Article
To read the full-text of this research, you can request a copy directly from the authors.

Abstract

Background and purpose: 1-Methyl-4-phenylpyridinium (MPP(+) ), a potent parkinsonizing agent in primates and rodents, is a blocker of mitochondrial complex I, therefore MPP(+) -induced parkinsonism is believed to depend largely on mitochondrial impairment. However, it has recently been proposed that other mechanisms may participate in MPP(+) -induced toxicity. We tackled this issue by probing the effects of an acute application of MPP(+) on substantia nigra pars compacta (SNc) dopamine (DA) neurons. Experimental approach: The effects of MPP(+) on SNc DA neurons in acute midbrain slices were investigated with electrophysiology techniques. Key results: MPP(+) (50 μM) was able to (i) hyperpolarize SNc DA neurons by ∼6 mV; (ii) cause an abrupt and marked (over 50%) reduction of the spontaneous activity; and (iii) inhibit the hyperpolarization-activated inward current (Ih ). MPP(+) shifted Ih activation curve towards negative potentials by ∼11 mV both in Wistar rats and in C57/BL6 mice. Inhibition was voltage- and concentration-dependent (Imax = 47%, IC50 = 7.74 μM). MPP(+) slowed Ih activation kinetics at all potentials. These effects were not dependent on (i) block of mitochondrial complex I/fall of ATP levels; (ii) activation of type 2 DA receptor; and (iii) alteration of cAMP metabolism. Finally, MPP(+) -dependent inhibition of Ih facilitated temporal summation of evoked EPSPs in SNc DA, but not in CA1 hippocampal neurons. Conclusion and implications: Reduced functionality of Ih in SNc DA neurons, via increased responsiveness towards synaptic excitation, might play a role in MPP(+) -induced parkinsonism and, possibly, in the pathogenesis of human Parkinson's.

No full-text available

Request Full-text Paper PDF

To read the full-text of this research,
you can request a copy directly from the authors.

... The authors conclude that astrocytic GABA release inhibits the pacemaker firing of DAergic neurons in PD models [7]. In line with this evidence, Masi and colleagues [55] similarly reported that MPP + reduces the spontaneous firing of SNpc DAergic neurons when acutely applied to midbrain slices. In addition, they show that MPP + directly inhibits the hyperpolarization-activated current (I h ), a hallmark of DAergic neurons [56] highly sensitive to pathological conditions of the DAergic system [8,57]. ...
... This effect produces an increase in the temporal summation of excitatory inputs to SNpc DAergic neurons, thus increasing spike probability and overall network excitability. Another study described the early and late effects of MPP + on SNpc DAergic neuron excitability [58], confirming the MPP + -mediated I h inhibition in Masi et al. [55]. However, this was not causative of firing inhibition/membrane potential hyperpolarization, as it was not prevented by I h blockade. ...
... Notwithstanding, functional properties of surviving nigral DAergic neurons were mostly preserved, with no overt alterations in the spontaneous firing nor in the intrinsic excitability in mutated rats [8]. Interestingly, spontaneous α-syn overexpression was associated with a decrease in the I h current, mediated by the HCN channels [8], an electrophysiological signature of nigral DAergic neurons, that is similarly reduced in the MPTP PD model [55,84], whereas the function of the DA D 2 autoreceptor and GABAergic GABA B were not affected [8]. ...
Article
Full-text available
The degeneration of nigral dopaminergic neurons is considered the hallmark of Parkinson’s disease (PD), and it is triggered by different factors, including mitochondrial dysfunction, Lewy body accumulation, neuroinflammation, excitotoxicity and metal accumulation. Despite the extensive literature devoted to unravelling the signalling pathways involved in neuronal degeneration, little is known about the functional impairments occurring in these cells during illness progression. Of course, it is not possible to obtain direct information on the properties of the dopaminergic cells in patients. However, several data are available in the literature reporting changes in the function of these cells in PD animal models. In the present manuscript, we focus on dopaminergic neuron functional properties and summarize shared or peculiar features of neuronal dysfunction in different PD animal models at different stages of the disease in an attempt to design a picture of the functional modifications occurring in nigral dopaminergic neurons during disease progression preceding their eventual death.
... In addition to the classical mitochondrial impairment mechanism, several studies have also described the effects of MPP + on neuronal electrophysiological activities. It hyperpolarized the membrane potential and reduced the spontaneous firing rate of dopaminergic neurons (Liss et al., 2005;Masi et al., 2013;Yee et al., 2014). However, the ionic mechanisms of this toxin on neuronal electrophysiological properties are not clear. ...
... However, the ionic mechanisms of this toxin on neuronal electrophysiological properties are not clear. Hyperpolarization-activated cyclic nucleotide-gated channels (HCN channels), ATP-sensitive potassium (K-ATP) channels and DAT are proposed to be related to the neurotoxicity, which still remain controversial (Liss et al., 2005;Masi et al., 2013;Yee et al., 2014). Therefore, in this study, we first elucidated the acute effects of MPP + on electrical activities in nigral dopaminergic neurons, including spontaneous and evoked firing, resting membrane potential and Sag. ...
... Liss et al. (2005) first reported that acute application of MPP + reduced pacing activity of dopaminergic neurons by inhibiting mitochondrial complex I and activating K-ATP channels. Later, Masi et al. (2013) proposed that the reduced functionality of I h in SNc dopaminergic neurons, via increased responsiveness towards synaptic excitation, might play a role in MPP +induced parkinsonism. These researches revealed the acute action of this toxic on electrophysiological properties of dopaminergic neurons. ...
Article
Ghrelin, a 28 amino acid brain-gut peptide, has attracted increasing attention for its neuroprotective effect in Parkinson’s disease (PD). In view of the pivotal role of excitability of dopaminergic neurons in substantia nigra pars compacta (SNc) in the function of nigrostriatal system, it is of great significance to elucidate the regulation of electrical activity of dopaminergic neurons by ghrelin, especially in PD pathogenesis. In this study, we tackled this issue by probing the effects of ghrelin on the electrophysiological properties of dopaminergic neurons in acute application of Methyl-4-phenylpyridinium (MPP⁺), a potent parkinsonizing agent in primates and rodents, with whole cell patch clamp technique. We first observed that MPP⁺ (10, 20 and 50 μM) inhibited the spontaneous firing activity of dopaminergic neurons with dose-dependent and time-dependent properties. MPP⁺ also hyperpolarized the membrane potential, inhibited the evoked firing activity and reduced the amplitude of the inward rectification characteristic (Sag) in dopaminergic neurons. Importantly, ghrelin (100 nM) could improve the above effects of MPP⁺ on the electrical activities of dopaminergic neurons. The potential mechanism of this phenomenon may be that ghrelin upregulated hyperpolarization-activated cyclic nucleotide-gated channel current (Ih) to antagonize the inhibition of MPP⁺ on Ih, thereby improving the electrical activities of dopaminergic neurons.
... More recently, another family of voltage-gated ion channels has been proposed as an intrinsic determinant of the vulnerable phenotype of SNc DAergic neurons. With brain slice patch clamp recordings, the authors of the present manuscript demonstrated that the PD-causing toxin MPP + inhibits Hyperpolarizationactivated cyclic nucleotide-gated (HCN) channels in SNc DAergic neurons (Masi et al., 2013). HCN channels conduct a hyperpolarization-activated, inward cationic current which promotes pace-making and controls synaptic excitability (Mercuri et al., 1995;Zolles et al., 2006;Masi et al., 2013). ...
... With brain slice patch clamp recordings, the authors of the present manuscript demonstrated that the PD-causing toxin MPP + inhibits Hyperpolarizationactivated cyclic nucleotide-gated (HCN) channels in SNc DAergic neurons (Masi et al., 2013). HCN channels conduct a hyperpolarization-activated, inward cationic current which promotes pace-making and controls synaptic excitability (Mercuri et al., 1995;Zolles et al., 2006;Masi et al., 2013). In SNc Daergic neurons, HCN current is carried by the HCN2 and 4 isoforms, which distinguish themselves from isoforms 1 and 3 for higher cAMP sensitivity (Neuhoff et al., 2002). ...
... In SNc Daergic neurons, HCN current is carried by the HCN2 and 4 isoforms, which distinguish themselves from isoforms 1 and 3 for higher cAMP sensitivity (Neuhoff et al., 2002). HCN current inhibition causes rapid reduction of autonomous spiking and marked increase in synaptic excitability (Masi et al., 2013). Furthermore, the electrophysiological effects exerted by pharmacological inhibition of HCN current, as well as the ensuing somatic Ca 2+ inflow, is larger in vulnerable SNc as compared to resistant VTA DAergic neurons (Masi et al., 2015). ...
Article
Full-text available
Progressive neuronal death is the key pathogenic event leading to clinical symptoms in neurodegenerative disorders (NDDs). Neuroprotective treatments are virtually unavailable, partly because of the marked internal heterogeneity of the mechanisms underlying pathology. Targeted neuroprotection would require deep mechanistic knowledge across the entire aetiological spectrum of each NDD and the development of tailored treatments. Although ideal, this strategy appears challenging, as it would require a degree of characterization of both the disease and the patient that is currently unavailable. The alternate strategy is to search for commonalities across molecularly distinct NDD forms and exploit these for the development of drugs with broad-spectrum efficacy. In this view, mounting evidence points to ionic mechanisms (IMs) as targets with potential therapeutic efficacy across distinct NDD subtypes. The scope of this review is to present clinical and preclinical evidence supporting the link between disruption of IMs and neuronal death in specific NDDs and to critically revise past and ongoing attempts of harnessing IMs for the development of neuroprotective treatments.
... Functional HCN channels have been found in distal dendrites of several neuronal populations. In dendritic compartments, active HCN channels reduce input resistance and accelerate the decay phase of Excitatory Post-Synaptic Potentials (EPSP), thereby limiting the amplitude and duration of single EPSPs and dampening temporal summation of EPSP trains in cortical [76,81], thalamic [82] and midbrain dopaminergic neurons [83]. Furthermore, thanks to an increasing soma-to-dendrite expression gradient found in CA1 hippocampal and cortical neurons, HCN channels normalize the duration of EPSPs originating at different distances from the soma, thus preventing loss of information [84][85]. ...
... Structure of other HCN channels modulators. minergic neurons [83]. MPP + , used at concentrations similar to those used to induce nigrostriatal degeneration in rodents, blocked I h in a dose-and voltage-dependent manner: 50 µM MPP + shifted I h activation curve towards negative potentials, and reduced I h amplitude to 44% of maximum at -75 mV, while the current was nearly unchanged at -135 mV. ...
... As mentioned in section 5, MPP + , a toxin capable of inducing PD-like selective nigrostriatal degeneration in primates and rodents, is a voltage-dependent inhibitor of HCN channels in SNc DA neurons in vitro. This block is effective at physiological potentials and results in increased dendritic excitability and overall responsiveness to excitatory synaptic inputs [83]. Altogether, these findings suggest that altered HCN channels expression or function in experimental PD models deserves in-depth scrutiny, especially within the context of differential vulnerability. ...
Article
Full-text available
Hyperpolarization-activated, cyclic nucleotide-gated (HCN) channels, the molecular correlate of the hyperpolarization-activated current (If/Ih), are membrane proteins which play an important role in several physiological processes and various pathological conditions. In the Sino Atrial Node (SAN) HCN4 is the target of ivabradine, a bradycardic agent that is, at the moment, the only drug which specifically blocks If. Nevertheless, several other pharmacological agents have been shown to modulate HCN channels, a property that may contribute to their therapeutic activity and/or to their side effects. HCN channels are considered potential targets for developing drugs to treat several important pathologies, but a major issue in this field is the discovery of isoform-selective compounds, owing to the wide distribution of these proteins into the central and peripheral nervous systems, heart and other peripheral tissues. This survey is focused on the compounds that have been shown, or have been designed, to interact with HCN channels and on their binding sites, with the aim to summarize current knowledge and possibly to unveil useful information to design new potent and selective modulators.
... However, it appears likely that an SNc-specific interplay of pathogenic determinants, rather than individual factors, underlies selective nigrostriatal degeneration (Brichta & Greengard, 2014). In this respect, we recently demonstrated that MPP+, the active metabolite of the parkinsonizing toxin MPTP, inhibits Ih at physiological potentials, thus promoting temporal summation of evoked EPSPs in SNc DA neurons in vitro (Masi et al., 2013). Ih is expressed at different magnitude among subsets of midbrain DA neurons, thus determining differential subthreshold membrane properties and pace-making control (Franz et al., 2000;Neuhoff et al., 2002). ...
... Block of GABAergic transmission was deliberately omitted in these experiments in order to assess the overall contribution of Ih on synaptic excitability of midbrain DA neurons. Moreover, block of GABAergic transmission did not affect EPSP kinetics in control conditions or after Ih suppression in rat SNc DA neurons (Masi et al., 2013). As shown in Figure ...
... Postsynaptic HCN channels affect dendritic excitability of SNc and VTA DA neurons differentially We have previously demonstrated that micromolar MPP+ inhibits Ih causing somatic EPSP summation in acute midbrain slices (Masi et al., 2013). The diverse expression and biophysical properties of HCN channels were previously associated to differential intrinsic electrical properties in characterized subsets of midbrain DA neurons (Franz et al., 2000;Neuhoff et al., 2002), but never investigated in the context of synaptic excitability. ...
