1
|
Ermiş M, Karakaş E, Erol H, Akcakavak G, Aci R, Ümit F, Karatas Ö, Çiftci G. Effect of autologous cytokine-rich serum and platelet-rich plasma administration on oxidative status, minerals and proinflammatory cytokines in brain and serum in cyclophosphamide-induced ovarian failure. J Mol Histol 2025; 56:159. [PMID: 40387948 DOI: 10.1007/s10735-025-10448-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Accepted: 05/03/2025] [Indexed: 05/20/2025]
Abstract
Cyclophosphamide (CP) is one of the most commonly used chemotherapy agents and carries a high risk of ovarian damage. This study aimed to evaluate the effects of autologous cytokine-rich serum (ACRS) and platelet-rich plasma (PRP) on brain oxidative status, mineral levels, and proinflammatory cytokines in rats with CP-induced ovarian failure. A total of 42 female Wistar rats (12-weeks-old) were used in the study. Six of these rats were allocated as donors, and the remaining 36 rats were randomly distributed into six groups (n = 6 per group). Group 1 received no treatment. On the 1st and 7th days, 75 mg/kg of CP was administered intraperitoneally to Groups 4, 5, and 6. On day 1, PRP was administered intraovarianly to Groups 2 and 5, while ACRS was administered intraovarianly to Groups 3 and 6. Additionally, PRP and ACRS were administered intraperitoneally to the respective groups on 7th and 14th days.The study was terminated at the end of the 31st day. Brain tissue and blood samples were collected for biochemical analyses and ovarian tissue samples were collected for histomorphological examinations. Morphological analysis using Hematoxylin-Eosin (HE) staining and immunohistochemical evaluation for AMH, α-SMA, and IL-1β were conducted on the ovaries. Proinflammatory cytokines and insulin levels were measured using ELISA test kits. TAS/TOS levels were assessed using Relassay Diagnostic kits. Biochemical parameters and mineral levels were measured using autoanalyzer. Histopathological evaluation revealed that follicular degeneration, congestion, hemorrhage, edema, and inflammatory cell infiltration, as well as the number of atretic follicles and IL-1β immunoreactivity, were observed at the highest levels in the CP group (Group 4). In contrast, the numbers of primordial, primary, secondary, and tertiary follicles, along with AMH and α-SMA immunoreactivity levels, were found to be the lowest in this group. However, positive therapeutic effects were observed in the CP-treated groups (Groups 5 and 6). In the serum, increased levels of AST, ALT, creatinine, glucose, LDL, TOS, Ca, Fe, Mg, IL-1β, IL-1α, TNF-α, and NF-kB were detected in the CP groups (G4, G5, G6) compared to the control groups (G1, G2, and G3). In brain tissue, a decrease of total protein and total cholesterol levels were observed in the CP groups (G4, G5, G6) compared to the control groups, while increases in Na, Cl, Fe, IL-1β, IL-1α, TNF-α, and NF-kB levels were detected. In conclusion, PRP and ACRS therapies from the patient's own blood have a potential as supportive or chemopreventive strategies with reduced side effects and treatment costs.
Collapse
Affiliation(s)
- Mustafa Ermiş
- Erciyes University Experimental Research Application and Research Center, Kayseri, Turkey
| | | | - Hanifi Erol
- Department of Surgery, Faculty of Veterinary Medicine, University of Erciyes, Kayseri, Turkey
| | - Gökhan Akcakavak
- Faculty of Veterinary Medicine, Department of Pathology, Aksaray University, Aksaray, Turkey
| | - Recai Aci
- Aydιn Adnan Menderes University, Söke Vocational School of Health Services, Aydιn, Turkey
| | - Furkan Ümit
- Department of Biochemistry, Faculty of Veterinary Medicine, University of Ondokuz Mayis, 55220, Atakum, Samsun, Turkey
| | - Özhan Karatas
- Faculty of Veterinary Medicine, Department of Pathology, Sivas Cumhuriyet University, Sivas, Turkey
| | - Gülay Çiftci
- Department of Biochemistry, Faculty of Veterinary Medicine, University of Ondokuz Mayis, 55220, Atakum, Samsun, Turkey.
| |
Collapse
|
2
|
Holstein DM, Saliba A, Lozano D, Kim J, Sharma K, Lechleiter JD. β-hydroxybutyrate enhances brain metabolism in normoglycemia and hyperglycemia, providing cerebroprotection in a mouse stroke model. J Cereb Blood Flow Metab 2025:271678X251334222. [PMID: 40219805 PMCID: PMC11993551 DOI: 10.1177/0271678x251334222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/20/2025] [Accepted: 03/26/2025] [Indexed: 04/14/2025]
Abstract
Hyperglycemia in poorly controlled diabetes is widely recognized as detrimental to organ dysfunction. However, the acute effects of hyperglycemia on brain metabolism and function are not fully understood. The potential protective benefit of ketone bodies on mitochondrial function in the brain has also not been well characterized. Here, we evaluated the acute effects of hyperglycemia and β-hydroxybutyrate (BHB) on brain metabolism by employing a novel approach leveraging adenosine triphosphate (ATP)-dependence of bioluminescence originating from luciferin-luciferase activity. Oxygen consumption rate was measured in ex vivo live brain punches to further evaluate mitochondrial function. Our data demonstrate that brain metabolism in mice is affected by acute exposure to high glucose. This short-term effect of glucose exposure was reduced by co-administration with the ketone body BHB. Additionally, we investigated the functional relevance of BHB using an in vivo photothrombotic stroke model to assess its cerebroprotective effects in presence or absence of acute hyperglycemia. BHB significantly reduced infarct size in the brain stroke model, providing functional evidence for its protective role in the brain. These findings suggest that BHB may effectively mitigate the adverse effects of metabolic stress and ischemic events on brain metabolism and function.
Collapse
Affiliation(s)
- Deborah M Holstein
- Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Afaf Saliba
- Center for Precision Medicine, University of Texas Health San Antonio, San Antonio, Texas, USA
- Division of Nephrology, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Damian Lozano
- Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Jiwan Kim
- Division of Nephrology, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Kumar Sharma
- Center for Precision Medicine, University of Texas Health San Antonio, San Antonio, Texas, USA
- Division of Nephrology, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - James D Lechleiter
- Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, Texas, USA
- Center for Precision Medicine, University of Texas Health San Antonio, San Antonio, Texas, USA
| |
Collapse
|
3
|
Li X, Zhu XH, Li Y, Wang T, Zhang G, Wiesner HM, Liang ZP, Chen W. Quantitative mapping of key glucose metabolic rates in the human brain using dynamic deuterium magnetic resonance spectroscopic imaging. PNAS NEXUS 2025; 4:pgaf072. [PMID: 40109558 PMCID: PMC11922071 DOI: 10.1093/pnasnexus/pgaf072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 02/10/2025] [Indexed: 03/22/2025]
Abstract
Deuterium (2H) magnetic resonance spectroscopic imaging (DMRSI) is a newly developed technology for assessing glucose metabolism by simultaneously measuring deuterium-labeled glucose and its downstream metabolites (1) and has a potential to provide a powerful neurometabolic imaging tool for quantitative studies of cerebral glucose metabolism involving multiple metabolic pathways in the human brain. In this work, we developed a dynamic DMRSI method that combines advanced radiofrequency coil and postprocessing techniques to substantially improve the imaging signal-to-noise ratio for detecting deuterated metabolites and enable robust dynamic DMRSI of the human brain at 7 T with very high resolution (HR; 0.7 cc nominal voxel and 2.5 min/image) and whole-brain coverage. Utilizing this capability, we were able to map and differentiate metabolite contents and dynamics throughout the human brain following oral administration of deuterated glucose. Furthermore, by introducing a sophisticated kinetic model, we demonstrated that three key cerebral metabolic rates of glucose consumption (CMRGlc), lactate production (CMRLac), and tricarboxylic acid (TCA) cycle (V TCA), as well as the maximum apparent rate of forward glucose transport (T max) can be simultaneously imaged in the human brain through a single dynamic DMRSI measurement. The results clearly show that the glucose transport, neurotransmitter turnover, CMRGlc, and V TCA are significantly higher in gray matter than in white matter in the human brain; and the mean metabolic rates and their ratios measured in this study are consistent with the values reported in the literature. The HR dynamic DMRSI methodology presented herein is of great significance and value for the quantitative assessment of human brain glucose metabolism, aerobic glycolysis, and metabolic reprogramming under physiopathological conditions.
Collapse
Affiliation(s)
- Xin Li
- Center for Magnetic Resonance Research (CMRR), Department of Radiology, University of Minnesota, 2021 6th St SE, Minneapolis, MN 55455, USA
| | - Xiao-Hong Zhu
- Center for Magnetic Resonance Research (CMRR), Department of Radiology, University of Minnesota, 2021 6th St SE, Minneapolis, MN 55455, USA
| | - Yudu Li
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Tao Wang
- Center for Magnetic Resonance Research (CMRR), Department of Radiology, University of Minnesota, 2021 6th St SE, Minneapolis, MN 55455, USA
| | - Guangle Zhang
- Center for Magnetic Resonance Research (CMRR), Department of Radiology, University of Minnesota, 2021 6th St SE, Minneapolis, MN 55455, USA
| | - Hannes M Wiesner
- Center for Magnetic Resonance Research (CMRR), Department of Radiology, University of Minnesota, 2021 6th St SE, Minneapolis, MN 55455, USA
| | - Zhi-Pei Liang
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Wei Chen
- Center for Magnetic Resonance Research (CMRR), Department of Radiology, University of Minnesota, 2021 6th St SE, Minneapolis, MN 55455, USA
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
4
|
Holstein DM, Saliba A, Lozano D, Kim J, Sharma K, Lechleiter JD. β-Hydroxybutyrate enhances brain metabolism in normoglycemia and hyperglycemia, providing cerebroprotection in a mouse stroke model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.19.639087. [PMID: 40060617 PMCID: PMC11888179 DOI: 10.1101/2025.02.19.639087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
Hyperglycemia in poorly controlled diabetes is widely recognized as detrimental to organ dysfunction. However, the acute effects of hyperglycemia on brain metabolism and function are not fully understood. The potential protective benefit of ketone bodies on mitochondrial function in the brain has also not been well characterized. Here, we evaluated the acute effects of hyperglycemia and β-hydroxybutyrate (BHB) on brain metabolism by employing a novel approach leveraging adenosine triphosphate (ATP)-dependence of bioluminescence originating from luciferin-luciferase activity. Oxygen consumption rate was measured in ex vivo live brain punches to further evaluate mitochondrial function. Additionally, we investigated the functional relevance of BHB using an in vivo photothrombotic stroke model to assess its cerebroprotective effects. Our data demonstrate that brain metabolism in mice is affected by acute exposure to high glucose, at a level similar to consuming food or a beverage with high sucrose. This short-term effect of glucose exposure was reduced by co-administration with the ketone body BHB. Moreover, BHB significantly reduced infarct size in the brain stroke model, providing evidence for its functional protective role in the brain. These findings suggest that BHB may effectively mitigate the adverse effects of metabolic stress and ischemic events on brain metabolism and function.
Collapse
|
5
|
Granov R, Vedad S, Wang SH, Durham A, Shah D, Pasinetti GM. The Role of the Neural Exposome as a Novel Strategy to Identify and Mitigate Health Inequities in Alzheimer's Disease and Related Dementias. Mol Neurobiol 2025; 62:1205-1224. [PMID: 38967905 PMCID: PMC11711138 DOI: 10.1007/s12035-024-04339-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 06/28/2024] [Indexed: 07/06/2024]
Abstract
With the continuous increase of the elderly population, there is an urgency to understand and develop relevant treatments for Alzheimer's disease and related dementias (ADRD). In tandem with this, the prevalence of health inequities continues to rise as disadvantaged communities fail to be included in mainstream research. The neural exposome poses as a relevant mechanistic approach and tool for investigating ADRD onset, progression, and pathology as it accounts for several different factors: exogenous, endogenous, and behavioral. Consequently, through the neural exposome, health inequities can be addressed in ADRD research. In this paper, we address how the neural exposome relates to ADRD by contributing to the discourse through defining how the neural exposome can be developed as a tool in accordance with machine learning. Through this, machine learning can allow for developing a greater insight into the application of transferring and making sense of experimental mouse models exposed to health inequities and potentially relate it to humans. The overall goal moving beyond this paper is to define a multitude of potential factors that can increase the risk of ADRD onset and integrate them to create an interdisciplinary approach to the study of ADRD and subsequently translate the findings to clinical research.
Collapse
Affiliation(s)
- Ravid Granov
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10019, USA
| | - Skyler Vedad
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10019, USA
| | - Shu-Han Wang
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10019, USA
| | - Andrea Durham
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10019, USA
| | - Divyash Shah
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10019, USA
| | - Giulio Maria Pasinetti
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10019, USA.
- Geriatrics Research, Education and Clinical Center, JJ Peters VA Medical Center, Bronx, NY, 10468, USA.
| |
Collapse
|
6
|
Arat-Çelik HE, Eslami Abriz A, Coello K, Vinberg M, Ceylan D. Evaluating Oxidative Stress Markers in At-Risk Individuals for Bipolar Disorder: A Systematic Review and Meta-Analysis. Neuropsychobiology 2024; 83:121-134. [PMID: 39293410 DOI: 10.1159/000540999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 08/15/2024] [Indexed: 09/20/2024]
Abstract
INTRODUCTION Bipolar disorder (BD), a mood disorder with recurrent affective episodes and a strong genetic basis is frequently associated with significant comorbidities, both physical and psychiatric, yet its neurobiology remains unclear. Recent evidence underscores oxidative stress as a pivotal factor linking BD to its comorbidities, prompting an investigation into whether this is a sign of a genetic vulnerability or a consequence of the disease. In this study, we systematically reviewed oxidative stress studies conducted on individuals at risk for BD. We performed a meta-analysis on studies examining oxidative DNA damage in these individuals. METHODS The literature was searched across the databases PubMed, Web of Science, Scopus, Ovid MEDLINE, and Cochrane to locate studies of oxidative stress markers in relatives of patients with BD compared with healthy controls (from 1946 to March 2024). Studies were considered for inclusion based on the following criteria: (i) involvement of first- or second-degree relatives of individuals diagnosed with BD, (ii) presence of a healthy control group, (iii) reporting of oxidative stress parameters for relatives, including mean and standard deviation or median and interquartile range (25-75%) values, and (iv) publication in the English language. Studies comparing the levels of 8-hydroxy-2'-deoxyguanosine (8-OH-dG) or its tautomer 8-oxo-7,8-dihydro-2'-deoxyguanosine (8-oxo-dG) in individuals at risk for BD with healthy controls were evaluated using a meta-analysis with the random-effects method. The risk of bias was evaluated using the Risk of Bias in Non-Randomized Studies of Exposure (ROBINS-E) tool. RESULTS Eleven studies were included in the systematic review and four studies for the meta-analysis. The meta-analysis included 543 individuals (first-degree relatives of individuals with BD = 238, control = 305). 8-OH-dG levels were found to be increased in first-degree relatives of individuals with BD compared to healthy controls (random effects: Hedges's g = 0.53, 95% CI = 0.36-0.71, p < 0.001). Findings of oxidative stress markers other than oxidative DNA damage in relatives of individuals with BD are limited and scarce. CONCLUSION In this meta-analysis, which consists of a limited number of studies, oxidative DNA damage seems to be a trait marker for BD. This finding could be associated with increased comorbidity and a higher risk of premature aging in individuals at risk for BD. However, further studies with larger sample sizes and longitudinal designs are warranted to confirm findings. Clarifying the changes in these markers from individuals at risk for the disorder throughout the course of the illness would help bridge the gap in understanding the role of oxidative pathways in the risk of BD.
Collapse
Affiliation(s)
| | - Aysan Eslami Abriz
- Research Center for Translational Medicine (KUTTAM), Affective Disorders Laboratory, Koc University, Istanbul, Turkey
- Graduate School of Health Sciences, Koç University, Istanbul, Turkey
| | - Klara Coello
- Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Maj Vinberg
- Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Copenhagen, Denmark
- The Early Multimodular Prevention and Intervention Research Institution (EMPIRI), Mental Health Centre, Copenhagen University Hospital - Mental Health Services CPH, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Deniz Ceylan
- Research Center for Translational Medicine (KUTTAM), Affective Disorders Laboratory, Koc University, Istanbul, Turkey
- Graduate School of Health Sciences, Koç University, Istanbul, Turkey
- Department of Psychiatry, School of Medicine, Koc University, Istanbul, Turkey
| |
Collapse
|
7
|
Bisle E, Varadarajan S, Kolassa IT. Vitamin-mediated interaction between the gut microbiome and mitochondria in depression: A systematic review-based integrated perspective. Brain Behav Immun Health 2024; 38:100790. [PMID: 38974216 PMCID: PMC11225645 DOI: 10.1016/j.bbih.2024.100790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 04/08/2024] [Accepted: 05/02/2024] [Indexed: 07/09/2024] Open
Abstract
Depression is one of the world's most prevalent mental disorders and its treatment remains suboptimal. Depression is a systemic disease with highly complex biological mechanisms. Emerging evidence points towards the involvement of mitochondria, microbiome and vitamins in its pathophysiology. Mitochondrial energy production was shown to be lowered in patients with depression. Mitochondrial energy production depends on vitamins, which are available from food, but are also synthesized by the gut microbiota. Several studies reported altered vitamin levels as well as changes in the gut microbiome composition and its vitamin metabolism in patients with depression. Therefore, the question of a connection between mitochondria and gut microbiome and vitamins influencing the mental health arises. This review aims to systematically investigate a combination of the topics - depression, mitochondria, microbiome, and vitamins - to generate an overview of a novel yet extremely complex and interconnected research field. A systematic literature search yielded 34 articles, and the results were summarized and bundled to develop this new integrative perspective on mitochondrial function mediated by the microbiome and microbiome-derived vitamins in depression. Furthermore, by discussing the research gaps this review aims to encourage innovative research approaches to better understand the biology of depression, which could result in optimized therapeutic approaches.
