Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Neurosci Lett ; 639: 59-67, 2017 02 03.
Article in English | MEDLINE | ID: mdl-28038937

ABSTRACT

While most membrane channels are only capable of passing small ions, certain non-selective cation channels have been recently shown to have the capacity to permeate large cations. The mechanisms underlying large molecule permeation are unclear, but this property has been exploited pharmacologically to target molecules, such as nerve conduction blockers, to specific subsets of pain-sensing neurons (nociceptors) expressing the heat-gated transient receptor potential (TRP) channel TRPV1. However, it is not clear if the principal mediator of cold stimuli TRPM8 is capable of mediating the permeation large molecules across cell membranes, suggesting that TRPM8-positive nerves cannot be similarly targeted. Here we show that both heterologous cells and native sensory neurons expressing TRPM8 channels allow the permeation of the large fluorescent cation Po-Pro3. Po-Pro3 influx is blocked by TRPM8-specific antagonism and when channel activity is desensitized. The effects of the potent agonist WS-12 are TRPM8-specific and dye uptake mediated by TRPM8 channels is similar to that observed with TRPV1. Lastly, we find that as with TRPV1, activation of TRPM8 channels can be used as a means to target intracellular uptake of cell-impermeable sodium channel blockers. In a neuronal cell line expressing TRPM8 channels, voltage-gated sodium currents are blocked in the presence of the cell-impermeable, charged lidocaine derivative QX-314 and WS-12. These results show that the ability of somatosensory TRP channels to promote the permeation of large cations also includes TRPM8, thereby suggesting that novel approaches to alter cold pain can also be employed via conduction block in TRPM8-positive sensory neurons.


Subject(s)
Nociceptors/drug effects , TRPM Cation Channels/metabolism , Transient Receptor Potential Channels/metabolism , Anesthetics, Local/pharmacology , Animals , Cell Line , Ganglia, Spinal/metabolism , Humans , Lidocaine/analogs & derivatives , Lidocaine/pharmacology , Pain/drug therapy , Pain/metabolism , Rats , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Thermosensing/drug effects , Thermosensing/physiology
2.
Sci Rep ; 6: 25626, 2016 05 12.
Article in English | MEDLINE | ID: mdl-27172999

ABSTRACT

We previously reported neuroprotective activity of the botanical anti-cancer drug candidate PBI-05204, a supercritical CO2 extract of Nerium oleander, in brain slice and in vivo models of ischemic stroke. We showed that one component of this neuroprotective activity is mediated through its principal cardiac glycoside constituent, oleandrin, via induction of the potent neurotrophic factor brain-derived neurotrophic factor (BDNF). However, we also noted that the concentration-relation for PBI-05204 in the brain slice oxygen-glucose deprivation (OGD) model is considerably broader than that for oleandrin as a single agent. We thus surmised that PBI-05204 contains an additional neuroprotective component(s), distinct from oleandrin. We report here that neuroprotective activity is also provided by the triterpenoid constituents of PBI-05204, notably oleanolic acid. We demonstrate that a sub-fraction of PBI-05204 (Fraction 0-4) containing oleanolic and other triterpenoids, but without cardiac glycosides, induces the expression of cellular antioxidant gene transcription programs regulated through antioxidant transcriptional response elements (AREs). Finally, we show that Fraction 0-4 provides broad neuroprotection in organotypic brain slice models for neurodegeneration driven by amyloid precursor protein (APP) and tau implicated in Alzheimer's disease and frontotemporal dementias, respectively, in addition to ischemic injury modeled by OGD.


Subject(s)
Antineoplastic Agents/pharmacology , Brain/drug effects , Heterocyclic Compounds, 4 or More Rings/pharmacology , Neurodegenerative Diseases/drug therapy , Plant Extracts/pharmacology , Stroke/drug therapy , Animals , Antineoplastic Agents/chemistry , Brain/metabolism , Brain/pathology , Chemical Fractionation/methods , Disease Models, Animal , Female , Glucose/metabolism , Humans , Male , Nerium/chemistry , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Oleanolic Acid/chemistry , Oleanolic Acid/pharmacology , Organ Culture Techniques , Oxygen/metabolism , Rats, Sprague-Dawley
3.
J Neurosci ; 33(30): 12543-52, 2013 Jul 24.
Article in English | MEDLINE | ID: mdl-23884957

