Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
eNeuro ; 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38997145

ABSTRACT

Chronic neuropathic pain can result from nervous system injury and can persist in the absence of external stimuli. Although ongoing pain characterizes the disorder, in many individuals, the intensity of this ongoing pain fluctuates dramatically. Previously, it was identified that functional magnetic resonance imaging (fMRI) signal covariations between the midbrain periaqueductal gray matter (PAG), rostral ventromedial medulla (RVM), and spinal trigeminal nucleus (SpV) are associated with moment-to-moment fluctuations in pain intensity in individuals with painful trigeminal neuropathy (PTN). Since this brainstem circuit is modulated by higher brain input, we sought to determine which cortical sites might be influencing this brainstem network during spontaneous fluctuations in pain intensity. Over 12 minutes, we recorded ongoing pain intensity in 24 PTN participants, and classified them as fluctuating (n=13) or stable (n=11). Using a PAG seed, we identified connections between the PAG and emotional-affective sites such as the hippocampal and posterior cingulate cortices, the sensory-discriminative posterior insula, and cognitive-affective sites such as the dorsolateral prefrontal (dlPFC) and subgenual anterior cingulate cortices that were altered dependent on spontaneous high and low pain intensity. Additionally, sliding-window functional connectivity analysis revealed that the dlPFC-PAG connection anticorrelated with perceived pain intensity over the entire 12-minute period. These findings reveal cortical systems underlying moment-to-moment changes in perceived pain in PTN, which likely cause dysregulation in the brainstem circuits previously identified, and consequently alter the appraisal of pain across time.Significance statement Whilst the intensity of an individual's chronic pain is often measured at a specific point in time, it is known that in a large proportion of individuals, pain intensity fluctuates dramatically from moment-to-moment. In individuals with chronic neuropathic pain, we found that these spontaneous pain intensity fluctuations are associated with neural function fluctuations, specifically of function reflected as neural connectivity between brainstem pain modulatory circuits and cortical regions, including the dorsolateral prefrontal and cingulate cortices. These findings raise the possibility that modulating brain regions such as the dorsolateral prefrontal cortex in individuals with fluctuating chronic pain may provide an avenue for analgesic treatment.

2.
Cereb Cortex ; 33(7): 4145-4155, 2023 03 21.
Article in English | MEDLINE | ID: mdl-36069972

ABSTRACT

Pain perception can be modulated by several factors. Phenomena like temporal summation leads to increased perceived pain, whereas behavioral conditioning can result in analgesic responses. Furthermore, during repeated, identical noxious stimuli, pain intensity can vary greatly in some individuals. Understanding these variations is important, given the increase in investigations that assume stable baseline pain for accurate response profiles, such as studies of analgesic mechanisms. We utilized functional magnetic resonance imaging to examine the differences in neural circuitry between individuals displaying consistent pain ratings and those who experienced variable pain during a series of identical noxious stimuli. We investigated 63 healthy participants: 31 were assigned to a "consistent" group, and 32 were assigned to a "variable" group dependent on pain rating variability. Variable pain ratings were associated with reduced signal intensity in the dorsolateral prefrontal cortex (dlPFC). Furthermore, the dlPFC connectivity with the primary somatosensory cortex and temperoparietal junction was significantly reduced in variable participants. Our results suggest that investigators should consider variability of baseline pain when investigating pain modulatory paradigms. Additionally, individuals with consistent and variable pain ratings differ in their dlPFC activity and connectivity with pain-sensitive regions during noxious stimulation, possibly reflecting the differences in attentional processing and catastrophizing during pain.


Subject(s)
Pain Perception , Pain , Humans , Pain Perception/physiology , Pain/diagnostic imaging , Pain Measurement , Attention , Brain/diagnostic imaging , Magnetic Resonance Imaging/methods , Prefrontal Cortex/physiology
3.
Redox Biol ; 38: 101798, 2021 01.
Article in English | MEDLINE | ID: mdl-33285412

ABSTRACT

Maintaining high frequency firing of narrow action potentials puts a large metabolic load on fast spiking (FS), perisomatic-inhibitory interneurons compared to their slow-spiking, dendrite targeting counterparts. Although the relationship of action potential (AP) firing and metabolism is firmly established, there is no single method to differentiate interneurons in situ based on their firing properties. In this study, we explore a novel strategy to easily identify the metabolically active FS cells among different classes of interneurons. We found that the oxidation of the fluorescent free radical marker 2,7-dichlorodihydrofluorescein (H2DCF) preferentially occurs in interneurons both in slice cultures and acute brain slices. Despite their morphological heterogeneity, almost all DCF-positive (DCF+) neurons belonged to the cluster of non-accommodating FS interneurons. Furthermore, all FS interneurons expressing parvalbumin (PV) both in slice cultures and in acute slices from tdTomato-PVCre transgenic mice were also DCF+. However, only half of the recorded DCF + cells were also PV+, indicating that H2DCF-oxidation occurs in different interneuron classes characterized by non-accomodating AP-firing. Comprehensively enhancing spontaneous neuronal activity led to mitochondrial oxidation of DCF in pyramidal cells as well as interneurons, suggesting that the apparent selectivity towards interneurons represents differences in the underlying metabolic load. While radical-scavenging, inhibition of APs or NO-synthesis, and iron chelation had no effect on the staining pattern, exposure to the complex-I inhibitor, rotenone, prevented interneuronal DCF accumulation. We conclude that H2DCF oxidation is independent of free radicals but correlates with the intensive oxidative energy metabolism and high mitochondrial mass in interneurons sharing the non-accommodating FS phenotype.