Collapse
Affiliation(s)
- Ellen Bisle
- Department of Clinical & Biological Psychology, Institute of Psychology & Education, Ulm University, Albert-Einstein-Allee 47, 89081, Ulm, Germany
| | - Suchithra Varadarajan
- Department of Clinical & Biological Psychology, Institute of Psychology & Education, Ulm University, Albert-Einstein-Allee 47, 89081, Ulm, Germany
| | - Iris-Tatjana Kolassa
- Department of Clinical & Biological Psychology, Institute of Psychology & Education, Ulm University, Albert-Einstein-Allee 47, 89081, Ulm, Germany
| |
Collapse
|
8
|
Yue WL, Ng KK, Liu S, Qian X, Chong JSX, Koh AJ, Ong MQW, Ting SKS, Ng ASL, Kandiah N, Yeo BTT, Zhou JH. Differential spatial working memory-related functional network reconfiguration in young and older adults. Netw Neurosci 2024; 8:395-417. [PMID: 38952809 PMCID: PMC11142455 DOI: 10.1162/netn_a_00358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 01/05/2024] [Indexed: 07/03/2024] Open
Abstract
Functional brain networks have preserved architectures in rest and task; nevertheless, previous work consistently demonstrated task-related brain functional reorganization. Efficient rest-to-task functional network reconfiguration is associated with better cognition in young adults. However, aging and cognitive load effects, as well as contributions of intra- and internetwork reconfiguration, remain unclear. We assessed age-related and load-dependent effects on global and network-specific functional reconfiguration between rest and a spatial working memory (SWM) task in young and older adults, then investigated associations between functional reconfiguration and SWM across loads and age groups. Overall, global and network-level functional reconfiguration between rest and task increased with age and load. Importantly, more efficient functional reconfiguration associated with better performance across age groups. However, older adults relied more on internetwork reconfiguration of higher cognitive and task-relevant networks. These reflect the consistent importance of efficient network updating despite recruitment of additional functional networks to offset reduction in neural resources and a change in brain functional topology in older adults. Our findings generalize the association between efficient functional reconfiguration and cognition to aging and demonstrate distinct brain functional reconfiguration patterns associated with SWM in aging, highlighting the importance of combining rest and task measures to study aging cognition.
Collapse
Affiliation(s)
- Wan Lin Yue
- Centre for Sleep and Cognition & Centre for Translational Magnetic Resonance Research, Yong Loo Lin School of Medicine, National University of Singapore
- Integrative Sciences and Engineering Programme, NUS Graduate School, National University of Singapore
| | - Kwun Kei Ng
- Centre for Sleep and Cognition & Centre for Translational Magnetic Resonance Research, Yong Loo Lin School of Medicine, National University of Singapore
| | - Siwei Liu
- Centre for Sleep and Cognition & Centre for Translational Magnetic Resonance Research, Yong Loo Lin School of Medicine, National University of Singapore
| | - Xing Qian
- Centre for Sleep and Cognition & Centre for Translational Magnetic Resonance Research, Yong Loo Lin School of Medicine, National University of Singapore
| | - Joanna Su Xian Chong
- Centre for Sleep and Cognition & Centre for Translational Magnetic Resonance Research, Yong Loo Lin School of Medicine, National University of Singapore
| | - Amelia Jialing Koh
- Centre for Sleep and Cognition & Centre for Translational Magnetic Resonance Research, Yong Loo Lin School of Medicine, National University of Singapore
| | - Marcus Qin Wen Ong
- Centre for Sleep and Cognition & Centre for Translational Magnetic Resonance Research, Yong Loo Lin School of Medicine, National University of Singapore
| | | | | | - Nagaendran Kandiah
- National Neuroscience Institute, Singapore
- Neuroscience and Behavioural Disorders Programme, Duke-NUS Medical School, Singapore
| | - B. T. Thomas Yeo
- Centre for Sleep and Cognition & Centre for Translational Magnetic Resonance Research, Yong Loo Lin School of Medicine, National University of Singapore
- Integrative Sciences and Engineering Programme, NUS Graduate School, National University of Singapore
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital
- Department of Electrical and Computer Engineering, N.1 Institute for Health and Memory Networks Program, National University of Singapore
| | - Juan Helen Zhou
- Centre for Sleep and Cognition & Centre for Translational Magnetic Resonance Research, Yong Loo Lin School of Medicine, National University of Singapore
- Integrative Sciences and Engineering Programme, NUS Graduate School, National University of Singapore
- Neuroscience and Behavioural Disorders Programme, Duke-NUS Medical School, Singapore
| |
Collapse
|
9
|
Yaghoobi A, Malekpour SA. Unraveling the genetic architecture of blood unfolded p-53 among non-demented elderlies: novel candidate genes for early Alzheimer's disease. BMC Genomics 2024; 25:440. [PMID: 38702606 PMCID: PMC11067101 DOI: 10.1186/s12864-024-10363-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 04/29/2024] [Indexed: 05/06/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a heritable neurodegenerative disease whose long asymptomatic phase makes the early diagnosis of it pivotal. Blood U-p53 has recently emerged as a superior predictive biomarker for AD in the early stages. We hypothesized that genetic variants associated with blood U-p53 could reveal novel loci and pathways involved in the early stages of AD. RESULTS We performed a blood U-p53 Genome-wide association study (GWAS) on 484 healthy and mild cognitively impaired subjects from the ADNI cohort using 612,843 Single nucleotide polymorphisms (SNPs). We performed a pathway analysis and prioritized candidate genes using an AD single-cell gene program. We fine-mapped the intergenic SNPs by leveraging a cell-type-specific enhancer-to-gene linking strategy using a brain single-cell multimodal dataset. We validated the candidate genes in an independent brain single-cell RNA-seq and the ADNI blood transcriptome datasets. The rs279686 between AASS and FEZF1 genes was the most significant SNP (p-value = 4.82 × 10-7). Suggestive pathways were related to the immune and nervous systems. Twenty-three candidate genes were prioritized at 27 suggestive loci. Fine-mapping of 5 intergenic loci yielded nine cell-specific candidate genes. Finally, 15 genes were validated in the independent single-cell RNA-seq dataset, and five were validated in the ADNI blood transcriptome dataset. CONCLUSIONS We underlined the importance of performing a GWAS on an early-stage biomarker of AD and leveraging functional omics datasets for pinpointing causal genes in AD. Our study prioritized nine genes (SORCS1, KIF5C, TMEFF2, TMEM63C, HLA-E, ATAT1, TUBB, ARID1B, and RUNX1) strongly implicated in the early stages of AD.
Collapse
Affiliation(s)
- Arash Yaghoobi
- School of Biological Sciences, Institute for Research in Fundamental Sciences (IPM), Tehran, 19395-5746, Iran
| | - Seyed Amir Malekpour
- School of Biological Sciences, Institute for Research in Fundamental Sciences (IPM), Tehran, 19395-5746, Iran.
| |
Collapse
|
10
|
Flønes IH, Toker L, Sandnes DA, Castelli M, Mostafavi S, Lura N, Shadad O, Fernandez-Vizarra E, Painous C, Pérez-Soriano A, Compta Y, Molina-Porcel L, Alves G, Tysnes OB, Dölle C, Nido GS, Tzoulis C. Mitochondrial complex I deficiency stratifies idiopathic Parkinson's disease. Nat Commun 2024; 15:3631. [PMID: 38684731 PMCID: PMC11059185 DOI: 10.1038/s41467-024-47867-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 04/15/2024] [Indexed: 05/02/2024] Open
Abstract
Idiopathic Parkinson's disease (iPD) is believed to have a heterogeneous pathophysiology, but molecular disease subtypes have not been identified. Here, we show that iPD can be stratified according to the severity of neuronal respiratory complex I (CI) deficiency, and identify two emerging disease subtypes with distinct molecular and clinical profiles. The CI deficient (CI-PD) subtype accounts for approximately a fourth of all cases, and is characterized by anatomically widespread neuronal CI deficiency, a distinct cell type-specific gene expression profile, increased load of neuronal mtDNA deletions, and a predilection for non-tremor dominant motor phenotypes. In contrast, the non-CI deficient (nCI-PD) subtype exhibits no evidence of mitochondrial impairment outside the dopaminergic substantia nigra and has a predilection for a tremor dominant phenotype. These findings constitute a step towards resolving the biological heterogeneity of iPD with implications for both mechanistic understanding and treatment strategies.
Collapse
Affiliation(s)
- Irene H Flønes
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021, Bergen, Norway
- Department of Clinical Medicine, University of Bergen, Pb 7804, 5020, Bergen, Norway
- K.G. Jebsen Center for Translational Research in Parkinson's disease, University of Bergen, Pb 7804, 5020, Bergen, Norway
| | - Lilah Toker
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021, Bergen, Norway
- Department of Clinical Medicine, University of Bergen, Pb 7804, 5020, Bergen, Norway
- K.G. Jebsen Center for Translational Research in Parkinson's disease, University of Bergen, Pb 7804, 5020, Bergen, Norway
| | - Dagny Ann Sandnes
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021, Bergen, Norway
- Department of Clinical Medicine, University of Bergen, Pb 7804, 5020, Bergen, Norway
| | - Martina Castelli
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021, Bergen, Norway
| | - Sepideh Mostafavi
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021, Bergen, Norway
- Department of Clinical Medicine, University of Bergen, Pb 7804, 5020, Bergen, Norway
| | - Njål Lura
- Mohn Medical Imaging and Visualization Centre, Department of Radiology, Haukeland University Hospital, Bergen, Norway
- Section for Radiology, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Omnia Shadad
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021, Bergen, Norway
- Department of Clinical Medicine, University of Bergen, Pb 7804, 5020, Bergen, Norway
| | - Erika Fernandez-Vizarra
- MRC-Mitochondrial Biology Unit, University of Cambridge, Hills Road, Cambridge, CB2 0XY, UK
- Veneto Institute of Molecular Medicine, 35131, Padova, Italy
| | - Cèlia Painous
- Parkinson's disease & Movement Disorders Unit, Neurology Service, Hospital Clínic I Universitari de Barcelona; IDIBAPS, CIBERNED (CB06/05/0018-ISCIII), ERN-RND, Institut Clínic de Neurociències (Maria de Maeztu excellence centre), Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Alexandra Pérez-Soriano
- Parkinson's disease & Movement Disorders Unit, Neurology Service, Hospital Clínic I Universitari de Barcelona; IDIBAPS, CIBERNED (CB06/05/0018-ISCIII), ERN-RND, Institut Clínic de Neurociències (Maria de Maeztu excellence centre), Universitat de Barcelona, Barcelona, Catalonia, Spain
- UParkinson - Sinapsi Neurología, Centre Mèdic Teknon Grup Hospitalari Quirón Salud, Barcelona, Spain
| | - Yaroslau Compta
- Parkinson's disease & Movement Disorders Unit, Neurology Service, Hospital Clínic I Universitari de Barcelona; IDIBAPS, CIBERNED (CB06/05/0018-ISCIII), ERN-RND, Institut Clínic de Neurociències (Maria de Maeztu excellence centre), Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Laura Molina-Porcel
- Alzheimer's disease and other cognitive disorders unit. Neurology Service, Hospital Clínic, Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Neurological Tissue Bank, Biobanc-Hospital Clínic-IDIBAPS, Barcelona, Spain
| | - Guido Alves
- The Norwegian Centre for Movement Disorders and Department of Neurology, Stavanger University Hospital, Pb 8100, 4068, Stavanger, Norway
- Department of Mathematics and Natural Sciences, University of Stavanger, 4062, Stavanger, Norway
| | - Ole-Bjørn Tysnes
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021, Bergen, Norway
- Department of Clinical Medicine, University of Bergen, Pb 7804, 5020, Bergen, Norway
| | - Christian Dölle
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021, Bergen, Norway
- Department of Clinical Medicine, University of Bergen, Pb 7804, 5020, Bergen, Norway
- K.G. Jebsen Center for Translational Research in Parkinson's disease, University of Bergen, Pb 7804, 5020, Bergen, Norway
| | - Gonzalo S Nido
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021, Bergen, Norway
- Department of Clinical Medicine, University of Bergen, Pb 7804, 5020, Bergen, Norway
- K.G. Jebsen Center for Translational Research in Parkinson's disease, University of Bergen, Pb 7804, 5020, Bergen, Norway
| | - Charalampos Tzoulis
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021, Bergen, Norway.
- Department of Clinical Medicine, University of Bergen, Pb 7804, 5020, Bergen, Norway.
- K.G. Jebsen Center for Translational Research in Parkinson's disease, University of Bergen, Pb 7804, 5020, Bergen, Norway.
| |
Collapse
|
11
|
D'Andrea L, Audano M, Pedretti S, Pelucchi S, Stringhi R, Imperato G, De Cesare G, Cambria C, Laporte MH, Zamboni N, Antonucci F, Di Luca M, Mitro N, Marcello E. Glucose-derived glutamate drives neuronal terminal differentiation in vitro. EMBO Rep 2024; 25:991-1021. [PMID: 38243137 PMCID: PMC10933318 DOI: 10.1038/s44319-023-00048-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 12/01/2023] [Accepted: 12/19/2023] [Indexed: 01/21/2024] Open
Abstract
Neuronal maturation is the phase during which neurons acquire their final characteristics in terms of morphology, electrical activity, and metabolism. However, little is known about the metabolic pathways governing neuronal maturation. Here, we investigate the contribution of the main metabolic pathways, namely glucose, glutamine, and fatty acid oxidation, during the maturation of primary rat hippocampal neurons. Blunting glucose oxidation through the genetic and chemical inhibition of the mitochondrial pyruvate transporter reveals that this protein is critical for the production of glutamate, which is required for neuronal arborization, proper dendritic elongation, and spine formation. Glutamate supplementation in the early phase of differentiation restores morphological defects and synaptic function in mitochondrial pyruvate transporter-inhibited cells. Furthermore, the selective activation of metabotropic glutamate receptors restores the impairment of neuronal differentiation due to the reduced generation of glucose-derived glutamate and rescues synaptic local translation. Fatty acid oxidation does not impact neuronal maturation. Whereas glutamine metabolism is important for mitochondria, it is not for endogenous glutamate production. Our results provide insights into the role of glucose-derived glutamate as a key player in neuronal terminal differentiation.
Collapse
Affiliation(s)
- Laura D'Andrea
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Via Giuseppe Balzaretti 9, 20133, Milan, Italy
| | - Matteo Audano
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Via Giuseppe Balzaretti 9, 20133, Milan, Italy
| | - Silvia Pedretti
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Via Giuseppe Balzaretti 9, 20133, Milan, Italy
| | - Silvia Pelucchi
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Via Giuseppe Balzaretti 9, 20133, Milan, Italy
| | - Ramona Stringhi
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Via Giuseppe Balzaretti 9, 20133, Milan, Italy
| | - Gabriele Imperato
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Via Giuseppe Balzaretti 9, 20133, Milan, Italy
| | - Giulia De Cesare
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Via Giuseppe Balzaretti 9, 20133, Milan, Italy
| | - Clara Cambria
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), Via F.lli Cervi 93, Segrate, 20054 Milan and via Vanvitelli 32, Milan, Italy
| | - Marine H Laporte
- Department of Molecular and Cellular Biology, University of Geneva, Geneva, Switzerland
| | - Nicola Zamboni
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Flavia Antonucci
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), Via F.lli Cervi 93, Segrate, 20054 Milan and via Vanvitelli 32, Milan, Italy
- Institute of Neuroscience, IN-CNR, Milan, Italy
| | - Monica Di Luca
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Via Giuseppe Balzaretti 9, 20133, Milan, Italy
| | - Nico Mitro
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Via Giuseppe Balzaretti 9, 20133, Milan, Italy.
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy.
| | - Elena Marcello
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Via Giuseppe Balzaretti 9, 20133, Milan, Italy.
| |
Collapse
|
12
|
Yahyazadeh A, Başak F, Demirel MA. Efficacy of coenzyme Q10 and curcumin on antioxidant enzyme activity and hippocampal alteration following exposure to cyclophosphamide in male rat. Tissue Cell 2024; 86:102296. [PMID: 38184921 DOI: 10.1016/j.tice.2023.102296] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 12/03/2023] [Accepted: 12/27/2023] [Indexed: 01/09/2024]
Abstract
Coenzyme Q10 (KQ10) and curcumin (KUR) supplements are extensively used for their potential antioxidant, anticancer, and antiapoptotic properties. The present study investigated the neuroprotective potential of KQ10 and KUR against the side effect of cyclophosphamide (SF) (150 mg/kg) on the hippocampus of male Wistar albino rats. Forty-nine 10-12 weeks old rats were randomly divided into seven groups: control, olive oil (OL), SF, KQ10, KUR, SF+KQ10, and SF+KUR. Our biochemical finding showed a significant decrease in superoxide dismutase (SOD) level in the SF group compared to the control group (p < 0.05). There was also a significant reduction in the total number of the hippocampal pyramidal neurons in the CA1, CA2, and CA1-3 regions in the SF group compared to the control group (p < 0.05). In the SF+KQ10 group, we found a significant increase in serum SOD level and the total number of the hippocampal pyramidal neurons in the CA1, CA2, and CA1-3 regions compared to the SF group (p < 0.05). Immunohistochemical and histopathological examination exhibited noteworthy findings in the hippocampus tissues. Our findings showed that KQ10 administration significantly mitigated the hippocampal alteration caused by SF through enhancing antioxidant enzyme activity and reducing apoptosis. However, we found no protective activity of KUR on the hippocampus tissue, which may be due to its weak antioxidative activity.
