Pub Date : 2025-01-01Epub Date: 2024-10-25DOI: 10.1177/0271678X241293537
Julia K Gundersen, Fernando Gonzalez-Ortiz, Thomas Karikari, Bjørn-Eivind Kirsebom, Katrin Mertes, Henrik Zetterberg, Hlin Kvartsberg, Ole Morten Rønning, Berglind Gísladóttir, Kaj Blennow, Tormod Fladby
Early imaging-based detection of acute ischemic stroke (AIS) has improved in the era of reperfusion therapy. Despite of this, prognosis of outcome after AIS remains a challenge. Therefore, parameters that support clinical decision making are sought. Blood-based biomarkers have the potential to provide valuable information in addition to the established prognostic factors. Neuronal biomarkers of acute or degenerative neuronal injury have shown to be reliably detected in plasma. These biomarkers are well-established in neurodegenerative pathology, such as Alzheimer's disease. In this study, we explored the association between stroke diameter and plasma biomarkers for neuronal injury and tau pathophysiology (brain-derived tau [BD-tau], phosphorylated-tau-217 [p-tau21] and neurofilament light [NfL]) in patients (n = 193) admitted to the acute ward, Akershus University Hospital. All patients received a final diagnosis of AIS, transient ischemic attack or stroke mimics. Blood samples were obtained the day after admission. We find that levels of BD-tau (p = .004) and NfL (p = .011) were higher after AIS than in patients with stroke mimics. The cortical stroke diameter correlated with BD-tau (tau-b = 0.64, p < .001) and p-tau217 (tau-b = 0.36, p = .003). Linear regression confirmed BD-tau to be the strongest variable associated with stroke diameter, pointing to the potential clinical value of plasma BD-tau in outcome prediction after AIS.
{"title":"Neuronal plasma biomarkers in acute ischemic stroke.","authors":"Julia K Gundersen, Fernando Gonzalez-Ortiz, Thomas Karikari, Bjørn-Eivind Kirsebom, Katrin Mertes, Henrik Zetterberg, Hlin Kvartsberg, Ole Morten Rønning, Berglind Gísladóttir, Kaj Blennow, Tormod Fladby","doi":"10.1177/0271678X241293537","DOIUrl":"10.1177/0271678X241293537","url":null,"abstract":"<p><p>Early imaging-based detection of acute ischemic stroke (AIS) has improved in the era of reperfusion therapy. Despite of this, prognosis of outcome after AIS remains a challenge. Therefore, parameters that support clinical decision making are sought. Blood-based biomarkers have the potential to provide valuable information in addition to the established prognostic factors. Neuronal biomarkers of acute or degenerative neuronal injury have shown to be reliably detected in plasma. These biomarkers are well-established in neurodegenerative pathology, such as Alzheimer's disease. In this study, we explored the association between stroke diameter and plasma biomarkers for neuronal injury and tau pathophysiology (brain-derived tau [BD-tau], phosphorylated-tau-217 [p-tau21] and neurofilament light [NfL]) in patients (n = 193) admitted to the acute ward, Akershus University Hospital. All patients received a final diagnosis of AIS, transient ischemic attack or stroke mimics. Blood samples were obtained the day after admission. We find that levels of BD-tau (p = .004) and NfL (p = .011) were higher after AIS than in patients with stroke mimics. The cortical stroke diameter correlated with BD-tau (tau-b = 0.64, p < .001) and p-tau217 (tau-b = 0.36, p = .003). Linear regression confirmed BD-tau to be the strongest variable associated with stroke diameter, pointing to the potential clinical value of plasma BD-tau in outcome prediction after AIS.</p>","PeriodicalId":15325,"journal":{"name":"Journal of Cerebral Blood Flow and Metabolism","volume":" ","pages":"77-84"},"PeriodicalIF":4.9,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11563507/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142501176","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
The brain's function of clearance and transport is closely related to the prognosis of acute ischemic stroke (AIS). In this study, we proposed a novel method, clearance rate of contrast extravasation (CROCE), to measure brain clearance and transport function in AIS patients undergoing endovascular therapy (EVT), and examined its association with cerebral edema and functional outcome. We conducted a pooled analysis of AIS patients of anterior circulation large vessel occlusion who underwent EVT in two academic hospitals. Patients who experienced contrast extravasation but not intracerebral hemorrhage following EVT were included. CROCE was defined as the mass of contrast agent cleared per hour on non-contrast CT (NCCT). Among the 215 patients finally included, we found that high CROCE was significantly associated with 90-day favorable functional outcome, and the association retained after adjustment for potential confounders. Different correlation analysis demonstrated a significant correlation between CROCE, cerebral edema, and functional outcome. Further mediation analysis revealed that cerebral edema mediated the effect of CROCE on functional outcome. These results revealed that CROCE may be a promising indicator of brain clearance function for patients who received EVT and had contrast extravasation.
