Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Mol Psychiatry ; 28(2): 767-779, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36203006

RESUMEN

Opioids are the frontline analgesics for managing various types of pain. Paradoxically, repeated use of opioid analgesics may cause an exacerbated pain state known as opioid-induced hyperalgesia (OIH), which significantly contributes to dose escalation and consequently opioid overdose. Neuronal malplasticity in pain circuits has been the predominant proposed mechanism of OIH expression. Although glial cells are known to become reactive in OIH animal models, their biological contribution to OIH remains to be defined and their activation mechanism remains to be elucidated. Here, we show that reactive astrocytes (a.k.a. astrogliosis) are critical for OIH development in both male and female mice. Genetic reduction of astrogliosis inhibited the expression of OIH and morphine-induced neural circuit polarization (NCP) in the spinal dorsal horn (SDH). We found that Wnt5a is a neuron-to-astrocyte signal that is required for morphine-induced astrogliosis. Conditional knock-out of Wnt5a in neurons or its co-receptor ROR2 in astrocytes blocked not only morphine-induced astrogliosis but also OIH and NCP. Furthermore, we showed that the Wnt5a-ROR2 signaling-dependent astrogliosis contributes to OIH via inflammasome-regulated IL-1ß. Our results reveal an important role of morphine-induced astrogliosis in OIH pathogenesis and elucidate a neuron-to-astrocyte intercellular Wnt signaling pathway that controls the astrogliosis.


Asunto(s)
Analgésicos Opioides , Hiperalgesia , Animales , Femenino , Masculino , Ratones , Astrocitos/metabolismo , Gliosis , Hiperalgesia/inducido químicamente , Hiperalgesia/genética , Hiperalgesia/metabolismo , Morfina , Dolor , Vía de Señalización Wnt
2.
Brain ; 145(11): 4108-4123, 2022 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-35040478

RESUMEN

Chronic pain is the most common neurological disorder of HIV patients. Multiple neuropathologies were identified in the pain pathway. Among them is the prominent astrocytic reaction (also know an astrogliosis). However, the pathogenic role and mechanism of the astrogliosis are unclear. Here, we show that the astrogliosis is crucial for the pain development induced by a key neurotoxic HIV protein gp120 and that a neuron-to-astrocyte Wnt5a signal controls the astrogliosis. Ablation of astrogliosis blocked the development of gp120-induced mechanical hyperalgesia, and concomitantly the expression of neural circuit polarization in the spinal dorsal horn. We demonstrated that conditional knockout of either Wnt5a in neurons or its receptor ROR2 in astrocytes abolished not only gp120-induced astrogliosis but also hyperalgesia and neural circuit polarization. Furthermore, we found that the astrogliosis promoted expression of hyperalgesia and NCP via IL-1ß regulated by a Wnt5a-ROR2-MMP2 axis. Our results shed light on the role and mechanism of astrogliosis in the pathogenesis of HIV-associated pain.


Asunto(s)
Infecciones por VIH , Hiperalgesia , Humanos , Hiperalgesia/metabolismo , Astrocitos/metabolismo , Gliosis , Infecciones por VIH/complicaciones , Infecciones por VIH/metabolismo , Médula Espinal/metabolismo , Dolor/inducido químicamente , Neuronas/metabolismo , Proteína Wnt-5a/metabolismo
3.
J Neurosci ; 39(42): 8408-8421, 2019 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-31471472