Collapse
Affiliation(s)
- Ahmad Yahyazadeh
- Department of Histology and Embryology, Faculty of Medicine, Karabuk University, Karabuk, Turkey.
| | - Feyza Başak
- Department of Histology and Embryology, Faculty of Medicine, Karabuk University, Karabuk, Turkey
| | - Mürşide Ayşe Demirel
- Laboratory Animals Breeding and Experimental Research Centre, Department of Basic Pharmaceutical Sciences, Faculty of Pharmacy, Gazi University, Turkey
| |
Collapse
|
13
|
Fang W, Liu L, Yin B, Ke L, Su Y, Liu F, Ma X, Di Q. Heat exposure intervention, anxiety level, and multi-omic profiles: A randomized crossover study. ENVIRONMENT INTERNATIONAL 2023; 181:108247. [PMID: 37871510 DOI: 10.1016/j.envint.2023.108247] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 09/22/2023] [Accepted: 10/03/2023] [Indexed: 10/25/2023]
Abstract
BACKGROUND Climate change has led to the frequent occurrence of high-temperature weather, which has various adverse effects on health, ranging from blood metabolism to systemic organ function. In particular, the sequelae of heat stress injury in most people are related to the nervous system. However, the mechanisms between heat stress and mental health conditions, especially heat stress and anxiety, remain unclear. OBJECTIVE We attempted to elucidate the effect of heat exposure intervention on anxiety levels in the population and its mechanism. METHODS We first carried out a randomized controlled trial in 20 college students in Beijing, China, to observe the results of the effects of heat exposure intervention on human anxiety. Then, we collected blood samples before and after heat exposure experiment and used metabolomic and transcriptomic approaches to quantify serum metabolites and ELISA measurements to explore the underlying mechanisms. RESULTS We found that even 1.5-hour heat exposure intervention significantly increased anxiety levels. Heat stress-induced anxiety was mediated by the activation of the HPA axis, inflammation, oxidative stress, and subsequently unbalanced neurotransmitters. Metabolites such as BDNF, GABA, and glucocorticoids released by the adrenal glands are biomarkers of heat stress-induced anxiety. CONCLUSIONS We have demonstrated a causal link between heat stress and anxiety, explored possible biological pathway between heat stress and anxiety. Heat stress can cause the activation of the HPA axis and lead to changes in the body's metabolism, resulting in a series of changes such as inflammation and oxidative stress, leading to anxiety. This study reveals hidden health cost of climate change that has been underexplored, and also reminds us the importance of immediate climate actions.
Collapse
Affiliation(s)
- Wen Fang
- Division of Sports Science & Physical Education, Tsinghua University, Beijing, China
| | - Linfeng Liu
- School of Medicine, Tsinghua University, Beijing, China
| | - Bo Yin
- School of Medicine, Tsinghua University, Beijing, China
| | - Limei Ke
- School of Medicine, Tsinghua University, Beijing, China
| | - Yao Su
- National Protein Science Facility, School of Life Science Tsinghua University, Beijing China
| | - Fang Liu
- National Protein Science Facility, School of Life Science Tsinghua University, Beijing China
| | - Xindong Ma
- Division of Sports Science & Physical Education, Tsinghua University, Beijing, China; IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China
| | - Qian Di
- Vanke School of Public Health, Tsinghua University, Beijing, China; Institute for Healthy China, Tsinghua University, Beijing, China.
| |
Collapse
|
14
|
Wang J, Zhang W, Xu H, Ellenbroek B, Dai J, Wang L, Yan C, Wang W. The Changes of Histone Methylation Induced by Adolescent Social Stress Regulate the Resting-State Activity in mPFC. RESEARCH (WASHINGTON, D.C.) 2023; 6:0264. [PMID: 38434244 PMCID: PMC10907022 DOI: 10.34133/research.0264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 10/15/2023] [Indexed: 03/05/2024]
Abstract
Early-life stress can lead to sustained alterations in regional resting-state brain functions, but the underlying molecular mechanism remains unclear. Stress can also induce sustained changes in epigenetic modifications across brain regions, which are not limited to a few genes; rather, they often tend to produce global levels of change. The functional implication of these changes also remains to be elucidated. We hypothesize that global epigenetic changes may partly modulate the resting-state functions of brain regions to influence behavior. To test this hypothesis, we used an adolescent social stress (ASS) model in mice and examined the relationship between epigenetic modifications and regional resting-state brain activity using resting-state functional magnetic resonance imaging (rs-fMRI). The results showed that, compared to the control mice, the stressed mice showed increased anxiety and social avoidance behaviors and greater levels of dimethylation of histone H3 at lysine 9 (H3K9me2) in the medial prefrontal cortex (mPFC). In addition, the resting-state activity represented by the amplitude of low-frequency fluctuation (ALFF) was significantly lower in the mPFC of stressed mice. To verify the relationship of H3K9me2 and ALFF, the specific inhibition of H3Kme2 was performed by using the drug UNC0642, which reversed the anxiety behavior induced by ASS and significantly increase the ALFF value of mPFC in both normal and ASS animals. Our study is the first to report an association between histone modifications and rs-fMRI findings, providing a new perspective for understanding of the significance of regional brain epigenetic changes and a possible molecular explanation for rs-fMRI findings.
Collapse
Affiliation(s)
- Jiesi Wang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
| | - Wei Zhang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Hang Xu
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
| | - Bart Ellenbroek
- School of Psychology, Victoria University of Wellington, Kelburn, Wellington 6012, New Zealand
| | - Jiajie Dai
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Li Wang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Chaogan Yan
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
- CAS Key Laboratory of Behavioral Science, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- CAS Key Laboratory of Behavioral Science, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
| | - Weiwen Wang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
- CAS Key Laboratory of Behavioral Science, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
15
|
Karnecki K, Świerczyński J, Steiner J, Krzyżanowska M, Kaliszan M, Gos T. The left-lateralisation of citrate synthase activity in the anterior cingulate cortex of male violent suicide victims. Eur Arch Psychiatry Clin Neurosci 2023; 273:1225-1232. [PMID: 36350374 PMCID: PMC10449962 DOI: 10.1007/s00406-022-01509-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 10/19/2022] [Indexed: 11/11/2022]
Abstract
The anterior cingulate cortex (AC) as a part of prefrontal cortex plays a crucial role in behavioural regulation, which is profoundly disturbed in suicide. Citrate synthase (CS) is a key enzyme of tricarboxylic acid cycle fundamental for brain energetics and neurotransmitter synthesis, which are deteriorated in suicidal behaviour. However, CS activity has not been yet studied in brain structures of suicide victims. CS activity assay was performed bilaterally on frozen samples of the rostral part of the AC of 24 violent suicide completers (21 males and 3 females) with unknown psychiatric diagnosis and 24 non-suicidal controls (20 males and 4 females). Compared to controls, suicide victims revealed decreased CS activity in the right AC, however, insignificant. Further statistical analysis of laterality index revealed the left-lateralisation of CS activity in the AC in male suicides compared to male controls (U-test P = 0.0003, corrected for multiple comparisons). The results were not confounded by postmortem interval, blood alcohol concentration, age, and brain weight. Our findings suggest that disturbed CS activity in the AC plays a role in suicide pathogenesis and correspond with our previous morphological and molecular studies of prefrontal regions in suicide.
Collapse
Affiliation(s)
- Karol Karnecki
- Department of Forensic Medicine, Medical University of Gdańsk, Ul. Dębowa 23, 80-204, Gdańsk, Poland
| | | | - Johann Steiner
- Department of Psychiatry, Otto von Guericke University, Magdeburg, Germany
| | - Marta Krzyżanowska
- Department of Forensic Medicine, Medical University of Gdańsk, Ul. Dębowa 23, 80-204, Gdańsk, Poland
| | - Michał Kaliszan
- Department of Forensic Medicine, Medical University of Gdańsk, Ul. Dębowa 23, 80-204, Gdańsk, Poland
| | - Tomasz Gos
- Department of Forensic Medicine, Medical University of Gdańsk, Ul. Dębowa 23, 80-204, Gdańsk, Poland.
| |
Collapse
|
16
|
Niess F, Strasser B, Hingerl L, Niess E, Motyka S, Hangel G, Krššák M, Gruber S, Spurny-Dworak B, Trattnig S, Scherer T, Lanzenberger R, Bogner W. Reproducibility of 3D MRSI for imaging human brain glucose metabolism using direct ( 2H) and indirect ( 1H) detection of deuterium labeled compounds at 7T and clinical 3T. Neuroimage 2023; 277:120250. [PMID: 37414233 PMCID: PMC11019874 DOI: 10.1016/j.neuroimage.2023.120250] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/25/2023] [Accepted: 06/23/2023] [Indexed: 07/08/2023] Open
Abstract
INTRODUCTION Deuterium metabolic imaging (DMI) and quantitative exchange label turnover (QELT) are novel MR spectroscopy techniques for non-invasive imaging of human brain glucose and neurotransmitter metabolism with high clinical potential. Following oral or intravenous administration of non-ionizing [6,6'-2H2]-glucose, its uptake and synthesis of downstream metabolites can be mapped via direct or indirect detection of deuterium resonances using 2H MRSI (DMI) and 1H MRSI (QELT), respectively. The purpose of this study was to compare the dynamics of spatially resolved brain glucose metabolism, i.e., estimated concentration enrichment of deuterium labeled Glx (glutamate+glutamine) and Glc (glucose) acquired repeatedly in the same cohort of subjects using DMI at 7T and QELT at clinical 3T. METHODS Five volunteers (4 m/1f) were scanned in repeated sessions for 60 min after overnight fasting and 0.8 g/kg oral [6,6'-2H2]-glucose administration using time-resolved 3D 2H FID-MRSI with elliptical phase encoding at 7T and 3D 1H FID-MRSI with a non-Cartesian concentric ring trajectory readout at clinical 3T. RESULTS One hour after oral tracer administration regionally averaged deuterium labeled Glx4 concentrations and the dynamics were not significantly different over all participants between 7T 2H DMI and 3T 1H QELT data for GM (1.29±0.15 vs. 1.38±0.26 mM, p=0.65 & 21±3 vs. 26±3 µM/min, p=0.22) and WM (1.10±0.13 vs. 0.91±0.24 mM, p=0.34 & 19±2 vs. 17±3 µM/min, p=0.48). Also, the observed time constants of dynamic Glc6 data in GM (24±14 vs. 19±7 min, p=0.65) and WM (28±19 vs. 18±9 min, p=0.43) dominated regions showed no significant differences. Between individual 2H and 1H data points a weak to moderate negative correlation was observed for Glx4 concentrations in GM (r=-0.52, p<0.001), and WM (r=-0.3, p<0.001) dominated regions, while a strong negative correlation was observed for Glc6 data GM (r=-0.61, p<0.001) and WM (r=-0.70, p<0.001). CONCLUSION This study demonstrates that indirect detection of deuterium labeled compounds using 1H QELT MRSI at widely available clinical 3T without additional hardware is able to reproduce absolute concentration estimates of downstream glucose metabolites and the dynamics of glucose uptake compared to 2H DMI data acquired at 7T. This suggests significant potential for widespread application in clinical settings especially in environments with limited access to ultra-high field scanners and dedicated RF hardware.
Collapse
Affiliation(s)
- Fabian Niess
- High Field MR Center, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Lazarettgasse 14, Vienna A-1090, Austria.
| | - Bernhard Strasser
- High Field MR Center, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Lazarettgasse 14, Vienna A-1090, Austria
| | - Lukas Hingerl
- High Field MR Center, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Lazarettgasse 14, Vienna A-1090, Austria
| | - Eva Niess
- High Field MR Center, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Lazarettgasse 14, Vienna A-1090, Austria; Christian Doppler Laboratory for MR Imaging Biomarkers (BIOMAK), Austria
| | - Stanislav Motyka
- High Field MR Center, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Lazarettgasse 14, Vienna A-1090, Austria; Christian Doppler Laboratory for MR Imaging Biomarkers (BIOMAK), Austria
| | - Gilbert Hangel
- High Field MR Center, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Lazarettgasse 14, Vienna A-1090, Austria; Department of Neurosurgery, Medical University of Vienna, Austria
| | - Martin Krššák
- Department of Medicine III, Division of Endocrinology and Metabolism, Medical University of Vienna, Austria
| | - Stephan Gruber
- High Field MR Center, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Lazarettgasse 14, Vienna A-1090, Austria; Christian Doppler Laboratory for MR Imaging Biomarkers (BIOMAK), Austria
| | - Benjamin Spurny-Dworak
- Department of Psychiatry and Psychotherapy, Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Austria
| | - Siegfried Trattnig
- High Field MR Center, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Lazarettgasse 14, Vienna A-1090, Austria; Institute for Clinical Molecular MRI, Karl Landsteiner Society, Pölten 3100St, Austria
| | - Thomas Scherer
- Department of Medicine III, Division of Endocrinology and Metabolism, Medical University of Vienna, Austria
| | - Rupert Lanzenberger
- Department of Psychiatry and Psychotherapy, Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Austria
| | - Wolfgang Bogner
- High Field MR Center, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Lazarettgasse 14, Vienna A-1090, Austria; Christian Doppler Laboratory for MR Imaging Biomarkers (BIOMAK), Austria
| |
Collapse
|
17
|
Bednarik P, Goranovic D, Svatkova A, Niess F, Hingerl L, Strasser B, Deelchand DK, Spurny-Dworak B, Krssak M, Trattnig S, Hangel G, Scherer T, Lanzenberger R, Bogner W. 1H magnetic resonance spectroscopic imaging of deuterated glucose and of neurotransmitter metabolism at 7 T in the human brain. Nat Biomed Eng 2023; 7:1001-1013. [PMID: 37106154 PMCID: PMC10861140 DOI: 10.1038/s41551-023-01035-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 03/30/2023] [Indexed: 04/29/2023]
Abstract
Impaired glucose metabolism in the brain has been linked to several neurological disorders. Positron emission tomography and carbon-13 magnetic resonance spectroscopic imaging (MRSI) can be used to quantify the metabolism of glucose, but these methods involve exposure to radiation, cannot quantify downstream metabolism, or have poor spatial resolution. Deuterium MRSI (2H-MRSI) is a non-invasive and safe alternative for the quantification of the metabolism of 2H-labelled substrates such as glucose and their downstream metabolic products, yet it can only measure a limited number of deuterated compounds and requires specialized hardware. Here we show that proton MRSI (1H-MRSI) at 7 T has higher sensitivity, chemical specificity and spatiotemporal resolution than 2H-MRSI. We used 1H-MRSI in five volunteers to differentiate glutamate, glutamine, γ-aminobutyric acid and glucose deuterated at specific molecular positions, and to simultaneously map deuterated and non-deuterated metabolites. 1H-MRSI, which is amenable to clinically available magnetic-resonance hardware, may facilitate the study of glucose metabolism in the brain and its potential roles in neurological disorders.
Collapse
Affiliation(s)
- Petr Bednarik
- High-Field MR Centre, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria.
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Amager and Hvidovre, Copenhagen, Denmark.
- Department of Radiology, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Amager and Hvidovre, Copenhagen, Denmark.
| | - Dario Goranovic
- High-Field MR Centre, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Alena Svatkova
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Amager and Hvidovre, Copenhagen, Denmark
- Department of Radiology, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Amager and Hvidovre, Copenhagen, Denmark
- Department of Medicine III, Division of Endocrinology and Metabolism, Medical University of Vienna, Vienna, Austria
| | - Fabian Niess
- High-Field MR Centre, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Lukas Hingerl
- High-Field MR Centre, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Bernhard Strasser
- High-Field MR Centre, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Dinesh K Deelchand
- Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, MN, USA
| | - Benjamin Spurny-Dworak
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Martin Krssak
- Department of Medicine III, Division of Endocrinology and Metabolism, Medical University of Vienna, Vienna, Austria
| | - Siegfried Trattnig
- High-Field MR Centre, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Gilbert Hangel
- High-Field MR Centre, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
- Department of Neurosurgery, Medical University of Vienna, Vienna, Austria
| | - Thomas Scherer
- Department of Medicine III, Division of Endocrinology and Metabolism, Medical University of Vienna, Vienna, Austria
| | - Rupert Lanzenberger
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Bogner
- High-Field MR Centre, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
18
|
Mayrhofer F, Hanson AM, Navedo MF, Xiang YK, Soulika AM, Deng W, Chechneva OV. Transfer of nuclear and ribosomal material from Sox10-lineage cells to neurons in the mouse brain. J Exp Med 2023; 220:e20221632. [PMID: 37067791 PMCID: PMC10114922 DOI: 10.1084/jem.20221632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 02/22/2023] [Accepted: 03/27/2023] [Indexed: 04/18/2023] Open
Abstract
Material transfer is an essential form of intercellular communication to exchange information and resources between cells. Material transfer between neurons and from glia to neurons has been demonstrated to support neuronal survival and activity. Understanding the extent of material transfer in the healthy nervous system is limited. Here we report that in the mouse central nervous system (CNS), neurons receive nuclear and ribosomal material of Sox10-lineage cell (SOL) origin. We show that transfer of SOL-derived material to neurons is region dependent, establishes during postnatal brain maturation, and dynamically responds to LPS-induced neuroinflammation in the adult mouse brain. We identified satellite oligodendrocyte-neuron pairs with loss of plasma membrane integrity between nuclei, suggesting direct material transfer. Together, our findings provide evidence of regionally coordinated transfer of SOL-derived nuclear and ribosomal material to neurons in the mouse CNS, with potential implications for the understanding and modulation of neuronal function and treatment of neurological disorders.