{"title":"Clearance rate of contrast extravasation after endovascular therapy is associated with functional outcome and mediated by cerebral edema.","authors":"Jiaqi Luo, Xiaolin Zhao, Mengxuan Xiao, Lihua Wei, Zhiliang Zhu, Bingbing Li, Zhong Ji, Yongming Wu, Zhenzhou Lin, Suyue Pan, Kaibin Huang","doi":"10.1177/0271678X241275763","DOIUrl":"10.1177/0271678X241275763","url":null,"abstract":"<p><p>The brain's function of clearance and transport is closely related to the prognosis of acute ischemic stroke (AIS). In this study, we proposed a novel method, clearance rate of contrast extravasation (CROCE), to measure brain clearance and transport function in AIS patients undergoing endovascular therapy (EVT), and examined its association with cerebral edema and functional outcome. We conducted a pooled analysis of AIS patients of anterior circulation large vessel occlusion who underwent EVT in two academic hospitals. Patients who experienced contrast extravasation but not intracerebral hemorrhage following EVT were included. CROCE was defined as the mass of contrast agent cleared per hour on non-contrast CT (NCCT). Among the 215 patients finally included, we found that high CROCE was significantly associated with 90-day favorable functional outcome, and the association retained after adjustment for potential confounders. Different correlation analysis demonstrated a significant correlation between CROCE, cerebral edema, and functional outcome. Further mediation analysis revealed that cerebral edema mediated the effect of CROCE on functional outcome. These results revealed that CROCE may be a promising indicator of brain clearance function for patients who received EVT and had contrast extravasation.</p>","PeriodicalId":15325,"journal":{"name":"Journal of Cerebral Blood Flow and Metabolism","volume":" ","pages":"66-76"},"PeriodicalIF":4.9,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11572124/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142004367","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-01Epub Date: 2024-10-18DOI: 10.1177/0271678X241290018
Richard J Maddock
1H-MRS investigators studying brain metabolite concentrations often attribute biological significance to correlations between calculated metabolite values within the same voxel. A recent report in this journal provides a valuable perspective on how statistical non-independence of such values can undermine biological interpretations of their correlations. However, careful examination of this issue suggests their critical analysis does not go far enough. Hong et al. claim that appropriate water normalization, unlike creatine normalization, eliminates the problem of spurious correlation. Both logical and empirical considerations show this is not the case. Correlations between water-normalized metabolite values are also prone to substantial spurious correlations.