RESUMEN

HIV-1 infection of the nervous system causes various neurological diseases, and synaptic degeneration is likely a critical step in the neuropathogenesis. Our prior studies revealed a significant decrease of synaptic protein, specifically in the spinal dorsal horn of patients with HIV-1 in whom pain developed, suggesting a potential contribution of synaptic degeneration to the pathogenesis of HIV-associated pain. However, the mechanism by which HIV-1 causes the spinal synaptic degeneration is unclear. Here, we identified a critical role of microglia in the synaptic degeneration. In primary cortical cultures (day in vitro 14) and spinal cords of 3- to 5-month-old mice (both sexes), microglial ablation inhibited gp120-induced synapse decrease. Fractalkine (FKN), a microglia activation chemokine specifically expressed in neurons, was upregulated by gp120, and knockout of the FKN receptor CX3CR1, which is predominantly expressed in microglia, protected synapses from gp120-induced toxicity. These results indicate that the neuron-to-microglia intercellular FKN/CX3CR1 signaling plays a role in gp120-induced synaptic degeneration. To elucidate the mechanism controlling this intercellular signaling, we tested the role of the Wnt/ß-catenin pathway in regulating FKN expression. Inhibition of Wnt/ß-catenin signaling blocked both gp120-induced FKN upregulation and synaptic degeneration, and gp120 stimulated Wnt/ß-catenin-regulated FKN expression via NMDA receptors (NMDARs). Furthermore, NMDAR antagonist APV, Wnt/ß-catenin signaling suppressor DKK1, or knockout of CX3CR1 alleviated gp120-induced mechanical allodynia in mice, suggesting a critical contribution of the Wnt/ß-catenin/FKN/CX3R1 pathway to gp120-induced pain. These findings collectively suggest that HIV-1 gp120 induces synaptic degeneration in the spinal pain neural circuit by activating microglia via Wnt3a/ß-catenin-regulated FKN expression in neurons.SIGNIFICANCE STATEMENT Synaptic degeneration develops in the spinal cord dorsal horn of HIV patients with chronic pain, but the patients without the pain disorder do not show this neuropathology, indicating a pathogenic contribution of the synaptic degeneration to the development of HIV-associated pain. However, the mechanism underlying the synaptic degeneration is unclear. We report here that HIV-1 gp120, a neurotoxic protein that is specifically associated with the manifestation of pain in HIV patients, induces synapse loss via microglia. Further studies elucidate that gp120 activates microglia by stimulating Wnt/ß-catenin-regulated fractalkine in neuron. The results demonstrate a critical role of microglia in the pathogenesis of HIV-associated synaptic degeneration in the spinal pain neural circuit.


Asunto(s)
Proteína gp120 de Envoltorio del VIH/farmacología , Microglía/efectos de los fármacos , Degeneración Nerviosa/metabolismo , Médula Espinal/efectos de los fármacos , Sinapsis/efectos de los fármacos , Animales , Receptor 1 de Quimiocinas CX3C/genética , Receptor 1 de Quimiocinas CX3C/metabolismo , Quimiocina CX3CL1/metabolismo , Ratones , Ratones Noqueados , Microglía/metabolismo , Neuralgia/metabolismo , Médula Espinal/metabolismo , Sinapsis/metabolismo , Regulación hacia Arriba/efectos de los fármacos
4.
Mol Pain ; 15: 1744806919840098, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30857460

RESUMEN

Chemotherapy-induced peripheral neuropathy (CIPN) is an adverse side effect of many anti-cancer chemotherapeutic treatments. CIPN often causes neuropathic pain in extremities, and oxidative stress has been shown to be a major contributing factor to this pain. In this study, we determined the site of oxidative stress associated with pain (specifically, mechanical hypersensitivity) in cisplatin- and paclitaxel-treated mouse models of CIPN and investigated the neurophysiological mechanisms accounting for the pain. C57BL/6N mice that received either cisplatin or paclitaxel (2 mg/kg, once daily on four alternate days) developed mechanical hypersensitivity to von Frey filament stimulations of their hindpaws. Cisplatin-induced mechanical hypersensitivity was inhibited by silencing of Transient Receptor Potential channels V1 (TRPV1)- or TRPA1-expressing afferents, whereas paclitaxel-induced mechanical hypersensitivity was attenuated by silencing of Aß fibers. Although systemic delivery of phenyl N-tert-butylnitrone, a reactive oxygen species scavenger, alleviated mechanical hypersensitivity in both cisplatin- and paclitaxel-treated mice, intraplantar phenyl N-tert-butylnitrone was effective only in cisplatin-treated mice, and intrathecal phenyl N-tert-butylnitrone, only in paclitaxel-treated mice. In a reactive oxygen species-dependent manner, the mechanosensitivity of Aδ/C fiber endings in the hindpaw skin was increased in cisplatin-treated mice, and the excitatory synaptic strength in the spinal dorsal horn was potentiated in paclitaxel-treated mice. Collectively, these results suggest that cisplatin-induced mechanical hypersensitivity is attributed to peripheral oxidative stress sensitizing mechanical nociceptors, whereas paclitaxel-induced mechanical hypersensitivity is due to central (spinal) oxidative stress maintaining central sensitization that abnormally produces pain in response to Aß fiber inputs.


