1
|
Gorgich EA, Heidari Z, Mahmoudzadeh-Sagheb H, Rustamzadeh A, Shabani A, Amirzadeh A, Haghi Ashtiani B. Brain Metabolite Profiles are Associated with Selective Neuronal Vulnerability and Underlying Mechanisms in Amyotrophic Lateral Sclerosis. ACS Chem Neurosci 2025; 16:1469-1480. [PMID: 40156516 DOI: 10.1021/acschemneuro.4c00593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/01/2025] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a lethal neurological syndrome accompanied by selective degeneration of somatic motor neurons and neurochemistry alterations. Nevertheless, eye movement's nuclei are relatively spared from ALS damage. This survey was to probe metabolite changes in the primary motor cortex (PMC) and interstitial nucleus of Cajal (INC) of ALS patients using proton magnetic resonance spectroscopy (1H-MRS). In this case-control study, 20 patients with ALS and 20 healthy controls underwent 1.5 T MRI and multivoxel 1H-MRS. 1H-MRS spectra to determine metabolite profiles including tNAA, mIns, tCr, tCho, and also tNAA/tCr, tNAA/tCho, and mIns/tNAA metabolite ratios from the PMC and INC were quantified via a point resolved spectroscopy pulse (PRESS) sequence in two groups. Further, the associations between 1H-MRS markers with forced vital capacity (FVC), ALS functional rating scale (ALSFRS-R), and disease progression rate (ΔFS) were investigated. In the PMC, tNAA and tNAA/tCr were significantly lower in ALS patients than the healthy controls, but mIns and mIns/tNAA were significantly greater in these patients (p < 0.05). In the INC, tCho and mIns concentrations, and mIns/tNAA ratio were significantly increased (p < 0.05) in ALS patients, while tNAA and tNAA/tCr ratio did not show significant discriminations between the two groups (p > 0.05). The PMC tNAA/Cr ratio is associated with ALSFRS-R (p = 0.001, r = 0.71), FVC (p = 0.03, r = 0.58), and ΔFS (p = 0.01, r = -0.33). The mIns/tNAA ratio in PMC is also associated with ΔFS (p = 0.02, r = 0.41). In the INC, tCho concentrations (p = 0.04, r = -0.54) and mIns/tNAA ratio (p = 0.02, r = -0.38) were negatively associated with ALSFRS-R and positively correlated with ΔFS (p = 0.01, r = 0.33) and (p = 0.001, r = 0.61), respectively. The study suggests that neurochemistry changes in ALS patients' brains are linked to selective neuronal vulnerability and the underlying pathophysiology of the disease.
Collapse
Affiliation(s)
- Enam Alhagh Gorgich
- Department of Anatomy, School of Medicine, Iranshahr University of Medical Sciences, Iranshahr 99166-43535, Iran
- Department of Histology, School of Medicine, Zahedan University of Medical Sciences, Zahedan 98167-43463, Iran
| | - Zahra Heidari
- Department of Histology, School of Medicine, Zahedan University of Medical Sciences, Zahedan 98167-43463, Iran
- Infectious Diseases and Tropical Medicine Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan 98167-43463, Iran
| | - Hamidreza Mahmoudzadeh-Sagheb
- Department of Histology, School of Medicine, Zahedan University of Medical Sciences, Zahedan 98167-43463, Iran
- Infectious Diseases and Tropical Medicine Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan 98167-43463, Iran
| | - Auob Rustamzadeh
- Department of Anatomical Sciences, School of Medicine, Qazvin University of Medical Sciences, Qazvin 34148-53135, Iran
| | - Arash Shabani
- Department of Advanced Imaging and Image Processing, Saadatabad Medical Imaging Center, Tehran 14496-14535, Iran
| | - Ali Amirzadeh
- Department of Advanced Imaging and Image Processing, Saadatabad Medical Imaging Center, Tehran 14496-14535, Iran
| | - Bahram Haghi Ashtiani
- Department of Neurology, Firoozgar Hospital, School of Medicine, Iran University of Medical Sciences, Tehran 15937-48711, Iran
| |
Collapse
|
2
|
Filippi M, Ghirelli A, Spinelli EG, Agosta F. A comprehensive update on neuroimaging endpoints in amyotrophic lateral sclerosis. Expert Rev Neurother 2025; 25:397-413. [PMID: 39985812 DOI: 10.1080/14737175.2025.2470324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 02/15/2025] [Accepted: 02/18/2025] [Indexed: 02/24/2025]
Abstract
INTRODUCTION There are currently few treatments approved for amyotrophic lateral sclerosis (ALS). Additionally, there remains a significant unmet need for reliable, standardized biomarkers to assess endpoints in clinical trials. Magnetic resonance imaging (MRI)- and positron emission tomography (PET)-derived metrics could help in patient selection and stratification, shortening trial duration and reducing costs. AREAS COVERED This review focuses on the potential use of neuroimaging endpoints in the context of ALS therapeutic trials, providing insights on structural and functional neuroimaging, plexus and muscle alterations, glial involvement and neuroinflammation, envisioning how these surrogates of disease progression could be implemented in clinical trials. A PubMed search covering the past 15 years was performed. EXPERT OPINION Neuroimaging is essential in understanding ALS pathophysiology, aiding in disease progression tracking and evaluating therapeutic interventions. High costs, limited accessibility, lack of standardization, and patient tolerability limit their use in routine ALS care. Addressing these obstacles is essential for fully harnessing neuroimaging potential in improving diagnostics and treatment in ALS.
Collapse
Affiliation(s)
- Massimo Filippi
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurophysiology Service, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Alma Ghirelli
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Edoardo Gioele Spinelli
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Federica Agosta
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
3
|
de Vries E, Hagbohm C, Ouellette R, Granberg T. Clinical 7 Tesla magnetic resonance imaging: Impact and patient value in neurological disorders. J Intern Med 2025; 297:244-261. [PMID: 39775908 PMCID: PMC11846079 DOI: 10.1111/joim.20059] [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] [Indexed: 01/11/2025]
Abstract
Magnetic resonance imaging (MRI) is a cornerstone of non-invasive diagnostics and treatment monitoring, particularly for diseases of the central nervous system. Although 1.5- and 3 Tesla (T) field strengths remain the clinical standard, the advent of 7 T MRI represents a transformative step forward, offering superior spatial resolution, contrast, and sensitivity for visualizing neuroanatomy, metabolism, and function. Recent innovations, including parallel transmission and deep learning-based reconstruction, have resolved many prior technical challenges of 7 T MRI, enabling its routine clinical use. This review examines the diagnostic impact, patient value, and practical considerations of 7 T MRI, emphasizing its role in facilitating earlier diagnoses and improving care in conditions, such as amyotrophic lateral sclerosis (ALS), epilepsy, multiple sclerosis (MS), dementia, parkinsonism, tumors, and vascular diseases. Based on insights from over 1200 clinical scans with a second-generation 7 T system, the review highlights disease-specific biomarkers such as the motor band sign in ALS and the new diagnostic markers in MS, the central vein sign, and paramagnetic rim lesions. The unparalleled ability of 7 T MRI to study neurological diseases ex vivo at ultra-high resolution is also explored, offering new opportunities to understand pathophysiology and identify novel treatment targets. Additionally, the review provides a clinical perspective on patient handling and safety considerations, addressing challenges and practicalities associated with clinical 7 T MRI. By bridging research and clinical practice, 7 T MRI has the potential to redefine neuroimaging and advance the understanding and management of complex neurological disorders.
Collapse
Affiliation(s)
- Elisabeth de Vries
- Department of NeuroradiologyKarolinska University HospitalStockholmSweden
- Department of Clinical NeuroscienceKarolinska InstitutetStockholmSweden
| | - Caroline Hagbohm
- Department of NeuroradiologyKarolinska University HospitalStockholmSweden
- Department of Clinical NeuroscienceKarolinska InstitutetStockholmSweden
| | - Russell Ouellette
- Department of NeuroradiologyKarolinska University HospitalStockholmSweden
- Department of Clinical NeuroscienceKarolinska InstitutetStockholmSweden
| | - Tobias Granberg
- Department of NeuroradiologyKarolinska University HospitalStockholmSweden
- Department of Clinical NeuroscienceKarolinska InstitutetStockholmSweden
| |
Collapse
|
4
|
Luo S, Wang X, Ma B, Liu D, Li L, Wang L, Ding N, Zou L, Wang J, Pan J, Sang D, Zhou H, Qu H, Lu Y, Yang L. Therapeutic potential of simvastatin in ALS: Enhanced axonal integrity and motor neuron survival through Apoa4 and Alb modulation. BIOMOLECULES & BIOMEDICINE 2025; 25:632-647. [PMID: 39569650 PMCID: PMC12010976 DOI: 10.17305/bb.2024.11218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/14/2024] [Accepted: 11/14/2024] [Indexed: 11/22/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by the selective death of motor neurons in the spinal cord, brainstem, and motor cortex. This study investigates the effects of simvastatin on the G93A-copper/zinc superoxide dismutase (G93ASOD1) transgenic mouse model of ALS. The experiment included three groups: C57BL/6 wild-type mice, C57BL/6J SOD1G93A mice treated with PBS (SOD1G93A + PBS), and C57BL/6J SOD1G93A mice treated with simvastatin (SOD1G93A + simvastatin). The primary endpoints were survival rates, body weight changes, performance in pole climbing and suspension tests, and neurological deficit scores. Pathological changes were assessed using hematoxylin and eosin staining, transmission electron microscopy, Nissl staining, and Masson staining. Proteomic and metabolomic analyses were performed to identify differentially expressed proteins (DEPs) and metabolites. Quantitative real-time polymerase chain reaction and western blotting were used to measure gene expression. Although there were no significant differences in survival rates, body weight, pole climbing, and suspension test performance, or neurological deficit scores between the SOD1G93A + simvastatin and SOD1G93A + PBS groups, simvastatin treatment improved axonal organization within the spinal cord, increased the number of neurons, and reduced cytoplasmic swelling and gastrocnemius fibrosis. A total of 47 DEPs and 13 differential metabolites were identified between the SOD1G93A + PBS and SOD1G93A + simvastatin groups. Notably, the expression levels of Apoa4 and Alb were elevated in the SOD1G93A + simvastatin group compared to the SOD1G93A + PBS group. Our results suggest that simvastatin may have potential therapeutic effects in ALS, likely involving the modulation of Apoa4 and Alb expression.
Collapse
Affiliation(s)
- Song Luo
- Department of Neurology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Xiaorui Wang
- Department of Neurology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Bo Ma
- Department of Neurology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Dongliang Liu
- Department of Neurology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Li Li
- Department of Neurology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Lijin Wang
- Department of Psychiatry, Bengbu Medical University, Bengbu, China
| | - Ning Ding
- Department of Hematology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Liangyu Zou
- Department of Neurology, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, Shenzhen, China
| | - Jie Wang
- International Medical Center (Department of Geriatric Medicine), Shenzhen University General Hospital, Shenzhen, China
| | - Jialin Pan
- Department of Internal Medicine, Second People’s Hospital, Longgang District, Shenzhen, China
| | - Daoqian Sang
- Department of Neurology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Huadong Zhou
- Department of Neurology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Hongdang Qu
- Department of Neurology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Yi Lu
- Department of Neurology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Lijuan Yang
- Department of Pediatrics, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| |
Collapse
|
5
|
Ghaderi S, Fatehi F, Kalra S, Okhovat AA, Nafissi S, Mohammadi S, Batouli SAH. Metabolite alterations in the left dorsolateral prefrontal cortex and its association with cognitive assessments in amyotrophic lateral sclerosis: A longitudinal magnetic resonance spectroscopy study. Brain Res Bull 2024; 219:111125. [PMID: 39542047 DOI: 10.1016/j.brainresbull.2024.111125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 10/12/2024] [Accepted: 11/10/2024] [Indexed: 11/17/2024]
Abstract
OBJECTIVE To characterize the longitudinal metabolite profile of the left dorsolateral prefrontal cortex (DLPFC) in amyotrophic lateral sclerosis (ALS) using magnetic resonance spectroscopy (MRS) and to examine its correlation with cognitive assessments. METHODS Thirteen patients at baseline and ten at follow-up, along with 14 age-, sex-, and handedness-matched healthy controls (HCs), were recruited. Three Tesla with a 64-channel coil, Point-RESolved Spectroscopy (PRESS) sequence (TR=1500 ms and TE=140 ms) was used. Metabolites in the left DLPFC were quantified using LCModel. Cognitive performance and functional impairment were assessed using the Edinburgh Cognitive and Behavioral ALS Screen (ECAS) and Revised ALS Functional Rating Scale (ALSFRS-R), respectively. Group comparisons were adjusted for multiple comparisons (p < 0.05, Bonferroni correction). The links between the brain metabolites and cognitive function were investigated using relevant correlation tests (Pearson's or Spearman's). RESULTS Our analysis revealed a significant difference in the choline-to-creatine ratio (tCho/tCr) among the three groups. Baseline ALS patients showed a higher tCho/tCr ratio than HCs (p = 0.033, Bonferroni-corrected). Interestingly, the total N-acetyl aspartate (tNAA)/tCr ratio, a marker of neuronal health, was strongly positively correlated with visuospatial cognitive scores at baseline and follow-up. Furthermore, at follow-up, tNAA/tCr was positively correlated with the total scores and specific sub-scores on the ECAS, encompassing both ALS-specific and non-specific cognitive domains. At follow-up, positive correlations emerged between tNAA/tCr and the total language and executive function scores. CONCLUSIONS Metabolite alterations and correlations with cognition were observed in the left DLPFC of ALS patients, supporting extra-motor involvement and its association with cognitive decline.
