a key role for arachidonic acid (AA)-mediated Ca(2+) entry in the initial stages of tumor angiogenesis in vitro. In this study we assessed the mechanism involved in AA-induced EC migration. We report that TRPV4, an AA-activated channel, is differentially expressed in EC derived from human breast carcinomas (BTEC) as compared with 'normal' EC (HMVEC). BTEC display a significant increase in TRPV4 expression, which was correlated with greater Ca(2+) entry, induced by AA or 4αPDD (a selective TRPV4 agonist) in the tumor-derived ECs. Wound-healing assays revealed a key role of TRPV4 in regulating cell migration of BTEC but not HMVEC. Knockdown of TRPV4 expression completely abolished AA-induced BTEC migration, suggesting that TRPV4 mediates the pro-angiogenic effects promoted by AA. Furthermore, pre-incubation of BTEC with AA induced actin remodeling and a subsequent increase in the surface expression of TRPV4. This was consistent with the increased plasma membrane localization of TRPV4 and higher AA-stimulated Ca(2+) entry in the migrating cells. Together, the data presented herein demonstrate that: (1) TRPV4 is differentially expressed in tumor-derived versus 'normal' EC; (2) TRPV4 has a critical role in the migration of tumor-derived but not 'normal' EC migration; and (3) AA induces actin remodeling in BTEC, resulting in a corresponding increase of TRPV4 expression in the plasma membrane. We suggest that the latter is critical for migration of EC and thus in promoting angiogenesis and tumor growth.
Collapse
Affiliation(s)
- A Fiorio Pla
- Department of Animal and Human Biology, University of Torino, Torino, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
91
|
Devesa I, Planells-Cases R, Fernández-Ballester G, González-Ros JM, Ferrer-Montiel A, Fernández-Carvajal A. Role of the transient receptor potential vanilloid 1 in inflammation and sepsis. J Inflamm Res 2011; 4:67-81. [PMID: 22096371 PMCID: PMC3218746 DOI: 10.2147/jir.s12978] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The transient receptor potential vanilloid 1 (TRPV1) is a thermoreceptor that responds to noxious temperatures, as well as to chemical agonists, such as vanilloids and protons. In addition, its channel activity is notably potentiated by proinflammatory mediators released upon tissue damage. The TRPV1 contribution to sensory neuron sensitization by proalgesic agents has signaled this receptor as a prime target for analgesic and anti-inflammatory drug intervention. However, TRPV1 antagonists have notably failed in clinical and preclinical studies because of their unwanted side effects. Recent reports have unveiled previously unrecognized anti-inflammatory and protective functions of TRPV1 in several diseases. For instance, this channel has been suggested to play an anti-inflammatory role in sepsis. Therefore, the use of potent TRPV1 antagonists as a general strategy to treat inflammation must be cautiously considered, given the deleterious effects that may arise from inhibiting the population of channels that have a protective function. The use of TRPV1 antagonists may be limited to treating those pathologies where enhanced receptor activity contributes to the inflamed state. Alternatively, therapeutic paradigms, such as reduction of inflammatory-mediated increase of receptor expression in the cell surface, may be a better strategy to prevent abrogation of the TRPV1 subpopulation involved in anti-inflammatory and protective processes.
Collapse
Affiliation(s)
- Isabel Devesa
- Instituto de Biología Molecular y Celular, Universidad Miguel Hernández, Alicante
| | | | | | | | | | | |
Collapse
|
92
|
Zsombok A, Gao H, Miyata K, Issa A, Derbenev AV. Immunohistochemical localization of transient receptor potential vanilloid type 1 and insulin receptor substrate 2 and their co-localization with liver-related neurons in the hypothalamus and brainstem. Brain Res 2011; 1398:30-9. [PMID: 21620379 DOI: 10.1016/j.brainres.2011.04.048] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2011] [Revised: 04/29/2011] [Accepted: 04/29/2011] [Indexed: 02/05/2023]
Abstract
The central nervous system plays an important role in the regulation of energy balance and glucose homeostasis mainly via controlling the autonomic output to the visceral organs. The autonomic output is regulated by hormones and nutrients to maintain adequate energy and glucose homeostasis. Insulin action is mediated via insulin receptors (IR) resulting in phosphorylation of insulin receptor substrates (IRS) inducing activation of downstream pathways. Furthermore, insulin enhances transient receptor potential vanilloid type 1 (TRPV1) mediated currents. Activation of the TRPV1 receptor increases excitatory neurotransmitter release in autonomic centers of the brain, thereby impacting energy and glucose homeostasis. The aim of this study is to determine co-expression of IRS2 and TRPV1 receptors in the paraventricular nucleus of the hypothalamus (PVN) and dorsal motor nucleus of the vagus (DMV) in the mouse brain as well as expression of IRS2 and TRPV1 receptors at liver-related preautonomic neurons pre-labeled with a trans-neural, viral tracer (PRV-152). The data indicate that IRS2 and TRPV1 receptors are present and co-express in the PVN and the DMV. A large portion (over 50%) of the liver-related preautonomic DMV and PVN neurons expresses IRS2. Moreover, the majority of liver-related DMV and PVN neurons also express TRPV1 receptors, suggesting that insulin and TRPV1 actions may affect liver-related preautonomic neurons.