Asunto(s)
Antineoplásicos Fitogénicos/efectos adversos , Neuralgia/etiología , Neuralgia/metabolismo , Estrés Oxidativo/efectos de los fármacos , Animales , Cisplatino/efectos adversos , Hiperalgesia/etiología , Hiperalgesia/metabolismo , Masculino , Ratones Endogámicos C57BL , Paclitaxel/efectos adversos , Especies Reactivas de Oxígeno/metabolismo , Canal Catiónico TRPA1/metabolismo , Canales de Potencial de Receptor Transitorio/metabolismo
5.
Mol Pain ; 14: 1744806918797032, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30152257

RESUMEN

Reactive oxygen species has been suggested as a key player in neuropathic pain, causing central sensitization by changing synaptic strengths in spinal dorsal horn neurons. However, it remains unclear as to what type of reactive oxygen species changes what aspect of synaptic strengths for central sensitization in neuropathic pain conditions. In this study, we investigated whether mitochondrial superoxide affects both excitatory and inhibitory synaptic strengths in spinal dorsal horn neurons after peripheral nerve injury. Upregulation of mitochondrial superoxide level by knockout of superoxide dismutase-2 exacerbated neuropathic mechanical hypersensitivity caused by L5 spinal nerve ligation, whereas downregulation of mitochondrial superoxide level by overexpression of superoxide dismutase-2 alleviated the hypersensitivity. In spinal nerve ligation condition, the frequency of miniature excitatory postsynaptic currents increased, while that of miniature inhibitory postsynaptic currents decreased in spinal dorsal horn neurons. Superoxide dismutase-2-knockout augmented, whereas superoxide dismutase-2-overexpression prevented, the spinal nerve ligation-increased miniature excitatory postsynaptic currents frequency. However, superoxide dismutase-2-knockout had no effect on the spinal nerve ligation-decreased miniature inhibitory postsynaptic current frequency, and superoxide dismutase-2-overexpression unexpectedly decreased miniature inhibitory postsynaptic current frequency in the normal condition. When applied to the spinal cord slice during in vitro recordings, mitoTEMPO, a specific scavenger of mitochondrial superoxide, reduced the spinal nerve ligation-increased miniature excitatory postsynaptic currents frequency but failed to normalize the spinal nerve ligation-decreased miniature inhibitory postsynaptic current frequency. These results suggest that in spinal dorsal horn neurons, high levels of mitochondrial superoxide increase excitatory synaptic strength after peripheral nerve injury and contribute to neuropathic mechanical hypersensitivity. However, mitochondrial superoxide does not seem to be involved in the decreased inhibitory synaptic strength in this neuropathic pain condition.


Asunto(s)
Potenciales Postsinápticos Excitadores/fisiología , Neuralgia/patología , Células del Asta Posterior/fisiología , Superóxido Dismutasa/metabolismo , Animales , Modelos Animales de Enfermedad , Estimulación Eléctrica , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/genética , Hiperalgesia , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuralgia/genética , Neuralgia/fisiopatología , Umbral del Dolor/fisiología , Técnicas de Placa-Clamp , Superóxido Dismutasa/genética , Factores de Tiempo
6.
Mol Pain ; 13: 1744806917713907, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28587509

RESUMEN

Abstract: Intradermally injected capsaicin induces secondary mechanical hyperalgesia and allodynia outside the primary (i.e., capsaicininjected) site. This secondary mechanical hypersensitivity is attributed to central sensitization in which reactive oxygen species (ROS) play a key role. We examined whether ROS would be differentially involved in secondary mechanical hyperalgesia and allodynia using a mouse intraplantar capsaicin injection model. In mice, capsaicin-induced secondary mechanical hyperalgesia outlasted its allodynia counterpart. Unlike the hyperalgesia, the allodynia was temporarily abolished by an anesthetic given at the capsaicin-injected site. The ROS scavenger phenyl-N-tert-butylnitrone slowed the development of both secondary mechanical hyperalgesia and allodynia when administered before intraplantar capsaicin injection, whereas it inhibited only the allodynia when administered after capsaicin had already induced secondary mechanical hyperalgesia and allodynia. Intrathecal injection of the ROS donor KO2 induced both mechanical hyperalgesia and allodynia with the former outlasting the latter. Metformin, an activator of redox-sensitive adenosine monophosphate-activated protein kinase, selectively inhibited capsaicin-induced secondary mechanical allodynia and intrathecal KO2-induced mechanical allodynia. These results suggest that ROS is required for rapid activation of central sensitization mechanisms for both secondary mechanical hyperalgesia and allodynia after intraplantar capsaicin injection. Once activated, the mechanism for the hyperalgesia is longlasting without being critically dependent on ongoing afferent activities arising from the capsaicin-injected site and the continuous presence of ROS. On the contrary, the ongoing afferent activities, ROS presence and adenosine monophosphate-activated protein kinase inhibition are indispensable for the maintenance mechanism for capsaicin-induced secondary mechanical allodynia.