Collapse
Affiliation(s)
- Sadegh Ghaderi
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Neuromuscular Research Center, Department of Neurology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Farzad Fatehi
- Neuromuscular Research Center, Department of Neurology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran; Neurology Department, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Sanjay Kalra
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada; Division of Neurology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Ali Asghar Okhovat
- Neuromuscular Research Center, Department of Neurology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahriar Nafissi
- Neuromuscular Research Center, Department of Neurology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Sana Mohammadi
- Neuromuscular Research Center, Department of Neurology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Amir Hossein Batouli
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Takamiya S, Malvea A, Ishaque AH, Pedro K, Fehlings MG. Advances in imaging modalities for spinal tumors. Neurooncol Adv 2024; 6:iii13-iii27. [PMID: 39430391 PMCID: PMC11485884 DOI: 10.1093/noajnl/vdae045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2024] Open
Abstract
The spinal cord occupies a narrow region and is tightly surrounded by osseous and ligamentous structures; spinal tumors can damage this structure and deprive patients of their ability to independently perform activities of daily living. Hence, imaging is vital for the prompt detection and accurate diagnosis of spinal tumors, as well as determining the optimal treatment and follow-up plan. However, many clinicians may not be familiar with the imaging characteristics of spinal tumors due to their rarity. In addition, spinal surgeons might not fully utilize imaging for the surgical planning and management of spinal tumors because of the complex heterogeneity of these lesions. In the present review, we focus on conventional and advanced spinal tumor imaging techniques. These imaging modalities include computed tomography, positron emission tomography, digital subtraction angiography, conventional and microstructural magnetic resonance imaging, and high-resolution ultrasound. We discuss the advantages and disadvantages of conventional and emerging imaging modalities, followed by an examination of cutting-edge medical technology to complement current needs in the field of spinal tumors. Moreover, machine learning and artificial intelligence are anticipated to impact the application of spinal imaging techniques. Through this review, we discuss the importance of conventional and advanced spinal tumor imaging, and the opportunity to combine advanced technologies with conventional modalities to better manage patients with these lesions.
Collapse
Affiliation(s)
- Soichiro Takamiya
- Division of Genetics and Development, Krembil Brain Institute, University Health Network, Toronto, Ontario, Canada
| | - Anahita Malvea
- Division of Neurosurgery and Spine Program, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Abdullah H Ishaque
- Division of Neurosurgery and Spine Program, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Division of Neurosurgery, Krembil Neuroscience Centre, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
| | - Karlo Pedro
- Division of Neurosurgery and Spine Program, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Division of Neurosurgery, Krembil Neuroscience Centre, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Michael G Fehlings
- Division of Genetics and Development, Krembil Brain Institute, University Health Network, Toronto, Ontario, Canada
- Division of Neurosurgery and Spine Program, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Division of Neurosurgery, Krembil Neuroscience Centre, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
7
|
Montanaro D, Vavla M, Frijia F, Coi A, Baratto A, Pasquariello R, Stefan C, Martinuzzi A. Metabolite profile in hereditary spastic paraplegia analyzed using magnetic resonance spectroscopy: a cross-sectional analysis in a longitudinal study. Front Neurosci 2024; 18:1416093. [PMID: 39193522 PMCID: PMC11347332 DOI: 10.3389/fnins.2024.1416093] [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: 04/11/2024] [Accepted: 07/11/2024] [Indexed: 08/29/2024] Open
Abstract
Background Hereditary Spastic Paraplegias (HSP) are genetic neurodegenerative disorders affecting the corticospinal tract. No established neuroimaging biomarker is associated with this condition. Methods A total of 46 patients affected by HSP, genetically and clinically evaluated and tested with SPRS scores, and 46 healthy controls (HC) matched by age and gender underwent a single-voxel Magnetic Resonance Spectroscopy sampling (MRS) of bilateral pre-central and pre-frontal regions. MRS data were analyzed cross-sectionally (at T0 and T1) and longitudinally (T0 vs. T1). Results Statistically significant data showed that T0 mI/Cr in the pre-central areas of HSP patients was higher than in HC. In the left (L) pre-central area, NAA/Cr was significantly lower in HSP than in HC. In the right (R) pre-frontal area, NAA/Cr was significantly lower in HSP patients than in HC. HSP SPG4 subjects had significantly lower Cho/Cr concentrations in the L pre-central area compared to HC. Among the HSP subjects, non-SPG4 patients had significantly higher mI/Cr in the L pre-central area compared to SPG4 patients. In the R pre-frontal area, NAA/Cr was reduced, and ml/Cr was higher in non-SPG4 patients compared to SPG4 patients. Comparing "pure" and "complex" forms, NAA/Cr was higher in pHSP than in cHSP in the R pre-central and R pre-frontal areas. The longitudinal analysis, which involved fewer patients (n = 30), showed an increase in mI/Cr concentration in the L pre-frontal area among HSP subjects with respect to baseline. The patients had significantly higher SPRS scores at follow-up, with a significant positive correlation between SPRS scores and mI/Cr in the L pre-central area, while in bilateral pre-frontal areas, lower SPRS scores corresponded to higher NAA/Cr concentrations. To explore the discriminating power of MRS in correctly identifying HSP and controls, an inference tree methodology classified HSP subjects and controls with an overall accuracy of 73.9%, a sensitivity of 87.0%, and a specificity of 60.9%. Conclusion This pilot study indicates that brain MRS is a valuable approach that could potentially serve as an objective biomarker in HSP.
Collapse
Affiliation(s)
- Domenico Montanaro
- U.O. Dipartimentale e Servizio Autonomo di Risonanza Magnetica, Dipartimento di Neuroscienze dell’Età Evolutiva, IRCCS Fondazione Stella Maris, Pisa, Italy
| | - Marinela Vavla
- Child and Adolescent Neuropsychiatric Unit, Department of Women’s and Children’s Health, University Hospital of Padua, Padova, Italy
- Department of Neurorehabilitation, IRCCS E. Medea Scientific Institute, Conegliano, Italy
| | - Francesca Frijia
- Bioengineering Unit, Fondazione Toscana G. Monasterio, Pisa, Italy
| | - Alessio Coi
- Unit of Epidemiology of Rare Diseases and Congenital Anomalies, Institute of Clinical Physiology, National Research Council, Pisa, Italy
| | - Alessandra Baratto
- Department of Radiology, S. Maria dei Battuti Hospital- Conegliano, Treviso, Italy
| | - Rosa Pasquariello
- U.O. Dipartimentale e Servizio Autonomo di Risonanza Magnetica, Dipartimento di Neuroscienze dell’Età Evolutiva, IRCCS Fondazione Stella Maris, Pisa, Italy
| | - Cristina Stefan
- Department of Neurorehabilitation, IRCCS E. Medea Scientific Institute, Conegliano, Italy
| | - Andrea Martinuzzi
- Department of Neurorehabilitation, IRCCS E. Medea Scientific Institute, Conegliano, Italy
| |
Collapse
|
8
|
Christidi F, Kleinerova J, Tan EL, Delaney S, Tacheva A, Hengeveld JC, Doherty MA, McLaughlin RL, Hardiman O, Siah WF, Chang KM, Lope J, Bede P. Limbic Network and Papez Circuit Involvement in ALS: Imaging and Clinical Profiles in GGGGCC Hexanucleotide Carriers in C9orf72 and C9orf72-Negative Patients. BIOLOGY 2024; 13:504. [PMID: 39056697 PMCID: PMC11273537 DOI: 10.3390/biology13070504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 06/26/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024]
Abstract
Background: While frontotemporal involvement is increasingly recognized in Amyotrophic lateral sclerosis (ALS), the degeneration of limbic networks remains poorly characterized, despite growing evidence of amnestic deficits, impaired emotional processing and deficits in social cognition. Methods: A prospective neuroimaging study was conducted with 204 individuals with ALS and 111 healthy controls. Patients were stratified for hexanucleotide expansion status in C9orf72. A deep-learning-based segmentation approach was implemented to segment the nucleus accumbens, hypothalamus, fornix, mammillary body, basal forebrain and septal nuclei. The cortical, subcortical and white matter components of the Papez circuit were also systematically evaluated. Results: Hexanucleotide repeat expansion carriers exhibited bilateral amygdala, hypothalamus and nucleus accumbens atrophy, and C9orf72 negative patients showed bilateral basal forebrain volume reductions compared to controls. Both patient groups showed left rostral anterior cingulate atrophy, left entorhinal cortex thinning and cingulum and fornix alterations, irrespective of the genotype. Fornix, cingulum, posterior cingulate, nucleus accumbens, amygdala and hypothalamus degeneration was more marked in C9orf72-positive ALS patients. Conclusions: Our results highlighted that mesial temporal and parasagittal subcortical degeneration is not unique to C9orf72 carriers. Our radiological findings were consistent with neuropsychological observations and highlighted the importance of comprehensive neuropsychological testing in ALS, irrespective of the underlying genotype.
Collapse
Affiliation(s)
- Foteini Christidi
- Computational Neuroimaging Group (CNG), School of Medicine, Trinity College Dublin, D08 W9RT Dublin, Ireland
| | - Jana Kleinerova
- Computational Neuroimaging Group (CNG), School of Medicine, Trinity College Dublin, D08 W9RT Dublin, Ireland
| | - Ee Ling Tan
- Computational Neuroimaging Group (CNG), School of Medicine, Trinity College Dublin, D08 W9RT Dublin, Ireland
| | - Siobhan Delaney
- Computational Neuroimaging Group (CNG), School of Medicine, Trinity College Dublin, D08 W9RT Dublin, Ireland
- Department of Neurology, St James’s Hospital, D08 KC95 Dublin, Ireland
| | - Asya Tacheva
- Computational Neuroimaging Group (CNG), School of Medicine, Trinity College Dublin, D08 W9RT Dublin, Ireland
- Department of Neurology, St James’s Hospital, D08 KC95 Dublin, Ireland
| | | | - Mark A. Doherty
- Smurfit Institute of Genetics, Trinity College Dublin, D08 W9RT Dublin, Ireland
| | | | - Orla Hardiman
- Computational Neuroimaging Group (CNG), School of Medicine, Trinity College Dublin, D08 W9RT Dublin, Ireland
| | - We Fong Siah
- Computational Neuroimaging Group (CNG), School of Medicine, Trinity College Dublin, D08 W9RT Dublin, Ireland
| | - Kai Ming Chang
- Computational Neuroimaging Group (CNG), School of Medicine, Trinity College Dublin, D08 W9RT Dublin, Ireland
| | - Jasmin Lope
- Computational Neuroimaging Group (CNG), School of Medicine, Trinity College Dublin, D08 W9RT Dublin, Ireland
| | - Peter Bede
- Computational Neuroimaging Group (CNG), School of Medicine, Trinity College Dublin, D08 W9RT Dublin, Ireland
- Department of Neurology, St James’s Hospital, D08 KC95 Dublin, Ireland
| |
Collapse
|
9
|
Sun W, Liu SH, Wei XJ, Sun H, Ma ZW, Yu XF. Potential of neuroimaging as a biomarker in amyotrophic lateral sclerosis: from structure to metabolism. J Neurol 2024; 271:2238-2257. [PMID: 38367047 DOI: 10.1007/s00415-024-12201-x] [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: 11/18/2023] [Revised: 01/14/2024] [Accepted: 01/16/2024] [Indexed: 02/19/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease characterized by motor neuron degeneration. The development of ALS involves metabolite alterations leading to tissue lesions in the nervous system. Recent advances in neuroimaging have significantly improved our understanding of the underlying pathophysiology of ALS, with findings supporting the corticoefferent axonal disease progression theory. Current studies on neuroimaging in ALS have demonstrated inconsistencies, which may be due to small sample sizes, insufficient statistical power, overinterpretation of findings, and the inherent heterogeneity of ALS. Deriving meaningful conclusions solely from individual imaging metrics in ALS studies remains challenging, and integrating multimodal imaging techniques shows promise for detecting valuable ALS biomarkers. In addition to giving an overview of the principles and techniques of different neuroimaging modalities, this review describes the potential of neuroimaging biomarkers in the diagnosis and prognostication of ALS. We provide an insight into the underlying pathology, highlighting the need for standardized protocols and multicenter collaborations to advance ALS research.
Collapse
Affiliation(s)
- Wei Sun
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Si-Han Liu
- Department of Mechanical Engineering, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Xiao-Jing Wei
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Hui Sun
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Zhen-Wei Ma
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Xue-Fan Yu
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
10
|
Mohammadi S, Ghaderi S, Fatehi F. MRI biomarkers and neuropsychological assessments of hippocampal and parahippocampal regions affected by ALS: A systematic review. CNS Neurosci Ther 2024; 30:e14578. [PMID: 38334254 PMCID: PMC10853901 DOI: 10.1111/cns.14578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 12/11/2023] [Accepted: 12/13/2023] [Indexed: 02/10/2024] Open
Abstract
BACKGROUND AND OBJECTIVE Amyotrophic lateral sclerosis (ALS) is a progressive motor and extra-motor neurodegenerative disease. This systematic review aimed to examine MRI biomarkers and neuropsychological assessments of the hippocampal and parahippocampal regions in patients with ALS. METHODS A systematic review was conducted in the Scopus and PubMed databases for studies published between January 2000 and July 2023. The inclusion criteria were (1) MRI studies to assess hippocampal and parahippocampal regions in ALS patients, and (2) studies reporting neuropsychological data in patients with ALS. RESULTS A total of 46 studies were included. Structural MRI revealed hippocampal atrophy, especially in ALS-FTD, involving specific subregions (CA1, dentate gyrus). Disease progression and genetic factors impacted atrophy patterns. Diffusion tensor imaging (DTI) showed increased mean diffusivity (MD), axial diffusivity (AD), radial diffusivity (RD), and decreased fractional anisotropy (FA) in the hippocampal tracts and adjacent regions, indicating loss of neuronal and white matter integrity. Functional MRI (fMRI) revealed reduced functional connectivity (FC) between the hippocampus, parahippocampus, and other regions, suggesting disrupted networks. Perfusion MRI showed hypoperfusion in parahippocampal gyri. Magnetic resonance spectroscopy (MRS) found changes in the hippocampus, indicating neuronal loss. Neuropsychological tests showed associations between poorer memory and hippocampal atrophy or connectivity changes. CA1-2, dentate gyrus, and fimbria atrophy were correlated with worse memory. CONCLUSIONS The hippocampus and the connected regions are involved in ALS. Hippocampal atrophy disrupted connectivity and metabolite changes correlate with cognitive and functional decline. Specific subregions can be particularly affected. The hippocampus is a potential biomarker for disease monitoring and prognosis.