Collapse
Affiliation(s)
- Andrea Zsombok
- Department of Physiology, Tulane University Health Science Center, New Orleans, LA 70112, USA.
| | | | | | | | | |
Collapse
|
93
|
Miura M, Sasaki M, Mizukoshi K, Shibasaki M, Izumi Y, Shimosato G, Amaya F. Peripheral sensitization caused by insulin-like growth factor 1 contributes to pain hypersensitivity after tissue injury. Pain 2011; 152:888-895. [PMID: 21296499 DOI: 10.1016/j.pain.2011.01.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2010] [Revised: 12/03/2010] [Accepted: 01/04/2011] [Indexed: 01/25/2023]
Abstract
Sensitization of primary afferent neurons is one of the most important components of pain hypersensitivity after tissue injury. Insulin-like growth factor 1 (IGF-1), involved in wound repair in injured tissue, also plays an important role in maintaining neuronal function. In the present study, we investigated the effect of tissue IGF-1 on nociceptive sensitivity of primary afferent neurons. Local administration of IGF-1 induced thermal and mechanical pain hypersensitivity in a dose-dependent manner, and was attenuated by IGF-1 receptor (IGF1R) inhibition. Tissue but not plasma IGF-1 levels, as determined by enzyme-linked immunosorbent assay, significantly increased after plantar incision. Immunohistochemistry revealed that IGF1R was predominantly expressed in neurons as well as in satellite glial cells in the dorsal root ganglion (DRG). Double-labeling immunohistochemistry showed that IGF1R expression colocalized with peripherin and TRPV1, but not with NF200 in DRG neurons. The IGF1R inhibitor successfully alleviated mechanical allodynia, heat hyperalgesia, and spontaneous pain behavior observed after plantar incision. Expression of phosphorylated Akt in DRG neurons significantly increased after plantar incision and was suppressed by IGF1R inhibition. These results demonstrate that increased tissue IGF-1 production sensitizes primary afferent neurons via the IGF1R/Akt pathway to facilitate pain hypersensitivity after tissue damage.
Collapse
Affiliation(s)
- Mayumi Miura
- Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kyoto, Japan Pain Mechanism Research Group, Kyoto, Japan
| | | | | | | | | | | | | |
Collapse
|
94
|
Planells-Cases R, Valente P, Ferrer-Montiel A, Qin F, Szallasi A. Complex regulation of TRPV1 and related thermo-TRPs: implications for therapeutic intervention. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 704:491-515. [PMID: 21290313 DOI: 10.1007/978-94-007-0265-3_27] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The capsaicin receptor TRPV1 (Transient Receptor Potential, Vanilloid family member 1), the founding member of the heat-sensitive TRP ("thermo-TRP") channel family, plays a pivotal role in pain transduction. There is mounting evidence that TRPV1 regulation is complex and is manifest at many levels, from gene expression through post-translational modification and formation of receptor heteromers to subcellular compartmentalization and association with regulatory proteins. These mechanisms are believed to be involved both in disease-related changes in TRPV1 expression, and the long-lasting refractory state, referred to as "desensitization", that follows TRPV1 agonist treatment. The signaling cascades that regulate TRPV1 and related thermo-TRP channels are only beginning to be understood. Here we review our current knowledge in this rapidly changing field. We propose that the complex regulation of TRPV1 may be exploited for therapeutic purposes, with the ultimate goal being the development of novel, innovative agents that target TRPV1 in diseased, but not healthy, tissues. Such compounds are expected to be devoid of the side-effects (e.g. hyperthermia and impaired noxious heat sensation) that plague the clinical use of existing TRPV1 antagonists.
Collapse
|
95
|
Charrua A, Avelino A, Cruz F. Modulation of urinary bladder innervation: TRPV1 and botulinum toxin A. Handb Exp Pharmacol 2011:345-374. [PMID: 21290235 DOI: 10.1007/978-3-642-16499-6_17] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
The persisting interest around neurotoxins such as vanilloids and botulinum toxin (BoNT) derives from their marked effect on detrusor overactivity refractory to conventional antimuscarinic treatments. In addition, both are administered by intravesical route. This offers three potential advantages. First, intravesical therapy is an easy way to provide high concentrations of pharmacological agents in the bladder tissue without causing unsuitable levels in other organs. Second, drugs effective on the bladder, but inappropriate for systemic administration, can be safely used as it is the case of vanilloids and BoNT. Third, the effects of one single treatment might be extremely longlasting, contributing to render these therapies highly attractive to patients despite the fact that the reasons to the prolonged effect are still incompletely understood. Attractive as it may be, intravesical pharmacological therapy should still be considered as a second-line treatment in patients refractory to conventional oral antimuscarinic therapy or who do not tolerate its systemic side effects. However, the increasing off-label use of these neurotoxins justifies a reappraisal of their pharmacological properties.
Collapse
Affiliation(s)
- Ana Charrua
- Institute of Histology and Embryology, Porto, Portugal
| | | | | |
Collapse
|
96
|
Regulation of TRP signalling by ion channel translocation between cell compartments. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 704:545-72. [PMID: 21290316 DOI: 10.1007/978-94-007-0265-3_30] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The TRP (transient receptor potential) family of ion channels is a heterogeneous family of calcium permeable cation channels that is subdivided into seven subfamilies: TRPC ("Canonical"), TRPV ("Vanilloid"), TRPM ("Melastatin"), TRPA ("Ankyrin"), TRPN ("NOMPC"), TRPP ("Polycystin"), and TRPML ("Mucolipin"). TRP-mediated ion currents across the cell membrane are determined by the single channel conductance, by the fraction of activated channels, and by the total amount of TRP channels present at the plasma membrane. In many cases, the amount of TRP channels at the plasma membrane is altered in response to physiological stimuli by translocation of channels to and from the plasma membrane. Regulated translocation has been described for channels of the TRPC, TRPV, TRPM, and TRPA family and is achieved by vesicular transport of these channels along cellular exocytosis and endocytosis pathways. This review summarizes the stimuli and signalling cascades involved in the translocation of TRP channels and highlights interactions of TRP channels with proteins of the endocytosis and exocytosis machineries.
Collapse
|
97
|
TRP channels and their implications in metabolic diseases. Pflugers Arch 2010; 461:211-23. [PMID: 21110037 DOI: 10.1007/s00424-010-0902-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2010] [Revised: 11/02/2010] [Accepted: 11/03/2010] [Indexed: 12/22/2022]
Abstract
The transient receptor potential (TRP) channel superfamily is composed of 28 nonselective cation channels that are ubiquitously expressed in many cell types and have considerable functional diversity. Although changes in TRP channel expression and function have been reported in cardiovascular disease and renal disorders, the pathogenic roles of TRP channels in metabolic diseases have not been systemically reviewed. In this review, we summarised the distribution of TRP channels in several metabolic tissues and discussed their roles in mediating and regulating various physiological and pathophysiological metabolic processes and diseases including diabetes, obesity, dyslipidaemia, metabolic syndrome, atherosclerosis, metabolic bone diseases and electrolyte disturbances. This review provides new insight into the involvement of TRP channels in the pathogenesis of metabolic disorders and implicates these channels as potential therapeutic targets for the management of metabolic diseases.
