1
|
Castelli S, Desideri E, Laureti L, Felice F, De Cristofaro A, Scaricamazza S, Lazzarino G, Ciriolo MR, Ciccarone F. N-acetylaspartate promotes glycolytic-to-oxidative fiber-type switch and resistance to atrophic stimuli in myotubes. Cell Death Dis 2024; 15:686. [PMID: 39300071 DOI: 10.1038/s41419-024-07047-0] [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: 04/09/2024] [Revised: 08/23/2024] [Accepted: 09/02/2024] [Indexed: 09/22/2024]
Abstract
N-acetylaspartate (NAA) is a neuronal metabolite that can be extruded in extracellular fluids and whose blood concentration increases in several neurodegenerative disorders, including amyotrophic lateral sclerosis (ALS). Aspartoacylase (ASPA) is the enzyme responsible for NAA breakdown. It is abundantly expressed in skeletal muscle and most other human tissues, but the role of NAA catabolism in the periphery is largely neglected. Here we demonstrate that NAA treatment of differentiated C2C12 muscle cells increases lipid turnover, mitochondrial biogenesis and oxidative metabolism at the expense of glycolysis. These effects were ascribed to NAA catabolism, as CRISPR/Cas9 ASPA KO cells are insensitive to NAA administration. Moreover, the metabolic switch induced by NAA was associated with an augmented resistance to atrophic stimuli. Consistently with in vitro results, SOD1-G93A ALS mice show an increase in ASPA levels in those muscles undergoing the glycolytic to oxidative switch during the disease course. The impact of NAA on the metabolism and resistance capability of myotubes supports a role for this metabolite in the phenotypical adaptations of skeletal muscle in neuromuscular disorders.
Collapse
Affiliation(s)
| | - Enrico Desideri
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Open University, Rome, Italy
| | - Leonardo Laureti
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Federica Felice
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | | | | | - Giacomo Lazzarino
- UniCamillus-Saint Camillus International University of Health and Medical Sciences, Rome, Italy
| | - Maria Rosa Ciriolo
- IRCCS San Raffaele Roma, Rome, Italy
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Fabio Ciccarone
- IRCCS San Raffaele Roma, Rome, Italy.
- Department of Biology, University of Rome Tor Vergata, Rome, Italy.
| |
Collapse
|
2
|
Sampedro-Viana A, Fernández-Rodicio S, Castillo J, Hervella P, Alonso-Alonso ML, Iglesias-Rey R. Assessment of Mannitol-Induced Chronic Blood-Brain Barrier Dysfunction In Vivo Using Magnetic Resonance. Int J Mol Sci 2024; 25:9792. [PMID: 39337280 PMCID: PMC11431755 DOI: 10.3390/ijms25189792] [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: 08/02/2024] [Revised: 09/05/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
The blood-brain barrier (BBB) is essential for protection and plays a crucial role in chronic neurological disorders like small-vessel disease and Alzheimer's disease. Its complexity poses significant challenges for effective diagnostics and treatments, highlighting the need for novel animal models and comprehensive BBB dysfunction studies. This study investigates chronic BBB dysfunction induction using osmotic disruption via mannitol in healthy adult male Sprague Dawley rats over 12 weeks. Group 1 received 1 bolus/week (2.0 g/kg), Group 2 received 3 boluses/week (1.5 g/kg), and Group 3 received 3 boluses/week (2.5 g/kg). BBB dysfunction was assessed using gadolinium (Gd) infusion and MRI to evaluate location, severity, evolution, and persistence. MR spectroscopy (MRS) examined the brain metabolism changes due to intravenous mannitol, with T2-weighted MRI assessing brain lesions. Biomarkers of neuroinflammation were analyzed in the highest mannitol dose group. Our data show chronic BBB dysfunction primarily in the cortex, hippocampus, and striatum, but not in the corpus callosum of rats under periodic mannitol dosing in groups 1 and 2. MRS identified a distinctive metabolite signature, including changes in alanine, choline, and N-acetyl aspartate in the striatum of Group 1. No significant differences were found in the serum levels of all pro- and anti-inflammatory cytokines analyzed in the high-dose Group 3. This study underscores the feasibility and implications of using osmotic disruption to model chronic BBB dysfunction, offering insights for future neuroprotection and therapeutic strategies research.
Collapse
Affiliation(s)
- Ana Sampedro-Viana
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Rúa Travesa da Choupana s/n, 15706 Santiago de Compostela, Spain
| | - Sabela Fernández-Rodicio
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Rúa Travesa da Choupana s/n, 15706 Santiago de Compostela, Spain
| | - José Castillo
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Rúa Travesa da Choupana s/n, 15706 Santiago de Compostela, Spain
| | - Pablo Hervella
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Rúa Travesa da Choupana s/n, 15706 Santiago de Compostela, Spain
| | - María Luz Alonso-Alonso
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Rúa Travesa da Choupana s/n, 15706 Santiago de Compostela, Spain
| | - Ramón Iglesias-Rey
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Rúa Travesa da Choupana s/n, 15706 Santiago de Compostela, Spain
| |
Collapse
|
3
|
Shamsesfandabadi P, Patel A, Liang Y, Shepard MJ, Wegner RE. Radiation-Induced Cognitive Decline: Challenges and Solutions. Cancer Manag Res 2024; 16:1043-1052. [PMID: 39183756 PMCID: PMC11345022 DOI: 10.2147/cmar.s441360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 08/14/2024] [Indexed: 08/27/2024] Open
Abstract
Radiation therapy, a common treatment for central nervous system cancers, can negatively impact cognitive function, resulting in radiation-induced cognitive decline (RICD). RICD involves a decline in cognitive abilities such as memory and attention, likely due to damage to brain white matter, inflammation, and oxidative stress. The multifactorial nature of RICD poses challenges including different mechanisms of injury (neurogenesis, oxidative stress and neuroinflammation, dendritic structure alterations and vascular effects) and confounding factors like advanced age, and pre-existing conditions. Despite these challenges, several potential solutions exist. Neuroprotective agents like antioxidants can mitigate radiation damage, while cognitive rehabilitation techniques such as cognitive training and memory strategies improve cognitive function. Advanced imaging techniques like magnetic resonance imaging (MRI) help identify vulnerable brain areas, and proton therapy offers precise targeting of cancer cells, sparing healthy tissue. Multidisciplinary care teams are crucial for managing RICD's cognitive and psychological effects. Personalized medicine, using genetic and molecular data, can identify high-risk patients and tailor treatments accordingly. Emerging therapies, including stem cell therapy and regenerative medicine, offer hope for repairing or replacing damaged brain tissue. Addressing RICD is vital for cancer survivors, necessitating consideration of cognitive function and provision of appropriate support and resources for those experiencing cognitive decline.
Collapse
Affiliation(s)
| | - Arpeet Patel
- Drexel University College of Medicine, Philadelphia, PA, USA
| | - Yun Liang
- Radiation Oncology department, Allegheny Health Network, Pittsburgh, PA, USA
| | - Matthew J Shepard
- Neurosurgery Department, Allegheny Health Network, Pittsburgh, PA, USA
| | - Rodney E Wegner
- Radiation Oncology department, Allegheny Health Network, Pittsburgh, PA, USA
| |
Collapse
|
4
|
Song Z, He J, Yu W, He C, Yang M, Li P, Li Z, Jian G, Cheng S. Exploring the multifaceted therapeutic mechanism of Schisanlactone E (XTS) in APP/PS1 mouse model of Alzheimer's disease through multi-omics analysis. Front Microbiol 2024; 15:1440564. [PMID: 39044957 PMCID: PMC11263214 DOI: 10.3389/fmicb.2024.1440564] [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: 05/29/2024] [Accepted: 06/28/2024] [Indexed: 07/25/2024] Open
Abstract
Background Schisanlactone E, also known as XueTongSu (XTS), is an active compound extracted from the traditional Tujia medicine Kadsura heteroclita ("XueTong"). Recent studies highlight its anti-inflammatory and antioxidant properties, yet the mechanisms of XTS's therapeutic effects on Alzheimer's disease (AD) are unclear. This study aims to elucidate the therapeutic efficacy and mechanisms of XTS in AD. Methods Ten C57BL/6 mice were assigned to the control group (NC), and twenty APP/PS1 transgenic mice were randomly divided into the model group (M) (10 mice) and the XTS treatment group (Tre) (10 mice). After an acclimatization period of 7 days, intraperitoneal injections were administered over a 60-day treatment period. The NC and M groups received saline, while the Tre group received XTS at 2 mg/kg. Learning and memory abilities were assessed using the Morris Water Maze (MWM) test. Histopathological changes were evaluated using hematoxylin and eosin (HE) and Nissl staining, and immunofluorescence was used to assess pathological products and glial cell activation. Cytokine levels (IL-1β, IL-6, TNF-α) in the hippocampus were quantified by qPCR. 16S rDNA sequencing analyzed gut microbiota metabolic alterations, and metabolomic analysis was performed on cortical samples. The KEGG database was used to analyze the regulatory mechanisms of XTS in AD treatment. Results XTS significantly improved learning and spatial memory in APP/PS1 mice and ameliorated histopathological changes, reducing Aβ plaque aggregation and glial cell activation. XTS decreased the expression of inflammatory cytokines IL-1β, IL-6, and TNF-α. It also enhanced gut microbiota diversity, notably increasing Akkermansia species, and modulated levels of metabolites such as isosakuranetin, 5-KETE, 4-methylcatechol, and sphinganine. Pathway analysis indicated that XTS regulated carbohydrate metabolism, neuroactive ligand-receptor interactions, and alanine, aspartate, and glutamate metabolism, mitigating gut microbiota dysbiosis and metabolic disturbances. Conclusion XTS ameliorates cognitive deficits, pathological changes, and inflammatory responses in APP/PS1 mice. It significantly modulates the gut microbiota, particularly increasing Akkermansia abundance, and influences levels of key metabolites in both the gut and brain. These findings suggest that XTS exerts anti-AD effects through the microbial-gut-brain axis (MGBA).
Collapse
Affiliation(s)
- Zhenyan Song
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jiawei He
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Wenjing Yu
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Chunxiang He
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Miao Yang
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Ping Li
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Ze Li
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Gonghui Jian
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Shaowu Cheng
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan University of Chinese Medicine, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
5
|
Saccaro LF, Tassone M, Tozzi F, Rutigliano G. Proton magnetic resonance spectroscopy of N-acetyl aspartate in first depressive episode and chronic major depressive disorder: A systematic review and meta-analysis. J Affect Disord 2024; 355:265-282. [PMID: 38554884 DOI: 10.1016/j.jad.2024.03.150] [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: 03/15/2023] [Revised: 03/20/2024] [Accepted: 03/25/2024] [Indexed: 04/02/2024]
Abstract
N-acetyl aspartate (NAA) is a marker of neuronal integrity and metabolism. Deficiency in neuronal plasticity and hypometabolism are implicated in Major Depressive Disorder (MDD) pathophysiology. To test if cerebral NAA concentrations decrease progressively over the MDD course, we conducted a pre-registered meta-analysis of Proton Magnetic Resonance Spectroscopy (1H-MRS) studies comparing NAA concentrations in chronic MDD (n = 1308) and first episode of depression (n = 242) patients to healthy controls (HC, n = 1242). Sixty-two studies were meta-analyzed using a random-effect model for each brain region. NAA concentrations were significantly reduced in chronic MDD compared to HC within the frontal lobe (Hedges' g = -0.330; p = 0.018), the occipital lobe (Hedges' g = -0.677; p = 0.007), thalamus (Hedges' g = -0.673; p = 0.016), and frontal (Hedges' g = -0.471; p = 0.034) and periventricular white matter (Hedges' g = -0.478; p = 0.047). We highlighted a gap of knowledge regarding NAA levels in first episode of depression patients. Sensitivity analyses indicated that antidepressant treatment may reverse NAA alterations in the frontal lobe. We highlighted field strength and correction for voxel grey matter as moderators of NAA levels detection. Future studies should assess NAA alterations in the early stages of the illness and their longitudinal progression.
Collapse
Affiliation(s)
- Luigi F Saccaro
- Department of Psychiatry, Faculty of Medicine, University of Geneva, Campus Biotech, 9 Chemin des Mines, 1202 Geneva, Switzerland; Department of Psychiatry, Geneva University Hospital, 1205 Geneva, Switzerland.
| | - Matteo Tassone
- Department of Pathology, University of Pisa, via Savi 10, 56126 Pisa, Italy
| | - Francesca Tozzi
- Bio@SNS laboratory, Scuola Normale Superiore, 56124 Pisa, Italy
| | - Grazia Rutigliano
- Department of Pathology, University of Pisa, via Savi 10, 56126 Pisa, Italy; Institute of Clinical Sciences, Imperial College London, MRI Steiner Unit, Hammersmith Hospital Campus, Du Cane Road, W12 0NN London, United Kingdom of Great Britain and Northern Ireland
| |
Collapse
|
6
|
Grønbæk-Thygesen M, Voutsinos V, Johansson KE, Schulze TK, Cagiada M, Pedersen L, Clausen L, Nariya S, Powell RL, Stein A, Fowler DM, Lindorff-Larsen K, Hartmann-Petersen R. Deep mutational scanning reveals a correlation between degradation and toxicity of thousands of aspartoacylase variants. Nat Commun 2024; 15:4026. [PMID: 38740822 DOI: 10.1038/s41467-024-48481-0] [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: 10/18/2023] [Accepted: 05/02/2024] [Indexed: 05/16/2024] Open
Abstract
Unstable proteins are prone to form non-native interactions with other proteins and thereby may become toxic. To mitigate this, destabilized proteins are targeted by the protein quality control network. Here we present systematic studies of the cytosolic aspartoacylase, ASPA, where variants are linked to Canavan disease, a lethal neurological disorder. We determine the abundance of 6152 of the 6260 ( ~ 98%) possible single amino acid substitutions and nonsense ASPA variants in human cells. Most low abundance variants are degraded through the ubiquitin-proteasome pathway and become toxic upon prolonged expression. The data correlates with predicted changes in thermodynamic stability, evolutionary conservation, and separate disease-linked variants from benign variants. Mapping of degradation signals (degrons) shows that these are often buried and the C-terminal region functions as a degron. The data can be used to interpret Canavan disease variants and provide insight into the relationship between protein stability, degradation and cell fitness.
Collapse
Affiliation(s)
- Martin Grønbæk-Thygesen
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Vasileios Voutsinos
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Kristoffer E Johansson
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Thea K Schulze
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Matteo Cagiada
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Line Pedersen
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Lene Clausen
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Snehal Nariya
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Rachel L Powell
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Amelie Stein
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Douglas M Fowler
- Department of Genome Sciences, University of Washington, Seattle, WA, USA.