Collapse
Affiliation(s)
- Sana Mohammadi
- Neuromuscular Research Center, Department of Neurology, Shariati HospitalTehran University of Medical SciencesTehranIran
- Department of Medical Sciences, School of MedicineIran University of Medical SciencesTehranIran
| | - Sadegh Ghaderi
- Neuromuscular Research Center, Department of Neurology, Shariati HospitalTehran University of Medical SciencesTehranIran
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in MedicineTehran University of Medical SciencesTehranIran
| | - Farzad Fatehi
- Neuromuscular Research Center, Department of Neurology, Shariati HospitalTehran University of Medical SciencesTehranIran
| |
Collapse
|
11
|
Lu T, Ding L, Zheng X, Li Y, Wei W, Liu W, Tao J, Xue X. Alisol A Exerts Neuroprotective Effects Against HFD-Induced Pathological Brain Aging via the SIRT3-NF-κB/MAPK Pathway. Mol Neurobiol 2024; 61:753-771. [PMID: 37659035 PMCID: PMC10861652 DOI: 10.1007/s12035-023-03592-5] [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: 01/17/2023] [Accepted: 08/16/2023] [Indexed: 09/05/2023]
Abstract
Chronic consumption of a high-fat diet (HFD) has profound effects on brain aging, which is mainly characterized by cognitive decline, inflammatory responses, and neurovascular damage. Alisol A (AA) is a triterpenoid with therapeutic potential for metabolic diseases, but whether it has a neuroprotective effect against brain aging caused by a HFD has not been investigated. Six-month-old male C57BL6/J mice were exposed to a HFD with or without AA treatment for 12 weeks. Behavioral tasks were used to assess the cognitive abilities of the mice. Neuroinflammation and changes in neurovascular structure in the brains were examined. We further assessed the mechanism by which AA exerts neuroprotective effects against HFD-induced pathological brain aging in vitro and in vivo. Behavioral tests showed that cognitive function was improved in AA-treated animals. AA treatment reduced microglia activation and inflammatory cytokine release induced by a HFD. Furthermore, AA treatment increased the number of hippocampal neurons, the density of dendritic spines, and the expression of tight junction proteins. We also demonstrated that AA attenuated microglial activation by targeting the SIRT3-NF-κB/MAPK pathway and ameliorated microglial activation-induced tight junction degeneration in endothelial cells and apoptosis in hippocampal neurons. The results of this study show that AA may be a promising agent for the treatment of HFD-induced brain aging.
Collapse
Affiliation(s)
- Taotao Lu
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350112, China
- Fujian Key Laboratory of Rehabilitation Techniques, Cognitive Rehabilitation, Fuzhou, 350112, China
| | - Linlin Ding
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350112, China
| | - Xiaoqing Zheng
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350112, China
| | - Yongxu Li
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350112, China
- The Affiliated Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine, No 13, Hudongzhi Road, Fuzhou City, 350003, Fujian Province, China
| | - Wei Wei
- Fujian Key Laboratory of Rehabilitation Techniques, Cognitive Rehabilitation, Fuzhou, 350112, China
- The Affiliated Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine, No 13, Hudongzhi Road, Fuzhou City, 350003, Fujian Province, China
| | - Weilin Liu
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350112, China
| | - Jing Tao
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350112, China
- Fujian Key Laboratory of Rehabilitation Techniques, Cognitive Rehabilitation, Fuzhou, 350112, China
| | - Xiehua Xue
- Fujian Key Laboratory of Rehabilitation Techniques, Cognitive Rehabilitation, Fuzhou, 350112, China.
- The Affiliated Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine, No 13, Hudongzhi Road, Fuzhou City, 350003, Fujian Province, China.
| |
Collapse
|
12
|
Dziadkowiak E, Koszewicz M, Podgórski P, Wieczorek M, Budrewicz S, Zimny A. Central nervous system involvement in chronic inflammatory demyelinating polyradiculoneuropathy-MRS and DTI study. Front Neurol 2024; 15:1301405. [PMID: 38333607 PMCID: PMC10850251 DOI: 10.3389/fneur.2024.1301405] [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: 09/24/2023] [Accepted: 01/04/2024] [Indexed: 02/10/2024] Open
Abstract
Objective The current research aimed to analyze the alterations within the motor cortex and pyramidal pathways and their association with the degree of damage within the peripheral nerve fibers in patients with chronic inflammatory demyelinating polyradiculoneuropathy (CIDP). To achieve that goal, we investigated the microstructural changes within the pyramidal white matter tracts using diffusion tensor imaging (DTI) parameters, evaluated metabolic alterations in both precentral gyri using magnetic resonance spectroscopy (MRS) ratios, and correlated them with the neurographic findings in patients with CIDP. Methods The spectroscopic ratios of NAA/Cr, Cho/Cr, and mI/Cr from both precentral gyri and the values of fractional anisotropy (FA), axial diffusivity (AD), and mean diffusivity (MD) from both of the corticospinal tracts were correlated with the results of neurological and neurographic findings. The comparison of DTI parameters between the patients and controls was performed using Student's t-test or the Mann-Whitney U test. Due to the lack of normal distribution of most variables, Spearman's Rho rank coefficient was used to test all correlations. All analyses were performed at a significant level of alpha = 0.05 using STATISTICA 13.3. Results Compared to the control group (CG), the patient group showed significantly lower ratios of NAA/Cr (1.66 ± 0.11 vs. 1.61 ± 0.15; p = 0.022), higher ratios of ml/Cr in the right precentral gyrus (0.57 ± 0.15 vs. 0.61 ± 0.08; p = 0.005), and higher levels of Cho/Cr within the left precentral gyrus (0.83 ± 0.09 vs. 0.88 ± 0.14, p = 0.012). The DTI parameters of MD from the right CST and AD from the right and left CSTs showed a strong positive correlation (0.52-0.53) with the sural sensory nerve action potential (SNAP) latency of the right sural nerve. There were no other significant correlations between other DTI and MRS parameters and neurographic results. Significance In our study, significant metabolic alterations were found in the precentral gyri in patients with CIDP without clinical symptoms of central nervous system involvement. The revealed changes reflected neuronal loss or dysfunction, myelin degradation, and increased gliosis. Our results suggest coexisting CNS damage in these patients and may provide a new insight into the still unknown pathomechanism of CIDP.
Collapse
Affiliation(s)
- Edyta Dziadkowiak
- Department of Neurology, Wroclaw Medical University, Borowska, Wrocław, Poland
| | - Magdalena Koszewicz
- Department of Neurology, Wroclaw Medical University, Borowska, Wrocław, Poland
| | - Przemysław Podgórski
- Department of General and Interventional Radiology and Neuroradiology, Wroclaw Medical University, Borowska, Wrocław, Poland
| | - Małgorzata Wieczorek
- Faculty of Earth Sciences and Environmental Management, University of Wroclaw, Uniwersytecki, Wrocław, Poland
| | - Sławomir Budrewicz
- Department of Neurology, Wroclaw Medical University, Borowska, Wrocław, Poland
| | - Anna Zimny
- Department of General and Interventional Radiology and Neuroradiology, Wroclaw Medical University, Borowska, Wrocław, Poland
| |
Collapse
|
13
|
Ghaderi S, Mohammadi S, Mohammadi M. Obstructive sleep apnea and attention deficits: A systematic review of magnetic resonance imaging biomarkers and neuropsychological assessments. Brain Behav 2023; 13:e3262. [PMID: 37743582 PMCID: PMC10636416 DOI: 10.1002/brb3.3262] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/07/2023] [Accepted: 09/12/2023] [Indexed: 09/26/2023] Open
Abstract
BACKGROUND AND OBJECTIVE Obstructive sleep apnea (OSA) is a common sleep disorder that causes intermittent hypoxia and sleep fragmentation, leading to attention impairment and other cognitive deficits. Magnetic resonance imaging (MRI) is a powerful modality that can reveal the structural and functional brain alterations associated with attention impairment in OSA patients. The objective of this systematic review is to identify and synthesize the evidence on MRI biomarkers and neuropsychological assessments of attention deficits in OSA patients. METHODS We searched the Scopus and PubMed databases for studies that used MRI to measure biomarkers related to attention alteration in OSA patients and reported qualitative and quantitative data on the association between MRI biomarkers and attention outcomes. We also included studies that found an association between neuropsychological assessments and MRI findings in OSA patients with attention deficits. RESULTS We included 19 studies that met our inclusion criteria and extracted the relevant data from each study. We categorized the studies into three groups based on the MRI modality and the cognitive domain they used: structural and diffusion tensor imaging MRI findings, functional, perfusion, and metabolic MRI findings, and neuropsychological assessment findings. CONCLUSIONS We found that OSA is associated with structural, functional, and metabolic brain alterations in multiple regions and networks that are involved in attention processing. Treatment with continuous positive airway pressure can partially reverse some of the brain changes and improve cognitive function in some domains and in some studies. This review suggests that MRI techniques and neuropsychological assessments can be useful tools for monitoring the progression and response to treatment of OSA patients.
Collapse
Affiliation(s)
- Sadegh Ghaderi
- Department of Neuroscience and Addiction StudiesSchool of Advanced Technologies in MedicineTehran University of Medical SciencesTehranIran
| | - Sana Mohammadi
- Department of Medical SciencesSchool of MedicineIran University of Medical SciencesTehranIran
| | - Mahdi Mohammadi
- Department of Medical Physics and Biomedical Engineering, School of MedicineTehran University of Medical SciencesTehranIran
| |
Collapse
|
14
|
Abstract
Although the past two decades have produced exciting discoveries in the genetics and pathology of amyotrophic lateral sclerosis (ALS), progress in developing an effective therapy remains slow. This review summarizes the critical discoveries and outlines the advances in disease characterization, diagnosis, imaging, and biomarkers, along with the current status of approaches to ALS care and treatment. Additional knowledge of the factors driving disease progression and heterogeneity will hopefully soon transform the care for patients with ALS into an individualized, multi-prong approach able to prevent disease progression sufficiently to allow for a dignified life with limited disability.
Collapse
Affiliation(s)
- Hristelina Ilieva
- Jefferson Weinberg ALS Center, Thomas Jefferson University, Philadelphia, PA, USA
| | | | - Justin Kwan
- National Institute of Neurological Disorders and Stroke, National Institute of Health, Bethesda, MD, USA
| |
Collapse
|
15
|
Stephenson D, Belfiore-Oshan R, Karten Y, Keavney J, Kwok DK, Martinez T, Montminy J, Müller MLTM, Romero K, Sivakumaran S. Transforming Drug Development for Neurological Disorders: Proceedings from a Multidisease Area Workshop. Neurotherapeutics 2023; 20:1682-1691. [PMID: 37823970 PMCID: PMC10684834 DOI: 10.1007/s13311-023-01440-x] [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] [Accepted: 09/06/2023] [Indexed: 10/13/2023] Open
Abstract
Neurological disorders represent some of the most challenging therapeutic areas for successful drug approvals. The escalating global burden of death and disability for such diseases represents a significant worldwide public health challenge, and the rate of failure of new therapies for chronic progressive disorders of the nervous system is higher relative to other non-neurological conditions. However, progress is emerging rapidly in advancing the drug development landscape in both rare and common neurodegenerative diseases. In October 2022, the Critical Path Institute (C-Path) and the US Food and Drug Administration (FDA) organized a Neuroscience Annual Workshop convening representatives from the drug development industry, academia, the patient community, government agencies, and regulatory agencies regarding the future development of tools and therapies for neurological disorders. This workshop focused on five chronic progressive diseases: Alzheimer's disease, Parkinson's disease, Huntington's disease, Duchenne muscular dystrophy, and inherited ataxias. This special conference report reviews the key points discussed during the three-day dynamic workshop, including shared learnings, and recommendations that promise to catalyze future advancement of novel therapies and drug development tools.
Collapse
|
16
|
Dey A, Luk CC, Ishaque A, Ta D, Srivastava O, Krebs D, Seres P, Hanstock C, Beaulieu C, Korngut L, Frayne R, Zinman L, Graham S, Genge A, Briemberg H, Kalra S. Motor cortex functional connectivity is associated with underlying neurochemistry in ALS. J Neurol Neurosurg Psychiatry 2023; 94:193-200. [PMID: 36379713 PMCID: PMC9985743 DOI: 10.1136/jnnp-2022-329993] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 10/18/2022] [Indexed: 11/16/2022]
Abstract
OBJECTIVE To identify structural and neurochemical properties that underlie functional connectivity impairments of the primary motor cortex (PMC) and how these relate to clinical findings in amyotrophic lateral sclerosis (ALS). METHODS 52 patients with ALS and 52 healthy controls, matched for age and sex, were enrolled from 5 centres across Canada for the Canadian ALS Neuroimaging Consortium study. Resting-state functional MRI, diffusion tensor imaging and magnetic resonance spectroscopy data were acquired. Functional connectivity maps, diffusion metrics and neurometabolite ratios were obtained from the analyses of the acquired multimodal data. A clinical assessment of foot tapping (frequency) was performed to examine upper motor neuron function in all participants. RESULTS Compared with healthy controls, the primary motor cortex in ALS showed reduced functional connectivity with sensory (T=5.21), frontal (T=3.70), temporal (T=3.80), putaminal (T=4.03) and adjacent motor (T=4.60) regions. In the primary motor cortex, N-acetyl aspartate (NAA, a neuronal marker) ratios and diffusion metrics (mean, axial and radial diffusivity, fractional anisotropy (FA)) were altered. Within the ALS cohort, foot tapping frequency correlated with NAA (r=0.347) and white matter FA (r=0.537). NAA levels showed associations with disturbed functional connectivity of the motor cortex. CONCLUSION In vivo neurochemistry may represent an effective imaging marker of impaired motor cortex functional connectivity in ALS.