Asunto(s)
Capsaicina/farmacología , Hiperalgesia/inducido químicamente , Dolor/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Modelos Animales de Enfermedad , Hiperalgesia/metabolismo , Inyecciones Espinales , Masculino , Ratones Endogámicos C57BL , Dimensión del Dolor/métodos , Médula Espinal/metabolismo
7.
J Biol Chem ; 290(8): 5167-5173, 2015 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-25561736

RESUMEN

PIEZO1 is a recently cloned eukaryotic cation-selective channel that opens with mechanical force. We found that extracellular protonation inhibits channel activation by ≈90% by increased occupancy in the closed or the inactivated state. Titration between pH 6.3 and 8.3 exhibited a pK of ≈6.9. The steepness of the titration data suggests positive cooperativity, implying the involvement of at least two protonation sites. Whole-cell recordings yielded results similar to patches, and pH 6.5 reduced whole-cell currents by >80%. The effects were reversible. To assess whether pH acts on the open or the inactivated state, we tested a double-mutant PIEZO1 that does not inactivate. Cell-attached patches and whole-cell currents from this mutant channel were pH-insensitive. Thus, protonation appears to be associated with domain(s) of the channel involved with inactivation. pH also did not affect mutant channels with point mutations at position 2456 that are known to exhibit slow inactivation. To determine whether the physical properties of the membrane are altered by pH and thereby affect channel gating, we measured patch capacitance during mechanical stimuli at pH 6.5 and 7.3. The rate constants for changes in patch capacitance were independent of pH, suggesting that bilayer mechanics are not involved. In summary, low pH stabilizes the inactivated state. This effect may be important when channels are activated under pathological conditions in which the pH is reduced, such as during ischemia.


Asunto(s)
Activación del Canal Iónico , Canales Iónicos/metabolismo , Células HEK293 , Humanos , Concentración de Iones de Hidrógeno , Canales Iónicos/química , Canales Iónicos/genética , Isquemia/genética , Isquemia/metabolismo , Estructura Terciaria de Proteína
8.
Proc Natl Acad Sci U S A ; 110(12): E1162-8, 2013 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-23487776

RESUMEN

Familial xerocytosis (HX) in humans is an autosomal disease that causes dehydration of red blood cells resulting in hemolytic anemia which has been traced to two individual mutations in the mechanosensitive ion channel, PIEZO1. Each mutation alters channel kinetics in ways that can explain the clinical presentation. Both mutations slowed inactivation and introduced a pronounced latency for activation. A conservative substitution of lysine for arginine (R2456K) eliminated inactivation and also slowed deactivation, indicating that this mutant's loss of charge is not responsible for HX. Fitting the current vs. pressure data to Boltzmann distributions showed that the half-activation pressure, P1/2, for M2225R was similar to that of WT, whereas mutations at position 2456 were left shifted. The absolute stress sensitivity was calibrated by cotransfection and comparison with MscL, a well-characterized mechanosensitive channel from bacteria that is driven by bilayer tension. The slope sensitivity of WT and mutant human PIEZO1 (hPIEZO1) was similar to that of MscL implying that the in-plane area increased markedly, by ∼6-20 nm(2) during opening. In addition to the behavior of individual channels, groups of hPIEZO1 channels could undergo simultaneous changes in kinetics including a loss of inactivation and a long (∼200 ms), silent latency for activation. These observations suggest that hPIEZO1 exists in spatial domains whose global properties can modify channel gating. The mutations that create HX affect cation fluxes in two ways: slow inactivation increases the cation flux, and the latency decreases it. These data provide a direct link between pathology and mechanosensitive channel dysfunction in nonsensory cells.


Asunto(s)
Anemia Hemolítica Congénita/metabolismo , Hidropesía Fetal/metabolismo , Canales Iónicos/metabolismo , Mecanotransducción Celular , Mutación Missense , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Anemia Hemolítica Congénita/genética , Anemia Hemolítica Congénita/patología , Anemia Hemolítica Congénita/fisiopatología , Escherichia coli , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Células HEK293 , Humanos , Hidropesía Fetal/genética , Hidropesía Fetal/patología , Hidropesía Fetal/fisiopatología , Canales Iónicos/genética , Cinética , Datos de Secuencia Molecular , Estructura Terciaria de Proteína
9.
Front Cell Neurosci ; 18: 1423410, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38957539

RESUMEN

Microglia are the resident macrophages of the central nervous system (CNS) that control brain development, maintain neural environments, respond to injuries, and regulate neuroinflammation. Despite their significant impact on various physiological and pathological processes across mammalian biology, there remains a notable gap in our understanding of how microglia perceive and transmit mechanical signals in both normal and diseased states. Recent studies have revealed that microglia possess the ability to detect changes in the mechanical properties of their environment, such as alterations in stiffness or pressure. These changes may occur during development, aging, or in pathological conditions such as trauma or neurodegenerative diseases. This review will discuss microglial Piezo1 mechanosensitive channels as potential therapeutic targets for Alzheimer's disease (AD). The structure, function, and modulation of Piezo1 will be discussed, as well as its role in facilitating microglial clearance of misfolded amyloid-ß (Aß) proteins implicated in the pathology of AD.