Collapse
Affiliation(s)
- Florian Mayrhofer
- Institute for Pediatric Regenerative Medicine, Shriners Children’s Northern California, Sacramento, CA, USA
| | - Angela M. Hanson
- Institute for Pediatric Regenerative Medicine, Shriners Children’s Northern California, Sacramento, CA, USA
| | - Manuel F. Navedo
- Department of Pharmacology, University of California, Davis, Davis, CA, USA
| | - Yang K. Xiang
- Department of Pharmacology, University of California, Davis, Davis, CA, USA
- Northern California Health Care System, Mather, CA, USA
| | - Athena M. Soulika
- Institute for Pediatric Regenerative Medicine, Shriners Children’s Northern California, Sacramento, CA, USA
- Department of Dermatology, University of California, Davis, Sacramento, CA, USA
| | - Wenbin Deng
- Institute for Pediatric Regenerative Medicine, Shriners Children’s Northern California, Sacramento, CA, USA
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, CA, USA
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Guangdong, China
| | - Olga V. Chechneva
- Institute for Pediatric Regenerative Medicine, Shriners Children’s Northern California, Sacramento, CA, USA
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, CA, USA
| |
Collapse
|
19
|
Niess F, Hingerl L, Strasser B, Bednarik P, Goranovic D, Niess E, Hangel G, Krššák M, Spurny-Dworak B, Scherer T, Lanzenberger R, Bogner W. Noninvasive 3-Dimensional 1 H-Magnetic Resonance Spectroscopic Imaging of Human Brain Glucose and Neurotransmitter Metabolism Using Deuterium Labeling at 3T : Feasibility and Interscanner Reproducibility. Invest Radiol 2023; 58:431-437. [PMID: 36735486 PMCID: PMC10184811 DOI: 10.1097/rli.0000000000000953] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 12/15/2022] [Indexed: 02/04/2023]
Abstract
OBJECTIVES Noninvasive, affordable, and reliable mapping of brain glucose metabolism is of critical interest for clinical research and routine application as metabolic impairment is linked to numerous pathologies, for example, cancer, dementia, and depression. A novel approach to map glucose metabolism noninvasively in the human brain has been presented recently on ultrahigh-field magnetic resonance (MR) scanners (≥7T) using indirect detection of deuterium-labeled glucose and downstream metabolites such as glutamate, glutamine, and lactate. The aim of this study was to demonstrate the feasibility to noninvasively detect deuterium-labeled downstream glucose metabolites indirectly in the human brain via 3-dimensional (3D) proton ( 1 H) MR spectroscopic imaging on a clinical 3T MR scanner without additional hardware. MATERIALS AND METHODS This prospective, institutional review board-approved study was performed in 7 healthy volunteers (mean age, 31 ± 4 years, 5 men/2 women) after obtaining written informed consent. After overnight fasting and oral deuterium-labeled glucose administration, 3D metabolic maps were acquired every ∼4 minutes with ∼0.24 mL isotropic spatial resolution using real-time motion-, shim-, and frequency-corrected echo-less 3D 1 H-MR spectroscopic Imaging on a clinical routine 3T MR system. To test the interscanner reproducibility of the method, subjects were remeasured on a similar 3T MR system. Time courses were analyzed using linear regression and nonparametric statistical tests. Deuterium-labeled glucose and downstream metabolites were detected indirectly via their respective signal decrease in dynamic 1 H MR spectra due to exchange of labeled and unlabeled molecules. RESULTS Sixty-five minutes after deuterium-labeled glucose administration, glutamate + glutamine (Glx) signal intensities decreased in gray/white matter (GM/WM) by -1.63 ± 0.3/-1.0 ± 0.3 mM (-13% ± 3%, P = 0.02/-11% ± 3%, P = 0.02), respectively. A moderate to strong negative correlation between Glx and time was observed in GM/WM ( r = -0.64, P < 0.001/ r = -0.54, P < 0.001), with 60% ± 18% ( P = 0.02) steeper slopes in GM versus WM, indicating faster metabolic activity. Other nonlabeled metabolites showed no significant changes. Excellent intrasubject repeatability was observed across scanners for static results at the beginning of the measurement (coefficient of variation 4% ± 4%), whereas differences were observed in individual Glx dynamics, presumably owing to physiological variation of glucose metabolism. CONCLUSION Our approach translates deuterium metabolic imaging to widely available clinical routine MR scanners without specialized hardware, offering a safe, affordable, and versatile (other substances than glucose can be labeled) approach for noninvasive imaging of glucose and neurotransmitter metabolism in the human brain.
Collapse
Affiliation(s)
- Fabian Niess
- From the High Field MR Center, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Lukas Hingerl
- From the High Field MR Center, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Bernhard Strasser
- From the High Field MR Center, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Petr Bednarik
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, University Hospital Amager and Hvidovre, Hvidovre, Denmark
- Department of Radiology, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Dario Goranovic
- From the High Field MR Center, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Eva Niess
- From the High Field MR Center, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Gilbert Hangel
- From the High Field MR Center, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
- Department of Neurosurgery
| | - Martin Krššák
- Department of Medicine III, Division of Endocrinology and Metabolism
| | - Benjamin Spurny-Dworak
- Department of Psychiatry and Psychotherapy, Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - Thomas Scherer
- Department of Medicine III, Division of Endocrinology and Metabolism
| | - Rupert Lanzenberger
- Department of Psychiatry and Psychotherapy, Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - Wolfgang Bogner
- From the High Field MR Center, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
20
|
Van Huynh T, Rethi L, Rethi L, Chen CH, Chen YJ, Kao YH. The Complex Interplay between Imbalanced Mitochondrial Dynamics and Metabolic Disorders in Type 2 Diabetes. Cells 2023; 12:1223. [PMID: 37174622 PMCID: PMC10177489 DOI: 10.3390/cells12091223] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 04/15/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a global burden, with an increasing number of people affected and increasing treatment costs. The advances in research and guidelines improve the management of blood glucose and related diseases, but T2DM and its complications are still a big challenge in clinical practice. T2DM is a metabolic disorder in which insulin signaling is impaired from reaching its effectors. Mitochondria are the "powerhouses" that not only generate the energy as adenosine triphosphate (ATP) using pyruvate supplied from glucose, free fatty acid (FFA), and amino acids (AA) but also regulate multiple cellular processes such as calcium homeostasis, redox balance, and apoptosis. Mitochondrial dysfunction leads to various diseases, including cardiovascular diseases, metabolic disorders, and cancer. The mitochondria are highly dynamic in adjusting their functions according to cellular conditions. The shape, morphology, distribution, and number of mitochondria reflect their function through various processes, collectively known as mitochondrial dynamics, including mitochondrial fusion, fission, biogenesis, transport, and mitophagy. These processes determine the overall mitochondrial health and vitality. More evidence supports the idea that dysregulated mitochondrial dynamics play essential roles in the pathophysiology of insulin resistance, obesity, and T2DM, as well as imbalanced mitochondrial dynamics found in T2DM. This review updates and discusses mitochondrial dynamics and the complex interactions between it and metabolic disorders.
Collapse
Affiliation(s)
- Tin Van Huynh
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Department of Interventional Cardiology, Thong Nhat Hospital, Ho Chi Minh City 700000, Vietnam
| | - Lekha Rethi
- School of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
- International Ph.D. Program for Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Lekshmi Rethi
- International Ph.D. Program for Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Chih-Hwa Chen
- School of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
- Department of Orthopedics, Taipei Medical University-Shuang Ho Hospital, New Taipei City 23561, Taiwan
- School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Yi-Jen Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 11031, Taiwan
| | - Yu-Hsun Kao
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Department of Medical Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
21
|
John SR, Krauskopf B, Osinga HM, Rubin JE. Slow negative feedback enhances robustness of square-wave bursting. J Comput Neurosci 2023; 51:239-261. [PMID: 37067661 PMCID: PMC10181982 DOI: 10.1007/s10827-023-00846-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 01/17/2023] [Accepted: 02/15/2023] [Indexed: 04/18/2023]
Abstract
Square-wave bursting is an activity pattern common to a variety of neuronal and endocrine cell models that has been linked to central pattern generation for respiration and other physiological functions. Many of the reduced mathematical models that exhibit square-wave bursting yield transitions to an alternative pseudo-plateau bursting pattern with small parameter changes. This susceptibility to activity change could represent a problematic feature in settings where the release events triggered by spike production are necessary for function. In this work, we analyze how model bursting and other activity patterns vary with changes in a timescale associated with the conductance of a fast inward current. Specifically, using numerical simulations and dynamical systems methods, such as fast-slow decomposition and bifurcation and phase-plane analysis, we demonstrate and explain how the presence of a slow negative feedback associated with a gradual reduction of a fast inward current in these models helps to maintain the presence of spikes within the active phases of bursts. Therefore, although such a negative feedback is not necessary for burst production, we find that its presence generates a robustness that may be important for function.
Collapse
Affiliation(s)
- Sushmita Rose John
- Department of Mathematics, University of Pittsburgh, 301 Thackeray Hall, Pittsburgh, 15260, PA, USA
| | - Bernd Krauskopf
- Department of Mathematics, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Hinke M Osinga
- Department of Mathematics, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.
| | - Jonathan E Rubin
- Department of Mathematics, University of Pittsburgh, 301 Thackeray Hall, Pittsburgh, 15260, PA, USA.
| |
Collapse
|
22
|
Niess F, Strasser B, Hingerl L, Niess E, Motyka S, Hangel G, Krššák M, Gruber S, Spurny-Dworak B, Trattnig S, Scherer T, Lanzenberger R, Bogner W. Reproducibility of 3D MRSI for imaging human brain glucose metabolism using direct ( 2 H) and indirect ( 1 H) detection of deuterium labeled compounds at 7T and clinical 3T. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.04.17.23288672. [PMID: 37131634 PMCID: PMC10153308 DOI: 10.1101/2023.04.17.23288672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Introduction Deuterium metabolic imaging (DMI) and quantitative exchange label turnover (QELT) are novel MR spectroscopy techniques for non-invasive imaging of human brain glucose and neurotransmitter metabolism with high clinical potential. Following oral or intravenous administration of non-ionizing [6,6'- 2 H 2 ]-glucose, its uptake and synthesis of downstream metabolites can be mapped via direct or indirect detection of deuterium resonances using 2 H MRSI (DMI) and 1 H MRSI (QELT), respectively. The purpose of this study was to compare the dynamics of spatially resolved brain glucose metabolism, i.e., estimated concentration enrichment of deuterium labeled Glx (glutamate+glutamine) and Glc (glucose) acquired repeatedly in the same cohort of subjects using DMI at 7T and QELT at clinical 3T. Methods Five volunteers (4m/1f) were scanned in repeated sessions for 60 min after overnight fasting and 0.8g/kg oral [6,6'- 2 H 2 ]-glucose administration using time-resolved 3D 2 H FID-MRSI with elliptical phase encoding at 7T and 3D 1 H FID-MRSI with a non-Cartesian concentric ring trajectory readout at clinical 3T. Results One hour after oral tracer administration regionally averaged deuterium labeled Glx 4 concentrations and the dynamics were not significantly different over all participants between 7T 2 H DMI and 3T 1 H QELT data for GM (1.29±0.15 vs. 1.38±0.26 mM, p=0.65 & 21±3 vs. 26±3 µM/min, p=0.22) and WM (1.10±0.13 vs. 0.91±0.24 mM, p=0.34 & 19±2 vs. 17±3 µM/min, p=0.48). Also, the observed time constants of dynamic Glc 6 data in GM (24±14 vs. 19±7 min, p=0.65) and WM (28±19 vs. 18±9 min, p=0.43) dominated regions showed no significant differences. Between individual 2 H and 1 H data points a weak to moderate negative correlation was observed for Glx 4 concentrations in GM (r=-0.52, p<0.001), and WM (r=-0.3, p<0.001) dominated regions, while a strong negative correlation was observed for Glc 6 data GM (r=- 0.61, p<0.001) and WM (r=-0.70, p<0.001). Conclusion This study demonstrates that indirect detection of deuterium labeled compounds using 1 H QELT MRSI at widely available clinical 3T without additional hardware is able to reproduce absolute concentration estimates of downstream glucose metabolites and the dynamics of glucose uptake compared to 2 H DMI data acquired at 7T. This suggests significant potential for widespread application in clinical settings especially in environments with limited access to ultra-high field scanners and dedicated RF hardware.
Collapse
Affiliation(s)
- Fabian Niess
- High Field MR Center, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna
| | - Bernhard Strasser
- High Field MR Center, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna
| | - Lukas Hingerl
- High Field MR Center, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna
| | - Eva Niess
- High Field MR Center, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna
- Christian Doppler Laboratory for MR Imaging Biomarkers (BIOMAK)
| | - Stanislav Motyka
- High Field MR Center, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna
- Christian Doppler Laboratory for MR Imaging Biomarkers (BIOMAK)
| | - Gilbert Hangel
- High Field MR Center, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna
- Department of Neurosurgery, Medical University of Vienna
| | - Martin Krššák
- Department of Medicine III, Division of Endocrinology and Metabolism, Medical University of Vienna
| | - Stephan Gruber
- High Field MR Center, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna
- Christian Doppler Laboratory for MR Imaging Biomarkers (BIOMAK)
| | - Benjamin Spurny-Dworak
- Department of Psychiatry and Psychotherapy, Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna
| | - Siegfried Trattnig
- High Field MR Center, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna
- Institute for Clinical Molecular MRI, Karl Landsteiner Society, 3100 St. Pölten, Austria
| | - Thomas Scherer
- Department of Medicine III, Division of Endocrinology and Metabolism, Medical University of Vienna
| | - Rupert Lanzenberger
- Department of Psychiatry and Psychotherapy, Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna
| | - Wolfgang Bogner
- High Field MR Center, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna
- Christian Doppler Laboratory for MR Imaging Biomarkers (BIOMAK)
| |
Collapse
|
23
|
Yu Y, Akif A, Herman P, Cao M, Rothman DL, Carson RE, Agarwal D, Evans AC, Hyder F. A 3D atlas of functional human brain energetic connectome based on neuropil distribution. Cereb Cortex 2023; 33:3996-4012. [PMID: 36104858 PMCID: PMC10068297 DOI: 10.1093/cercor/bhac322] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 11/13/2022] Open
Abstract
The human brain is energetically expensive, yet the key factors governing its heterogeneous energy distributions across cortical regions to support its diversity of functions remain unexplored. Here, we built up a 3D digital cortical energy atlas based on the energetic costs of all neuropil activities into a high-resolution stereological map of the human cortex with cellular and synaptic densities derived, respectively, from ex vivo histological staining and in vivo PET imaging. The atlas was validated with PET-measured glucose oxidation at the voxel level. A 3D cortical activity map was calculated to predict the heterogeneous activity rates across all cortical regions, which revealed that resting brain is indeed active with heterogeneous neuronal activity rates averaging around 1.2 Hz, comprising around 70% of the glucose oxidation of the cortex. Additionally, synaptic density dominates spatial patterns of energetics, suggesting that the cortical energetics rely heavily on the distribution of synaptic connections. Recent evidence from functional imaging studies suggests that some cortical areas act as hubs (i.e., interconnecting distinct and functionally active regions). An inverse allometric relationship was observed between hub metabolic rates versus hub volumes. Hubs with smaller volumes have higher synapse density, metabolic rate, and activity rates compared to nonhubs. The open-source BrainEnergyAtlas provides a granular framework for exploring revealing design principles in energy-constrained human cortical circuits across multiple spatial scales.
Collapse
Affiliation(s)
- Yuguo Yu
- Shanghai Artificial Intelligence Laboratory, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Research Institute of Intelligent and Complex Systems, Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai 200032, China
| | - Adil Akif
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| | - Peter Herman
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA
- Magnetic Resonance Research Center, Yale University, New Haven, CT 06520, USA
| | - Miao Cao
- Shanghai Artificial Intelligence Laboratory, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Research Institute of Intelligent and Complex Systems, Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai 200032, China
| | - Douglas L Rothman
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA
- Magnetic Resonance Research Center, Yale University, New Haven, CT 06520, USA
| | - Richard E Carson
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA
- PET Center, Yale University, New Haven, CT 06520, USA
| | - Divyansh Agarwal
- Department of Surgery, MGH, Harvard University, Boston, MA 02114, USA
| | - Alan C Evans
- Montreal Neurological Institute, McGill University, Montreal, Quebec H3A 0G4, Canada
| | - Fahmeed Hyder
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA
- Magnetic Resonance Research Center, Yale University, New Haven, CT 06520, USA
| |
Collapse
|
24
|
Kang BS, Choi BY, Kho AR, Lee SH, Hong DK, Park MK, Lee SH, Lee CJ, Yang HW, Woo SY, Park SW, Kim DY, Park JB, Chung WS, Suh SW. Effects of Pyruvate Kinase M2 (PKM2) Gene Deletion on Astrocyte-Specific Glycolysis and Global Cerebral Ischemia-Induced Neuronal Death. Antioxidants (Basel) 2023; 12:491. [PMID: 36830049 PMCID: PMC9952809 DOI: 10.3390/antiox12020491] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/04/2023] [Accepted: 02/14/2023] [Indexed: 02/17/2023] Open
Abstract
Ischemic stroke is caused by insufficient blood flow to the brain. Astrocytes have a role in bidirectionally converting pyruvate, generated via glycolysis, into lactate and then supplying it to neurons through astrocyte-neuron lactate shuttle (ANLS). Pyruvate kinase M2 (PKM2) is an enzyme that dephosphorylates phosphoenolpyruvate to pyruvate during glycolysis in astrocytes. We hypothesized that a reduction in lactate supply in astrocyte PKM2 gene deletion exacerbates neuronal death. Mice harboring a PKM2 gene deletion were established by administering tamoxifen to Aldh1l1-CreERT2; PKM2f/f mice. Upon development of global cerebral ischemia, mice were immediately injected with sodium l-lactate (250 mg/kg, i.p.). To verify our hypothesis, we compared oxidative damage, microtubule disruption, ANLS disruption, and neuronal death between the gene deletion and control subjects. We observed that PKM2 gene deletion increases the degree of neuronal damage and impairment of lactate metabolism in the hippocampal region after GCI. The lactate administration groups showed significantly reduced neuronal death and increases in neuron survival and cognitive function. We found that lactate supply via the ANLS in astrocytes plays a crucial role in maintaining energy metabolism in neurons. Lactate administration may have potential as a therapeutic tool to prevent neuronal damage following ischemic stroke.