Article
Full-text available
The selective vulnerability of Substantia Nigra pars compacta (SNc) dopaminergic (DA) neurons is an enigmatic trait of Parkinson's disease (PD), especially if compared to the remarkable resistance of closely related DA neurons in the neighboring Ventral Tegmental Area (VTA). Overall evidence indicates that specific electrophysiological, metabolic and molecular factors underlie SNc vulnerability, although many pieces of the puzzle are still missing. In this respect, we recently demonstrated that 1-methyl-4-phenylpyridinium (MPP+), the active metabolite of the parkinsonizing toxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), alters the electrophysiological properties of SNc DA neurons in vitro, by inhibiting the Hyperpolarization-activated current (Ih). Here, we present an electrophysiological investigation of the functional role of Ih in the integration of synaptic inputs in identified SNc and VTA DA neurons, comparatively, in acute midbrain slices from TH-GFP mice. We show that pharmacological suppression of Ih increases the amplitude and decay time of Excitatory Postsynaptic Potentials (EPSPs), leading to temporal summation of multiple excitatory potentials at somatic level. Importantly, these effects are quantitatively more evident in SNc DA neurons. We conclude that Ih regulates the responsiveness to excitatory synaptic transmission in SNc and VTA DA neurons differentially. Finally, we present the hypothesis that Ih loss of function may be linked to PD trigger mechanisms, such as mitochondrial failure and ATP depletion, and act in concert with SNc-specific synaptic connectivity to promote selective vulnerability.This article is protected by copyright. All rights reserved.
... Neurons were held at −70 mV using constant current injection to suppress spontaneous firing and to achieve comparable conditions before and after the application of ZD 7288 (30 μM). Bath application of ZD 7288 reduced the sag and I h (Harris & Constanti, 1995;Neuhoff et al. 2002;Masi et al. 2013), increased the input resistance and hyperpolarized DA neurons. ...
... However, the effect was not different between the two types of dendrite. An increase of temporal summation consecutive to an indirect inhibition of I h has been reported previously using electrically evoked EPSPs (Masi et al. 2013), indicating that the effect of I h can be observed both on artificial and on electrically evoked multiple EPSPs. ...
... In the pathophysiological context, a study using an in vitro model of Parkinson's disease shows an increase in temporal summation in nigral DA neurons in the presence of a neurotoxin implicated in the inhibition of I h , which in turn may drive elevated excitability (Masi et al. 2013). Remarkably, alterations of h-channels were also reported for external globus pallidus neurons where they influence the transition between bursting and pacemaking . ...
Article
Key points: The hyperpolarization-activated cation current Ih is expressed in dopamine neurons of the substantia nigra, but the subcellular distribution of the current and its role in synaptic integration remain unknown. We used cell-attached patch recordings to determine the localization profile of Ih along the somatodendritic axis of nigral dopamine neurons in slices from young rats. Ih density is higher in axon-bearing dendrites, in a membrane area close to the axon origin, than in the soma and axon-lacking dendrites. Dual current-clamp recordings revealed a similar contribution of Ih to the waveform of single excitatory postsynaptic potentials throughout the somatodendritic domain. The Ih blocker ZD 7288 increased the temporal summation in all dendrites with a comparable effect in axon- and non-axon dendrites. The strategic position of Ih in the proximity of the axon may influence importantly transitions between pacemaker and bursting activities and consequently the downstream release of dopamine. Abstract: Dendrites of most neurons express voltage-gated ion channels in their membrane. In combination with passive properties, active currents confer to dendrites a high computational potential. The hyperpolarization-activated cation current Ih present in the dendrites of some pyramidal neurons affects their membrane and integration properties, synaptic plasticity and higher functions such as memory. A gradient of increasing h-channel density towards distal dendrites has been found to be responsible for the location independence of excitatory postsynaptic potential (EPSP) waveform and temporal summation in cortical and hippocampal pyramidal cells. However, reports on other cell types revealed that smoother gradients or even linear distributions of Ih can achieve homogeneous temporal summation. Although the existence of a robust, slowly activating Ih current has been repeatedly demonstrated in nigral dopamine neurons, its subcellular distribution and precise role in synaptic integration are unknown. Using cell-attached patch-clamp recordings, we find a higher Ih current density in the axon-bearing dendrite than in the soma or in dendrites without axon in nigral dopamine neurons. Ih is mainly concentrated in the dendritic membrane area surrounding the axon origin and decreases with increasing distances from this site. Single EPSPs and temporal summation are similarly affected by blockade of Ih in axon- and non-axon-bearing dendrites. The presence of Ih close to the axon is pivotal to control the integrative functions and the output signal of dopamine neurons and may consequently influence the downstream coding of movement.
... It has previously been shown that MPP + inhibits pacemaker activity of SNc neurons due to mitochondrial Complex I inhibition and activation of ATP-sensitive potassium (K ATP ) channels, and that silencing of these neurons through K ATP channel activation promotes neural damage (Liss et al., 2005). However, a recent study reported that MPP + -induced cell membrane hyperpolarisation and rapid inhibition of firing of SNc neurons do not involve K ATP channel activation, and that these effects solely depend on the direct ability of MPP + to block hyperpolarisation-activated cyclic nucleotide-gated (HCN) channels (Masi et al., 2013). ...
... The bath concentration of MPP + (20 mM) was chosen to be comparable with the effects described in two previous studies which were also conducted in midbrain slices (Liss et al., 2005; 10 mM; Masi et al., 2013;50 mM). In intracellular recording experiments, DA (30 mM, 10 s) was applied through a U-tube rapid perfusion system (Fenwick et al., 1982). ...
... We first tested the effects of MPP + on the firing frequency of dopaminergic and non-dopaminergic SNc neurons using singleunit recording. Consistent with previous studies (Liss et al., 2005;Masi et al., 2013), MPP + (20 mM; 30 min) slowed or stopped the spontaneous firing of dopaminergic neurons (from baseline followed by complete cessation of firing following MPP + (20 mM, 30 min) application. A2: Normalised data demonstrating that while the firing rate of dopaminergic neurons (n = 19) was decreased, the firing of non-dopaminergic neurons (n = 5) was enhanced by the neurotoxin. ...
... In MitoPark mice, a wellestablished animal model of PD, a reduction in the HCN current density in SNc dopaminergic neurons was observed, followed by the initiation of epileptic seizures (Good et al., 2011). Moreover, recent research has reported some evidence of a relationship between PD pathogenesis in humans and dysfunctional HCN channels by analyzing electrophysiological data obtained from models treated with the toxic chemical 1-methyl-4-phenylpyridinium (MPP+), which can induce PD-like selective degeneration of nigral dopaminergic neurons, suggesting that HCN blockage from MPP+ treatment may increase synaptic excitability (Masi et al., 2013). ...
... The heterogeneous distribution of HCN channel isoforms enables efficient summation of EPSPs (Berger et al., 2003;Benarroch, 2013). Neurological conditions including epilepsy and PD have been related to dysfunctional HCN channels that impact basal ganglia circuitry, neuronal firing patterns, and neuronal excitability (Kase and Imoto, 2012;Masi et al., 2013;Chang et al., 2019;Kessi et al., 2022). For the purpose of creating novel therapies for these disorders, it seems critical to fully comprehend the function and control of HCN channels. ...
Article
Full-text available
Transmembrane proteins known as hyperpolarization-activated cyclic nucleotide-gated (HCN) channels control the movement of Na⁺ and K⁺ ions across cellular membranes. HCN channels are known to be involved in crucial physiological functions in regulating neuronal excitability and rhythmicity, and pacemaker activity in the heart. Although HCN channels have been relatively well investigated in the brain, their distribution and function in the retina have received less attention, remaining their physiological roles to be comprehensively understood. Also, because recent studies reported HCN channels have been somewhat linked with the dysfunction of photoreceptors which are affected by retinal diseases, investigating HCN channels in the retina may offer valuable insights into disease mechanisms and potentially contribute to identifying novel therapeutic targets for retinal degenerative disorders. This paper endeavors to summarize the existing literature on the distribution and function of HCN channels reported in the vertebrate retinas of various species and discuss the potential implications for the treatment of retinal diseases. Then, we recapitulate current knowledge regarding the function and regulation of HCN channels, as well as their relevance to various neurological disorders.
... In a genetic mitochondrial model of Parkinson's disease, the HCN channel functions reduce significantly in midbrain dopaminergic neurons (Good et al., 2011). Furthermore, application of 1-Methyl-4phenylpyridinium (MPP + ) inhibits the activity of HCN channels in substantia nigra dopaminergic neurons (Masi et al., 2013). ...
... It is known that HCN channels are involved in the integration of dendritic excitatory glutamatergic synaptic inputs in a variety of neurons. Activation of HCN channels reduce the amplitude and time course of excitatory postsynaptic potentials (Williams and Stuart, 2000;Angelo et al., 2007;Bullis et al., 2007;Ying et al., 2007;Masi et al., 2013). As our previous studies in normal rats, the interaction of HCN channels with glutamatergic transmission may be one of the possible reasons in ZD7288-induced increase in firing rate (Chen et al., 2015). ...
Article
Full-text available
The globus pallidus occupies a critical position in the indirect pathway of the basal ganglia motor control system. Hyperpolarization-activated cyclic-nucleotide gated (HCN) channels play an important role in the modulation of neuronal excitability. In vivo extracellular single unit recording, behavioral test and immunohistochemistry were performed to explore the possible modulation of endogenous HCN channels in the globus pallidus under parkinsonian states. In MPTP parkinsonian mice, micro-pressure application of the selective HCN channel antagonist, ZD7288, decreased the firing rate in 10 out of the 28 pallidal neurons, while increased the firing rate in another 15 out of the 28 neurons. In 6-OHDA parkinsonian rats, ZD7288 also bidirectionally regulated the spontaneous firing activity of the globus pallidus neurons. The proportion of pallidal neurons with ZD7288-induced slowing of firing rate tended to reduce in both parkinsonian animals. Morphological studies revealed a weaker staining of HCN channels in the globus pallidus under parkinsonian state. Finally, behavioral study demonstrated that intrapallidal microinjection of ZD7288 alleviated locomotor deficits in MPTP parkinsonian mice. These results suggest that endogenous HCN channels modulate the activities of pallidal neurons under parkinsonian states.
... In agreement with this, CB1Rs have been shown to play a pivotal role in the control of movement and pathogenesis of some movement disorders such as PD by modulating GABA, glutamate, DA and other neurotransmitters throughout the BG. Additionally, dysfunctional HCN channels which mediates Ih currents have been suggested to be involved in different experimental models of PD 55,56 , and evidence for a potentially relevant role of Ih currents in the pathogenesis of PD has grown considerably 43 . Our study has attempted to clarify a link between Ih and CB1R in an animal model of PD. ...
Preprint
Full-text available
Based on data from our lab and others, the endocannabinoid system (ECB S ) appears to be involved in PD-related processes. Therefore, we compared the motor and non-motor effects of the cannabinoid receptor type 1 (CB1R) WIN 55,212-2 (WIN) and selective antagonist (AM251), on motor and non-motor symptoms (NMS) of PD in a mice model generated by an intracerebroventricular (i.c.v.) injection of 6-hydroxydopamine (6-OHDA). To provide further knowledge about the link of CB1R with hyperpolarization-activated current (Ih), we conducted ex vivo investigations in the ventral tegmental area (VTA). In the current study, pharmacological blockage of CB1R ameliorated explorative behaviors, balance, muscle strength, and passive avoidance memory deficits induced by 6-OHDA, however, anxious, and depressive-like behaviors were heightened. 6-OHDA exposure induced severe alterations in the spontaneous and evoked firing behavior of DA neurons as evidenced by a significant increase in the mean number of spikes and a decrease in half-width, respectively. Interestingly, an increase in the amplitude of the sag voltage and in the amplitude of the steady state Ih currents was seen. WIN exacerbated 6-OHDA-induced actions by further reducing the spike half-width and increasing the firing frequency, as well as increasing sEPSP amplitudes. The effects of 6-OHDA on sag voltage, Ih currents amplitude, and firing frequency were reversed by administration of AM251. These results suggest that ECBs might underlie some of the 6-OHDA-induced electrophysiological alterations in VTA DA neurons in this animal model of PD and antagonist of this receptor could be effective in modulating the devastating effects of 6-OHDA.
... Dysfunctional HCN channels are reported to mediate MPP + -induced toxicity (Masi et al., 2013). The mitochondrial failure and ATP depletion caused by MPP + results in the opening of K-ATP that eventually inhibits HCN channels. ...
Article
Parkinson’s disease (PD) is primarily associated with the progressive neurodegeneration of the dopaminergic neurons in the substantia nigra region of the brain. The resulting motor symptoms are managed with the help of dopamine replacement therapies. However, these therapeutics do not prevent the neurodegeneration underlying the disease and therefore lose their effectiveness in managing disease symptoms over time. Thus, there is an urgent need to develop newer therapeutics for the benefit of patients. The release of dopamine and the firing activity of substantia nigra neurons is regulated by several ion channels that act in concert. Dysregulations of these channels cause the aberrant movement of various ions in the intracellular milieu. This eventually leads to disruption of intracellular signalling cascades, alterations in cellular homeostasis, and bioenergetic deficits. Therefore, ion channels play a central role in driving the high vulnerability of dopaminergic neurons to degenerate during PD. Targeting ion channels offers an attractive mechanistic strategy to combat the process of neurodegeneration. In this review, we highlight the evidence pointing to the role of various ion channels in driving the PD processes. In addition, we also discuss the various drugs or compounds that target the ion channels and have shown neuroprotective potential in the in-vitro and in-vivo models of PD. We also discuss the current clinical status of various drugs targeting the ion channels in the context of PD.
... Using brain slice patch clamp recordings, it has been reported that MPP+ causes I h inhibition in SNc DA neurons in a dose-dependent manner. Of note, MPP + -induced I h block increases synaptic excitability as measured in terms of excitatory postsynaptic potentials amplitude [95]. When I h is blocked with selective drugs, the magnitude of excitatory synaptic response, as well as the associated calcium response, is significantly greater in vulnerable (i.e., SNc) compared to resistant (i.e., VTA) DA populations [96], and local pharmacological blockade of I h causes selective nigrostriatal DA degeneration [25]. ...
