Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 707
Filter
1.
Brain Behav ; 14(7): e3607, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39010690

ABSTRACT

BACKGROUND: Pathologic perivascular spaces (PVS), the fluid-filled compartments surrounding brain vasculature, may underlie cognitive decline in Parkinson's disease (PD). However, whether this impacts specific cognitive domains has not been investigated. OBJECTIVES: This study examined the relationship of PVS volume at baseline with domain-specific and global cognitive change over 2 years in PD individuals. METHODS: A total of 39 individuals with PD underwent 3T T1w magnetic resonance imaging to determine PVS volume fraction (PVS volume normalized to total regional volume) within (i) centrum semiovale, (ii) prefrontal white matter (medial orbitofrontal, rostral middle frontal, and superior frontal), and (iii) basal ganglia. A neuropsychological battery included assessment of cognitive domains and global cognitive function at baseline and after 2 years. RESULTS: Higher basal ganglia PVS at baseline was associated with greater decline in attention, executive function, and global cognition scores. CONCLUSIONS: While previous reports have associated elevated PVS volume in the basal ganglia with decline in global cognition in PD, our findings show such decline may affect the attention and executive function domains.


Subject(s)
Attention , Basal Ganglia , Cognitive Dysfunction , Executive Function , Magnetic Resonance Imaging , Parkinson Disease , Humans , Parkinson Disease/diagnostic imaging , Parkinson Disease/pathology , Parkinson Disease/physiopathology , Basal Ganglia/diagnostic imaging , Basal Ganglia/pathology , Basal Ganglia/physiopathology , Executive Function/physiology , Female , Male , Aged , Middle Aged , Attention/physiology , Cognitive Dysfunction/physiopathology , Cognitive Dysfunction/etiology , Cognitive Dysfunction/diagnostic imaging , Cognitive Dysfunction/pathology , Glymphatic System/diagnostic imaging , Glymphatic System/pathology , Glymphatic System/physiopathology , Neuropsychological Tests , White Matter/diagnostic imaging , White Matter/pathology , White Matter/physiopathology
3.
Fluids Barriers CNS ; 21(1): 55, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38987813

ABSTRACT

BACKGROUND: Studies indicate that brain clearance via the glymphatic system is impaired in idiopathic normal pressure hydrocephalus (INPH). This has been suggested to result from reduced cerebrospinal fluid (CSF) turnover, which could be caused by a reduced CSF formation rate. The aim of this study was to determine the formation rate of CSF in a cohort of patients investigated for INPH and compare this to a historical control cohort. METHODS: CSF formation rate was estimated in 135 (75 ± 6 years old, 64/71 men/women) patients undergoing investigation for INPH. A semiautomatic CSF infusion investigation (via lumbar puncture) was performed. CSF formation rate was assessed by downregulating and steadily maintaining CSF pressure at a zero level. During the last 10 min, the required outflow to maintain zero pressure, i.e., CSF formation rate, was continuously measured. The values were compared to those of a historical reference cohort from a study by Ekstedt in 1978. RESULTS: Mean CSF formation rate was 0.45 ± 0.15 ml/min (N = 135), equivalent to 27 ± 9 ml/hour. There was no difference in the mean (p = 0.362) or variance (p = 0.498) of CSF formation rate between the subjects that were diagnosed as INPH (N = 86) and those who were not (N = 43). The CSF formation rate in INPH was statistically higher than in the reference cohort (0.46 ± 0.15 vs. 0.40 ± 0.08 ml/min, p = 0.005), but the small difference was probably not physiologically relevant. There was no correlation between CSF formation rate and baseline CSF pressure (r = 0.136, p = 0.115, N = 135) or age (-0.02, p = 0.803, N = 135). CONCLUSIONS: The average CSF formation rate in INPH was not decreased compared to the healthy reference cohort, which does not support reduced CSF turnover. This emphasizes the need to further investigate the source and routes of the flow in the glymphatic system and the cause of the suggested impaired glymphatic clearance in INPH.