Collapse
Affiliation(s)
- Beom-Seok Kang
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Bo-Young Choi
- Department of Physical Education, Hallym University, Chuncheon 24252, Republic of Korea
- Institute of Sport Science, Hallym University, Chuncheon 24252, Republic of Korea
| | - A-Ra Kho
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, College of Medicine, Johns Hopkins University School, Baltimore, MD 21205, USA
- Department of Neurology, College of Medicine, Johns Hopkins University School, Baltimore, MD 21205, USA
| | - Song-Hee Lee
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Dae-Ki Hong
- Department of Pathology and Laboratory Medicine, College of Medicine, Emory University School, Atlanta, GA 30322, USA
| | - Min-Kyu Park
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Si-Hyun Lee
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Chang-Juhn Lee
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Hyeun-Wook Yang
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Seo-Young Woo
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Se-Wan Park
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Dong-Yeon Kim
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Jae-Bong Park
- Department of Biochemistry, College of Medicine, Chuncheon 24252, Republic of Korea
| | - Won-Suk Chung
- Department of Biological Sciences and KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology, Daejeon 34051, Republic of Korea
| | - Sang-Won Suh
- Department of Physiology, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| |
Collapse
|
25
|
Ma T, Chang RCC, Macauley SL. Editorial: Metabolic signaling dysregulation and cognitive impairments in aging and Alzheimer's disease, volume II. Front Aging Neurosci 2023; 15:1150101. [PMID: 36819722 PMCID: PMC9936185 DOI: 10.3389/fnagi.2023.1150101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 01/25/2023] [Indexed: 02/05/2023] Open
Affiliation(s)
- Tao Ma
- Department of Internal Medicine, Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, United States
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
- Department of Neurobiology and Anatomy, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Raymond Chuen-Chung Chang
- Laboratory of Neurodegenerative Diseases, LKS Faculty of Medicine, School of Biomedical Science, The University of Hong Kong, Pok Fu Lam, Hong Kong SAR, China
| | - Shannon L. Macauley
- Department of Internal Medicine, Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, United States
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
26
|
Hafiz R, Gandhi TK, Mishra S, Prasad A, Mahajan V, Natelson BH, Di X, Biswal BB. Assessing functional connectivity differences and work-related fatigue in surviving COVID-negative patients. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2022.02.01.478677. [PMID: 35132408 PMCID: PMC8820653 DOI: 10.1101/2022.02.01.478677] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The Coronavirus Disease 2019 (COVID-19) has affected all aspects of life around the world. Neuroimaging evidence suggests the novel coronavirus can attack the central nervous system (CNS), causing cerebro-vascular abnormalities in the brain. This can lead to focal changes in cerebral blood flow and metabolic oxygen consumption rate in the brain. However, the extent and spatial locations of brain alterations in COVID-19 survivors are largely unknown. In this study, we have assessed brain functional connectivity (FC) using resting-state functional MRI (RS-fMRI) in 38 (25 males) COVID patients two weeks after hospital discharge, when PCR negative and 31 (24 males) healthy subjects. FC was estimated using independent component analysis (ICA) and dual regression. When compared to the healthy group, the COVID group demonstrated significantly enhanced FC in the basal ganglia and precuneus networks (family wise error (fwe) corrected, pfwe < 0.05), while, on the other hand, reduced FC in the language network (pfwe < 0.05). The COVID group also experienced higher fatigue levels during work, compared to the healthy group (p < 0.001). Moreover, within the precuneus network, we noticed a significant negative correlation between FC and fatigue scores across groups (Spearman's ρ = -0.47, p = 0.001, r2 = 0.22). Interestingly, this relationship was found to be significantly stronger among COVID survivors within the left parietal lobe, which is known to be structurally and functionally associated with fatigue in other neurological disorders.
Collapse
Affiliation(s)
- Rakibul Hafiz
- Department of Biomedical Engineering, New Jersey Institute of Technology (NJIT), 323 Dr Martin Luther King Jr Blvd, Newark, NJ 07102, USA
| | - Tapan Kumar Gandhi
- Department of Electrical Engineering, Indian Institute of Technology (IIT), Block II, IIT Delhi Main Rd, IIT Campus, Hauz Khas, New Delhi, Delhi 110016, India
| | - Sapna Mishra
- Department of Electrical Engineering, Indian Institute of Technology (IIT), Block II, IIT Delhi Main Rd, IIT Campus, Hauz Khas, New Delhi, Delhi 110016, India
| | - Alok Prasad
- Internal Medicine, Irene Hospital & Senior Consultant Medicine, Metro Heart and Super-specialty Hospital, New Delhi, India
| | - Vidur Mahajan
- Centre for Advanced Research in Imaging, Neuroscience & Genomics, Mahajan Imaging, New Delhi, India
| | - Benjamin H. Natelson
- Pain and Fatigue Study Center, Department of Neurology, Icahn School of Medicine at Mount Sinai, NY, USA
| | - Xin Di
- Department of Biomedical Engineering, New Jersey Institute of Technology (NJIT), 323 Dr Martin Luther King Jr Blvd, Newark, NJ 07102, USA
| | - Bharat B. Biswal
- Department of Biomedical Engineering, New Jersey Institute of Technology (NJIT), 323 Dr Martin Luther King Jr Blvd, Newark, NJ 07102, USA
| |
Collapse
|
27
|
Greeck VB, Williams SK, Haas J, Wildemann B, Fairless R. Alterations in Lymphocytic Metabolism-An Emerging Hallmark of MS Pathophysiology? Int J Mol Sci 2023; 24:ijms24032094. [PMID: 36768415 PMCID: PMC9917089 DOI: 10.3390/ijms24032094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/21/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disease of the central nervous system (CNS) characterised by acute inflammation and subsequent neuro-axonal degeneration resulting in progressive neurological impairment. Aberrant immune system activation in the periphery and subsequent lymphocyte migration to the CNS contribute to the pathophysiology. Recent research has identified metabolic dysfunction as an additional feature of MS. It is already well known that energy deficiency in neurons caused by impaired mitochondrial oxidative phosphorylation results in ionic imbalances that trigger degenerative pathways contributing to white and grey matter atrophy. However, metabolic dysfunction in MS appears to be more widespread than the CNS. This review focuses on recent research assessing the metabolism and mitochondrial function in peripheral immune cells of MS patients and lymphocytes isolated from murine models of MS. Emerging evidence suggests that pharmacological modulation of lymphocytic metabolism may regulate their subtype differentiation and rebalance pro- and anti-inflammatory functions. As such, further understanding of MS immunometabolism may aid the identification of novel treatments to specifically target proinflammatory immune responses.
Collapse
Affiliation(s)
- Viktoria B. Greeck
- Department of Neurology, University Clinic Heidelberg, 69120 Heidelberg, Germany
- Clinical Cooperation Unit (CCU) Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Sarah K. Williams
- Department of Neurology, University Clinic Heidelberg, 69120 Heidelberg, Germany
- Clinical Cooperation Unit (CCU) Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Jürgen Haas
- Department of Neurology, University Clinic Heidelberg, 69120 Heidelberg, Germany
| | - Brigitte Wildemann
- Department of Neurology, University Clinic Heidelberg, 69120 Heidelberg, Germany
| | - Richard Fairless
- Department of Neurology, University Clinic Heidelberg, 69120 Heidelberg, Germany
- Clinical Cooperation Unit (CCU) Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Correspondence:
| |
Collapse
|
28
|
Zhang F, Khan AF, Ding L, Yuan H. Network organization of resting-state cerebral hemodynamics and their aliasing contributions measured by functional near-infrared spectroscopy. J Neural Eng 2023; 20:016012. [PMID: 36535032 PMCID: PMC9855663 DOI: 10.1088/1741-2552/acaccb] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 12/05/2022] [Accepted: 12/19/2022] [Indexed: 12/23/2022]
Abstract
Objective. Spontaneous fluctuations of cerebral hemodynamics measured by functional magnetic resonance imaging (fMRI) are widely used to study the network organization of the brain. The temporal correlations among the ultra-slow, <0.1 Hz fluctuations across the brain regions are interpreted as functional connectivity maps and used for diagnostics of neurological disorders. However, despite the interest narrowed in the ultra-slow fluctuations, hemodynamic activity that exists beyond the ultra-slow frequency range could contribute to the functional connectivity, which remains unclear.Approach. In the present study, we have measured the brain-wide hemodynamics in the human participants with functional near-infrared spectroscopy (fNIRS) in a whole-head, cap-based and high-density montage at a sampling rate of 6.25 Hz. In addition, we have acquired resting state fMRI scans in the same group of participants for cross-modal evaluation of the connectivity maps. Then fNIRS data were deliberately down-sampled to a typical fMRI sampling rate of ∼0.5 Hz and the resulted differential connectivity maps were subject to a k-means clustering.Main results. Our diffuse optical topographical analysis of fNIRS data have revealed a default mode network (DMN) in the spontaneous deoxygenated and oxygenated hemoglobin changes, which remarkably resemble the same fMRI network derived from participants. Moreover, we have shown that the aliased activities in the down-sampled optical signals have altered the connectivity patterns, resulting in a network organization of aliased functional connectivity in the cerebral hemodynamics.Significance.The results have for the first time demonstrated that fNIRS as a broadly accessible modality can image the resting-state functional connectivity in the posterior midline, prefrontal and parietal structures of the DMN in the human brain, in a consistent pattern with fMRI. Further empowered by the fast sampling rate of fNIRS, our findings suggest the presence of aliased connectivity in the current understanding of the human brain organization.
Collapse
Affiliation(s)
- Fan Zhang
- Stephenson School of Biomedical Engineering, The University of Oklahoma, Norman, OK 73019, United States of America
| | - Ali F Khan
- Stephenson School of Biomedical Engineering, The University of Oklahoma, Norman, OK 73019, United States of America
| | - Lei Ding
- Stephenson School of Biomedical Engineering, The University of Oklahoma, Norman, OK 73019, United States of America
- Institute for Biomedical Engineering, Science and Technology, The University of Oklahoma, Norman, OK 73019, United States of America
| | - Han Yuan
- Stephenson School of Biomedical Engineering, The University of Oklahoma, Norman, OK 73019, United States of America
- Institute for Biomedical Engineering, Science and Technology, The University of Oklahoma, Norman, OK 73019, United States of America
| |
Collapse
|
29
|
Carmichael O. The Role of fMRI in Drug Development: An Update. ADVANCES IN NEUROBIOLOGY 2023; 30:299-333. [PMID: 36928856 DOI: 10.1007/978-3-031-21054-9_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
Functional magnetic resonance imaging (fMRI) of the brain is a technology that holds great potential for increasing the efficiency of drug development for the central nervous system (CNS). In preclinical studies and both early- and late-phase human trials, fMRI has the potential to improve cross-species translation of drug effects, help to de-risk compounds early in development, and contribute to the portfolio of evidence for a compound's efficacy and mechanism of action. However, to date, the utilization of fMRI in the CNS drug development process has been limited. The purpose of this chapter is to explore this mismatch between potential and utilization. This chapter provides introductory material related to fMRI and drug development, describes what is required of fMRI measurements for them to be useful in a drug development setting, lists current capabilities of fMRI in this setting and challenges faced in its utilization, and ends with directions for future development of capabilities in this arena. This chapter is the 5-year update of material from a previously published workshop summary (Carmichael et al., Drug DiscovToday 23(2):333-348, 2018).
Collapse
Affiliation(s)
- Owen Carmichael
- Pennington Biomedical Research Center, Baton Rouge, LA, USA.
| |
Collapse
|
30
|
Krycer JR, Nayler SP. A Survey of the Metabolic Landscape of the Developing Cerebellum at Single-Cell Resolution. CEREBELLUM (LONDON, ENGLAND) 2022; 21:838-850. [PMID: 35767214 DOI: 10.1007/s12311-022-01415-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/09/2022] [Indexed: 06/15/2023]
Abstract
The use of cell-culture models to investigate development and disease of the cerebellum is a recent advance, facilitated by the discovery that patterning of precursors is capable of giving rise to cells with specific neuronal identity. Pluripotent stem cell-derived organoids, which exhibit self-organisational characteristics reminiscent of early cerebellar tissue, present a number of challenges including recapitulation of conditions resembling the mature brain. An understanding of the processes driving fetal and postnatal maturation is required to reproduce these conditions in vitro and advance the capability of the system to model adult-onset disease. A key tool for achieving this is single-cell RNA sequencing, which enables visualisation of key transcriptional features of subpopulations comprising tissues. Here, we explore and compare available single-cell RNA sequencing data derived from the developing human cerebellum and its synthetic, in vitro counterpart (stem cell-derived cerebellar organoids). We focus on performing a qualitative assessment of the expression of key metabolic pathway genes, given recent findings exemplifying tissue-specific metabolic activity, including hypoxia and metabolic shifts associated with neuronal expansion. Signatures indicative of known cell type-specific metabolic differences, such as the astrocyte-neuron lactate shuttle and glutamate-glutamine cycle were evident at a transcriptional level. Cerebellar tissue and cerebellar organoids showed a number of behavioural similarities, including HIF1 signalling, which may serve to drive expansion of granule cell progenitors in both settings. We further highlight numerous differences between cultured organoids and native tissue which may provide clarity on the state of metabolic state following differentiation of organoids, providing the future framework to test and further hypotheses regarding promoting maturation. Overall, this analysis provides insight into understanding the state of in vitro models of the cerebellum, a critical factor required for modelling susceptibility of various cell types to cerebellar disease.
Collapse
Affiliation(s)
- James R Krycer
- Queensland Institute of Medical Research Berghofer Research Institute, Herston, QLD, Australia
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
| | - Sam P Nayler
- Queensland Institute of Medical Research Berghofer Research Institute, Herston, QLD, Australia.
| |
Collapse
|
31
|
Ma J, Hua XY, Zheng MX, Wu JJ, Huo BB, Xing XX, Gao X, Zhang H, Xu JG. Brain Metabolic Network Redistribution in Patients with White Matter Hyperintensities on MRI Analyzed with an Individualized Index Derived from 18F-FDG-PET/MRI. Korean J Radiol 2022; 23:986-997. [PMID: 36098344 PMCID: PMC9523232 DOI: 10.3348/kjr.2022.0320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/01/2022] [Accepted: 08/02/2022] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE Whether metabolic redistribution occurs in patients with white matter hyperintensities (WMHs) on magnetic resonance imaging (MRI) is unknown. This study aimed 1) to propose a measure of the brain metabolic network for an individual patient and preliminarily apply it to identify impaired metabolic networks in patients with WMHs, and 2) to explore the clinical and imaging features of metabolic redistribution in patients with WMHs. MATERIALS AND METHODS This study included 50 patients with WMHs and 70 healthy controls (HCs) who underwent 18F-fluorodeoxyglucose-positron emission tomography/MRI. Various global property parameters according to graph theory and an individual parameter of brain metabolic network called "individual contribution index" were obtained. Parameter values were compared between the WMH and HC groups. The performance of the parameters in discriminating between the two groups was assessed using the area under the receiver operating characteristic curve (AUC). The correlation between the individual contribution index and Fazekas score was assessed, and the interaction between age and individual contribution index was determined. A generalized linear model was fitted with the individual contribution index as the dependent variable and the mean standardized uptake value (SUVmean) of nodes in the whole-brain network or seven classic functional networks as independent variables to determine their association. RESULTS The means ± standard deviations of the individual contribution index were (0.697 ± 10.9) × 10-3 and (0.0967 ± 0.0545) × 10-3 in the WMH and HC groups, respectively (p < 0.001). The AUC of the individual contribution index was 0.864 (95% confidence interval, 0.785-0.943). A positive correlation was identified between the individual contribution index and the Fazekas scores in patients with WMHs (r = 0.57, p < 0.001). Age and individual contribution index demonstrated a significant interaction effect on the Fazekas score. A significant direct association was observed between the individual contribution index and the SUVmean of the limbic network (p < 0.001). CONCLUSION The individual contribution index may demonstrate the redistribution of the brain metabolic network in patients with WMHs.