Article
Full-text available
Hyperpolarization-activated cyclic nucleotide–gated (HCN) channels are expressed as four different isoforms (HCN1-4) in the heart and in the central and peripheral nervous systems. In the voltage range of activation, HCN channels carry an inward current mediated by Na+ and K+, termed If in the heart and Ih in neurons. Altered function of HCN channels, mainly HCN4, is associated with sinus node dysfunction and other arrhythmias such as atrial fibrillation, ventricular tachycardia, and atrioventricular block. In recent years, several data have also shown that dysfunctional HCN channels, in particular HCN1, but also HCN2 and HCN4, can play a pathogenic role in epilepsy; these include experimental data from animal models, and data collected over genetic mutations of the channels identified and characterized in epileptic patients. In the central nervous system, alteration of the Ih current could predispose to the development of neurodegenerative diseases such as Parkinson’s disease; since HCN channels are widely expressed in the peripheral nervous system, their dysfunctional behavior could also be associated with the pathogenesis of neuropathic pain. Given the fundamental role played by the HCN channels in the regulation of the discharge activity of cardiac and neuronal cells, the modulation of their function for therapeutic purposes is under study since it could be useful in various pathological conditions. Here we review the present knowledge of the HCN-related channelopathies in cardiac and neurological diseases, including clinical, genetic, therapeutic, and physiopathological aspects.
... Briefly, the slices were preconditioned with NMDA or DHPG and 24h later sEPSCs were recorded from CA1 pyramidal neurons. The slices were then removed from the culture disc and placed in a recording chamber mounted on an upright microscope (Nikon Eclipse E600 FN) equipped with IR-DIC optics and an infrared camera (Hamamatsu) for visually guided experiments (Masi, Narducci, Landucci, Moroni, & Mannaioni, 2013). Specifically, inward currents were recorded in CA1 pyramidal cells for 5 min under control condition (basal level), for 5 min during the application of 0.3 µM AMPA and for 5 min after the wash for each experimental condition. ...
Article
We investigated the molecular events triggered by NMDA and 3,5‐dihydroxyphenylglycine (DHPG) preconditioning, that lead to neuroprotection against excitotoxic insults (AMPA or oxygen and glucose deprivation) in rat organotypic hippocampal slices, with particular attention on glutamate receptors and on cannabinoid system. We firstly evaluated the protein expression of NMDA and AMPA receptor subunits after preconditioning using western blot analysis performed in post‐synaptic densities. We observed that following NMDA, but not DHPG preconditioning, the expression of GluA1 was significantly reduced and this reduction appeared to be associated with the internalization of AMPA receptors. Whole‐cell voltage clamp recordings on CA1 pyramidal neurons of organotypic slices show that 24 hr after exposure to NMDA and DHPG preconditioning, AMPA‐induced currents were significantly reduced. To clarify the mechanisms induced by DHPG preconditioning, we then investigated the involvement of the endocannabinoid system. Exposure of slices to the CB1 antagonist AM251 prevented the development of tolerance to AMPA toxicity induced by DHPG but not NMDA. Accordingly, the MAG‐lipase inhibitor URB602, that increases arachidonoylglycerol (2‐AG) content, but not the FAAH inhibitor URB597, that limits the degradation of anandamide, was also able to induce tolerance versus AMPA and OGD toxicity, suggesting that 2‐AG is responsible for the DHPG‐induced tolerance. In conclusion, preconditioning with NMDA or DHPG promotes differential neuroprotective mechanisms: NMDA by internalization of GluA1‐AMPA receptors, DHPG by producing the endocannabinoid 2‐AG. image
... In addition, it has been reported that 1-methyl-4-phenylpyridinium, a toxin capable of inducing PD-like selective nigrostriatal degeneration in primates and rodents, is a voltage-dependent inhibitor of HCN channels in SNc DA neurons. The 1-methyl-4phenylpyridinium-induced HCN block results in increased dendritic excitability and overall responsiveness to excitatory synaptic inputs (Masi et al., 2013). In agreement with relative HCN expression levels, the effect of HCN block on synaptic excitability is significantly greater in SNc compared with VTA DA neurons (Masi et al., 2015). ...
Article
Full-text available
The hyperpolarization-activated cyclic-nucleotide-gated (HCN) proteins are voltage-dependent ion channels, conducting both Na<sup>+</sup> and K<sup>+</sup>, blocked by millimolar concentrations of extracellular Cs+ and modulated by cyclic nucleotides (mainly cAMP) that contribute crucially to the pacemaker activity in cardiac nodal cells and subsidiary pacemakers. Over the last decades, much attention has focused on HCN current in non-pacemaker cells and its potential role in triggering arrhythmias. In fact, in addition to pacemakers, HCN current is constitutively present in the human atria and has since long been proposed to sustain atrial arrhythmias associated to different cardiac pathologies or triggered by various modulatory signals (catecholamines, serotonin, natriuretic peptides). The properties of atrial and ventricular I <sub>f</sub> and its modulation by pharmacological interventions has been object of intense study, including the synthesis and characterization of new compounds able to block preferentially HCN1, HCN2 or HCN4 isoforms. Altogether, clues emerge for opportunities of future pharmacological strategies exploiting the unique properties of this channel family: the prevalence of different HCN subtypes in organs and tissues, the possibility to target HCN gain- or loss-of-function associated with disease, the feasibility of novel isoform-selective drugs as well as the discovery of HCN-mediated effects for old medicines.
... In addition, it has been reported that 1-methyl-4-phenylpyridinium, a toxin capable of inducing PD-like selective nigrostriatal degeneration in primates and rodents, is a voltage-dependent inhibitor of HCN channels in SNc DA neurons. The 1-methyl-4phenylpyridinium-induced HCN block results in increased dendritic excitability and overall responsiveness to excitatory synaptic inputs (Masi et al., 2013). In agreement with relative HCN expression levels, the effect of HCN block on synaptic excitability is significantly greater in SNc compared with VTA DA neurons (Masi et al., 2015). ...
Article
Hyperpolarization-activated, cyclic nucleotide-gated (HCN) channels are important members of the voltage-gated pore loop channels family.They show unique features: they open at hyperpolarizing potential, carry a mixed Na/K current, and are regulated by cyclic nucleotides. Four different isoforms have been cloned (HCN1-4) that can assemble to form homo- or heterotetramers, characterized by different biophysical properties. These proteins are widely distributed throughout the body and involved in different physiologic processes, the most important being the generation of spontaneous electrical activity in the heart and theregulation of synaptic transmission in the brain. Their role in heart rate, neuronal pacemaking, dendritic integration, learning and memory, and visual and pain perceptions has been extensively studied; these channels have been found also in some peripheral tissues, where their functions still need to be fully elucidated. Genetic defects and altered expression of HCN channels are linked to several pathologies, which makes these proteins attractive targets for translational research; at themoment only one drug (ivabradine), which specifically blocks the hyperpolarization-activated current, is clinically available. This review discusses current knowledge about HCN channels, starting from their biophysical properties, origin, and developmental features, to (patho)physiologic role in different tissues and pharmacological modulation, ending with their present and future relevance as drug targets. © 2017 by The American Society for Pharmacology and Experimental Therapeutics.
... The HCN blocker ZD7288 (50 mM for 10 0 ) fully abolished Ih current (Fig. 1B top-right). Interestingly, Ih pharmacological suppression had no discernible effect on Resting Membrane Potential (RMP) (À54.8 ± 2.0 mV to À55.1 ± 3.2 mV, n ¼ 6, p ¼ 0.7, ctrl vs ZD7288, respectively; Fig. 1B bottom-right) showing that the control of the RMP in small DRG neurons differs from that of other neuronal cell types (Biel et al., 2009;Masi et al., 2013). ...
Article
Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels have a key role in the control of cellular excitability. HCN2, a subgroup of the HCN family channels, are heavily expressed in small dorsal root ganglia (DRG) neurons and their activation seems to be important in the determination of pain intensity. Intracellular elevation of cAMP levels activates HCN-mediated current (Ih) and small DRG neurons excitability. GPR35, a Gi/o coupled receptor, is highly expressed in small DRG neurons, and we hypothesized that its activation, mediated by endogenous or exogenous ligands, could lead to pain control trough a reduction of Ih current. Patch clamp recordings were carried out in primary cultures of rat DRG neurons and the effects of GPR35 activation on Ih current and neuronal excitability were studied in control conditions and after adenylate cyclase activation with either forskolin or prostaglandin E2 (PGE2). We found that both kynurenic acid (KYNA) and zaprinast, the endogenous and synthetic GPR35 agonist respectively, were able to antagonize the forskolin-induced depolarization of resting membrane potential by reducing Ih-mediated depolarization. Similar results were obtained when PGE2 was used to activate adenylate cyclase and to increase Ih current and the overall neuronal excitability. Finally, we tested the analgesic effect of both GPR35 agonists in an in vivo model of PGE2-induced thermal hyperalgesia. In accord with the hypothesis, both KYNA and zaprinast showed a dose dependent analgesic effect. In conclusion, GPR35 activation leads to a reduced excitability of small DRG neurons in vitro and causes a dose-dependent analgesia in vivo. GPR35 agonists, by reducing adenylate cyclase activity and inhibiting Ih in DRG neurons may represent a promising new group of analgesic drugs.
... In addition, it has been reported that 1-methyl-4-phenylpyridinium, a toxin capable of inducing PD-like selective nigrostriatal degeneration in primates and rodents, is a voltage-dependent inhibitor of HCN channels in SNc DA neurons. The 1-methyl-4phenylpyridinium-induced HCN block results in increased dendritic excitability and overall responsiveness to excitatory synaptic inputs ( Masi et al., 2013). In agreement with relative HCN expression levels, the effect of HCN block on synaptic excitability is significantly greater in SNc compared with VTA DA neurons ( Masi et al., 2015). ...
Article
The hyperpolarization-activated cyclic nucleotide-gated (HCN) channels play an important role in the generation of pacemaker activity of cardiac sinoatrial node cells and immature cardiomyocytes. HCN channels are also present in adult atrial and ventricular cardiomyocytes, where the physiological role is currently under investigation. In different cardiac pathologies, dysfunctional HCN channels have been suggested to be a direct cause of rhythm disorders. While loss-of-function mutations of HCN channels are associated with sinus bradycardia, HCN channel gain-of-function in atrial fibrillation, ventricular hypertrophy and failure might help enhance ectopic electrical activity and promote arrhythmogenesis. Blockade of HCN channels with ivabradine, a selective bradycardic agent currently available for clinical use, improves cardiac performance and counteracts functional remodeling in experimental hypertrophy. Accordingly, ivabradine ameliorates clinical outcome in patients with chronic heart failure. Novel compounds with enhanced selectivity for cardiac HCN channel isoforms are being studied as potential candidates for new drug development.
... For electrophysiology experiments, rat organotypic hippocampal slices were removed from the culture disc and placed in a recording chamber mounted on an upright microscope (Nikon Eclipse E600 FN) equipped with IR-DIC optics and an infrared camera (Hamamatsu) for visually guided experiments (Hamill et al., 2009;Masi et al., 2013). Slices were continuously bathed with oxygenated aCSF maintained at 33/ 34°C with a TC344B temperature controller (Warner). ...
Article
An excessive activation of poly(ADP-ribose) polymerases (PARPs) may trigger a form of neuronal death similar to that occurring in neurodegenerative disorders. To investigate this process, we exposed organotypic hippocampal slices to N-methyl-N'-nitro-N'-nitrosoguanidine (MNNG, 100 μM for 5min), an alkylating agent widely used to activate PARP-1. MNNG induced a pattern of degeneration of the CA1 pyramidal cells morphologically similar to that observed after a brief period of oxygen and glucose deprivation (OGD). MNNG exposure was also associated with a dramatic increase in PARP-activity and a robust decrease in NAD(+) and ATP content. These effects were prevented by PARP-1 but not PARP-2 inhibitors. In our experimental conditions, cell death was not mediated by AIF translocation (parthanatos) or caspase-dependent apoptotic processes. Furthermore, we found that PARP activation was followed by a significant deterioration of neuronal membrane properties. Using electrophysiological recordings we firstly investigated the suggested ability of ADP-ribose to open TRPM2 channels in MNNG-induced cells death, but the results we obtained showed that TRPM2 channels are not involved. We then studied the involvement of glutamate receptor-ion channel complex and we found that NBQX, a selective AMPA receptor antagonist, was able to effectively prevent CA1 neuronal loss while MK801, a NMDA antagonist, was not active. Moreover, we observed that MNNG treatment increased the ratio of GluA1/GluA2 AMPAR subunit expression, which was associated with an inward rectification of the IV relationship of AMPA sEPSCs in the CA1 but not in the CA3 subfield. Accordingly, 1-naphthyl acetyl spermine (NASPM), a selective blocker of Ca(2+)- permeable GluA2-lacking AMPA receptors, reduced MNNG-induced CA1 pyramidal cell death. In conclusion, our results show that activation of the nuclear enzyme PARP-1 may change the expression of membrane proteins and Ca(2+) permeability of AMPA channels, thus affecting the function and survival of CA1 pyramidal cells.
... recording of eEPSCs, electrodes were filled with (in mM) 140 Kgluconate , 10 HEPES, 7 NaCl, 4 Mg-ATP and 0.3 Na 3 -GTP. EPSCs were evoked from a holding potential of –60 mV by stimulation at a frequency of 0.06 Hz [28]. Stimuli were delivered through a bipolar stimulating electrode (240 µm spacing, FHC Inc., Bowdoinham, ME, USA) placed in the stratum radiatum within 100 µm of the patched cell. ...
Article
Full-text available
Limited information is available on the brain expression and role of GPR35, a Gi/o coupled receptor activated by kynurenic acid (KYNA). In mouse cultured astrocytes, we detected GPR35 transcript using RT-PCR and we found that KYNA (0.1 to 100 µM) decreased forskolin (FRSK)-induced cAMP production (p<0.05). Both CID2745687 (3 µM, CID), a recently described GPR35 antagonist, and GPR35 gene silencing significantly prevented the action of KYNA on FRSK-induced cAMP production. In these cultures, we then evaluated whether GPR35 activation was able to modulate intracellular Ca(2+) concentration ([Ca(2+)]i ) and [Ca(2+)]i fluxes. We found that both KYNA and zaprinast, a phosphodiesterase (PDE) inhibitor and GPR35 agonist, did not modify either basal or peaks of [Ca(2+)]i induced by challenging the cells with ATP (30 µM). However, the [Ca(2+)]i plateau phase following peak was significantly attenuated by these compounds in a store-operated Ca(2+) channel (SOC)-independent manner. The activation of GPR35 by KYNA and zaprinast was also studied at the CA3-CA1 synapse in the rat hippocampus. Evoked excitatory post synaptic currents (eEPSCs) were recorded from CA1 pyramidal neurons in acute brain slices. The action of KYNA on GPR35 was pharmacologically isolated by using NMDA and α7 nicotinic receptor blockers and resulted in a significant reduction of eEPSC amplitude. This effect was prevented in the presence of CID. Moreover, zaprinast reduced eEPSC amplitude in a PDE5- and cGMP-independent mechanism, thus suggesting that glutamatergic transmission in this area is modulated by GPR35. In conclusion, GPR35 is expressed in cultured astrocytes and its activation modulates cAMP production and [Ca(2+)]i. GPR35 activation may contribute to KYNA effects on the previously reported decrease of brain extracellular glutamate levels and reduction of excitatory transmission.
Chapter
Neurons are excitable cells. They use ions and electrical signaling to talk to each to other, and when they talk to each other, neurons control behavior. The Oxford Handbook of Neuronal Ion Channels is an accessible reference describing the nature and properties of ion channels in neurons. The book explains how ion channels open and close, how they can be selective for specific ions, and how they give rise to action potentials. There are in-depth chapters discussing specific classes of ion channels: potassium channels, sodium channels, neurotransmitter-gated ion channels, and other specialized channels. Throughout the handbook, important insight is provided on the contribution ion channels make to neuronal excitability and to synaptic transmission. The handbook goes further to discuss a group of human diseases such as epilepsy, pain, and migraines that can be caused by ion channel dysfunction called channelopathies. For neuroscientists, biophysicists, and neuropharmacologists, this handbook is a valuable reference of ion channel biology and function.
Article
It is widely acknowledged that ethanol (EtOH) can alter many neuronal functions, including synaptic signaling, firing discharge, and membrane excitability, through its interaction with multiple membrane proteins and intracellular pathways. Previous work has demonstrated that EtOH enhances the firing rate of hippocampal GABAergic interneurons and thus the presynaptic GABA release at CA1 and CA3 inhibitory synapses through a positive modulation of the hyperpolarization-activated cyclic nucleotide-gated cation (HCN) channels. Activation of HCN channels produce an inward current, commonly called Ih, which plays an essential role in generating/regulating specific neuronal activities in GABAergic interneurons and principal glutamatergic pyramidal neurons such as those in the CA3 subregion. Since the direct effect of EtOH on HCN channels expressed in CA3 pyramidal neurons was not thoroughly elucidated, we investigated the possible interaction between EtOH and HCN channels and the impact on excitability and postsynaptic integration of these neurons. Patch-clamp recordings were performed in single CA3 pyramidal neurons from acute male rat coronal hippocampal slices. Our results show that EtOH modulates HCN-mediated Ih in a concentration-dependent and bi-directional manner, with a positive modulation at lower (20 mM) and an inhibitory action at higher (60-80 mM) concentrations. The modulation of Ih by EtOH was mimicked by forskolin, antagonized by different drugs that selectively interfere with the AC/cAMP/PKA intracellular pathway, as well as by the selective HCN inhibitor ZD7288. Altogether, these data further support the evidence that HCN channels may represent an important molecular target through which EtOH may regulate neuronal activity.
Article
Full-text available
Background: The accumulation of α-synuclein (α-syn) fibrils in intraneuronal inclusions called Lewy bodies and Lewy neurites is a pathological signature of Parkinson’s disease (PD). Although several aspects linked to α-syn–dependent pathology (concerning its spreading, aggregation, and activation of inflammatory and neurodegenerative processes) have been under intense investigation, less attention has been devoted to the real impact of α-syn overexpression on structural and functional properties of substantia nigra pars compacta (SNpc) dopamine (DA) neurons, particularly at tardive stages of α-syn buildup, despite this has obvious relevance to comprehending mechanisms beyond PD progression. Objectives: We aimed to determine the consequences of a prolonged α-syn overexpression on somatodendritic morphology and functions of SNpc DA neurons. Methods: We performed immunohistochemistry, stereological DA cell counts, analyses of dendritic arborization, ex vivo patch-clamp recordings, and in vivo DA microdialysis measurements in a 12- to 13-month-old transgenic rat model overexpressing the full-length human α-syn (Snca+/+) and age-matched wild-type rats. Results: Aged Snca+/+ rats have mild loss of SNpc DA neurons and decreased basal DA levels in the SN. Residual nigral DA neurons display smaller soma and compromised dendritic arborization and, in parallel, increased firing activity, switch in firing mode, and hyperexcitability associated with hypofunction of fast activating/ inactivating voltage-gated K+ channels and Ca2+- and voltage-activated large conductance K+ channels. These intrinsic currents underlie the repolarization/ afterhyperpolarization phase of action potentials, thus affecting neuronal excitability. Conclusions: Besides clarifying α-syn–induced pathological landmarks, such evidence reveals compensatory functional mechanisms that nigral DA neurons could adopt during PD progression to counteract neurodegeneration
Article
Recent evidence from genetic and pharmacological animal models of Parkinson’s disease (PD) suggests alteration in activity of hyperpolarization-activated cyclic nucleotide-gated channels (HCN) occurs following dopamine (DA) depletion. Further, based on data from our lab and others, the endocannabinoid system (ECBS) appears to be involved in PD-related processes. Therefore, we compared the motor and non-motor effects of an intracerebroventricular (i.c.v.) injection of the cannabinoid receptor type 1 (CB1R) agonist WIN 55,212-2 (WIN) and selective antagonist AM251 (AM) on motor and non-motor symptoms (NMS) of PD in a mouse model generated by an i.c.v. injection of 6-hydroxydopamine (6-OHDA). To provide further knowledge about the link between CB1R and the hyperpolarization-activated current (Ih), we conducted ex vivo investigations in the ventral tegmental area (VTA). In the current study, pharmacological antagonism of CB1R ameliorated explorative behaviors, balance, muscle strength, and passive avoidance memory deficits induced by 6-OHDA, however, anxious, and depressive-like behaviors were heightened. The AM was also effective in reducing the 6-OHDA-induced TH level deficit. 6-OHDA exposure induced severe alterations in the spontaneous and evoked firing behavior of DA neurons, as evidenced by a significant increase in the mean number of spikes and a decrease in spike half-width, respectively. Interestingly, an increase in the amplitude of the sag voltage and in the amplitude of the steady state Ih current was seen. Consistent with an effect of increasing Ih, WIN exacerbated 6-OHDA-induced actions by further reducing the spike half-width and increasing the firing frequency. In addition, greater amplitudes of sEPSPs were elicited. The effects of 6-OHDA on sag voltage, Ih current amplitude, and firing frequency were reversed by administration of AM. These results suggest that ECBs might be involved in some of the 6-OHDA-induced electrophysiological alterations in VTA DA neurons in this animal model of PD. In addition, the CB1R antagonistic mechanism could be effective in modulating the devastating effects of 6-OHDA.
Article
Full-text available
Neurodegenerative diseases such as Parkinson's disease (PD), Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), and spinal muscular atrophy (SMA) are chronic, progressive, and age-associated neurological disorders characterized by neuronal deterioration in specific brain regions. Although the specific pathological mechanisms underlying these disorders have remained elusive, ion channel dysfunction has become increasingly accepted as a potential mechanism for neurodegenerative diseases. Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are encoded by the HCN1-4 gene family and conduct the hyperpolarization-activated current (Ih). These channels play important roles in modulating cellular excitability, rhythmic activity, dendritic integration, and synaptic transmission. In the present review, we first provide a comprehensive picture of the role of HCN channels in PD by summarizing their role in the regulation of neuronal activity in PD-related brain regions. Dysfunction of Ih may participate in 1-methyl-4-phenylpyridinium (MPP+)-induced toxicity and represent a pathogenic mechanism in PD. Given current reports of the critical role of HCN channels in neuroinflammation and depression, we also discussed the putative contribution of HCN channels in inflammatory processes and non-motor symptoms in PD. In the second section, we summarize how HCN channels regulate the formation of β-amyloid peptide in AD and the role of these channels in learning and memory. Finally, we briefly discuss the effects of HCN channels in ALS and SMA based on existing discoveries.
Article
Full-text available
Hyperpolarization-activated cyclic nucleotide-gated cation current (I h ) plays important roles in the achievement of many physiological/pathological functions in the nervous system by modulating the electrophysiological activities, such as the rebound (spike) to hyperpolarization stimulations, subthreshold membrane resonance to sinusoidal currents, and spike-timing precision to stochastic factors. In the present paper, with increasingg h (conductance ofI h ), the rebound (spike) and subthreshold resonance appear and become stronger, and the variability of the interspike intervals (ISIs) becomes lower, i.e., the enhancement of spike-timing precision, which are simulated in a conductance-based theoretical model and well explained by the nonlinear concept of bifurcation. With increasingg h , the stable node to stable focus, to coexistence behavior, and to firing via the codimension-1 bifurcations (Hopf bifurcation, saddle-node bifurcation, saddle-node bifurcations on an invariant circle, and saddle homoclinic orbit) and codimension-2 bifurcations such as Bogdanov-Takens (BT) point related to the transition between saddle-node and Hopf bifurcations, are acquired with 1- and 2-parameter bifurcation analysis. The decrease of variability of ISIs with increasingg h is induced by the fast decrease of the standard deviation of ISIs, which is related to the increase of the capacity of resisting noisy disturbance due to the firing becomes far away from the bifurcation point. The enhancement of the rebound (spike) with increasingg h builds up a relationship to the decrease of the capacity of resisting disturbance like the hyperpolarization stimulus as the resting state approaches the bifurcation point. The "typical"-resonance and non-resonance appear in the parameter region of the stable focus and node far away from the bifurcation points, respectively. The complex or "strange" dynamics, such as the "weak"-resonance for the stable node near the transition point between the stable node and focus and the non-resonance for the stable focus close to the codimension-1 and -2 bifurcation points, are discussed.
Article
Full-text available
Differential vulnerability between Substantia Nigra pars compacta (SNpc) and Ventral Tegmental Area (VTA) dopaminergic (DAergic) neurons is a hallmark of Parkinson's disease (PD). Understanding the molecular bases of this key histopathological aspect would foster the development of much-needed disease-modifying therapies. Non-heterogeneous DAergic degeneration is present in both toxin-based and genetic animal models, suggesting that cellular specificity, rather than causing factors, constitutes the background for differential vulnerability. In this regard, we previously demonstrated that MPP+, a neurotoxin able to cause selective nigrostriatal degeneration in animal rodents and primates, inhibits the Hyperpolarization-activated current (Ih) in SNpc DAergic neurons and that pharmacological Ih antagonism causes potentiation of evoked Excitatory post-synaptic potentials (EPSPs). Of note, the magnitude of such potentiation is greater in the SNpc subfield, consistent with higher Ih density. In the present work, we show that Ih block-induced synaptic potentiation leads to the amplification of somatic calcium responses (SCRs) in vitro. This effect is specific for the SNpc subfield and largely mediated by L-Type calcium channels, as indicated by sensitivity to the CaV 1 blocker isradipine. Furthermore, Ih is downregulated by low intracellular ATP and determines the efficacy of GABAergic inhibition in SNpc DAergic neurons. Finally, we show that stereotaxic administration of Ih blockers causes SNpc-specific neurodegeneration and hemiparkinsonian motor phenotype in rats. During PD progression, Ih downregulation may result from mitochondrial dysfunction and, in concert with PD-related disinhibition of excitatory inputs, determine a SNpc-specific disease pathway.
Article
The presence of α-synuclein (α-syn) in Lewy bodies and Lewy neurites is an important characteristic of the neurodegenerative processes of substantia nigra pars compacta (SNpc) dopaminergic (DAergic) neurons in Parkinson's disease (PD) and other synucleinopathies. Here we report that Berlin-Druckrey rats carrying a spontaneous mutation in the 3' untranslated region of α-syn mRNA (m/m rats) display a marked accumulation of α-syn in the mesencephalic area, striatum and frontal cortex, accompanied to severe dysfunctions in the dorsolateral striatum. Despite a small reduction in the number of SNpc and ventral tegmental area DAergic cells, the surviving dopaminergic neurons of the m/m rats do not show clear-cut alterations of the spontaneous and evoked firing activity, DA responses and somatic amphetamine-induced firing inhibition. Interestingly, mutant DAergic neurons display diminished whole-cell Ih conductance and a reduced frequency of spontaneous excitatory synaptic currents. By contrast, m/m rats show a severe impairment of DA and glutamate release in the dorsolateral striatum, as revealed by amperometric measure of DA currents and by electrophysiological recordings of glutamatergic synaptic events in striatal medium spiny neurons. These functional impairments are paralleled by a decreased expression of the DA transporter and VGluT1 proteins in the same area. Thus, together with α-syn overload in the mesencephalic region, striatum and frontal cortex, the main functional alterations occur in the DAergic and glutamatergic terminals in the dorsal striatum of the m/m rats.
Article
Full-text available
Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are expressed as four different isoforms (HCN1-4) in the heart and in the central and peripheral nervous systems. HCN channels are activated by membrane hyperpolarization at voltages close to resting membrane potentials and carry the hyperpolarization-activated current, dubbed If (funny current) in heart and Ih in neurons. HCN channels contribute in several ways to neuronal activity and are responsible for many important cellular functions, including cellular excitability, generation and modulation of rhythmic activity, dendritic integration, transmission of synaptic potentials and plasticity phenomena. Because of their role, defective HCN channels are natural candidates in the search for potential causes of neurological disorders in humans. Several data, including growing evidence that some forms of epilepsy are associated with HCN mutations, support the notion of an involvement of dysfunctional HCN channels in different experimental models of the disease. Additionally, some anti-epileptic drugs are known to modify the activity of the Ih current. HCN channels are widely expressed in the peripheral nervous system and recent evidence has highlighted the importance of the HCN2 isoform in the transmission of pain. HCN channels are also present in the midbrain system, where they finely regulate the activity of dopaminergic neurons, and a potential role of these channels in the pathogenesis of Parkinson’s disease has recently emerged. The function of HCN channels is regulated by specific accessory proteins, which control the correct expression and modulation of the neuronal Ih current. Alteration of these proteins can severely interfere with the physiological channel function, potentially predisposing to pathological conditions. In this review we address the present knowledge of the association between HCN dysfunctions and neurological diseases, including clinical, genetic and physiopathological aspects.
Article
Full-text available
Neurodegenerative diseases (NDDs) involve years of gradual preclinical progression. It is widely anticipated that in order to be effective, treatments should target early stages of disease, but we lack conceptual frameworks to identify and treat early manifestations relevant to disease progression. Here we discuss evidence that a focus on physiological features of neuronal subpopulations most vulnerable to NDDs, and how those features are affected in disease, points to signaling pathways controlling excitation in selectively vulnerable neurons, and to mechanisms regulating calcium and energy homeostasis. These hypotheses could be tested in neuronal stress tests involving animal models or patient-derived iPS cells. Copyright © 2015 Elsevier Inc. All rights reserved.
Article
Full-text available
Motor cortex is a key brain center involved in motor control in rodents and other mammals, but specific intracortical mechanisms at the microcircuit level are largely unknown. Neuronal expression of hyperpolarization-activated current (I(h)) is cell class specific throughout the nervous system, but in neocortex, where pyramidal neurons are classified in various ways, a systematic pattern of expression has not been identified. We tested whether I(h) is differentially expressed among projection classes of pyramidal neurons in mouse motor cortex. I(h) expression was high in corticospinal neurons and low in corticostriatal and corticocortical neurons, a pattern mirrored by mRNA levels for HCN1 and Trip8b subunits. Optical mapping experiments showed that I(h) attenuated glutamatergic responses evoked across the apical and basal dendritic arbors of corticospinal but not corticostriatal neurons. Due to I(h), corticospinal neurons resonated, with a broad peak at ∼4 Hz, and were selectively modulated by α-adrenergic stimulation. I(h) reduced the summation of short trains of artificial excitatory postsynaptic potentials (EPSPs) injected at the soma, and similar effects were observed for short trains of actual EPSPs evoked from layer 2/3 neurons. I(h) narrowed the coincidence detection window for EPSPs arriving from separate layer 2/3 inputs, indicating that the dampening effect of I(h) extended to spatially disperse inputs. To test the role of corticospinal I(h) in transforming EPSPs into action potentials, we transfected layer 2/3 pyramidal neurons with channelrhodopsin-2 and used rapid photostimulation across multiple sites to synaptically drive spiking activity in postsynaptic neurons. Blocking I(h) increased layer 2/3-driven spiking in corticospinal but not corticostriatal neurons. Our results imply that I(h)-dependent synaptic integration in corticospinal neurons constitutes an intracortical control mechanism, regulating the efficacy with which local activity in motor cortex is transferred to downstream circuits in the spinal cord. We speculate that modulation of I(h) in corticospinal neurons could provide a microcircuit-level mechanism involved in translating action planning into action execution.
Article
Full-text available
This protocol describes our method of producing a reliable mouse model of Parkinson's disease (PD) using the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). We discuss the particulars of the model, provide key references and outline what investigators need to know to develop the MPTP mouse model of PD safely and successfully. Completion of this protocol depends on the regimen of MPTP used and on the actual planned studies, which often range from 7 to 30 d. This protocol calls for implementation of safety measures and for the acquisition of several pieces of equipment, which are a one-time investment worth making if one elects to use this model on a regular basis.
Article
Full-text available
Journal of Cerebral Blood Flow & Metabolism stands at the interface between basic and clinical neurovascular research, and features research on experimental, theoretical, and clinical aspects of brain circulation, metabolism and imaging. The journal is relevant to any physician or scientist with an interest in brain function, cerebrovascular disease, cerebral vascular regulation and brain metabolism, including neurologists, neurochemists, physiologists, pharmacologists, anesthesiologists, neuroradiologists, neurosurgeons, neuropathologists and neuroscientists.
Article
Full-text available
The onset of spontaneous seizures in the pilocarpine model of epilepsy causes a hyperpolarized shift in the voltage-dependent activation of hyperpolarization-activated cyclic nucleotide-gated (HCN) channel-mediated current (Ih) in CA1 hippocampal pyramidal neuron dendrites, contributing to neuronal hyperexcitability and possibly to epileptogenesis. However, the specific mechanisms by which spontaneous seizures cause downregulation of HCN channel gating are yet unknown. We asked whether the seizure-dependent downregulation of HCN channel gating was due to altered phosphorylation signaling mediated by the phosphatase calcineurin (CaN) or the kinase p38 mitogen-activated protein kinase (p38 MAPK). We first found that CaN inhibition upregulated HCN channel gating and reduced neuronal excitability under normal conditions, showing that CaN is a strong modulator of HCN channels. We then found that an in vitro model of seizures (1 h in 0 Mg2+ and 50 microM bicuculline at 35-37 degrees C) reproduced the HCN channel gating change seen in vivo. Pharmacological inhibition of CaN or activation of p38 MAPK partially reversed the in vitro seizure-induced hyperpolarized shift in HCN channel gating, and the shift was fully reversed by the combination of CaN inhibition and p38 MAPK activation. We then demonstrated enhanced CaN activity as well as reduced p38 MAPK activity in vivo in the CA1 hippocampal area of chronically epileptic animals. Pharmacological reversal of these phosphorylation changes restored HCN channel gating downregulation and neuronal hyperexcitability in epileptic tissue to control levels. Together, these results suggest that alteration of two different phosphorylation pathways in epilepsy contributes to the downregulation of HCN channel gating, which consequently produces neuronal hyperexcitability and thus may be a target for novel antiepileptic therapies.
Article
Full-text available
The initial reports regarding a cytotoxic role of calcium ions were published over 30 years ago. In neurons, calcium ions can gain entry into the cell through several mechanisms. These include the over-activation of glutamate receptors (NMDA, AMPA, KA) or of a range of channels and transporters (TRPM2, TRPM7, NCX, ASICs, CaV1.2, and hemichannels). Potentially toxic cytoplasmic calcium concentrations can also occur due to release from internal stores, either through physical damage to mitochondria and the endoplasmic reticulum, or a malfunction of receptors and channels present in their membranes. Such increases of cytoplasmic calcium concentrations can trigger a range of downstream neurotoxic cascades, including the uncoupling mitochondrial electron transfer from ATP synthesis, and the activation and overstimulation of enzymes such as calpains and other proteases, protein kinases, nitric oxide synthase (NOS), calcineurin and endonucleases. Despite the toxic role of calcium, drugs designed to block its entry into neurons have all failed to have any beneficial effects in clinical trials. We suggest that blocking certain receptors and ion channels is unlikely to be a useful therapeutic strategy due to potential deleterious side effects. However, identifying those that are most responsible for cell death and their downstream signalling pathways may lead to improved strategies for treating ischemic and excitotoxic disorders.
Article
Full-text available
Inhibition of mitochondrial complex I is one of the leading hypotheses for dopaminergic neuron death associated with Parkinson's disease (PD). To test this hypothesis genetically, we used a mouse strain lacking functional Ndufs4, a gene encoding a subunit required for complete assembly and function of complex I. Deletion of the Ndufs4 gene abolished complex I activity in midbrain mesencephalic neurons cultured from embryonic day (E) 14 mice, but did not affect the survival of dopaminergic neurons in culture. Although dopaminergic neurons were more sensitive than other neurons in these cultures to cell death induced by rotenone, MPP⁺, or paraquat treatments, the absence of complex I activity did not protect the dopaminergic neurons, as would be expected if these compounds act by inhibiting complex 1. In fact, the dopaminergic neurons were more sensitive to rotenone. These data suggest that dopaminergic neuron death induced by treatment with rotenone, MPP⁺, or paraquat is independent of complex I inhibition. • apoptosis • cell death • dopamine neurons • Ndufs4 • Parkinson's disease
Article
Full-text available
The presence of dopamine receptors in olfactory receptor neurons (ORNs) suggests that odor sensitivity may be modulated by neurotransmitters at the level of primary sensory neurons. Using standard patch-clamp techniques on rat ORNs, we found that 1 microM dopamine, 500 microM SQ 22536 (SQ, an adenylyl cyclase inhibitor), 20 and 50 microM quinpirole (a selective dopamine D2 receptor agonist), and 1 mM adenosine 3', 5'-cyclic monophosphate (cAMP) modulate the hyperpolarization-activated current Ih. On hyperpolarizing from a holding potential of -58 mV, a small Cs+-sensitive inwardly rectifying current (Ih) was observed. Increases in extracellular K+ increased Ih amplitude without shifting its voltage dependence of activation, whereas increases in temperature produced an increase in Ih amplitude and a hyperpolarizing shift in the activation curve. Application of 1 microM dopamine reversibly shifted Ih activation to more negative potentials and decreased Ih current amplitudes. These effects were blocked by concomitant application of dopamine with sulpiride, a selective dopamine D2 receptor antagonist. The effects of dopamine were mimicked by quinpirole. Quinpirole (20 microM) decreased Ih current amplitude, but was without effect on Ih voltage dependence of activation. However, 50 microM quinpirole produced both a reduction of Ih peak currents and a hyperpolarizing shift in the activation curve for Ih. External application of the adenylyl cyclase inhibitor SQ 22536 produced a reversible decrease in peak currents but had no effect on Ih voltage dependence of activation, whereas internal application of cAMP shifted Ih activation to more depolarized potentials. Because Ih modulates cell excitability and spike frequency adaptation, our findings support a role for dopamine in modulating the sensitivity and output of rat ORNs to odorants.
Article
Full-text available
Most mammalian central neurons receive synaptic input over complicated dendritic arbors. Therefore, timing of synaptic information should vary with synapse location. However, I report that temporal summation at CA1 pyramidal somata does not depend on the location of synaptic input. This spatial normalization of temporal integration requires a dendritic hyperpolarization-activated current (Ih). Shaping of synaptic activity by deactivating a nonuniform Ih could counterbalance filtering by dendrites and effectively remove location-dependent variability in temporal integration, thus enhancing synchronization of neuronal populations and functional capabilities of the hippocampal CA1 region.
Article
Full-text available
We describe an electrophysiological preparation of the neuromuscular junction of the nematode C. elegans, which adds to its considerable genetic and genomic resources. Mutant analysis, pharmacology and patch-clamp recording showed that the body wall muscles of wild-type animals expressed a GABA receptor and two acetylcholine receptors. The muscle GABA response was abolished in animals lacking the GABA receptor gene unc-49. One acetylcholine receptor was activated by the nematocide levamisole. This response was eliminated in mutants lacking either the unc-38 or unc-29 genes, which encode alpha and non-alpha acetylcholine receptor subunits, respectively. The second, previously undescribed, acetylcholine receptor was activated by nicotine, desensitized rapidly and was selectively blocked by dihydro-beta-erythroidine, thus explaining the residual motility of unc-38 and unc-29 mutants. By recording spontaneous endogenous currents and selectively eliminating each of these receptors, we demonstrated that all three receptor types function at neuromuscular synapses.
Article
Full-text available
Dopaminergic (DA) midbrain neurons in the substantia nigra (SN) and ventral tegmental area (VTA) are involved in various brain functions such as voluntary movement and reward and are targets in disorders such as Parkinson's disease and schizophrenia. To study the functional properties of identified DA neurons in mouse midbrain slices, we combined patch-clamp recordings with either neurobiotin cell-filling and triple labeling confocal immunohistochemistry, or single-cell RT-PCR. We discriminated four DA subpopulations based on anatomical and neurochemical differences: two calbindin D28-k (CB)-expressing DA populations in the substantia nigra (SN/CB+) or ventral tegmental area (VTA/CB+), and respectively, two calbindin D28-k negative DA populations (SN/CB-, VTA/CB-). VTA/CB+ DA neurons displayed significantly faster pacemaker frequencies with smaller afterhyperpolarizations compared with other DA neurons. In contrast, all four DA populations possessed significant differences in I(h) channel densities and I(h) channel-mediated functional properties like sag amplitudes and rebound delays in the following order: SN/CB- --> VTA/CB- --> SN/CB+ --> VTA/CB+. Single-cell RT-multiplex PCR experiments demonstrated that differential calbindin but not calretinin expression is associated with differential I(h) channel densities. Only in SN/CB- DA neurons, however, I(h) channels were actively involved in pacemaker frequency control. In conclusion, diversity within the DA system is not restricted to distinct axonal projections and differences in synaptic connectivity, but also involves differences in postsynaptic conductances between neurochemically and topographically distinct DA neurons.
Article
Full-text available
The selective degeneration of dopaminergic (DA) midbrain neurons in the substantia nigra (SN) is a hallmark of Parkinson disease. DA neurons in the neighboring ventral tegmental area (VTA) are significantly less affected. The mechanisms for this differential vulnerability of DA neurons are unknown. We identified selective activation of ATP-sensitive potassium (K-ATP) channels as a potential mechanism. We show that in response to parkinsonism-inducing toxins, electrophysiological activity of SN DA neurons, but not VTA DA neurons, is lost owing to activation of K-ATP channels. This selective K-ATP channel activation is controlled by differences in mitochondrial uncoupling between SN and VTA DA neurons. Genetic inactivation of the K-ATP channel pore-forming subunit Kir6.2 resulted in a selective rescue of SN but not VTA DA neurons in two mechanistically distinct mouse models of dopaminergic degeneration, the neurotoxicological 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) model and the mutant weaver mouse. Thus, K-ATP channel activation has an unexpected role in promoting death of DA neurons in chronic disease.
Article
Full-text available
Why dopamine-containing neurons of the brain's substantia nigra pars compacta die in Parkinson's disease has been an enduring mystery. Our studies suggest that the unusual reliance of these neurons on L-type Ca(v)1.3 Ca2+ channels to drive their maintained, rhythmic pacemaking renders them vulnerable to stressors thought to contribute to disease progression. The reliance on these channels increases with age, as juvenile dopamine-containing neurons in the substantia nigra pars compacta use pacemaking mechanisms common to neurons not affected in Parkinson's disease. These mechanisms remain latent in adulthood, and blocking Ca(v)1.3 Ca2+ channels in adult neurons induces a reversion to the juvenile form of pacemaking. Such blocking ('rejuvenation') protects these neurons in both in vitro and in vivo models of Parkinson's disease, pointing to a new strategy that could slow or stop the progression of the disease.
Article
: Previous studies from this laboratory demonstrated that (+)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-imine maleate (MK-801), an N-methyl-D-aspartate (NMDA) receptor antagonist, did not prevent neurotoxicity to dopaminergic neurons in mice produced by systemic treatment with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). However, Turski et al. [Nature349, 414–418 (1991)] reported that extended treatment of rats with NMDA receptor antagonists (six injections at 4-h intervals) did prevent the loss of nigral dopaminergic neurons resulting from an intranigral infusion of 1-methyl-4-phenylpyridinium (MPP+), the neurotoxic metabolite of MPTP. The present studies examined if a similar extended treatment with MK-801 would protect mice from the neurotoxicity of systemically administered MPTP. Six intraperitoneal injections of MK-801 given at 4-h intervals did not protect mice against the MPTP-induced neostriatal dopamine loss measured 1 week after treatment. In other experiments, designed to replicate and expand on the results of Turski et al. (1991), the extended treatment of rats with MK-801 did not prevent MPP+-induced cell loss in the infused substantia nigra pars compacta or the dopamine depletion in the ipsilateral neostriatum at 7-11 days after MPP+ infusion. These results do not support the hypothesis that NMDA receptors are involved with MPTP/MPP+-induced neurodegeneration.
Article
abstract Intracellular electrophysiological recordings in current- and voltage-clamp mode were obtained from dopaminergic neurons of the rat mesencephalon in an in vitro slice preparation. In current-clamp mode, a time-dependent anomalous rectification (TDR) of the membrane was observed in response to hyperpolarizing current pulses. In single-electrode voltage-clamp mode, a slowly developing inward current (lh) underlying the TDR was studied by hyperpolarizing voltage commands from a holding potential of -50 to -60 mV. lh started to be activated at -69 mV, was fully activated at -129 to -141 mV, with half-maximal activation at -87 mV, and showed no inactivation with time. The time course of development of Ih followed a single exponential, and its time constant was voltage-dependent. At -81 mV, lh activated with a time constant of 379 2 47.6 ms, whereas at -129 mV lh activated with a time constant of 65 ? 2.2 ms. Its estimated reversal potential was -35 ± 4 mV. Raising the extracellular concentration of K+ from 2.5 to 6.5 and to 12.5 mM increased the amplitude of lh while reducing the extracellular concentration of Na+ from 153.2 to 27.2 mM caused a reduction in amplitude of lh. Bath application of caesium (1–5 mM) reversibly reduced or blocked the TDR/lh. Perfusion of tetrodotoxin (0.5–1 M), tetraethylammonium (10–20 mM) or barium (0.3–2 mM) did not significantly affect lh. lh was also present in cells impaled with CsCI-filled electrodes. When lh was substantially reduced by extracellular caesium (1 mM) the firing rate of the dopaminergic cells, which consisted of a spontaneous pacemaker discharge of action potentials, was not clearly changed. In addition, the holding current in voltage-clamp experiments at -50 to -60 mV was not affected by 1 mM caesium. We conclude that although the lh current is a typical feature of the dopaminergic neurons, it is neither a significant factor underlying the spontaneous pacemaker activity nor does it contribute substantially to the setting of the normal resting potential level of the membrane. On the other hand, since lh starts at voltages lower than or equal to -69 mV (below firing threshold), it may play a modulatory role in the cell's excitability by limiting the amplitude and duration of any prolonged hyperpolarizing events in the dopaminergic cells.
Article
In this study, the neuroprotective effect of Dimocarpus longan Lour. flower water extract (LFWE) was investigated. First, an in vitro study showed that LFWE concentration-dependently inhibited lipid peroxidation of brain homogenates incubated at 37 °C. The antioxidative activity of LFWE was more potent than that of glutathione or Trolox. Furthermore, an ex vivo study found that the basal lipid peroxidation (0 °C) and lipid peroxidation incubated at 37 °C were lower in the brain homogenates of LFWE-treated (500 mg/day) rats, indicating that the brain of LFWE-treated rats was more resistant to oxidative stress. Moreover, a Parkinsonian animal model was employed to demonstrate that oral administration of LFWE (125-500 mg/kg/day) dose-dependently attenuated 1-methyl-4-phenylpyridinium (MPP(+))-induced neurotoxicity in the nigrostriatal dopaminergic system of rat brain. In conclusion, this study suggests that LFWE is antioxidative, anti-inflammatory, and anti-apoptotic. Furthermore, oral administration of LFWE appears to be useful in preventing and/or treating central nervous system neurodegenerative diseases, including Parkinsonism.
Article
The hippocampus is an integral brain region for affective disorders. TRIP8b knockout mice lacking functional HCN channels as well as both HCN1 and HCN2 knockout mice have been shown to display antidepressant-like behaviors. The mechanisms or brain regions involved in these alterations in behavior, however, are not clear. We developed a lentiviral shRNA system to examine whether knockdown of HCN1 protein in the dorsal hippocampal CA1 region is sufficient to produce antidepressant-like effects. We found that knockdown of HCN1 channels increased cellular excitability and resulted in physiological changes consistent with a reduction of I(h). Rats infused with lentiviral shRNA-HCN1 in the dorsal hippocampal CA1 region displayed antidepressant- and anxiolytic-like behaviors associated with widespread enhancement of hippocampal activity and upregulation of BDNF-mTOR signaling pathways. Our results suggest that HCN1 protein could be a potential target for treatment of anxiety and depression disorders.
Article
The neurotoxin MPTP has been used to create an animal model of Parkinson's disease in the mouse, in part, because it causes a significant loss of dopaminergic neurons in the substantia nigra (nucleus A9). The purpose of the present study was to determine whether MPTP also causes degeneration of midbrain dopaminergic neurons in nuclei A8 and A10 in the mouse, as occurs in humans with Parkinson's disease. Two commonly used strains of mice were used: FVB/N and C57BL/6. MPTP was administered in cumulative doses of 50—300 mg/kg. Seven days later, dopamine concentrations were measured in the striatum using high performance liquid chromatography, and midbrain dopaminergic neurons were identified using an antibody against tyrosine hydroxylase. The cell locations were mapped with a computer imaging system. In the FVB/N strain, there was a dose-dependent decrease in striatal dopamine concentrations. Although the highest dose (300 mg/kg) caused an 86% reduction in striatal dopamine concentrations, there was only a moderate and non-significant loss of midbrain dopaminergic neurons. In the C57BL/6 strain, however, a high dose of MPTP (240 mg/kg) caused a significant reduction in both striatal dopamine concentrations (95%), and midbrain dopaminergic cells; 69% loss of nucleus A8 cells, 75% loss of nucleus A9 cells, and in nucleus A10 subnuclei there was 42% loss of ventral tegmental area cells, 55% loss of interfascicular nucleus cells, and no loss of cells in the central linear nucleus. These data (1) provide further evidence for differential susceptibility to MPTP toxicity among different mouse strains, (2) indicate that a significant depletion of striatal dopamine is not necessarily due to degeneration of midbrain dopaminergic neurons, (3) provide the precise locations of midbrain dopaminergic cells that are vulnerable to MPTP, which will aid future studies that seek to determine the mechanism/s by which MPTP selectively destroys only certain midbrain dopaminergic neurons, and (4) indicate that MPTP produces midbrain dopaminergic neuronal degeneration in the same nuclei in the C57BL16 mouse that degenerate in humans with Parkinson's disease..
Article
Hyperpolarization-activated currents (Ih) are key players in shaping rhythmic neuronal activity. Although candidate genes for Ih channels have been cloned (HCN1–HCN4), the subunit composition of different native Ih channels is unknown. We used a combined patch-clamp and qualitative single-cell reverse transcription multiplex polymerase chain reaction (RT-mPCR) approach to analyse HCN1–4 coexpression profiles in four neuronal populations in mouse CNS. Coexpression of HCN2, HCN3 and HCN4 mRNA was detected in single neurons of all four neuronal cell types analysed. In contrast, HCN1 mRNA was detected in neocortical and hippocampal pyramidal neurons but not in dopaminergic midbrain and thalamocortical neurons. HCN1 expression was correlated with significantly faster activation kinetics on the level of individual neurons. Semiquantitative single-cell RT-mPCR analysis demonstrated that HCN1 mRNA expression is at least eightfold higher in cortical neurons than subcortical neurons. We show that single neurons possess complex coexpression patterns of Ih candidate genes. Alternative expression of HCN1 is likely to be an important molecular determinant to generate the different neuronal Ih channel species adapted to tune either subcortical or cortical network activity.
Article
Animal models of Parkinson's disease (PD) have been widely used in the past four decades to investigate the pathogenesis and pathophysiology of this neurodegenerative disorder. These models have been classically based on the systemic or local (intracerebral) administration of neutoxins that are able to replicate most of the pathological and phenotypic features of PD in mammals (i.e. rodents or primates). In the last decade, the advent of the 'genetic era' of PD has provided a phenomenal enrichment of the research possibilities in this field, with the development of various mammalian (mice and, more recently, rats) and non-mammalian transgenic models that replicate most of the disease-causing mutations identified for monogenic forms of familial PD. Both toxic and transgenic classes of animal PD models have their own specificities and limitations, which must be carefully taken into consideration when choosing the model to be used. If a substantial and reproducible nigrostriatal lesion is required (e.g. for testing therapeutic interventions aimed at counteracting PD-related cell death), a classic toxic model such as one based on the administration of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine or 6-hydroxydopamine will adequately serve the purpose. On the other hand, if selected molecular mechanisms of PD pathogenesis must be investigated, transgenic models will offer invaluable insights. Therefore, until the 'perfect' model is developed, indications to use one model or another will depend on the specific objectives that are being pursued.
Article
Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels have a key role in the control of heart rate and neuronal excitability. Ivabradine is the first compound acting on HCN channels to be clinically approved for the treatment of angina pectoris. HCN channels may offer excellent opportunities for the development of novel anticonvulsant, anaesthetic and analgesic drugs. In support of this idea, some well-established drugs that act on the central nervous system - including lamotrigine, gabapentin and propofol - have been found to modulate HCN channel function. This Review gives an up-to-date summary of compounds acting on HCN channels, and discusses strategies to further explore the potential of these channels for therapeutic intervention.
Article
British Journal of Pharmacology (BJP) is pleased to publish a new set of guidelines for reporting research involving animals, simultaneously with several other journals; the 'ARRIVE' guidelines (Animals in Research: Reporting In Vivo Experiments). This editorial summarizes the background to the guidelines, gives our view of their significance, considers aspects of specific relevance to pharmacology, re-states BJP's guidelines for authors on animal experiments and indicates our commitment to carrying on discussion of this important topic. We also invite feedback via the British Pharmacological Society website.
Article
The ventral tegmental area (VTA) is the source of dopaminergic projections innervating cortical structures and ventral forebrain. Dysfunction of this mesocorticolimbic system is critically involved in psychiatric disorders such as addiction and schizophrenia. Changes in VTA dopamine (DA) neuronal activity can alter neurotransmitter release at target regions which modify information processing in the reward circuit. Here we studied the effect of alpha-2 noradrenergic receptor activation on the hyperpolarization-activated cation current (I(h)) in DA neurons of the rat VTA. Brain slice preparations using whole-cell current and voltage-clamp techniques were employed. Clonidine and UK14304 (alpha-2 receptor selective agonists) were found to decrease I(h) amplitude and to slow its rate of activation indicating a negative shift in the current's voltage dependence. Two non-subtype-selective alpha-2 receptor antagonists, yohimbine and RS79948, prevented the effects of alpha-2 receptor activation. RX821002, a noradrenergic antagonist specific for alpha-2A and alpha-2D did not prevent I(h) inhibition. This result suggests that clonidine might be acting via an alpha-2C subtype since this receptor is the most abundant variant in the VTA. Analysis of a second messenger system associated with the alpha-2 receptor revealed that I(h) inhibition is independent of cyclic AMP (cAMP) and resulted from the activation of protein kinase C. It is suggested that the alpha-2 mediated hyperpolarizing shift in I(h) voltage dependence can facilitate the transition from pacemaker firing to afferent-driven burst activity. This transition may play a key role on the changes in synaptic plasticity that occurs in the mesocorticolimbic system under pathological conditions.
Article
Rotenone is a toxin used to generate animal models of Parkinson's disease; however, the mechanisms of toxicity in substantia nigra pars compacta (SNc) neurons have not been well characterized. We have investigated rotenone (0.05-1 microm) effects on SNc neurons in acute rat midbrain slices, using whole-cell patch-clamp recording combined with microfluorometry. Rotenone evoked a tolbutamide-sensitive outward current (94 +/- 15 pA) associated with increases in intracellular [Ca(2+)] ([Ca(2+)](i)) (73.8 +/- 7.7 nm) and intracellular [Na(+)] (3.1 +/- 0.6 mm) (all with 1 microm). The outward current was not affected by a high ATP level (10 mm) in the patch pipette but was decreased by Trolox. The [Ca(2+)](i) rise was abolished by removing extracellular Ca(2+), and attenuated by Trolox and a transient receptor potential M2 (TRPM2) channel blocker, N-(p-amylcinnamoyl) anthranilic acid. Other effects included mitochondrial depolarization (rhodamine-123) and increased mitochondrial reactive oxygen species (ROS) production (MitoSox), which was also abolished by Trolox. A low concentration of rotenone (5 nm) that, by itself, did not evoke a [Ca(2+)](i) rise resulted in a large (46.6 +/- 25.3 nm) Ca(2+) response when baseline [Ca(2+)](i) was increased by a 'priming' protocol that activated voltage-gated Ca(2+) channels. There was also a positive correlation between 'naturally' occurring variations in baseline [Ca(2+)](i) and the rotenone-induced [Ca(2+)](i) rise. This correlation was not seen in non-dopaminergic neurons of the substantia nigra pars reticulata (SNr). Our results show that mitochondrial ROS production is a key element in the effect of rotenone on ATP-gated K(+) channels and TRPM2-like channels in SNc neurons, and demonstrate, in these neurons (but not in the SNr), a large potentiation of rotenone-induced [Ca(2+)](i) rise by a small increase in baseline [Ca(2+)](i).
Article
Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels comprise a small subfamily of proteins within the superfamily of pore-loop cation channels. In mammals, the HCN channel family comprises four members (HCN1-4) that are expressed in heart and nervous system. The current produced by HCN channels has been known as I(h) (or I(f) or I(q)). I(h) has also been designated as pacemaker current, because it plays a key role in controlling rhythmic activity of cardiac pacemaker cells and spontaneously firing neurons. Extensive studies over the last decade have provided convincing evidence that I(h) is also involved in a number of basic physiological processes that are not directly associated with rhythmicity. Examples for these non-pacemaking functions of I(h) are the determination of the resting membrane potential, dendritic integration, synaptic transmission, and learning. In this review we summarize recent insights into the structure, function, and cellular regulation of HCN channels. We also discuss in detail the different aspects of HCN channel physiology in the heart and nervous system. To this end, evidence on the role of individual HCN channel types arising from the analysis of HCN knockout mouse models is discussed. Finally, we provide an overview of the impact of HCN channels on the pathogenesis of several diseases and discuss recent attempts to establish HCN channels as drug targets.
Article
The pathogenesis of Parkinson's disease is not fully understood, but there is evidence that excitotoxic mechanisms contribute to the pathology. However, data supporting a role for excitotoxicity in the pathophysiology of the disease are controversial and sparse. The goal of this study was to determine whether changes in glutamate signaling and uptake contribute to the demise of dopaminergic neurons in the substantia nigra. Mice were treated chronically with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and probenecid or vehicle (probenecid or saline alone). Extracellular levels of glutamate in the substantia nigra were substantially increased, and there was an increase in the affinity, but no change in the velocity, of glutamate transport after MPTP/probenecid treatment compared to vehicle controls. In addition, the substantia nigra showed two types of programmed death, apoptosis (type I) and autophagic (type II) cell death. These data suggest that increased glutamate signaling could be an important mechanism for the death of dopaminergic neurons and trigger the induction of programmed cell death in the chronic MPTP/probenecid model.
Article
Parkinson's disease (PD) is a neurodegenerative disease that mainly affects dopaminergic (DA-ergic) neurons in the substantia nigra pars compacta (SNc). Glutamate modulates neuronal excitability, and a high concentration of glutamatergic receptors is found on DA-ergic neurons in the SNc. Paraquat (PQ) is a putative causative agent for PD. Its effects on synaptic glutamate transmission in SNc DA-ergic neurons were evaluated using whole-cell voltage-clamp recording in brain slices from 7- to 14-day-old Wistar rats. In the presence of bicuculline (BIC), strychnine, and dl-aminophosphonovaleric acid, PQ reversibly suppressed AMPA receptor-mediated evoked excitatory postsynaptic currents (eEPSCs) in a concentration-dependent manner (P<0.05). In the presence of tetrodotoxin (1 microM), PQ (50 microM) significantly reduced the amplitudes, but not the frequencies, of miniature EPSCs in the SNc, suggesting PQ inhibited eEPSCs through a postsynaptic mechanism. Exogenous application of AMPA to induce AMPA-mediated inward currents excluded involvement of a presynaptic response. The AMPA-induced currents in the SNc were significantly reduced by PQ (50 microM) to 74% of control levels (P<0.05), supporting that PQ acts on postsynaptic AMPA receptors. No effect of PQ on eEPSCs was seen in the LD thalamic nucleus and hippocampus, showing PQ specifically inhibited DA-ergic neurons in the SNc. Our results demonstrate a novel mechanism of action of PQ on glutamate-gated postsynaptic AMPA receptors in SNc DA-ergic neurons. This effect may attenuate the excitability and function of DA-ergic neurons in the SNc, which may contribute to the pathogenesis of PD.
Article
Previous studies from this laboratory demonstrated that (+)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-imine maleate (MK-801), an N-methyl-D-aspartate (NMDA) receptor antagonist, did not prevent neurotoxicity to dopaminergic neurons in mice produced by systemic treatment with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). However, Turski et al. [Nature 349, 414-418 (1991)] reported that extended treatment of rats with NMDA receptor antagonists (six injections at 4-h intervals) did prevent the loss of nigral dopaminergic neurons resulting from an intranigral infusion of 1-methyl-4-phenylpyridinium (MPP+), the neurotoxic metabolite of MPTP. The present studies examined if a similar extended treatment with MK-801 would protect mice from the neurotoxicity of systemically administered MPTP. Six intraperitoneal injections of MK-801 given at 4-h intervals did not protect mice against the MPTP-induced neostriatal dopamine loss measured 1 week after treatment. In other experiments, designed to replicate and expand on the results of Turski et al. (1991), the extended treatment of rats with MK-801 did not prevent MPP(+)-induced cell loss in the infused substantia nigra pars compacta or the dopamine depletion in the ipsilateral neostriatum at 7-11 days after MPP+ infusion. These results do not support the hypothesis that NMDA receptors are involved with MPTP/MPP(+)-induced neurodegeneration.
Article
The administration of the 1-methyl-4-phenylpyridinium ion (MPP+) stereotaxically into the left neostriatum or left median forebrain bundle of female rats resulted in a very large and highly significant loss of dopamine and of its metabolites in the left neostriatum. The effect of MPP+ on neostriatal dopamine content was in general considerably greater than its effect on serotonin or on several amino acids. These results are consistent with the premise that MPP+, formed from 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) by the enzyme monoamine oxidase B, may be responsible for the toxicity observed after MPTP administration.
Article
1. Intracellular recordings were made from neurones in the substantia nigra zona compacta in slices of rat mesencephalon in vitro. The majority of neurones fired action potentials spontaneously at 0.2-5.6 Hz. Dopamine, applied either by superfusion or from the tip of a pressurized pipette, prevented spontaneous action potential firing and hyperpolarized the membrane. 2. When the membrane potential was held negative to the threshold for action potential firing, the hyperpolarization evoked by dopamine was accompanied by a fall in input resistance. Under voltage clamp, dopamine produced an outward membrane current associated with an increase in membrane conductance. The effects of superfused dopamine on firing rate, membrane potential and membrane current were concentration dependent in the range 1-100 microM. 3. The reversal potential for the hyperpolarizations and the outward currents produced by dopamine was -109.7 +/- 1.7 mV (n = 12) when the potassium concentration was 2.5 mM and -74.0 +/- 5.0 mV (n = 4) when the potassium concentration was 10.5 mM. The change in reversal potentials in these and intermediate potassium concentrations was described by the Nernst equation. 4. The outward current induced by dopamine was reversibly reduced by barium (100-300 microM) and by high concentrations of tetraethylammonium (greater than or equal to 10 mM). Calcium-free solutions with cobalt (0.5-2 mM) did not reduce the current in response to dopamine during the first 5 min of their application. Currents and hyperpolarizations caused by dopamine were unaffected by tetrodotoxin (1 microM). 5. The hyperpolarization produced by dopamine was mimicked by the D2 receptor agonist quinpirole (LY 171555, 0.1-3 microM) and was blocked by the D2 receptor agonists domperidone and (-)-sulpiride. Agonists and antagonists at D1 receptors had no effect. 6. (-)-Sulpiride (30 nM-30 microM) produced a progressive shift to the right in the concentration-response curve to either dopamine or quinpirole. Schild analysis of the antagonism between (-)-sulpiride and quinpirole suggested competitive antagonism with a dissociation equilibrium constant for (-)-sulpiride of about 13 nM. 7. It is concluded that dopamine acts on D2 receptors on neurones of the rat substantia nigra pars compacta to increase the membrane potassium conductance.
Article
MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) causes a Parkinson's disease-like syndrome in man, monkeys, and mice. We studied the effects of MPTP and its metabolite, MPP+, on neuronal properties and synaptic transmission in isolated slices of guinea-pig hippocampus using intra- and extracellular recording methods. Addition of MPTP to the superfusate (50 to 100 microM) produced the following effects: Excitatory postsynaptic potentials and extracellularly recorded population spikes, evoked by stimulation of the Schaffer collaterals were increased in amplitude during the application period (30 min). Within 30 min of washing in normal solution, synaptic transmission was blocked, although axonal population action potentials could still be elicited. The block of synaptic transmission was prevented by prior incubation in pargyline, an inhibitor of monoamine oxidase. The membrane potential and resistance of single pyramidal neurons were virtually unaffected; action potentials elicited by depolarizing intracellular current pulses were also unchanged. MPP+ (50 microM) blocked synaptic transmission during the application period by a pargyline-in-sensitive mechanism. These results suggest that MPP+ blocks synaptic transmission in the hippocampus at a presynaptic site. This effect may be relevant for the acute action of MPTP and may provide some insight into its chronic action on nigrostriatal neurons.
Article
The neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) is rapidly metabolized to a 1-methyl-4-phenylpyridinium species (MPP+) in the squirrel monkey. After administration of toxic doses of MPTP, the concentration of MPP+ in the substantia nigra appears to increase during the first 72 hours, reaching the highest concentration of any central nervous system (CNS) tissue studied. In contrast, the concentration of this compound in other brain areas suggested time dependent elimination during the same period. Pretreatment of animals with the monoamine oxidase (MAO) inhibitor pargyline blocks both the neurotoxic action and the biotransformation of MPTP. In animals given pargyline and MPTP, initial MPTP levels are much higher in all brain regions than in those not receiving pargyline, but by 12 hours, MPTP levels had fallen rapidly in all regions except the substantia nigra and the eye. It may be that the selective toxicity of MPTP is related in some way to the accumulation of its oxidized metabolite in the substantia nigra.
Article
MPTP given to mice in 4 daily doses (20 mg/kg s.c.) resulted in 56-70% depletion of striatal dopamine 1 week after the last dose. Pretreatment with deprenyl or MD 240928, selective inhibitors of monoamine oxidase type B, or with amfonelic acid or nomifensine, selective inhibitors of dopamine uptake, prevented the depletion of striatal dopamine. In contrast, pretreatment with alpha-methyl-tyrosine, Ro 4-1284 or haloperidol did not prevent the depletion of striatal dopamine by MPTP. The results are compatible with the view that dopamine itself is not involved in the neurotoxic effect of MPTP but that MPP+, a metabolite of MPTP formed by MAO-B and accumulated by the dopamine uptake carrier, is responsible for the neurotoxicity.
Article
Intracellular electrophysiological recordings in current- and voltage-clamp mode were obtained from dopaminergic neurons of the rat mesencephalon in an in vitro slice preparation. In current-clamp mode, a time-dependent anomalous rectification (TDR) of the membrane was observed in response to hyperpolarizing current pulses. In single-electrode voltage-clamp mode, a slowly developing inward current (Ih) underlying the TDR was studied by hyperpolarizing voltage commands from a holding potential of -50 to -60 mV. Ih started to be activated at approximately -69 mV, was fully activated at -129 to -141 mV, with half-maximal activation at -87 mV, and showed no inactivation with time. The time course of development of Ih followed a single exponential, and its time constant was voltage-dependent. At -81 mV, Ih activated with a time constant of 379 +/- 47.6 ms, whereas at -129 mV Ih activated with a time constant of 65 +/- 2.2 ms. Its estimated reversal potential was -35 +/- 4 mV. Raising the extracellular concentration of K+ from 2.5 to 6.5 and to 12.5 mM increased the amplitude of Ih while reducing the extracellular concentration of Na+ from 153.2 to 27.2 mM caused a reduction in amplitude of Ih. Bath application of caesium (1-5 mM) reversibly reduced or blocked the TDR/Ih. Perfusion of tetrodotoxin (0.5-1 microM), tetraethylammonium (10-20 mM) or barium (0.3-2 mM) did not significantly affect Ih. Ih was also present in cells impaled with CsCl-filled electrodes.(ABSTRACT TRUNCATED AT 250 WORDS)
Article
1. Whole-cell patch-clamp recordings were made from dopamine-containing ventral tegmental area neurones in slices of rat midbrain. An inward current (Ih) was activated by hyperpolarization from -60 mV. 2. Dopamine (30 microM) reduced the amplitude of Ih by 10-30% at potentials from -70 to -120 mV. The effect was concentration dependent, mimicked by the D2 agonist quinpirole, and prevented by the D2 antagonist (-)-sulpiride. Baclofen (0.3-3 microM) also inhibited Ih; this action was antagonized by 2-hydroxysaclofen but not by (-)-sulpiride. The decrease in Ih resulted from a reduction in the maximal current with no change in the voltage dependence. 3. The action of dopamine was unaffected by cadmium (200 microM), forskolin (10 microM), the adenylyl cyclase inhibitor 2',3'-dideoxyadenosine (100 microM), or by intracellular solution containing cyclic AMP (2 mM). 4. Ih was progressively reduced during the first 5-10 min of recording with electrodes containing guanosine 5'-O-(3-thiotriphosphate); after this time, dopamine had no further effect. 5. It is concluded that agonists acting at D2 receptors and GABAB receptors reduce Ih in ventral tegmental neurones.
Article
The effects of ZD 7288, a ”bradycardic” agent, in young rat hippocampal slices in vitro were studied. ZD 7288 (1–1000 μM) reduced the hyperpolarization-activated current (I h) in CA1 pyramidal neurons by a voltage-independent blocking mechanism. Under current-clamp conditions, the bradycardic agent (10 μM) caused membrane hyperpolarization (by 5.9 ± 0.5 mV) and a reduction of membrane conductance (by 17.9 ± 4.1%). These data are consistent with the block of an inward current which is active at rest. The drug-induced hyperpolarization depressed the cell’s excitability by increasing the threshold current necessary to induce firing. When the drug-induced hyperpolarization was compensated for by injection of a tonic depolarizing current, ZD 7288 caused a reduction of the inhibitory post-synaptic potential (IPSP) in EPSP-IPSP sequences. Since Cs+, another known blocker of I h, is able to reverse long-term depression (LTD) of the CA3-CA1 synapse in hippocampal slices, we tested the effect of ZD 7288 on synaptic transmission. We found that ZD 7288 did not significantly modify LTD, suggesting that Cs+-induced inhibition of LTD maintenance is not directly related to block of I h.
Article
Although controversial, studies with methamphetamine and MPTP suggest a link between glutamate-mediated excitotoxicity and degeneration of dopamine cells. Both compounds are thought to create a metabolic stress. To further explore glutamate actions in DA degeneration, we investigated the effects of other metabolic inhibitors. In mesencephalic cultures, DA cell loss produced by 3-NPA or malonate was potentiated by NMDA and prevented by MK-801. In vivo, striatal DA loss produced by intranigral infusions of malonate was also potentiated by intranigral NMDA and prevented by systemic MK-801. In contrast, systemic MK-801 did not prevent DA loss produced by intrastriatal malonate. Intrastriatal MK-801 or CGS 19755 did attenuate DA loss in METH-treated mice, but was confounded by the findings that METH-induced hyperthermia, an important component in toxicity, was also attenuated. Taken together, the data support the hypothesis of NMDA receptor involvement in degeneration of DA neurons. Furthermore, the data also suggest that this interaction is likely to occur in the substantia nigra rather than in the striatum.
Article
Whole-cell ruptured-patch and perforated-patch recordings were used in principal neurons of the rat substantia nigra pars compacta (SNc) to study the effect of catecholamines both on the hyperpolarization-activated cationic (Ih) and the inwardly rectifying potassium (I(Kir)) currents. In internal potassium, a 2 min bath application of noradrenaline (NA; 50 microM) or dopamine (DA; 50 microM) both inhibited Ih and induced an outward current associated with an increase in I(Kir) conductance. These two effects recovered poorly after wash-out. Protein kinase A (PKA), protein kinase C (PKC) and phosphatases 1 and 2A inhibitors did not modify the NA and DA effects on the amplitude of Ih and I(Kir) currents. They also had no effect on the recovery of the catecholamine responses. In perforated-patch experiments, NA and DA also induced an inhibition of Ih and revealed an outward current associated with an increase in conductance. However, both effects recovered in less than 5 min following the wash-out. These results indicate that neither PKA, PKC, nor phosphatases 1 or 2A were required in the NA and DA modulation of these two currents and that an intracellular factor, that could be either washed-out or inversely up-regulated in the ruptured-patch configuration, was implicated in the recovery of both effects. In the presence of external barium (300 microM) or internal caesium which both blocked the outward current and the increase in conductance, neither NA nor DA affected Ih, suggesting that the effect on Ih observed is secondary to the activation of the I(Kir) channels. Increasing chloride conductance of the cell by activation of GABA(A) receptors also induced an inhibition of Ih. All together these results suggest that the NA or DA induced inhibition of Ih could result from an occlusion of Ih by a space-clamp effect.
Article
Most mammalian central neurons receive synaptic input over complicated dendritic arbors. Therefore, timing of synaptic information should vary with synapse location. However, I report that temporal summation at CA1 pyramidal somata does not depend on the location of synaptic input. This spatial normalization of temporal integration requires a dendritic hyperpolarization-activated current (lh). Shaping of synaptic activity by deactivating a nonuniform lh could counterbalance filtering by dendrites and effectively remove location-dependent variability in temporal integration, thus enhancing synchronization of neuronal populations and functional capabilities of the hippocampal CA1 region.
Article
Morphological and electrophysiological characteristics of dopaminergic and non-dopaminergic neurons in the substantia nigra and their postsynaptic responses to stimulation of the tegmental pedunculopontine nucleus were studied in rat organotypic triple cultures. These cultures consisted of the subthalamic nucleus explant, ventral mesencephalic explant, inclusive of the substantia nigra and the mesopontine tegmentum explant, inclusive of the tegmental pedunculopontine nucleus, prepared from one- to two-day-old rats. Intracellular sharp and whole-cell recordings were obtained from three- to eight-week-old organotypic cultures. Recorded neurons were identified as dopaminergic and non-dopaminergic neurons with tyrosine hydroxylase immunohistochemistry. Dopaminergic neurons had long duration action potentials, prominent afterhyperpolarization, time-dependent inward and outward rectification and strong frequency adaptation. Spontaneous firing patterns varied from regular, irregular to burst firing. Non-dopaminergic neurons had short duration action potentials, in general no rectifying currents, and maintained high firing frequencies. Spontaneous firing patterns in these neurons were irregular or burst firing. Morphological analysis of the recorded neurons labeled with neurobiotin revealed that non-dopaminergic neurons had more extensive arborization of higher-order dendrites than dopaminergic neurons. Dopaminergic and non-dopaminergic neurons receive glutamatergic and cholinergic excitatory inputs from the tegmental pedunculopontine nucleus. These results indicate that morphological and electrophysiological characteristics of substantia nigra neurons in the organotypic culture are generally similar to those reported in in vitro slice and in vivo studies. However, spontaneous activities of dopamine neurons observed in the organotypic culture preparation more closely resemble those in in vivo preparation compared to in vitro preparation.
Article
The cause of Parkinson's disease (PD) is unknown, but reduced activity of complex I of the electron-transport chain has been implicated in the pathogenesis of both mitochondrial permeability transition pore-induced Parkinsonism and idiopathic PD. We developed a novel model of PD in which chronic, systemic infusion of rotenone, a complex-I inhibitor, selectively kills dopaminergic nerve terminals and causes retrograde degeneration of substantia nigra neurons over a period of months. The distribution of dopaminergic pathology replicates that seen in PD, and the slow time course of neurodegeneration mimics PD more accurately than current models. Our model should enhance our understanding of neurodegeneration in PD. Metabolic impairment depletes ATP, depresses Na+/K(+)-ATPase activity, and causes graded neuronal depolarization. This relieves the voltage-dependent Mg2+ block of the N-methyl-D-aspartate (NMDA) subtype of the glutamate receptor, which is highly permeable to Ca2+. Consequently, innocuous levels of glutamate become lethal via secondary excitotoxicity. Mitochondrial impairment also disrupts cellular Ca2+ homoeostasis. Moreover, the facilitation of NMDA-receptor function leads to further mitochondrial dysfunction. To a large part, this occurs because Ca2+ entering neurons through NMDA receptors has 'privileged' access to mitochondria, where it causes free-radical production and mitochondrial depolarization. Thus there may be a feed-forward cycle wherein mitochondrial dysfunction causes NMDA-receptor activation, which leads to further mitochondrial impairment. In this scenario, NMDA-receptor antagonists may be neuroprotective.
Article
A previous investigation has suggested that the hyperpolarization-activated cation current Ih does not contribute to the spontaneous firing of midbrain dopaminergic neurons. This conclusion was reached using Cs(-1). We have re-examined this question with extracellular recordings in slices using the more specific blocker ZD7288. In two-thirds of the cells, low concentrations of ZD7288 induced a decrease of the spontaneous firing. The maximal inhibition was about 40% and the mean IC50 was 1.6 microM. This effect was probably direct because it persisted in the presence of antagonists of various receptors. These concentrations of ZD7288 had no effect in the remaining one third of the examined cells. However, the highest concentration of ZD7288 (300 microM) abolished the firing of all dopaminergic neurons, probably by a mechanism unrelated to the blockade of Ih. We conclude that Ih controls to a certain extent the firing of a majority of midbrain dopaminergic neurons.
Article
Hyperpolarization-activated cation currents (I(h)) contribute to various physiological properties and functions in the brain, including neuronal pacemaker activity, setting of resting membrane potential, and dendritic integration of synaptic input. Four subunits of the Hyperpolarization-activated and Cyclic-Nucleotide-gated nonselective cation channels (HCN1-4), which generate I(h), have been cloned recently. To better understand the functional diversity of I(h) in the brain, we examined precise immunohistochemical localization of four HCNs in the rat brain. Immunoreactivity for HCN1 showed predominantly cortical distribution, being intense in the neocortex, hippocampus, superior colliculus, and cerebellum, whereas those for HCN3 and HCN4 exhibited subcortical distribution mainly concentrated in the hypothalamus and thalamus, respectively. Immunoreactivity for HCN2 had a widespread distribution throughout the brain. Double immunofluorescence revealed colocalization of immunoreactivity for HCN1 and HCN2 in distal dendrites of pyramidal cells in the hippocampus and neocortex. At the electron microscopic level, immunogold particles for HCN1 and HCN2 had similar distribution patterns along plasma membrane of dendritic shafts in layer I of the neocortex and stratum lacunosum moleculare of the hippocampal CA1 area, suggesting that these subunits could form heteromeric channels. Our results further indicate that HCNs are localized not only in somato-dendritic compartments but also in axonal compartments of neurons. Immunoreactivity for HCNs often occurred in preterminal rather than terminal portions of axons and in specific populations of myelinated axons. We also found HCN2-immunopositive oligodendrocytes including perineuronal oligodendrocytes throughout the brain. These results support previous electrophysiological findings and further suggest unexpected roles of I(h) channels in the brain.
Article
Neuronal subthreshold excitability and firing behaviour are markedly influenced by the activation and deactivation of the somato-dendritic hyperpolarization-activated cation current (Ih). Here, we evaluated possible contributions of Ih to hyperexcitability in an animal model of absence seizures (WAG/Rij rats). We investigated pyramidal neurons of the somatosensory neocortex, the site of generation of spike-wave discharges. Ih-mediated functions in neurons from WAG/Rij rats, Wistar rats (sharing the same genetic background with WAG/Rij, but less epilepsy-prone) and ACI rats (an inbred strain, virtually free of seizures) were compared. We complemented whole-cell recordings from layer 2-3 pyramidal neurons with immunohistochemistry, Western blot and RT-PCR analysis of the h-channel subunits HCN1-4. The fast component of Ih activation in WAG/Rij neurons was significantly reduced (50% reduction in the h-current density) and four times slower than in neurons from nonepileptic Wistar or ACI rats. The results showing decreases in currents corresponded to a 34% reduction in HCN1 protein in the WAG/Rij compared to the Wistar neocortex, but HCN1 mRNA showed stable expression. The other three Ih subunit mRNAs and proteins (HCN2-4) were not affected. The alterations in Ih magnitude and kinetics of gating in WAG/Rij neurons may contribute to augmented excitatory postsynaptic potentials, the increase in their temporal summation and the facilitation of burst firing of these neurons because each of these effects could be mimicked by the selective Ih antagonist ZD 7288. We suggest that the deficit in Ih-mediated functions may contribute to the development and onset of spontaneously occurring hyperexcitability in a rat model of absence seizures.
Article
The biochemical and cellular changes that occur following administration of 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP) are remarkably similar to that seen in idiopathic Parkinson's disease (PD). In this review, we detail the molecular activities of this compound from peripheral intoxication through its various biotransformations. In addition, we detail the interplay that occurs between the different cellular compartments (neurons and glia) that eventually consort to kill substantia nigra pars compacta (SNpc) neurons.
Article
Hyperpolarization-activated, cyclic-nucleotide-gated (HCN) channels mediate the depolarizing cation current (termed I(h) or I(f)) that initiates spontaneous rhythmic activity in heart and brain. This function critically depends on the reliable opening of HCN channels in the subthreshold voltage-range. Here we show that activation of HCN channels at physiologically relevant voltages requires interaction with phosphoinositides such as phosphatidylinositol-4,5-bisphosphate (PIP(2)). PIP(2) acts as a ligand that allosterically opens HCN channels by shifting voltage-dependent channel activation approximately 20 mV toward depolarized potentials. Allosteric gating by PIP(2) occurs in all HCN subtypes and is independent of the action of cyclic nucleotides. In CNS neurons and cardiomyocytes, enzymatic degradation of phospholipids results in reduced channel activation and slowing of the spontaneous firing rate. These results demonstrate that gating by phospholipids is essential for the pacemaking activity of HCN channels in cardiac and neuronal rhythmogenesis.
Article
Formidable challenges for Parkinson's disease (PD) research are to understand the processes underlying nigrostriatal degeneration and how to protect dopamine neurons. Fundamental research relies on good animal models that demonstrate the pathological hallmarks and motor deficits of PD. Using a chronic regimen of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine and probenecid (MPTP/p) in mice, dopamine cell loss exceeds 60%, extracellular glutamate is elevated, cytoplasmic inclusions are formed and inflammation is chronic. Nevertheless, isradipine, an L-type calcium-channel blocker, attenuates the degeneration. These data support the validity of the MPTP/p model for unravelling the degenerative processes in PD and testing therapies that slow their progress.
NC3Rs Reporting Guidelines Working Group
  • C Kilkenny
  • W Browne
  • Ic Cuthill
  • M Emerson
  • Dg Altman
Kilkenny C, Browne W, Cuthill IC, Emerson M, Altman DG (2010). NC3Rs Reporting Guidelines Working Group. Br J Pharmacol 160: 1577–1579.
NC3Rs Reporting Guidelines Working Group
  • Kilkenny