Subject(s)
Cerebrospinal Fluid , Glymphatic System , Hydrocephalus, Normal Pressure , Humans , Male , Female , Glymphatic System/physiopathology , Hydrocephalus, Normal Pressure/cerebrospinal fluid , Hydrocephalus, Normal Pressure/physiopathology , Aged , Cerebrospinal Fluid/physiology , Aged, 80 and over , Cohort Studies , Spinal Puncture , Cerebrospinal Fluid Pressure/physiology , Middle Aged
4.
Eur J Sport Sci ; 24(7): 975-986, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38956796

ABSTRACT

The neurological effects and underlying pathophysiological mechanisms of sports-related concussion (SRC) in active young boxers remain poorly understood. This study aims to investigate the impairment of white matter microstructure and assess changes in glymphatic function following SRC by utilizing neurite orientation dispersion and density imaging (NODDI) on young boxers who have sustained SRC. A total of 60 young participants were recruited, including 30 boxers diagnosed with SRC and 30 healthy individuals engaging in regular exercise. The assessment of whole-brain white matter damage was conducted using diffusion metrics, while the evaluation of glymphatic function was performed through diffusion tensor imaging (DTI) analysis along the perivascular space (DTI-ALPS) index. A two-sample t-test was utilized to examine group differences in DTI and NODDI metrics. Spearman correlation and generalized linear mixed models were employed to investigate the relationship between clinical assessments of SRC and NODDI measurements. Significant alterations were observed in DTI and NODDI metrics among young boxers with SRC. Additionally, the DTI-ALPS index in the SRC group exhibited a significantly higher value than that of the control group (left side: 1.58 vs. 1.48, PFDR = 0.009; right side: 1.61 vs. 1.51, PFDR = 0.02). Moreover, it was observed that the DTI-ALPS index correlated with poorer cognitive test results among boxers in this study population. Repetitive SRC in active young boxers is associated with diffuse white matter injury and glymphatic dysfunction, highlighting the detrimental impact on brain health. These findings highlight the importance of long-term monitoring of the neurological health of boxers.


Subject(s)
Boxing , Brain Concussion , Diffusion Tensor Imaging , Glymphatic System , Neurites , White Matter , Humans , White Matter/diagnostic imaging , White Matter/pathology , Glymphatic System/diagnostic imaging , Male , Brain Concussion/diagnostic imaging , Brain Concussion/physiopathology , Adolescent , Neurites/physiology , Boxing/injuries , Boxing/physiology , Female , Case-Control Studies , Young Adult , Athletic Injuries/diagnostic imaging , Athletic Injuries/physiopathology
5.
Sci Rep ; 14(1): 13911, 2024 06 17.
Article in English | MEDLINE | ID: mdl-38886538

ABSTRACT

Previous studies have demonstrated associations between enlarged perivascular spaces (EPVS) and dementias such as Alzheimer's disease. However, an association between EPVS and dementia with Lewy bodies (DLB) has not yet been clarified. We performed a cross-sectional analysis of our prospective study cohort of 109 participants (16 with DLB). We assessed cognitive function, pulse wave velocity (PWV), and brain magnetic resonance imaging features. The relationships between EPVS and DLB were evaluated using multivariable logistic regression analyses. Compared with the non-dementia group, the DLB group was more likely to have EPVS in the basal ganglia. Compared with participants without EPVS, those with EPVS were older and had cognitive impairment and high PWV. In multivariable analyses, EPVS in the basal ganglia was independently associated with DLB. High PWV was also independently associated with EPVS in both the basal ganglia and centrum semiovale. High PWV may cause cerebrovascular pulsatility, leading to accelerated EPVS in DLB participants.


Subject(s)
Glymphatic System , Lewy Body Disease , Pulse Wave Analysis , Humans , Lewy Body Disease/physiopathology , Lewy Body Disease/diagnostic imaging , Lewy Body Disease/pathology , Female , Male , Aged , Glymphatic System/diagnostic imaging , Glymphatic System/physiopathology , Glymphatic System/pathology , Cross-Sectional Studies , Magnetic Resonance Imaging , Prospective Studies , Aged, 80 and over , Basal Ganglia/diagnostic imaging , Basal Ganglia/physiopathology , Basal Ganglia/pathology
6.
Int J Geriatr Psychiatry ; 39(6): e6104, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38877354

ABSTRACT

The central nervous system (CNS) is widely recognized as the only organ system without lymphatic capillaries to promote the removal of interstitial metabolic by-products. Thus, the newly identified glymphatic system which provides a pseudolymphatic activity in the nervous system has been focus of latest research in neurosciences. Also, findings reported that, sleep stimulates the elimination actions of glymphatic system and is linked to normal brain homeostatis. The CNS is cleared of potentially hazardous compounds via the glymphatic system, particularly during sleep. Any age-related alterations in brain functioning and pathophysiology of various neurodegenerative illnesses indicates the disturbance of the brain's glymphatic system. In this context, ß-amyloid as well as tau leaves the CNS through the glymphatic system, it's functioning and CSF discharge markedly altered in elderly brains as per many findings. Thus, glymphatic failure may have a potential mechanism which may be therapeutically targetable in several neurodegenerative and age-associated cognitive diseases. Therefore, there is an urge to focus for more research into the connection among glymphatic system and several potential brain related diseases. Here, in our current review paper, we reviewed current research on the glymphatic system's involvement in a number of prevalent neurodegenerative and neuropsychiatric diseases and, we also discussed several therapeutic approaches, diet and life style modifications which might be used to acquire a more thorough performance and purpose of the glymphatic system to decipher novel prospects for clinical applicability for the management of these diseases.