{"title":"Statistical non-independence of brain metabolite concentrations whether normalized to creatine or water.","authors":"Richard J Maddock","doi":"10.1177/0271678X241290018","DOIUrl":"10.1177/0271678X241290018","url":null,"abstract":"<p><p>1H-MRS investigators studying brain metabolite concentrations often attribute biological significance to correlations between calculated metabolite values within the same voxel. A recent report in this journal provides a valuable perspective on how statistical non-independence of such values can undermine biological interpretations of their correlations. However, careful examination of this issue suggests their critical analysis does not go far enough. Hong et al. claim that appropriate water normalization, unlike creatine normalization, eliminates the problem of spurious correlation. Both logical and empirical considerations show this is not the case. Correlations between water-normalized metabolite values are also prone to substantial spurious correlations.</p>","PeriodicalId":15325,"journal":{"name":"Journal of Cerebral Blood Flow and Metabolism","volume":" ","pages":"196-198"},"PeriodicalIF":4.9,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11563549/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142466526","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-01Epub Date: 2024-07-28DOI: 10.1177/0271678X241264401
Yangyang Liu, Xiang Li, Chang Cao, Haojie Ding, Xuan Shi, Juyi Zhang, Haiying Li
Blood-brain barrier (BBB) damage significantly affects the prognosis of ischemic stroke patients. This project employed multi-omics analysis to identify key factors regulating BBB disruption during cerebral ischemia-reperfusion. An integrated analysis of three transcriptome sequencing datasets from mouse middle cerebral artery occlusion/reperfusion (MCAO/R) models identified eight downregulated genes in endothelial cells. Additionally, transcriptome analysis of BBB (cortex) and non-BBB (lung) endothelium of E13.5 mice revealed 2,102 upregulated genes potentially associated with BBB integrity. The eight downregulated genes were intersected with the 2,102 BBB-related genes and mapped using single-cell RNA sequencing data, revealing that solute carrier family 22 member 8 (Slc22a8) is specifically expressed in endothelial cells and pericytes and significantly decreases after MCAO/R. This finding was validated in the mouse MCAO/R model at both protein and mRNA levels in this study. External overexpression of Slc22a8 using a lentivirus carrying Tie2 improved Slc22a8 and tight junction protein levels and reduced BBB leakage after MCAO/R, accompanied by Wnt/β-catenin signaling activation. In conclusion, this study suggested that MCAO/R-induced downregulation of Slc22a8 expression may be a crucial mechanism underlying BBB disruption. Interventions that promote Slc22a8 expression or enhance its function hold promise for improving the prognosis of patients with cerebral ischemia.
{"title":"Critical role of Slc22a8 in maintaining blood-brain barrier integrity after experimental cerebral ischemia-reperfusion.","authors":"Yangyang Liu, Xiang Li, Chang Cao, Haojie Ding, Xuan Shi, Juyi Zhang, Haiying Li","doi":"10.1177/0271678X241264401","DOIUrl":"10.1177/0271678X241264401","url":null,"abstract":"<p><p>Blood-brain barrier (BBB) damage significantly affects the prognosis of ischemic stroke patients. This project employed multi-omics analysis to identify key factors regulating BBB disruption during cerebral ischemia-reperfusion. An integrated analysis of three transcriptome sequencing datasets from mouse middle cerebral artery occlusion/reperfusion (MCAO/R) models identified eight downregulated genes in endothelial cells. Additionally, transcriptome analysis of BBB (cortex) and non-BBB (lung) endothelium of E13.5 mice revealed 2,102 upregulated genes potentially associated with BBB integrity. The eight downregulated genes were intersected with the 2,102 BBB-related genes and mapped using single-cell RNA sequencing data, revealing that solute carrier family 22 member 8 (Slc22a8) is specifically expressed in endothelial cells and pericytes and significantly decreases after MCAO/R. This finding was validated in the mouse MCAO/R model at both protein and mRNA levels in this study. External overexpression of Slc22a8 using a lentivirus carrying Tie2 improved Slc22a8 and tight junction protein levels and reduced BBB leakage after MCAO/R, accompanied by Wnt/β-catenin signaling activation. In conclusion, this study suggested that MCAO/R-induced downregulation of Slc22a8 expression may be a crucial mechanism underlying BBB disruption. Interventions that promote Slc22a8 expression or enhance its function hold promise for improving the prognosis of patients with cerebral ischemia.</p>","PeriodicalId":15325,"journal":{"name":"Journal of Cerebral Blood Flow and Metabolism","volume":" ","pages":"85-101"},"PeriodicalIF":4.9,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11572098/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141788197","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-01Epub Date: 2024-08-07DOI: 10.1177/0271678X241270241
Helena Justić, Anja Barić, Martina Ratko, Iva Šimunić, Marin Radmilović, Marta Pongrac, Siniša Škokić, Marina Dobrivojević Radmilović
The activation of the bradykinin type 2 receptor is intricately involved in acute post-ischemic inflammatory responses. However, its precise role in different stages of ischemic injury, especially in the chronic phase, remains unclear. Following simultaneous cerebral and retinal ischemia, bradykinin type 2 receptor knockout mice and their controls were longitudinally monitored for 35 days via magnetic resonance imaging, fundus photography, fluorescein angiography, behavioral assessments, vascular permeability measurements, and immunohistochemistry, as well as glycemic status assessments. Without impacting the lesion size, bradykinin type 2 receptor deficiency reduced acute cerebral vascular permeability preventing the loss of pericytes and tight junctions. In the chronic phase of ischemia, however, it resulted in increased astrogliosis and cortical neuronal loss, as well as higher functional deficits. The retinal findings demonstrated a similar pattern. Bradykinin type 2 receptor deficiency delayed, but exacerbated the development of retinal necrosis, increased subacute vascular permeability, and promoted retinal ganglion cell loss in the chronic phase of ischemia. This investigation sheds light on the temporal dynamic of bradykinin type 2 receptor effects in ischemia, pointing to a therapeutic potential in the subacute and chronic phases of ischemic injury.
