Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
NPJ Parkinsons Dis ; 8(1): 166, 2022 Nov 30.
Article in English | MEDLINE | ID: mdl-36450732

ABSTRACT

The dual-hit hypothesis of Parkinson's disease (PD) originally postulated that a neurotropic pathogen leads to formation of α-synuclein pathology in the olfactory bulb (OB) and dorsal motor nucleus of the vagus (DMV) and then invades the brain from these two entry points. Little work has been conducted to validate an important underlying premise for the dual-hit hypothesis, namely that the initial Lewy pathology does arise simultaneously in the OB and the enteric nervous system (ENS) plexuses and DMV at the earliest disease stage. We conducted a focused re-analysis of two postmortem datasets, which included large numbers of mild Lewy body disease (LBD) cases. We found that cases with α-synuclein pathology restricted to the peripheral autonomic nervous system and/or lower brainstem (early body-first LBD cases) very rarely had any OB pathology, suggesting that Lewy pathology commonly arises in the ENS without concomitant involvement of the OB. In contrast, cases with mild amygdala-predominant Lewy pathology (early brain-first LBD cases) nearly always showed OB pathology. This is compatible with the first pathology being triggered in the OB or amygdala followed by secondary spreading to connected structures, but without early involvement of the ENS or lower brainstem. These observations support that the pathologic process starts in either the olfactory bulb or the ENS, but rarely in the olfactory bulb and gut simultaneously. More studies on neuropathological datasets are warranted to reproduce these findings. The agreement between the revised single-hit hypothesis and the recently proposed brain-first vs. body-first model of LBD is discussed.

2.
Parkinsonism Relat Disord ; 104: 21-25, 2022 11.
Article in English | MEDLINE | ID: mdl-36198248

ABSTRACT

INTRODUCTION: The peripheral autonomic nervous system may be involved years before onset of motor symptoms in some patients with Parkinson's disease (PD). Specific imaging techniques to quantify the cholinergic nervous system in peripheral organs are an unmet need. We tested the hypothesis that patients with PD display decreased [18F]FEOBV uptake in peripheral organs - a sign of parasympathetic denervation. METHODS: We included 15 PD patients and 15 age- and sex matched healthy controls for a 70 min whole-body dynamic positron emission tomography (PET) acquisition. Compartmental modelling was used for tracer kinetic analyses of adrenal gland, pancreas, myocardium, spleen, renal cortex, muscle and colon. Standard uptake values (SUV) at 60-70 min post injection were also extracted for these organs. Additionally, SUVs were also determined in the total colon, prostate, parotid and submandibular glands. RESULTS: We found no statistically significant difference of [18F]FEOBV binding parameters in any organs between patients with PD and healthy controls, although trends were observed. The pancreas SUV showed a 14% reduction in patients (P = 0.021, not statistically significant after multiple comparison correction). We observed a trend towards lower SUVs in the pancreas, colon, adrenal gland, and myocardium of PD patients with versus without probable REM sleep behavior disorder. CONCLUSION: [18F]FEOBV PET may not be a sensitive marker for parasympathetic degeneration in patients with PD.


Subject(s)
Parkinson Disease , REM Sleep Behavior Disorder , Male , Humans , Parkinson Disease/diagnostic imaging , Piperidines , Positron-Emission Tomography/methods , Parasympathectomy
3.
Front Aging Neurosci ; 14: 909273, 2022.
Article in English | MEDLINE | ID: mdl-35966779