Subject(s)
Interneurons , Parvalbumins , Action Potentials , Animals , Mice , Mice, Transgenic , Pyramidal Cells
4.
Neuroimage Clin ; 19: 167-173, 2018.
Article in English | MEDLINE | ID: mdl-30035014

ABSTRACT

The neural mechanisms underlying the development and maintenance of chronic pain following nerve injury remain unclear. There is growing evidence that chronic neuropathic pain is associated with altered thalamic firing patterns, thalamocortical dysrhythmia and altered infra-slow oscillations in ascending pain pathways. Preclinical and post-mortem human studies have revealed that neuropathic pain is associated with prolonged astrocyte activation in the dorsal horn and we have suggested that this may result in altered gliotransmission, which results in altered resting neural rhythm in the ascending pain pathway. Evidence of astrocyte activation above the level of the dorsal horn in living humans is lacking and direct measurement of astrocyte activation in living humans is not possible, however, there is evidence that regional alterations in T2 relaxation times are indicative of astrogliosis. The aim of this study was to use T2 relaxometry to explore regional brain anatomy of the ascending pain pathway in individuals with chronic orofacial neuropathic pain. We found that in individuals with trigeminal neuropathic pain, decreases in T2 relaxation times occurred in the region of the spinal trigeminal nucleus and primary somatosensory cortex, as well as in higher order processing regions such as the dorsolateral prefrontal, cingulate and hippocampal/parahippocampal cortices. We speculate that these regional changes in T2 relaxation times reflect prolonged astrocyte activation, which results in altered brain rhythm and ultimately the constant perception of pain. Blocking prolonged astrocyte activation may be effective in preventing and even reversing the development of chronic pain following neural injury.


Subject(s)
Brain/physiopathology , Chronic Pain/physiopathology , Neuralgia/physiopathology , Relaxation/physiology , Adult , Brain/metabolism , Chronic Pain/metabolism , Female , Humans , Magnetic Resonance Imaging/methods , Male , Middle Aged , Pain Measurement/methods
5.
Phys Ther Sport ; 33: 12-17, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29935429

ABSTRACT

OBJECTIVES: To establish patterns of subsequent injury in U18 rugby, to quantify the burden of within season injury recurrence. DESIGN: Secondary analysis of prospective data. SETTING: 28 Schools in Ireland. PARTICIPANTS: 825 male rugby players (aged 15-18 years). MAIN OUTCOME MEASURES: Subsequent injuries were classified as: new, local or recurrent (same site and type as index injury). All recurrent injuries were sub-grouped by body part and diagnosis. Burden was based on frequency, days lost and injury proportion ratios. RESULTS: A total of 426 injuries were eligible for analysis, of which, 121 were subsequent injuries. The majority of subsequent injuries involved a different body part than their index injury. There were n = 23 cases of within season recurrence. 78% of recurrences occurred within 2 months of return to play. Recurrent injuries comprised 5% of all injuries and their cumulative time loss was 1073 days. Recurrent injury to the ankle ligaments, lumbar muscles and concussions carried the greatest burden. CONCLUSION: The burden of recurrent injury in U18 rugby is lower than in the professional game. However, this population could benefit from targeted secondary prevention efforts including reconsideration of return-to-play protocols for ankle sprain, lumbar muscles and potentially concussion.


Subject(s)
Athletic Injuries/epidemiology , Football/injuries , Adolescent , Humans , Incidence , Ireland , Male , Recurrence , Return to Sport
6.
Neuroimage Clin ; 17: 222-231, 2018.
Article in English | MEDLINE | ID: mdl-29159039

ABSTRACT

It has been proposed that pain competes with other attention-demanding stimuli for cognitive resources, and many chronic pain patients display significant attention and mental flexibility deficits. These alterations may result from disruptions in the functioning of the default mode network (DMN) which plays a critical role in attention, memory, prospection and self-processing, and recent investigations have found alterations in DMN function in multiple chronic pain conditions. Whilst it has been proposed that these DMN alterations are a characteristic of pain that is chronic in nature, we recently reported altered oscillatory activity in the DMN during an acute, 5  minute noxious stimulus in healthy control subjects. We therefore hypothesize that altered DMN activity patterns will not be restricted to those in chronic pain but instead will also occur in healthy individuals during tonic noxious stimuli. We used functional magnetic resonance imaging to measure resting state infra-slow oscillatory activity and functional connectivity in patients with chronic orofacial pain at rest and in healthy controls during a 20-minute tonic pain stimulus. We found decreases in oscillatory activity in key regions of the DMN in patients with chronic pain, as well as in healthy controls during tonic pain in addition to changes in functional connectivity between the posterior cingulate cortex and areas of the DMN in both groups. The results show that similar alterations in DMN function occur in healthy individuals during acute noxious stimuli as well as in individuals with chronic pain. These DMN changes may reflect the presence of pain per se and may underlie alterations in attentional processes that occur in the presence of pain.