Subject(s)
Glymphatic System , Neurodegenerative Diseases , Humans , Glymphatic System/physiopathology , Glymphatic System/physiology , Neurodegenerative Diseases/physiopathology , Neurodegenerative Diseases/metabolism , Brain/physiopathology , Brain/metabolism , Amyloid beta-Peptides/metabolism
7.
Int J Mol Sci ; 25(11)2024 May 24.
Article in English | MEDLINE | ID: mdl-38891923

ABSTRACT

The ocular glymphatic system subserves the bidirectional polarized fluid transport in the optic nerve, whereby cerebrospinal fluid from the brain is directed along periarterial spaces towards the eye, and fluid from the retina is directed along perivenous spaces following upon its axonal transport across the glial lamina. Fluid homeostasis and waste removal are vital for retinal function, making the ocular glymphatic fluid pathway a potential route for targeted manipulation to combat blinding ocular diseases such as age-related macular degeneration, diabetic retinopathy, and glaucoma. Several lines of work investigating the bidirectional ocular glymphatic transport with varying methodologies have developed diverging mechanistic models, which has created some confusion about how ocular glymphatic transport should be defined. In this review, we provide a comprehensive summary of the current understanding of the ocular glymphatic system, aiming to address misconceptions and foster a cohesive understanding of the topic.


Subject(s)
Glymphatic System , Humans , Glymphatic System/physiology , Glymphatic System/metabolism , Animals , Optic Nerve/metabolism , Optic Nerve/physiology , Retina/metabolism , Retina/physiology , Eye/metabolism , Glaucoma/metabolism , Glaucoma/physiopathology , Glaucoma/pathology
8.
Acta Neurochir (Wien) ; 166(1): 274, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38904802

ABSTRACT

The discovery of the glymphatic system has fundamentally altered our comprehension of cerebrospinal fluid transport and the removal of waste from brain metabolism. In the past decade, since its initial characterization, research on the glymphatic system has surged exponentially. Its potential implications for central nervous system disorders have sparked significant interest in the field of neurosurgery. Nonetheless, ongoing discussions and debates persist regarding the concept of the glymphatic system, and our current understanding largely relies on findings from experimental animal studies. This review aims to address several key inquiries: What methodologies exist for evaluating glymphatic function in humans today? What is the current evidence supporting the existence of a human glymphatic system? Can the glymphatic system be considered distinct from the meningeal-lymphatic system? What is the human evidence for glymphatic-meningeal lymphatic system failure in neurosurgical diseases? Existing literature indicates a paucity of techniques available for assessing glymphatic function in humans. Thus far, intrathecal contrast-enhanced magnetic resonance imaging (MRI) has shown the most promising results and have provided evidence for the presence of a glymphatic system in humans, albeit with limitations. It is, however, essential to recognize the interconnection between the glymphatic and meningeal lymphatic systems, as they operate in tandem. There are some human studies demonstrating deteriorations in glymphatic function associated with neurosurgical disorders, enriching our understanding of their pathophysiology. However, the translation of this knowledge into clinical practice is hindered by the constraints of current glymphatic imaging modalities.


Subject(s)
Glymphatic System , Humans , Glymphatic System/physiology , Glymphatic System/surgery , Neurosurgical Procedures/methods , Meninges/surgery , Animals , Magnetic Resonance Imaging/methods
9.
CNS Neurosci Ther ; 30(6): e14803, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38887168

ABSTRACT

The glymphatic system is cerebrospinal fluid-brain tissue fluid exchange flow mediated by aquaporin-4 (AQP4) on the end feet of astrocytes for a system, which is capable of rapidly removing brain metabolites and thus maintaining brain homeostasis, and is known as the central immune system. Dysfunction of the glymphatic system causes accumulation of misfolded and highly phosphorylated proteins (amyloid-ß and Tau proteins), which destabilizes the proteins, and the body's neuroinflammatory factors are altered causing aging of the immune system and leading to neurodegenerative diseases. Damage to the glymphatic system and aging share common manifestations, as well as unstudied biological mechanisms that are also linked, such as mitochondria, oxidative stress, chronic inflammation, and sleep. In this paper, we first summarize the structure, function, and research methods of the glymphatic system and the relationship between the glymphatic system and the peripheral immune system, and second, sort out and summarize the factors of the glymphatic system in removing metabolites and resolving aging-related diseases and factors affecting aging, to explore its related biological mechanisms, and moreover, to provide a new way of thinking for treating or intervening aging-related diseases.