Collapse
Affiliation(s)
- Jie Ma
- Center of Rehabilitation Medicine, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xu-Yun Hua
- Department of Traumatology and Orthopedics, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Mou-Xiong Zheng
- Department of Traumatology and Orthopedics, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jia-Jia Wu
- Center of Rehabilitation Medicine, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Bei-Bei Huo
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiang-Xin Xing
- Center of Rehabilitation Medicine, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xin Gao
- Panoramic Medical Imaging Diagnostic Center, Shanghai, China
| | - Han Zhang
- School of Biomedical Engineering, ShanghaiTech University, Shanghai, China.
| | - Jian-Guang Xu
- Center of Rehabilitation Medicine, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Engineering Research Center of Traditional Chinese Medicine Intelligent Rehabilitation, Ministry of Education, Shanghai, China.
| |
Collapse
|
32
|
Replay, the default mode network and the cascaded memory systems model. Nat Rev Neurosci 2022; 23:628-640. [PMID: 35970912 DOI: 10.1038/s41583-022-00620-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/07/2022] [Indexed: 12/25/2022]
Abstract
The spontaneous replay of patterns of activity related to past experiences and memories is a striking feature of brain activity, as is the coherent activation of sets of brain areas - particularly those comprising the default mode network (DMN) - during rest. We propose that these two phenomena are strongly intertwined and that their potential functions overlap. In the 'cascaded memory systems model' that we outline here, we hypothesize that the DMN forms the backbone for the propagation of replay, mediating interactions between the hippocampus and the neocortex that enable the consolidation of new memories. The DMN may also independently ignite replay cascades, which support reactivation of older memories or high-level semantic representations. We suggest that transient cortical activations, inducing long-range correlations across the neocortex, are a key mechanism supporting a hierarchy of representations that progresses from simple percepts to semantic representations of causes and, finally, to whole episodes.
Collapse
|
33
|
Wiehler A, Branzoli F, Adanyeguh I, Mochel F, Pessiglione M. A neuro-metabolic account of why daylong cognitive work alters the control of economic decisions. Curr Biol 2022; 32:3564-3575.e5. [PMID: 35961314 DOI: 10.1016/j.cub.2022.07.010] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 05/27/2022] [Accepted: 07/06/2022] [Indexed: 12/22/2022]
Abstract
Behavioral activities that require control over automatic routines typically feel effortful and result in cognitive fatigue. Beyond subjective report, cognitive fatigue has been conceived as an inflated cost of cognitive control, objectified by more impulsive decisions. However, the origins of such control cost inflation with cognitive work are heavily debated. Here, we suggest a neuro-metabolic account: the cost would relate to the necessity of recycling potentially toxic substances accumulated during cognitive control exertion. We validated this account using magnetic resonance spectroscopy (MRS) to monitor brain metabolites throughout an approximate workday, during which two groups of participants performed either high-demand or low-demand cognitive control tasks, interleaved with economic decisions. Choice-related fatigue markers were only present in the high-demand group, with a reduction of pupil dilation during decision-making and a preference shift toward short-delay and little-effort options (a low-cost bias captured using computational modeling). At the end of the day, high-demand cognitive work resulted in higher glutamate concentration and glutamate/glutamine diffusion in a cognitive control brain region (lateral prefrontal cortex [lPFC]), relative to low-demand cognitive work and to a reference brain region (primary visual cortex [V1]). Taken together with previous fMRI data, these results support a neuro-metabolic model in which glutamate accumulation triggers a regulation mechanism that makes lPFC activation more costly, explaining why cognitive control is harder to mobilize after a strenuous workday.
Collapse
Affiliation(s)
- Antonius Wiehler
- Motivation, Brain and Behavior Lab, Paris Brain Institute (ICM), Pitié-Salpêtrière Hospital, Paris, France; Center for NeuroImaging Research (CENIR), Paris Brain Institute (ICM), Pitié-Salpêtrière Hospital, Paris, France; Sorbonne Universités, Inserm U1127, CNRS U7225, Paris, France; Department of Psychiatry, Service Hospitalo-Universitaire, Groupe Hospitalier Universitaire Paris Psychiatrie & Neurosciences, Paris, France; Sorbonne Universités, Inserm U1127, CNRS U7225, Paris, France.
| | - Francesca Branzoli
- Center for NeuroImaging Research (CENIR), Paris Brain Institute (ICM), Pitié-Salpêtrière Hospital, Paris, France; Sorbonne Universités, Inserm U1127, CNRS U7225, Paris, France
| | - Isaac Adanyeguh
- Sorbonne Universités, Inserm U1127, CNRS U7225, Paris, France
| | - Fanny Mochel
- Sorbonne Universités, Inserm U1127, CNRS U7225, Paris, France; Assistance Publique - hôpitaux de Paris, Pitié-Salpêtrière Hospital, Department of Genetics, Paris, France
| | - Mathias Pessiglione
- Motivation, Brain and Behavior Lab, Paris Brain Institute (ICM), Pitié-Salpêtrière Hospital, Paris, France; Center for NeuroImaging Research (CENIR), Paris Brain Institute (ICM), Pitié-Salpêtrière Hospital, Paris, France; Sorbonne Universités, Inserm U1127, CNRS U7225, Paris, France; Department of Psychiatry, Service Hospitalo-Universitaire, Groupe Hospitalier Universitaire Paris Psychiatrie & Neurosciences, Paris, France.
| |
Collapse
|
34
|
Chen JJ, Uthayakumar B, Hyder F. Mapping oxidative metabolism in the human brain with calibrated fMRI in health and disease. J Cereb Blood Flow Metab 2022; 42:1139-1162. [PMID: 35296177 PMCID: PMC9207484 DOI: 10.1177/0271678x221077338] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Conventional functional MRI (fMRI) with blood-oxygenation level dependent (BOLD) contrast is an important tool for mapping human brain activity non-invasively. Recent interest in quantitative fMRI has renewed the importance of oxidative neuroenergetics as reflected by cerebral metabolic rate of oxygen consumption (CMRO2) to support brain function. Dynamic CMRO2 mapping by calibrated fMRI require multi-modal measurements of BOLD signal along with cerebral blood flow (CBF) and/or volume (CBV). In human subjects this "calibration" is typically performed using a gas mixture containing small amounts of carbon dioxide and/or oxygen-enriched medical air, which are thought to produce changes in CBF (and CBV) and BOLD signal with minimal or no CMRO2 changes. However non-human studies have demonstrated that the "calibration" can also be achieved without gases, revealing good agreement between CMRO2 changes and underlying neuronal activity (e.g., multi-unit activity and local field potential). Given the simpler set-up of gas-free calibrated fMRI, there is evidence of recent clinical applications for this less intrusive direction. This up-to-date review emphasizes technological advances for such translational gas-free calibrated fMRI experiments, also covering historical progression of the calibrated fMRI field that is impacting neurological and neurodegenerative investigations of the human brain.
Collapse
Affiliation(s)
- J Jean Chen
- Medical Biophysics, University of Toronto, Toronto, Canada.,Rotman Research Institute, Baycrest, Toronto, Canada
| | - Biranavan Uthayakumar
- Medical Biophysics, University of Toronto, Toronto, Canada.,Sunnybrook Research Institute, Toronto, Canada
| | - Fahmeed Hyder
- Magnetic Resonance Research Center (MRRC), Yale University, New Haven, Connecticut, USA.,Department of Radiology, Yale University, New Haven, Connecticut, USA.,Quantitative Neuroscience with Magnetic Resonance (QNMR) Research Program, Yale University, New Haven, Connecticut, USA.,Department of Biomedical Engineering, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
35
|
Nordin K, Gorbach T, Pedersen R, Panes Lundmark V, Johansson J, Andersson M, McNulty C, Riklund K, Wåhlin A, Papenberg G, Kalpouzos G, Bäckman L, Salami A. DyNAMiC: A prospective longitudinal study of dopamine and brain connectomes: A new window into cognitive aging. J Neurosci Res 2022; 100:1296-1320. [PMID: 35293013 PMCID: PMC9313590 DOI: 10.1002/jnr.25039] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 01/18/2022] [Accepted: 02/16/2022] [Indexed: 11/07/2022]
Abstract
Concomitant exploration of structural, functional, and neurochemical brain mechanisms underlying age-related cognitive decline is crucial in promoting healthy aging. Here, we present the DopamiNe, Age, connectoMe, and Cognition (DyNAMiC) project, a multimodal, prospective 5-year longitudinal study spanning the adult human lifespan. DyNAMiC examines age-related changes in the brain's structural and functional connectome in relation to changes in dopamine D1 receptor availability (D1DR), and their associations to cognitive decline. Critically, due to the complete lack of longitudinal D1DR data, the true trajectory of one of the most age-sensitive dopamine systems remains unknown. The first DyNAMiC wave included 180 healthy participants (20-80 years). Brain imaging included magnetic resonance imaging assessing brain structure (white matter, gray matter, iron), perfusion, and function (during rest and task), and positron emission tomography (PET) with the [11 C]SCH23390 radioligand. A subsample (n = 20, >65 years) was additionally scanned with [11 C]raclopride PET measuring D2DR. Age-related variation was evident for multiple modalities, such as D1DR; D2DR, and performance across the domains of episodic memory, working memory, and perceptual speed. Initial analyses demonstrated an inverted u-shaped association between D1DR and resting-state functional connectivity across cortical network nodes, such that regions with intermediate D1DR levels showed the highest levels of nodal strength. Evident within each age group, this is the first observation of such an association across the adult lifespan, suggesting that emergent functional architecture depends on underlying D1DR systems. Taken together, DyNAMiC is the largest D1DR study worldwide, and will enable a comprehensive examination of brain mechanisms underlying age-related cognitive decline.
Collapse
Affiliation(s)
- Kristin Nordin
- Umeå Center for Functional Brain Imaging (UFBI)Umeå UniversityUmeåSweden
- Department of Integrative Medical BiologyUmeå UniversityUmeåSweden
- Wallenberg Centre for Molecular MedicineUmeå UniversityUmeåSweden
- Present address:
Aging Research CenterKarolinska Institutet & Stockholm UniversityStockholm11330Sweden
| | - Tetiana Gorbach
- Umeå Center for Functional Brain Imaging (UFBI)Umeå UniversityUmeåSweden
- Department of Integrative Medical BiologyUmeå UniversityUmeåSweden
- Wallenberg Centre for Molecular MedicineUmeå UniversityUmeåSweden
- Umeå School of Business, Economics and StatisticsUmeå UniversityUmeåSweden
| | - Robin Pedersen
- Umeå Center for Functional Brain Imaging (UFBI)Umeå UniversityUmeåSweden
- Department of Integrative Medical BiologyUmeå UniversityUmeåSweden
- Wallenberg Centre for Molecular MedicineUmeå UniversityUmeåSweden
| | - Vania Panes Lundmark
- Umeå Center for Functional Brain Imaging (UFBI)Umeå UniversityUmeåSweden
- Department of Integrative Medical BiologyUmeå UniversityUmeåSweden
| | - Jarkko Johansson
- Umeå Center for Functional Brain Imaging (UFBI)Umeå UniversityUmeåSweden
- Wallenberg Centre for Molecular MedicineUmeå UniversityUmeåSweden
- Department of Radiation SciencesUmeå UniversityUmeåSweden
| | - Micael Andersson
- Umeå Center for Functional Brain Imaging (UFBI)Umeå UniversityUmeåSweden
- Department of Integrative Medical BiologyUmeå UniversityUmeåSweden
| | - Charlotte McNulty
- Umeå Center for Functional Brain Imaging (UFBI)Umeå UniversityUmeåSweden
- Department of Integrative Medical BiologyUmeå UniversityUmeåSweden
- Wallenberg Centre for Molecular MedicineUmeå UniversityUmeåSweden
| | - Katrine Riklund
- Umeå Center for Functional Brain Imaging (UFBI)Umeå UniversityUmeåSweden
- Department of Radiation SciencesUmeå UniversityUmeåSweden
| | - Anders Wåhlin
- Umeå Center for Functional Brain Imaging (UFBI)Umeå UniversityUmeåSweden
- Department of Radiation SciencesUmeå UniversityUmeåSweden
| | - Goran Papenberg
- Aging Research CenterKarolinska Institutet & Stockholm UniversityStockholmSweden
| | - Grégoria Kalpouzos
- Aging Research CenterKarolinska Institutet & Stockholm UniversityStockholmSweden
| | - Lars Bäckman
- Aging Research CenterKarolinska Institutet & Stockholm UniversityStockholmSweden
| | - Alireza Salami
- Umeå Center for Functional Brain Imaging (UFBI)Umeå UniversityUmeåSweden
- Department of Integrative Medical BiologyUmeå UniversityUmeåSweden
- Wallenberg Centre for Molecular MedicineUmeå UniversityUmeåSweden
- Aging Research CenterKarolinska Institutet & Stockholm UniversityStockholmSweden
| |
Collapse
|
36
|
Koush Y, Rothman DL, Behar KL, de Graaf RA, Hyder F. Human brain functional MRS reveals interplay of metabolites implicated in neurotransmission and neuroenergetics. J Cereb Blood Flow Metab 2022; 42:911-934. [PMID: 35078383 PMCID: PMC9125492 DOI: 10.1177/0271678x221076570] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 12/26/2021] [Accepted: 01/05/2022] [Indexed: 01/28/2023]
Abstract
While functional MRI (fMRI) localizes brain activation and deactivation, functional MRS (fMRS) provides insights into the underlying metabolic conditions. There is much interest in measuring task-induced and resting levels of metabolites implicated in neuroenergetics (e.g., lactate, glucose, or β-hydroxybutyrate (BHB)) and neurotransmission (e.g., γ-aminobutyric acid (GABA) or pooled glutamate and glutamine (Glx)). Ultra-high magnetic field (e.g., 7T) has boosted the fMRS quantification precision, reliability, and stability of spectroscopic observations using short echo-time (TE) 1H-MRS techniques. While short TE 1H-MRS lacks sensitivity and specificity for fMRS at lower magnetic fields (e.g., 3T or 4T), most of these metabolites can also be detected by J-difference editing (JDE) 1H-MRS with longer TE to filter overlapping resonances. The 1H-MRS studies show that JDE can detect GABA, Glx, lactate, and BHB at 3T, 4T and 7T. Most recently, it has also been demonstrated that JDE 1H-MRS is capable of reliable detection of metabolic changes in different brain areas at various magnetic fields. Combining fMRS measurements with fMRI is important for understanding normal brain function, but also clinically relevant for mechanisms and/or biomarkers of neurological and neuropsychiatric disorders. We provide an up-to-date overview of fMRS research in the last three decades, both in terms of applications and technological advances. Overall the emerging fMRS techniques can be expected to contribute substantially to our understanding of metabolism for brain function and dysfunction.
Collapse
Affiliation(s)
- Yury Koush
- Magnetic Resonance Research Center, Department of Radiology & Biomedical Imaging, Yale University, New Haven, CT, USA
| | - Douglas L Rothman
- Magnetic Resonance Research Center, Department of Radiology & Biomedical Imaging, Yale University, New Haven, CT, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Kevin L Behar
- Magnetic Resonance Research Center, Department of Radiology & Biomedical Imaging, Yale University, New Haven, CT, USA
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | - Robin A de Graaf
- Magnetic Resonance Research Center, Department of Radiology & Biomedical Imaging, Yale University, New Haven, CT, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Fahmeed Hyder
- Magnetic Resonance Research Center, Department of Radiology & Biomedical Imaging, Yale University, New Haven, CT, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| |
Collapse
|
37
|
Mariman JJ, Lorca E, Biancardi C, Burgos P, Álvarez-Ruf J. Brain’s Energy After Stroke: From a Cellular Perspective Toward Behavior. Front Integr Neurosci 2022; 16:826728. [PMID: 35651830 PMCID: PMC9149581 DOI: 10.3389/fnint.2022.826728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 03/18/2022] [Indexed: 11/13/2022] Open
Abstract
Stroke is a neurological condition that impacts activity performance and quality of life for survivors. While neurological impairments after the event explain the performance of patients in specific activities, the origin of such impairments has traditionally been explained as a consequence of structural and functional damage to the nervous system. However, there are important mechanisms related to energy efficiency (trade-off between biological functions and energy consumption) at different levels that can be related to these impairments and restrictions: first, at the neuronal level, where the availability of energy resources is the initial cause of the event, as well as determines the possibilities of spontaneous recovery. Second, at the level of neural networks, where the “small world” operation of the network is compromised after the stroke, implicating a high energetic cost and inefficiency in the information transfer, which is related to the neurological recovery and clinical status. Finally, at the behavioral level, the performance limitations are related to the highest cost of energy or augmented energy expenditure during the tasks to maintain the stability of the segment, system, body, and finally, the behavior of the patients. In other words, the postural homeostasis. In this way, we intend to provide a synthetic vision of the energy impact of stroke, from the particularities of the operation of the nervous system, its implications, as one of the determinant factors in the possibilities of neurological, functional, and behavioral recovery of our patients.
Collapse
Affiliation(s)
- Juan José Mariman
- Laboratorio de Cognición y Comportamiento Sensoriomotor, Departamento de Kinesiología, Facultad de Artes y Educación Física, Universidad Metropolitana de Ciencias de la Educación, Santiago, Chile
- Departamento de Kinesiología, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Enrique Lorca
- Laboratorio de Cognición y Comportamiento Sensoriomotor, Departamento de Kinesiología, Facultad de Artes y Educación Física, Universidad Metropolitana de Ciencias de la Educación, Santiago, Chile
- Escuela de Enfermería, Facultad de Medicina, Universidad Finis Terrae, Santiago, Chile
| | - Carlo Biancardi
- Biomechanics Lab, Departamento de Ciencias Biológicas, CENUR Litoral Norte, Universidad de la República, Paysandú, Uruguay
| | - Pablo Burgos
- Departamento de Kinesiología, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Departamento de Neurociencias, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Joel Álvarez-Ruf
- Laboratorio de Cognición y Comportamiento Sensoriomotor, Departamento de Kinesiología, Facultad de Artes y Educación Física, Universidad Metropolitana de Ciencias de la Educación, Santiago, Chile
- Laboratorio de Biomecánica Clínica, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
- *Correspondence: Joel Álvarez-Ruf,
| |
Collapse
|
38
|
Geßner C, Krüger A, Folkow LP, Fehrle W, Mikkelsen B, Burmester T. Transcriptomes Suggest That Pinniped and Cetacean Brains Have a High Capacity for Aerobic Metabolism While Reducing Energy-Intensive Processes Such as Synaptic Transmission. Front Mol Neurosci 2022; 15:877349. [PMID: 35615068 PMCID: PMC9126210 DOI: 10.3389/fnmol.2022.877349] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 04/19/2022] [Indexed: 11/24/2022] Open
Abstract
The mammalian brain is characterized by high energy expenditure and small energy reserves, making it dependent on continuous vascular oxygen and nutritional supply. The brain is therefore extremely vulnerable to hypoxia. While neurons of most terrestrial mammals suffer from irreversible damage after only short periods of hypoxia, neurons of the deep-diving hooded seal (Cystophora cristata) show a remarkable hypoxia-tolerance. To identify the molecular mechanisms underlying the intrinsic hypoxia-tolerance, we excised neurons from the visual cortices of hooded seals and mice (Mus musculus) by laser capture microdissection. A comparison of the neuronal transcriptomes suggests that, compared to mice, hooded seal neurons are endowed with an enhanced aerobic metabolic capacity, a reduced synaptic transmission and an elevated antioxidant defense. Publicly available whole-tissue brain transcriptomes of the bowhead whale (Balaena mysticetus), long-finned pilot whale (Globicephala melas), minke whale (Balaenoptera acutorostrata) and killer whale (Orcinus orca), supplemented with 2 newly sequenced long-finned pilot whales, suggest that, compared to cattle (Bos taurus), the cetacean brain also displays elevated aerobic capacity and reduced synaptic transmission. We conclude that the brain energy balance of diving mammals is preserved during diving, due to reduced synaptic transmission that limits energy expenditure, while the elevated aerobic capacity allows efficient use of oxygen to restore energy balance during surfacing between dives.
