Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Neurophysiol ; 129(5): 1259-1277, 2023 05 01.
Article in English | MEDLINE | ID: mdl-37073966

ABSTRACT

Lambert-Eaton myasthenic syndrome (LEMS) is an autoimmune-mediated neuromuscular disease thought to be caused by autoantibodies against P/Q-type voltage-gated calcium channels (VGCCs), which attack and reduce the number of VGCCs within transmitter release sites (active zones; AZs) at the neuromuscular junction (NMJ), resulting in neuromuscular weakness. However, patients with LEMS also have antibodies to other neuronal proteins, and about 15% of patients with LEMS are seronegative for antibodies against VGCCs. We hypothesized that a reduction in the number of P/Q-type VGCCs alone is not sufficient to explain LEMS effects on transmitter release. Here, we used a computational model to study a variety of LEMS-mediated effects on AZ organization and transmitter release constrained by electron microscopic, pharmacological, immunohistochemical, voltage imaging, and electrophysiological observations. We show that models of healthy AZs can be modified to predict the transmitter release and short-term facilitation characteristics of LEMS and that in addition to a decrease in the number of AZ VGCCs, disruption in the organization of AZ proteins, a reduction in AZ number, a reduction in the amount of synaptotagmin, and the compensatory expression of L-type channels outside the remaining AZs are important contributors to LEMS-mediated effects on transmitter release. Furthermore, our models predict that antibody-mediated removal of synaptotagmin in combination with disruption in AZ organization alone could mimic LEMS effects without the removal of VGCCs (a seronegative model). Overall, our results suggest that LEMS pathophysiology may be caused by a collection of pathological alterations to AZs at the NMJ, rather than by a simple loss of VGCCs.NEW & NOTEWORTHY We used a computational model of the active zone (AZ) in the mammalian neuromuscular junction to investigate Lambert-Eaton myasthenic syndrome (LEMS) pathophysiology. This model suggests that disruptions in presynaptic active zone organization and protein content (particularly synaptotagmin), beyond the simple removal of presynaptic calcium channels, play an important role in LEMS pathophysiology.


Subject(s)
Lambert-Eaton Myasthenic Syndrome , Animals , Humans , Lambert-Eaton Myasthenic Syndrome/pathology , Calcium Channels/metabolism , Neuromuscular Junction/metabolism , Neurons/metabolism , Calcium Channels, Q-Type , Synaptotagmins , Mammals/metabolism
2.
Nat Biomed Eng ; 6(9): 1057-1073, 2022 09.
Article in English | MEDLINE | ID: mdl-36038771

ABSTRACT

Many crowded biomolecular structures in cells and tissues are inaccessible to labelling antibodies. To understand how proteins within these structures are arranged with nanoscale precision therefore requires that these structures be decrowded before labelling. Here we show that an iterative variant of expansion microscopy (the permeation of cells and tissues by a swellable hydrogel followed by isotropic hydrogel expansion, to allow for enhanced imaging resolution with ordinary microscopes) enables the imaging of nanostructures in expanded yet otherwise intact tissues at a resolution of about 20 nm. The method, which we named 'expansion revealing' and validated with DNA-probe-based super-resolution microscopy, involves gel-anchoring reagents and the embedding, expansion and re-embedding of the sample in homogeneous swellable hydrogels. Expansion revealing enabled us to use confocal microscopy to image the alignment of pre-synaptic calcium channels with post-synaptic scaffolding proteins in intact brain circuits, and to uncover periodic amyloid nanoclusters containing ion-channel proteins in brain tissue from a mouse model of Alzheimer's disease. Expansion revealing will enable the further discovery of previously unseen nanostructures within cells and tissues.