- Department of Bioengineering, University of Washington, Seattle, WA, USA.
| | - Kresten Lindorff-Larsen
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Copenhagen, Denmark.
| | - Rasmus Hartmann-Petersen
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
7
|
Yang S, Zhao J, Liu X, Wang J, Gu M, Cai C, Niu H, Chen L, Hua W. Metabolomics Profiling Predicts Ventricular Arrhythmia in Patients with an Implantable Cardioverter Defibrillator. J Cardiovasc Transl Res 2024; 17:91-101. [PMID: 37556036 DOI: 10.1007/s12265-023-10413-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 07/04/2023] [Indexed: 08/10/2023]
Abstract
Implantable cardioverter defibrillators (ICDs) reduce sudden cardiac death (SCD) when patients experience life-threatening ventricular arrhythmias (LTVA). However, current strategies determining ICD patient selection and risk stratification are inefficient. We used metabolomics to assess whether dysregulated metabolites are associated with LTVA and identify potential biomarkers. Baseline plasma samples were collected from 72 patients receiving ICDs. Over a median follow-up of 524.0 days (range 239.0-705.5), LTVA occurred in 23 (31.9%) patients (22 effective ICD treatments and 1 SCD). After confounding risk factors adjustment for age, smoking, secondary prevention, and creatine kinase MB, 23 metabolites were significantly associated with LTVA. Pathway analysis revealed LTVA associations with disrupted metabolism of glycine, serine, threonine, and branched chain amino acids. Pathway enrichment analysis identified a panel of 6 metabolites that potentially predicted LTVA, with an area under the receiver operating characteristic curve of 0.8. Future studies are necessary on biological mechanisms and potential clinical use.
Collapse
Affiliation(s)
- Shengwen Yang
- Heart Center & Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Junhan Zhao
- Arrhythmia Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xi Liu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Jing Wang
- Arrhythmia Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Min Gu
- Arrhythmia Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chi Cai
- Arrhythmia Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hongxia Niu
- Arrhythmia Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liang Chen
- Department of Cardiac surgery, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Hua
- Arrhythmia Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
8
|
Weerasekera A, Knight PC, Alshelh Z, Morrissey EJ, Kim M, Zhang Y, Napadow V, Anzolin A, Torrado-Carvajal A, Edwards RR, Ratai EM, Loggia ML. Thalamic neurometabolite alterations in chronic low back pain: a common phenomenon across musculoskeletal pain conditions? Pain 2024; 165:126-134. [PMID: 37578456 PMCID: PMC10841327 DOI: 10.1097/j.pain.0000000000003002] [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: 03/22/2023] [Accepted: 06/14/2023] [Indexed: 08/15/2023]
Abstract
ABSTRACT Recently, we showed that patients with knee osteoarthritis (KOA) demonstrate alterations in the thalamic concentrations of several metabolites compared with healthy controls: higher myo-inositol (mIns), lower N-acetylaspartate (NAA), and lower choline (Cho). Here, we evaluated whether these metabolite alterations are specific to KOA or could also be observed in patients with a different musculoskeletal condition, such as chronic low back pain (cLBP). Thirty-six patients with cLBP and 20 healthy controls were scanned using 1 H-magnetic resonance spectroscopy (MRS) and a PRESS (Point RESolved Spectroscopy) sequence with voxel placement in the left thalamus. Compared with healthy controls, patients with cLBP demonstrated lower absolute concentrations of NAA ( P = 0.0005) and Cho ( P < 0.05) and higher absolute concentrations of mIns ( P = 0.01) when controlling for age, as predicted by our previous work in KOA. In contrast to our KOA study, mIns levels in this population did not significantly correlate with pain measures (eg, pain severity or duration). However, exploratory analyses revealed that NAA levels in patients were negatively correlated with the severity of sleep disturbance ( P < 0.01), which was higher in patients compared with healthy controls ( P < 0.001). Additionally, also in patients, both Cho and mIns levels were positively correlated with age ( P < 0.01 and P < 0.05, respectively). Altogether, these results suggest that thalamic metabolite changes may be common across etiologically different musculoskeletal chronic pain conditions, including cLBP and KOA, and may relate to symptoms often comorbid with chronic pain, such as sleep disturbance. The functional and clinical significance of these brain changes remains to be fully understood.
Collapse
Affiliation(s)
- Akila Weerasekera
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- Department of Radiology, Harvard Medical School, Boston, MA, USA
| | - Paulina C. Knight
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | - Zeynab Alshelh
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- Department of Radiology, Harvard Medical School, Boston, MA, USA
| | - Erin J. Morrissey
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | - Minhae Kim
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | - Yi Zhang
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Vitaly Napadow
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- Department of Radiology, Harvard Medical School, Boston, MA, USA
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Harvard Medical School, Boston, MA, USA
| | - Alessandra Anzolin
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- Department of Radiology, Harvard Medical School, Boston, MA, USA
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Harvard Medical School, Boston, MA, USA
| | - Angel Torrado-Carvajal
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- Department of Radiology, Harvard Medical School, Boston, MA, USA
- Medical Image Analysis and Biometry Laboratory, Universidad Rey Juan Carlos, Madrid, Spain
| | - Robert R. Edwards
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Eva-Maria Ratai
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- Department of Radiology, Harvard Medical School, Boston, MA, USA
| | - Marco L. Loggia
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- Department of Radiology, Harvard Medical School, Boston, MA, USA
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
9
|
Baranovicova E, Kalenska D, Kaplan P, Kovalska M, Tatarkova Z, Lehotsky J. Blood and Brain Metabolites after Cerebral Ischemia. Int J Mol Sci 2023; 24:17302. [PMID: 38139131 PMCID: PMC10743907 DOI: 10.3390/ijms242417302] [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: 10/27/2023] [Revised: 12/01/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
The study of an organism's response to cerebral ischemia at different levels is essential to understanding the mechanism of the injury and protection. A great interest is devoted to finding the links between quantitative metabolic changes and post-ischemic damage. This work aims to summarize the outcomes of the most studied metabolites in brain tissue-lactate, glutamine, GABA (4-aminobutyric acid), glutamate, and NAA (N-acetyl aspartate)-regarding their biological function in physiological conditions and their role after cerebral ischemia/reperfusion. We focused on ischemic damage and post-ischemic recovery in both experimental-including our results-as well as clinical studies. We discuss the role of blood glucose in view of the diverse impact of hyperglycemia, whether experimentally induced, caused by insulin resistance, or developed as a stress response to the cerebral ischemic event. Additionally, based on our and other studies, we analyze and critically discuss post-ischemic alterations in energy metabolites and the elevation of blood ketone bodies observed in the studies on rodents. To complete the schema, we discuss alterations in blood plasma circulating amino acids after cerebral ischemia. So far, no fundamental brain or blood metabolite(s) has been recognized as a relevant biological marker with the feasibility to determine the post-ischemic outcome or extent of ischemic damage. However, studies from our group on rats subjected to protective ischemic preconditioning showed that these animals did not develop post-ischemic hyperglycemia and manifested a decreased metabolic infringement and faster metabolomic recovery. The metabolomic approach is an additional tool for understanding damaging and/or restorative processes within the affected brain region reflected in the blood to uncover the response of the whole organism via interorgan metabolic communications to the stressful cerebral ischemic challenge.
Collapse
Affiliation(s)
- Eva Baranovicova
- Biomedical Center BioMed, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia;
| | - Dagmar Kalenska
- Department of Anatomy, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia
| | - Peter Kaplan
- Department of Medical Biochemistry, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia (Z.T.)
| | - Maria Kovalska
- Department of Histology and Embryology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia
| | - Zuzana Tatarkova
- Department of Medical Biochemistry, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia (Z.T.)
| | - Jan Lehotsky
- Department of Medical Biochemistry, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia (Z.T.)
| |
Collapse
|
10
|
de Oliveira AC, Benchimol M, Benchimol I, Chimelli L, de Oliveira-Souza R. Delayed recovery from ataxic dementia following liposuction. Neurocase 2023; 29:174-179. [PMID: 38704615 DOI: 10.1080/13554794.2024.2346982] [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: 08/24/2023] [Accepted: 04/15/2024] [Indexed: 05/06/2024]
Abstract
A 19-year-old student developed hypoventilation and cyanosis at the end of a cosmetic liposuction procedure. She was awake, but severely abulic, disoriented, and unable to stand and walk due to severe locomotor ataxia. Neuropsychological evaluation showed psychomotor slowness, and deficits in memory encoding and retrieval, and on executive, and visuospatial and visuoperceptual tests; oral comprehension and constructional praxis were spared. ¹H-MRS showed a reduction of NAA. A year later, her cognitive and neurological exam, and NAA returned to normal, and she resumed her normal life. The severity of the acute manifestations of hypoxic encephalopathy not always entail a poor prognosis.
Collapse
Affiliation(s)
- Ana Clara de Oliveira
- Graduation in Internal Medicine, The Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcos Benchimol
- Department of Internal Medicine, The Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ilana Benchimol
- Graduation in Internal Medicine, The Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leila Chimelli
- Department of Neuropathology, State Institute of the Brain, Rio de Janeiro, Brazil
| | - Ricardo de Oliveira-Souza
- Department of Neuropathology, The D'Or Institute for Research & Education, Rio de Janeiro, Brazil
- Department of Neurology, The Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
11
|
Huang Y, Chen J, Lu J, Luo H, Ying N, Dong W, Lin M, Zheng H. Transient neonatal hyperglycemia induces metabolic shifts in the rat hippocampus: a 1H NMR-based metabolomics analysis. Metab Brain Dis 2023; 38:2281-2288. [PMID: 37358727 DOI: 10.1007/s11011-023-01255-x] [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: 02/09/2023] [Accepted: 06/13/2023] [Indexed: 06/27/2023]
Abstract
Diabetes has been reported to induce brain metabolic disturbance, but the effect of transient neonatal hyperglycemia (TNH) on brain metabolism remains unclear. Herein the rats were treated with a single intraperitoneal injection of 100 µg/g body weight of streptozotocin within 12 h after birth and displayed a typical clinical characteristic of TNH. Then we used NMR-based metabolomics to examine the metabolic changes in the hippocampus between TNH and normal control (Ctrl) rats at postnatal 7 days (P7) and 21 days (P21). The results show that TNH rats had significantly increased levels of N-acetyl aspartate, glutamine, aspartate and choline in the hippocampus relative to Ctrl rats at P7. Moreover, we found that the levels of alanine, myo-inositol and choline were significantly lower in TNH rats, although their blood glucose levels have been recovered to the normal level at P21. Therefore, our results suggest that TNH may have a long-term effect on hippocampal metabolic changes mainly involving neurotransmitter metabolism and choline metabolism.
Collapse
Affiliation(s)
- Yinli Huang
- Department of Endocrinology, Pingyang Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325400, China
| | - Junli Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Jiahui Lu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Hanqi Luo
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Na Ying
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Wei Dong
- Department of Endocrinology, Pingyang Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325400, China
| | - Minjie Lin
- Department of Endocrinology, Pingyang Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325400, China
| | - Hong Zheng
- Department of Endocrinology, Pingyang Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325400, China.
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
12
|
Vaziri S, Liu H, Xie E, Ratiney H, Sdika M, Lupo JM, Xu D, Li Y. Evaluation of deep learning models for quality control of MR spectra. Front Neurosci 2023; 17:1219343. [PMID: 37706154 PMCID: PMC10495580 DOI: 10.3389/fnins.2023.1219343] [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: 05/09/2023] [Accepted: 08/10/2023] [Indexed: 09/15/2023] Open
Abstract
Purpose While 3D MR spectroscopic imaging (MRSI) provides valuable spatial metabolic information, one of the hurdles for clinical translation is its interpretation, with voxel-wise quality control (QC) as an essential and the most time-consuming step. This work evaluates the accuracy of machine learning (ML) models for automated QC filtering of individual spectra from 3D healthy control and patient datasets. Methods A total of 53 3D MRSI datasets from prior studies (30 neurological diseases, 13 brain tumors, and 10 healthy controls) were included in the study. Three ML models were evaluated: a random forest classifier (RF), a convolutional neural network (CNN), and an inception CNN (ICNN) along with two hybrid models: CNN + RF, ICNN + RF. QC labels used for training were determined manually through consensus of two MRSI experts. Normalized and cropped real-valued spectra was used as input. A cross-validation approach was used to separate datasets into training/validation/testing sets of aggregated voxels. Results All models achieved a minimum AUC of 0.964 and accuracy of 0.910. In datasets from neurological disease and controls, the CNN model produced the highest AUC (0.982), while the RF model achieved the highest AUC in patients with brain tumors (0.976). Within tumor lesions, which typically exhibit abnormal metabolism, the CNN AUC was 0.973 while that of the RF was 0.969. Data quality inference times were on the order of seconds for an entire 3D dataset, offering drastic time reduction compared to manual labeling. Conclusion ML methods accurately and rapidly performed automated QC. Results in tumors highlights the applicability to a variety of metabolic conditions.
Collapse
Affiliation(s)
- Sana Vaziri
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, United States
| | - Huawei Liu
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, United States
| | - Emily Xie
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, United States
| | - Hélène Ratiney
- Univ Lyon, INSA-Lyon, Université Claude Bernard Lyon 1, UJM-Saint Etienne, CNRS, Inserm, CREATIS UMR 5220, Lyon, France
| | - Michaël Sdika
- Univ Lyon, INSA-Lyon, Université Claude Bernard Lyon 1, UJM-Saint Etienne, CNRS, Inserm, CREATIS UMR 5220, Lyon, France
| | - Janine M. Lupo
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, United States
- UC San Francisco/UC Berkeley Graduate Program in Bioengineering, San Francisco, CA, United States
| | - Duan Xu
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, United States
- UC San Francisco/UC Berkeley Graduate Program in Bioengineering, San Francisco, CA, United States
| | - Yan Li
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
13
|
Reitlo LS, Mihailovic JM, Stensvold D, Wisløff U, Hyder F, Håberg AK. Hippocampal neurochemicals are associated with exercise group and intensity, psychological health, and general cognition in older adults. GeroScience 2023; 45:1667-1685. [PMID: 36626020 PMCID: PMC10400748 DOI: 10.1007/s11357-022-00719-9] [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: 09/08/2022] [Accepted: 12/24/2022] [Indexed: 01/11/2023] Open
Abstract
Based on the premise that physical activity/exercise impacts hippocampal structure and function, we investigated if hippocampal metabolites for neuronal viability and cell membrane density (i.e., N-acetyl aspartate (NAA), choline (Cho), creatine (Cr)) were higher in older adults performing supervised exercise compared to following national physical activity guidelines. Sixty-three participants (75.3 ± 1.9 years after 3 years of intervention) recruited from the Generation 100 study (NCT01666340_date:08.16.2012) were randomized into a supervised exercise group (SEG) performing twice weekly moderate- to high-intensity training, and a control group (CG) following national physical activity guidelines of ≥ 30-min moderate physical activity ≥ 5 days/week. Hippocampal body and head volumes and NAA, Cho, and Cr levels were acquired at 3T with magnetic resonance imaging and spectroscopic imaging. Sociodemographic data, peak oxygen uptake (VO2peak), exercise characteristics, psychological health, and cognition were recorded. General linear models were used to assess group differences and associations corrected for age, sex, education, and hippocampal volume. Both groups adhered to their training, where SEG trained at higher intensity. SEG had significantly lower NAA/Cr in hippocampal body than CG (p = 0.04). Across participants, higher training intensity was associated with lower Cho/Cr in hippocampal body (p < 0.001). Change in VO2peak, increasing VO2peak from baseline to 3 years, or VO2peak at 3 years were not associated with hippocampal neurochemicals. Lower NAA/Cr in hippocampal body was associated with poorer psychological health and slightly higher cognitive scores. Thus, following the national physical activity guidelines and not training at the highest intensity level were associated with the best neurochemical profile in the hippocampus at 3 years.
