Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Ann Neurol ; 82(6): 961-971, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29171917

ABSTRACT

OBJECTIVE: Weakness induced by critical illness (intensive care unit acquired weakness) is a major cause of disability in patients and is currently untreatable. We recently identified a defect in repetitive firing of lower motor neurons as a novel contributor to intensive care unit acquired weakness. To develop therapy for intensive care unit acquired weakness, it was necessary to determine the mechanism underlying the defect in repetitive firing. METHODS: Both computer simulation and in vivo dynamic voltage clamp of spinal motor neurons in septic rats were employed to explore potential mechanisms underlying defective repetitive firing. RESULTS: Our results suggest alteration in subthreshold voltage-activated currents might be the mechanism underlying defective repetitive firing. It has been shown previously that pharmacologic activation of serotonin receptors on motor neurons increases motor neuron excitability, in part by enhancing subthreshold voltage-activated inward currents. Administration of a U.S. Food and Drug Administration-approved serotonin agonist (lorcaserin) to septic rats greatly improved repetitive firing and motor unit force generation. INTERPRETATION: Our findings suggest activation of serotonin receptors with lorcaserin may provide the first ever therapy for intensive care unit acquired weakness in patients. Ann Neurol 2017;82:961-971.


Subject(s)
Action Potentials/physiology , Computer Simulation , Motor Neurons/physiology , Muscle Weakness/physiopathology , Sepsis/physiopathology , Serotonin Receptor Agonists/therapeutic use , Action Potentials/drug effects , Animals , Benzazepines/pharmacology , Motor Neurons/drug effects , Muscle Weakness/drug therapy , Rats , Sepsis/drug therapy , Serotonin Receptor Agonists/pharmacology , Treatment Outcome
2.
J Neurophysiol ; 115(6): 3146-55, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27075540

ABSTRACT

Skeletal muscle force can be transmitted to the skeleton, not only via its tendons of origin and insertion but also through connective tissues linking the muscle belly to surrounding structures. Through such epimuscular myofascial connections, length changes of a muscle may cause length changes within an adjacent muscle and hence, affect muscle spindles. The aim of the present study was to investigate the effects of epimuscular myofascial forces on feedback from muscle spindles in triceps surae muscles of the rat. We hypothesized that within an intact muscle compartment, muscle spindles not only signal length changes of the muscle in which they are located but can also sense length changes that occur as a result of changing the length of synergistic muscles. Action potentials from single afferents were measured intra-axonally in response to ramp-hold release (RHR) stretches of an agonistic muscle at different lengths of its synergist, as well as in response to synergist RHRs. A decrease in force threshold was found for both soleus (SO) and lateral gastrocnemius afferents, along with an increase in length threshold for SO afferents. In addition, muscle spindle firing could be evoked by RHRs of the synergistic muscle. We conclude that muscle spindles not only signal length changes of the muscle in which they are located but also local length changes that occur as a result of changing the length and relative position of synergistic muscles.


Subject(s)
Action Potentials/physiology , Muscle Spindles/physiology , Muscle, Skeletal/cytology , Analysis of Variance , Animals , Biomechanical Phenomena , Female , Isometric Contraction , Models, Biological , Muscle, Skeletal/physiology , Rats , Rats, Wistar , Stress, Mechanical
3.
Exp Neurol ; 282: 1-8, 2016 08.
Article in English | MEDLINE | ID: mdl-27118372

ABSTRACT

The mechanisms by which sepsis triggers intensive care unit acquired weakness (ICUAW) remain unclear. We previously identified difficulty with motor unit recruitment in patients as a novel contributor to ICUAW. To study the mechanism underlying poor recruitment of motor units we used the rat cecal ligation and puncture model of sepsis. We identified striking dysfunction of alpha motor neurons during repetitive firing. Firing was more erratic, and often intermittent. Our data raised the possibility that reduced excitability of motor neurons was a significant contributor to weakness induced by sepsis. In this study we quantified the contribution of reduced motor neuron excitability and compared its magnitude to the contributions of myopathy, neuropathy and failure of neuromuscular transmission. We injected constant depolarizing current pulses (5s) into the soma of alpha motor neurons in the lumbosacral spinal cord of anesthetized rats to trigger repetitive firing. In response to constant depolarization, motor neurons in untreated control rats fired at steady and continuous firing rates and generated smooth and sustained tetanic motor unit force as expected. In contrast, following induction of sepsis, motor neurons were often unable to sustain firing throughout the 5s current injection such that force production was reduced. Even when firing, motor neurons from septic rats fired erratically and discontinuously, leading to irregular production of motor unit force. Both fast and slow type motor neurons had similar disruption of excitability. We followed rats after recovery from sepsis to determine the time course of resolution of the defect in motor neuron excitability. By one week, rats appeared to have recovered from sepsis as they had no piloerection and appeared to be in no distress. The defects in motor neuron repetitive firing were still striking at 2weeks and, although improved, were present at one month. We infer that rats suffered from weakness due to reduced motor neuron excitability for weeks after resolution of sepsis. To assess whether additional contributions from myopathy, neuropathy and defects in neuromuscular transmission contributed to the reduction in force generation, we measured whole-muscle force production in response to electrical stimulation of the muscle nerve. We found no abnormality in force generation that would suggest the presence of myopathy, neuropathy or defective neuromuscular transmission. These data suggest disruption of repetitive firing of motor neurons is an important contributor to weakness induced by sepsis in rats and raise the possibility that reduced motor neuron excitability contributes to disability that persists after resolution of sepsis.