Collapse
Affiliation(s)
- Avyarthana Dey
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada.,Division of Neurology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Collin C Luk
- Division of Neurology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Abdullah Ishaque
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada.,Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Daniel Ta
- Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Ojas Srivastava
- Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Dennell Krebs
- Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Peter Seres
- Department of Biomedical Engineering, University of Alberta, Edmonton, Alberta, Canada
| | - Chris Hanstock
- Department of Biomedical Engineering, University of Alberta, Edmonton, Alberta, Canada
| | - Christian Beaulieu
- Department of Biomedical Engineering, University of Alberta, Edmonton, Alberta, Canada
| | - Lawrence Korngut
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Richard Frayne
- Seaman Family Magnetic Resonance Research Centre, Foothills Medical Centre, Alberta Health Services, Calgary, Alberta, Canada.,Department of Radiology, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Lorne Zinman
- Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Simon Graham
- Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Angela Genge
- The Montreal Neurological Institute and Hospital, McGill University, Montreal, Québec, Canada
| | - Hannah Briemberg
- Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Sanjay Kalra
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada .,Division of Neurology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | | |
Collapse
|
17
|
Hippocampal Metabolic Alterations in Amyotrophic Lateral Sclerosis: A Magnetic Resonance Spectroscopy Study. Life (Basel) 2023; 13:life13020571. [PMID: 36836928 PMCID: PMC9965919 DOI: 10.3390/life13020571] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 02/15/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
BACKGROUND Magnetic resonance spectroscopy (MRS) in amyotrophic lateral sclerosis (ALS) has been overwhelmingly applied to motor regions to date and our understanding of frontotemporal metabolic signatures is relatively limited. The association between metabolic alterations and cognitive performance in also poorly characterised. MATERIAL AND METHODS In a multimodal, prospective pilot study, the structural, metabolic, and diffusivity profile of the hippocampus was systematically evaluated in patients with ALS. Patients underwent careful clinical and neurocognitive assessments. All patients were non-demented and exhibited normal memory performance. 1H-MRS spectra of the right and left hippocampi were acquired at 3.0T to determine the concentration of a panel of metabolites. The imaging protocol also included high-resolution T1-weighted structural imaging for subsequent hippocampal grey matter (GM) analyses and diffusion tensor imaging (DTI) for the tractographic evaluation of the integrity of the hippocampal perforant pathway zone (PPZ). RESULTS ALS patients exhibited higher hippocampal tNAA, tNAA/tCr and tCho bilaterally, despite the absence of volumetric and PPZ diffusivity differences between the two groups. Furthermore, superior memory performance was associated with higher hippocampal tNAA/tCr bilaterally. Both longer symptom duration and greater functional disability correlated with higher tCho levels. CONCLUSION Hippocampal 1H-MRS may not only contribute to a better academic understanding of extra-motor disease burden in ALS, but given its sensitive correlations with validated clinical metrics, it may serve as practical biomarker for future clinical and clinical trial applications. Neuroimaging protocols in ALS should incorporate MRS in addition to standard structural, functional, and diffusion sequences.
Collapse
|
18
|
Chen X, Zhou L, Cui C, Sun J. Evolving markers in amyotrophic lateral sclerosis. Adv Clin Chem 2023. [DOI: 10.1016/bs.acc.2023.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/15/2023]
|
19
|
Ashhurst JF, Tu S, Timmins HC, Kiernan MC. Progress, development, and challenges in amyotrophic lateral sclerosis clinical trials. Expert Rev Neurother 2022; 22:905-913. [PMID: 36543326 DOI: 10.1080/14737175.2022.2161893] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Amyotrophic Lateral Sclerosis (ALS) brings unique challenges to a clinical trial setting, due in part to relatively low disease prevalence coupled with a poor prognosis, in addition to the complexities linked to disease heterogeneity. As critical understanding of the disease develops, particularly in relation to clinical phenotype and the mechanisms of disease progression, so too new concepts evolve in relation to clinical trials, including the advent of precision therapy, targeted to subgroups of ALS patients. AREAS COVERED Individualized, or precision medicine in ALS recognizes the heterogeneous nature of the disease and utilizes information such as the clinical phenotype of the disease, clinical biomarkers, and genotyping to promote a tailored approach to treatment. Separate to these considerations, the present review will discuss clinical trial design and how this can be improved to better match patient and investigator needs in ALS clinical trials. EXPERT OPINION Precision therapy will promote a more focused treatment approach, with the goal of improving clinical outcomes for ALS patients. An increased community awareness of ALS, coupled with significant industry and philanthropic funding for ALS research, is accelerating this process.
Collapse
Affiliation(s)
| | - Sicong Tu
- Brain and Mind Centre, University of Sydney, Camperdown, Australia
| | - Hannah C Timmins
- Brain and Mind Centre, University of Sydney, Camperdown, Australia
| | - Matthew C Kiernan
- Brain and Mind Centre, University of Sydney, Camperdown, Australia.,Department of Neurology, Royal Prince Alfred Hospital, Sydney, Australia
| |
Collapse
|
20
|
Reddy DS, Abeygunaratne HN. Experimental and Clinical Biomarkers for Progressive Evaluation of Neuropathology and Therapeutic Interventions for Acute and Chronic Neurological Disorders. Int J Mol Sci 2022; 23:11734. [PMID: 36233034 PMCID: PMC9570151 DOI: 10.3390/ijms231911734] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/27/2022] [Accepted: 09/28/2022] [Indexed: 11/27/2022] Open
Abstract
This article describes commonly used experimental and clinical biomarkers of neuronal injury and neurodegeneration for the evaluation of neuropathology and monitoring of therapeutic interventions. Biomarkers are vital for diagnostics of brain disease and therapeutic monitoring. A biomarker can be objectively measured and evaluated as a proxy indicator for the pathophysiological process or response to therapeutic interventions. There are complex hurdles in understanding the molecular pathophysiology of neurological disorders and the ability to diagnose them at initial stages. Novel biomarkers for neurological diseases may surpass these issues, especially for early identification of disease risk. Validated biomarkers can measure the severity and progression of both acute neuronal injury and chronic neurological diseases such as epilepsy, migraine, Alzheimer's disease, Parkinson's disease, Huntington's disease, traumatic brain injury, amyotrophic lateral sclerosis, multiple sclerosis, and other brain diseases. Biomarkers are deployed to study progression and response to treatment, including noninvasive imaging tools for both acute and chronic brain conditions. Neuronal biomarkers are classified into four core subtypes: blood-based, immunohistochemical-based, neuroimaging-based, and electrophysiological biomarkers. Neuronal conditions have progressive stages, such as acute injury, inflammation, neurodegeneration, and neurogenesis, which can serve as indices of pathological status. Biomarkers are critical for the targeted identification of specific molecules, cells, tissues, or proteins that dramatically alter throughout the progression of brain conditions. There has been tremendous progress with biomarkers in acute conditions and chronic diseases affecting the central nervous system.
Collapse
Affiliation(s)
- Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA
- Institute of Pharmacology and Neurotherapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA
- Intercollegiate School of Engineering Medicine, Texas A&M University, Houston, TX 77030, USA
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX 77843, USA
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Hasara Nethma Abeygunaratne
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA
- Institute of Pharmacology and Neurotherapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA
| |
Collapse
|
21
|
Juengling FD, Wuest F, Kalra S, Agosta F, Schirrmacher R, Thiel A, Thaiss W, Müller HP, Kassubek J. Simultaneous PET/MRI: The future gold standard for characterizing motor neuron disease-A clinico-radiological and neuroscientific perspective. Front Neurol 2022; 13:890425. [PMID: 36061999 PMCID: PMC9428135 DOI: 10.3389/fneur.2022.890425] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 07/20/2022] [Indexed: 01/18/2023] Open
Abstract
Neuroimaging assessment of motor neuron disease has turned into a cornerstone of its clinical workup. Amyotrophic lateral sclerosis (ALS), as a paradigmatic motor neuron disease, has been extensively studied by advanced neuroimaging methods, including molecular imaging by MRI and PET, furthering finer and more specific details of the cascade of ALS neurodegeneration and symptoms, facilitated by multicentric studies implementing novel methodologies. With an increase in multimodal neuroimaging data on ALS and an exponential improvement in neuroimaging technology, the need for harmonization of protocols and integration of their respective findings into a consistent model becomes mandatory. Integration of multimodal data into a model of a continuing cascade of functional loss also calls for the best attempt to correlate the different molecular imaging measurements as performed at the shortest inter-modality time intervals possible. As outlined in this perspective article, simultaneous PET/MRI, nowadays available at many neuroimaging research sites, offers the perspective of a one-stop shop for reproducible imaging biomarkers on neuronal damage and has the potential to become the new gold standard for characterizing motor neuron disease from the clinico-radiological and neuroscientific perspectives.
Collapse
Affiliation(s)
- Freimut D. Juengling
- Division of Oncologic Imaging, University of Alberta, Edmonton, AB, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Faculty of Medicine, University Bern, Bern, Switzerland
| | - Frank Wuest
- Division of Oncologic Imaging, University of Alberta, Edmonton, AB, Canada
| | - Sanjay Kalra
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Department of Neurology, University of Alberta, Edmonton, AB, Canada
| | - Federica Agosta
- Division of Neuroscience, San Raffaele Scientific Institute, University Vita Salute San Raffaele, Milan, Italy
| | - Ralf Schirrmacher
- Division of Oncologic Imaging, University of Alberta, Edmonton, AB, Canada
- Medical Isotope and Cyclotron Facility, University of Alberta, Edmonton, AB, Canada
| | - Alexander Thiel
- Lady Davis Institute for Medical Research, Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Wolfgang Thaiss
- Department of Nuclear Medicine, University of Ulm Medical Center, Ulm, Germany
- Department of Diagnostic and Interventional Radiology, University of Ulm Medical Center, Ulm, Germany
| | - Hans-Peter Müller
- Department of Neurology, Ulm University Medical Center, Ulm, Germany
| | - Jan Kassubek
- Department of Neurology, Ulm University Medical Center, Ulm, Germany
| |
Collapse
|
22
|
Müller HP, Nagel AM, Keidel F, Wunderlich A, Hübers A, Gast LV, Ludolph AC, Beer M, Kassubek J. Relaxation-weighted 23Na magnetic resonance imaging maps regional patterns of abnormal sodium concentrations in amyotrophic lateral sclerosis. Ther Adv Chronic Dis 2022; 13:20406223221109480. [PMID: 35837670 PMCID: PMC9274400 DOI: 10.1177/20406223221109480] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 06/01/2022] [Indexed: 11/30/2022] Open
Abstract
Objectives: Multiparametric magnetic resonance imaging (MRI) is established as a
technical instrument for the characterisation of patients with amyotrophic
lateral sclerosis (ALS). The contribution of relaxation-weighted sodium
(23NaR) MRI remains to be defined. The aim of this study is
to apply 23NaR MRI to investigate brain sodium homeostasis and
map potential alterations in patients with ALS as compared with healthy
controls. Materials and Methods: Seventeen patients with ALS (mean age 61.1 ± 11.4 years, m/f = 9/8) and 10
healthy control subjects (mean age 60.3 ± 15.3 years, m/f = 6/4) were
examined by 23NaR MRI at 3 T. Regional sodium maps were obtained
by the calculation of the weighted difference from two image data sets with
different echo times (TE1 = 0.3 ms, TE2 = 25 ms).
Voxel-based analysis of the relaxation-weighted maps, together with
23Na concentration maps for comparison, was performed. Results: ROI-based analyses of relaxation-weighted brain sodium concentration maps
demonstrated increased sodium concentrations in the upper corticospinal
tracts and in the frontal lobes in patients with ALS; no differences between
ALS patients and controls were found in reference ROIs, where no involvement
in ALS-associated neurodegeneration could be anticipated. Conclusion: 23NaR MRI mapped regional alterations within disease-relevant
areas in ALS which correspond to the stages of the central nervous system
(CNS) pathology, providing evidence that the technique is a potential
biological marker of the cerebral neurodegenerative process in ALS.
Collapse
Affiliation(s)
| | - Armin M Nagel
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Franziska Keidel
- Department of Diagnostic and Interventional Radiology, University of Ulm, Ulm, Germany
| | - Arthur Wunderlich
- Department of Diagnostic and Interventional Radiology, University of Ulm, Ulm, Germany
| | | | - Lena V Gast
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Albert C Ludolph
- Department of Neurology, University of Ulm, Ulm, Germany German Center for Neurodegenerative Diseases (DZNE), Ulm, Germany
| | - Meinrad Beer
- Department of Diagnostic and Interventional Radiology, University of Ulm, Ulm, Germany
| | - Jan Kassubek
- Department of Neurology, University of Ulm, Oberer Eselsberg 45, Ulm 89081, Germany
| |
Collapse
|
23
|
Reyes-Leiva D, Dols-Icardo O, Sirisi S, Cortés-Vicente E, Turon-Sans J, de Luna N, Blesa R, Belbin O, Montal V, Alcolea D, Fortea J, Lleó A, Rojas-García R, Illán-Gala I. Pathophysiological Underpinnings of Extra-Motor Neurodegeneration in Amyotrophic Lateral Sclerosis: New Insights From Biomarker Studies. Front Neurol 2022; 12:750543. [PMID: 35115992 PMCID: PMC8804092 DOI: 10.3389/fneur.2021.750543] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 12/09/2021] [Indexed: 11/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD) lie at opposing ends of a clinical, genetic, and neuropathological continuum. In the last decade, it has become clear that cognitive and behavioral changes in patients with ALS are more frequent than previously recognized. Significantly, these non-motor features can impact the diagnosis, prognosis, and management of ALS. Partially overlapping neuropathological staging systems have been proposed to describe the distribution of TAR DNA-binding protein 43 (TDP-43) aggregates outside the corticospinal tract. However, the relationship between TDP-43 inclusions and neurodegeneration is not absolute and other pathophysiological processes, such as neuroinflammation (with a prominent role of microglia), cortical hyperexcitability, and synaptic dysfunction also play a central role in ALS pathophysiology. In the last decade, imaging and biofluid biomarker studies have revealed important insights into the pathophysiological underpinnings of extra-motor neurodegeneration in the ALS-FTLD continuum. In this review, we first summarize the clinical and pathophysiological correlates of extra-motor neurodegeneration in ALS. Next, we discuss the diagnostic and prognostic value of biomarkers in ALS and their potential to characterize extra-motor neurodegeneration. Finally, we debate about how biomarkers could improve the diagnosis and classification of ALS. Emerging imaging biomarkers of extra-motor neurodegeneration that enable the monitoring of disease progression are particularly promising. In addition, a growing arsenal of biofluid biomarkers linked to neurodegeneration and neuroinflammation are improving the diagnostic accuracy and identification of patients with a faster progression rate. The development and validation of biomarkers that detect the pathological aggregates of TDP-43 in vivo are notably expected to further elucidate the pathophysiological underpinnings of extra-motor neurodegeneration in ALS. Novel biomarkers tracking the different aspects of ALS pathophysiology are paving the way to precision medicine approaches in the ALS-FTLD continuum. These are essential steps to improve the diagnosis and staging of ALS and the design of clinical trials testing novel disease-modifying treatments.