{"title":"The temporal dynamic of bradykinin type 2 receptor effects reveals its neuroprotective role in the chronic phase of cerebral and retinal ischemic injury.","authors":"Helena Justić, Anja Barić, Martina Ratko, Iva Šimunić, Marin Radmilović, Marta Pongrac, Siniša Škokić, Marina Dobrivojević Radmilović","doi":"10.1177/0271678X241270241","DOIUrl":"10.1177/0271678X241270241","url":null,"abstract":"<p><p>The activation of the bradykinin type 2 receptor is intricately involved in acute post-ischemic inflammatory responses. However, its precise role in different stages of ischemic injury, especially in the chronic phase, remains unclear. Following simultaneous cerebral and retinal ischemia, bradykinin type 2 receptor knockout mice and their controls were longitudinally monitored for 35 days via magnetic resonance imaging, fundus photography, fluorescein angiography, behavioral assessments, vascular permeability measurements, and immunohistochemistry, as well as glycemic status assessments. Without impacting the lesion size, bradykinin type 2 receptor deficiency reduced acute cerebral vascular permeability preventing the loss of pericytes and tight junctions. In the chronic phase of ischemia, however, it resulted in increased astrogliosis and cortical neuronal loss, as well as higher functional deficits. The retinal findings demonstrated a similar pattern. Bradykinin type 2 receptor deficiency delayed, but exacerbated the development of retinal necrosis, increased subacute vascular permeability, and promoted retinal ganglion cell loss in the chronic phase of ischemia. This investigation sheds light on the temporal dynamic of bradykinin type 2 receptor effects in ischemia, pointing to a therapeutic potential in the subacute and chronic phases of ischemic injury.</p>","PeriodicalId":15325,"journal":{"name":"Journal of Cerebral Blood Flow and Metabolism","volume":" ","pages":"153-170"},"PeriodicalIF":4.9,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11572167/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141901947","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-01Epub Date: 2024-08-07DOI: 10.1177/0271678X241270531
Harm Jan van der Horn, Andrew B Dodd, Tracey V Wick, Cidney Robertson-Benta, Jessica R McQuaid, Erik B Erhardt, Samuel D Miller, Divyasree Sasi Kumar, Upasana Nathaniel, Josef M Ling, Sephira G Ryman, Andrei A Vakhtin, Robert E Sapien, John P Phillips, Richard A Campbell, Andrew R Mayer
Cerebrovascular dysfunction following mild traumatic brain injury (mTBI) is understudied relative to other microstructural injuries, especially during neurodevelopment. The blood-oxygen level dependent response was used to investigate cerebrovascular reactivity (CVR) in response to hypercapnia following pediatric mTBI (pmTBI; ages 8-18 years), as well as pseudocontinuous arterial spin labeling to measure cerebral blood flow (CBF). Data were collected ∼1-week (N = 107) and 4 months (N = 73) post-injury. Sex- and age-matched healthy controls (HC) underwent identical examinations at comparable time points (N = 110 and N = 91). Subtle clinical and cognitive deficits existed at ∼1 week that resolved for some, but not all domains at 4 months post-injury. At both visits, pmTBI showed an increased maximal fit between end-tidal CO2 regressor and the cerebrovascular response across multiple regions (primarily fronto-temporal), as well as increased latency to maximal fit in independent regions (primarily posterior). Hypoperfusion was also noted within the bilateral cerebellum. A biphasic relationship existed between CVR amplitude and age (i.e., positive until 14.5 years, negative thereafter) in both gray and white matter, but these neurodevelopment effects did not moderate injury effects. CVR metrics were not associated with post-concussive symptoms or cognitive deficits. In conclusion, cerebrovascular dysfunction may persist for up to four months following pmTBI.