Subject(s)
Action Potentials/physiology , Motor Neurons/physiology , Muscle Weakness/etiology , Muscle Weakness/pathology , Sepsis/complications , Spinal Cord/pathology , Analysis of Variance , Animals , Disease Models, Animal , Electric Stimulation , Electromyography , Patch-Clamp Techniques , Rats , Synaptic Transmission/physiology
4.
J Neurophysiol ; 109(7): 1775-81, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23303860

ABSTRACT

Many critically ill patients in intensive care units suffer from an infection-induced whole body inflammatory state known as sepsis, which causes severe weakness in patients who survive. The mechanisms by which sepsis triggers intensive care unit-acquired weakness (ICUAW) remain unclear. Currently, research into ICUAW is focused on dysfunction of the peripheral nervous system. During electromyographic studies of patients with ICUAW, we noticed that recruitment was limited to few motor units, which fired at low rates. The reduction in motor unit rate modulation suggested that functional impairment within the central nervous system contributes to ICUAW. To understand better the mechanism underlying reduced firing motor unit firing rates, we moved to the rat cecal ligation and puncture model of sepsis. In isoflurane-anesthetized rats, we studied the response of spinal motoneurons to injected current to determine their capacity for initiating and firing action potentials repetitively. Properties of single action potentials and passive membrane properties of motoneurons from septic rats were normal, suggesting excitability was normal. However, motoneurons exhibited striking dysfunction during repetitive firing. The sustained firing that underlies normal motor unit activity and smooth force generation was slower, more erratic, and often intermittent in septic rats. Our data are the first to suggest that reduced excitability of neurons within the central nervous system may contribute to ICUAW.


Subject(s)
Action Potentials , Motor Neurons/physiology , Systemic Inflammatory Response Syndrome/physiopathology , Animals , Critical Illness , Disease Models, Animal , Humans , Membrane Potentials , Muscle Weakness/etiology , Muscle Weakness/physiopathology , Rats , Spinal Cord/physiopathology , Systemic Inflammatory Response Syndrome/complications
5.
J Clin Invest ; 119(5): 1150-8, 2009 May.
Article in English | MEDLINE | ID: mdl-19425168

ABSTRACT

Neuropathy and myopathy can cause weakness during critical illness. To determine whether reduced excitability of peripheral nerves, rather than degeneration, is the mechanism underlying acute neuropathy in critically ill patients, we prospectively followed patients during the acute phase of critical illness and early recovery and assessed nerve conduction. During the period of early recovery from critical illness, patients recovered from neuropathy within days. This rapidly reversible neuropathy has not to our knowledge been previously described in critically ill patients and may be a novel type of neuropathy. In vivo intracellular recordings from dorsal root axons in septic rats revealed reduced action potential amplitude, demonstrating that reduced excitability of nerve was the mechanism underlying neuropathy. When action potentials were triggered by hyperpolarizing pulses, their amplitudes largely recovered, indicating that inactivation of sodium channels was an important contributor to reduced excitability. There was no depolarization of axon resting potential in septic rats, which ruled out a contribution of resting potential to the increased inactivation of sodium channels. Our data suggest that a hyperpolarized shift in the voltage dependence of sodium channel inactivation causes increased sodium inactivation and reduced excitability. Acquired sodium channelopathy may be the mechanism underlying acute neuropathy in critically ill patients.


Subject(s)
Channelopathies/physiopathology , Critical Illness , Peripheral Nervous System Diseases/physiopathology , Polyneuropathies/physiopathology , Sodium Channels/physiology , Action Potentials/physiology , Animals , Cell Membrane/physiology , Electric Impedance , Electrolytes/blood , Electromyography , Female , Humans , Membrane Potentials/physiology , Motor Neurons/pathology , Motor Neurons/physiology , Muscle Weakness/etiology , Muscle Weakness/physiopathology , Muscular Diseases/etiology , Muscular Diseases/physiopathology , Neural Conduction/physiology , Peripheral Nervous System Diseases/etiology , Polyneuropathies/etiology , Rats , Rats, Wistar , Sensory Receptor Cells/pathology , Sensory Receptor Cells/physiology , Sepsis/complications
SELECTION OF CITATIONS
SEARCH DETAIL
...