10.
Biophys J ; 105(4): 880-6, 2013 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-23972840

RESUMEN

PIEZO1 is an inactivating eukaryotic cation-selective mechanosensitive ion channel. Two sites have been located in the channel that when individually mutated lead to xerocytotic anemia by slowing inactivation. By introducing mutations at two sites, one associated with xerocytosis and the other artificial, we were able to remove inactivation. The double mutant (DhPIEZO1) has a substitution of arginine for methionine (M2225R) and lysine for arginine (R2456K). The loss of inactivation was accompanied by ∼30-mmHg shift of the activation curve to lower pressures and slower rates of deactivation. The slope sensitivity of gating was the same for wild-type and mutants, indicating that the dimensional changes between the closed and open state are unaffected by the mutations. The unitary channel conductance was unchanged by mutations, so these sites are not associated with pore. DhPIEZO1 was reversibly inhibited by the peptide GsMTx4 that acted as a gating modifier. The channel kinetics were solved using complex stimulus waveforms and the data fit to a three-state loop in detailed balance. The reaction had two pressure-dependent rates, closed to open and inactivated to closed. Pressure sensitivity of the opening rate with no sensitivity of the closing rate means that the energy barrier between them is located near the open state. Mutant cycle analysis of inactivation showed that the two sites interacted strongly, even though they are postulated to be on opposite sides of the membrane.


Asunto(s)
Activación del Canal Iónico , Canales Iónicos/química , Canales Iónicos/metabolismo , Anemia Hemolítica Congénita/genética , Fenómenos Biomecánicos , Células HEK293 , Humanos , Hidropesía Fetal/genética , Canales Iónicos/genética , Cinética , Mutación , Presión , Termodinámica
11.
Front Cell Neurosci ; 17: 1200946, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37305437

RESUMEN

Piezo1 mechanosensitive ion channel (MSC) plays a significant role in human physiology. Despite several research on the function and expression of Piezo1 in the nervous system, its electrophysiological properties in neuroinflammatory astrocytes remain unknown. We tested whether astrocytic neuroinflammatory state regulates Piezo1 using electrical recordings, calcium imaging, and wound healing assays on cultured astrocytes. In this study, we determined whether neuroinflammatory condition regulates astrocytic Piezo1 currents in astrocytes. First, we performed electrophysiological recordings on the mouse cerebellum astrocytes (C8-S) under lipopolysaccharide (LPS)-induced neuroinflammatory condition. We found that LPS treatment significantly increased MSC currents in C8-S. The half-maximal pressure of LPS treated MSC currents was left-shifted but the slope sensitivity was not altered by LPS treatment. LPS-induced increase of MSC currents were further augmented by Piezo1 agonist, Yoda1 but were normalized by Piezo1 inhibitor, GsMTx4. Furthermore, silencing Piezo1 in LPS treated C8-S normalized not only MSC currents but also calcium influx and cell migration velocity. Together, our results show that LPS sensitized Piezo1 channel in C8-S astrocytes. These findings will suggest that astrocytic Piezo1 is a determinant of neuroinflammation pathogenesis and may in turn become the foundation of further research into curing several neuronal illnesses and injury related inflammation of neuronal cells.

12.
Biophys J ; 101(11): 2645-51, 2011 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-22261052

RESUMEN

The ability to form gigaseals is essential for patch-clamp electrophysiology; however, ion channels located in the seal can produce measureable currents. To explore the expected properties of channels in the seal (i.e., rim channels), we created a mathematical model. The seal was a two-dimensional cable filled with saline and bounded on one side by membrane (with resistance and capacitance) and on the other side by glass (nonconductive and noncapacitive). We included ion depletion/accumulation around the channels. The channels were ohmic with a conductance that increased with the concentration of permeant ions. The aqueous seal thickness was set nominally to 1 nm. Imaging with fluorescent dyes in the pipette showed that the hydrophilic dye Alexa 488 is impermeant, but lipophilic FM1-43 labels the seal. The model showed that to obtain high-resistance seals, the conductivity of the seal media has to be <10% that of the bath. Stimulus voltages decreased with distance down the seal. In agreement with results in the literature, channels in the seal can produce currents similar to those in the pipette-spanning dome. The transition times of currents are slower due to membrane capacitance. If channel densities are uniform, patch currents are dominated by channels in the dome.