{"title":"Alterations of cerebrovascular reactivity following pediatric mild traumatic brain injury are independent of neurodevelopmental changes.","authors":"Harm Jan van der Horn, Andrew B Dodd, Tracey V Wick, Cidney Robertson-Benta, Jessica R McQuaid, Erik B Erhardt, Samuel D Miller, Divyasree Sasi Kumar, Upasana Nathaniel, Josef M Ling, Sephira G Ryman, Andrei A Vakhtin, Robert E Sapien, John P Phillips, Richard A Campbell, Andrew R Mayer","doi":"10.1177/0271678X241270531","DOIUrl":"10.1177/0271678X241270531","url":null,"abstract":"<p><p>Cerebrovascular dysfunction following mild traumatic brain injury (mTBI) is understudied relative to other microstructural injuries, especially during neurodevelopment. The blood-oxygen level dependent response was used to investigate cerebrovascular reactivity (CVR) in response to hypercapnia following pediatric mTBI (pmTBI; ages 8-18 years), as well as pseudocontinuous arterial spin labeling to measure cerebral blood flow (CBF). Data were collected ∼1-week (N = 107) and 4 months (N = 73) post-injury. Sex- and age-matched healthy controls (HC) underwent identical examinations at comparable time points (N = 110 and N = 91). Subtle clinical and cognitive deficits existed at ∼1 week that resolved for some, but not all domains at 4 months post-injury. At both visits, pmTBI showed an increased maximal fit between end-tidal CO<sub>2</sub> regressor and the cerebrovascular response across multiple regions (primarily fronto-temporal), as well as increased latency to maximal fit in independent regions (primarily posterior). Hypoperfusion was also noted within the bilateral cerebellum. A biphasic relationship existed between CVR amplitude and age (i.e., positive until 14.5 years, negative thereafter) in both gray and white matter, but these neurodevelopment effects did not moderate injury effects. CVR metrics were not associated with post-concussive symptoms or cognitive deficits. In conclusion, cerebrovascular dysfunction may persist for up to four months following pmTBI.</p>","PeriodicalId":15325,"journal":{"name":"Journal of Cerebral Blood Flow and Metabolism","volume":" ","pages":"125-139"},"PeriodicalIF":4.9,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11572250/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141901926","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-01Epub Date: 2024-08-07DOI: 10.1177/0271678X241270528
Laura Schönberg, Abdalla Z Mohamed, Qiang Yu, Richard A Kwiatek, Peter Del Fante, Vince D Calhoun, Zack Y Shan
Neurological symptoms are central to Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS), yet its underlying neurophysiological mechanisms remain elusive. We examined a neglected aspect of task-based functional MRI, focusing on how blood oxygenation level-dependent (BOLD) signals alter during cognitive tasks in ME/CFS. This prospective observational study utilised MRI scans on ME/CFS participants and healthy controls (HCs) with sedentary lifestyles (ACTRN12622001095752). Participants completed two blocks of a Symbol Digit Modalities Test, with 30 trials per block split into two sets. The fMRI signal changes between blocks and sets were compared within and between groups. Thirty-four ME/CFS participants (38 years ± 10; 27 women) and 34 HCs (38 ± 10; 27 women), were evaluated. In the second task block, ME/CFS participants exhibited increased activation in the right postcentral gyrus, contrasting with decreased activation in multiple regions in HCs. These results were further confirmed by significantly higher bilateral dynamic changes (2nd vs 1st set) in the motor, sensory and cognitive cortex in ME/CFS compared to HCs and significant correlations between those changes in the left primary motor cortex with fatigue severities. BOLD adaptation, potentially improving energy economy, was absent in ME/CFS, which may provide an underlying neurophysiological process in ME/CFS.