Collapse
Affiliation(s)
- David Reyes-Leiva
- Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, CIBERER, Valencia, Spain
| | - Oriol Dols-Icardo
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Sonia Sirisi
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Elena Cortés-Vicente
- Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, CIBERER, Valencia, Spain
| | - Janina Turon-Sans
- Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, CIBERER, Valencia, Spain
| | - Noemi de Luna
- Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, CIBERER, Valencia, Spain
| | - Rafael Blesa
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Olivia Belbin
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Victor Montal
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Daniel Alcolea
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Juan Fortea
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Alberto Lleó
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Ricard Rojas-García
- Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, CIBERER, Valencia, Spain
| | - Ignacio Illán-Gala
- Sant Pau Memory Unit, Department of Neurology, Biomedical Research Institute Sant Pau, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
- *Correspondence: Ignacio Illán-Gala
| |
Collapse
|
24
|
Matthews DC, Mao X, Dowd K, Tsakanikas D, Jiang CS, Meuser C, Andrews RD, Lukic AS, Lee J, Hampilos N, Shafiian N, Sano M, David Mozley P, Fillit H, McEwen BS, Shungu DC, Pereira AC. Riluzole, a glutamate modulator, slows cerebral glucose metabolism decline in patients with Alzheimer's disease. Brain 2021; 144:3742-3755. [PMID: 34145880 PMCID: PMC8719848 DOI: 10.1093/brain/awab222] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 04/07/2021] [Accepted: 05/22/2021] [Indexed: 11/14/2022] Open
Abstract
Dysregulation of glutamatergic neural circuits has been implicated in a cycle of toxicity, believed among the neurobiological underpinning of Alzheimer's disease. Previously, we reported preclinical evidence that the glutamate modulator riluzole, which is FDA approved for the treatment of amyotrophic lateral sclerosis, has potential benefits on cognition, structural and molecular markers of ageing and Alzheimer's disease. The objective of this study was to evaluate in a pilot clinical trial, using neuroimaging biomarkers, the potential efficacy and safety of riluzole in patients with Alzheimer's disease as compared to placebo. A 6-month phase 2 double-blind, randomized, placebo-controlled study was conducted at two sites. Participants consisted of males and females, 50 to 95 years of age, with a clinical diagnosis of probable Alzheimer's disease, and Mini-Mental State Examination between 19 and 27. Ninety-four participants were screened, 50 participants who met inclusion criteria were randomly assigned to receive 50 mg riluzole (n = 26) or placebo (n = 24) twice a day. Twenty-two riluzole-treated and 20 placebo participants completed the study. Primary end points were baseline to 6 months changes in (i) cerebral glucose metabolism as measured with fluorodeoxyglucose-PET in prespecified regions of interest (hippocampus, posterior cingulate, precuneus, lateral temporal, inferior parietal, frontal); and (ii) changes in posterior cingulate levels of the neuronal viability marker N-acetylaspartate as measured with in vivo proton magnetic resonance spectroscopy. Secondary outcome measures were neuropsychological testing for correlation with neuroimaging biomarkers and in vivo measures of glutamate in posterior cingulate measured with magnetic resonance spectroscopy as a potential marker of target engagement. Measures of cerebral glucose metabolism, a well-established Alzheimer's disease biomarker and predictor of disease progression, declined significantly less in several prespecified regions of interest with the most robust effect in posterior cingulate, and effects in precuneus, lateral temporal, right hippocampus and frontal cortex in riluzole-treated participants in comparison to the placebo group. No group effect was found in measures of N-acetylaspartate levels. A positive correlation was observed between cognitive measures and regional cerebral glucose metabolism. A group × visit interaction was observed in glutamate levels in posterior cingulate, potentially suggesting engagement of glutamatergic system by riluzole. In vivo glutamate levels positively correlated with cognitive performance. These findings support our main primary hypothesis that cerebral glucose metabolism would be better preserved in the riluzole-treated group than in the placebo group and provide a rationale for more powered, longer duration studies of riluzole as a potential intervention for Alzheimer's disease.
Collapse
Affiliation(s)
| | - Xiangling Mao
- Department of Radiology, Weill Cornell Medicine, New York, NY 10021, USA
| | | | | | | | - Caroline Meuser
- Department of Psychiatry, Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | | | - Ana S Lukic
- ADM Diagnostics Inc., Northbrook, IL 60062, USA
| | - Jihyun Lee
- Department of Radiology, Weill Cornell Medicine, New York, NY 10021, USA
| | - Nicholas Hampilos
- Department of Radiology, Weill Cornell Medicine, New York, NY 10021, USA
| | - Neeva Shafiian
- Department of Neurology, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Mary Sano
- Department of Psychiatry, Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - P David Mozley
- Department of Radiology, Weill Cornell Medicine, New York, NY 10021, USA
| | - Howard Fillit
- Alzheimer's Drug Discovery Foundation, New York, NY 10019, USA
| | | | - Dikoma C Shungu
- Department of Radiology, Weill Cornell Medicine, New York, NY 10021, USA
| | - Ana C Pereira
- The Rockefeller University, New York, NY 10065, USA
- Department of Neurology, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| |
Collapse
|
25
|
Weise D, Menze I, Metelmann MCF, Woost TB, Classen J, Otto Pelz J. Multimodal assessment of autonomic dysfunction in amyotrophic lateral sclerosis. Eur J Neurol 2021; 29:715-723. [PMID: 34748270 DOI: 10.1111/ene.15177] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 10/05/2021] [Accepted: 10/20/2021] [Indexed: 01/15/2023]
Abstract
BACKGROUND AND PURPOSE Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder with predominant progressive degeneration of motor neurons and motor deficits, but non-motor symptoms (NMS) such as cognitive and behavioural deficits are frequent and underestimated in current diagnostic pathways. Autonomic dysfunction has occasionally been described, although its frequency and relevance are unclear. The aim of this study was to investigate the role of the autonomic nervous system in ALS using a multimodal approach. METHODS Thirty-seven ALS patients and 40 healthy sex- and age-matched controls were included. NMS were studied with the NMS assessment scale for Parkinson's disease and an autonomic subscale was calculated. Cardioautonomic innervation at rest and whilst standing was assessed by different parameters of heart rate variability. Morphological changes (cross-sectional area) of the vagus and median nerves for control were measured with high-resolution ultrasound. RESULTS Non-motor symptoms in general were more frequent in ALS patients and correlated inversely with the ALS Functional Rating Scale whereas the autonomic subscore of the NMS assessment scale for Parkinson's disease did not differ between the two groups and was not related to functional impairment. Cardioautonomic assessment solely revealed an increased heart rate at rest in ALS patients, whereas the other heart rate variability parameters did not differ from controls. Structural sonographic investigation of the vagus and median nerves was similar in both groups. CONCLUSIONS Using a multimodal approach evidence was found for a rather mild cardio-sympathetic overactivity in ALS patients. Overall, autonomic dysfunction seems to be subtle and is not related to the functional state of ALS patients.
Collapse
Affiliation(s)
- David Weise
- Department of Neurology, Leipzig University Hospital, Leipzig, Germany.,Department of Neurology, Asklepios Fachklinikum Stadtroda, Stadtroda, Germany
| | - Ina Menze
- Department of Neurology, Leipzig University Hospital, Leipzig, Germany
| | | | - Timo B Woost
- Department of Neurology, Leipzig University Hospital, Leipzig, Germany.,Department of Psychiatry and Psychotherapy, Center for Psychosocial Medicine, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Joseph Classen
- Department of Neurology, Leipzig University Hospital, Leipzig, Germany
| | - Johann Otto Pelz
- Department of Neurology, Leipzig University Hospital, Leipzig, Germany
| |
Collapse
|
26
|
Caldwell S, Rothman DL. 1H Magnetic Resonance Spectroscopy to Understand the Biological Basis of ALS, Diagnose Patients Earlier, and Monitor Disease Progression. Front Neurol 2021; 12:701170. [PMID: 34512519 PMCID: PMC8429815 DOI: 10.3389/fneur.2021.701170] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 08/03/2021] [Indexed: 11/13/2022] Open
Abstract
At present, limited biomarkers exist to reliably understand, diagnose, and monitor the progression of amyotrophic lateral sclerosis (ALS), a fatal neurological disease characterized by motor neuron death. Standard MRI technology can only be used to exclude a diagnosis of ALS, but 1H-MRS technology, which measures neurochemical composition, may provide the unique ability to reveal biomarkers that are specific to ALS and sensitive enough to diagnose patients at early stages in disease progression. In this review, we present a summary of current theories of how mitochondrial energetics and an altered glutamate/GABA neurotransmitter flux balance play a role in the pathogenesis of ALS. The theories are synthesized into a model that predicts how pathogenesis impacts glutamate and GABA concentrations. When compared with the results of all MRS studies published to date that measure the absolute concentrations of these neurochemicals in ALS patients, results were variable. However, when normalized for neuronal volume using the MRS biomarker N-acetyl aspartate (NAA), there is clear evidence for an elevation of neuronal glutamate in nine out of thirteen studies reviewed, an observation consistent with the predictions of the model of increased activity of glutamatergic neurons and excitotoxicity. We propose that this increase in neuronal glutamate concentration, in combination with decreased neuronal volume, is specific to the pathology of ALS. In addition, when normalized to glutamate levels, there is clear evidence for a decrease in neuronal GABA in three out of four possible studies reviewed, a finding consistent with a loss of inhibitory regulation contributing to excessive neuronal excitability. The combination of a decreased GABA/Glx ratio with an elevated Glx/NAA ratio may enhance the specificity for 1H-MRS detection of ALS and ability to monitor glutamatergic and GABAergic targeted therapeutics. Additional longitudinal studies calculating the exact value of these ratios are needed to test these hypotheses and understand how ratios may change over the course of disease progression. Proposed modifications to the experimental design of the reviewed 1H MRS studies may also increase the sensitivity of the technology to changes in these neurochemicals, particularly in early stages of disease progression.
Collapse
Affiliation(s)
- Sarah Caldwell
- Departments of Radiology and Biomedical Engineering, Magnetic Resonance Research Center, Yale University School of Medicine, New Haven, CT, United States
| | - Douglas L Rothman
- Departments of Radiology and Biomedical Engineering, Magnetic Resonance Research Center, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
27
|
Ta D, Ishaque A, Srivastava O, Hanstock C, Seres P, Eurich DT, Luk C, Briemberg H, Frayne R, Genge AL, Graham SJ, Korngut L, Zinman L, Kalra S. Progressive Neurochemical Abnormalities in Cognitive and Motor Subgroups of Amyotrophic Lateral Sclerosis: A Prospective Multicenter Study. Neurology 2021; 97:e803-e813. [PMID: 34426551 PMCID: PMC8397589 DOI: 10.1212/wnl.0000000000012367] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 05/19/2021] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To evaluate progressive cerebral degeneration in amyotrophic lateral sclerosis (ALS) by assessing alterations in N-acetylaspartate (NAA) ratios in the motor and prefrontal cortex within clinical subgroups of ALS. METHODS Seventy-six patients with ALS and 59 healthy controls were enrolled in a prospective, longitudinal, multicenter study in the Canadian ALS Neuroimaging Consortium. Participants underwent serial clinical evaluations and magnetic resonance spectroscopy at baseline and 4 and 8 months using a harmonized protocol across 5 centers. NAA ratios were quantified in the motor cortex and prefrontal cortex. Patients were stratified into subgroups based on disease progression rate, upper motor neuron (UMN) signs, and cognitive status. Linear mixed models were used for baseline and longitudinal comparisons of NAA metabolite ratios. RESULTS Patients with ALS had reduced NAA ratios in the motor cortex at baseline (p < 0.001). Ratios were lower in those with more rapid disease progression and greater UMN signs (p < 0.05). A longitudinal decline in NAA ratios was observed in the motor cortex in the rapidly progressing (p < 0.01) and high UMN burden (p < 0.01) cohorts. The severity of UMN signs did not change significantly over time. NAA ratios were reduced in the prefrontal cortex only in cognitively impaired patients (p < 0.05); prefrontal cortex metabolites did not change over time. CONCLUSIONS Progressive degeneration of the motor cortex in ALS is associated with more aggressive clinical presentations. These findings provide biological evidence of variable spatial and temporal cerebral degeneration linked to the disease heterogeneity of ALS. The use of standardized imaging protocols may have a role in clinical trials for patient selection or subgrouping. CLASSIFICATION OF EVIDENCE This study provides Class II evidence that MRS NAA metabolite ratios of the motor cortex are associated with more rapid disease progression and greater UMN signs in patients with ALS. TRIAL REGISTRATION INFORMATION ClinicalTrials.gov Identifier: NCT02405182.