Asunto(s)
Fenómenos Electrofisiológicos , Canales Iónicos/metabolismo , Modelos Biológicos , Difusión , Conductividad Eléctrica , Activación del Canal Iónico/fisiología , Canales de Potasio/metabolismo
13.
Biochemistry ; 50(29): 6295-300, 2011 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-21696149

RESUMEN

Cells can respond to mechanical stress by gating mechanosensitive ion channels (MSCs). The cloning of Piezo1, a eukaryotic cation selective MSC, defines a new system for studying mechanical transduction at the cellular level. Because Piezo1 has electrophysiological properties similar to those of endogenous cationic MSCs that are selectively inhibited by the peptide GsMTx4, we tested whether the peptide targets Piezo1 activity. Extracellular GsMTx4 at micromolar concentrations reversibly inhibited ∼80% of the mechanically induced current of outside-out patches from transfected HEK293 cells. The inhibition was voltage insensitive, and as seen with endogenous MSCs, the mirror image d enantiomer inhibited like the l. The rate constants for binding and unbinding based on Piezo1 current kinetics provided association and dissociation rates of 7.0 × 10(5) M(-1) s(-1) and 0.11 s(-1), respectively, and a K(D) of ∼155 nM, similar to values previously reported for endogenous MSCs. Consistent with predicted gating modifier behavior, GsMTx4 produced an ∼30 mmHg rightward shift in the pressure-gating curve and was active on closed channels. In contrast, streptomycin, a nonspecific inhibitor of cationic MSCs, showed the use-dependent inhibition characteristic of open channel block. The peptide did not block currents of the mechanical channel TREK-1 on outside-out patches. Whole-cell Piezo1 currents were also reversibly inhibited by GsMTx4, and although the off rate was nearly identical to that of outside-out patches, differences were observed for the on rate. The ability of GsMTx4 to target the mechanosensitivity of Piezo1 supports the use of this channel in high-throughput screens for pharmacological agents and diagnostic assays.


Asunto(s)
Canales Iónicos/antagonistas & inhibidores , Mecanotransducción Celular/efectos de los fármacos , Péptidos/farmacología , Venenos de Araña/farmacología , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intercelular , Activación del Canal Iónico/efectos de los fármacos , Canales Iónicos/metabolismo , Cinética
14.
Front Synaptic Neurosci ; 13: 748929, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34867259

RESUMEN

Opioids are widely used for pain relief; however, chronic opioid use causes a paradoxical state of enhanced pain sensitivity, termed "Opioid-induced hyperalgesia (OIH)." Despite the clinical importance of OIH, the detailed mechanism by which it enhances pain sensitivity remains unclear. In this study, we tested whether repeated morphine induces a neuronal circuit polarization in the mouse spinal dorsal horn (SDH). Transgenic mice expressing GFP to neurokinin 1 receptor-expressing neurons (sNK1Rn) and GABAergic interneurons (sGABAn) that received morphine [20 mg/kg, once daily for four consecutive days (i.p.)] developed mechanical hypersensitivity. Repeated morphine altered synaptic strengths in the SDH as a specific cell-type but not in a gender-dependent manner. In sNK1Rn and non-tonic firing neurons, repeated morphine treatment significantly increased frequency of spontaneous excitatory postsynaptic current (sEPSC) and evoked EPSC (eEPSC). In addition, repeated morphine treatment significantly decreased evoked inhibitory postsynaptic current (eIPSC) in sNK1Rn. Conversely, in sGABAn and tonic firing neurons, repeated morphine treatment significantly decreased sEPSC frequency and eEPSC, but had no change of eIPSC in sGABAn. Interestingly, repeated morphine treatment significantly decreased neuronal rheobase of sNK1Rn but had no effect on sGABAn. These findings suggest that spinal neuronal circuit polarization maybe the mechanism of OIH and identify a potential therapeutic mechanism to prevent or treat opioid-induced pain.

15.
Neuroscience ; 428: 132-139, 2020 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-31917342

RESUMEN

Since 1967, spinal cord stimulation (SCS) has been used to manage chronic intractable pain of the trunk and limbs. Compared to traditional high-intensity, low-frequency (<100 Hz) SCS that is thought to produce paresthesia and pain relief by stimulating large myelinated fibers in the dorsal column (DC), low-intensity, high-frequency (10 kHz) SCS has demonstrated long-term pain relief without generation of paresthesia. To understand this paresthesia-free analgesic mechanism of 10 kHz SCS, we examined whether 10 kHz SCS at intensities below sensory thresholds would modulate spinal dorsal horn (DH) neuronal function in a neuron type-dependent manner. By using in vivo and ex vivo electrophysiological approaches, we found that low-intensity (sub-sensory threshold) 10 kHz SCS, but not 1 kHz or 5 kHz SCS, selectively activates inhibitory interneurons in the spinal DH. This study suggests that low-intensity 10 kHz SCS may inhibit pain sensory processing in the spinal DH by activating inhibitory interneurons without activating DC fibers, resulting in paresthesia-free pain relief.