{"title":"Absence of BOLD adaptation in chronic fatigue syndrome revealed by task functional MRI.","authors":"Laura Schönberg, Abdalla Z Mohamed, Qiang Yu, Richard A Kwiatek, Peter Del Fante, Vince D Calhoun, Zack Y Shan","doi":"10.1177/0271678X241270528","DOIUrl":"10.1177/0271678X241270528","url":null,"abstract":"<p><p>Neurological symptoms are central to Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS), yet its underlying neurophysiological mechanisms remain elusive. We examined a neglected aspect of task-based functional MRI, focusing on how blood oxygenation level-dependent (BOLD) signals alter during cognitive tasks in ME/CFS. This prospective observational study utilised MRI scans on ME/CFS participants and healthy controls (HCs) with sedentary lifestyles (ACTRN12622001095752). Participants completed two blocks of a Symbol Digit Modalities Test, with 30 trials per block split into two sets. The fMRI signal changes between blocks and sets were compared within and between groups. Thirty-four ME/CFS participants (38 years ± 10; 27 women) and 34 HCs (38 ± 10; 27 women), were evaluated. In the second task block, ME/CFS participants exhibited increased activation in the right postcentral gyrus, contrasting with decreased activation in multiple regions in HCs. These results were further confirmed by significantly higher bilateral dynamic changes (2nd vs 1st set) in the motor, sensory and cognitive cortex in ME/CFS compared to HCs and significant correlations between those changes in the left primary motor cortex with fatigue severities. BOLD adaptation, potentially improving energy economy, was absent in ME/CFS, which may provide an underlying neurophysiological process in ME/CFS.</p>","PeriodicalId":15325,"journal":{"name":"Journal of Cerebral Blood Flow and Metabolism","volume":" ","pages":"115-124"},"PeriodicalIF":4.9,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141901925","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-24DOI: 10.1177/0271678X241309783
Eleonora Cavallari, Elena Lorenzi, Enza Di Gregorio, Giuseppe Ferrauto, Silvio Aime, Giorgio Vallortigara, Angelo Bifone
This study presents the first in vivo measurement of transcytolemmal water exchange in the brain using a novel Magnetic Resonance technique. We extend previous applications of Chemical Exchange Saturation Transfer (CEST) to examine water exchange across cellular membranes in late-stage chicken embryo brains. The immature blood-brain barrier at this stage allows Gadolinium-Based Contrast Agents (GBCAs) to penetrate the brain's interstitial space, sensitizing the CEST effect to water exchange between intra- and extracellular environments. Exchange rates were measured in the awake brain and under different anaesthetic regimens, including isoflurane and ketamine/xylazine. Results show that brain water exchange is dominated by activity-dependent mechanisms, with anaesthesia reducing exchange rates by over an order of magnitude. These findings suggest that anaesthetics may impact neuronal and glial function by interfering with active transport mechanisms, potentially altering brain water homeostasis. This study highlights the utility of CEST MRI for studying dynamic biological processes in vivo.
本研究首次利用新型磁共振技术对大脑中的跨细胞膜水交换进行了活体测量。我们扩展了以往化学交换饱和转移(CEST)技术的应用,研究了晚期鸡胚大脑中跨细胞膜的水交换。这个阶段的血脑屏障尚未成熟,钆基造影剂(GBCA)可以穿透大脑间隙,使 CEST 效应对细胞内和细胞外环境之间的水交换敏感。在清醒的大脑和不同的麻醉方案(包括异氟醚和氯胺酮/恶嗪)下测量了交换率。结果表明,脑水交换主要受活动依赖机制的影响,麻醉会使交换率降低一个数量级以上。这些发现表明,麻醉剂可能会通过干扰活性转运机制来影响神经元和神经胶质细胞的功能,从而可能改变脑水平衡。这项研究凸显了 CEST MRI 在研究体内动态生物过程方面的实用性。
{"title":"In vivo assessment of the influence of general anaesthetics on transmembrane water cycling in the brain.","authors":"Eleonora Cavallari, Elena Lorenzi, Enza Di Gregorio, Giuseppe Ferrauto, Silvio Aime, Giorgio Vallortigara, Angelo Bifone","doi":"10.1177/0271678X241309783","DOIUrl":"10.1177/0271678X241309783","url":null,"abstract":"<p><p>This study presents the first in vivo measurement of transcytolemmal water exchange in the brain using a novel Magnetic Resonance technique. We extend previous applications of Chemical Exchange Saturation Transfer (CEST) to examine water exchange across cellular membranes in late-stage chicken embryo brains. The immature blood-brain barrier at this stage allows Gadolinium-Based Contrast Agents (GBCAs) to penetrate the brain's interstitial space, sensitizing the CEST effect to water exchange between intra- and extracellular environments. Exchange rates were measured in the awake brain and under different anaesthetic regimens, including isoflurane and ketamine/xylazine. Results show that brain water exchange is dominated by activity-dependent mechanisms, with anaesthesia reducing exchange rates by over an order of magnitude. These findings suggest that anaesthetics may impact neuronal and glial function by interfering with active transport mechanisms, potentially altering brain water homeostasis. This study highlights the utility of CEST MRI for studying dynamic biological processes in vivo.</p>","PeriodicalId":15325,"journal":{"name":"Journal of Cerebral Blood Flow and Metabolism","volume":" ","pages":"271678X241309783"},"PeriodicalIF":4.9,"publicationDate":"2024-12-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11669147/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142885358","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Chronic cerebral hypoperfusion (CCH) is a crucial mechanism causing vascular cognitive impairment (VCI). Choline is metabolized by gut microbiota into trimethylamine N-oxide (TMAO), a risk factor of cardiovascular diseases and cognitive impairment. However, the impact of choline-TMAO pathway on CCH-induced VCI is elusive. We performed a cross-sectional clinical study to investigate the relationship between the choline-TMAO pathway and cognitive outcome and used a bilateral common carotid artery occlusion rat model to explore the effect of a choline-rich diet on cognition and underlying mechanisms. Plasma choline and TMAO levels were negatively correlated with cognitive scores in CCH patients. A choline-rich diet exacerbated CCH-induced cognitive impairment by encouraging the proliferation of choline-metabolizing bacteria in the gut and subsequent generation of TMAO. The choline-TMAO pathway, mediated by gut microbiota, exacerbates cognitive impairment induced by CCH. Targeted dietary choline regulation based on gut microbiota modulation may ameliorate long-term cognitive impairment.
{"title":"Gut microbiota-mediated choline metabolism exacerbates cognitive impairment induced by chronic cerebral hypoperfusion.","authors":"Xiao Li, Yueran Ren, Xuxuan Gao, Huidi Wang, Jiafeng Zhang, Jiahui Xie, Jingru Liang, Boxin Zhao, Hongwei Zhou, Jia Yin","doi":"10.1177/0271678X241309777","DOIUrl":"https://doi.org/10.1177/0271678X241309777","url":null,"abstract":"<p><p>Chronic cerebral hypoperfusion (CCH) is a crucial mechanism causing vascular cognitive impairment (VCI). Choline is metabolized by gut microbiota into trimethylamine N-oxide (TMAO), a risk factor of cardiovascular diseases and cognitive impairment. However, the impact of choline-TMAO pathway on CCH-induced VCI is elusive. We performed a cross-sectional clinical study to investigate the relationship between the choline-TMAO pathway and cognitive outcome and used a bilateral common carotid artery occlusion rat model to explore the effect of a choline-rich diet on cognition and underlying mechanisms. Plasma choline and TMAO levels were negatively correlated with cognitive scores in CCH patients. A choline-rich diet exacerbated CCH-induced cognitive impairment by encouraging the proliferation of choline-metabolizing bacteria in the gut and subsequent generation of TMAO. The choline-TMAO pathway, mediated by gut microbiota, exacerbates cognitive impairment induced by CCH. Targeted dietary choline regulation based on gut microbiota modulation may ameliorate long-term cognitive impairment.</p>","PeriodicalId":15325,"journal":{"name":"Journal of Cerebral Blood Flow and Metabolism","volume":" ","pages":"271678X241309777"},"PeriodicalIF":4.9,"publicationDate":"2024-12-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142884853","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-18DOI: 10.1177/0271678X241299985
Julie L Proctor, Su Xu, Sijia Guo, Boris Piskoun, Catriona Miller, Steven Roys, Rao P Gullapalli, Gary Fiskum
Aircraft cabins are routinely pressurized to the equivalent of 8000 ft altitude. Exposure of lab animals to aeromedical evacuation relevant hypobaria after traumatic brain injury worsens neurological outcomes, which is paradoxically exacerbated by hyperoxia. This study tested the hypothesis that exposure of rats to hypobaria following cortical impact reduces cerebral blood flow, increases neuroinflammation, and alters brain neurochemistry. Rats were exposed to simulated ground (normobaric) or air (hypobaric 8000 ft) transport, under normoxia or hyperoxia, 24 hr after trauma. Hypobaria exposure resulted in lower cerebral blood flow to the contralateral cortex and bilateral thalamus during flight and increased delayed cortical inflammation (ED1 immunoreactivity) at 14 days post injury. Impacted rats exposed to hypobaria had higher cortical creatine levels compared rats maintained at sea level. Exposure to the combination of hyperoxia and hypobaria resulted in the greatest reduction in cortical blood flow and total creatine during flight which persisted up to two weeks. In conclusion, hypoperfusion during hypobaria exposure could contribute to worsening of neuroinflammation and neurochemical imbalances. The presence of excessive O2 during hypobaria results in long-term suppression of cerebral blood flow, indicating that supplemental O2 should be titrated during hypobaria to maintain normoxia.
{"title":"Aeromedical evacuation-relevant hypobaria following traumatic brain injury in rats contributes to cerebral blood flow depression, altered neurochemistry, and increased neuroinflammation.","authors":"Julie L Proctor, Su Xu, Sijia Guo, Boris Piskoun, Catriona Miller, Steven Roys, Rao P Gullapalli, Gary Fiskum","doi":"10.1177/0271678X241299985","DOIUrl":"10.1177/0271678X241299985","url":null,"abstract":"<p><p>Aircraft cabins are routinely pressurized to the equivalent of 8000 ft altitude. Exposure of lab animals to aeromedical evacuation relevant hypobaria after traumatic brain injury worsens neurological outcomes, which is paradoxically exacerbated by hyperoxia. This study tested the hypothesis that exposure of rats to hypobaria following cortical impact reduces cerebral blood flow, increases neuroinflammation, and alters brain neurochemistry. Rats were exposed to simulated ground (normobaric) or air (hypobaric 8000 ft) transport, under normoxia or hyperoxia, 24 hr after trauma. Hypobaria exposure resulted in lower cerebral blood flow to the contralateral cortex and bilateral thalamus during flight and increased delayed cortical inflammation (ED1 immunoreactivity) at 14 days post injury. Impacted rats exposed to hypobaria had higher cortical creatine levels compared rats maintained at sea level. Exposure to the combination of hyperoxia and hypobaria resulted in the greatest reduction in cortical blood flow and total creatine during flight which persisted up to two weeks. In conclusion, hypoperfusion during hypobaria exposure could contribute to worsening of neuroinflammation and neurochemical imbalances. The presence of excessive O<sub>2</sub> during hypobaria results in long-term suppression of cerebral blood flow, indicating that supplemental O<sub>2</sub> should be titrated during hypobaria to maintain normoxia.</p>","PeriodicalId":15325,"journal":{"name":"Journal of Cerebral Blood Flow and Metabolism","volume":" ","pages":"271678X241299985"},"PeriodicalIF":4.9,"publicationDate":"2024-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11656461/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142854460","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}