Subject(s)
Aging , Glymphatic System , Humans , Glymphatic System/physiology , Glymphatic System/metabolism , Aging/physiology , Aging/metabolism , Animals , Astrocytes/metabolism , Brain/metabolism , Aquaporin 4/metabolism
10.
Cereb Cortex ; 34(6)2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38912605

ABSTRACT

Glymphatic dysfunction has been correlated with cognitive decline, with a higher choroid plexus volume (CPV) being linked to a slower glymphatic clearance rate. Nevertheless, the interplay between CPV, glymphatic function, and cognitive impairment in white matter hyperintensities (WMHs) has not yet been investigated. In this study, we performed neuropsychological assessment, T1-weighted three-dimensional (3D-T1) images, and diffusion tensor imaging (DTI) in a cohort of 206 WMHs subjects and 43 healthy controls (HCs) to further explore the relationship. The DTI analysis along the perivascular space (DTI-ALPS) index, as a measure of glymphatic function, was calculated based on DTI. Severe WMHs performed significantly worse in information processing speed (IPS) than other three groups, as well as in executive function than HCs and mild WMHs. Additionally, severe WMHs demonstrated lower DTI-ALPS index and higher CPV than HCs and mild WMHs. Moderate WMHs displayed higher CPV than HCs and mild WMHs. Mini-Mental State Examination, IPS, and executive function correlated negatively with CPV but positively with DTI-ALPS index in WMHs patients. Glymphatic function partially mediated the association between CPV and IPS, indicating a potential mechanism for WMHs-related cognitive impairment. CPV may act as a valuable prognostic marker and glymphatic system as a promising therapeutic target for WMHs-related cognitive impairment.


Subject(s)
Choroid Plexus , Cognitive Dysfunction , Diffusion Tensor Imaging , Glymphatic System , White Matter , Humans , Male , Female , Choroid Plexus/diagnostic imaging , Choroid Plexus/pathology , Choroid Plexus/physiopathology , White Matter/diagnostic imaging , White Matter/pathology , Aged , Glymphatic System/diagnostic imaging , Glymphatic System/pathology , Glymphatic System/physiopathology , Middle Aged , Cognitive Dysfunction/diagnostic imaging , Cognitive Dysfunction/physiopathology , Cognitive Dysfunction/pathology , Neuropsychological Tests , Magnetic Resonance Imaging/methods , Processing Speed
11.
Cell Death Dis ; 15(6): 448, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38918408

ABSTRACT

Multiple sevoflurane exposures may damage the developing brain. The neuroprotective function of dexmedetomidine has been widely confirmed in animal experiments and human studies. However, the effect of dexmedetomidine on the glymphatic system has not been clearly studied. We hypothesized that dexmedetomidine could alleviate sevoflurane-induced circulatory dysfunction of the glymphatic system in young mice. Six-day-old C57BL/6 mice were exposed to 3% sevoflurane for 2 h daily, continuously for 3 days. Intraperitoneal injection of either normal saline or dexmedetomidine was administered before every anaesthesia. Meanwhile the circulatory function of glymphatic system was detected by tracer injection at P8 and P32. On P30-P32, behavior tests including open field test, novel object recognition test, and Y-maze test were conducted. Primary astrocyte cultures were established and treated with the PI3K activator 740Y-P, dexmedetomidine, and small interfering RNA (siRNA) to silence ΔFosB. We propose for the first time that multiple exposure to sevoflurane induces circulatory dysfunction of the glymphatic system in young mice. Dexmedetomidine improves the circulatory capacity of the glymphatic system in young mice following repeated exposure to sevoflurane through the PI3K/AKT/ΔFosB/AQP4 signaling pathway, and enhances their long-term learning and working memory abilities.


Subject(s)
Aquaporin 4 , Dexmedetomidine , Glymphatic System , Mice, Inbred C57BL , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Sevoflurane , Signal Transduction , Animals , Dexmedetomidine/pharmacology , Sevoflurane/pharmacology , Sevoflurane/adverse effects , Glymphatic System/drug effects , Glymphatic System/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Mice , Phosphatidylinositol 3-Kinases/metabolism , Aquaporin 4/metabolism , Aquaporin 4/genetics , Signal Transduction/drug effects , Astrocytes/drug effects , Astrocytes/metabolism , Male
12.
Sci Rep ; 14(1): 12966, 2024 06 05.
Article in English | MEDLINE | ID: mdl-38839864

ABSTRACT

The inflow of CSF into perivascular spaces (PVS) in the brain is crucial for clearing waste molecules. Inefficiency in PVS flow leads to neurodegeneration. Failure of PVS flushing is associated with CSF flow impairment in the intracranial hydrodynamic condition of CSF hypo-pulsatility. However, enlarged PVS (ePVS), a finding indicative of PVS flow dysfunction, is also present in patients with derangement of CSF dynamics characterized by CSF hyper-pulsatility, which increases CSF flow. Intriguingly, two opposite intracranial hydrodynamic conditions would lead to the same result of impairing the PVS flushing. To investigate this issue, we assessed the subsistence of a dysfunctional interplay between CSF and PVS flows and, if the case, the mechanisms preventing a hyper-pulsatile brain from providing an effective PVS flushing. We analyzed the association between phase contrast MRI aqueductal CSF stroke volume (aqSV), a proxy of CSF pulsatility, and the burden of ePVS in chronic adult hydrocephalus, a disease involving a broad spectrum of intracranial hydrodynamics disturbances. In the 147 (85 males, 62 females) patients, the age at diagnosis ranged between 28 and 88 years (median 73 years). Ninety-seven patients had tri-ventriculomegaly and 50 tetra-ventriculomegaly. According to the extent of ePVS, 113 patients had a high ePVS burden, while 34 had a low ePVS burden. aqSV, which ranged between 0 and 562 µL (median 86 µL), was increased with respect to healthy subjects. Patients presenting with less ePVS burden had higher aqSV (p < 0.002, corrected for the multiple comparisons) than those with higher ePVS burden. The present study confirmed the association between CSF dynamics and PVS flow disturbances and demonstrated this association in intracranial hyper-pulsatility. Further studies should investigate the association between PVS flow failure and CSF hypo- and hyper-pulsatility as responsible/co-responsible for glymphatic failure in other neurodegenerative diseases, particularly in diseases in which CSF disturbances can be corrected, as in chronic adult hydrocephalus.


Subject(s)
Glymphatic System , Hydrocephalus , Magnetic Resonance Imaging , Humans , Hydrocephalus/cerebrospinal fluid , Hydrocephalus/physiopathology , Hydrocephalus/pathology , Male , Female , Aged , Middle Aged , Adult , Glymphatic System/physiopathology , Glymphatic System/pathology , Aged, 80 and over , Cerebrospinal Fluid , Hydrodynamics , Stroke Volume , Cerebral Aqueduct/pathology , Cerebral Aqueduct/physiopathology , Chronic Disease
14.
Neurology ; 103(1): e209529, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38833652

ABSTRACT

BACKGROUND AND OBJECTIVES: Idiopathic intracranial hypertension (IIH) is a neurologic disorder characterized by symptoms of elevated intracranial pressure in the absence of a clear cause. There is a developing theory that IIH may, in part, be related to abnormal cerebral glymphatic clearance. In addition, transverse sinus stenosis (TSS) is a common finding in IIH of unclear pathophysiologic significance. Similarly, whether or not TSS is associated with glymphatic outflow in IIH is unknown. The aim of this investigation was to explore the possible association between glymphatic outflow and extent of TSS in patients with IIH. METHODS: The study cohort consisted of patients with IIH and healthy controls who were retrospectively identified from our tertiary care institution located in upstate New York from 2016 to 2023. Patients with IIH were included if they had brain MRIs completed with sufficient sequences for analysis. Brain MRIs were computationally analyzed using diffusion tensor imaging analysis along the perivascular space technique to quantify the glymphatic function in patients with IIH. Glymphatic clearance, the primary outcome, was then correlated with the degree of TSS on MR venography using 2 different scoring systems, the 'Farb score' and 'Carvalho score.' RESULTS: Overall, 81 patients with IIH (70 [86%] female, mean age 29.8 years [SD: 8.2 years], mean BMI 41 [SD: 8.4]) and 10 normal controls were identified with sufficient imaging. Based on the Carvalho TSS score, IIH patients without TSS had significantly lower glymphatic clearance than healthy controls (mean ALPS index: 1.196 [SD: 0.05] vs 1.238 [SD: 0.04], respectively; p = 0.018). Furthermore, IIH patients with TSS had significantly lower glymphatic outflow than healthy controls (1.129 [SD: 0.07] vs 1.238 [SD: 0.04], respectively; p < 0.0001) and IIH patients without TSS (1.129 [SD: 0.07] vs 1.196 [SD: 0.05], respectively; p < 0.0001). In addition, there was a significant association between increasing extent of TSS and declining glymphatic clearance (p < 0.0001, R = 0.62). Finally, IIH patients with severe TSS had significantly lower glymphatic flow than IIH patients with mild stenosis (1.121 [SD: 0.07] vs 1.178 [SD: 0.05], respectively; p < 0.0001). These findings were similarly recapitulated using the Farb TSS scoring system. DISCUSSION: These preliminary findings suggest that the extent of TSS is associated with the degree of glymphatic clearance in IIH, providing novel insights into IIH pathophysiology. Further research is required to clarify the possible causal relationship between TSS and impaired glymphatic clearance in IIH.