Collapse
|
98
|
Anand U, Otto WR, Anand P. Sensitization of capsaicin and icilin responses in oxaliplatin treated adult rat DRG neurons. Mol Pain 2010; 6:82. [PMID: 21106058 PMCID: PMC3003244 DOI: 10.1186/1744-8069-6-82] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2010] [Accepted: 11/24/2010] [Indexed: 12/22/2022] Open
Abstract
Background Oxaliplatin chemotherapy induced neuropathy is a dose related cumulative toxicity that manifests as tingling, numbness, and chronic pain, compromising the quality of life and leading to discontinued chemotherapy. Patients report marked hypersensitivity to cold stimuli at early stages of treatment, when sensory testing reveals cold and heat hyperalgesia. This study examined the morphological and functional effects of oxaliplatin treatment in cultured adult rat DRG neurons. Results 48 hour exposure to oxaliplatin resulted in dose related reduction in neurite length, density, and number of neurons compared to vehicle treated controls, using Gap43 immunostaining. Neurons treated acutely with 20 μg/ml oxaliplatin showed significantly higher signal intensity for cyclic AMP immunofluorescence (160.5 ± 13 a.u., n = 3, P < 0.05), compared to controls (120.3 ± 4 a.u.). Calcium imaging showed significantly enhanced capsaicin (TRPV1 agonist), responses after acute 20 μg/ml oxaliplatin treatment where the second of paired capsaicin responses increased from 80.7 ± 0.6% without oxaliplatin, to 171.26 ± 29% with oxaliplatin, (n = 6 paired t test, P < 0.05); this was reduced to 81.42 ± 8.1% (P < 0.05), by pretretreatment with the cannabinoid CB2 receptor agonist GW 833972. Chronic oxaliplatin treatment also resulted in dose related increases in capsaicin responses. Similarly, second responses to icilin (TRPA1/TRPM8 agonist), were enhanced after acute (143.85 ± 7%, P = 0.004, unpaired t test, n = 3), and chronic (119.7 ± 11.8%, P < 0.05, n = 3) oxaliplatin treatment, compared to control (85.3 ± 1.7%). Responses to the selective TRPM8 agonist WS-12 were not affected. Conclusions Oxaliplatin treatment induces TRP sensitization mediated by increased intracellular cAMP, which may cause neuronal damage. These effects may be mitigated by co-treatment with adenylyl cyclase inhibitors, like CB2 agonists, to alleviate the neurotoxic effects of oxaliplatin.
Collapse
Affiliation(s)
- Uma Anand
- Histopathology Unit, Cancer Research UK, London Research Institute, 44 Lincoln's Inn Fields, London WC1A 3PX, UK.
| | | | | |
Collapse
|
99
|
Premkumar LS, Sikand P. TRPV1: a target for next generation analgesics. Curr Neuropharmacol 2010; 6:151-63. [PMID: 19305794 PMCID: PMC2647151 DOI: 10.2174/157015908784533888] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2007] [Revised: 10/17/2007] [Accepted: 11/11/2007] [Indexed: 12/11/2022] Open
Abstract
Transient Receptor Potential Vanilloid 1 (TRPV1) is a Ca2+ permeant non-selective cation channel expressed in a subpopulation of primary afferent neurons. TRPV1 is activated by physical and chemical stimuli. It is critical for the detection of nociceptive and thermal inflammatory pain as revealed by the deletion of the TRPV1 gene. TRPV1 is distributed in the peripheral and central terminals of the sensory neurons and plays a role in initiating action potentials at the nerve terminals and modulating neurotransmitter release at the first sensory synapse, respectively. Distribution of TRPV1 in the nerve terminals innervating blood vessels and in parts of the CNS that are not subjected to temperature range that is required to activate TRPV1 suggests a role beyond a noxious thermal sensor. Presently, TRPV1 is being considered as a target for analgesics through evaluation of different antagonists. Here, we will discuss the distribution and the functions of TRPV1, potential use of its agonists and antagonists as analgesics and highlight the functions that are not related to nociceptive transmission that might lead to adverse effects.
Collapse
Affiliation(s)
- Louis S Premkumar
- Department of Pharmacology, Southern Illinois University School of Medicine Springfield, IL 62702, USA.
| | | |
Collapse
|
100
|
Huang J, Zhang X, McNaughton PA. Inflammatory pain: the cellular basis of heat hyperalgesia. Curr Neuropharmacol 2010; 4:197-206. [PMID: 18615146 DOI: 10.2174/157015906778019554] [Citation(s) in RCA: 142] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2006] [Revised: 06/30/2006] [Accepted: 06/30/2006] [Indexed: 12/20/2022] Open
Abstract
Injury or inflammation release a range of inflammatory mediators that increase the sensitivity of sensory neurons to noxious thermal or mechanical stimuli. The heat- and capsaicin-gated channel TRPV1, which is an important detector of multiple noxious stimuli, plays a critical role in the development of thermal hyperalgesia induced by a wide range of inflammatory mediators. We review here recent findings on the molecular mechanisms of sensitisation of TRPV1 by inflammatory mediators, including bradykinin, ATP, NGF and prostaglandins. We describe the signalling pathways believed to be involved in the potentiation of TRPV1, and our current understanding of how inflammatory mediators couple to these pathways.
Collapse
Affiliation(s)
- Jiehong Huang
- Department of Pharmacology, University of Cambridge, Cambridge, CB2 1PD, United Kingdom
| | | | | |
Collapse
|