Subject(s)
Acute Pain/physiopathology , Brain/physiopathology , Chronic Pain/physiopathology , Acute Pain/diagnostic imaging , Adult , Attention/physiology , Brain/diagnostic imaging , Brain Mapping , Chronic Pain/diagnostic imaging , Facial Pain/physiopathology , Female , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Neural Pathways/diagnostic imaging , Neural Pathways/physiopathology
7.
Br J Sports Med ; 51(7): 600-606, 2017 Apr.
Article in English | MEDLINE | ID: mdl-26701931

ABSTRACT

OBJECTIVE: To examine injury patterns in adolescent rugby players and determine factors associated with injury risk. DESIGN: Prospective injury surveillance study. SETTING: N=28 Grammar Schools in Ulster, Ireland (2014-2015 playing season). PARTICIPANTS: 825 adolescent rugby players, across in 28 school first XV rugby squads; mean age 16.9 years. MAIN OUTCOME MEASURES: Injuries were classified by body part and diagnosis, and injury incidence using injuries per 1000 match hours of exposure. HRs for injury were calculated through Cox proportional hazard regression after correction for influential covariates. RESULTS: A total of n=426 injuries were reported across the playing season. Over 50% of injuries occurred in the tackle situation or during collisions (270/426), with few reported during set plays. The 3 most common injury sites were head/face (n=102, 23.9%), clavicle/shoulder (n=65, 15.3%) and the knee (n=56, 13.1%). Sprain (n=133, 31.2%), concussion (n=81, 19%) and muscle injury (n=65, 15.3%) were the most common diagnoses. Injury incidence is calculated at 29.06 injuries per 1000 match hours. There were no catastrophic injuries. A large percentage of injuries (208/424) resulted in absence from play for more than 28 days. Concussion carried the most significant time out from play (n=33; 15.9%), followed by dislocations of the shoulder (n=22; 10.6%), knee sprains (n=19, 9.1%), ankle sprains (n=14, 6.7%), hand/finger/thumb (n=11; 5.3%). 36.8% of participants in the study (304/825) suffered at least one injury during the playing season. Multivariate models found higher risk of injury (adjusted HR (AHR); 95% CI) with: higher age (AHR 1.45; 1.14 to 1.83), heavier weight (AHR 1.32; 1.04 to 1.69), playing representative rugby (AHR 1.42; 1.06 to 1.90) and undertaking regular strength training (AHR 1.65; 1.11 to 2.46). Playing for a lower ranked team (AHR 0.67; 0.49 to 0.90) and wearing a mouthguard (AHR 0.70; 0.54 to 0.92) were associated with lower risk of injury. CONCLUSIONS: There was a high incidence of severe injuries, with concussion, ankle and knee ligament injuries and upper limb fractures/dislocations causing greatest time loss. Players were compliant with current graduated return-to-play regulations following concussion. Physical stature and levels of competition were important risk factors and there was limited evidence for protective equipment.


Subject(s)
Athletic Injuries/epidemiology , Football/injuries , Adolescent , Brain Concussion/epidemiology , Fractures, Bone/epidemiology , Humans , Incidence , Ireland , Male , Multivariate Analysis , Muscle, Skeletal/injuries , Proportional Hazards Models , Prospective Studies , Risk Factors , Schools , Sprains and Strains/epidemiology
8.
Cogn Affect Behav Neurosci ; 16(3): 561-9, 2016 06.
Article in English | MEDLINE | ID: mdl-27007610

ABSTRACT

The temperament dimension of harm avoidance defines an individual's biological tendency to exhibit altering levels of anxious, inhibiting, and cautious behavior. High harm avoidance and anxiety are highly comorbid, likely due to activity in similar neural circuitries involving the dorsal raphe nucleus. Despite the many investigations that have explored personality factors and brain function, none have determined the influence of ongoing activity within dorsal raphe networks on harm avoidance. The aim of this study was to explore such a relationship. In 62 healthy subjects, a series of 180 functional magnetic resonance images covering the entire brain were collected, and each subject completed the 240-item TCI-R questionnaire. Independent component analyses were performed to define the dorsal raphe network and then to determine the regions significantly correlated with harm avoidance. The independent component analyses revealed three signal intensity fluctuation maps encompassing the dorsal raphe nucleus, showing interactions with regions of the amygdala, hippocampus, nucleus accumbens, and prefrontal, insular, and cingulate cortices. Within these systems, the resting signal intensity was significantly coupled to harm avoidance in the bilateral basal amygdala, bilateral ventral hippocampus, bilateral insula, bilateral nucleus accumbens, and medial prefrontal cortex. Note that we could not measure serotonergic output, but instead measured signal changes in the dorsal raphe that likely reflect synaptic activity. These data provide evidence that at rest, signal intensity fluctuations within the dorsal raphe networks are related to harm avoidance. Given the strong relationship between harm avoidance and anxiety-like behaviors, it is possible that ongoing activity within this identified neural circuitry can contribute to an individual developing anxiety disorders.