ABSTRACT

Aging is the biggest risk factor for developing Parkinson's disease (PD), the second most common neurodegenerative disorder. Several animal models have been developed to explore the pathophysiology underlying neurodegeneration and the initiation and spread of alpha-synuclein-related PD pathology, and to investigate biomarkers and therapeutic strategies. However, bench-to-bedside translation of preclinical findings remains suboptimal and successful disease-modifying treatments remain to be discovered. Despite aging being the main risk factor for developing idiopathic PD, most studies employ young animals in their experimental set-up, hereby ignoring age-related cellular and molecular mechanisms at play. Consequently, studies in young animals may not be an accurate reflection of human PD, limiting translational outcomes. Recently, it has been shown that aged animals in PD research demonstrate a higher susceptibility to developing pathology and neurodegeneration, and present with a more disseminated and accelerated disease course, compared to young animals. Here we review recent advances in the investigation of the role of aging in preclinical PD research, including challenges related to aged animal models that are limiting widespread use. Overall, current findings indicate that the use of aged animals may be required to account for age-related interactions in PD pathophysiology. Thus, although the use of older animals has disadvantages, a model that better represents clinical disease within the elderly would be more beneficial in the long run, as it will increase translational value and minimize the risk of therapies failing during clinical studies. Furthermore, we provide recommendations to manage the challenges related to aged animal models.

4.
Front Aging Neurosci ; 14: 907293, 2022.
Article in English | MEDLINE | ID: mdl-35693346

ABSTRACT

Pathogenic alpha-synuclein (asyn) aggregates are a defining feature of neurodegenerative synucleinopathies, which include Parkinson's disease, Lewy body dementia, pure autonomic failure and multiple system atrophy. Early accurate differentiation between these synucleinopathies is challenging due to the highly heterogeneous clinical profile at early prodromal disease stages. Therefore, diagnosis is often made in late disease stages when a patient presents with a broad range of motor and non-motor symptoms easing the differentiation. Increasing data suggest the clinical heterogeneity seen in patients is explained by the presence of distinct asyn strains, which exhibit variable morphologies and pathological functions. Recently, asyn seed amplification assays (PMCA and RT-QuIC) and conformation-specific ligand assays have made promising progress in differentiating between synucleinopathies in prodromal and advanced disease stages. Importantly, the cellular environment is known to impact strain morphology. And, asyn aggregate pathology can propagate trans-synaptically along the brain-body axis, affecting multiple organs and propagating through multiple cell types. Here, we present our hypothesis that the changing cellular environments, an asyn seed may encounter during its brain-to-body or body-to-brain propagation, may influence the structure and thereby the function of the aggregate strains developing within the different cells. Additionally, we aim to review strain characteristics of the different synucleinopathies in clinical and preclinical studies. Future preclinical animal models of synucleinopathies should investigate if asyn strain morphology is altered during brain-to-body and body-to-brain spreading using these seeding amplification and conformation-specific assays. Such findings would greatly deepen our understanding of synucleinopathies and the potential link between strain and phenotypic variability, which may enable specific diagnosis of different synucleinopathies in the prodromal phase, creating a large therapeutic window with potential future applications in clinical trials and personalized therapeutics.

5.
EJNMMI Res ; 12(1): 17, 2022 Apr 01.
Article in English | MEDLINE | ID: mdl-35362761

ABSTRACT

BACKGROUND: The autonomic nervous system is frequently affected in some neurodegenerative diseases, including Parkinson's disease and Dementia with Lewy bodies. In vivo imaging methods to visualize and quantify the peripheral cholinergic nervous system are lacking. By using [18F]FEOBV PET, we here describe the peripheral distribution of the specific cholinergic marker, vesicular acetylcholine transporters (VAChT), in human subjects. We included 15 healthy subjects aged 53-86 years for 70 min dynamic PET protocol of peripheral organs. We performed kinetic modelling of the adrenal gland, pancreas, myocardium, renal cortex, spleen, colon, and muscle using an image-derived input function from the aorta. A metabolite correction model was generated from venous blood samples. Three non-linear compartment models were tested. Additional time-activity curves from 6 to 70 min post injection were generated for prostate, thyroid, submandibular-, parotid-, and lacrimal glands. RESULTS: A one-tissue compartment model generated the most robust fits to the data. Total volume-of-distribution rank order was: adrenal gland > pancreas > myocardium > spleen > renal cortex > muscle > colon. We found significant linear correlations between total volumes-of-distribution and standard uptake values in most organs. CONCLUSION: High [18F]FEOBV PET signal was found in structures with known cholinergic activity. We conclude that [18F]FEOBV PET is a valid tool for estimating VAChT density in human peripheral organs. Simple static images may replace kinetic modeling in some organs and significantly shorten scan duration. Clinical Trial Registration Trial registration: NCT, NCT03554551. Registered 31 May 2018. https://clinicaltrials.gov/ct2/show/NCT03554551?term=NCT03554551&draw=2&rank=1 .

6.
Biomolecules ; 12(2)2022 01 20.
Article in English | MEDLINE | ID: mdl-35204668

ABSTRACT

Alpha-synucleinopathies include Parkinson's disease, dementia with Lewy bodies, pure autonomic failure and multiple system atrophy. These are all progressive neurodegenerative diseases that are characterized by pathological misfolding and accumulation of the protein alpha-synuclein (αsyn) in neurons, axons or glial cells in the brain, but also in other organs. The abnormal accumulation and propagation of pathogenic αsyn across the autonomic connectome is associated with progressive loss of neurons in the brain and peripheral organs, resulting in motor and non-motor symptoms. To date, no cure is available for synucleinopathies, and therapy is limited to symptomatic treatment of motor and non-motor symptoms upon diagnosis. Recent advances using passive immunization that target different αsyn structures show great potential to block disease progression in rodent studies of synucleinopathies. However, passive immunotherapy in clinical trials has been proven safe but less effective than in preclinical conditions. Here we review current achievements of passive immunotherapy in animal models of synucleinopathies. Furthermore, we propose new research strategies to increase translational outcome in patient studies, (1) by using antibodies against immature conformations of pathogenic αsyn (monomers, post-translationally modified monomers, oligomers and protofibrils) and (2) by focusing treatment on body-first synucleinopathies where damage in the brain is still limited and effective immunization could potentially stop disease progression by blocking the spread of pathogenic αsyn from peripheral organs to the brain.


Subject(s)
Synucleinopathies , alpha-Synuclein , Animals , Humans , Immunization, Passive , Lewy Bodies/metabolism , Models, Animal , alpha-Synuclein/metabolism
7.
Neurobiol Dis ; 163: 105599, 2022 02.
Article in English | MEDLINE | ID: mdl-34952161

ABSTRACT

Alpha-synuclein aggregates are the hallmark pathology of Parkinson's disease, which can propagate in a stereotypical pattern along the brain-body axis. Parkinson's disease patients not only display heterogeneous symptoms but also show variable patterns of alpha-synuclein pathology and affected neuronal systems during the disease course, complicating early and accurate diagnosis. Emerging data from post-mortem and imaging studies strongly suggest that disease heterogeneity could, at least in part, be explained by variable disease onset site, i.e. brain or body. This has led to the recently hypothesized formulation of two Parkinson's disease-subtypes, a body-first subtype where pathogenic alpha-synuclein arises in the body and spreads to the brain, and a brain-first subtype where pathogenic alpha-synuclein arises in the brain and spreads to the body. From a preclinical perspective, several animal models have been adapted or developed to reproduce Parkinson's disease-like pathology in the brain or periphery aiming to address the site of disease onset. Here, we review the current rodent and primate models that aim to reproduce Parkinson's disease pathology development and spreading in the brain and/or body and discuss the value and shortcomings of these models for the development of potential future applications in clinical trials and personalized medicine.


Subject(s)
Brain/physiopathology , Disease Models, Animal , Parkinson Disease/physiopathology , alpha-Synuclein/metabolism , Animals , Brain/metabolism , Parkinson Disease/metabolism
8.
Neurobiol Dis ; 161: 105557, 2021 12.
Article in English | MEDLINE | ID: mdl-34763110

ABSTRACT

Aggregation of alpha-synuclein into inclusion bodies, termed Lewy pathology, is a defining feature of Parkinson's disease (PD) and Dementia with Lewy bodies (DLB). In the majority of post mortem cases, the distribution of Lewy pathology seems to follow two overarching patterns: a caudo-rostral pattern with relatively more pathology in the brainstem than in the telencephalon, and an amygdala-centered pattern with the most abundant pathology in the "center of the brain", including the amygdala, entorhinal cortex, and substantia nigra, and relatively less pathology in the lower brainstem and spinal autonomic nuclei. The recent body-first versus brain-first model of Lewy Body Disorders proposes that the initial pathogenic alpha-synuclein in some patients originates in the enteric nervous system with secondary spreading to the brain; and in other patients originates inside the CNS with secondary spreading to the lower brainstem and peripheral autonomic nervous system. Here, we use two existing post mortem datasets to explore the possibility that clinical body-first and brain-first subtypes are equivalent to the caudo-rostral and amygdala-centered patterns of Lewy pathology seen at post mortem.


Subject(s)
Lewy Body Disease , Parkinson Disease , Brain/metabolism , Brain Stem/metabolism , Humans , Lewy Body Disease/pathology , Parkinson Disease/pathology , Substantia Nigra/metabolism , alpha-Synuclein/metabolism
9.
J Parkinsons Dis ; 11(4): 1677-1687, 2021.
Article in English | MEDLINE | ID: mdl-34334424

ABSTRACT

BACKGROUND: We have hypothesized that Parkinson's disease (PD) comprises two subtypes. Brain-first, where pathogenic α-synuclein initially forms unilaterally in one hemisphere leading to asymmetric nigrostriatal degeneration, and body-first with initial enteric pathology, which spreads through overlapping vagal innervation leading to more symmetric brainstem involvement and hence more symmetric nigrostriatal degeneration. Isolated REM sleep behaviour disorder has been identified as a strong marker of the body-first type. OBJECTIVE: To analyse striatal asymmetry in [18F]FDOPA PET and [123I]FP-CIT DaT SPECT data from iRBD patients, de novo PD patients with RBD (PD+RBD) and de novo PD patients without RBD (PD-RBD). These groups were defined as prodromal body-first, de novo body-first, and de novo brain-first, respectively. METHODS: We included [18F]FDOPA PET scans from 21 iRBD patients, 11 de novo PD+RBD, 22 de novo PD-RBD, and 18 controls subjects. Also, [123I]FP-CIT DaT SPECT data from iRBD and de novo PD patients with unknown RBD status from the PPPMI dataset was analysed. Lowest putamen specific binding ratio and putamen asymmetry index (AI) was defined. RESULTS: Nigrostriatal degeneration was significantly more symmetric in patients with RBD versus patients without RBD or with unknown RBD status in both FDOPA (p = 0.001) and DaT SPECT (p = 0.001) datasets. CONCLUSION: iRBD subjects and de novo PD+RBD patients present with significantly more symmetric nigrostriatal dopaminergic degeneration compared to de novo PD-RBD patients. The results support the hypothesis that body-first PD is characterized by more symmetric distribution most likely due to more symmetric propagation of pathogenic α-synuclein compared to brain-first PD.


Subject(s)
Parkinson Disease , REM Sleep Behavior Disorder , Brain/diagnostic imaging , Brain/metabolism , Dopamine/chemistry , Dopamine Plasma Membrane Transport Proteins/chemistry , Dopamine Plasma Membrane Transport Proteins/metabolism , Humans , Parkinson Disease/diagnostic imaging , REM Sleep Behavior Disorder/diagnostic imaging , Tomography, Emission-Computed, Single-Photon , alpha-Synuclein/metabolism
10.
Brain ; 144(6): 1853-1868, 2021 07 28.
Article in English | MEDLINE | ID: mdl-33880502

ABSTRACT

Neuronal aggregates of misfolded alpha-synuclein protein are found in the brain and periphery of patients with Parkinson's disease. Braak and colleagues have hypothesized that the initial formation of misfolded alpha-synuclein may start in the gut, and then spread to the brain via peripheral autonomic nerves hereby affecting several organs, including the heart and intestine. Age is considered the greatest risk factor for Parkinson's disease, but the effect of age on the formation of pathology and its propagation has not been studied in detail. We aimed to investigate whether propagation of alpha-synuclein pathology from the gut to the brain is more efficient in old versus young wild-type rats, upon gastrointestinal injection of aggregated alpha-synuclein. Our results demonstrate a robust age-dependent gut-to-brain and brain-to-gut spread of alpha-synuclein pathology along the sympathetic and parasympathetic nerves, resulting in age-dependent dysfunction of the heart and stomach, as observed in patients with Parkinson's disease. Moreover, alpha-synuclein pathology is more densely packed and resistant to enzymatic digestion in old rats, indicating an age-dependent maturation of alpha-synuclein aggregates. Our study is the first to provide a detailed investigation of alpha-synuclein pathology in several organs within one animal model, including the brain, skin, heart, intestine, spinal cord and autonomic ganglia. Taken together, our findings suggest that age is a crucial factor for alpha-synuclein aggregation and complete propagation to heart, stomach and skin, similar to patients. Given that age is the greatest risk factor for human Parkinson's disease, it seems likely that older experimental animals will yield the most relevant and reliable findings. These results have important implications for future research to optimize diagnostics and therapeutics in Parkinson's disease and other age-associated synucleinopathies. Increased emphasis should be placed on using aged animals in preclinical studies and to elucidate the nature of age-dependent interactions.


Subject(s)
Aging/pathology , Primary Dysautonomias/etiology , alpha-Synuclein/toxicity , Aging/metabolism , Animals , Autonomic Nervous System/drug effects , Autonomic Nervous System/metabolism , Autonomic Nervous System/pathology , Brain/pathology , Duodenum/drug effects , Duodenum/pathology , Kidney/pathology , Muscle, Skeletal/pathology , Myocardium/pathology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Primary Dysautonomias/metabolism , Primary Dysautonomias/pathology , Protein Aggregation, Pathological/pathology , Rats, Inbred F344 , Skin/pathology , Spinal Cord/pathology , Stomach/drug effects , Stomach/pathology
11.
Parkinsonism Relat Disord ; 75: 63-69, 2020 06.
Article in English | MEDLINE | ID: mdl-32480309

ABSTRACT

INTRODUCTION: Noradrenergic denervation is thought to aggravate motor dysfunction in Parkinson's disease (PD). In a previous PET study with the norepinephrine transporter (NART) ligand 11C-MeNER, we detected reduced NART binding in primary sensorimotor cortex (M1S1) of PD patients. Idiopathic rapid-eye-movement sleep behaviour disorder (iRBD) is a phenotype of prodromal PD. Using 11C-MeNER PET, we investigated whether iRBD patients showed similar NART binding reductions in M1S1 cortex as PD patients. Additionally, we investigated whether 11C-MeNER binding and loss of nigrostriatal dopamine storage capacity measured with 18F-DOPA PET were correlated. METHODS: 17 iRBD patients, 16 PD patients with (PDRBD+) and 14 without RBD (PDRBD-), and 25 control subjects underwent 11C-MeNER PET. iRBD patients also had 18F-DOPA PET. Volume-of-interest analyses and voxel-level statistical parametric mapping were performed. RESULTS: Partial-volume corrected 11C-MeNER binding potential (BPND) values in M1S1 differed across the groups (P = 0.022) with the iRBD and PDRBD+ groups showing significant reductions (controls vs. iRBD P = 0.007; control vs. PDRBD+P = 0.008). Voxel-wise comparisons confirmed reductions of M1S1 11C-MeNER binding in PD and iRBD patients. Significant correlation was seen between putaminal 18F-DOPA uptake and thalamic 11C-MeNER binding in iRBD patients (r2 = 0.343, P = 0.013). CONCLUSIONS: This study found altered noradrenergic neurotransmission in the M1S1 cortex of iRBD patients. The observed reduction of M1S1 11C-MeNER binding in iRBD may represent noradrenergic terminal degeneration or physiological down-regulation of NARTs in this prodromal phenotype of PD. The correlation between thalamic 11C-MeNER binding and putaminal 18F-DOPA binding suggests that these neurotransmitter systems degenerate in parallel in the iRBD phenotype of prodromal PD.


Subject(s)
Norepinephrine/metabolism , Parkinson Disease/metabolism , Putamen/metabolism , REM Sleep Behavior Disorder/metabolism , Sensorimotor Cortex/metabolism , Thalamus/metabolism , Aged , Dihydroxyphenylalanine/analogs & derivatives , Female , Humans , Male , Middle Aged , Morpholines , Parkinson Disease/complications , Parkinson Disease/diagnostic imaging , Positron-Emission Tomography , Putamen/diagnostic imaging , REM Sleep Behavior Disorder/diagnostic imaging , REM Sleep Behavior Disorder/etiology , Sensorimotor Cortex/diagnostic imaging , Thalamus/diagnostic imaging
12.
J Parkinsons Dis ; 9(s2): S281-S295, 2019.
Article in English | MEDLINE | ID: mdl-31498132

ABSTRACT

Parkinson's disease (PD) is a highly heterogeneous disorder, which probably consists of multiple subtypes. Aggregation of misfolded alpha-synuclein and propagation of these proteinacious aggregates through interconnected neural networks is believed to be a crucial pathogenetic factor. It has been hypothesized that the initial pathological alpha-synuclein aggregates originate in the enteric or peripheral nervous system (PNS) and invade the central nervous system (CNS) via retrograde vagal transport. However, evidence from neuropathological studies suggests that not all PD patients can be reconciled with this hypothesis. Importantly, a small fraction of patients do not show pathology in the dorsal motor nucleus of the vagus. Here, it is hypothesized that PD can be divided into a PNS-first and a CNS-first subtype. The former is tightly associated with REM sleep behavior disorder (RBD) during the prodromal phase and is characterized by marked autonomic damage before involvement of the dopaminergic system. In contrast, the CNS-first phenotype is most often RBD-negative during the prodromal phase and characterized by nigrostriatal dopaminergic dysfunction prior to involvement of the autonomic PNS. The existence of these subtypes is supported by in vivo imaging studies of RBD-positive and RBD-negative patient groups and by histological evidence- reviewed herein. The present proposal provides a fresh hypothesis-generating framework for future studies into the etiopathogenesis of PD and seems capable of explaining a number of discrepant findings in the neuropathological literature.


Subject(s)
Autonomic Nervous System , Central Nervous System , Dopamine/metabolism , Parkinson Disease , Prodromal Symptoms , alpha-Synuclein/metabolism , Autonomic Nervous System/metabolism , Autonomic Nervous System/pathology , Autonomic Nervous System/physiopathology , Central Nervous System/metabolism , Central Nervous System/pathology , Central Nervous System/physiopathology , Humans , Parkinson Disease/etiology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Parkinson Disease/physiopathology
13.
Acta Neuropathol ; 138(4): 535-550, 2019 10.
Article in English | MEDLINE | ID: mdl-31254094

ABSTRACT

The conversion of endogenous alpha-synuclein (asyn) to pathological asyn-enriched aggregates is a hallmark of Parkinson's disease (PD). These inclusions can be detected in the central and enteric nervous system (ENS). Moreover, gastrointestinal symptoms can appear up to 20 years before the diagnosis of PD. The dual-hit hypothesis posits that pathological asyn aggregation starts in the ENS, and retrogradely spreads to the brain. In this study, we tested this hypothesis by directly injecting preformed asyn fibrils into the duodenum wall of wild-type rats and transgenic rats with excess levels of human asyn. We provide a meticulous characterization of the bacterial artificial chromosome (BAC) transgenic rat model with respect to initial propagation of pathological asyn along the parasympathetic and sympathetic pathways to the brainstem, by performing immunohistochemistry at early time points post-injection. Induced pathology was observed in all key structures along the sympathetic and parasympathetic pathways (ENS, autonomic ganglia, intermediolateral nucleus of the spinal cord (IML), heart, dorsal motor nucleus of the vagus, and locus coeruleus (LC)) and persisted for at least 4 months post-injection. In contrast, asyn propagation was not detected in wild-type rats, nor in vehicle-injected BAC rats. The presence of pathology in the IML, LC, and heart indicate trans-synaptic spread of the pathology. Additionally, the observed asyn inclusions in the stomach and heart may indicate secondary anterograde propagation after initial retrograde spreading. In summary, trans-synaptic propagation of asyn in the BAC rat model is fully compatible with the "body-first hypothesis" of PD etiopathogenesis. To our knowledge, this is the first animal model evidence of asyn propagation to the heart, and the first indication of bidirectional asyn propagation via the vagus nerve, i.e., duodenum-to-brainstem-to-stomach. The BAC rat model could be very valuable for detailed mechanistic studies of the dual-hit hypothesis, and for studies of disease modifying therapies targeting early pathology in the gastrointestinal tract.


Subject(s)
Brain/metabolism , Neurons/metabolism , Parkinson Disease/pathology , Synapses/metabolism , alpha-Synuclein/metabolism , Animals , Autonomic Nervous System/metabolism , Autonomic Nervous System/pathology , Brain/pathology , Disease Models, Animal , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/pathology , Neurons/pathology , Parkinson Disease/metabolism , Rats , Rats, Sprague-Dawley , Spinal Cord/metabolism , Spinal Cord/pathology , Synapses/pathology , alpha-Synuclein/administration & dosage
14.
Neuroimage ; 146: 1050-1061, 2017 02 01.
Article in English | MEDLINE | ID: mdl-27825979

ABSTRACT

The substantia nigra pars reticulata (SNr) and external globus pallidus (GPe) constitute the two major output targets of the rodent striatum. Both the SNr and GPe converge upon thalamic relay nuclei (directly or indirectly, respectively), and are traditionally modeled as functionally antagonistic relay inputs. However, recent anatomical and functional studies have identified unanticipated circuit connectivity in both the SNr and GPe, demonstrating their potential as far more than relay nuclei. In the present study, we employed simultaneous deep brain stimulation and functional magnetic resonance imaging (DBS-fMRI) with cerebral blood volume (CBV) measurements to functionally and unbiasedly map the circuit- and network level connectivity of the SNr and GPe. Sprague-Dawley rats were implanted with a custom-made MR-compatible stimulating electrode in the right SNr (n=6) or GPe (n=7). SNr- and GPe-DBS, conducted across a wide range of stimulation frequencies, revealed a number of surprising evoked responses, including unexpected CBV decreases within the striatum during DBS at either target, as well as GPe-DBS-evoked positive modulation of frontal cortex. Functional connectivity MRI revealed global modulation of neural networks during DBS at either target, sensitive to stimulation frequency and readily reversed following cessation of stimulation. This work thus contributes to a growing literature demonstrating extensive and unanticipated functional connectivity among basal ganglia nuclei.


Subject(s)
Globus Pallidus/physiology , Pars Reticulata/physiology , Animals , Brain/physiology , Brain Mapping/methods , Electric Stimulation , Magnetic Resonance Imaging , Male , Neural Pathways/physiology , Rats, Sprague-Dawley
15.
Sci Rep ; 6: 31613, 2016 09 07.
Article in English | MEDLINE | ID: mdl-27601003

ABSTRACT

Deep brain stimulation of the nucleus accumbens (NAc-DBS) is an emerging therapy for diverse, refractory neuropsychiatric diseases. Although DBS therapy is broadly hypothesized to work through large-scale neural modulation, little is known regarding the neural circuits and networks affected by NAc-DBS. Using a healthy, sedated rat model of NAc-DBS, we employed both evoked- and functional connectivity (fc) MRI to examine the functional circuit and network changes achieved by electrical NAc stimulation. Optogenetic-fMRI experiments were also undertaken to evaluate the circuit modulation profile achieved by selective stimulation of NAc neurons. NAc-DBS directly modulated neural activity within prefrontal cortex and a large number of subcortical limbic areas (e.g., amygdala, lateral hypothalamus), and influenced functional connectivity among sensorimotor, executive, and limbic networks. The pattern and extent of circuit modulation measured by evoked-fMRI was relatively insensitive to DBS frequency. Optogenetic stimulation of NAc cell bodies induced a positive fMRI signal in the NAc, but no other detectable downstream responses, indicating that therapeutic NAc-DBS might exert its effect through antidromic stimulation. Our study provides a comprehensive mapping of circuit and network-level neuromodulation by NAc-DBS, which should facilitate our developing understanding of its therapeutic mechanisms of action.


Subject(s)
Deep Brain Stimulation/methods , Magnetic Resonance Imaging/methods , Nucleus Accumbens/physiology , Optogenetics/methods , Amygdala/physiology , Animals , Brain Mapping , Hypothalamic Area, Lateral/physiology , Male , Nerve Net/physiology , Neural Pathways/physiology , Neurons/physiology , Nucleus Accumbens/cytology , Prefrontal Cortex/physiology , Rats, Sprague-Dawley
16.
PLoS One ; 10(7): e0133245, 2015.
Article in English | MEDLINE | ID: mdl-26193653

ABSTRACT

Deep Brain Stimulation (DBS) is a promising treatment for neurological and psychiatric disorders. The mechanism of action and the effects of electrical fields administered to the brain by means of an electrode remain to be elucidated. The effects of DBS have been investigated primarily by electrophysiological and neurochemical studies, which lack the ability to investigate DBS-related responses on a whole-brain scale. Visualization of whole-brain effects of DBS requires functional imaging techniques such as functional Magnetic Resonance Imaging (fMRI), which reflects changes in blood oxygen level dependent (BOLD) responses throughout the entire brain volume. In order to visualize BOLD responses induced by DBS, we have developed an MRI-compatible electrode and an acquisition protocol to perform DBS during BOLD fMRI. In this study, we investigate whether DBS during fMRI is valuable to study local and whole-brain effects of hippocampal DBS and to investigate the changes induced by different stimulation intensities. Seven rats were stereotactically implanted with a custom-made MRI-compatible DBS-electrode in the right hippocampus. High frequency Poisson distributed stimulation was applied using a block-design paradigm. Data were processed by means of Independent Component Analysis. Clusters were considered significant when p-values were <0.05 after correction for multiple comparisons. Our data indicate that real-time hippocampal DBS evokes a bilateral BOLD response in hippocampal and other mesolimbic structures, depending on the applied stimulation intensity. We conclude that simultaneous DBS and fMRI can be used to detect local and whole-brain responses to circuit activation with different stimulation intensities, making this technique potentially powerful for exploration of cerebral changes in response to DBS for both preclinical and clinical DBS.


Subject(s)
Deep Brain Stimulation , Hippocampus/physiology , Animals , Functional Neuroimaging , Magnetic Resonance Imaging , Male , Rats , Rats, Sprague-Dawley
17.
Mol Imaging Biol ; 17(3): 373-83, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25361593

ABSTRACT

PURPOSE: The effects of deep brain stimulation (DBS) have been studied primarily by cellular studies, which lack the ability to elucidate DBS-related responses on a whole-brain scale. 2-Deoxy-2-[(18)F]fluoro-D-glucose positron emission tomography ([(18)F]FDG-PET) reflects changes in neural activity throughout the entire brain volume. The aim of this study was to investigate the whole-brain effect of DBS on the glucose utilization in healthy rats. PROCEDURES: Seven rats were implanted with a DBS electrode in the right hippocampus and injected with [(18)F]FDG to measure the glucose metabolism during DBS. RESULTS: Analysis reveals significant DBS-induced decreases in the glucose metabolism in the bilateral hippocampus and other limbic structures. CONCLUSIONS: This study demonstrates that DBS exhibits not only a local effect around the electrode tip but also in other limbic regions. [(18)F]FDG-PET studies have the potential to provide better insight into the mechanism of action of DBS by simultaneously observing activity at multiple sites in the brain.


Subject(s)
Deep Brain Stimulation , Glucose/chemistry , Hippocampus/diagnostic imaging , Limbic System/diagnostic imaging , Animals , Disease Models, Animal , Electrodes , Epilepsy/diagnostic imaging , Epilepsy/physiopathology , Fluorodeoxyglucose F18/chemistry , Glucose/metabolism , Hippocampus/metabolism , Limbic System/metabolism , Male , Multimodal Imaging , Neuroimaging , Neurons/metabolism , Poisson Distribution , Positron-Emission Tomography , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...