Collapse
Affiliation(s)
- Cornelia Geßner
- Institute of Zoology, University of Hamburg, Hamburg, Germany
| | - Alena Krüger
- Institute of Zoology, University of Hamburg, Hamburg, Germany
| | - Lars P. Folkow
- Department of Arctic and Marine Biology, UiT The Arctic University of Norway, Tromsø, Norway
| | - Wilfrid Fehrle
- Institute of Pathology With the Sections Molecular Pathology and Cytopathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | | |
Collapse
|
39
|
Averill LA, Jiang L, Purohit P, Coppoli A, Averill CL, Roscoe J, Kelmendi B, De Feyter HM, de Graaf RA, Gueorguieva R, Sanacora G, Krystal JH, Rothman DL, Mason GF, Abdallah CG. Prefrontal Glutamate Neurotransmission in PTSD: A Novel Approach to Estimate Synaptic Strength in Vivo in Humans. CHRONIC STRESS (THOUSAND OAKS, CALIF.) 2022; 6:24705470221092734. [PMID: 35434443 PMCID: PMC9008809 DOI: 10.1177/24705470221092734] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 03/22/2022] [Indexed: 11/16/2022]
Abstract
Background Trauma and chronic stress are believed to induce and exacerbate psychopathology by disrupting glutamate synaptic strength. However, in vivo in human methods to estimate synaptic strength are limited. In this study, we established a novel putative biomarker of glutamatergic synaptic strength, termed energy-per-cycle (EPC). Then, we used EPC to investigate the role of prefrontal neurotransmission in trauma-related psychopathology. Methods Healthy controls (n = 18) and patients with posttraumatic stress (PTSD; n = 16) completed 13C-acetate magnetic resonance spectroscopy (MRS) scans to estimate prefrontal EPC, which is the ratio of neuronal energetic needs per glutamate neurotransmission cycle (VTCA/VCycle). Results Patients with PTSD were found to have 28% reduction in prefrontal EPC (t = 3.0; df = 32, P = .005). There was no effect of sex on EPC, but age was negatively associated with prefrontal EPC across groups (r = -0.46, n = 34, P = .006). Controlling for age did not affect the study results. Conclusion The feasibility and utility of estimating prefrontal EPC using 13C-acetate MRS were established. Patients with PTSD were found to have reduced prefrontal glutamatergic synaptic strength. These findings suggest that reduced glutamatergic synaptic strength may contribute to the pathophysiology of PTSD and could be targeted by new treatments.
Collapse
Affiliation(s)
- Lynnette A. Averill
- National Center for PTSD – Clinical Neurosciences Division, US
Department of Veterans Affairs, West Haven, CT, USA,Michael E. DeBakey VA Medical Center, Houston, TX, USA,Menninger Department of Psychiatry, Baylor College of Medicine, Houston, TX, USA,Department of Psychiatry, Yale University School of
Medicine, New Haven, CT, USA
| | - Lihong Jiang
- Yale Magnetic Resonance Research Center, Department of Radiology and
Biomedical Imaging, Yale University School of
Medicine, New Haven, CT, USA
| | - Prerana Purohit
- National Center for PTSD – Clinical Neurosciences Division, US
Department of Veterans Affairs, West Haven, CT, USA,Department of Psychiatry, Yale University School of
Medicine, New Haven, CT, USA
| | - Anastasia Coppoli
- Yale Magnetic Resonance Research Center, Department of Radiology and
Biomedical Imaging, Yale University School of
Medicine, New Haven, CT, USA
| | - Christopher L. Averill
- National Center for PTSD – Clinical Neurosciences Division, US
Department of Veterans Affairs, West Haven, CT, USA,Michael E. DeBakey VA Medical Center, Houston, TX, USA,Menninger Department of Psychiatry, Baylor College of Medicine, Houston, TX, USA,Department of Psychiatry, Yale University School of
Medicine, New Haven, CT, USA
| | - Jeremy Roscoe
- National Center for PTSD – Clinical Neurosciences Division, US
Department of Veterans Affairs, West Haven, CT, USA,Department of Psychiatry, Yale University School of
Medicine, New Haven, CT, USA
| | - Benjamin Kelmendi
- National Center for PTSD – Clinical Neurosciences Division, US
Department of Veterans Affairs, West Haven, CT, USA,Department of Psychiatry, Yale University School of
Medicine, New Haven, CT, USA
| | - Henk M. De Feyter
- Yale Magnetic Resonance Research Center, Department of Radiology and
Biomedical Imaging, Yale University School of
Medicine, New Haven, CT, USA
| | - Robin A de Graaf
- Yale Magnetic Resonance Research Center, Department of Radiology and
Biomedical Imaging, Yale University School of
Medicine, New Haven, CT, USA
| | - Ralitza Gueorguieva
- Department of Biostatistics, School of Public Health, Yale University School of
Medicine, New Haven, CT, USA
| | - Gerard Sanacora
- National Center for PTSD – Clinical Neurosciences Division, US
Department of Veterans Affairs, West Haven, CT, USA,Department of Psychiatry, Yale University School of
Medicine, New Haven, CT, USA
| | - John H. Krystal
- National Center for PTSD – Clinical Neurosciences Division, US
Department of Veterans Affairs, West Haven, CT, USA,Department of Psychiatry, Yale University School of
Medicine, New Haven, CT, USA
| | - Douglas L. Rothman
- Yale Magnetic Resonance Research Center, Department of Radiology and
Biomedical Imaging, Yale University School of
Medicine, New Haven, CT, USA
| | - Graeme F. Mason
- Department of Psychiatry, Yale University School of
Medicine, New Haven, CT, USA,Yale Magnetic Resonance Research Center, Department of Radiology and
Biomedical Imaging, Yale University School of
Medicine, New Haven, CT, USA
| | - Chadi G. Abdallah
- National Center for PTSD – Clinical Neurosciences Division, US
Department of Veterans Affairs, West Haven, CT, USA,Michael E. DeBakey VA Medical Center, Houston, TX, USA,Menninger Department of Psychiatry, Baylor College of Medicine, Houston, TX, USA,Department of Psychiatry, Yale University School of
Medicine, New Haven, CT, USA,Core for Advanced Magnetic Resonance Imaging (CAMRI), Baylor College of Medicine, Houston, TX, USA,Chadi G. Abdallah, Menninger Department of
Psychiatry, Baylor College of Medicine, 1977 Butler Blvd, E4187, Houston, TX
77030, USA.
| |
Collapse
|
40
|
Zarei M, Xie D, Jiang F, Bagirov A, Huang B, Raj A, Nagarajan S, Guo S. High activity and high functional connectivity are mutually exclusive in resting state zebrafish and human brains. BMC Biol 2022; 20:84. [PMID: 35410342 PMCID: PMC8996543 DOI: 10.1186/s12915-022-01286-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 03/28/2022] [Indexed: 01/21/2023] Open
Abstract
Background The structural connectivity of neurons in the brain allows active neurons to impact the physiology of target neuron types with which they are functionally connected. While the structural connectome is at the basis of functional connectome, it is the functional connectivity measured through correlations between time series of individual neurophysiological events that underlies behavioral and mental states. However, in light of the diverse neuronal cell types populating the brain and their unique connectivity properties, both neuronal activity and functional connectivity are heterogeneous across the brain, and the nature of their relationship is not clear. Here, we employ brain-wide calcium imaging at cellular resolution in larval zebrafish to understand the principles of resting state functional connectivity. Results We recorded the spontaneous activity of >12,000 neurons in the awake resting state forebrain. By classifying their activity (i.e., variances of ΔF/F across time) and functional connectivity into three levels (high, medium, low), we find that highly active neurons have low functional connections and highly connected neurons are of low activity. This finding holds true when neuronal activity and functional connectivity data are classified into five instead of three levels, and in whole brain spontaneous activity datasets. Moreover, such activity-connectivity relationship is not observed in randomly shuffled, noise-added, or simulated datasets, suggesting that it reflects an intrinsic brain network property. Intriguingly, deploying the same analytical tools on functional magnetic resonance imaging (fMRI) data from the resting state human brain, we uncover a similar relationship between activity (signal variance over time) and functional connectivity, that is, regions of high activity are non-overlapping with those of high connectivity. Conclusions We found a mutually exclusive relationship between high activity (signal variance over time) and high functional connectivity of neurons in zebrafish and human brains. These findings reveal a previously unknown and evolutionarily conserved brain organizational principle, which has implications for understanding disease states and designing artificial neuronal networks. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-022-01286-3.
Collapse
Affiliation(s)
- Mahdi Zarei
- Department of Bioengineering and Therapeutic Sciences, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, USA.
| | - Dan Xie
- Department of Bioengineering and Therapeutic Sciences, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, USA.,Department of Pharmaceutical Chemistry, University of California, San Francisco, USA
| | - Fei Jiang
- Department of Epidemiology and Biostatistics, University of California, San Francisco, USA
| | - Adil Bagirov
- Faculty of Science and Technology, Federation University Australia, Ballarat, Victoria, Australia
| | - Bo Huang
- Department of Pharmaceutical Chemistry, University of California, San Francisco, USA.,Chan Zuckerberg Biohub, San Francisco, CA, 94143, USA
| | - Ashish Raj
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, USA
| | - Srikantan Nagarajan
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, USA
| | - Su Guo
- Department of Bioengineering and Therapeutic Sciences, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, USA. .,Programs in Human Genetics and Biological Sciences, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, USA.
| |
Collapse
|
41
|
Jackson JG, Krizman E, Takano H, Lee M, Choi GH, Putt ME, Robinson MB. Activation of Glutamate Transport Increases Arteriole Diameter in v ivo: Implications for Neurovascular Coupling. Front Cell Neurosci 2022; 16:831061. [PMID: 35308116 PMCID: PMC8930833 DOI: 10.3389/fncel.2022.831061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 01/24/2022] [Indexed: 11/21/2022] Open
Abstract
In order to meet the energetic demands of cell-to-cell signaling, increases in local neuronal signaling are matched by a coordinated increase in local blood flow, termed neurovascular coupling. Multiple different signals from neurons, astrocytes, and pericytes contribute to this control of blood flow. Previously, several groups demonstrated that inhibition/ablation of glutamate transporters attenuates the neurovascular response. However, it was not determined if glutamate transporter activation was sufficient to increase blood flow. Here, we used multiphoton imaging to monitor the diameter of fluorescently labeled cortical arterioles in anesthetized C57/B6J mice. We delivered vehicle, glutamate transporter substrates, or a combination of a glutamate transporter substrate with various pharmacologic agents via a glass micropipette while simultaneously visualizing changes in arteriole diameter. We developed a novel image analysis method to automate the measurement of arteriole diameter in these time-lapse analyses. Using this workflow, we first conducted pilot experiments in which we focally applied L-glutamate, D-aspartate, or L-threo-hydroxyaspartate (L-THA) and measured arteriole responses as proof of concept. We subsequently applied the selective glutamate transport substrate L-THA (applied at concentrations that do not activate glutamate receptors). We found that L-THA evoked a significantly larger dilation than that observed with focal saline application. This response was blocked by co-application of the potent glutamate transport inhibitor, L-(2S,3S)-3-[3-[4-(trifluoromethyl)-benzoylamino]benzyloxy]-aspartate (TFB-TBOA). Conversely, we were unable to demonstrate a reduction of this effect through co-application of a cocktail of glutamate and GABA receptor antagonists. These studies provide the first direct evidence that activation of glutamate transport is sufficient to increase arteriole diameter. We explored potential downstream mechanisms mediating this transporter-mediated dilation by using a Ca2+ chelator or inhibitors of reversed-mode Na+/Ca2+ exchange, nitric oxide synthetase, or cyclo-oxygenase. The estimated effects and confidence intervals suggested some form of inhibition for a number of these inhibitors. Limitations to our study design prevented definitive conclusions with respect to these downstream inhibitors; these limitations are discussed along with possible next steps. Understanding the mechanisms that control blood flow are important because changes in blood flow/energy supply are implicated in several neurodegenerative disorders and are used as a surrogate measure of neuronal activity in widely used techniques such as functional magnetic resonance imaging (fMRI).
Collapse
Affiliation(s)
- Joshua G. Jackson
- Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Pediatrics, University of Pennsylvania, Philadelphia, PA, United States
| | - Elizabeth Krizman
- Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Pediatrics, University of Pennsylvania, Philadelphia, PA, United States
| | - Hajime Takano
- Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, United States
| | - Meredith Lee
- Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Grace H. Choi
- Department of Biostatistics, Epidemiology & Informatics, University of Pennsylvania, Philadelphia, PA, United States
| | - Mary E. Putt
- Department of Biostatistics, Epidemiology & Informatics, University of Pennsylvania, Philadelphia, PA, United States
| | - Michael B. Robinson
- Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
42
|
Finger Tapping as a Biomarker to Classify Cognitive Status in 80+-Year-Olds. J Pers Med 2022; 12:jpm12020286. [PMID: 35207773 PMCID: PMC8878665 DOI: 10.3390/jpm12020286] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 02/10/2022] [Accepted: 02/11/2022] [Indexed: 12/10/2022] Open
Abstract
This study examined the association between finger tapping and cognitive function in a group of 225 elderly participants (116 males; age 79–92 years; M = 82.5; SD = 2.4). Finger tapping was assessed in two conditions: self-selected pace and fast pace. Based on cognitive assessments, including the MoCA and CERA-NP test battery, participants were classified as cognitively healthy individuals (CHI), participants with mild cognitive impairments (MCI), and those with possible MCI (pMCI). Results of the analyses show significant differences between groups, sex and the group × sex interaction in four parameters for the self-selected pace condition and eight parameters for the fast pace condition. These parameters were used for classification by means of linear discriminant analysis (LDA). The first LDA component showed significant differences between CHI and pMCI and between CHI and MCI. Furthermore, the second LDA component showed significant differences between CHI and pMCI as well as between pMCI and MCI. Nevertheless, the algorithm correctly classified only 50% of participants, regardless of group, suggesting that tapping parameters are only partially useful for classification in early stages of dementia. We discuss these findings in terms of the diadochokinetic nature of finger tapping as associated with the age-related degeneration of cortical and subcortical motor areas.
Collapse
|
43
|
Aerobic Glycolysis: A DeOxymoron of (Neuro)Biology. Metabolites 2022; 12:metabo12010072. [PMID: 35050194 PMCID: PMC8780167 DOI: 10.3390/metabo12010072] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/09/2022] [Accepted: 01/11/2022] [Indexed: 11/17/2022] Open
Abstract
The term ‘aerobic glycolysis’ has been in use ever since Warburg conducted his research on cancer cells’ proliferation and discovered that cells use glycolysis to produce adenosine triphosphate (ATP) rather than the more efficient oxidative phosphorylation (oxphos) pathway, despite an abundance of oxygen. When measurements of glucose and oxygen utilization by activated neural tissue indicated that glucose was consumed without an accompanied oxygen consumption, the investigators who performed those measurements also termed their discovery ‘aerobic glycolysis’. Red blood cells do not contain mitochondria and, therefore, produce their energy needs via glycolysis alone. Other processes within the central nervous system (CNS) and additional organs and tissues (heart, muscle, and so on), such as ion pumps, are also known to utilize glycolysis only for the production of ATP necessary to support their function. Unfortunately, the phenomenon of ‘aerobic glycolysis’ is an enigma wherever it is encountered, thus several hypotheses have been produced in attempts to explain it; that is, whether it occurs in cancer cells, in activated neural tissue, or during postprandial or exercise metabolism. Here, it is argued that, where the phenomenon in neural tissue is concerned, the prefix ‘aerobic’ in the term ‘aerobic glycolysis’ should be removed. Data collected over the past three decades indicate that L-lactate, the end product of the glycolytic pathway, plays an essential role in brain energy metabolism, justifying the elimination of the prefix ‘aerobic’. Similar justification is probably appropriate for other tissues as well.