Collapse
Affiliation(s)
- Line S Reitlo
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Jelena M Mihailovic
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| | - Dorthe Stensvold
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Ulrik Wisløff
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- School of Human Movement and Nutrition Science, University of Queensland, Brisbane, Australia
| | - Fahmeed Hyder
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| | - Asta Kristine Håberg
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway.
- Department of Radiology and Nuclear Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway.
| |
Collapse
|
14
|
Strefeler A, Jan M, Quadroni M, Teav T, Rosenberg N, Chatton JY, Guex N, Gallart-Ayala H, Ivanisevic J. Molecular insights into sex-specific metabolic alterations in Alzheimer's mouse brain using multi-omics approach. Alzheimers Res Ther 2023; 15:8. [PMID: 36624525 PMCID: PMC9827669 DOI: 10.1186/s13195-023-01162-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 01/02/2023] [Indexed: 01/11/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is a progressive neurodegenerative disorder that is characterized by altered cellular metabolism in the brain. Several of these alterations have been found to be exacerbated in females, known to be disproportionately affected by AD. We aimed to unravel metabolic alterations in AD at the metabolic pathway level and evaluate whether they are sex-specific through integrative metabolomic, lipidomic, and proteomic analysis of mouse brain tissue. METHODS We analyzed male and female triple-transgenic mouse whole brain tissue by untargeted mass spectrometry-based methods to obtain a molecular signature consisting of polar metabolite, complex lipid, and protein data. These data were analyzed using multi-omics factor analysis. Pathway-level alterations were identified through joint pathway enrichment analysis or by separately evaluating lipid ontology and known proteins related to lipid metabolism. RESULTS Our analysis revealed significant AD-associated and in part sex-specific alterations across the molecular signature. Sex-dependent alterations were identified in GABA synthesis, arginine biosynthesis, and in alanine, aspartate, and glutamate metabolism. AD-associated alterations involving lipids were also found in the fatty acid elongation pathway and lysophospholipid metabolism, with a significant sex-specific effect for the latter. CONCLUSIONS Through multi-omics analysis, we report AD-associated and sex-specific metabolic alterations in the AD brain involving lysophospholipid and amino acid metabolism. These findings contribute to the characterization of the AD phenotype at the molecular level while considering the effect of sex, an overlooked yet determinant metabolic variable.
Collapse
Affiliation(s)
- Abigail Strefeler
- grid.9851.50000 0001 2165 4204Metabolomics Unit, Faculty of Biology and Medicine, University de Lausanne, Lausanne, Switzerland
| | - Maxime Jan
- grid.9851.50000 0001 2165 4204Bioinformatics Competence Center, Faculty of Biology and Medicine, University de Lausanne, Lausanne, Switzerland
| | - Manfredo Quadroni
- grid.9851.50000 0001 2165 4204Protein Analysis Facility, Faculty of Biology and Medicine, University de Lausanne, Lausanne, Switzerland
| | - Tony Teav
- grid.9851.50000 0001 2165 4204Metabolomics Unit, Faculty of Biology and Medicine, University de Lausanne, Lausanne, Switzerland
| | - Nadia Rosenberg
- grid.9851.50000 0001 2165 4204Department of Fundamental Neurosciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Jean-Yves Chatton
- grid.9851.50000 0001 2165 4204Department of Fundamental Neurosciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Nicolas Guex
- grid.9851.50000 0001 2165 4204Bioinformatics Competence Center, Faculty of Biology and Medicine, University de Lausanne, Lausanne, Switzerland
| | - Hector Gallart-Ayala
- grid.9851.50000 0001 2165 4204Metabolomics Unit, Faculty of Biology and Medicine, University de Lausanne, Lausanne, Switzerland
| | - Julijana Ivanisevic
- grid.9851.50000 0001 2165 4204Metabolomics Unit, Faculty of Biology and Medicine, University de Lausanne, Lausanne, Switzerland
| |
Collapse
|
15
|
Talati A, van Dijk MT, Pan L, Hao X, Wang Z, Gameroff M, Dong Z, Kayser J, Shankman S, Wickramaratne PJ, Posner J, Weissman MM. Putamen Structure and Function in Familial Risk for Depression: A Multimodal Imaging Study. Biol Psychiatry 2022; 92:932-941. [PMID: 36038379 PMCID: PMC9872322 DOI: 10.1016/j.biopsych.2022.06.035] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 06/14/2022] [Accepted: 06/16/2022] [Indexed: 01/26/2023]
Abstract
BACKGROUND The putamen has been implicated in depressive disorders, but how its structure and function increase depression risk is not clearly understood. Here, we examined how putamen volume, neuronal density, and mood-modulated functional activity relate to family history and prospective course of depression. METHODS The study includes 115 second- and third-generation offspring at high or low risk for depression based on the presence or absence of major depressive disorder in the first generation. Offspring were followed longitudinally using semistructured clinical interviews blinded to their familial risk; putamen structure, neuronal integrity, and functional activation were indexed by structural magnetic resonance imaging (MRI), proton magnetic resonance spectroscopy (N-acetylaspartate/creatine ratio), and functional MRI activity modulated by valence and arousal components of a mood induction task, respectively. RESULTS After adjusting for covariates, the high-risk individuals had lower putamen volume (standardized betas, β-left = -0.17, β-right = -0.15, ps = .002), N-acetylaspartate/creatine ratio (β-left= -0.40, β-right= -0.37, ps < .0001), and activation modulated by valence (β-left = -0.22, β-right = -0.27, ps < .05) than low-risk individuals. Volume differences were greater at younger ages, and N-acetylaspartate/creatine ratio differences were greater at older ages. Lower putamen volume also predicted major depressive disorder episodes up to 8 years after the scan (β-left = -0.72, p = .013; β-right = -0.83, p = .037). Magnetic resonance spectroscopy and task functional MRI measures were modestly correlated (0.27 ≤ r ≤ 0.33). CONCLUSIONS Findings demonstrate abnormalities in putamen structure and function in individuals at high risk for major depressive disorder. Future studies should focus on this region as a potential biomarker for depressive illness, noting meanwhile that differences attributable to family history may peak at different ages based on which MRI modality is being used to assay them.
Collapse
Affiliation(s)
- Ardesheer Talati
- Department of Psychiatry, Columbia University Irving Medical Center and Vagelos College of Physicians and Surgeons, Columbia University, New York, New York; Division of Translational Epidemiology, New York State Psychiatric Institute, New York, New York.
| | - Milenna T van Dijk
- Department of Psychiatry, Columbia University Irving Medical Center and Vagelos College of Physicians and Surgeons, Columbia University, New York, New York; Division of Translational Epidemiology, New York State Psychiatric Institute, New York, New York
| | - Lifang Pan
- Department of Psychiatry, Columbia University Irving Medical Center and Vagelos College of Physicians and Surgeons, Columbia University, New York, New York; Division of Translational Epidemiology, New York State Psychiatric Institute, New York, New York
| | - Xuejun Hao
- Department of Psychiatry, Columbia University Irving Medical Center and Vagelos College of Physicians and Surgeons, Columbia University, New York, New York; Division of Translational Epidemiology, New York State Psychiatric Institute, New York, New York
| | - Zhishun Wang
- Department of Psychiatry, Columbia University Irving Medical Center and Vagelos College of Physicians and Surgeons, Columbia University, New York, New York; Division of Translational Imaging, New York State Psychiatric Institute, New York, New York
| | - Marc Gameroff
- Department of Psychiatry, Columbia University Irving Medical Center and Vagelos College of Physicians and Surgeons, Columbia University, New York, New York; Division of Translational Epidemiology, New York State Psychiatric Institute, New York, New York
| | - Zhengchao Dong
- Department of Psychiatry, Columbia University Irving Medical Center and Vagelos College of Physicians and Surgeons, Columbia University, New York, New York; Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, New York
| | - Jürgen Kayser
- Department of Psychiatry, Columbia University Irving Medical Center and Vagelos College of Physicians and Surgeons, Columbia University, New York, New York; Division of Translational Epidemiology, New York State Psychiatric Institute, New York, New York
| | - Stewart Shankman
- Department of Psychiatry and Behavioral Sciences, Northwestern University, Chicago, Illinois
| | - Priya J Wickramaratne
- Department of Psychiatry, Columbia University Irving Medical Center and Vagelos College of Physicians and Surgeons, Columbia University, New York, New York; Division of Translational Epidemiology, New York State Psychiatric Institute, New York, New York; Department of Biostatistics, Columbia University Mailman School of Public Health, New York, New York
| | - Jonathan Posner
- Department of Psychiatry, Duke University, Durham, North Carolina
| | - Myrna M Weissman
- Department of Psychiatry, Columbia University Irving Medical Center and Vagelos College of Physicians and Surgeons, Columbia University, New York, New York; Division of Translational Epidemiology, New York State Psychiatric Institute, New York, New York; Department of Epidemiology, Columbia University Mailman School of Public Health, New York, New York
| |
Collapse
|
16
|
Frank SM, Becker M, Qi A, Geiger P, Frank UI, Rosedahl LA, Malloni WM, Sasaki Y, Greenlee MW, Watanabe T. Efficient learning in children with rapid GABA boosting during and after training. Curr Biol 2022; 32:5022-5030.e7. [PMID: 36384138 DOI: 10.1016/j.cub.2022.10.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 09/10/2022] [Accepted: 10/11/2022] [Indexed: 11/17/2022]
Abstract
It is generally thought that children learn more efficiently than adults. One way to accomplish this is to have learning rapidly stabilized such that it is not interfered with by subsequent learning. Although γ-aminobutyric acid (GABA) plays an important role in stabilization, it has been reported that GABAergic inhibitory processing is not fully matured yet in children compared with adults. Does this finding indicate that more efficient learning in children is not due to more rapid stabilization? Here, we measured the concentration of GABA in early visual cortical areas in a time-resolved fashion before, during, and after visual perceptual learning (VPL) within subjects using functional MRS (fMRS) and then compared the concentrations between children (8 to 11 years old) and adults (18 to 35 years old). We found that children exhibited a rapid boost of GABA during visual training that persisted after training ended, whereas the concentration of GABA in adults remained unchanged. Moreover, behavioral experiments showed that children exhibited rapid development of resilience to retrograde interference, which indicates that children stabilize VPL much faster than adults. These results together suggest that inhibitory processing in children's brains is more dynamic and adapts more quickly to stabilize learning than in adults, making learning more efficient in children.
Collapse
Affiliation(s)
- Sebastian M Frank
- University of Regensburg, Institute for Experimental Psychology, Universitätsstraße 31, 93053 Regensburg, Germany; Brown University, Department of Cognitive, Linguistic and Psychological Sciences, 190 Thayer Street, Providence, RI 02912, USA.
| | - Markus Becker
- University of Regensburg, Institute for Experimental Psychology, Universitätsstraße 31, 93053 Regensburg, Germany
| | - Andrea Qi
- Brown University, Department of Cognitive, Linguistic and Psychological Sciences, 190 Thayer Street, Providence, RI 02912, USA
| | - Patricia Geiger
- University of Regensburg, Institute for Experimental Psychology, Universitätsstraße 31, 93053 Regensburg, Germany
| | - Ulrike I Frank
- University of Regensburg, Institute for Experimental Psychology, Universitätsstraße 31, 93053 Regensburg, Germany
| | - Luke A Rosedahl
- Brown University, Department of Cognitive, Linguistic and Psychological Sciences, 190 Thayer Street, Providence, RI 02912, USA
| | - Wilhelm M Malloni
- University of Regensburg, Institute for Experimental Psychology, Universitätsstraße 31, 93053 Regensburg, Germany
| | - Yuka Sasaki
- Brown University, Department of Cognitive, Linguistic and Psychological Sciences, 190 Thayer Street, Providence, RI 02912, USA
| | - Mark W Greenlee
- University of Regensburg, Institute for Experimental Psychology, Universitätsstraße 31, 93053 Regensburg, Germany
| | - Takeo Watanabe
- Brown University, Department of Cognitive, Linguistic and Psychological Sciences, 190 Thayer Street, Providence, RI 02912, USA.
| |
Collapse
|
17
|
Baranovicova E, Hnilicova P, Kalenska D, Kaplan P, Kovalska M, Tatarkova Z, Tomascova A, Lehotsky J. Metabolic Changes Induced by Cerebral Ischemia, the Effect of Ischemic Preconditioning, and Hyperhomocysteinemia. Biomolecules 2022; 12:554. [PMID: 35454143 PMCID: PMC9032340 DOI: 10.3390/biom12040554] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/01/2022] [Accepted: 04/07/2022] [Indexed: 12/12/2022] Open
Abstract
1H Nuclear Magnetic Resonance (NMR) metabolomics is one of the fundamental tools in the fast-developing metabolomics field. It identifies and quantifies the most abundant metabolites, alterations of which can describe energy metabolism, activated immune response, protein synthesis and catabolism, neurotransmission, and many other factors. This paper summarizes our results of the 1H NMR metabolomics approach to characterize the distribution of relevant metabolites and their alterations induced by cerebral ischemic injury or its combination with hyperhomocysteinemia in the affected tissue and blood plasma in rodents. A decrease in the neurotransmitter pool in the brain tissue likely follows the disordered feasibility of post-ischemic neurotransmission. This decline is balanced by the increased tissue glutamine level with the detected impact on neuronal health. The ischemic injury was also manifested in the metabolomic alterations in blood plasma with the decreased levels of glycolytic intermediates, as well as a post-ischemically induced ketosis-like state with increased plasma ketone bodies. As the 3-hydroxybutyrate can act as a likely neuroprotectant, its post-ischemic increase can suggest its supporting role in balancing ischemic metabolic dysregulation. Furthermore, the 1H NMR approach revealed post-ischemically increased 3-hydroxybutyrate in the remote organs, such as the liver and heart, as well as decreased myocardial glutamate. Ischemic preconditioning, as a proposed protective strategy, was manifested in a lower extent of metabolomic changes and/or their faster recovery in a longitudinal study. The paper also summarizes the pre- and post-ischemic metabolomic changes in the rat hyperhomocysteinemic models. Animals are challenged with hyperglycemia and ketosis-like state. A decrease in several amino acids in plasma follows the onset and progression of hippocampal neuropathology when combined with ischemic injury. The 1H NMR metabolomics approach also offers a high potential for metabolites in discriminatory analysis in the search for potential biomarkers of ischemic injury. Based on our results and the literature data, this paper presents valuable findings applicable in clinical studies and suggests the precaution of a high protein diet, especially foods which are high in Met content and low in B vitamins, in the possible risk of human cerebrovascular neuropathology.