Subject(s)
Glymphatic System , Pseudotumor Cerebri , Transverse Sinuses , Humans , Female , Male , Glymphatic System/diagnostic imaging , Glymphatic System/physiopathology , Adult , Pseudotumor Cerebri/physiopathology , Pseudotumor Cerebri/diagnostic imaging , Retrospective Studies , Constriction, Pathologic/diagnostic imaging , Constriction, Pathologic/physiopathology , Transverse Sinuses/diagnostic imaging , Young Adult , Middle Aged , Magnetic Resonance Imaging , Diffusion Tensor Imaging
15.
Neurology ; 102(12): e209483, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38833653

ABSTRACT

BACKGROUND AND OBJECTIVES: Although the potential role of enlarged perivascular spaces (EPVSs) in Parkinson disease (PD) is increasingly recognized, whether EPVSs located in different anatomical regions exert differential effects on clinical manifestation remains uncertain. We investigated the regional EPVS burden and its association with cognition and neuropsychiatric symptoms (NPSs) in newly diagnosed PD population. METHODS: In this retrospective, cross-sectional study, EPVS in the temporal lobe (T-EPVS), centrum semiovale (CS-EPVS), and basal ganglia (BG-EPVS) were visually rated in drug-naive patients with PD who underwent magnetic resonance imaging, dopamine transporter (DAT) scans, neuropsychological assessments, and Neuropsychiatric Inventory Questionnaire at baseline. Cognitive performance, NPS burden, vascular risk factors, small vessel disease (SVD) imaging markers, and DAT availability were compared across groups dichotomized by their regional EPVS burden (cutoff for high-degree vs low-degree: >10 for T-EPVS/BG-EPVS and >20 for CS-EPVS). RESULTS: A total of 480 patients with PD (123 without cognitive impairment, 291 with mild cognitive impairment, and 66 with dementia) were included. The proportion of high-degree T-EPVS (p for trend <0.001) and BG-EPVS (p for trend = 0.001) exhibited an increasing trend across the cognitive spectrum, corresponding to worsening cognition. Compared with the low-degree group, the high-degree BG-EPVS group showed higher SVD burden (moderate-to-severe white matter hyperintensity [14.8% vs 40.5%, p < 0.001], lacune [10.3% vs 30.7%, p < 0.001], and cerebral microbleeds [8.1% vs 22.2%, p < 0.001]), greater atrophy in cortical gray matter (40.73% ± 1.09% vs 39.96% ± 1.20% of intracranial volume, p < 0.001), and lower cognitive performance (in language [-0.22 ± 1.18 vs -0.53 ± 1.29, p = 0.013], and visual memory domains [-0.24 ± 0.97 vs -0.61 ± 0.96, p = 0.009]). The high-degree T-EPVS group presented with greater NPS burden in decreased motivation (0.61 ± 1.78 vs 1.35 ± 2.36, p = 0.007), affective dysregulation (0.88 ± 2.13 vs 2.36 ± 3.53, p < 0.001), and impulse dyscontrol (0.43 ± 1.67 vs 1.74 ± 4.29, p < 0.001), compared with the low-degree T-EPVS group. Meanwhile, the burden of CS-EPVS did not reveal any differences in cognition or NPS. DISCUSSION: BG-EPVS and T-EPVS seem to exert differential effects on cognition and NPS in patients with PD. Investigating the EPVS profile in distinct anatomical regions may be useful in disentangling the heterogeneity within PD.


Subject(s)
Glymphatic System , Parkinson Disease , Humans , Parkinson Disease/diagnostic imaging , Parkinson Disease/complications , Parkinson Disease/pathology , Parkinson Disease/psychology , Male , Female , Aged , Cross-Sectional Studies , Middle Aged , Retrospective Studies , Glymphatic System/diagnostic imaging , Glymphatic System/pathology , Cognitive Dysfunction/diagnostic imaging , Cognitive Dysfunction/etiology , Cognitive Dysfunction/pathology , Magnetic Resonance Imaging , Cognition/physiology , Neuropsychological Tests , Dopamine Plasma Membrane Transport Proteins/metabolism
17.
Radiology ; 311(3): e232274, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38888481

ABSTRACT

Background The role of perivascular space (PVS) dysfunction in obstructive sleep apnea (OSA) requires further study. Purpose To compare MRI indexes of PVS across patients with differing severities of OSA and relate them with disease characteristics and treatment. Materials and Methods This single-center prospective study included healthy controls (HCs) and patients with complaints of snoring who underwent MRI and cognitive evaluation between June 2021 and December 2022. Participants with complaints of snoring were classified into four groups (snoring, mild OSA, moderate OSA, and severe OSA). PVS networks were assessed at MRI using PVS volume fraction, extracellular free water (FW), and diffusion tensor imaging analysis along the PVS (DTI-ALPS). One-way analysis of variance and Pearson correlation were used for analysis. Alterations in PVS indexes and cognitive performance after treatment were assessed in 15 participants with moderate OSA. Results A total of 105 participants (mean age, 33.4 years ± 8.9 [SD]; 80 males) and 50 HCs (mean age, 37.0 years ± 8.6; 33 males) were included. Higher mean PVS volume fraction was observed in participants with severe OSA (n = 23) than in patients with mild OSA (n = 36) (0.11 vs 0.10; P = .03). Participants with severe OSA exhibited higher mean FW index (0.11) than both HCs (0.10; P < .001) and patients with mild OSA (0.10; P = .003). All patient groups had lower DTI-ALPS than HCs (range, 1.5-1.9 vs 2.1; all P < .001). DTI-ALPS correlated with cognitive performance on the Stroop Color and Word Test (r range, -0.23 to -0.24; P value range, .003-.005). After treatment, PVS indexes changed (P value range, <.001 to .01) and cognitive performance improved (P value range, <.001 to .03). Conclusion Differences in PVS indexes were observed among participants with differing severities of OSA and HCs. Indexes correlated with measures of cognitive function, and changes in indexes and improvement in cognitive performance were observed after treatment in participants with moderate OSA. © RSNA, 2024 Supplemental material is available for this article. See also the editorial by Port in this issue.


Subject(s)
Cognitive Dysfunction , Magnetic Resonance Imaging , Sleep Apnea, Obstructive , Humans , Sleep Apnea, Obstructive/diagnostic imaging , Sleep Apnea, Obstructive/complications , Male , Female , Prospective Studies , Adult , Magnetic Resonance Imaging/methods , Cognitive Dysfunction/diagnostic imaging , Cognitive Dysfunction/etiology , Glymphatic System/diagnostic imaging , Diffusion Tensor Imaging/methods , Middle Aged
18.
Brain ; 147(6): 2214-2229, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38802114

ABSTRACT

Mild traumatic brain injury (mTBI) has emerged as a potential risk factor for the development of neurodegenerative conditions such as Alzheimer's disease and chronic traumatic encephalopathy. Blast mTBI, caused by exposure to a pressure wave from an explosion, is predominantly experienced by military personnel and has increased in prevalence and severity in recent decades. Yet the underlying pathology of blast mTBI is largely unknown. We examined the expression and localization of AQP4 in human post-mortem frontal cortex and observed distinct laminar differences in AQP4 expression following blast exposure. We also observed similar laminar changes in AQP4 expression and localization and delayed impairment of glymphatic function that emerged 28 days following blast injury in a mouse model of repetitive blast mTBI. In a cohort of veterans with blast mTBI, we observed that blast exposure was associated with an increased burden of frontal cortical MRI-visible perivascular spaces, a putative neuroimaging marker of glymphatic perivascular dysfunction. These findings suggest that changes in AQP4 and delayed glymphatic impairment following blast injury may render the post-traumatic brain vulnerable to post-concussive symptoms and chronic neurodegeneration.


Subject(s)
Aquaporin 4 , Blast Injuries , Glymphatic System , Adult , Aged , Animals , Female , Humans , Male , Mice , Middle Aged , Aquaporin 4/metabolism , Blast Injuries/complications , Blast Injuries/pathology , Blast Injuries/metabolism , Brain Concussion/metabolism , Brain Concussion/complications , Brain Concussion/pathology , Brain Concussion/physiopathology , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/pathology , Frontal Lobe/metabolism , Frontal Lobe/pathology , Frontal Lobe/diagnostic imaging , Glymphatic System/metabolism , Glymphatic System/pathology , Magnetic Resonance Imaging , Mice, Inbred C57BL , Veterans
19.
J Neurol Sci ; 461: 123056, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38772058

ABSTRACT

FMR1 premutation carriers (55-200 CGG repeats) are at risk of developing fragile X-associated tremor/ataxia syndrome (FXTAS), a neurodegenerative disorder associated with motor and cognitive impairment. Bilateral hyperintensities of the middle cerebellar peduncles (MCP sign) are the major radiological hallmarks of FXTAS. In the general population, enlarged perivascular spaces (PVS) are biomarkers of small vessel disease and glymphatic dysfunction and are associated with cognitive decline. Our aim was to determine if premutation carriers show higher ratings of PVS than controls and whether enlarged PVS are associated with motor and cognitive impairment, MRI features of neurodegeneration, cerebrovascular risk factors and CGG repeat length. We evaluated 655 MRIs (1-10 visits/participant) from 229 carriers (164 with FXTAS and 65 without FXTAS) and 133 controls. PVS in the basal ganglia (BG-EPVS), centrum semiovale, and midbrain were evaluated with a semiquantitative scale. Mixed-effects models were used for statistical analysis adjusting for age. In carriers with FXTAS, we revealed that (1) BG-PVS ratings were higher than those of controls and carriers without FXTAS; (2) BG-PVS severity was associated with brain atrophy, white matter hyperintensities, enlarged ventricles, FXTAS stage and abnormal gait; (3) age-related increase in BG-PVS was associated with cognitive dysfunction; and (4) PVS ratings of all three regions showed robust associations with CGG repeat length and were higher in carriers with the MCP sign than carriers without the sign. This study demonstrates clinical relevance of PVS in FXTAS especially in the basal ganglia region and suggests microangiopathy and dysfunctional cerebrospinal fluid circulation in FXTAS physiopathology.


Subject(s)
Ataxia , Fragile X Mental Retardation Protein , Fragile X Syndrome , Glymphatic System , Magnetic Resonance Imaging , Tremor , Humans , Male , Fragile X Syndrome/genetics , Fragile X Syndrome/diagnostic imaging , Fragile X Syndrome/pathology , Middle Aged , Aged , Fragile X Mental Retardation Protein/genetics , Tremor/genetics , Tremor/diagnostic imaging , Tremor/pathology , Ataxia/genetics , Ataxia/diagnostic imaging , Ataxia/pathology , Glymphatic System/diagnostic imaging , Glymphatic System/pathology , Risk Factors , Heterozygote , Cerebrovascular Disorders/genetics , Cerebrovascular Disorders/diagnostic imaging , Cerebrovascular Disorders/pathology , Cognitive Dysfunction/genetics , Cognitive Dysfunction/diagnostic imaging , Cognitive Dysfunction/pathology , Cognitive Dysfunction/etiology , Brain/diagnostic imaging , Brain/pathology
20.
Cell Mol Life Sci ; 81(1): 239, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38801464

ABSTRACT

The brain's network of perivascular channels for clearance of excess fluids and waste plays a critical role in the pathogenesis of several neurodegenerative diseases including cerebral amyloid angiopathy (CAA). CAA is the main cause of hemorrhagic stroke in the elderly, the most common vascular comorbidity in Alzheimer's disease and also implicated in adverse events related to anti-amyloid immunotherapy. Remarkably, the mechanisms governing perivascular clearance of soluble amyloid ß-a key culprit in CAA-from the brain to draining lymphatics and systemic circulation remains poorly understood. This knowledge gap is critically important to bridge for understanding the pathophysiology of CAA and accelerate development of targeted therapeutics. The authors of this review recently converged their diverse expertise in the field of perivascular physiology to specifically address this problem within the framework of a Leducq Foundation Transatlantic Network of Excellence on Brain Clearance. This review discusses the overarching goal of the consortium and explores the evidence supporting or refuting the role of impaired perivascular clearance in the pathophysiology of CAA with a focus on translating observations from rodents to humans. We also discuss the anatomical features of perivascular channels as well as the biophysical characteristics of fluid and solute transport.


Subject(s)
Amyloid beta-Peptides , Brain , Cerebral Amyloid Angiopathy , Humans , Brain/metabolism , Brain/pathology , Cerebral Amyloid Angiopathy/metabolism , Cerebral Amyloid Angiopathy/pathology , Animals , Amyloid beta-Peptides/metabolism , Glymphatic System/metabolism , Glymphatic System/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...