ABSTRACT

Chronic pain associated with injury or disease can result from dysfunction of sensory afferents whereby the threshold for activation of pain-sensing neurons (nociceptors) is lowered. Neurotrophic factors control nociceptor development and survival, but also induce sensitization through activation of their cognate receptors, attributable, in part, to the modulation of ion channel function. Thermal pain is mediated by channels of the transient receptor potential (TRP) family, including the cold and menthol receptor TRPM8. Although it has been shown that TRPM8 is involved in cold hypersensitivity, the molecular mechanisms underlying this pain modality are unknown. Using microarray analyses to identify mouse genes enriched in TRPM8 neurons, we found that the glial cell line-derived neurotrophic factor (GDNF) family receptor GFRα3 is expressed in a subpopulation of TRPM8 sensory neurons that have the neurochemical profile of cold nociceptors. Moreover, we found that artemin, the specific GFRα3 ligand that evokes heat hyperalgesia, robustly sensitized cold responses in a TRPM8-dependent manner in mice. In contrast, GFRα1 and GFRα2 are not coexpressed with TRPM8 and their respective ligands GDNF and neurturin did not induce cold pain, whereas they did evoke heat hyperalgesia. Nerve growth factor induced mild cold sensitization, consistent with TrkA expression in TRPM8 neurons. However, bradykinin failed to alter cold sensitivity even though its receptor expresses in a subset of TRPM8 neurons. These results show for the first time that only select neurotrophic factors induce cold sensitization through TRPM8 in vivo, unlike the broad range of proalgesic agents capable of promoting heat hyperalgesia.


Subject(s)
Chronic Pain/physiopathology , Cold Temperature/adverse effects , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Nerve Tissue Proteins/metabolism , TRPM Cation Channels/genetics , Animals , Chronic Pain/genetics , Female , Ganglia, Spinal/cytology , Ganglia, Spinal/physiology , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Hot Temperature , Hyperalgesia/genetics , Hyperalgesia/physiopathology , Male , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Nociception/physiology , Oligonucleotide Array Sequence Analysis , Sensory Receptor Cells/physiology , TRPM Cation Channels/metabolism , Transcriptional Activation/physiology , Trigeminal Ganglion/cytology , Trigeminal Ganglion/physiology
4.
Am J Physiol Endocrinol Metab ; 305(1): E78-88, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23651844

ABSTRACT

Blood glucose concentration is tightly regulated by the rate of insulin secretion and clearance, a process partially controlled by sensory neurons serving as metabolic sensors in relevant tissues. The activity of these neurons is regulated by the products of metabolism which regulate transmitter release, and recent evidence suggests that neuronally expressed ion channels of the transient receptor potential (TRP) family function in this critical process. Here, we report the novel finding that the cold and menthol-gated channel TRPM8 is necessary for proper insulin homeostasis. Mice lacking TRPM8 respond normally to a glucose challenge while exhibiting prolonged hypoglycemia in response to insulin. Additionally, Trpm8-/- mice have increased rates of insulin clearance compared with wild-type animals and increased expression of insulin-degrading enzyme in the liver. TRPM8 channels are not expressed in the liver, but TRPM8-expressing sensory afferents innervate the hepatic portal vein, suggesting a TRPM8-mediated neuronal control of liver insulin clearance. These results demonstrate that TRPM8 is a novel regulator of serum insulin and support the role of sensory innervation in metabolic homeostasis.


Subject(s)
Blood Glucose/metabolism , Hypoglycemia/genetics , Insulin/metabolism , Sensory Receptor Cells/metabolism , TRPM Cation Channels/genetics , Animals , Bacteriocin Plasmids , Diabetes Mellitus, Experimental/metabolism , Homeostasis/physiology , Hypoglycemia/metabolism , Insulin-Secreting Cells/metabolism , Liver/blood supply , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Portal Vein/innervation , Rats , TRPM Cation Channels/metabolism
5.
J Neurosci ; 33(7): 2837-48, 2013 Feb 13.
Article in English | MEDLINE | ID: mdl-23407943

ABSTRACT

Many primary sensory neurons are polymodal, responding to multiple stimulus modalities (chemical, thermal, or mechanical), yet each modality is recognized differently. Although polymodality implies that stimulus encoding occurs in higher centers, such as the spinal cord or brain, recent sensory neuron ablation studies find that behavioral responses to different modalities require distinct subpopulations, suggesting the existence of modality-specific labeled lines at the level of the sensory afferent. Here we provide evidence that neurons expressing TRPM8, a cold- and menthol-gated channel required for normal cold responses in mammals, represents a labeled line solely for cold sensation. We examined the behavioral significance of conditionally ablating TRPM8-expressing neurons in adult mice, finding that, like animals lacking TRPM8 channels (Trpm8(-/-)), animals depleted of TRPM8 neurons ("ablated") are insensitive to cool to painfully cold temperatures. Ablated animals showed little aversion to noxious cold and did not distinguish between cold and a preferred warm temperature, a phenotype more profound than that of Trpm8(-/-) mice which exhibit only partial cold-avoidance and -preference behaviors. In addition to acute responses, cold pain associated with inflammation and nerve injury was significantly attenuated in ablated and Trpm8(-/-) mice. Moreover, cooling-induced analgesia after nerve injury was abolished in both genotypes. Last, heat, mechanical, and proprioceptive behaviors were normal in ablated mice, demonstrating that TRPM8 neurons are dispensable for other somatosensory modalities. Together, these data show that, although some limited cold sensitivity remains in Trpm8(-/-) mice, TRPM8 neurons are required for the breadth of behavioral responses evoked by cold temperatures.