Collapse
Affiliation(s)
- Eva Baranovicova
- Biomedical Center BioMed, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia; (E.B.); (P.H.); (A.T.)
| | - Petra Hnilicova
- Biomedical Center BioMed, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia; (E.B.); (P.H.); (A.T.)
| | - Dagmar Kalenska
- Department of Anatomy, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia;
| | - Peter Kaplan
- Department of Medical Biochemistry, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia; (P.K.); (Z.T.)
| | - Maria Kovalska
- Department of Histology and Embryology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia;
| | - Zuzana Tatarkova
- Department of Medical Biochemistry, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia; (P.K.); (Z.T.)
| | - Anna Tomascova
- Biomedical Center BioMed, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia; (E.B.); (P.H.); (A.T.)
| | - Jan Lehotsky
- Department of Medical Biochemistry, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01 Martin, Slovakia; (P.K.); (Z.T.)
| |
Collapse
|
18
|
Effects of Tai Chi on biomarkers and their implication to neurorehabilitation – a systemic review. Eur J Integr Med 2022. [DOI: 10.1016/j.eujim.2021.101391] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
19
|
Therapeutic treatment with the anti-inflammatory drug candidate MW151 may partially reduce memory impairment and normalizes hippocampal metabolic markers in a mouse model of comorbid amyloid and vascular pathology. PLoS One 2022; 17:e0262474. [PMID: 35081152 PMCID: PMC8791470 DOI: 10.1371/journal.pone.0262474] [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: 05/07/2021] [Accepted: 12/24/2021] [Indexed: 12/03/2022] Open
Abstract
Alzheimer’s disease (AD) is the leading cause of dementia in the elderly, but therapeutic options are lacking. Despite long being able to effectively treat the ill-effects of pathology present in various rodent models of AD, translation of these strategies to the clinic has so far been disappointing. One potential contributor to this situation is the fact that the vast majority of AD patients have other dementia-contributing comorbid pathologies, the most common of which are vascular in nature. This situation is modeled relatively infrequently in basic AD research, and almost never in preclinical studies. As part of our efforts to develop small molecule, anti-inflammatory therapeutics for neurological injury and disease, we have recently been exploring potentially promising treatments in preclinical multi-morbidity contexts. In the present study, we generated a mouse model of mixed amyloid and hyperhomocysteinemia (HHcy) pathology in which to test the efficacy of one of our anti-inflammatory compounds, MW151. HHcy can cause cerebrovascular damage and is an independent risk factor for both AD dementia and vascular contributions to cognitive impairment and dementia. We found that MW151 was able to partially rescue hippocampal-dependent spatial memory and learning deficits in this comorbidity context, and further, that the benefit is associated with a normalization of hippocampal metabolites detectable via magnetic resonance spectroscopy. These findings provide evidence that MW151 in particular, and potentially anti-inflammatory treatment more generally, may be beneficial in AD patients with comorbid vascular pathology.
Collapse
|
20
|
Moss HG, Wolf LG, Coker-Bolt P, Ramakrishnan V, Aljuhani T, Yazdani M, Brown TR, Jensen JH, Jenkins DD. Quantitative Diffusion and Spectroscopic Neuroimaging Combined with a Novel Early-Developmental Assessment Improves Models for 1-Year Developmental Outcomes. AJNR Am J Neuroradiol 2022; 43:139-145. [PMID: 34949592 PMCID: PMC8757543 DOI: 10.3174/ajnr.a7370] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 09/27/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND AND PURPOSE Preterm infants are at risk for overt and silent CNS injury, with developmental consequences that are difficult to predict. The novel Specific Test of Early Infant Motor Performance, administered in preterm infants at term age, is indicative of later developmental gross motor and cognitive scores at 12 months. Here, we assessed whether functional performance on this early assessment correlates with CNS integrity via MR spectroscopy or diffusional kurtosis imaging and whether these quantitative neuroimaging methods improve predictions for future 12-month developmental scores. MATERIALS AND METHODS MR spectroscopy and quantitative diffusion MR imaging data were acquired in preterm infants (n = 16) at term. Testing was performed at term and 3 months using the Specific Test of Early Infant Motor Performance and the Bayley Scales of Infant and Toddler Development, Third Edition, at 12 months. We modeled the relationship of MR spectroscopy and diffusion MR imaging data with both test scores via multiple linear regression. RESULTS MR spectroscopy NAA ratios at a TE of 270 ms in the frontal WM and basal ganglia and kurtosis metrics in major WM tracts correlated strongly with total Specific Test of Early Infant Motor Performance scores. The addition of MR spectroscopy and diffusion separately improved the functional predictions of 12-month outcomes. CONCLUSIONS Microstructural integrity of the major WM tracts and metabolism in the basal ganglia and frontal WM strongly correlate with early developmental performance, suggesting that the Specific Test of Early Infant Motor Performance reflects CNS integrity after preterm birth. This study demonstrates that combining quantitative neuroimaging and early functional movement improves the prediction of 12-month outcomes in premature infants.
Collapse
Affiliation(s)
- H G Moss
- From the Department of Neuroscience (H.G.M., J.H.J.)
- Center for Biomedical Imaging (H.G.M., T.R.B., J.H.J., D.D.J.)
| | - L G Wolf
- Department of Pediatrics (L.G.W., D.D.J.)
| | - P Coker-Bolt
- Division of Occupational Therapy (P.C.-B., T.A.), College of Health Sciences
| | | | - T Aljuhani
- Division of Occupational Therapy (P.C.-B., T.A.), College of Health Sciences
- Division of Public Health Sciences (V.R., T.A.)
| | - M Yazdani
- Department of Radiology and Radiological Science (M.Y., T.R.B., J.H.J.), Medical University of South Carolina, Charleston, South Carolina
| | - T R Brown
- Center for Biomedical Imaging (H.G.M., T.R.B., J.H.J., D.D.J.)
- Department of Radiology and Radiological Science (M.Y., T.R.B., J.H.J.), Medical University of South Carolina, Charleston, South Carolina
| | - J H Jensen
- From the Department of Neuroscience (H.G.M., J.H.J.)
- Center for Biomedical Imaging (H.G.M., T.R.B., J.H.J., D.D.J.)
- Department of Radiology and Radiological Science (M.Y., T.R.B., J.H.J.), Medical University of South Carolina, Charleston, South Carolina
| | - D D Jenkins
- Center for Biomedical Imaging (H.G.M., T.R.B., J.H.J., D.D.J.)
- Department of Pediatrics (L.G.W., D.D.J.)
| |
Collapse
|
21
|
Luna R, Talanki Manjunatha R, Bollu B, Jhaveri S, Avanthika C, Reddy N, Saha T, Gandhi F. A Comprehensive Review of Neuronal Changes in Diabetics. Cureus 2021; 13:e19142. [PMID: 34868777 PMCID: PMC8628358 DOI: 10.7759/cureus.19142] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/30/2021] [Indexed: 12/11/2022] Open
Abstract
There has been an exponential rise in diabetes mellitus (DM) cases on a global scale. Diabetes affects almost every system of the body, and the nervous system is no exception. Although the brain is dependent on glucose, providing it with the energy required for optimal functionality, glucose also plays a key role in the regulation of oxidative stress, cell death, among others, which furthermore contribute to the pathophysiology of neurological disorders. The variety of biochemical processes engaged in this process is only matched by the multitude of clinical consequences resulting from it. The wide-ranging effects on the central and peripheral nervous system include, but are not limited to axonopathies, neurodegenerative diseases, neurovascular diseases, and general cognitive impairment. All language search was conducted on MEDLINE, COCHRANE, EMBASE, and GOOGLE SCHOLAR till September 2021. The following search strings and Medical Subject Headings (MeSH terms) were used: "Diabetes Mellitus," "CNS," "Diabetic Neuropathy," and "Insulin." We explored the literature on diabetic neuropathy, covering its epidemiology, pathophysiology with the respective molecular pathways, clinical consequences with a special focus on the central nervous system and finally, measures to prevent and treat neuronal changes. Diabetes is slowly becoming an epidemic, rapidly increasing the clinical burden on account of its wide-ranging complications. This review focuses on the neuronal changes occurring in diabetes such as the impact of hyperglycemia on brain function and structure, its association with various neurological disorders, and a few diabetes-induced peripheral neuropathic changes. It is an attempt to summarize the relevant literature about neuronal consequences of DM as treatment options available today are mostly focused on achieving better glycemic control; further research on novel treatment options to prevent or delay the progression of neuronal changes is still needed.
Collapse
Affiliation(s)
- Rudy Luna
- Neurofisiología, Instituto Nacional de Neurologia y Neurocirugia, CDMX, MEX
| | | | | | | | - Chaithanya Avanthika
- Medicine and Surgery; Pediatrics, Karnataka Institute of Medical Sciences, Hubli, IND
| | - Nikhil Reddy
- Internal Medicine, Kamineni Academy of Medical Science and Research Centre, Hyderabad, IND
| | - Tias Saha
- Internal Medicine, Diabetic Association Medical College, Faridpur, BGD
| | - Fenil Gandhi
- Medicine, Shree Krishna Hospital, Anand, IND
- Research Project Associate, Memorial Sloan Kettering Cancer Center, New York, USA
| |
Collapse
|
22
|
Oxidative Dysregulation in Early Life Stress and Posttraumatic Stress Disorder: A Comprehensive Review. Brain Sci 2021; 11:brainsci11060723. [PMID: 34072322 PMCID: PMC8228973 DOI: 10.3390/brainsci11060723] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/24/2021] [Accepted: 05/26/2021] [Indexed: 12/30/2022] Open
Abstract
Traumatic stress may chronically affect master homeostatic systems at the crossroads of peripheral and central susceptibility pathways and lead to the biological embedment of trauma-related allostatic trajectories through neurobiological alterations even decades later. Lately, there has been an exponential knowledge growth concerning the effect of traumatic stress on oxidative components and redox-state homeostasis. This extensive review encompasses a detailed description of the oxidative cascade components along with their physiological and pathophysiological functions and a systematic presentation of both preclinical and clinical, genetic and epigenetic human findings on trauma-related oxidative stress (OXS), followed by a substantial synthesis of the involved oxidative cascades into specific and functional, trauma-related pathways. The bulk of the evidence suggests an imbalance of pro-/anti-oxidative mechanisms under conditions of traumatic stress, respectively leading to a systemic oxidative dysregulation accompanied by toxic oxidation byproducts. Yet, there is substantial heterogeneity in findings probably relative to confounding, trauma-related parameters, as well as to the equivocal directionality of not only the involved oxidative mechanisms but other homeostatic ones. Accordingly, we also discuss the trauma-related OXS findings within the broader spectrum of systemic interactions with other major influencing systems, such as inflammation, the hypothalamic-pituitary-adrenal axis, and the circadian system. We intend to demonstrate the inherent complexity of all the systems involved, but also put forth associated caveats in the implementation and interpretation of OXS findings in trauma-related research and promote their comprehension within a broader context.
Collapse
|
23
|
Bangma DF, Tucha O, Tucha L, De Deyn PP, Koerts J. How well do people living with neurodegenerative diseases manage their finances? A meta-analysis and systematic review on the capacity to make financial decisions in people living with neurodegenerative diseases. Neurosci Biobehav Rev 2021; 127:709-739. [PMID: 34058557 DOI: 10.1016/j.neubiorev.2021.05.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 05/18/2021] [Accepted: 05/20/2021] [Indexed: 12/13/2022]
Abstract
Self and proxy reported questionnaires indicate that people living with a neurodegenerative disease (NDD) have more difficulties with financial decision-making (FDM) than healthy controls. Self-reports, however, rely on adequate insight into everyday functioning and might, therefore, be less reliable. The present study provides a comprehensive overview and meta-analysis of studies evaluating FDM in people living with an NDD. For this, the reliability of performance-based tests to consistently identify FDM difficulties in people living with an NDD compared to healthy controls is evaluated. Furthermore, the associations between FDM and disease severity, performances on standard measures of cognition and demographics are evaluated. All 47 included articles, consistently reported lower performances on performance-based FDM tests of people living with an NDD (including Alzheimer's disease, mild cognitive impairment, frontotemporal dementia, Parkinson's disease, multiple sclerosis or Huntington's disease) compared to healthy controls. The majority of studies, however, focused on Alzheimer's disease and mild cognitive impairment (k = 38). FDM performance appears to be related to cognitive decline, specifically in working memory, processing speed and numeracy.
Collapse
Affiliation(s)
- Dorien F Bangma
- Department of Clinical and Developmental Neuropsychology, University of Groningen, Groningen, the Netherlands; Department of Psychology, University of Amsterdam, Amsterdam, the Netherlands
| | - Oliver Tucha
- Department of Clinical and Developmental Neuropsychology, University of Groningen, Groningen, the Netherlands; Department of Department of Psychiatry and Psychotherapy, University Medical Center Rostock, Rostock, Germany; Department of Psychology, Maynooth University, National University of Ireland, Maynooth, Ireland
| | - Lara Tucha
- Department of Department of Psychiatry and Psychotherapy, University Medical Center Rostock, Rostock, Germany
| | - Peter P De Deyn
- Department of Neurology and Alzheimer Center Groningen, University Medical Center Groningen, Groningen, the Netherlands; Institute Born-Bunge, University of Antwerp, Antwerp, Belgium; Department of Neurology and Memory Clinic, Middelheim General Hospital (ZNA), Antwerp, Belgium
| | - Janneke Koerts
- Department of Clinical and Developmental Neuropsychology, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
24
|
Bahadur Patel A, Veeraiah P, Shameem M, Mahesh Kumar J, Saba K. Impaired GABAergic and glutamatergic neurometabolic activity in aged mice brain as measured by 1 H-[ 13 C]-NMR spectroscopy. FASEB J 2021; 35:e21321. [PMID: 33543543 DOI: 10.1096/fj.202001704rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 12/07/2020] [Accepted: 12/14/2020] [Indexed: 01/04/2023]
Abstract
Healthy aging is associated with a decline in cognitive function, and is a major risk factor for many neurodegenerative diseases. Although, there are several evidence that brain mitochondrial function is altered with aging its significance at the cellular level is elusive. In this study, we have investigated mitochondrial TCA cycle and neurotransmitter cycle fluxes associated with glutamatergic, GABAergic neurons and astroglia in the cerebral cortex and hippocampus of young (6 months) and aged (24 months) C57BL6 mice by using 1 H-[13 C]-NMR spectroscopy together with timed infusion of 13 C-labeled glucose and acetate. The ratio VCyc /VTCA was determined from a steady-state [2-13 C]acetate experiment. Metabolic fluxes were obtained by fitting a three-compartment metabolic model to 13 C turnover of amino acids from glucose. Levels of glutamate, aspartate and taurine were reduced in the cerebral cortex, while glutamine and choline were elevated in the hippocampus of aged mice. Interestingly, the rate of acetate oxidation increased in the cerebral cortex, while the flux of mitochondrial TCA cycle of glutamatergic neurons decreased in the cerebral cortex (P < .0001) and hippocampus (P = .025) of aged mice. The glutamate-glutamine neurotransmitter cycle flux was reduced in the cerebral cortex (P < .0001). The GABAergic TCA cycle flux was reduced in the cerebral cortex (P = .0008), while GABA-glutamine neurotransmitter cycling flux was also reduced in the cerebral cortex (P = .011) and hippocampus (P = .042) of aged brain. In conclusion, the reduction in excitatory and inhibitory neurotransmitter activity of glutamatergic and GABAergic neurons in the cerebral cortex and hippocampus correlates qualitatively with declined cognitive function in aged mice.