Collapse
Affiliation(s)
- Daniel Ta
- From the Neuroscience and Mental Health Institute (D.T., A.I., O.S., S.K.), Department of Biomedical Engineering (C.H., P.S.), School of Public Health (D.T.E.), and Division of Neurology (C.L., S.K.), University of Alberta, Edmonton; Division of Neurology (H.B.), University of British Columbia, Vancouver; Seaman Family MR Centre (R.F.) and Hotchkiss Brain Institute (R.F., L.K.), University of Calgary, Alberta; Montreal Neurological Institute (A.L.G.), McGill University, Quebec; and Sunnybrook Health Sciences Centre (S.J.G., L.Z.), University of Toronto, Ontario, Canada.
| | - Abdullah Ishaque
- From the Neuroscience and Mental Health Institute (D.T., A.I., O.S., S.K.), Department of Biomedical Engineering (C.H., P.S.), School of Public Health (D.T.E.), and Division of Neurology (C.L., S.K.), University of Alberta, Edmonton; Division of Neurology (H.B.), University of British Columbia, Vancouver; Seaman Family MR Centre (R.F.) and Hotchkiss Brain Institute (R.F., L.K.), University of Calgary, Alberta; Montreal Neurological Institute (A.L.G.), McGill University, Quebec; and Sunnybrook Health Sciences Centre (S.J.G., L.Z.), University of Toronto, Ontario, Canada
| | - Ojas Srivastava
- From the Neuroscience and Mental Health Institute (D.T., A.I., O.S., S.K.), Department of Biomedical Engineering (C.H., P.S.), School of Public Health (D.T.E.), and Division of Neurology (C.L., S.K.), University of Alberta, Edmonton; Division of Neurology (H.B.), University of British Columbia, Vancouver; Seaman Family MR Centre (R.F.) and Hotchkiss Brain Institute (R.F., L.K.), University of Calgary, Alberta; Montreal Neurological Institute (A.L.G.), McGill University, Quebec; and Sunnybrook Health Sciences Centre (S.J.G., L.Z.), University of Toronto, Ontario, Canada
| | - Chris Hanstock
- From the Neuroscience and Mental Health Institute (D.T., A.I., O.S., S.K.), Department of Biomedical Engineering (C.H., P.S.), School of Public Health (D.T.E.), and Division of Neurology (C.L., S.K.), University of Alberta, Edmonton; Division of Neurology (H.B.), University of British Columbia, Vancouver; Seaman Family MR Centre (R.F.) and Hotchkiss Brain Institute (R.F., L.K.), University of Calgary, Alberta; Montreal Neurological Institute (A.L.G.), McGill University, Quebec; and Sunnybrook Health Sciences Centre (S.J.G., L.Z.), University of Toronto, Ontario, Canada
| | - Peter Seres
- From the Neuroscience and Mental Health Institute (D.T., A.I., O.S., S.K.), Department of Biomedical Engineering (C.H., P.S.), School of Public Health (D.T.E.), and Division of Neurology (C.L., S.K.), University of Alberta, Edmonton; Division of Neurology (H.B.), University of British Columbia, Vancouver; Seaman Family MR Centre (R.F.) and Hotchkiss Brain Institute (R.F., L.K.), University of Calgary, Alberta; Montreal Neurological Institute (A.L.G.), McGill University, Quebec; and Sunnybrook Health Sciences Centre (S.J.G., L.Z.), University of Toronto, Ontario, Canada
| | - Dean T Eurich
- From the Neuroscience and Mental Health Institute (D.T., A.I., O.S., S.K.), Department of Biomedical Engineering (C.H., P.S.), School of Public Health (D.T.E.), and Division of Neurology (C.L., S.K.), University of Alberta, Edmonton; Division of Neurology (H.B.), University of British Columbia, Vancouver; Seaman Family MR Centre (R.F.) and Hotchkiss Brain Institute (R.F., L.K.), University of Calgary, Alberta; Montreal Neurological Institute (A.L.G.), McGill University, Quebec; and Sunnybrook Health Sciences Centre (S.J.G., L.Z.), University of Toronto, Ontario, Canada
| | - Collin Luk
- From the Neuroscience and Mental Health Institute (D.T., A.I., O.S., S.K.), Department of Biomedical Engineering (C.H., P.S.), School of Public Health (D.T.E.), and Division of Neurology (C.L., S.K.), University of Alberta, Edmonton; Division of Neurology (H.B.), University of British Columbia, Vancouver; Seaman Family MR Centre (R.F.) and Hotchkiss Brain Institute (R.F., L.K.), University of Calgary, Alberta; Montreal Neurological Institute (A.L.G.), McGill University, Quebec; and Sunnybrook Health Sciences Centre (S.J.G., L.Z.), University of Toronto, Ontario, Canada
| | - Hannah Briemberg
- From the Neuroscience and Mental Health Institute (D.T., A.I., O.S., S.K.), Department of Biomedical Engineering (C.H., P.S.), School of Public Health (D.T.E.), and Division of Neurology (C.L., S.K.), University of Alberta, Edmonton; Division of Neurology (H.B.), University of British Columbia, Vancouver; Seaman Family MR Centre (R.F.) and Hotchkiss Brain Institute (R.F., L.K.), University of Calgary, Alberta; Montreal Neurological Institute (A.L.G.), McGill University, Quebec; and Sunnybrook Health Sciences Centre (S.J.G., L.Z.), University of Toronto, Ontario, Canada
| | - Richard Frayne
- From the Neuroscience and Mental Health Institute (D.T., A.I., O.S., S.K.), Department of Biomedical Engineering (C.H., P.S.), School of Public Health (D.T.E.), and Division of Neurology (C.L., S.K.), University of Alberta, Edmonton; Division of Neurology (H.B.), University of British Columbia, Vancouver; Seaman Family MR Centre (R.F.) and Hotchkiss Brain Institute (R.F., L.K.), University of Calgary, Alberta; Montreal Neurological Institute (A.L.G.), McGill University, Quebec; and Sunnybrook Health Sciences Centre (S.J.G., L.Z.), University of Toronto, Ontario, Canada
| | - Angela L Genge
- From the Neuroscience and Mental Health Institute (D.T., A.I., O.S., S.K.), Department of Biomedical Engineering (C.H., P.S.), School of Public Health (D.T.E.), and Division of Neurology (C.L., S.K.), University of Alberta, Edmonton; Division of Neurology (H.B.), University of British Columbia, Vancouver; Seaman Family MR Centre (R.F.) and Hotchkiss Brain Institute (R.F., L.K.), University of Calgary, Alberta; Montreal Neurological Institute (A.L.G.), McGill University, Quebec; and Sunnybrook Health Sciences Centre (S.J.G., L.Z.), University of Toronto, Ontario, Canada
| | - Simon J Graham
- From the Neuroscience and Mental Health Institute (D.T., A.I., O.S., S.K.), Department of Biomedical Engineering (C.H., P.S.), School of Public Health (D.T.E.), and Division of Neurology (C.L., S.K.), University of Alberta, Edmonton; Division of Neurology (H.B.), University of British Columbia, Vancouver; Seaman Family MR Centre (R.F.) and Hotchkiss Brain Institute (R.F., L.K.), University of Calgary, Alberta; Montreal Neurological Institute (A.L.G.), McGill University, Quebec; and Sunnybrook Health Sciences Centre (S.J.G., L.Z.), University of Toronto, Ontario, Canada
| | - Lawrence Korngut
- From the Neuroscience and Mental Health Institute (D.T., A.I., O.S., S.K.), Department of Biomedical Engineering (C.H., P.S.), School of Public Health (D.T.E.), and Division of Neurology (C.L., S.K.), University of Alberta, Edmonton; Division of Neurology (H.B.), University of British Columbia, Vancouver; Seaman Family MR Centre (R.F.) and Hotchkiss Brain Institute (R.F., L.K.), University of Calgary, Alberta; Montreal Neurological Institute (A.L.G.), McGill University, Quebec; and Sunnybrook Health Sciences Centre (S.J.G., L.Z.), University of Toronto, Ontario, Canada
| | - Lorne Zinman
- From the Neuroscience and Mental Health Institute (D.T., A.I., O.S., S.K.), Department of Biomedical Engineering (C.H., P.S.), School of Public Health (D.T.E.), and Division of Neurology (C.L., S.K.), University of Alberta, Edmonton; Division of Neurology (H.B.), University of British Columbia, Vancouver; Seaman Family MR Centre (R.F.) and Hotchkiss Brain Institute (R.F., L.K.), University of Calgary, Alberta; Montreal Neurological Institute (A.L.G.), McGill University, Quebec; and Sunnybrook Health Sciences Centre (S.J.G., L.Z.), University of Toronto, Ontario, Canada
| | - Sanjay Kalra
- From the Neuroscience and Mental Health Institute (D.T., A.I., O.S., S.K.), Department of Biomedical Engineering (C.H., P.S.), School of Public Health (D.T.E.), and Division of Neurology (C.L., S.K.), University of Alberta, Edmonton; Division of Neurology (H.B.), University of British Columbia, Vancouver; Seaman Family MR Centre (R.F.) and Hotchkiss Brain Institute (R.F., L.K.), University of Calgary, Alberta; Montreal Neurological Institute (A.L.G.), McGill University, Quebec; and Sunnybrook Health Sciences Centre (S.J.G., L.Z.), University of Toronto, Ontario, Canada.
| |
Collapse
|
28
|
Mitolo M, Stanzani-Maserati M, Manners DN, Capellari S, Testa C, Talozzi L, Poda R, Oppi F, Evangelisti S, Gramegna LL, Magarelli S, Pantieri R, Liguori R, Lodi R, Tonon C. The Combination of Metabolic Posterior Cingulate Cortical Abnormalities and Structural Asymmetries Improves the Differential Diagnosis Between Primary Progressive Aphasia and Alzheimer's Disease. J Alzheimers Dis 2021; 82:1467-1473. [PMID: 34151798 DOI: 10.3233/jad-210211] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Differential diagnosis between primary progressive aphasia (PPA) and Alzheimer's disease (AD) could be difficult if based on clinical grounds alone. We evaluated the combination of proton MR spectroscopy of posterior cingulate cortex (PCC) and quantitative structural imaging asymmetries to differentiate PPA from AD patients. A greater left-lateralized temporo-parietal atrophy (higher accuracy for the PCC, 81.4%) and metabolic neurodegenerative changes in PCC (accuracy 76.8%) was demonstrated in PPA versus AD. The combined multiparametric approach increased the accuracy to 94%in the differential diagnosis between these two neurodegenerative diseases.
Collapse
Affiliation(s)
- Micaela Mitolo
- Functional and Molecular Neuroimaging Unit, IRCCS Istituto delle Scienze Neurologiche di Bologna, OUC Neurologia, Bologna, Italy
| | | | - David N Manners
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy
| | - Sabina Capellari
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna, Italy.,Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy
| | - Claudia Testa
- Department of Physics and Astronomy, University of Bologna, Bologna, Italy
| | - Lia Talozzi
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy
| | - Roberto Poda
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna, Italy
| | - Federico Oppi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna, Italy
| | - Stefania Evangelisti
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy
| | - Laura L Gramegna
- Functional and Molecular Neuroimaging Unit, IRCCS Istituto delle Scienze Neurologiche di Bologna, OUC Neurologia, Bologna, Italy.,Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy
| | - Silvia Magarelli
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Neurologia, Bologna, Italy
| | - Roberta Pantieri
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Neurologia, Bologna, Italy
| | - Rocco Liguori
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna, Italy.,Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy
| | - Raffaele Lodi
- Functional and Molecular Neuroimaging Unit, IRCCS Istituto delle Scienze Neurologiche di Bologna, OUC Neurologia, Bologna, Italy.,Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy
| | - Caterina Tonon
- Functional and Molecular Neuroimaging Unit, IRCCS Istituto delle Scienze Neurologiche di Bologna, OUC Neurologia, Bologna, Italy.,Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
29
|
Sassani M, Alix JJ, McDermott CJ, Baster K, Hoggard N, Wild JM, Mortiboys HJ, Shaw PJ, Wilkinson ID, Jenkins TM. Magnetic resonance spectroscopy reveals mitochondrial dysfunction in amyotrophic lateral sclerosis. Brain 2021; 143:3603-3618. [PMID: 33439988 DOI: 10.1093/brain/awaa340] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 07/16/2020] [Accepted: 08/08/2020] [Indexed: 12/16/2022] Open
Abstract
Mitochondrial dysfunction is postulated to be central to amyotrophic lateral sclerosis (ALS) pathophysiology. Evidence comes primarily from disease models and conclusive data to support bioenergetic dysfunction in vivo in patients is currently lacking. This study is the first to assess mitochondrial dysfunction in brain and muscle in individuals living with ALS using 31P-magnetic resonance spectroscopy (MRS), the modality of choice to assess energy metabolism in vivo. We recruited 20 patients and 10 healthy age and gender-matched control subjects in this cross-sectional clinico-radiological study. 31P-MRS was acquired from cerebral motor regions and from tibialis anterior during rest and exercise. Bioenergetic parameter estimates were derived including: ATP, phosphocreatine, inorganic phosphate, adenosine diphosphate, Gibbs free energy of ATP hydrolysis (ΔGATP), phosphomonoesters, phosphodiesters, pH, free magnesium concentration, and muscle dynamic recovery constants. Linear regression was used to test for associations between brain data and clinical parameters (revised amyotrophic functional rating scale, slow vital capacity, and upper motor neuron score) and between muscle data and clinico-neurophysiological measures (motor unit number and size indices, force of contraction, and speed of walking). Evidence for primary dysfunction of mitochondrial oxidative phosphorylation was detected in the brainstem where ΔGATP and phosphocreatine were reduced. Alterations were also detected in skeletal muscle in patients where resting inorganic phosphate, pH, and phosphomonoesters were increased, whereas resting ΔGATP, magnesium, and dynamic phosphocreatine to inorganic phosphate recovery were decreased. Phosphocreatine in brainstem correlated with respiratory dysfunction and disability; in muscle, energy metabolites correlated with motor unit number index, muscle power, and speed of walking. This study provides in vivo evidence for bioenergetic dysfunction in ALS in brain and skeletal muscle, which appears clinically and electrophysiologically relevant. 31P-MRS represents a promising technique to assess the pathophysiology of mitochondrial function in vivo in ALS and a potential tool for future clinical trials targeting bioenergetic dysfunction.
Collapse
Affiliation(s)
- Matilde Sassani
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - James J Alix
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Christopher J McDermott
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Kathleen Baster
- Statistical Service Unit, University of Sheffield, Sheffield, UK
| | - Nigel Hoggard
- Academic Unit of Radiology, University of Sheffield, Sheffield, UK
| | - Jim M Wild
- Academic Unit of Radiology, University of Sheffield, Sheffield, UK
| | - Heather J Mortiboys
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Pamela J Shaw
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Iain D Wilkinson
- Academic Unit of Radiology, University of Sheffield, Sheffield, UK
| | - Thomas M Jenkins
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| |
Collapse
|
30
|
Gafson AR, Barthélemy NR, Bomont P, Carare RO, Durham HD, Julien JP, Kuhle J, Leppert D, Nixon RA, Weller RO, Zetterberg H, Matthews PM. Neurofilaments: neurobiological foundations for biomarker applications. Brain 2020; 143:1975-1998. [PMID: 32408345 DOI: 10.1093/brain/awaa098] [Citation(s) in RCA: 204] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 12/20/2019] [Accepted: 01/20/2020] [Indexed: 12/11/2022] Open
Abstract
Interest in neurofilaments has risen sharply in recent years with recognition of their potential as biomarkers of brain injury or neurodegeneration in CSF and blood. This is in the context of a growing appreciation for the complexity of the neurobiology of neurofilaments, new recognition of specialized roles for neurofilaments in synapses and a developing understanding of mechanisms responsible for their turnover. Here we will review the neurobiology of neurofilament proteins, describing current understanding of their structure and function, including recently discovered evidence for their roles in synapses. We will explore emerging understanding of the mechanisms of neurofilament degradation and clearance and review new methods for future elucidation of the kinetics of their turnover in humans. Primary roles of neurofilaments in the pathogenesis of human diseases will be described. With this background, we then will review critically evidence supporting use of neurofilament concentration measures as biomarkers of neuronal injury or degeneration. Finally, we will reflect on major challenges for studies of the neurobiology of intermediate filaments with specific attention to identifying what needs to be learned for more precise use and confident interpretation of neurofilament measures as biomarkers of neurodegeneration.
Collapse
Affiliation(s)
- Arie R Gafson
- Department of Brain Sciences, Imperial College, London, UK
| | - Nicolas R Barthélemy
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
| | - Pascale Bomont
- ATIP-Avenir team, INM, INSERM, Montpellier University, Montpellier, France
| | - Roxana O Carare
- Clinical Neurosciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Heather D Durham
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Québec, Canada
| | - Jean-Pierre Julien
- Department of Psychiatry and Neuroscience, Laval University, Quebec, Canada.,CERVO Brain Research Center, 2601 Chemin de la Canardière, Québec, QC, G1J 2G3, Canada
| | - Jens Kuhle
- Neurologic Clinic and Policlinic, Departments of Medicine, Biomedicine and Clinical Research, University Hospital Basel, University of Basel, Basel, Switzerland
| | - David Leppert
- Neurologic Clinic and Policlinic, Departments of Medicine, Biomedicine and Clinical Research, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Ralph A Nixon
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, 10962, USA.,Departments of Psychiatry, New York University School of Medicine, New York, NY, 10016, USA.,Neuroscience Institute, New York University School of Medicine, New York, NY, 10016, USA.,Department of Cell Biology, New York University School of Medicine, New York, NY, 10016, USA
| | - Roy O Weller
- Clinical Neurosciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Henrik Zetterberg
- University College London Queen Square Institute of Neurology, London, UK.,UK Dementia Research Institute at University College London, London, UK.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Paul M Matthews
- Department of Brain Sciences, Imperial College, London, UK.,UK Dementia Research Institute at Imperial College, London
| |
Collapse
|
31
|
Andronesi OC, Nicholson K, Jafari-Khouzani K, Bogner W, Wang J, Chan J, Macklin EA, Levine-Weinberg M, Breen C, Schwarzschild MA, Cudkowicz M, Rosen BR, Paganoni S, Ratai EM. Imaging Neurochemistry and Brain Structure Tracks Clinical Decline and Mechanisms of ALS in Patients. Front Neurol 2020; 11:590573. [PMID: 33343494 PMCID: PMC7744722 DOI: 10.3389/fneur.2020.590573] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 11/03/2020] [Indexed: 12/27/2022] Open
Abstract
Background: Oxidative stress and protein aggregation are key mechanisms in amyotrophic lateral sclerosis (ALS) disease. Reduced glutathione (GSH) is the most important intracellular antioxidant that protects neurons from reactive oxygen species. We hypothesized that levels of GSH measured by MR spectroscopic imaging (MRSI) in the motor cortex and corticospinal tract are linked to clinical trajectory of ALS patients. Objectives: Investigate the value of GSH imaging to probe clinical decline of ALS patients in combination with other neurochemical and structural parameters. Methods: Twenty-four ALS patients were imaged at 3 T with an advanced MR protocol. Mapping GSH levels in the brain is challenging, and for this purpose, we used an optimized spectral-edited 3D MRSI sequence with real-time motion and field correction to image glutathione and other brain metabolites. In addition, our imaging protocol included (i) an adiabatic T1ρ sequence to image macromolecular fraction of brain parenchyma, (ii) diffusion tensor imaging (DTI) for white matter tractography, and (iii) high-resolution anatomical imaging. Results: We found GSH in motor cortex (r = −0.431, p = 0.04) and corticospinal tract (r = −0.497, p = 0.016) inversely correlated with time between diagnosis and imaging. N-Acetyl-aspartate (NAA) in motor cortex inversely correlated (r = −0.416, p = 0.049), while mean water diffusivity (r = 0.437, p = 0.033) and T1ρ (r = 0.482, p = 0.019) positively correlated with disease progression measured by imputed change in revised ALS Functional Rating Scale. There is more decrease in the motor cortex than in the white matter for GSH compared to NAA, glutamate, and glutamine. Conclusions: Our study suggests that a panel of biochemical and structural imaging biomarkers defines a brain endophenotype, which can be used to time biological events and clinical progression in ALS patients. Such a quantitative brain endophenotype may stratify ALS patients into more homogeneous groups for therapeutic interventions compared to clinical criteria.
Collapse
Affiliation(s)
- Ovidiu C Andronesi
- Department of Radiology, A. A. Martinos Center for Biomedical Imaging, Harvard Medical School, Massachusetts General Hospital, Boston, MA, United States
| | - Katharine Nicholson
- Neurological Clinical Research Institute (NCRI), Massachusetts General Hospital, Boston, MA, United States
| | - Kourosh Jafari-Khouzani
- Department of Radiology, A. A. Martinos Center for Biomedical Imaging, Harvard Medical School, Massachusetts General Hospital, Boston, MA, United States
| | - Wolfgang Bogner
- High Field MR Centre, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Jing Wang
- Department of Radiology, A. A. Martinos Center for Biomedical Imaging, Harvard Medical School, Massachusetts General Hospital, Boston, MA, United States.,Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - James Chan
- Biostatistics Center, Massachusetts General Hospital, Boston, MA, United States
| | - Eric A Macklin
- Biostatistics Center, Massachusetts General Hospital, Boston, MA, United States
| | - Mark Levine-Weinberg
- Neurological Clinical Research Institute (NCRI), Massachusetts General Hospital, Boston, MA, United States
| | - Christopher Breen
- Neurological Clinical Research Institute (NCRI), Massachusetts General Hospital, Boston, MA, United States
| | | | - Merit Cudkowicz
- Neurological Clinical Research Institute (NCRI), Massachusetts General Hospital, Boston, MA, United States
| | - Bruce R Rosen
- Department of Radiology, A. A. Martinos Center for Biomedical Imaging, Harvard Medical School, Massachusetts General Hospital, Boston, MA, United States
| | - Sabrina Paganoni
- Neurological Clinical Research Institute (NCRI), Massachusetts General Hospital, Boston, MA, United States.,Spaulding Rehabilitation Hospital, Boston, MA, United States
| | - Eva-Maria Ratai
- Department of Radiology, A. A. Martinos Center for Biomedical Imaging, Harvard Medical School, Massachusetts General Hospital, Boston, MA, United States
| |
Collapse
|
32
|
Pioro EP, Turner MR, Bede P. Neuroimaging in primary lateral sclerosis. Amyotroph Lateral Scler Frontotemporal Degener 2020; 21:18-27. [PMID: 33602015 DOI: 10.1080/21678421.2020.1837176] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 10/08/2020] [Accepted: 10/09/2020] [Indexed: 12/15/2022]
Abstract
Increased interest in the underlying pathogenesis of primary lateral sclerosis (PLS) and its relationship to amyotrophic lateral sclerosis (ALS) has corresponded to a growing number of CNS imaging studies, especially in the past decade. Both its rarity and uncertainty of definite diagnosis prior to 4 years from symptom onset have resulted in PLS being less studied than ALS. In this review, we highlight most relevant papers applying magnetic resonance imaging (MRI), magnetic resonance spectroscopy (MRS), and positron emission tomography (PET) to analyzing CNS changes in PLS, often in relation to ALS. In patients with PLS, mostly brain, but also spinal cord has been evaluated since significant neurodegeneration is essentially restricted to upper motor neuron (UMN) structures and related pathways. Abnormalities of cortex and subcortical white matter tracts have been identified by structural and functional MRI and MRS studies, while metabolic and cell-specific changes in PLS brain have been revealed using various PET radiotracers. Future neuroimaging studies will continue to explore the interface between the PLS-ALS continuum, identify more changes unique to PLS, apply novel MRI and MRS sequences showing greater structural and neurochemical detail, as well as expand the repertoire of PET radiotracers that reveal various cellular pathologies. Neuroimaging has the potential to play an important role in the evaluation of novel therapies for patients with PLS.
Collapse
Affiliation(s)
- Erik P Pioro
- Section of ALS & Related Disorders, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Martin R Turner
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Peter Bede
- Computational Neuroimaging Group, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
33
|
Chipika RH, Christidi F, Finegan E, Li Hi Shing S, McKenna MC, Chang KM, Karavasilis E, Doherty MA, Hengeveld JC, Vajda A, Pender N, Hutchinson S, Donaghy C, McLaughlin RL, Hardiman O, Bede P. Amygdala pathology in amyotrophic lateral sclerosis and primary lateral sclerosis. J Neurol Sci 2020; 417:117039. [PMID: 32713609 DOI: 10.1016/j.jns.2020.117039] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/19/2020] [Accepted: 07/13/2020] [Indexed: 12/26/2022]
Abstract
Temporal lobe studies in motor neuron disease overwhelmingly focus on white matter alterations and cortical grey matter atrophy. Reports on amygdala involvement are conflicting and the amygdala is typically evaluated as single structure despite consisting of several functionally and cytologically distinct nuclei. A prospective, single-centre, neuroimaging study was undertaken to comprehensively characterise amygdala pathology in 100 genetically-stratified ALS patients, 33 patients with PLS and 117 healthy controls. The amygdala was segmented into groups of nuclei using a Bayesian parcellation algorithm based on a probabilistic atlas and shape deformations were additionally assessed by vertex analyses. The accessory basal nucleus (p = .021) and the cortical nucleus (p = .022) showed significant volume reductions in C9orf72 negative ALS patients compared to controls. The lateral nucleus (p = .043) and the cortico-amygdaloid transition (p = .024) were preferentially affected in C9orf72 hexanucleotide carriers. A trend of total volume reduction was identified in C9orf72 positive ALS patients (p = .055) which was also captured in inferior-medial shape deformations on vertex analyses. Our findings highlight that the amygdala is affected in ALS and our study demonstrates the selective involvement of specific nuclei as opposed to global atrophy. The genotype-specific patterns of amygdala involvement identified by this study are consistent with the growing literature of extra-motor clinical features. Mesial temporal lobe pathology in ALS is not limited to hippocampal pathology but, as a key hub of the limbic system, the amygdala is also affected in ALS.
Collapse
Affiliation(s)
- Rangariroyashe H Chipika
- Computational Neuroimaging Group, Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland
| | - Foteini Christidi
- Computational Neuroimaging Group, Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland; Department of Neurology, Aeginition Hospital, University of Athens, Greece
| | - Eoin Finegan
- Computational Neuroimaging Group, Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland
| | - Stacey Li Hi Shing
- Computational Neuroimaging Group, Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland
| | - Mary Clare McKenna
- Computational Neuroimaging Group, Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland
| | - Kai Ming Chang
- Computational Neuroimaging Group, Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland; Electronics and Computer Science, University of Southampton, Southampton SO17 1BJ, United Kingdom
| | - Efstratios Karavasilis
- Computational Neuroimaging Group, Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland; 2nd Department of Radiology, Attikon University Hospital, University of Athens, Athens, Greece
| | - Mark A Doherty
- Complex Trait Genomics Laboratory, Smurfit Institute of Genetics, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland
| | - Jennifer C Hengeveld
- Complex Trait Genomics Laboratory, Smurfit Institute of Genetics, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland
| | - Alice Vajda
- Complex Trait Genomics Laboratory, Smurfit Institute of Genetics, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland
| | - Niall Pender
- Department of psychology, Beaumont Hospital Dublin, Ireland
| | - Siobhan Hutchinson
- Department of Neurology, St James's Hospital, James's St, Ushers, Dublin 8 D08 NHY1, Ireland
| | - Colette Donaghy
- Department of Neurology, Belfast, Western Health & Social Care Trust, UK
| | - Russell L McLaughlin
- Complex Trait Genomics Laboratory, Smurfit Institute of Genetics, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland
| | - Orla Hardiman
- Computational Neuroimaging Group, Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland
| | - Peter Bede
- Computational Neuroimaging Group, Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland.
| |
Collapse
|
34
|
Hanstock C, Sun K, Choi C, Eurich D, Camicioli R, Johnston W, Kalra S. Spectroscopic markers of neurodegeneration in the mesial prefrontal cortex predict survival in ALS. Amyotroph Lateral Scler Frontotemporal Degener 2020; 21:246-251. [PMID: 32067510 DOI: 10.1080/21678421.2020.1727926] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background and objective: N-acetylaspartate (NAA) and myo-inositol (mIns) are spectroscopic markers of neuronal integrity and astrogliosis, respectively. We performed a survival analysis to determine the prognostic value of the NAA/mIns metabolite ratio in ALS after a period of two and five years. Methods: Twenty-four patients with ALS (two with ALS-FTD) were recruited to participate in a high-field MR spectroscopy study of the mesial prefrontal cortex. Univariate and multivariate Cox proportional hazards analyses were used to assess NAA/mIns as a predictor of survival alongside other demographic and clinical measures. Census dates were set at two and five years after the time of MR scan for each patient. Survival curves were calculated using the Kaplan-Meier method. Results: After a five-year observation period, 19 patients had died and five were still alive. Median survival time from date of scan was 1.95 years. Univariate and multivariate Cox analysis showed NAA/mIns to be a significant independent predictor of survival at two years after scanning, but not at five years. Conclusion: Cerebral degeneration in the mesial prefrontal cortex as detected by the NAA/mIns metabolite ratio is predictive of survival in ALS in a time-dependent manner.
Collapse
Affiliation(s)
- Chris Hanstock
- Department of Biomedical Engineering, University of Alberta, Edmonton, AB, Canada
| | - Kerry Sun
- Department of Medicine, Division of Neurology, University of Alberta, Edmonton, AB, Canada
| | - Changho Choi
- South-Western Medical Center, University of Texas, Dallas, TX, USA
| | - Dean Eurich
- School of Public Health, University of Alberta, Edmonton, AB, Canada, and
| | - Richard Camicioli
- Department of Medicine, Division of Neurology, University of Alberta, Edmonton, AB, Canada
| | - Wendy Johnston
- Department of Medicine, Division of Neurology, University of Alberta, Edmonton, AB, Canada
| | - Sanjay Kalra
- Department of Biomedical Engineering, University of Alberta, Edmonton, AB, Canada.,Department of Medicine, Division of Neurology, University of Alberta, Edmonton, AB, Canada.,Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
35
|
Conway ME. Alzheimer's disease: targeting the glutamatergic system. Biogerontology 2020; 21:257-274. [PMID: 32048098 PMCID: PMC7196085 DOI: 10.1007/s10522-020-09860-4] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 01/29/2020] [Indexed: 12/21/2022]
Abstract
Alzheimer’s disease (AD) is a debilitating neurodegenerative disease that causes a progressive decline in memory, language and problem solving. For decades mechanism-based therapies have primarily focused on amyloid β (Aβ) processing and pathways that govern neurofibrillary tangle generation. With the potential exception to Aducanumab, a monotherapy to target Aβ, clinical trials in these areas have been challenging and have failed to demonstrate efficacy. Currently, the prescribed therapies for AD are those that target the cholinesterase and glutamatergic systems that can moderately reduce cognitive decline, dependent on the individual. In the brain, over 40% of neuronal synapses are glutamatergic, where the glutamate level is tightly regulated through metabolite exchange in neuronal, astrocytic and endothelial cells. In AD brain, Aβ can interrupt effective glutamate uptake by astrocytes, which evokes a cascade of events that leads to neuronal swelling, destruction of membrane integrity and ultimately cell death. Much work has focussed on the post-synaptic response with little insight into how glutamate is regulated more broadly in the brain and the influence of anaplerotic pathways that finely tune these mechanisms. The role of blood branched chain amino acids (BCAA) in regulating neurotransmitter profiles under disease conditions also warrant discussion. Here, we review the importance of the branched chain aminotransferase proteins in regulating brain glutamate and the potential consequence of dysregulated metabolism in the context of BCAA or glutamate accumulation. We explore how the reported benefits of BCAA supplementation or restriction in improving cognitive function in other neurological diseases may have potential application in AD. Given that memantine, the glutamate receptor agonist, shows clinical relevance it is now timely to research related pathways, an understanding of which could identify novel approaches to treatment of AD.
Collapse
Affiliation(s)
- Myra E Conway
- Faculty of Health and Applied Sciences, University of the West of England, Coldharbour Lane, Bristol, BS16 1QY, UK. .,Faculty of Health and Life Sciences, University of the West of England, Coldharbour Lane, Bristol, BS16 1QY, UK.
| |
Collapse
|
36
|
Kassubek J, Müller HP. Advanced neuroimaging approaches in amyotrophic lateral sclerosis: refining the clinical diagnosis. Expert Rev Neurother 2020; 20:237-249. [PMID: 31937156 DOI: 10.1080/14737175.2020.1715798] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Introduction: In the last decade, multiparametric magnetic resonance imaging (MRI) has achieved tremendous advances in applications to amyotrophic lateral sclerosis (ALS) to increase the understanding of the associated pathophysiology. The aim of this review is to summarize recent progress in the development of MRI-based techniques aiming to support the clinical diagnosis in ALS.Areas covered: The review of structural and functional MRI applications to ALS and its variants (restricted phenotypes) is focused on the potential of MRI techniques which contribute to the diagnostic work-up of patients with the clinical presentation of a motor neuron disease. The potential of specific MRI methods for patient diagnosis and monitoring is discussed, and the future design of clinical MRI applications to ALS is conceptualized.Expert opinion: Current multiparametric MRI allows for the use as a clinical biological marker and a technical instrument in the clinical diagnosis of patients with ALS and also of patients with ALS variants. Composite neuroimaging indices of specific anatomical areas derived from different MRI techniques might guide in the diagnostic applications to ALS. Such a development of ALS-specific MRI-based composite scores with sufficient discriminative power versus ALS mimics at an individual level requires standardized advanced protocols and comprehensive analysis approaches.
Collapse
Affiliation(s)
- Jan Kassubek
- Department of Neurology, University of Ulm, Ulm, Germany
| | | |
Collapse
|
37
|
Bede P, Chipika RH, Finegan E, Li Hi Shing S, Doherty MA, Hengeveld JC, Vajda A, Hutchinson S, Donaghy C, McLaughlin RL, Hardiman O. Brainstem pathology in amyotrophic lateral sclerosis and primary lateral sclerosis: A longitudinal neuroimaging study. NEUROIMAGE-CLINICAL 2019; 24:102054. [PMID: 31711033 PMCID: PMC6849418 DOI: 10.1016/j.nicl.2019.102054] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 10/10/2019] [Accepted: 10/21/2019] [Indexed: 01/06/2023]
Abstract
Computational neuroimaging captures focal brainstem pathology in motor neuron diseases in contrast to both healthy- and disease controls. ALS patients exhibit progressive medulla oblongata, pontine and mesencephalic volume loss over time. Brainstem atrophy in ALS and PLS is dominated by medulla oblongata volume reductions. Vertex analyses of ALS patients reveal flattening of the medullary pyramids bilaterally. Morphometric analyses in ALS detect density reductions in the mesencephalic crura consistent with corticospinal tract degeneration.
Background Brainstem pathology is a hallmark feature of ALS, yet most imaging studies focus on cortical grey matter alterations and internal capsule white matter pathology. Brainstem imaging in ALS provides a unique opportunity to appraise descending motor tract degeneration and bulbar lower motor neuron involvement. Methods A prospective longitudinal imaging study has been undertaken with 100 patients with ALS, 33 patients with PLS, 30 patients with FTD and 100 healthy controls. Volumetric, vertex and morphometric analyses were conducted correcting for demographic factors to characterise disease-specific patterns of brainstem pathology. Using a Bayesian segmentation algorithm, the brainstem was segmented into the medulla, pons and mesencephalon to measure regional volume reductions, shape analyses were performed to ascertain the atrophy profile of each study group and region-of-interest morphometry was used to evaluate focal density alterations. Results ALS and PLS patients exhibit considerable brainstem atrophy compared to both disease- and healthy controls. Volume reductions in ALS and PLS are dominated by medulla oblongata pathology, but pontine atrophy can also be detected. In ALS, vertex analyses confirm the flattening of the medullary pyramids bilaterally in comparison to healthy controls and widespread pontine shape deformations in contrast to PLS. The ALS cohort exhibit bilateral density reductions in the mesencephalic crura in contrast to healthy controls, central pontine atrophy compared to disease controls, peri-aqueduct mesencephalic and posterior pontine changes in comparison to PLS patients. Conclus ions: Computational brainstem imaging captures the degeneration of both white and grey matter components in ALS. Our longitudinal data indicate progressive brainstem atrophy over time, underlining the biomarker potential of quantitative brainstem measures in ALS. At a time when a multitude of clinical trials are underway worldwide, there is an unprecedented need for accurate biomarkers to monitor disease progression and detect response to therapy. Brainstem imaging is a promising addition to candidate biomarkers of ALS and PLS.
Collapse
Affiliation(s)
- Peter Bede
- Computational Neuroimaging Group, Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland.
| | - Rangariroyashe H Chipika
- Computational Neuroimaging Group, Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland
| | - Eoin Finegan
- Computational Neuroimaging Group, Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland
| | - Stacey Li Hi Shing
- Computational Neuroimaging Group, Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland
| | - Mark A Doherty
- Complex Trait Genomics Laboratory, Smurfit Institute of Genetics, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland
| | - Jennifer C Hengeveld
- Complex Trait Genomics Laboratory, Smurfit Institute of Genetics, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland
| | - Alice Vajda
- Complex Trait Genomics Laboratory, Smurfit Institute of Genetics, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland
| | - Siobhan Hutchinson
- Department of Neurology, St James's Hospital, James's St, Ushers, Dublin 8 D08 NHY1, Ireland
| | - Colette Donaghy
- Department of Neurology, Western Health & Social Care Trust, Belfast, UK
| | - Russell L McLaughlin
- Complex Trait Genomics Laboratory, Smurfit Institute of Genetics, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland
| | - Orla Hardiman
- Computational Neuroimaging Group, Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland
| |
Collapse
|
38
|
Fortanier E, Grapperon AM, Le Troter A, Verschueren A, Ridley B, Guye M, Attarian S, Ranjeva JP, Zaaraoui W. Structural Connectivity Alterations in Amyotrophic Lateral Sclerosis: A Graph Theory Based Imaging Study. Front Neurosci 2019; 13:1044. [PMID: 31632235 PMCID: PMC6783612 DOI: 10.3389/fnins.2019.01044] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 09/17/2019] [Indexed: 12/15/2022] Open
Abstract
Background Amyotrophic lateral sclerosis (ALS) is a relentlessly progressive neurodegenerative disorder. Diffusion magnetic resonance imagining (MRI) studies have consistently showed widespread alterations in both motor and non-motor brain regions. However, connectomics and graph theory based approaches have shown inconsistent results. Hub-centered lesion patterns and their impact on local and large-scale brain networks remain to be established. The objective of this work is to characterize topological properties of structural brain connectivity in ALS using an array of local, global and hub-based network metrics. Materials and Methods Magnetic resonance imagining data were acquired from 25 patients with ALS and 26 age-matched healthy controls. Structural network graphs were constructed from diffusion tensor MRI. Network-based statistics (NBS) and graph theory metrics were used to compare structural networks without a priori regions of interest. Results Patients with ALS exhibited global network alterations with decreased global efficiency (Eglob) (p = 0.03) and a trend of reduced whole brain mean degree (p = 0.05) compared to controls. Six nodes showed significantly decreased mean degree in ALS: left postcentral gyrus, left interparietal and transverse parietal sulcus, left calcarine sulcus, left occipital temporal medial and lingual sulcus, right precentral gyrus and right frontal inferior sulcus (p < 0.01). Hub distribution was comparable between the two groups. There was no selective hub vulnerability or topological reorganization centered on these regions as the hub disruption index (κ) was not significant for the relevant metrics (degree, local efficiency and betweenness centrality). Using NBS, we identified an impaired motor subnetwork of 11 nodes and 10 edges centered on the precentral and the paracentral nodes (p < 0.01). Significant clinical correlations were identified between degree in the frontal area and the disease progression rate of ALS patients (p < 0.01). Conclusion Our study provides evidence that alterations of structural connectivity in ALS are primarily driven by node degree and white matter tract degeneration within an extended network around the precentral and the paracentral areas without hub-centered reorganization.
Collapse
Affiliation(s)
- Etienne Fortanier
- Aix Marseille Univ, CNRS, CRMBM, Marseille, France.,APHM, Hôpital de la Timone, Referral Centre for Neuromuscular Diseases and ALS, Marseille, France.,APHM, Hôpital de la Timone, CEMEREM, Marseille, France
| | - Aude-Marie Grapperon
- Aix Marseille Univ, CNRS, CRMBM, Marseille, France.,APHM, Hôpital de la Timone, Referral Centre for Neuromuscular Diseases and ALS, Marseille, France.,APHM, Hôpital de la Timone, CEMEREM, Marseille, France
| | - Arnaud Le Troter
- Aix Marseille Univ, CNRS, CRMBM, Marseille, France.,APHM, Hôpital de la Timone, CEMEREM, Marseille, France
| | - Annie Verschueren
- Aix Marseille Univ, CNRS, CRMBM, Marseille, France.,APHM, Hôpital de la Timone, Referral Centre for Neuromuscular Diseases and ALS, Marseille, France.,APHM, Hôpital de la Timone, CEMEREM, Marseille, France
| | - Ben Ridley
- Aix Marseille Univ, CNRS, CRMBM, Marseille, France.,APHM, Hôpital de la Timone, CEMEREM, Marseille, France
| | - Maxime Guye
- Aix Marseille Univ, CNRS, CRMBM, Marseille, France.,APHM, Hôpital de la Timone, CEMEREM, Marseille, France
| | - Shahram Attarian
- APHM, Hôpital de la Timone, Referral Centre for Neuromuscular Diseases and ALS, Marseille, France.,Aix Marseille Univ, INSERM, GMGF, Marseille, France
| | - Jean-Philippe Ranjeva
- Aix Marseille Univ, CNRS, CRMBM, Marseille, France.,APHM, Hôpital de la Timone, CEMEREM, Marseille, France
| | - Wafaa Zaaraoui
- Aix Marseille Univ, CNRS, CRMBM, Marseille, France.,APHM, Hôpital de la Timone, CEMEREM, Marseille, France
| |
Collapse
|