Subject(s)
Avoidance Learning/drug effects , Dorsal Raphe Nucleus/drug effects , Rest/physiology , Serotonin/pharmacology , Temperament/drug effects , Adult , Aged , Amygdala/physiopathology , Anxiety/drug therapy , Anxiety/physiopathology , Anxiety Disorders/drug therapy , Avoidance Learning/physiology , Dorsal Raphe Nucleus/physiopathology , Female , Hippocampus/drug effects , Hippocampus/physiopathology , Humans , Male , Middle Aged , Prefrontal Cortex/drug effects , Prefrontal Cortex/physiopathology , Temperament/physiology , Young Adult
9.
Eur J Pain ; 20(4): 639-51, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26392220

ABSTRACT

BACKGROUND: Pain catastrophizing significantly affects an individual's experience of pain. High pain catastrophizing is associated with increased fear avoidance behaviours, pain intensity and disability. The aim of this investigation was to determine the effect of pain catastrophizing on ongoing brain activity and movement-evoked brain activity during acute orofacial muscle pain. METHODS: Thirty-four healthy, pain-free subjects were recruited. In 17 subjects, the effect of catastrophizing on regional brain activity was determined. In 19 subjects, functional magnetic resonance imaging was used to determine the effects of pain catastrophizing on brain activation patterns during jaw movements in the presence of ongoing pain. RESULTS: We found that in the presence of pain, catastrophizing was significantly correlated with activity in multi-sensory integrative brain regions, including the dorsolateral and medial prefrontal cortices. Importantly, this relationship did not exist when subjects were not experiencing pain. In addition, during repetitive open-close jaw movements in the presence of pain, activity in the primary motor cortex, cerebellar cortex and the trigeminal motor nucleus was positively correlated with pain catastrophizing scores. In contrast, in the dorsolateral prefrontal cortex, as pain catastrophizing scores increased, the magnitude of signal intensity change during jaw movements decreased. Again, no such relationships occurred when the individual was not in pain. CONCLUSIONS: These data show that during pain, catastrophic thinking has a significant impact on activity in motor and sensory integrative regions. Reducing negative coping strategies may be an effective means in reducing fear avoidance behaviours and the intensity of ongoing pain.


Subject(s)
Brain/physiopathology , Catastrophization/physiopathology , Catastrophization/psychology , Facial Pain/psychology , Motor Activity/physiology , Myalgia/psychology , Adult , Case-Control Studies , Facial Pain/physiopathology , Female , Humans , Magnetic Resonance Imaging , Male , Myalgia/physiopathology , Pain Measurement/methods , Young Adult
10.
Neuroimage ; 124(Pt A): 54-62, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26343321

ABSTRACT

Conditioned pain modulation is a powerful analgesic mechanism, occurring when a painful stimulus is inhibited by a second painful stimulus delivered at a different body location. Reduced conditioned pain modulation capacity is associated with the development of some chronic pain conditions and the effectiveness of some analgesic medications. Human lesion studies show that the circuitry responsible for conditioned pain modulation lies within the caudal brainstem, although the precise nuclei in humans remain unknown. We employed brain imaging to determine brainstem sites responsible for conditioned pain modulation in 54 healthy individuals. In all subjects, 8 noxious heat stimuli (test stimuli) were applied to the right side of the mouth and brain activity measured using functional magnetic resonance imaging. This paradigm was then repeated. However, following the fourth noxious stimulus, a separate noxious stimulus, consisting of an intramuscular injection of hypertonic saline into the leg, was delivered (conditioning stimulus). During this test and conditioning stimulus period, 23 subjects displayed conditioned pain modulation analgesia whereas 31 subjects did not. An individual's analgesic ability was not influenced by gender, pain intensity levels of the test or conditioning stimuli or by psychological variables such as pain catastrophizing or fear of pain. Brain images were processed using SPM8 and the brainstem isolated using the SUIT toolbox. Significant increases in signal intensity were determined during each test stimulus and compared between subjects that did and did not display CPM analgesia (p<0.05, small volume correction). The expression of analgesia was associated with reduction in signal intensity increases during each test stimulus in the presence of the conditioning stimulus in three brainstem regions: the caudalis subdivision of the spinal trigeminal nucleus, i.e., the primary synapse, the region of the subnucleus reticularis dorsalis and in the dorsolateral pons in the region of the parabrachial nucleus. Furthermore, the magnitudes of these signal reductions in all three brainstem regions were significantly correlated to analgesia magnitude. Defining conditioned pain modulation circuitry provides a framework for the future investigations into the neural mechanisms responsible for the maintenance of persistent pain conditions thought to involve altered analgesic circuitry.