Collapse
Affiliation(s)
- Anant Bahadur Patel
- NMR Microimaging and Spectroscopy, CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Pandichelvam Veeraiah
- NMR Microimaging and Spectroscopy, CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
| | - Mohammad Shameem
- NMR Microimaging and Spectroscopy, CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
| | - Jerald Mahesh Kumar
- NMR Microimaging and Spectroscopy, CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
| | - Kamal Saba
- NMR Microimaging and Spectroscopy, CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
25
|
Xu P, Ning J, Jiang Q, Li C, Yan J, Zhao L, Gao H, Zheng H. Region-specific metabolic characterization of the type 1 diabetic brain in mice with and without cognitive impairment. Neurochem Int 2020; 143:104941. [PMID: 33333211 DOI: 10.1016/j.neuint.2020.104941] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 12/08/2020] [Accepted: 12/12/2020] [Indexed: 11/28/2022]
Abstract
Type 1 diabetes (T1D) has been reported to cause cognitive decline, but brain metabolic changes during this process are still far from being fully understood. Here, we found that streptozotocin (STZ)-induced T1D mice exhibited impaired learning and memory at 11 weeks after STZ treatment but not at 3 weeks. Therefore, we studied metabolic alterations in six different brain regions of T1D mice with and without cognitive decline, and attempted to identify key metabolic pathways related to diabetic cognitive dysfunction. The results demonstrate that lactate had already increased in all brain regions of T1D mice prior to cognitive decline, but a decreased TCA cycle was only observed in hippocampus, cortex and striatum of T1D mice with cognitive impairment. Reduced N-acetylaspartate and choline were found in all brain regions of T1D mice, irrespective of cognitive decline. In addition, disrupted neurotransmitter metabolism was noted to occur in T1D mice before cognitive deficit. Of note, we found that the level of uridine was significantly reduced in cerebellum, cortex, hypothalamus and midbrain of T1D mice when cognitive decline was presented. Therefore, brain region-specific metabolic alterations may comprise possible biomarkers for the early-diagnosis and monitoring of diabetic cognitive decline. Moreover, down-regulated TCA cycle and pyrimidine metabolism could be closely related to T1D-associated cognitive impairment.
Collapse
Affiliation(s)
- Pengtao Xu
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Jie Ning
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Qiaoying Jiang
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Chen Li
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Junjie Yan
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Liangcai Zhao
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Hongchang Gao
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
| | - Hong Zheng
- Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
26
|
Kirov II, Sollberger M, Davitz MS, Glodzik L, Soher BJ, Babb JS, Monsch AU, Gass A, Gonen O. Global brain volume and N-acetyl-aspartate decline over seven decades of normal aging. Neurobiol Aging 2020; 98:42-51. [PMID: 33232854 DOI: 10.1016/j.neurobiolaging.2020.10.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 09/07/2020] [Accepted: 10/24/2020] [Indexed: 10/23/2022]
Abstract
We characterize the whole-brain N-acetyl-aspartate (WBNAA) and brain tissue fractions across the adult lifespan and test the hypothesis that, despite age-related atrophy, neuronal integrity (reflected by WBNAA) is preserved in normal aging. Two-hundred-and-seven participants: 133 cognitively intact older adults (73.6 ± 7.4 mean ± standard deviation, range: 60-90 year old) and 84 young (37.9 ± 11, range: 21-59 year old) were scanned with proton magnetic resonance spectroscopy and T1-weighted MRI. Their WBNAA, fractional brain parenchyma, and gray and white matter volumes (fBPV, fGM, and fWM) were compared and modeled as functions of age and sex. Compared with young, older-adults' WBNAA was lower by ~35%, and fBPV, fGM and fWM were lower by ~10%. Linear regressions found 0.5%/year WBNAA and 0.2%/year fBPV and fGM declines, whereas fWM rose to age ~40 years, and declined thereafter. fBPV and fGM were 1.8% and 4% higher in women, with no sex decline rates difference. We conclude that contrary to our hypothesis, atrophy was accompanied by WBNAA decline. Across the entire age range, women's brains showed less atrophy than men's. Formulas to estimate WBNAA and brain tissue fractions in healthy adults are provided to help differentiate normal from abnormal aging.
Collapse
Affiliation(s)
- Ivan I Kirov
- Department of Radiology, Center for Advanced Imaging Innovation and Research (CAI(2)R), Bernard and Irene Schwartz Center for Biomedical Imaging, New York University Grossman School of Medicine, New York, NY, USA
| | - Marc Sollberger
- University Department of Geriatric Medicine FELIX PLATTER, Memory Clinic, Basel, Switzerland; Department of Neurology, University Hospital, Basel, Switzerland
| | - Matthew S Davitz
- Department of Radiology, Center for Advanced Imaging Innovation and Research (CAI(2)R), Bernard and Irene Schwartz Center for Biomedical Imaging, New York University Grossman School of Medicine, New York, NY, USA
| | - Lidia Glodzik
- Department of Radiology, Center for Advanced Imaging Innovation and Research (CAI(2)R), Bernard and Irene Schwartz Center for Biomedical Imaging, New York University Grossman School of Medicine, New York, NY, USA
| | - Brian J Soher
- Department of Radiology, Duke University Medical Center, Durham, NC, USA
| | - James S Babb
- Department of Radiology, Center for Advanced Imaging Innovation and Research (CAI(2)R), Bernard and Irene Schwartz Center for Biomedical Imaging, New York University Grossman School of Medicine, New York, NY, USA
| | - Andreas U Monsch
- University Department of Geriatric Medicine FELIX PLATTER, Memory Clinic, Basel, Switzerland
| | - Achim Gass
- Department of Neurology/Neuroimaging, University Hospital Mannheim, University of Heidelberg, Mannheim, Germany
| | - Oded Gonen
- Department of Radiology, Center for Advanced Imaging Innovation and Research (CAI(2)R), Bernard and Irene Schwartz Center for Biomedical Imaging, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
27
|
Pépin J, de Longprez L, Trovero F, Brouillet E, Valette J, Flament J. Complementarity of gluCEST and 1 H-MRS for the study of mouse models of Huntington's disease. NMR IN BIOMEDICINE 2020; 33:e4301. [PMID: 32198958 DOI: 10.1002/nbm.4301] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 03/06/2020] [Accepted: 03/07/2020] [Indexed: 06/10/2023]
Abstract
Identification of relevant biomarkers is fundamental to understand biological processes of neurodegenerative diseases and to evaluate therapeutic efficacy. Atrophy of brain structures has been proposed as a biomarker, but it provides little information about biochemical events related to the disease. Here, we propose to identify early and relevant biomarkers by combining biological specificity provided by 1 H-MRS and high spatial resolution offered by gluCEST imaging. For this, two different genetic mouse models of Huntington's disease (HD)-the Ki140CAG model, characterized by a slow progression of the disease, and the R6/1 model, which mimics the juvenile form of HD-were used. Animals were scanned at 11.7 T using a protocol combining 1 H-MRS and gluCEST imaging. We measured a significant decrease in levels of N-acetyl-aspartate, a metabolite mainly located in the neuronal compartment, in HD animals, and the decrease seemed to be correlated with disease severity. In addition, variations of tNAA levels were correlated with striatal volumes in both models. Significant variations of glutamate levels were also observed in Ki140CAG but not in R6/1 mice. Thanks to its high resolution, gluCEST provided complementary insights, and we highlighted alterations in small brain regions such as the corpus callosum in Ki140CAG mice, whereas the glutamate level was unchanged in the whole brain of R6/1 mice. In this study, we showed that 1 H-MRS can provide key information about biological processes occurring in vivo but was limited by the spatial resolution. On the other hand, gluCEST may finely point to alterations in unexpected brain regions, but it can also be blind to disease processes when glutamate levels are preserved. This highlights in a practical context the complementarity of the two methods to study animal models of neurodegenerative diseases and to identify relevant biomarkers.
Collapse
Affiliation(s)
- Jérémy Pépin
- Commissariat à l'Energie Atomique (CEA), Direction de la Recherche Fondamentale (DRF), Institut de biologie François Jacob, Molecular Imaging Research Center (MIRCen), Université Paris-Saclay, Fontenay-aux-Roses, France
- Centre National de la Recherche Scientifique (CNRS), Université Paris-Sud, Université Paris-Saclay, UMR 9199, Neurodegenerative Diseases Laboratory, Fontenay-aux-Roses, France
| | - Lucie de Longprez
- Commissariat à l'Energie Atomique (CEA), Direction de la Recherche Fondamentale (DRF), Institut de biologie François Jacob, Molecular Imaging Research Center (MIRCen), Université Paris-Saclay, Fontenay-aux-Roses, France
- Centre National de la Recherche Scientifique (CNRS), Université Paris-Sud, Université Paris-Saclay, UMR 9199, Neurodegenerative Diseases Laboratory, Fontenay-aux-Roses, France
| | | | - Emmanuel Brouillet
- Commissariat à l'Energie Atomique (CEA), Direction de la Recherche Fondamentale (DRF), Institut de biologie François Jacob, Molecular Imaging Research Center (MIRCen), Université Paris-Saclay, Fontenay-aux-Roses, France
- Centre National de la Recherche Scientifique (CNRS), Université Paris-Sud, Université Paris-Saclay, UMR 9199, Neurodegenerative Diseases Laboratory, Fontenay-aux-Roses, France
| | - Julien Valette
- Commissariat à l'Energie Atomique (CEA), Direction de la Recherche Fondamentale (DRF), Institut de biologie François Jacob, Molecular Imaging Research Center (MIRCen), Université Paris-Saclay, Fontenay-aux-Roses, France
- Centre National de la Recherche Scientifique (CNRS), Université Paris-Sud, Université Paris-Saclay, UMR 9199, Neurodegenerative Diseases Laboratory, Fontenay-aux-Roses, France
| | - Julien Flament
- Commissariat à l'Energie Atomique (CEA), Direction de la Recherche Fondamentale (DRF), Institut de biologie François Jacob, Molecular Imaging Research Center (MIRCen), Université Paris-Saclay, Fontenay-aux-Roses, France
- Centre National de la Recherche Scientifique (CNRS), Université Paris-Sud, Université Paris-Saclay, UMR 9199, Neurodegenerative Diseases Laboratory, Fontenay-aux-Roses, France
| |
Collapse
|
28
|
Lin AL, Parikh I, Yanckello LM, White RS, Hartz AMS, Taylor CE, McCulloch SD, Thalman SW, Xia M, McCarty K, Ubele M, Head E, Hyder F, Sanganahalli BG. APOE genotype-dependent pharmacogenetic responses to rapamycin for preventing Alzheimer's disease. Neurobiol Dis 2020; 139:104834. [PMID: 32173556 PMCID: PMC7486698 DOI: 10.1016/j.nbd.2020.104834] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 02/21/2020] [Accepted: 03/11/2020] [Indexed: 01/07/2023] Open
Abstract
The ε4 allele of Apolipoprotein (APOE4) is the strongest genetic risk factor for Alzheimer's disease (AD), the most common form of dementia. Cognitively normal APOE4 carriers have developed amyloid beta (Aβ) plaques and cerebrovascular, metabolic and structural deficits decades before showing the cognitive impairment. Interventions that can inhibit Aβ retention and restore the brain functions to normal would be critical to prevent AD for the asymptomatic APOE4 carriers. A major goal of the study was to identify the potential usefulness of rapamycin (Rapa), a pharmacological intervention for extending longevity, for preventing AD in the mice that express human APOE4 gene and overexpress Aβ (the E4FAD mice). Another goal of the study was to identify the potential pharmacogenetic differences in response to rapamycin between the E4FAD and E3FAD mice, the mice with human APOE ε3 allele. We used multi-modal MRI to measure in vivo cerebral blood flow (CBF), neurotransmitter levels, white matter integrity, water content, cerebrovascular reactivity (CVR) and somatosensory response; used behavioral assessments to determine cognitive function; used biochemistry assays to determine Aβ retention and blood-brain barrier (BBB) functions; and used metabolomics to identify brain metabolic changes. We found that in the E4FAD mice, rapamycin normalized bodyweight, restored CBF (especially in female), BBB activity for Aβ transport, neurotransmitter levels, neuronal integrity and free fatty acid level, and reduced Aβ retention, which were not observe in the E3FAD-Rapa mice. In contrast, E3FAD-Rapa mice had lower CVR responses, lower anxiety and reduced glycolysis in the brain, which were not seen in the E4FAD-Rapa mice. Further, rapamycin appeared to normalize lipid-associated metabolism in the E4FAD mice, while slowed overall glucose-associated metabolism in the E3FAD mice. Finally, rapamycin enhanced overall water content, water diffusion in white matter, and spatial memory in both E3FAD and E4FAD mice, but did not impact the somatosensory responses under hindpaw stimulation. Our findings indicated that rapamycin was able to restore brain functions and reduce AD risk for young, asymptomatic E4FAD mice, and there were pharmacogenetic differences between the E3FAD and E4FAD mice. As the multi-modal MRI methods used in the study are readily to be used in humans and rapamycin is FDA-approved, our results may pave a way for future clinical testing of the pharmacogenetic responses in humans with different APOE alleles, and potentially using rapamycin to prevent AD for asymptomatic APOE4 carriers.
Collapse
Affiliation(s)
- Ai-Ling Lin
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States of America; Department of Pharmacology and Nutritional Science, University of Kentucky, Lexington, KY, United States of America; Department of Neuroscience, University of Kentucky, Lexington, KY, United States of America; F. Joseph Halcomb III, Department of Biomedical Engineering, University of Kentucky, Lexington, KY, United States of America.
| | - Ishita Parikh
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States of America; Oklahoma Medical Research Foundation, Oklahoma City, OK, United States of America
| | - Lucille M Yanckello
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States of America; Department of Pharmacology and Nutritional Science, University of Kentucky, Lexington, KY, United States of America
| | - Renee S White
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States of America; Georgetown College, Georgetown, KY, United States of America
| | - Anika M S Hartz
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States of America; Department of Pharmacology and Nutritional Science, University of Kentucky, Lexington, KY, United States of America
| | - Chase E Taylor
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States of America
| | | | - Scott W Thalman
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States of America; F. Joseph Halcomb III, Department of Biomedical Engineering, University of Kentucky, Lexington, KY, United States of America
| | - Mengfan Xia
- Department of Pharmacology and Nutritional Science, University of Kentucky, Lexington, KY, United States of America
| | - Katie McCarty
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States of America
| | - Margo Ubele
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States of America
| | - Elizabeth Head
- Department of Pathology & Laboratory Medicine, University of California, Irvine, CA, United States of America; University of California Irvine Institute for Memory Impairments and Neurological Disorders, Irvine, CA, United States of America
| | - Fahmeed Hyder
- Magnetic Resonance Research Center, Yale University, New Haven, CT, United States of America; Quantitative Neuroscience with Magnetic Resonance Core Center, Yale University, New Haven, CT, United States of America; Department of Diagnostic Radiology, Yale University, New Haven, CT, United States of America; Department of Biomedical Engineering, Yale University, New Haven, CT, United States of America
| | - Basavaraju G Sanganahalli
- Magnetic Resonance Research Center, Yale University, New Haven, CT, United States of America; Quantitative Neuroscience with Magnetic Resonance Core Center, Yale University, New Haven, CT, United States of America; Department of Diagnostic Radiology, Yale University, New Haven, CT, United States of America; Department of Biomedical Engineering, Yale University, New Haven, CT, United States of America
| |
Collapse
|
29
|
Woodcock EA, Hillmer AT, Mason GF, Cosgrove KP. Imaging Biomarkers of the Neuroimmune System among Substance Use Disorders: A Systematic Review. MOLECULAR NEUROPSYCHIATRY 2019; 5:125-146. [PMID: 31312635 PMCID: PMC6597912 DOI: 10.1159/000499621] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 03/13/2019] [Indexed: 12/14/2022]
Abstract
There is tremendous interest in the role of the neuroimmune system and inflammatory processes in substance use disorders (SUDs). Imaging biomarkers of the neuroimmune system in vivo provide a vital translational bridge between preclinical and clinical research. Herein, we examine two imaging techniques that measure putative indices of the neuroimmune system and review their application among SUDs. Positron emission tomography (PET) imaging of 18 kDa translocator protein availability is a marker associated with microglia. Proton magnetic resonance spectroscopy quantification of myo-inositol levels is a putative glial marker found in astrocytes. Neuroinflammatory responses are initiated and maintained by microglia and astrocytes, and thus represent important imaging markers. The goal of this review is to summarize neuroimaging findings from the substance use literature that report data using these markers and discuss possible mechanisms of action. The extant literature indicates abused substances exert diverse and complex neuroimmune effects. Moreover, drug effects may change across addiction stages, i.e. the neuroimmune effects of acute drug administration may differ from chronic use. This burgeoning field has considerable potential to improve our understanding and treatment of SUDs. Future research is needed to determine how targeting the neuroimmune system may improve treatment outcomes.
Collapse
Affiliation(s)
| | | | | | - Kelly P. Cosgrove
- Departments of Psychiatry, and of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
30
|
Hollinger KR, Alt J, Rais R, Kaplin AI, Slusher BS. The NAAG’ing Concerns of Modeling Human Alzheimer’s Disease in Mice. J Alzheimers Dis 2019; 68:939-945. [DOI: 10.3233/jad-181251] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Kristen R. Hollinger
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
- Johns Hopkins Drug Discovery, Johns Hopkins University, Baltimore, MD, USA
| | - Jesse Alt
- Johns Hopkins Drug Discovery, Johns Hopkins University, Baltimore, MD, USA
| | - Rana Rais
- Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
- Johns Hopkins Drug Discovery, Johns Hopkins University, Baltimore, MD, USA
| | - Adam I. Kaplin
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Barbara S. Slusher
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
- Johns Hopkins Drug Discovery, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
31
|
The Impact of Early Neuroimaging and Developmental Assessment in a Preterm Infant Diagnosed with Cerebral Palsy. Case Rep Pediatr 2019; 2019:9612507. [PMID: 30881719 PMCID: PMC6383416 DOI: 10.1155/2019/9612507] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 01/09/2019] [Indexed: 11/18/2022] Open
Abstract
Premature infants are at risk for cerebral palsy (CP) that is typically diagnosed between 18-24 months. We present a case study of an infant who was discharged from the neonatal intensive care unit (NICU) without obvious neurological deficits but was later diagnosed with hemiplegic CP. The infant was enrolled in an infant motor study, which included neuroimaging and developmental motor assessments. At term, anatomical MRI showed bilateral periventricular leukomalacia, abnormal brain metabolites in frontal white matter via MR spectroscopy (MRS), and low fractional anisotropy (FA) values obtained from diffusional kurtosis imaging (DKI) in several cortical white matter tracts compared to a group of typically developing infants without neuroimaging abnormalities. In addition, the infant scored below average on a developmental assessment administered at term and three months as well as on the standard Bayley III assessment at 12 months. Abnormal neuroimaging and low scores on the early developmental assessment prompted referral for intervention services at two months. With intensive therapy, by 45 months, the infant was average in self-care, mobility, and communication skills, although below average in visual motor and gross motor coordination. This case highlights the clinical impact of early detection and referral using combined neuroimaging and developmental testing.
Collapse
|
32
|
Zhang Y, Liu Z, Ji B, Liu L, Wu S, Liu X, Wang S, Wang L. Metabolite Profile of Alzheimer's Disease in the Frontal Cortex as Analyzed by HRMAS 1H NMR. Front Aging Neurosci 2019; 10:424. [PMID: 30687076 PMCID: PMC6333733 DOI: 10.3389/fnagi.2018.00424] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 12/06/2018] [Indexed: 12/14/2022] Open
Abstract
Background: Investigation on neurochemical changes in the frontal cortex in individuals with Alzheimer’s disease (AD) and different Apolipoprotein E (APOE) genotypes, using ex vivo solid-state high-resolution NMR analysis, may lead to a better understanding of the neurochemistry associated with AD as well as new AD-specific metabolite biomarkers that might potentially improve the clinical diagnosis of AD. Methods: Intact tissue samples of the frontal cortex were obtained from 11 patients and 11 age-matched non-demented controls. Metabolite profiles in all samples were analyzed ex vivo, using solid-state high-resolution magic angle spinning (HRMAS) 600 MHz 1H nuclear magnetic resonance (NMR). A logistic regression analysis was used to rank metabolites based on their level of contribution in differentiating the AD patient tissues and the controls, and different AD-associated APOE genotypes (APOE ε4 vs. APOE ε3). Results: Tissue samples from the AD patients showed significantly lower NAA/Cr (p = 0.011), Ace/Cr (p = 0.027), GABA/Cr (p = 0.005), Asp/Cr (p < 0.0001), mI/Cr (p < 0.0001), and Tau/Cr (p = 0.021), and higher PCho/Cr (p < 0.0001), GPCho/Cr (p < 0.0001), and α&β-Glc/Cr (p < 0.0001) than the controls did. Specifically, a newly observed resonance at 3.71 ppm, referred to as α&β-Glc, was observed in 90.9% of the AD samples (10/11). Samples with APOE ε4 also exhibited higher PCho/Cr (p = 0.0002), GPCho/Cr (p = 0.0001), α&β-Glc/Cr (p < 0.0001), and lower Asp/Cr (p = 0.004) and GABA/Cr (p = 0.04) than the samples with APOE ε3 did. In the logistic regression analysis, PCho, GPCho, ASP, and α&β-Glc were found to be the most relevant metabolites for differentiating the AD patient tissues and the controls, and different APOE genotypes. Conclusion: HRMAS 1H NMR with high spectral resolution and sensitivity offers a powerful tool to gain quantitative information on AD associated neurochemical changes. There are important neurochemical differences in the frontal cortex between the AD patient tissues and the controls, and between those with different APOE genotypes. The resonance (α&β-Glc) found at 3.71 ppm in the AD patient tissues may be further investigated for its potential in the diagnosis and monitoring of AD.
Collapse
Affiliation(s)
- Yuzhong Zhang
- Department of Radiology, The People's Hospital of Longhua, Shenzhen, China
| | - Zhou Liu
- Graduate School, Medical College of Nanchang University, Nanchang, China.,Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, United States
| | - Bing Ji
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, United States
| | - Lijian Liu
- Graduate School, Medical College of Nanchang University, Nanchang, China
| | - Shaoxiong Wu
- Department of Chemistry, NMR Research Center, Emory University, Atlanta, GA, United States
| | - Xiaowu Liu
- Yiwei Medical Technology, Inc., Shenzhen, China
| | - Silun Wang
- Yiwei Medical Technology, Inc., Shenzhen, China
| | - Liya Wang
- Graduate School, Medical College of Nanchang University, Nanchang, China.,Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
33
|
Soeiro-de-Souza MG, Otaduy MCG, Machado-Vieira R, Moreno RA, Nery FG, Leite C, Lafer B. Lithium-associated anterior cingulate neurometabolic profile in euthymic Bipolar I disorder: A 1H-MRS study. J Affect Disord 2018; 241:192-199. [PMID: 30130684 DOI: 10.1016/j.jad.2018.08.039] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 08/07/2018] [Accepted: 08/09/2018] [Indexed: 01/28/2023]
Abstract
OBJECTIVE In the treatment of Bipolar disorder (BD), achieving euthymia is highly complex and usually requires a combination of mood stabilizers. The mechanism of action in stabilizing mood has not been fully elucidated, but alterations in N-Acetylaspartate (NAA), Myo-Inositol (mI) and Choline (Cho) have been implicated. Proton magnetic resonance spectroscopy (1H-MRS) is the gold standard technique for measuring brain NAA, Cho and mI in vivo. The objective of this study was to investigate the association of lithium use in BD type I and brain levels of NAA, mI and Cho in the (anterior cingulate cortex) ACC. METHODS 129 BD type I subjects and 79 healthy controls (HC) were submitted to a 3-Tesla brain magnetic resonance imaging scan (1H-MRS) using a PRESS ACC single voxel (8cm3) sequence. RESULTS BD patients exhibited higher NAA and Cho levels compared to HC. Lithium prescription was associated with lower mI (combination + monotherapy) and higher NAA levels (monotherapy). CONCLUSION The results observed add to the knowledge about the mechanisms of action of mood stabilizers on brain metabolites during euthymia. Additionally, the observed decrease in mI levels associated with lithium monotherapy is an in vivo finding that supports the inositol-depletion hypothesis of lithium pharmacodynamics.
Collapse
Affiliation(s)
- Marcio Gerhardt Soeiro-de-Souza
- Mood Disorders Unit (GRUDA), Department and Institute of Psychiatry, University of Sao Paulo, Brazil; Genetics and Pharmacogenetics Unit (PROGENE), Department and Institute of Psychiatry, University of Sao Paulo, Brazil.
| | - Maria Concepcion Garcia Otaduy
- Laboratory of Magnetic Resonance LIM44, Department and Institute of Radiology, University of São Paulo (InRad-FMUSP), Brazil
| | | | - Ricardo Alberto Moreno
- Mood Disorders Unit (GRUDA), Department and Institute of Psychiatry, University of Sao Paulo, Brazil
| | - Fabiano G Nery
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, USA
| | - Claudia Leite
- Laboratory of Magnetic Resonance LIM44, Department and Institute of Radiology, University of São Paulo (InRad-FMUSP), Brazil
| | - Beny Lafer
- Bipolar Disorders Program (PROMAN), Department and Institute of Psychiatry, University of São Paulo, Brazil
| |
Collapse
|
34
|
Jin P, Pan Y, Pan Z, Xu J, Lin M, Sun Z, Chen M, Xu M. Alzheimer-like brain metabolic and structural features in cholesterol-fed rabbit detected by magnetic resonance imaging. Lipids Health Dis 2018; 17:61. [PMID: 29587752 PMCID: PMC5870103 DOI: 10.1186/s12944-018-0705-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Accepted: 03/09/2018] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Hypercholesterolemia is known to increase the risk of AD in later life, the purpose of this study is to illustrate brain metabolic and structural changes in a cholesterol-fed rabbit model of Alzheimer's Disease (AD) by using clinical 3 T Magnetic Resonance Imaging (MRI). METHODS The Institutional Animal Care and Use Committee of Zhejiang Chinese Medical University approved the study. Totally 16 Japanese White Rabbits (JWR) were randomly divided into 2 groups including normal control group fed with routine diet (group NC) and high cholesterol diet group (group CD) fed a 2% cholesterol diet with 0.24 ppm copper in the drinking water for 12 weeks. Magnetic resonance spectroscopy (MRS) and structural image of rabbit brain were performed by using a 3 Tesla (T) MRI scanner with an 8 channel Rabbit coil. The chemical metabolites were identified by LC Model including N-acetylaspartate (NAA), creatine (Cr), glutamate (Glu), glutamine (Gln), Glycerophosphatidylcholine (GPC), phosphorylcholine (PCH), and myoinositol (MI). The relative concentrations (/Cr) were analyzed. Additionally, Amyloid-β (Aβ) accumulation in the brain was measured postmortem. For comparisons of MR and Aβ data between groups, two-tailed t-tests were performed. RESULTS The ratio of NAA/Cr (0.76 ± 0.10) and Glu/Cr (0.90 ± 0.14) in group CD were lower than those in the group NC (0.87 ± 0.06, 1.13 ± 0.22, respectively, P < 0.05). Compared to the group NC (2.88 ± 0.09 cm3, 0.63 ± 0.08 cm3, respectively), the cortical and hippocampal volumes (2.60 ± 0.14 cm3 and 0.47 ± 0.07 cm3, respectively) of rabbits brain decreased in the group CD while the third and lateral ventricular volumes enlarged (44.56 ± 6.01 mm3 vs 31.40 ± 6.14 mm3, 261.40 ± 30.98 mm3 vs 153.81 ± 30.08 mm3, P < 0.05). These metabolic and structural changes were additionally accompanied by the significant increase of Aβ1-42 in the cortex and hippocampus (163.60 ± 16.26 pg/mg and 215.20 ± 69.86 pg/mg, respectively, P < 0.05). CONCLUSION High cholesterol diet can induce the brain metabolic and structural changes of the rabbit including lowered level of NAA and Glu and the atrophy of the brain which were similar to those of human AD.