Asunto(s)
Potenciales de Acción/fisiología , Neuronas/fisiología , Células del Asta Posterior/fisiología , Médula Espinal/fisiología , Animales , Masculino , Manejo del Dolor/métodos , Dimensión del Dolor/métodos , Ratas Sprague-Dawley , Estimulación de la Médula Espinal/métodos
16.
PLoS One ; 13(11): e0207309, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30462693

RESUMEN

PIEZO1 and PIEZO2 are mechanosensitive channels (MSCs) important for cellular function and mutations in them lead to human disorders. We examined how functional heteromers form between subunits of PIEZO1 using the mutants E2117K, E2117D, and E2117A. Homomers of E2117K do not conduct. E2117A homomers have low conductance with rapid inactivation, and those of E2117D have high conductance with slow inactivation. Pairing E2117K with E2117D or E2117A with E2117D gave rise to new channel species representing heteromers with distinct conductances. Whole-cell currents from co-expression of E2117A and E2117D fit well with a linear-combination model of homomeric channel currents suggesting that functional channels do not form from freely-diffusing, randomly-mixed monomers in-vitro. Whole-cell current from coexpressed PIEZO1/PIEZO2 also fit as a linear combination of homomer currents. High-resolution optical images of fluorescently-tagged channels support this interpretation because coexpressed subunits segregate into discrete domains.


Asunto(s)
Canales Iónicos/metabolismo , Mutación Missense , Multimerización de Proteína , Sustitución de Aminoácidos , Células HEK293 , Humanos , Canales Iónicos/genética
17.
PLoS One ; 11(3): e0151289, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26963637

RESUMEN

PIEZO1 is a mechanosensitive eukaryotic cation-selective channel that rapidly inactivates in a voltage-dependent manner. We previously showed that a fluorescent protein could be encoded within the hPIEZO1 sequence without loss of function. In this work, we split the channel into two at this site and asked if coexpression would produce a functional channel or whether gating and permeation might be contained in either segment. The split protein was expressed in two segments by a bicistronic plasmid where the first segment spanned residues 1 to 1591, and the second segment spanned 1592 to 2521. When the "split protein" is coexpressed, the parts associate to form a normal channel. We measured the whole-cell, cell-attached and outside-out patch currents in transfected HEK293 cells. Indentation produced whole-cell currents monotonic with the stimulus. Single channel recordings showed voltage-dependent inactivation. The Boltzmann activation curve for outside-out patches had a slope of 8.6/mmHg vs 8.1 for wild type, and a small leftward shift in the midpoint (32 mmHg vs 41 mmHg). The association of the two channel domains was confirmed by FRET measurements of mCherry on the N-terminus and EGFP on the C-terminus. Neither of the individual protein segments produced current when expressed alone.


Asunto(s)
Canales Iónicos/metabolismo , Expresión Génica , Células HEK293 , Humanos , Activación del Canal Iónico , Canales Iónicos/química , Canales Iónicos/genética , Cinética , Modelos Moleculares , Mutación , Estructura Terciaria de Proteína
18.
Nat Commun ; 7: 10366, 2016 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-26785635

RESUMEN

Mechanosensitive ion channels are force-transducing enzymes that couple mechanical stimuli to ion flux. Understanding the gating mechanism of mechanosensitive channels is challenging because the stimulus seen by the channel reflects forces shared between the membrane, cytoskeleton and extracellular matrix. Here we examine whether the mechanosensitive channel PIEZO1 is activated by force-transmission through the bilayer. To achieve this, we generate HEK293 cell membrane blebs largely free of cytoskeleton. Using the bacterial channel MscL, we calibrate the bilayer tension demonstrating that activation of MscL in blebs is identical to that in reconstituted bilayers. Utilizing a novel PIEZO1-GFP fusion, we then show PIEZO1 is activated by bilayer tension in bleb membranes, gating at lower pressures indicative of removal of the cortical cytoskeleton and the mechanoprotection it provides. Thus, PIEZO1 channels must sense force directly transmitted through the bilayer.