Subject(s)
Brain Stem/physiopathology , Pain Perception/physiology , Pain/physiopathology , Adult , Brain Mapping , Female , Hot Temperature , Humans , Magnetic Resonance Imaging , Male , Pain/psychology , Pain Measurement , Young Adult
11.
Neuroimage ; 117: 258-66, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-25979666

ABSTRACT

Accumulated evidence from experimental animal models suggests that neuroplastic changes at the dorsal horn are critical for the maintenance of various chronic musculoskeletal pain conditions. However, to date, no study has specifically investigated whether neuroplastic changes also occur at this level in humans. Using brain imaging techniques, we sought to determine whether anatomical changes were present in the medullary dorsal horn (spinal trigeminal nucleus caudalis) in subjects with the chronic musculoskeletal pain. In twenty-two subjects with painful temporomandibular disorders (TMDs) and forty pain-free controls voxel based morphometry of T1-weighted anatomical images and diffusion tensor images were used to assess regional grey matter volume and microstructural changes within the brainstem and, in addition, the integrity of ascending pain pathways. Voxel based morphometry revealed significant regional grey matter volume decreases in the medullary dorsal horn, in conjunction with alterations in diffusivity properties, namely an increase in mean diffusivity, in TMD subjects. Volumetric and mean diffusivity changes also occurred in TMD subjects in regions of the descending pain modulation system, including the midbrain periaqueductal grey matter and nucleus raphe magnus. Finally, tractography revealed altered diffusivity properties, namely decreased fractional anisotropy, in the root entry zone of the trigeminal nerve, the spinal trigeminal tract and the ventral trigeminothalamic tracts of TMD subjects. These data reveal that chronic musculoskeletal pain in humans is associated with discrete alterations in the anatomy of the medullary dorsal horn, as well as its afferent and efferent projections. These neural changes may be critical for the maintenance of pathological pain.


Subject(s)
Brain Stem/pathology , Gray Matter/pathology , Magnetic Resonance Imaging/methods , Temporomandibular Joint Dysfunction Syndrome/pathology , Trigeminal Caudal Nucleus/pathology , Adult , Aged , Chronic Pain/pathology , Diffusion Tensor Imaging , Female , Humans , Male , Middle Aged , Spinal Cord Dorsal Horn/pathology
12.
Pain ; 155(5): 1027-1036, 2014 May.
Article in English | MEDLINE | ID: mdl-24530612

ABSTRACT

There is increasing evidence relating thalamic changes to the generation and/or maintenance of neuropathic pain. We have recently reported that neuropathic orofacial pain is associated with altered thalamic anatomy, biochemistry, and activity, which may result in disturbed thalamocortical oscillatory circuits. Despite this evidence, it is possible that these thalamic changes are not responsible for the presence of pain per se, but result as a consequence of the injury. To clarify this subject, we compared brain activity and biochemistry in 12 people with below-level neuropathic pain after complete thoracic spinal cord injury with 11 people with similar injuries and no neuropathic pain and 21 age- and gender-matched healthy control subjects. Quantitative arterial spinal labelling was used to measure thalamic activity, and magnetic resonance spectroscopy was used to determine changes in neuronal variability quantifying N-acetylaspartate and alterations in inhibitory function quantifying gamma amino butyric acid. This study revealed that the presence of neuropathic pain is associated with significant changes in thalamic biochemistry and neuronal activity. More specifically, the presence of neuropathic pain after spinal cord injury is associated with significant reductions in thalamic N-acetylaspartate, gamma amino butyric acid content, and blood flow in the region of the thalamic reticular nucleus. Spinal cord injury on its own did not account for these changes. These findings support the hypothesis that neuropathic pain is associated with altered thalamic structure and function, which may disturb central processing and play a key role in the experience of neuropathic pain.


Subject(s)
Neuralgia/physiopathology , Spinal Cord Injuries/physiopathology , Thalamus/physiopathology , Adult , Aged , Aspartic Acid/analogs & derivatives , Aspartic Acid/metabolism , Female , Functional Neuroimaging , Humans , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Male , Middle Aged , Neuralgia/etiology , Neuralgia/metabolism , Pain Measurement , Spin Labels , Spinal Cord Injuries/complications , Spinal Cord Injuries/metabolism , Thalamus/metabolism
13.
Cochlear Implants Int ; 14 Suppl 4: S27-31, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24533760

ABSTRACT

The assessment process is critical in deciding whether a profoundly deaf child with cochlear nerve deficiency (CND) will be suitable for a cochlear or auditory brainstem implant (ABI). Magnetic resonance imaging (MRI) using submillimetric T2 weighted gradient echo or turbo spin echo sequences is mandatory for all profoundly deaf children to diagnose CND. Evidence of audition on behavioural or electrophysiological tests following both auditory and electrical stimulation sometimes allows identification of significant auditory tissue not visible on MRI. In particular electric auditory brainstem response (EABR) testing may allow some quantification of auditory tissue and help decide whether a cochlear implant will be beneficial. Age and cognitive development are the most critical factors in determining ABI benefit. Hearing outcomes from both cochlear implants and ABIs are variable and likely to be limited in children with CND. A proportion of children will get no benefit. Usually the implants would be expected to provide recognition of environmental sounds and understanding of simple phonetics. Most children will not develop normal speech and they will often need to learn to communicate with sign language. The ABI involves a major neurosurgical procedure and at present the long term outcomes are unknown. It is therefore essential that parents who are considering this intervention have plenty of time to consider all aspects and the opportunity for in depth discussion.


Subject(s)
Auditory Brain Stem Implantation/methods , Cochlear Implantation/methods , Cochlear Implants , Deafness/surgery , Language Development , Vestibulocochlear Nerve Diseases/surgery , Adolescent , Child , Child Language , Child, Preschool , Cochlear Nucleus/physiology , Deafness/diagnosis , Deafness/physiopathology , Evoked Potentials, Auditory, Brain Stem/physiology , Humans , Infant , Magnetic Resonance Imaging , Neuronal Plasticity , Phonetics , Round Window, Ear/physiology , Speech , Speech Perception , Tomography, X-Ray Computed , Vestibulocochlear Nerve Diseases/diagnosis , Vestibulocochlear Nerve Diseases/physiopathology
14.
Pain ; 151(2): 384-393, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20732744

ABSTRACT

The conscious perception of somatosensory stimuli is thought to be located in the contralateral cerebral cortex. However, recent human brain imaging investigations in the spinal system report bilateral primary somatosensory cortex (SI) activations during unilateral noxious stimuli and that this ipsilateral spinal representation may be independent of transcallosal connections. In the trigeminal system, there is primate evidence for an ipsilateral somatosensory pathway through the thalamus to the face SI. However, the organization of the trigeminal nociceptive pathway in the human is not clear. The aim of this study was to determine whether noxious stimuli applied to the face are transmitted to the cerebral cortex by bilateral pathways. We used functional magnetic resonance imaging (fMRI) to compare ipsilateral and contralateral activation of the thalamus, SI and secondary somatosensory cortex (SII) during muscle and cutaneous orofacial pain and innocuous facial stimulation in healthy human subjects. We found that both muscle and cutaneous noxious stimuli, from injections of hypertonic saline into the right masseter or overlying skin, evoked bilateral increases in signal intensity in the region encompassing the ventral posterior thalamus as well as the face region of SI and SII. In contrast, innocuous unilateral brushing of the lower lip evoked a strict contralateral ventroposterior thalamic activation, but bilateral activation of SI and SII. These data indicate that, in contrast to innocuous inputs from the face, noxious information ascends bilaterally to the face SI through the ventroposterior thalamus in humans.


Subject(s)
Functional Laterality/physiology , Mouth/innervation , Muscle, Skeletal/innervation , Pain/pathology , Thalamus/physiopathology , Trigeminal Nerve/physiopathology , Adult , Brain Mapping , Evoked Potentials, Somatosensory/drug effects , Female , Humans , Image Processing, Computer-Assisted/methods , Magnetic Resonance Imaging , Male , Middle Aged , Oxygen , Pain/chemically induced , Pain Measurement/methods , Pain Threshold/drug effects , Saline Solution, Hypertonic/adverse effects , Somatosensory Cortex/blood supply , Somatosensory Cortex/physiopathology , Thalamus/blood supply , Young Adult
15.
Cereb Cortex ; 20(6): 1409-19, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19815621

ABSTRACT

Persistent neuropathic pain commonly occurs following spinal cord injury (SCI). It remains one of the most challenging management problems in this condition. In order to develop more effective treatments, a better understanding of the neural changes associated with neuropathic SCI pain is required. The aim of this investigation was to use diffusion tensor imaging (DTI) to determine if persistent neuropathic pain following SCI is associated with changes in regional brain anatomy and connectivity. In 23 subjects with complete thoracic SCI, 12 with below-level neuropathic pain and 11 without pain, and 45 healthy control subjects, a series of whole-brain DTI scans were performed. The mean diffusivity (MD) of each voxel was calculated and values compared between groups. This analysis revealed that neuropathic pain following SCI is associated with significant differences in regional brain anatomy. These anatomical changes were located in pain-related regions as well as regions of the classic reward circuitry, that is, the nucleus accumbens and orbitofrontal, dorsolateral prefrontal, and posterior parietal cortices. The right posterior parietal cortex projected to most regions that displayed an anatomical change. Analysis of the fiber tracts connecting areas of MD differences revealed no significance differences in MD values between the SCI pain, SCI no pain, and control groups.


Subject(s)
Brain/anatomy & histology , Brain/physiopathology , Nerve Net/anatomy & histology , Nerve Net/physiopathology , Pain, Intractable/physiopathology , Spinal Cord Injuries/physiopathology , Adult , Aged , Diffusion Tensor Imaging , Female , Functional Laterality/physiology , Humans , Male , Middle Aged , Neural Pathways/anatomy & histology , Neural Pathways/physiopathology , Neuralgia/etiology , Neuralgia/physiopathology , Neuronal Plasticity/physiology , Pain, Intractable/etiology , Spinal Cord Injuries/complications , Young Adult
16.
Exp Brain Res ; 195(2): 317-24, 2009 May.
Article in English | MEDLINE | ID: mdl-19365628

ABSTRACT

There is evidence in experimental animals that deep and superficial pain exert differential effects on cutaneous sympathetic activity. Skin sympathetic nerve activity (SSNA) was recorded from the common peroneal nerve of awake human subjects and injections of 0.5 ml hypertonic saline was made into the tibialis anterior muscle (causing a deep, dull ache) or 0.2 ml into the overlying skin (causing a sharp burning pain) at unexpected times. Both deep and superficial pain caused increases in SSNA immediately on injection and preceding the onset of pain for both muscle and skin pain (10.1 +/- 2.4 vs. 15.3 +/- 5.3 s; muscle versus skin, respectively). SSNA increases were short lasting (104.2 +/- 13.4 vs. 81.8 +/- 11.7 s; muscle versus skin pain) and did not follow muscle and skin pain profiles. Sweat release occurred following both intramuscular and subcutaneous injections of hypertonic saline. While muscle or skin pain invariably caused changes in skin blood flow as well as increases in sweat release, skin blood flow increased in females and decreased in males. We conclude that both acute muscle and skin pain cause an increase in SSNA, sweat release and gender-dependent changes in skin blood flow.


Subject(s)
Pain/physiopathology , Peroneal Nerve/physiology , Skin/innervation , Sympathetic Nervous System/physiopathology , Adolescent , Female , Humans , Male , Muscle, Skeletal/physiopathology , Pain/chemically induced , Pain Measurement , Regional Blood Flow , Saline Solution, Hypertonic , Severity of Illness Index , Sex Characteristics , Skin/blood supply , Sweat/physiology , Young Adult
17.
Pain ; 141(1-2): 52-9, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19027233

ABSTRACT

The most obvious impairments associated with spinal cord injury (SCI) are loss of sensation and motor control. However, many subjects with SCI also develop persistent neuropathic pain below the injury which is often severe, debilitating and refractory to treatment. The underlying mechanisms of persistent neuropathic SCI pain remain poorly understood. Reports in amputees describing phantom limb pain demonstrate a positive correlation between pain intensity and the amount of primary somatosensory cortex (S1) reorganization. Of note, this S1 reorganization has also been shown to reverse with pain reduction. It is unknown whether a similar association between S1 reorganization and pain intensity exists in subjects with SCI. The aim of this investigation was to determine whether the degree of S1 reorganization following SCI correlated with on-going neuropathic pain intensity. In 20 complete SCI subjects (10 with neuropathic pain, 10 without neuropathic pain) and 21 control subjects without SCI, the somatosensory cortex was mapped using functional magnetic resonance imaging during light brushing of the right little finger, thumb and lip. S1 reorganization was demonstrated in SCI subjects with the little finger activation point moving medially towards the S1 region that would normally innervate the legs. The amount of S1 reorganization in subjects with SCI significantly correlated with on-going pain intensity levels. This study provides evidence of a link between the degree of cortical reorganization and the intensity of persistent neuropathic pain following SCI. Strategies aimed at reversing somatosensory cortical reorganization may have therapeutic potential in central neuropathic pain.


Subject(s)
Neuralgia/etiology , Neuronal Plasticity/physiology , Somatosensory Cortex/physiopathology , Spinal Cord Injuries/complications , Spinal Cord Injuries/pathology , Adult , Brain Mapping , Female , Functional Laterality , Humans , Image Processing, Computer-Assisted/methods , Linear Models , Magnetic Resonance Imaging/methods , Male , Middle Aged , Oxygen/blood , Pain Measurement/methods , Severity of Illness Index , Somatosensory Cortex/blood supply , Young Adult
18.
J Physiol ; 587(1): 183-93, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-19015194

ABSTRACT

Human studies conducted more than half a century ago have suggested that superficial pain induces excitatory effects on the sympathetic nervous system, resulting in increases in blood pressure (BP) and heart rate (HR), whereas deep pain is believed to cause vasodepression. To date, no studies have addressed whether deep or superficial pain produces such differential effects on muscle sympathetic nerve activity (MSNA). Using microneurography we recorded spontaneous MSNA from the common peroneal nerve in 13 awake subjects. Continuous blood pressure was recorded by radial arterial tonometry. Deep pain was induced by intramuscular injection of 0.5 ml hypertonic saline (5%) into the tibialis anterior muscle, superficial pain by subcutaneous injection of 0.2 ml hypertonic saline into the overlying skin. Muscle pain, with a mean rating of 4.9 +/- 0.8 (S.E.M.) on a 0-10 visual analog scale (VAS) and lasting on average 358 +/- 32 s, caused significant increases in MSNA (43.9 +/- 10.0%), BP (5.4 +/- 1.1%) and HR (7.0 +/- 2.0%) - not the expected decreases. Skin pain, rated at 4.9 +/- 0.6 and lasting 464 +/- 54 s, also caused significant increases in MSNA (38.2 +/- 12.8%), BP (5.1 +/- 2.1%) and HR (5.6 +/- 2.0%). The high-frequency (HF) to low-frequency (LF) ratio of heart rate variability (HRV) increased from 1.54 +/- 0.25 to 2.90 +/- 0.45 for muscle pain and 2.80 +/- 0.52 for skin pain. Despite the different qualities of deep (dull and diffuse) and superficial (burning and well-localized) pain, we conclude that pain originating in muscle and skin does not exert a differential effect on muscle sympathetic nerve activity, both causing an increase in MSNA and an increase in the LF:HF ratio of HRV. Whether this holds true for longer lasting experimental pain remains to be seen.


Subject(s)
Muscle, Skeletal/physiopathology , Pain/physiopathology , Sympathetic Nervous System/physiopathology , Adult , Blood Pressure , Female , Heart Rate , Humans , Male , Muscle, Skeletal/innervation , Pain/etiology , Pain Measurement , Saline Solution, Hypertonic/administration & dosage , Saline Solution, Hypertonic/toxicity , Skin/innervation , Skin/physiopathology
19.
Cereb Cortex ; 19(1): 224-32, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18483004

ABSTRACT

A debilitating consequence of complete spinal cord injury (SCI) is the loss of motor control. Although the goal of most SCI treatments is to re-establish neural connections, a potential complication in restoring motor function is that SCI may result in anatomical and functional changes in brain areas controlling motor output. Some animal investigations show cell death in the primary motor cortex following SCI, but similar anatomical changes in humans are not yet established. The aim of this investigation was to use voxel-based morphometry (VBM) and diffusion tensor imaging (DTI) to determine if SCI in humans results in anatomical changes within motor cortices and descending motor pathways. Using VBM, we found significantly lower gray matter volume in complete SCI subjects compared with controls in the primary motor cortex, the medial prefrontal, and adjacent anterior cingulate cortices. DTI analysis revealed structural abnormalities in the same areas with reduced gray matter volume and in the superior cerebellar cortex. In addition, tractography revealed structural abnormalities in the corticospinal and corticopontine tracts of the SCI subjects. In conclusion, human subjects with complete SCI show structural changes in cortical motor regions and descending motor tracts, and these brain anatomical changes may limit motor recovery following SCI.


Subject(s)
Diffusion Magnetic Resonance Imaging , Efferent Pathways/pathology , Motor Cortex/pathology , Neuronal Plasticity , Spinal Cord Injuries/pathology , Spinal Cord/pathology , Thoracic Vertebrae/injuries , Adult , Humans , Middle Aged , Thoracic Vertebrae/pathology , Young Adult
20.
Pain ; 128(1-2): 20-30, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17011704

ABSTRACT

The insula is involved in processing noxious information. It is consistently activated by acute noxious stimuli, can elicit pain on stimulation, and lesions encompassing the insula can alter pain perception. Anatomical tracing, electrophysiological and functional brain imaging investigations have suggested that the insula is somatotopically organized with respect to noxious cutaneous inputs. It has also recently been revealed that the anterior insula displays differential activation during cutaneous compared with muscle pain. Given this difference, it is important to determine if an insula somatotopy also exists for muscle pain. Using high-resolution functional magnetic resonance imaging (fMRI) we compared insula activation patterns in 23 subjects during muscle and cutaneous pain induced in the right leg and forearm. Group and frequency analyses revealed somatotopically organized signal increases in the posterior contralateral (left) and ipsilateral (right) anterior insula. Within the posterior contralateral insula, signal increases during both cutaneous and muscle forearm pain were located lateral and anterior to those evoked by leg pain, whereas in the ipsilateral anterior insula the pattern was reversed. Furthermore, within the ipsilateral anterior insula, muscle pain activated a region anterior to that activated by cutaneous pain. This somatotopic organization may be crucial for pain localization or other aspects of the pain experience that differ depending on both stimulation site and type of tissue activated. This study reveals that the insula is organized somatopically with respect to muscle and cutaneous pain and that this organization is further separated according to the tissue in which the pain originates.


Subject(s)
Cerebral Cortex/physiopathology , Evoked Potentials, Motor , Evoked Potentials, Somatosensory , Muscle, Skeletal/physiopathology , Nerve Net/physiopathology , Pain/physiopathology , Skin/physiopathology , Adult , Animals , Brain Mapping/methods , Female , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Neural Pathways/physiopathology , Skin/innervation
SELECTION OF CITATIONS
SEARCH DETAIL
...