Collapse
Affiliation(s)
- Ping Jin
- The First Affiliated Hospital of Zhejiang Chinese Medical University, No. 54 Youdian Road, Shangcheng District, Hangzhou, Zhejiang 310006 People’s Republic of China
- The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang China
| | - Yongming Pan
- Laboratory Animal Research Center/Comparative Medical Research Institute, Zhejiang Chinese Medical University, No 548 Binwen Road, Binjiang District, Hangzhou, 310053 China
| | - Zhiyong Pan
- The First Affiliated Hospital of Zhejiang Chinese Medical University, No. 54 Youdian Road, Shangcheng District, Hangzhou, Zhejiang 310006 People’s Republic of China
| | - Jianqin Xu
- Laboratory Animal Research Center/Comparative Medical Research Institute, Zhejiang Chinese Medical University, No 548 Binwen Road, Binjiang District, Hangzhou, 310053 China
| | - Min Lin
- The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang China
| | - Zhichao Sun
- The First Affiliated Hospital of Zhejiang Chinese Medical University, No. 54 Youdian Road, Shangcheng District, Hangzhou, Zhejiang 310006 People’s Republic of China
| | - Minli Chen
- Laboratory Animal Research Center/Comparative Medical Research Institute, Zhejiang Chinese Medical University, No 548 Binwen Road, Binjiang District, Hangzhou, 310053 China
| | - Maosheng Xu
- The First Affiliated Hospital of Zhejiang Chinese Medical University, No. 54 Youdian Road, Shangcheng District, Hangzhou, Zhejiang 310006 People’s Republic of China
| |
Collapse
|
35
|
Macey PM, Sarma MK, Prasad JP, Ogren JA, Aysola R, Harper RM, Thomas MA. Obstructive sleep apnea is associated with altered midbrain chemical concentrations. Neuroscience 2017; 363:76-86. [PMID: 28893651 PMCID: PMC5983363 DOI: 10.1016/j.neuroscience.2017.09.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 08/25/2017] [Accepted: 09/03/2017] [Indexed: 12/28/2022]
Abstract
Obstructive sleep apnea (OSA) is accompanied by altered structure and function in cortical, limbic, brainstem, and cerebellar regions. The midbrain is relatively unexamined, but contains many integrative nuclei which mediate physiological functions that are disrupted in OSA. We therefore assessed the chemistry of the midbrain in OSA in this exploratory study. We used a recently developed accelerated 2D magnetic resonance spectroscopy (2D-MRS) technique, compressed sensing-based 4D echo-planar J-resolved spectroscopic imaging (4D-EP-JRESI), to measure metabolites in the midbrain of 14 OSA (mean age±SD:54.6±10.6years; AHI:35.0±19.4; SAO2 min:83±7%) and 26 healthy control (50.7±8.5years) subjects. High-resolution T1-weighted scans allowed voxel localization. MRS data were processed with custom MATLAB-based software, and metabolite ratios calculated with respect to the creatine peak using a prior knowledge fitting (ProFit) algorithm. The midbrain in OSA showed decreased N-acetylaspartate (NAA; OSA:1.24±0.43, Control:1.47±0.41; p=0.03; independent samples t-test), a marker of neuronal viability. Increased levels in OSA over control subjects appeared in glutamate (Glu; OSA:1.23±0.57, Control:0.98±0.33; p=0.03), ascorbate (Asc; OSA:0.56±0.28, Control:0.42±0.20; (50.7±8.5years; p=0.03), and myo-inositol (mI; OSA:0.96±0.48, Control:0.72±0.35; p=0.03). No differences between groups appeared in γ-aminobutyric acid (GABA) or taurine. The midbrain in OSA patients shows decreased NAA, indicating neuronal injury or dysfunction. Higher Glu levels may reflect excitotoxic processes and astrocyte activation, and higher mI is also consistent with glial activation. Higher Asc levels may result from oxidative stress induced by intermittent hypoxia in OSA. Additionally, Asc and Glu are involved with glutamatergic processes, which are likely upregulated in the midbrain nuclei of OSA patients. The altered metabolite levels help explain dysfunction and structural deficits in the midbrain of OSA patients.
Collapse
Key Words
- Asc, ascorbate
- Asp, aspartate
- Ch, choline
- GABA, gamma-aminobutyric acid
- GPC, glycerophosphorylcholine
- GSH, glutathione
- Gln, glutamine
- Glu, glutamate
- Gly, glycine
- NAA, N-acetylaspartate
- NAAG, N-acetylaspartate glutamate
- PCh, phosphocholine
- PE, phosphoethanolamine
- Scy, scyllo-inositol
- Tau, taurine
- Thr, threonine
- autonomic
- intermittent hypoxia
- mI, myo-inositol
- magnetic resonance spectroscopy
- periaqueductal gray
- respiration
- sleep-disordered breathing
Collapse
Affiliation(s)
- Paul M Macey
- School of Nursing, David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, CA 90095, United States; Brain Research Institute, David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, CA 90095, United States.
| | - Manoj K Sarma
- Department of Radiological Sciences, David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, CA 90095, United States
| | - Janani P Prasad
- School of Nursing, David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, CA 90095, United States
| | - Jennifer A Ogren
- Department of Neurobiology, David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, CA 90095, United States
| | - Ravi Aysola
- Department of Medicine (Division of Pulmonary and Critical Care), David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, CA 90095, United States
| | - Ronald M Harper
- Brain Research Institute, David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, CA 90095, United States; Department of Neurobiology, David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, CA 90095, United States
| | - M Albert Thomas
- Department of Radiological Sciences, David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, CA 90095, United States
| |
Collapse
|
36
|
Altered Local Spontaneous Brain Activity in Juvenile Myoclonic Epilepsy: A Preliminary Resting-State fMRI Study. Neural Plast 2015; 2016:3547203. [PMID: 26823984 PMCID: PMC4707362 DOI: 10.1155/2016/3547203] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Revised: 10/10/2015] [Accepted: 10/26/2015] [Indexed: 12/05/2022] Open
Abstract
Purpose. The purpose of this study was to evaluate the regional synchronization of brain in patients with juvenile myoclonic epilepsy (JME). Methods. Resting-state fMRI data were acquired from twenty-one patients with JME and twenty-two healthy subjects. Regional homogeneity (ReHo) was used to analyze the spontaneous activity in whole brain. Two-sample t-test was performed to detect the ReHo difference between two groups. Correlations between the ReHo values and features of seizures were calculated further. Key Findings. Compared with healthy controls, patients showed significantly increased ReHo in bilateral thalami and motor-related cortex regions and a substantial reduction of ReHo in cerebellum and occipitoparietal lobe. In addition, greater ReHo value in the left paracentral lobule was linked to the older age of onset in patients. Significance. These findings implicated the abnormality of thalamomotor cortical network in JME which were associated with the genesis and propagation of epileptiform activity. Moreover, our study supported that the local brain spontaneous activity is a potential tool to investigate the epileptic activity and provided important insights into understanding the pathophysiological mechanisms of JME.
Collapse
|
37
|
N -acetyl-aspartyl-glutamate and inhibition of glutamate carboxypeptidases protects against soman-induced neuropathology. Neurotoxicology 2015; 48:180-91. [DOI: 10.1016/j.neuro.2015.03.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 02/26/2015] [Accepted: 03/13/2015] [Indexed: 12/31/2022]
|
38
|
Koundal S, Gandhi S, Kaur T, Khushu S. Neurometabolic and structural alterations in rat brain due to acute hypobaric hypoxia: in vivo 1H MRS at 7 T. NMR IN BIOMEDICINE 2014; 27:341-347. [PMID: 24395642 DOI: 10.1002/nbm.3068] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Revised: 12/03/2013] [Accepted: 12/04/2013] [Indexed: 06/03/2023]
Abstract
In response to hypobaric hypoxia (HH), which occurs at high altitude, the brain undergoes deleterious changes at the structural and metabolite level. In vivo T2 weighted imaging (T2WI) and (1)H-MRS was performed to understand the structural and metabolic changes in the hippocampus region of rat brain. Data were acquired pre-exposure (baseline controls), immediately after exposure and subsequently at the first, fourth, seventh and 14th days post exposure at normoxia. T2 weighted images of rat brain showed hyperintensity in the CA2/CA3 region of the hippocampus 7 d after acute HH, which persisted till 14 d, probably indicating structural changes in the hippocampus. (1)H-MRS results showed no change in metabolite level immediately after acute HH exposure, but on the first day of normoxia the myo-inositol level was significantly decreased, possibly due to altered astrocyte metabolism. Metabolic alterations showing an increase in choline and decrease in glutamate on the fourth day of normoxia may be seen as a process of demyelination and loss of glutamate pool respectively. On the seventh and 14th days of normoxia, decreases in N-acetylaspartate, creatine and glutamine + glutamate were observed, which might be due to decreased viability of glutamatergic neurons. In vivo (1)H-MRS demonstrated early neurometabolic changes prior to probable structural changes post acute HH exposure. The extension of these studies will help in early risk assessment, developing intervention and strategies for combating HH related changes.
Collapse
Affiliation(s)
- Sunil Koundal
- NMR Research Centre, Institute of Nuclear Medicine and Allied Sciences (INMAS), Timarpur, Delhi, India
| | | | | | | |
Collapse
|
39
|
Jeter CB, Hergenroeder GW, Hylin MJ, Redell JB, Moore AN, Dash PK. Biomarkers for the diagnosis and prognosis of mild traumatic brain injury/concussion. J Neurotrauma 2013; 30:657-70. [PMID: 23062081 DOI: 10.1089/neu.2012.2439] [Citation(s) in RCA: 156] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Mild traumatic brain injury (mTBI) results from a transfer of mechanical energy into the brain from traumatic events such as rapid acceleration/deceleration, a direct impact to the head, or an explosive blast. Transfer of energy into the brain can cause structural, physiological, and/or functional changes in the brain that may yield neurological, cognitive, and behavioral symptoms that can be long-lasting. Because mTBI can cause these symptoms in the absence of positive neuroimaging findings, its diagnosis can be subjective and often is based on self-reported neurological symptoms. Further, proper diagnosis can be influenced by the motivation to conceal or embellish signs and/or an inability of the patient to notice subtle dysfunctions or alterations of consciousness. Therefore, appropriate diagnosis of mTBI would benefit from objective indicators of injury. Concussion and mTBI are often used interchangeably, with concussion being primarily used in sport medicine, whereas mTBI is used in reference to traumatic injury. This review provides a critical assessment of the status of current biomarkers for the diagnosis of human mTBI. We review the status of biomarkers that have been tested in TBI patients with injuries classified as mild, and introduce a new concept for the discovery of biomarkers (termed symptophenotypes) to predict common and unique symptoms of concussion. Finally, we discuss the need for biomarker/biomarker signatures that can detect mTBI in the context of polytrauma, and to assess the consequences of repeated injury on the development of secondary injury syndrome, prolongation of post-concussion symptoms, and chronic traumatic encephalopathy.
Collapse
Affiliation(s)
- Cameron B Jeter
- Department of Diagnostic and Biomedical Sciences, The University of Texas School of Dentistry at Houston, Houston, Texas, USA
| | | | | | | | | | | |
Collapse
|
40
|
Durazzo TC, Mon A, Gazdzinski S, Meyerhoff DJ. Chronic cigarette smoking in alcohol dependence: associations with cortical thickness and N-acetylaspartate levels in the extended brain reward system. Addict Biol 2013; 18:379-91. [PMID: 22070867 DOI: 10.1111/j.1369-1600.2011.00407.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Chronic smoking in alcohol dependence is associated with abnormalities in brain morphology and metabolite levels in large lobar regions (e.g. frontal lobe). Here, we evaluated if these abnormalities are specifically apparent in several cortical and select subcortical components of the extended brain reward system (BRS), a network that is critically involved in the development and maintenance of all forms of addictive disorders. We studied 33 non-smoking and 43 smoking alcohol-dependent individuals (ALC) with 1 week of abstinence and 42 non-smoking Controls. At 1.5 Tesla, we obtained regional measures of cortical thickness and N-acetylaspartate (NAA; a surrogate marker of neuronal integrity) concentration in major components of the BRS as well as the corresponding measures throughout the cortex. Smoking ALC and non-smoking ALC demonstrated decreased thickness compared with Controls in the dorsolateral prefrontal cortex (DLPFC), insula, orbitofrontal cortex (OFC), the total BRS, total frontal cortex and global cortex. Smoking ALC had significantly decreased thickness compared to non-smoking ALC in the ACC, insula, the total BRS and total frontal cortex. Smoking ALC had also lower NAA concentrations than both non-smoking ALC and Controls in the DLPFC, insula, superior corona radiata and the total BRS. Alcohol consumption and common medical and psychiatric co-morbidities did not mediate differences between smoking and non-smoking ALC. This dual modality magnetic resonance (MR) study indicated that chronic smoking in ALC was associated with significant cortical thinning and NAA abnormalities in anterior brain regions that are implicated in the development and maintenance of addictive disorders.
Collapse
Affiliation(s)
- Timothy C Durazzo
- Center for Imaging of Neurodegenerative Diseases (CIND), San Francisco VA Medical Center, San Francisco, CA 94121, USA.
| | | | | | | |
Collapse
|
41
|
Glodzik L, Wu WE, Babb JS, Achtnichts L, Amann M, Sollberger M, Monsch AU, Gass A, Gonen O. The whole-brain N-acetylaspartate correlates with education in normal adults. Psychiatry Res 2012. [PMID: 23177924 PMCID: PMC3508436 DOI: 10.1016/j.pscychresns.2012.04.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
N-acetylaspartate (NAA) is an index of neuronal integrity. We hypothesized that in healthy subjects its whole brain concentration (WBNAA) may be related to formal educational attainment, a common proxy for cognitive reserve. To test this hypothesis, 97 middle aged to elderly subjects (51-89 years old, 38% women) underwent brain magnetic resonance imaging and non-localizing proton spectroscopy. Their WBNAA was obtained by dividing their whole-head NAA amount by the brain volume. Intracranial volume and fractional brain volume, a metric of brain atrophy, were also determined. Each subject's educational attainment was the sum of his/her years of formal education. In the entire group higher education was associated with larger intracranial volume. The relationship between WBNAA and education was observed only in younger (51-70 years old) participants. In this group, education explained 21% of the variance in WBNAA. More WBNAA was related to more years of formal education in adults and younger elders. Prospective studies can determine whether this relationship reflects a true advantage from years of training versus innate characteristics predisposing a subject to higher achievements later in life. We propose that late-life WBNAA may be more affected by other factors acting at midlife and later.
Collapse
Affiliation(s)
- Lidia Glodzik
- Department of Psychiatry, New York University School of Medicine, New York, NY, USA
| | - William E. Wu
- Department of Radiology, New York University School of Medicine, New York, NY, USA
| | - James S. Babb
- Department of Radiology, New York University School of Medicine, New York, NY, USA
| | - Lutz Achtnichts
- Department of Neurology, University Hospital Basel, Petersgraben 4, CH-4031 Basel, Switzerland
| | - Michael Amann
- Department of Neurology, University Hospital Basel, Petersgraben 4, CH-4031 Basel, Switzerland,Department of Neuroradiology, University Hospital Basel, Petersgraben 4, CH-4031 Basel, Switzerland
| | - Marc Sollberger
- Department of Neurology, University Hospital Basel, Petersgraben 4, CH-4031 Basel, Switzerland,Department of Geriatrics, University Hospital Basel, Petersgraben 4, CH-4031 Basel, Switzerland
| | - Andreas U. Monsch
- Department of Geriatrics, University Hospital Basel, Petersgraben 4, CH-4031 Basel, Switzerland
| | - Achim Gass
- Department of Neurology, University Hospital Basel, Petersgraben 4, CH-4031 Basel, Switzerland
| | - Oded Gonen
- Department of Radiology, New York University School of Medicine, New York, NY, USA
| |
Collapse
|
42
|
Hajek T, Bauer M, Pfennig A, Cullis J, Ploch J, O’Donovan C, Bohner G, Klingebiel R, Young LT, MacQueen GM, Alda M. Large positive effect of lithium on prefrontal cortex N-acetylaspartate in patients with bipolar disorder: 2-centre study. J Psychiatry Neurosci 2012; 37:185-92. [PMID: 22353634 PMCID: PMC3341410 DOI: 10.1503/jpn.110097] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
BACKGROUND Neuroprotective effects of lithium (Li) have been well documented in tissue cultures and animal models, whereas human data continue to be limited. Previous studies investigating the association between Li treatment and brain N-acetylaspartate (NAA), a putative neuronal marker, showed mixed results because of methodological heterogeneity. METHODS To investigate the effects of Li on prefrontal cortex NAA levels, we compared patients with bipolar disorder from specialized Li clinics in Berlin and Halifax with at least 2 years of ongoing Li treatment (Li group), patients with lifetime Li exposure of less than 3 months more than 2 years ago (non-Li group) and healthy controls. Participants in both patient groups had at least 10 years of illness and 5 episodes. We measured left prefrontal NAA levels using 1.5-T magnetic resonance spectroscopy. RESULTS We enrolled 27 participants in the Li, 16 in the non-Li and 21 in the healthy control groups. The non-Li group had lower prefrontal NAA levels than the Li group (t41 = -3.44, corrected p < 0.01) or control participants (t35 = -2.91, corrected p < 0.05), who did not differ from the Li group (t46 = -0.14, p = 0.89). The same pattern of prefrontal NAA differences was replicated in both sites. In addition, there was a negative correlation between prefrontal NAA and duration of illness in the non-Li group (r = -0.60, p = 0.019) but not in the Li group (r = 0.07, p = 0.74). LIMITATIONS Study limitations include the crosssectional design and exposure to other medications. CONCLUSION Whereas patients with bipolar disorder, substantial illness burden and limited lifetime Li exposure had significantly lower prefrontal NAA levels than controls, Li-treated patients with similar illness burden showed prefrontal NAA levels comparable to those of healthy controls. These findings provide indirect support for neuroprotective effects of Li and for negative effects of illness burden on prefrontal NAA levels in patients with bipolar disorder.