Subject(s)
Analgesia , Cold Temperature , Pain/physiopathology , Sensory Receptor Cells/metabolism , TRPM Cation Channels/biosynthesis , Animals , Behavior, Animal/physiology , Cell Lineage/genetics , Cell Lineage/physiology , Diphtheria Toxin/pharmacology , Hand Strength/physiology , Hot Temperature , Immunohistochemistry , Mice , Mice, Knockout , Microarray Analysis , Nerve Fibers/physiology , Pain/chemically induced , Pain/psychology , Pain Insensitivity, Congenital/genetics , Physical Stimulation , Proprioception/physiology , Real-Time Polymerase Chain Reaction , Sensory Receptor Cells/physiology , TRPM Cation Channels/genetics , Thermosensing/genetics , Thermosensing/physiology
6.
PLoS One ; 6(9): e25894, 2011.
Article in English | MEDLINE | ID: mdl-21984952

ABSTRACT

TRPM8 (Transient Receptor Potential Melastatin-8) is a cold- and menthol-gated ion channel necessary for the detection of cold temperatures in the mammalian peripheral nervous system. Functioning TRPM8 channels are required for behavioral responses to innocuous cool, noxious cold, injury-evoked cold hypersensitivity, cooling-mediated analgesia, and thermoregulation. Because of these various roles, the ability to pharmacologically manipulate TRPM8 function to alter the excitability of cold-sensing neurons may have broad impact clinically. Here we examined a novel compound, PBMC (1-phenylethyl-4-(benzyloxy)-3-methoxybenzyl(2-aminoethyl)carbamate) which robustly and selectively inhibited TRPM8 channels in vitro with sub-nanomolar affinity, as determined by calcium microfluorimetry and electrophysiology. The actions of PBMC were selective for TRPM8, with no functional effects observed for the sensory ion channels TRPV1 and TRPA1. PBMC altered TRPM8 gating by shifting the voltage-dependence of menthol-evoked currents towards positive membrane potentials. When administered systemically to mice, PBMC treatment produced a dose-dependent hypothermia in wildtype animals while TRPM8-knockout mice remained unaffected. This hypothermic response was reduced at lower doses, whereas responses to evaporative cooling were still significantly attenuated. Lastly, systemic PBMC also diminished cold hypersensitivity in inflammatory and nerve-injury pain models, but was ineffective against oxaliplatin-induced neuropathic cold hypersensitivity, despite our findings that TRPM8 is required for the cold-related symptoms of this pathology. Thus PBMC is an attractive compound that serves as a template for the formulation of highly specific and potent TRPM8 antagonists that will have utility both in vitro and in vivo.


Subject(s)
Analgesics/therapeutic use , TRPM Cation Channels/metabolism , Thermosensing/drug effects , Animals , Cytophotometry , Electrophysiology , Mice , Organoplatinum Compounds/therapeutic use , Oxaliplatin , TRPA1 Cation Channel , TRPM Cation Channels/antagonists & inhibitors , TRPV Cation Channels/metabolism , Transient Receptor Potential Channels/metabolism
7.
Am J Physiol Regul Integr Comp Physiol ; 300(6): R1278-87, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21411765

ABSTRACT

The proper detection of environmental temperatures is essential for the optimal growth and survival of organisms of all shapes and phyla, yet only recently have the molecular mechanisms for temperature sensing been elucidated. The discovery of temperature-sensitive ion channels of the transient receptor potential (TRP) superfamily has been pivotal in explaining how temperatures are sensed in vivo, and here we will focus on the lone member of this cohort, TRPM8, which has been unequivocally shown to be cold sensitive. TRPM8 is expressed in somatosensory neurons that innervate peripheral tissues such as the skin and oral cavity, and recent genetic evidence has shown it to be the principal transducer of cool and cold stimuli. It is remarkable that this one channel, unlike other thermosensitive TRP channels, is associated with both innocuous and noxious temperature transduction, as well as cold hypersensitivity during injury and, paradoxically, cold-mediated analgesia. With ongoing research, the field is getting closer to answering a number of fundamental questions regarding this channel, including the cellular mechanisms of TRPM8 modulation, the molecular context of TRPM8 expression, as well as the full extent of the role of TRPM8 in cold signaling in vivo. These findings will further our understanding of basic thermotransduction and sensory coding, and may have important implications for treatments for acute and chronic pain.


Subject(s)
Cold Temperature , Signal Transduction/physiology , TRPM Cation Channels/physiology , Animals , Humans , Mice , Mice, Knockout , Models, Animal , Sensory Receptor Cells/physiology , TRPM Cation Channels/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...