Subject(s)
Microscopy , Nanostructures , Animals , Brain/metabolism , Calcium Channels/metabolism , DNA/metabolism , Hydrogels , Mice , Microscopy/methods , Proteins/metabolism
3.
Biomolecules ; 12(6)2022 05 24.
Article in English | MEDLINE | ID: mdl-35740866

ABSTRACT

The mouse neuromuscular junction (NMJ) has long been used as a model synapse for the study of neurotransmission in both healthy and disease states of the NMJ. Neurotransmission from these neuromuscular nerve terminals occurs at highly organized structures called active zones (AZs). Within AZs, the relationships between the voltage-gated calcium channels and docked synaptic vesicles govern the probability of acetylcholine release during single action potentials, and the short-term plasticity characteristics during short, high frequency trains of action potentials. Understanding these relationships is important not only for healthy synapses, but also to better understand the pathophysiology of neuromuscular diseases. In particular, we are interested in Lambert-Eaton myasthenic syndrome (LEMS), an autoimmune disorder in which neurotransmitter release from the NMJ decreases, leading to severe muscle weakness. In LEMS, the reduced neurotransmission is traditionally thought to be caused by the antibody-mediated removal of presynaptic voltage-gated calcium channels. However, recent experimental data and AZ computer simulations have predicted that a disruption in the normally highly organized active zone structure, and perhaps autoantibodies to other presynaptic proteins, contribute significantly to pathological effects in the active zone and the characteristics of chemical transmitters.


Subject(s)
Autoimmune Diseases , Lambert-Eaton Myasthenic Syndrome , Animals , Autoantibodies , Autoimmune Diseases/pathology , Calcium Channels , Lambert-Eaton Myasthenic Syndrome/pathology , Mice , Neuromuscular Junction/pathology
4.
Neuron ; 107(4): 667-683.e9, 2020 08 19.
Article in English | MEDLINE | ID: mdl-32616470

ABSTRACT

Presynaptic CaV2 channels are essential for Ca2+-triggered exocytosis. In addition, there are two competing models for their roles in synapse structure. First, Ca2+ channels or Ca2+ entry may control synapse assembly. Second, active zone proteins may scaffold CaV2s to presynaptic release sites, and synapse structure is CaV2 independent. Here, we ablated all three CaV2s using conditional knockout in cultured hippocampal neurons or at the calyx of Held, which abolished evoked exocytosis. Compellingly, synapse and active zone structure, vesicle docking, and transsynaptic nano-organization were unimpaired. Similarly, long-term blockade of action potentials and Ca2+ entry did not disrupt active zone assembly. Although CaV2 knockout impaired the localization of ß subunits, α2δ-1 localized normally. Rescue with CaV2 restored exocytosis, and CaV2 active zone targeting depended on the intracellular C-terminus. We conclude that synapse assembly is independent of CaV2s or Ca2+ entry through them. Instead, active zone proteins recruit and anchor CaV2s via CaV2 C-termini.


Subject(s)
Calcium Channels/metabolism , Presynaptic Terminals/metabolism , Synapses/metabolism , Synaptic Transmission/physiology , Animals , Calcium/metabolism , Calcium Channels/genetics , Exocytosis/physiology , Mice, Knockout , Neurons/metabolism , Synaptic Vesicles/metabolism
5.
J Neurophysiol ; 119(4): 1340-1355, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29357458

ABSTRACT

We have investigated the impact of transmitter release site (active zone; AZ) structure on synaptic function by physically rearranging the individual AZ elements in a previously published frog neuromuscular junction (NMJ) AZ model into the organization observed in a mouse NMJ AZ. We have used this strategy, purposefully without changing the properties of AZ elements between frog and mouse models (even though there are undoubtedly differences between frog and mouse AZ elements in vivo), to directly test how structure influences function at the level of an AZ. Despite a similarly ordered ion channel array substructure within both frog and mouse AZs, frog AZs are much longer and position docked vesicles in a different location relative to AZ ion channels. Physiologically, frog AZs have a lower probability of transmitter release compared with mouse AZs, and frog NMJs facilitate strongly during short stimulus trains in contrast with mouse NMJs that depress slightly. Using our computer modeling approach, we found that a simple rearrangement of the AZ building blocks of the frog model into a mouse AZ organization could recapitulate the physiological differences between these two synapses. These results highlight the importance of simple AZ protein organization to synaptic function. NEW & NOTEWORTHY A simple rearrangement of the basic building blocks in the frog neuromuscular junction model into a mouse transmitter release site configuration predicted the major physiological differences between these two synapses, suggesting that transmitter release site structure and organization is a strong predictor of function.


Subject(s)
Ion Channels , Models, Neurological , Neuromuscular Junction/physiology , Synaptic Transmission/physiology , Synaptic Vesicles , Animals , Female , Male , Mice , Rana pipiens
6.
Ann N Y Acad Sci ; 1412(1): 73-81, 2018 01.
Article in English | MEDLINE | ID: mdl-29125190

ABSTRACT

Lambert-Eaton myasthenic syndrome (LEMS) is an autoimmune disorder caused by antibodies directed against the voltage-gated calcium channels that provide the calcium ion flux that triggers acetylcholine release at the neuromuscular junction. To study the pathophysiology of LEMS and test candidate therapeutic strategies, a passive-transfer animal model has been developed in mice, which can be created by daily intraperitoneal injections of LEMS patient serum or IgG into mice for 2-4 weeks. Results from studies of the mouse neuromuscular junction have revealed that each synapse has hundreds of transmitter release sites but that the probability for release at each one is likely to be low. LEMS further reduces this low probability such that transmission is no longer effective at triggering a muscle contraction. The LEMS-mediated attack reduces the number of presynaptic calcium channels, disorganizes transmitter release sites, and results in the homeostatic upregulation of other calcium channel types. Symptomatic treatment is focused on increasing the probability of release from dysfunctional release sites. Current treatment uses the potassium channel blocker 3,4-diaminopyridine (DAP) to broaden the presynaptic action potential, providing more time for calcium channels to open. Current research is focused on testing new calcium channel gating modifiers that work synergistically with DAP.


Subject(s)
Lambert-Eaton Myasthenic Syndrome/etiology , Animals , Autoantigens , Carcinoma, Small Cell/etiology , Disease Models, Animal , Humans , Immunization, Passive , Lambert-Eaton Myasthenic Syndrome/pathology , Lambert-Eaton Myasthenic Syndrome/therapy , Lung Neoplasms/etiology , Mice , Neuromuscular Junction/pathology , Neuromuscular Junction/physiopathology , Neurotransmitter Agents/physiology
7.
Mol Neurobiol ; 52(1): 456-63, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25195700

ABSTRACT

Lambert-Eaton myasthenic syndrome (LEMS) is an autoimmune disease that disrupts the normally reliable neurotransmission at the neuromuscular junction (NMJ). This disruption is thought to result from an autoantibody-mediated removal of a subset of the P/Q-type Ca(2+) channels involved with neurotransmitter release. With less neurotransmitter release at the NMJ, LEMS patients experience debilitating muscle weakness. The underlying cause of LEMS in slightly more than half of all patients is small cell lung cancer, and cancer therapy is the priority for these patients. In the remaining cases, the cause of LEMS is unknown, and these patients often rely on symptomatic treatment options, as there is no cure. However, current symptomatic treatment options, such as 3,4-diaminopyridine (3,4-DAP), can have significant dose-limiting side effects; thus, additional treatment approaches would benefit LEMS patients. Recent studies introduced a novel Ca(2+) channel agonist (GV-58) as a potential therapeutic alternative for LEMS. Additionally, this work has shown that GV-58 and 3,4-DAP interact in a supra-additive manner to completely restore the magnitude of neurotransmitter release at the NMJs of a LEMS mouse model. In this review, we discuss synaptic mechanisms for reliability at the NMJ and how these mechanisms are disrupted in LEMS. We then discuss the current treatment options for LEMS patients, while also considering recent work demonstrating the therapeutic potential of GV-58 alone and in combination with 3,4-DAP.


Subject(s)
Lambert-Eaton Myasthenic Syndrome/physiopathology , Lambert-Eaton Myasthenic Syndrome/therapy , Synapses/pathology , Animals , Humans , Lambert-Eaton Myasthenic Syndrome/drug therapy , Neuromuscular Junction/drug effects , Neuromuscular Junction/physiopathology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Synapses/drug effects
8.
J Physiol ; 592(16): 3687-96, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25015919

ABSTRACT

Lambert-Eaton myasthenic syndrome (LEMS) is an autoimmune disorder in which a significant fraction of the presynaptic P/Q-type Ca(2+) channels critical to the triggering of neurotransmitter release at the neuromuscular junction (NMJ) are thought to be removed. There is no cure for LEMS, and the current most commonly used symptomatic treatment option is a potassium channel blocker [3,4-diaminopyridine (3,4-DAP)] that does not completely reverse symptoms and can have dose-limiting side-effects. We previously reported the development of a novel Ca(2+) channel agonist, GV-58, as a possible alternative treatment strategy for LEMS. In this study, we tested the hypothesis that the combination of GV-58 and 3,4-DAP will elicit a supra-additive increase in neurotransmitter release at LEMS model NMJs. First, we tested GV-58 in a cell survival assay to assess potential effects on cyclin-dependent kinases (Cdks) and showed that GV-58 did not affect cell survival at the relevant concentrations for Ca(2+) channel effects. Then, we examined the voltage dependence of GV-58 effects on Ca(2+) channels using patch clamp techniques; this showed the effects of GV-58 to be dependent upon Ca(2+) channel opening. Based on this mechanism, we predicted an interaction between 3,4-DAP and GV-58. We tested this hypothesis using a mouse passive transfer model of LEMS. Using intracellular electrophysiological ex vivo recordings, we demonstrated that a combined application of 3,4-DAP plus GV-58 had a supra-additive effect that completely reversed the deficit in neurotransmitter release magnitude at LEMS model NMJs. This reversal contrasts with the less significant improvement observed with either compound alone. Our data indicate that a combination of 3,4-DAP and GV-58 represents a promising treatment option for LEMS and potentially for other disorders of the NMJ.


Subject(s)
4-Aminopyridine/analogs & derivatives , Calcium Channel Agonists/pharmacology , Lambert-Eaton Myasthenic Syndrome/metabolism , Neuromuscular Junction/drug effects , Potassium Channel Blockers/pharmacology , Purines/pharmacology , Synaptic Potentials , Thiophenes/pharmacology , 4-Aminopyridine/pharmacology , Amifampridine , Animals , Cell Line, Tumor , Drug Synergism , Female , Humans , Lambert-Eaton Myasthenic Syndrome/physiopathology , Mice , Neuromuscular Junction/physiopathology , Purines/therapeutic use , Thiophenes/therapeutic use
9.
J Neurosci ; 33(25): 10559-67, 2013 Jun 19.
Article in English | MEDLINE | ID: mdl-23785168

ABSTRACT

We developed a novel calcium (Ca(2+)) channel agonist that is selective for N- and P/Q-type Ca(2+) channels, which are the Ca(2+) channels that regulate transmitter release at most synapses. We have shown that this new molecule (GV-58) slows the deactivation of channels, resulting in a large increase in presynaptic Ca(2+) entry during activity. GV-58 was developed as a modification of (R)-roscovitine, which was previously shown to be a Ca(2+) channel agonist, in addition to its known cyclin-dependent kinase activity. In comparison with the parent molecule, (R)-roscovitine, GV-58 has a ∼20-fold less potent cyclin-dependent kinase antagonist effect, a ∼3- to 4-fold more potent Ca(2+) channel agonist effect, and ∼4-fold higher efficacy as a Ca(2+) channel agonist. We have further evaluated GV-58 in a passive transfer mouse model of Lambert-Eaton myasthenic syndrome and have shown that weakened Lambert-Eaton myasthenic syndrome-model neuromuscular synapses are significantly strengthened following exposure to GV-58. This new Ca(2+) channel agonist has potential as a lead compound in the development of new therapeutic approaches to a variety of disorders that result in neuromuscular weakness.


Subject(s)
Calcium Channel Agonists/therapeutic use , Lambert-Eaton Myasthenic Syndrome/drug therapy , Purines/therapeutic use , Thiophenes/therapeutic use , Action Potentials/drug effects , Adult , Aged , Animals , Cell Line , Cyclin-Dependent Kinases/antagonists & inhibitors , Data Interpretation, Statistical , Electrophysiological Phenomena/drug effects , Humans , Mice , Mice, Transgenic , Middle Aged , Neuromuscular Junction/drug effects , Neurotransmitter Agents/metabolism , Patch-Clamp Techniques , Phosphotransferases/metabolism , Roscovitine
10.
Trends Neurosci ; 36(1): 14-22, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23102681

ABSTRACT

The frog neuromuscular junction (NMJ) is a strong and reliable synapse because, during activation, sufficient neurotransmitter is released to trigger a postsynaptic action potential (AP). Recent evidence supports the hypothesis that this reliability emerges from the assembly of thousands of unreliable single vesicle release sites. The mechanisms that govern this unreliability include a paucity of voltage-gated calcium channels, a low probability of calcium channel opening during an AP, and the rare triggering of synaptic vesicle fusion even when a calcium channel does open and allows calcium flux. Here, we discuss the evidence that these unreliable single vesicle release sites may be the fundamental building blocks of many types of synapses in both the peripheral and central nervous system (PNS and CNS, respectively).


Subject(s)
Central Nervous System/physiology , Neuromuscular Junction/physiology , Synapses/physiology , Synaptic Transmission/physiology , Animals , Humans
11.
Ann N Y Acad Sci ; 1275: 85-91, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23278582

ABSTRACT

Lambert-Eaton myasthenic syndrome (LEMS) causes neuromuscular weakness as a result of an autoimmune attack on the calcium channels that normally regulate chemical transmitter release at the neuromuscular junction. Currently there are limited treatment options for patients with this and other forms of neuromuscular weakness. A novel, first-in-class calcium channel agonist that is selective for the types of voltage-gated calcium channels that regulate transmitter release at neuromuscular synapses has recently been developed. This compound (GV-58) slows deactivation (closing) of the channel, resulting in a large increase in total calcium entry during motor nerve action potential activity. This new calcium channel agonist is currently being evaluated for the treatment of neuromuscular weakness. Potential applications include development as single therapeutics, or for combination treatments.


Subject(s)
Calcium Channel Agonists/therapeutic use , Lambert-Eaton Myasthenic Syndrome/drug therapy , Neuromuscular Diseases/drug therapy , Purines/therapeutic use , Thiophenes/therapeutic use , Humans , Roscovitine
12.
ACS Med Chem Lett ; 3(12): 985-90, 2012 Dec 13.
Article in English | MEDLINE | ID: mdl-24936234

ABSTRACT

The acute effect of the potent cyclin-dependent kinase (cdk) inhibitor (R)-roscovitine on Ca(2+) channels inspired the development of structural analogues as a potential treatment for motor nerve terminal dysfunction. On the basis of a versatile chlorinated purine scaffold, we have synthesized ca. 20 derivatives and characterized their N-type Ca(2+) channel agonist action. Agents that showed strong agonist effects were also characterized in a kinase panel for their off-target effects. Among several novel compounds with diminished cdk activity, we identified a new lead structure with a 4-fold improved N-type Ca(2+) channel agonist effect and a 22-fold decreased cdk2 activity as compared to (R)-roscovitine. This compound was selective for agonist activity on N- and P/Q-type over L-type calcium channels.

SELECTION OF CITATIONS
SEARCH DETAIL