Collapse
Affiliation(s)
- Tomas Hajek
- Department of Psychiatry, Dalhousie University, Halifax, NS.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Rigotti DJ, Inglese M, Kirov II, Gorynski E, Perry NN, Babb JS, Herbert J, Grossman RI, Gonen O. Two-year serial whole-brain N-acetyl-L-aspartate in patients with relapsing-remitting multiple sclerosis. Neurology 2012; 78:1383-9. [PMID: 22517095 DOI: 10.1212/wnl.0b013e318253d609] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVES To test the hypotheses that 1) patients with relapsing-remitting multiple sclerosis (RR-MS) exhibit a quantifiable decline in their whole-brain concentration of the neural marker N-acetyl-L-aspartate (WBNAA), that is 2) more sensitive than clinical changes and 3) may provide a practical outcome measure for proof-of-concept and larger phase III clinical trials. METHODS Nineteen patients (5 men and 14 women) with clinically definite RR-MS, who were 33 ± 5 years old (mean ± SD), had a disease duration of 47 ± 28 months, and had a median Expanded Disability Status Scale (EDSS) score of 1.0 (range 0-5.5), underwent MRI and proton magnetic resonance spectroscopy ((1)H-MRS) semiannually for 2 years (5 time points). Eight matched control subjects underwent the protocol annually (3 time points). Their global N-acetyl-L-aspartate (1)H-MRS signal was converted into absolute amounts by phantom replacement and into WBNAA by dividing with the brain parenchymal volume, V(B), from MRI segmentation. RESULTS The baseline WBNAA of the patients (10.5 ± 1.7 mM) was significantly lower than that of the controls (12.3 ± 1.3 mM; p < 0.002) and declined significantly (5%/year, p < 0.002) vs that for the controls who did not show a decline (0.4%/year, p > 0.7). Likewise, V(B) values of the patients also declined significantly (0.5%/year, p < 0.0001), whereas those of the controls did not (0.2%/year, p = 0.08). The mean EDSS score of the patients increased insignificantly from 1.0 to 1.5 (range 0-6.0) and did not correlate with V(B) or WBNAA. CONCLUSIONS WBNAA of patients with RR-MS declined significantly at both the group and individual levels over a 2-year time period common in clinical trials. Because of the small sample sizes required to establish power, WBNAA can be incorporated into future studies.
Collapse
Affiliation(s)
- D J Rigotti
- Department of Radiology, New York University School of Medicine, New York, NY, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
The heterogeneity of epilepsy syndromes and pathologies creates a great challenge for the search for biomarkers. Not surprisingly, identification of a marker that is specific and sensitive for a given epileptogenic pathology remains an unmet need. There have, however, been several studies of major epileptogenic etiologies like traumatic brain injury that aimed to identify molecular markers in blood and cerebrospinal fluid that predict outcome, by using proteomics and metabolomics. Unfortunately, epileptogenesis has not been analyzed as an outcome measure. Another question to be explored is whether a palette of molecular markers is needed, rather than a single molecule, with each marker probing a different component of epileptogenic pathology. Further, perhaps multiple biomarker platforms (e.g., imaging, proteomics, electrophysiology) should be used in combination and/or in a defined temporal sequence.
Collapse
Affiliation(s)
- Asla Pitkänen
- Department of Neurobiology, AI Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.
| | | |
Collapse
|
45
|
Wu WE, Gass A, Glodzik L, Babb JS, Hirsch J, Sollberger M, Achtnichts L, Amann M, Monsch AU, Gonen O. Whole brain N-acetylaspartate concentration is conserved throughout normal aging. Neurobiol Aging 2012; 33:2440-7. [PMID: 22245316 DOI: 10.1016/j.neurobiolaging.2011.12.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2011] [Revised: 12/01/2011] [Accepted: 12/03/2011] [Indexed: 01/24/2023]
Abstract
We hypothesize that normal aging implies neuronal durability, reflected by age-independent concentrations of their marker--the amino acid derivative N-acetylaspartate (NAA). To test this, we obtained the whole-brain and whole-head N-acetylaspartate concentrations (WBNAA and WHNAA) with proton magnetic resonance (MR) spectroscopy; and the fractional brain parenchyma volume (fBPV)--a metric of atrophy, by segmenting the magnetic resonance image (MRI) from 42 (18 male) healthy young (31.9 ± 5.8 years old) and 100 (64 male, 72.6 ± 7.3 years old) cognitively normal elderly. The 12.8 ± 1.9 mM WBNAA of the young was not significantly different from the 13.1 ± 3.1 mM in the elderly (p > 0.05). In contrast, both fBPV (87.3 ± 4.7% vs. 74.8 ± 4.8%) and WHNAA (11.1 ± 1.7 mM vs. 9.8 ± 2.4 mM) were significantly higher in the young (approximately 14%; p < 0.0001 for both). The similarity in mean WBNAA between 2 cohorts 4 decades of normal aging apart suggests that neuronal integrity is maintained across the lifespan. Clinically, WBNAA could be used as a marker for normal (hence, also abnormal) brain aging. In contrast, WHNAA and fBPV seem age-related suggesting that brain atrophy may occur without compromising the remaining tissue.
Collapse
Affiliation(s)
- William E Wu
- Department of Radiology, New York University School of Medicine, New York, NY, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
2D MR Spectroscopy Combined with Prior-Knowledge Fitting Is Sensitive to HCV-Associated Cerebral Metabolic Abnormalities. Int J Hepatol 2012; 2012:179365. [PMID: 22844602 PMCID: PMC3403451 DOI: 10.1155/2012/179365] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2012] [Revised: 05/05/2012] [Accepted: 05/19/2012] [Indexed: 11/18/2022] Open
Abstract
There is an evidence of neurocognitive dysfunction even in the absence of advanced liver disease in chronic hepatitis C virus (HCV) infection. Brain metabolism has been investigated non-invasively using one-dimensional (1D) in vivo Magnetic Resonance Spectroscopy (MRS) over three decades. Even though highly concentrated cerebral metabolites (N-acetylaspartate, creatine, choline, glutamate/glutamine, myo-inositol) have been detected using MRS, other metabolites at low concentrations (~1-3 mM or less) including glutathione, aspartate and GABA are quite difficult to observe using 1D MRS. In order to resolve overlapping resonances from a number of metabolites, a remedy is to add a second spectral dimension to the existing 1D MRS. Localized two-dimensional correlated spectroscopy (L-COSY) has been developed over the last decade to enhance the spectral dispersion by using the second spectral dimension. We have evaluated this L-COSY technique in the frontal white/gray matter regions of 14 HCV+ (mean age of 56.2 years) and 14 HCV- (mean age of 46.6 years) subjects. Our preliminary results showed significantly increased myo-inositol and glutathione in the HCV+ compared to the HCV- subjects. Hence, glutathione and myo-inositol should be considered along with other metabolites as important markers of inflammation.
Collapse
|
47
|
Determinants of the enzymatic activity and the subcellular localization of aspartate N-acetyltransferase. Biochem J 2011; 441:105-12. [DOI: 10.1042/bj20111179] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Aspartate N-acetyltransferase (NAT8L, N-acetyltransferase 8-like), the enzyme that synthesizes N-acetylaspartate, is membrane-bound and is at least partially associated with the ER (endoplasmic reticulum). The aim of the present study was to determine which regions of the protein are important for its catalytic activity and its subcellular localization. Transfection of truncated forms of NAT8L into HEK (human embryonic kidney)-293T cells indicated that the 68 N-terminal residues (regions 1 and 2) have no importance for the catalytic activity and the subcellular localization of this enzyme, which was exclusively associated with the ER. Mutation of conserved residues that precede (Arg81 and Glu101, in region 3) or follow (Asp168 and Arg220, in region 5) the putative membrane region (region 4) markedly affected the kinetic properties, suggesting that regions 3 and 5 form the catalytic domain and that the membrane region has a loop structure. Evidence is provided for the membrane region comprising α-helices and the catalytic site being cytosolic. Transfection of chimaeric proteins in which GFP (green fluorescent protein) was fused to different regions of NAT8L indicated that the membrane region (region 4) is necessary and sufficient to target NAT8L to the ER. Thus NAT8L is targeted to the ER membrane by a hydrophobic loop that connects two regions of the catalytic domain.
Collapse
|
48
|
Zhang M, Lu J, Jiao L, Ma Q, Li K. Proton magnetic resonance spectroscopy in patients with symptomatic unilateral internal carotid artery / middle cerebral artery stenosis or occlusion. J Magn Reson Imaging 2011; 34:910-6. [PMID: 21774028 DOI: 10.1002/jmri.22706] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2010] [Accepted: 06/06/2011] [Indexed: 11/10/2022] Open
Abstract
PURPOSE To explore the value of proton magnetic resonance spectroscopy imaging ((1)H-MRSI) in patients with stenosis or occlusion of the internal carotid artery (ICA) / middle cerebral artery (MCA). MATERIALS AND METHODS Fifty noninfarcted patients with stenosis or occlusion of unilateral ICA/MCA were included in our study. In the meantime, 25 patients with cerebral infarction and 25 healthy control subjects were enrolled. All patients and healthy control subjects underwent (1)H-MRSI. Cerebral metabolic changes were studied in the noninfarcted patients and compared with the infarcted patients as well as healthy control subjects. RESULTS In 50 noninfarcted patients N-acetylaspartate (NAA) decreased and choline increased in the ischemic hemisphere compared with the contralateral side and control subjects. Lactate peaks were observed in 12 patients. The metabolic changes were relatively slight but were associated with metabolic disruption in infarcted patients. There were relationships between metabolic abnormalities and neurological status of the noninfarcted patients. CONCLUSION (1)H-MRSI can demonstrate abnormal metabolic changes in cerebral tissues with no infarction, while with ICA/MCA may show stenosis or occlusion at an early stage, which may help guide treatment decisions and preoperative evaluation.
Collapse
Affiliation(s)
- Miao Zhang
- Department of Radiology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | | | | | | | | |
Collapse
|
49
|
Quantitative multivoxel proton MR spectroscopy study of brain metabolites in patients with amnestic mild cognitive impairment: a pilot study. Neuroradiology 2011; 54:451-8. [PMID: 21739221 DOI: 10.1007/s00234-011-0900-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Accepted: 06/16/2011] [Indexed: 10/18/2022]
Abstract
INTRODUCTION The purpose of this study is to investigate brain metabolic changes in patients with amnestic mild cognitive impairment (aMCI) using multivoxel proton MR spectroscopy ((1)H-MVS). METHODS Fourteen aMCI patients and fifteen healthy control subjects participated in this experiment. All MR measurements were acquired using a 1.5-T GE scanner. (1)H-MVS point resolved spectroscopy (2D PROBE-CSI PRESS) pulse sequence (TE = 35 ms; TR = 1,500 ms; phase × frequency, 18 × 18) was used for acquiring MRS data. All data were post-processed using Spectroscopy Analysis by General Electric software and linear combination of model (LCModel). The absolute concentrations of N-acetylaspartate (NAA), choline (Cho), myoinositol (MI), creatine (Cr), and the metabolite ratios of NAA/Cr, Cho/Cr, MI/Cr, and NAA/MI were measured bilaterally in the posterior cingulate gyrus (PCG), inferior precuneus (Pr), paratrigonal white matter (PWM), dorsal thalamus (DT), and lentiform nucleus (LN). RESULTS Patients with aMCI displayed significantly lower NAA levels in the bilateral PCG (p < 0.01), PWM (p < 0.05), and left inferior Pr (p < 0.05). The metabolite ratio of NAA/MI was decreased in the bilateral PCG (p < 0.01) and PWM (p < 0.05) and in the left DT (p < 0.01). NAA/Cr was decreased in the left PCG (p < 0.01), DT (p < 0.05), right PWM (p < 0.05), and LN (p < 0.05). However, MI/Cr was elevated in the right PCG (p < 0.01) and left PWM (p < 0.05). Significantly increased Cho level was also evident in the left PWM (p < 0.05). CONCLUSIONS Our observations of decreased NAA, NAA/Cr, and NAA/MI, in parallel with increased Cho and MI/Cr might be characteristic of aMCI patients.
Collapse
|
50
|
Song Z, Ge D, Ishii K, Yamada H, Toriumi K, Watanabe H, Nabeshima T, Fukushima T. Determination of N-acetylaspartic acid concentration in the mouse brain using HPLC with fluorescence detection. Biomed Chromatogr 2011; 26:147-51. [DOI: 10.1002/bmc.1639] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2011] [Revised: 03/18/2011] [Accepted: 03/21/2011] [Indexed: 11/10/2022]
Affiliation(s)
- Ziyu Song
- Department of Analytical Chemistry, Faculty of Pharmaceutical Sciences; Toho University; 2-2-1 Miyama, Funabashi-shi; Chiba; 274-8510; Japan
| | - Dan Ge
- Department of Analytical Chemistry, Faculty of Pharmaceutical Sciences; Toho University; 2-2-1 Miyama, Funabashi-shi; Chiba; 274-8510; Japan
| | - Kana Ishii
- Department of Analytical Chemistry, Faculty of Pharmaceutical Sciences; Toho University; 2-2-1 Miyama, Funabashi-shi; Chiba; 274-8510; Japan
| | - Hiroshi Yamada
- Department of Analytical Chemistry, Faculty of Pharmaceutical Sciences; Toho University; 2-2-1 Miyama, Funabashi-shi; Chiba; 274-8510; Japan
| | - Kazuya Toriumi
- Department of Chemical Pharmacology; Meijo University Graduate School of Pharmaceutical Sciences; Nagoya; 468-8503; Japan
| | - Hiroyuki Watanabe
- Department of Chemical Pharmacology; Meijo University Graduate School of Pharmaceutical Sciences; Nagoya; 468-8503; Japan
| | - Toshitaka Nabeshima
- Department of Chemical Pharmacology; Meijo University Graduate School of Pharmaceutical Sciences; Nagoya; 468-8503; Japan
| | - Takeshi Fukushima
- Department of Analytical Chemistry, Faculty of Pharmaceutical Sciences; Toho University; 2-2-1 Miyama, Funabashi-shi; Chiba; 274-8510; Japan
| |
Collapse
|