Collapse
|
44
|
Annunziato M, Eeza MNH, Bashirova N, Lawson A, Matysik J, Benetti D, Grosell M, Stieglitz JD, Alia A, Berry JP. An integrated systems-level model of the toxicity of brevetoxin based on high-resolution magic-angle spinning nuclear magnetic resonance (HRMAS NMR) metabolic profiling of zebrafish embryos. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 803:149858. [PMID: 34482148 DOI: 10.1016/j.scitotenv.2021.149858] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 08/18/2021] [Accepted: 08/19/2021] [Indexed: 06/13/2023]
Abstract
Brevetoxins (PbTx) are a well-recognized group of neurotoxins associated with harmful algal blooms, and specifically recurrent "Florida Red Tides," in marine waters that are linked to impacts on both human and ecosystem health including well-documented "fish kills" and marine mammal mortalities in affected coastal waters. Understanding mechanisms and pathways of PbTx toxicity enables identification of relevant biomarkers to better understand these environmental impacts, and improve monitoring efforts, in relation to this toxin. Toward a systems-level understanding of toxicity, and identification of potential biomarkers, high-resolution magic angle spinning nuclear magnetic resonance (HRMAS NMR) was utilized for metabolic profiling of zebrafish (Danio rerio) embryos, as an established toxicological model, exposed to PbTx-2 (the most common congener in marine waters). Metabolomics studies were, furthermore, complemented by an assessment of the toxicity of PbTx-2 in embryonic stages of zebrafish and mahi-mahi (Coryphaena hippurus), the latter representing an ecologically and geographically relevant marine species of fish, which identified acute embryotoxicity at environmentally relevant (i.e., parts-per-billion) concentrations in both species. HRMAS NMR analysis of intact zebrafish embryos exposed to sub-lethal concentrations of PbTx-2 afforded well-resolved spectra, and in turn, identification of 38 metabolites of which 28 were found to be significantly altered, relative to controls. Metabolites altered by PbTx-2 exposure specifically included those associated with (1) neuronal excitotoxicity, as well as associated neural homeostasis, and (2) interrelated pathways of carbohydrate and energy metabolism. Metabolomics studies, thereby, enabled a systems-level model of PbTx toxicity which integrated multiple metabolic, molecular and cellular pathways, in relation to environmentally relevant concentrations of the toxin, providing insight to not only targets and mechanisms, but potential biomarkers pertinent to environmental risk assessment and monitoring strategies.
Collapse
Affiliation(s)
- Mark Annunziato
- Institute of Environment, Department of Chemistry and Biochemistry, Florida International University, 3000 NE 151st Street, North Miami, FL 33181, USA
| | - Muhamed N H Eeza
- Institute of Medical Physics and Biophysics, University of Leipzig, Leipzig, Germany; Institute for Analytical Chemistry, University of Leipzig, Leipzig, Germany
| | - Narmin Bashirova
- Institute of Medical Physics and Biophysics, University of Leipzig, Leipzig, Germany; Institute for Analytical Chemistry, University of Leipzig, Leipzig, Germany
| | - Ariel Lawson
- Institute of Environment, Department of Chemistry and Biochemistry, Florida International University, 3000 NE 151st Street, North Miami, FL 33181, USA
| | - Jörg Matysik
- Institute for Analytical Chemistry, University of Leipzig, Leipzig, Germany
| | - Daniel Benetti
- Rosenstiel School of Marine and Atmospheric Sciences, University of Miami, Miami, FL, USA
| | - Martin Grosell
- Rosenstiel School of Marine and Atmospheric Sciences, University of Miami, Miami, FL, USA
| | - John D Stieglitz
- Rosenstiel School of Marine and Atmospheric Sciences, University of Miami, Miami, FL, USA
| | - A Alia
- Institute of Medical Physics and Biophysics, University of Leipzig, Leipzig, Germany; Leiden Institute of Chemistry, Leiden University, 2333 Leiden, the Netherlands.
| | - John P Berry
- Institute of Environment, Department of Chemistry and Biochemistry, Florida International University, 3000 NE 151st Street, North Miami, FL 33181, USA; Biomolecular Science Institute, Florida International University, 11200 SW 8th Street, Miami, FL 33199, USA.
| |
Collapse
|
45
|
Abd Elkader AM, Labib S, Taha TF, Althobaiti F, Aldhahrani A, Salem HM, Saad A, Ibrahim FM. Phytogenic compounds from avocado (Persea americana L.) extracts; antioxidant activity, amylase inhibitory activity, therapeutic potential of type 2 diabetes. Saudi J Biol Sci 2021; 29:1428-1433. [PMID: 35280591 PMCID: PMC8913550 DOI: 10.1016/j.sjbs.2021.11.031] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 10/25/2021] [Accepted: 11/17/2021] [Indexed: 12/13/2022] Open
Abstract
Diabetes is a worldwide public health disease. Currently, the most effective way to treat diabetes is to mitigate postprandial hyperglycemia by inhibiting carbohydrate hydrolysis enzymes in the digestive system. Plant extracts are rich in bioactive compounds, which can be used in diabetes treatment. This study aims to evaluate the polyphenols content in ethanolic extracts of avocado fruit and leaves (Persea americana Mill.). Additionally, their antioxidant activity using DPPH, while the inhibition ability of α-amylase was examined by reacting different amounts of the extracts with α-amylase compared to acarbose as standard inhibitor. The active compounds were detected in the extracts by LC/MS. The obtained results showed that the leaf extract recorded a significant content of total phenolic compounds compared to the fruit extract (178.95 and 145.7 mg GAE /g dry weight, respectively). The total flavonoid values ranged from 32.5 to 70.08 mg QE/g dry weight of fruit and leaves extracts, respectively. Twenty-six phytogenic compounds were detected in leaf and fruit extract by LC/MS. These compounds belong to fatty acids, sterols, triterpenes, phenolic acids, and flavonoids. The antioxidant activity of the extracts is due to the exist of phytogenic compounds, i.e., polyphenols and flavonoids. The antioxidant activity increased in a concentration dependant manner. Avocado fruit extract (1000 µg/mL) scavenged 95% of DPPḢ while leaf extract rummaged 91.03% of free radicals compared with Vit C and BHT. Additionally, higher α-amylase inhibitory activity was observed in fruit extract than the leaf extract, where the fruit and leaf extract (1000 μg/ml) inhibited the enzyme by 92.13% and 88.95%, respectively. The obtained results showed that the ethanolic extracts of avocado could have a significant impact on human health due to their high content of polyphenols.
Collapse
|
46
|
Krämer SD, Schuhmann MK, Schadt F, Israel I, Samnick S, Volkmann J, Fluri F. Changes of cerebral network activity after invasive stimulation of the mesencephalic locomotor region in a rat stroke model. Exp Neurol 2021; 347:113884. [PMID: 34624326 DOI: 10.1016/j.expneurol.2021.113884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 09/02/2021] [Accepted: 10/02/2021] [Indexed: 11/29/2022]
Abstract
Motor deficits after stroke reflect both, focal lesion and network alterations in brain regions distant from infarction. This remote network dysfunction may be caused by aberrant signals from cortical motor regions travelling via mesencephalic locomotor region (MLR) to other locomotor circuits. A method for modulating disturbed network activity is deep brain stimulation. Recently, we have shown that high frequency stimulation (HFS) of the MLR in rats has restored gait impairment after photothrombotic stroke (PTS). However, it remains elusive which cerebral regions are involved by MLR-stimulation and contribute to the improvement of locomotion. Seventeen male Wistar rats underwent photothrombotic stroke of the right sensorimotor cortex and implantation of a microelectrode into the right MLR. 2-[18F]Fluoro-2-deoxyglucose ([18F]FDG)-positron emission tomography (PET) was conducted before stroke and thereafter, on day 2 and 3 after stroke, without and with MLR-HFS, respectively. [18F]FDG-PET imaging analyses yielded a reduced glucose metabolism in the right cortico-striatal thalamic loop after PTS compared to the state before intervention. When MLR-HFS was applied after PTS, animals exhibited a significantly higher uptake of [18F]FDG in the right but not in the left cortico-striatal thalamic loop. Furthermore, MLR-HFS resulted in an elevated glucose metabolism of right-sided association cortices related to the ipsilateral sensorimotor cortex. These data support the concept of diaschisis i.e., of dysfunctional brain areas distant to a focal lesion and suggests that MLR-HFS can reverse remote network effects following PTS in rats which otherwise may result in chronic motor symptoms.
Collapse
Affiliation(s)
- Stefanie D Krämer
- Radiopharmaceutical Sciences/Biopharmacy, Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | | | - Fabian Schadt
- Department of Nuclear Medicine, Interdisciplinary PET center, University Hospital Würzburg, Würzburg, Germany
| | - Ina Israel
- Department of Nuclear Medicine, Interdisciplinary PET center, University Hospital Würzburg, Würzburg, Germany
| | - Samuel Samnick
- Department of Nuclear Medicine, Interdisciplinary PET center, University Hospital Würzburg, Würzburg, Germany
| | - Jens Volkmann
- Department of Neurology, University Hospital Würzburg, Würzburg, Germany
| | - Felix Fluri
- Department of Neurology, University Hospital Würzburg, Würzburg, Germany.
| |
Collapse
|
47
|
Peptidyl-prolyl cis-trans isomerase A participates in the selenium transport into the rat brain. J Biol Inorg Chem 2021; 26:933-945. [PMID: 34550449 DOI: 10.1007/s00775-021-01903-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 09/08/2021] [Indexed: 10/20/2022]
Abstract
Selenium, an essential micronutrient, plays vital roles in the brain. Selenoprotein P (SELENOP), a major plasma selenoprotein, is thought to transport selenium to the brain. However, Selenop-knockout mice fed a diet containing an adequate amount of selenium shows no objective neurological dysfunction which is observed in the selenium-deficient diet-fed Selenop-knockout mice. This fact indicated that selenium from low-mass selenium-source compounds can be transported by SELENOP-independent alternative pathways to the brain. In this study, to obtain the basic information about the SELENOP-independent transport pathways, we performed ex vivo experiments in which the rat brain cell membrane fraction was analyzed to find selenium-binding and/or -interactive proteins using its reactive metabolic intermediate, selenotrisulfide (STS), and MALDI TOF-mass spectrometry. Several membrane proteins with the cysteine (C) thiol were found to be reactive with STS through the thiol-exchange reaction. One of the C-containing proteins in the brain cell membrane fraction was identified as peptidyl-prolyl cis-trans isomerase (PPIase) A from tryptic fragmentation experiments and database search. Among the 4 C residues in rat PPIase A, 21st C was proved to react with STS by assessment using C mutated recombinant proteins. PPIase A is ubiquitously expressed and also associates with a variety of biologically important events such as immunomodulation, intracellular signaling, transcriptional regulation and protein trafficking. Consequently, PPIase A was thought to participate in the selenium transport into the rat brain.
Collapse
|
48
|
Sadoun A, Chauhan T, Zhang YF, Gallois Y, Marx M, Deguine O, Barone P, Strelnikov K. Intensity patterns at the peaks of brain activity in fMRI and PET are highly correlated with neural models of spatial integration. Eur J Neurosci 2021; 54:7141-7151. [PMID: 34550613 PMCID: PMC9291889 DOI: 10.1111/ejn.15469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 09/12/2021] [Accepted: 09/15/2021] [Indexed: 11/30/2022]
Abstract
Spatial integration during the brain's cognitive activity prompts changes in energy used by different neuroglial populations. Nevertheless, the organisation of such integration in 3D ‐brain activity remains undescribed from a quantitative standpoint. In response, we applied a cross‐correlation between brain activity and integrative models, which yielded a deeper understanding of information integration in functional brain mapping. We analysed four datasets obtained via fundamentally different neuroimaging techniques (functional magnetic resonance imaging [fMRI] and positron emission tomography [PET]) and found that models of spatial integration with an increasing input to each step of integration were significantly more correlated with brain activity than models with a constant input to each step of integration. In addition, marking the voxels with the maximal correlation, we found exceptionally high intersubject consistency with the initial brain activity at the peaks. Our method demonstrated for the first time that the network of peaks of brain activity is organised strictly according to the models of spatial integration independent of neuroimaging techniques. The highest correlation with models integrating an increasing at each step input suggests that brain activity reflects a network of integrative processes where the results of integration in some neuroglial populations serve as an input to other neuroglial populations.
Collapse
Affiliation(s)
- Amirouche Sadoun
- UMR 5549, Centre National de la Recherche Scientifique, Université de Toulouse 3, Toulouse, France.,Centre de Recherche Cerveau et Cognition, Université de Toulouse 3, Université Paul Sabatier, Toulouse, France
| | - Tushar Chauhan
- UMR 5549, Centre National de la Recherche Scientifique, Université de Toulouse 3, Toulouse, France.,Centre de Recherche Cerveau et Cognition, Université de Toulouse 3, Université Paul Sabatier, Toulouse, France
| | - Yi Fan Zhang
- UMR 5549, Centre National de la Recherche Scientifique, Université de Toulouse 3, Toulouse, France.,Centre de Recherche Cerveau et Cognition, Université de Toulouse 3, Université Paul Sabatier, Toulouse, France
| | - Yohan Gallois
- Service d'Oto-Rhino-Laryngologie et Oto-Neurologie, CHU de Toulouse, Université de Toulouse 3, Toulouse, France
| | - Mathieu Marx
- Service d'Oto-Rhino-Laryngologie et Oto-Neurologie, CHU de Toulouse, Université de Toulouse 3, Toulouse, France
| | - Olivier Deguine
- UMR 5549, Centre National de la Recherche Scientifique, Université de Toulouse 3, Toulouse, France.,Centre de Recherche Cerveau et Cognition, Université de Toulouse 3, Université Paul Sabatier, Toulouse, France.,Service d'Oto-Rhino-Laryngologie et Oto-Neurologie, CHU de Toulouse, Université de Toulouse 3, Toulouse, France
| | - Pascal Barone
- UMR 5549, Centre National de la Recherche Scientifique, Université de Toulouse 3, Toulouse, France.,Centre de Recherche Cerveau et Cognition, Université de Toulouse 3, Université Paul Sabatier, Toulouse, France
| | - Kuzma Strelnikov
- UMR 5549, Centre National de la Recherche Scientifique, Université de Toulouse 3, Toulouse, France.,Centre de Recherche Cerveau et Cognition, Université de Toulouse 3, Université Paul Sabatier, Toulouse, France.,Service d'Oto-Rhino-Laryngologie et Oto-Neurologie, CHU de Toulouse, Université de Toulouse 3, Toulouse, France
| |
Collapse
|
49
|
Ahmad SS, Younis K, Philippe J, Aschner M, Khan H. Strategic approaches to target the enzymes using natural compounds for the management of Alzheimer's disease: A review. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2021; 21:610-620. [PMID: 34382514 DOI: 10.2174/1871527320666210811160007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 06/21/2021] [Accepted: 07/18/2021] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disease. It is clinically characterized by memory loss and intellectual decrease, among other neurological deficits. The etiology of AD is not completely understood but includes amyloid plaques and intracellular helical filaments as well as neurofibrillary tangles with hyperphosphorylated tau protein. AD is also associated with alterations in amyloid processing genes, such as PSEN1 or PSEN2 and APP. The modulation immune system, cholesterol metabolism, and synaptic vesicle endocytosis have all been shown to remediate AD. In this review, enzymes such as AChE, BuChE, β-secretase, γ-secretase, MAO, and RAGE are discussed as potential targets for AD treatment. The aim of this review was to addresses the molecular mechanisms as well as various genetic factors in AD etiology. The use of natural compounds against these targets might be beneficial for the management of AD.
Collapse
Affiliation(s)
- Syed Sayeed Ahmad
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541. Korea
| | - Kaiser Younis
- Department of Bioengineering, Faculty of Engineering, Integral University, Lucknow. India
| | - Jeandet Philippe
- Research Unit "Induced Resistance and Plant Bioprotection", EA 4707, SFR Condorcet FR CNRS 3417, Faculty of Sciences University of Reims Champagne-Ardenne, PO Box 1039, 51687 Reims Cedex 2. France
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461. United States
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan 23200. Pakistan
| |
Collapse
|
50
|
Jiménez-Balado J, Eich TS. GABAergic dysfunction, neural network hyperactivity and memory impairments in human aging and Alzheimer's disease. Semin Cell Dev Biol 2021; 116:146-159. [PMID: 33573856 PMCID: PMC8292162 DOI: 10.1016/j.semcdb.2021.01.005] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/25/2021] [Accepted: 01/30/2021] [Indexed: 02/07/2023]
Abstract
In this review, we focus on the potential role of the γ-aminobutyric acidergic (GABAergic) system in age-related episodic memory impairments in humans, with a particular focus on Alzheimer's disease (AD). Well-established animal models have shown that GABA plays a central role in regulating and synchronizing neuronal signaling in the hippocampus, a brain area critical for episodic memory that undergoes early and significant morphologic and functional changes in the course of AD. Neuroimaging research in humans has documented hyperactivity in the hippocampus and losses of resting state functional connectivity in the Default Mode Network, a network that itself prominently includes the hippocampus-presaging episodic memory decline in individuals at-risk for AD. Apolipoprotein ε4, the highest genetic risk factor for AD, is associated with GABAergic dysfunction in animal models, and episodic memory impairments in humans. In combination, these findings suggest that GABA may be the linchpin in a complex system of factors that eventually leads to the principal clinical hallmark of AD: episodic memory loss. Here, we will review the current state of literature supporting this hypothesis. First, we will focus on the molecular and cellular basis of the GABAergic system and its role in memory and cognition. Next, we report the evidence of GABA dysregulations in AD and normal aging, both in animal models and human studies. Finally, we outline a model of GABAergic dysfunction based on the results of functional neuroimaging studies in humans, which have shown hippocampal hyperactivity to episodic memory tasks concurrent with and even preceding AD diagnosis, along with factors that may modulate this association.
Collapse
Affiliation(s)
- Joan Jiménez-Balado
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Teal S Eich
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|