Asunto(s)
Citoesqueleto/metabolismo , Canales Iónicos/metabolismo , Supervivencia Celular/fisiología , Células HEK293 , Humanos , Membrana Dobles de Lípidos/metabolismo
19.
PLoS One ; 10(5): e0125503, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25955826

RESUMEN

Members of the eukaryotic PIEZO family (the human orthologs are noted hPIEZO1 and hPIEZO2) form cation-selective mechanically-gated channels. We characterized the selectivity of human PIEZO1 (hPIEZO1) for alkali ions: K+, Na+, Cs+ and Li+; organic cations: TMA and TEA, and divalents: Ba2+, Ca2+, Mg2+ and Mn2+. All monovalent ions permeated the channel. At a membrane potential of -100 mV, Cs+, Na+ and K+ had chord conductances in the range of 35-55 pS with the exception of Li+, which had a significantly lower conductance of ~ 23 pS. The divalents decreased the single-channel permeability of K+, presumably because the divalents permeated slowly and occupied the open channel for a significant fraction of the time. In cell-attached mode, 90 mM extracellular divalents had a conductance for inward currents carried by the divalents of: 25 pS for Ba2+ and 15 pS for Ca2+ at -80 mV and 10 pS for Mg2+ at -50 mV. The organic cations, TMA and TEA, permeated slowly and attenuated K+ currents much like the divalents. As expected, the channel K+ conductance increased with K+ concentration saturating at ~ 45 pS and the KD of K+ for the channel was 32 mM. Pure divalent ion currents were of lower amplitude than those with alkali ions and the channel opening rate was lower in the presence of divalents than in the presence of monovalents. Exposing cells to the actin disrupting reagent cytochalasin D increased the frequency of openings in cell-attached patches probably by reducing mechanoprotection.


Asunto(s)
Canales Iónicos/metabolismo , Potenciales de la Membrana/fisiología , Metales Alcalinos/metabolismo , Metales Alcalinotérreos/metabolismo , Cationes Bivalentes , Cationes Monovalentes , Permeabilidad de la Membrana Celular/efectos de los fármacos , Citocalasina D/farmacología , Expresión Génica , Células HEK293 , Humanos , Canales Iónicos/genética , Transporte Iónico/efectos de los fármacos , Potenciales de la Membrana/efectos de los fármacos , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Técnicas de Placa-Clamp , Plásmidos/química , Plásmidos/metabolismo , Compuestos de Amonio Cuaternario/metabolismo , Tetraetilamonio/metabolismo , Transfección
20.
PLoS One ; 8(8): e72894, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24023653

RESUMEN

Mutations that lead to muscular dystrophy often create deficiencies in cytoskeletal support of the muscle sarcolemma causing hyperactive mechanosensitive cation channel (MSC) activity and elevated intracellular Ca(2+). Caveolae are cholesterol-rich microdomains that form mechanically deformable invaginations of the sarcolemma. Mutations to caveolin-3, the main scaffolding protein of caveolae in muscle, cause Limbe-Girdle muscular dystrophy. Using genetic and acute chemical perturbations of developing myotubes we investigated whether caveolae are functionally linked to MSCs. MSC sensitivity was assayed using suction application to patches and probe-induced indentation during whole-cell recordings. Membrane mechanical stress in patches was monitored using patch capacitance/impedance. Cholesterol depletion disrupted caveolae and caused a large increase in MSC current. It also decreased the membrane mechanical relaxation time, likely reflecting cytoskeleton dissociation from the bilayer. Reduction of Cav3 expression with miRNA also increased MSC current and decreased patch relaxation time. In contrast Cav3 overexpression produced a small decrease in MSC currents. To acutely and specifically inhibit Cav3 interactions, we made a chimeric peptide containing the antennapedia membrane translocation domain and the Cav3 scaffolding domain (A-CSD3). A-CSD3 action was time dependent initially producing a mild Ca(2+) leak and increased MSC current, while longer exposures decreased MSC currents coinciding with increased patch stiffening. Images of GFP labeled Cav3 in patches showed that Cav3 doesn't enter the pipette, showing patch composition differed from the cell surface. However, disruption via cholesterol depletion caused Cav3 to become uniformly distributed over the sarcolemma and Cav3 appearance in the patch dome. The whole-cell indentation currents elicited under the different caveolae modifying conditions mirror the patch response supporting the role of caveolae in MSC function. These studies show that normal expression levels of Cav3 are mechanoprotective to the sarcolemma through multiple mechanisms, and Cav3 upregulation observed in some dystrophies may compensate for other mechanical deficiencies.


Asunto(s)
Caveolas/metabolismo , Activación del Canal Iónico , Canales Iónicos/metabolismo , Mecanotransducción Celular , Fibras Musculares Esqueléticas/metabolismo , Animales , Calcio/metabolismo , Caveolina 3/metabolismo , Diferenciación Celular , Citoesqueleto/metabolismo , Distroglicanos/metabolismo , Homeostasis , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Técnicas de Placa-Clamp , Unión Proteica , Sarcolema/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA