1
|
Atkinson RAK, Collins JM, Sreedharan J, King AE, Fernandez-Martos CM. Alterations to metabolic hormones in amyotrophic lateral sclerosis and frontotemporal dementia postmortem human tissue. J Neuropathol Exp Neurol 2024; 83:907-916. [PMID: 38917432 PMCID: PMC11487092 DOI: 10.1093/jnen/nlae054] [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] [Indexed: 06/27/2024] Open
Abstract
Metabolic changes are observed in patients with both amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Although regulation of metabolic processes in the CNS is predominantly carried out within the hypothalamus, extra-hypothalamic CNS areas contain metabolic hormone receptors, including those for leptin (LEPR), insulin (INSR), and neuropeptide Y (NPY), indicating that they may play a role in biological processes underlying pathogenic disease processes. The status of these hormones within regions vulnerable in ALS/FTD is not well described. This study sought to determine whether the expression of these hormones and their receptors is altered in pathology-rich regions in cases of human FTD (superior frontal gyrus and insular cortex) and ALS (primary motor cortex and lumbar spinal cord) with TDP-43 pathology compared to matched healthy controls. LEPR mRNA was increased within the superior frontal gyrus of FTD cases and within primary motor cortex and lumbar spinal cord of ALS cases; INSR mRNA was increased in superior frontal gyrus and insular cortex of FTD cases. NPY protein was decreased in primary motor cortex and lumbar spinal cord of ALS cases. Our results demonstrate that metabolic hormones undergo complex alterations in ALS and FTD and suggest that these hormones could play critical roles in the pathogenesis of these diseases.
Collapse
Affiliation(s)
- Rachel A K Atkinson
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Jessica M Collins
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Jemeen Sreedharan
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, United Kingdom
| | - Anna E King
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Carmen M Fernandez-Martos
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
- Department of Health and Pharmaceutical Sciences, School of Pharmacy, Universidad CEU-San Pablo, CEU Universities, Madrid, Spain
| |
Collapse
|
2
|
Tu S, Vucic S, Kiernan MC. Pathological insights derived from neuroimaging in amyotrophic lateral sclerosis: emerging clinical applications. Curr Opin Neurol 2024; 37:577-584. [PMID: 38958573 DOI: 10.1097/wco.0000000000001295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
PURPOSE OF REVIEW Neuroimaging has been instrumental in shaping current understanding of the pathoanatomical signature of amyotrophic lateral sclerosis (ALS) across clinically well defined patient cohorts. The potential utility of imaging as an objective disease marker, however, remains poorly defined. RECENT FINDINGS Increasingly advanced quantitative and computational imaging studies have highlighted emerging clinical applications for neuroimaging as a complementary clinical modality for diagnosis, monitoring, and modelling disease propagation. Multimodal neuroimaging has demonstrated novel approaches for capturing primary motor disease. Extra-motor subcortical dysfunction is increasingly recognized as key modulators of disease propagation. SUMMARY The neural signature of cortical and subcortical dysfunction in ALS has been well defined at the population level. Objective metrics of focal primary motor dysfunction are increasingly sensitive and translatable to the individual patient level. Integrity of extra-motor subcortical abnormalities are recognized to represent critical pathways of the ALS disease 'connectome', predicting pathological spread. Neuroimaging plays a pivotal role in capturing upper motor neuron pathology in ALS. Their potential clinical role as objective disease markers for disease classification, longitudinal monitoring, and prognosis in ALS have become increasingly well defined.
Collapse
Affiliation(s)
- Sicong Tu
- Brain and Mind Centre
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney
- Neuroscience Research Australia
| | - Steve Vucic
- Brain and Nerve Research Center, The University of Sydney, Sydney, New South Wales, Australia
| | - Matthew C Kiernan
- Brain and Mind Centre
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney
- Neuroscience Research Australia
| |
Collapse
|
3
|
Clarke AJ, Brodtmann A, Irish M, Mowszowski L, Radford K, Naismith SL, Mok VC, Kiernan MC, Halliday GM, Ahmed RM. Risk factors for the neurodegenerative dementias in the Western Pacific region. THE LANCET REGIONAL HEALTH. WESTERN PACIFIC 2024; 50:101051. [PMID: 39399869 PMCID: PMC11471060 DOI: 10.1016/j.lanwpc.2024.101051] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/04/2024] [Accepted: 03/12/2024] [Indexed: 10/15/2024]
Abstract
The Western Pacific Region (WPR) is characterized by a group of socioeconomically, culturally, and geopolitically heterogenous countries and represents a microcosm of the global endemic of neurodegeneration. This review will chart the known risk factors for dementia across the WPR. We explore the intersection between the established risk factors for dementia including the biomedical and lifestyle (cardiovascular and metabolic disease, sleep, hearing loss, depression, alcohol, smoking, traumatic brain injury, genetics) and social determinants (social disadvantage, limited education, systemic racism) as well as incorporate neuroimaging data, where available, to predict disease progression in the WPR. In doing so, we highlight core risk factors for dementia in the WPR, as well as geographical epicentres at heightened risk for dementia, to orient future research towards addressing these disparities.
Collapse
Affiliation(s)
- Antonia J. Clarke
- Department of Neurosciences, Monash University, Melbourne, VIC 3004 Australia
| | - Amy Brodtmann
- Department of Neurosciences, Monash University, Melbourne, VIC 3004 Australia
| | - Muireann Irish
- Brain and Mind Centre, The University of Sydney, Sydney, NSW 2050 Australia
| | - Loren Mowszowski
- Brain and Mind Centre, The University of Sydney, Sydney, NSW 2050 Australia
- Department of Neurology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Kylie Radford
- Department of Neurology, Royal Prince Alfred Hospital, Sydney, Australia
- The University of New South Wales, Sydney, NSW 2031 Australia
| | - Sharon L. Naismith
- Brain and Mind Centre, The University of Sydney, Sydney, NSW 2050 Australia
| | | | - Matthew C. Kiernan
- Brain and Mind Centre, The University of Sydney, Sydney, NSW 2050 Australia
- Department of Neurology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Glenda M. Halliday
- Brain and Mind Centre, The University of Sydney, Sydney, NSW 2050 Australia
- Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Sydney, NSW 2050 Australia
| | - Rebekah M. Ahmed
- Brain and Mind Centre, The University of Sydney, Sydney, NSW 2050 Australia
- Department of Neurology, Royal Prince Alfred Hospital, Sydney, Australia
| |
Collapse
|
4
|
Shaw SR, Horne KS, Piguet O, Ahmed RM, Whitton AE, Irish M. Profiles of motivational impairment and their relationship to functional decline in frontotemporal dementia. J Neurol 2024; 271:4963-4971. [PMID: 38758282 PMCID: PMC11319612 DOI: 10.1007/s00415-024-12430-0] [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: 01/22/2024] [Revised: 04/05/2024] [Accepted: 05/08/2024] [Indexed: 05/18/2024]
Abstract
Motivational disturbances are pervasive in frontotemporal dementia (FTD) and impact negatively on everyday functioning. Despite mounting evidence of anhedonia in FTD, it remains unclear how such changes fit within the broader motivational symptom profile of FTD, or how anhedonia relates to functional outcomes. Here we sought to comprehensively characterize motivational disturbances in FTD and their respective relationships with functional impairment. A cross-sectional study design was used including 211 participants-68 behavioral-variant FTD (bvFTD), 32 semantic dementia (SD), 43 Alzheimer's disease (AD), and 68 healthy older control participants. Anhedonia severity was measured using the Snaith-Hamilton Pleasure Scale while severity of apathy was assessed across Emotional, Executive, and Initiation dimensions using the Dimensional Apathy Scale. Functional impairment was established using the FTD Functional Rating Scale (FRS). Distinct motivational profiles emerged in each dementia syndrome: a domain-general motivational impairment in bvFTD; a predominantly anhedonic profile in SD; and more pronounced initiation and executive apathy in AD. Correlation analyses revealed differential associations between motivational symptoms and severity of functional impairment in each group. Executive apathy was associated with functional impairment in bvFTD, while anhedonia was strongly correlated with functional decline in SD. Finally, executive and emotional apathy were associated with functional decline in AD. Our study indicates distinct profiles of apathy and anhedonia in FTD syndromes, which in turn are differentially associated with functional decline. This detailed characterization of motivational phenotypes can inform patient stratification for targeted interventions to improve functional outcomes.
Collapse
Affiliation(s)
- Siobhán R Shaw
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
- School of Psychology, The University of Sydney, Sydney, NSW, Australia
| | - Kristina S Horne
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
- School of Psychology, The University of Sydney, Sydney, NSW, Australia
| | - Olivier Piguet
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
- School of Psychology, The University of Sydney, Sydney, NSW, Australia
| | - Rebekah M Ahmed
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
- Memory and Cognition Clinic, Department of Clinical Neurosciences, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Alexis E Whitton
- Black Dog Institute, University of New South Wales, Sydney, NSW, Australia
| | - Muireann Irish
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia.
- School of Psychology, The University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
5
|
Ding M, Xu W, Pei G, Li P. Long way up: rethink diseases in light of phase separation and phase transition. Protein Cell 2024; 15:475-492. [PMID: 38069453 PMCID: PMC11214837 DOI: 10.1093/procel/pwad057] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 11/24/2023] [Indexed: 07/02/2024] Open
Abstract
Biomolecular condensation, driven by multivalency, serves as a fundamental mechanism within cells, facilitating the formation of distinct compartments, including membraneless organelles that play essential roles in various cellular processes. Perturbations in the delicate equilibrium of condensation, whether resulting in gain or loss of phase separation, have robustly been associated with cellular dysfunction and physiological disorders. As ongoing research endeavors wholeheartedly embrace this newly acknowledged principle, a transformative shift is occurring in our comprehension of disease. Consequently, significant strides have been made in unraveling the profound relevance and potential causal connections between abnormal phase separation and various diseases. This comprehensive review presents compelling recent evidence that highlight the intricate associations between aberrant phase separation and neurodegenerative diseases, cancers, and infectious diseases. Additionally, we provide a succinct summary of current efforts and propose innovative solutions for the development of potential therapeutics to combat the pathological consequences attributed to aberrant phase separation.
Collapse
Affiliation(s)
- Mingrui Ding
- State Key Laboratory of Membrane Biology & Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
- NuPhase Therapeutics, Beijing 100083, China
| | - Weifan Xu
- State Key Laboratory of Membrane Biology & Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
- NuPhase Therapeutics, Beijing 100083, China
| | - Gaofeng Pei
- State Key Laboratory of Membrane Biology & Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Pilong Li
- State Key Laboratory of Membrane Biology & Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| |
Collapse
|
6
|
Hu Y, Hruscha A, Pan C, Schifferer M, Schmidt MK, Nuscher B, Giera M, Kostidis S, Burhan Ö, van Bebber F, Edbauer D, Arzberger T, Haass C, Schmid B. Mis-localization of endogenous TDP-43 leads to ALS-like early-stage metabolic dysfunction and progressive motor deficits. Mol Neurodegener 2024; 19:50. [PMID: 38902734 PMCID: PMC11188230 DOI: 10.1186/s13024-024-00735-7] [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: 11/22/2023] [Accepted: 05/23/2024] [Indexed: 06/22/2024] Open
Abstract
BACKGROUND The key pathological signature of ALS/ FTLD is the mis-localization of endogenous TDP-43 from the nucleus to the cytoplasm. However, TDP-43 gain of function in the cytoplasm is still poorly understood since TDP-43 animal models recapitulating mis-localization of endogenous TDP-43 from the nucleus to the cytoplasm are missing. METHODS CRISPR/Cas9 technology was used to generate a zebrafish line (called CytoTDP), that mis-locates endogenous TDP-43 from the nucleus to the cytoplasm. Phenotypic characterization of motor neurons and the neuromuscular junction was performed by immunostaining, microglia were immunohistochemically localized by whole-mount tissue clearing and muscle ultrastructure was analyzed by scanning electron microscopy. Behavior was investigated by video tracking and quantitative analysis of swimming parameters. RNA sequencing was used to identify mis-regulated pathways with validation by molecular analysis. RESULTS CytoTDP fish have early larval phenotypes resembling clinical features of ALS such as progressive motor defects, neurodegeneration and muscle atrophy. Taking advantage of zebrafish's embryonic development that solely relys on yolk usage until 5 days post fertilization, we demonstrated that microglia proliferation and activation in the hypothalamus is independent from food intake. By comparing CytoTDP to a previously generated TDP-43 knockout line, transcriptomic analyses revealed that mis-localization of endogenous TDP-43, rather than TDP-43 nuclear loss of function, leads to early onset metabolic dysfunction. CONCLUSIONS The new TDP-43 model mimics the ALS/FTLD hallmark of progressive motor dysfunction. Our results suggest that functional deficits of the hypothalamus, the metabolic regulatory center, might be the primary cause of weight loss in ALS patients. Cytoplasmic gain of function of endogenous TDP-43 leads to metabolic dysfunction in vivo that are reminiscent of early ALS clinical non-motor metabolic alterations. Thus, the CytoTDP zebrafish model offers a unique opportunity to identify mis-regulated targets for therapeutic intervention early in disease progression.
Collapse
Affiliation(s)
- Yiying Hu
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Metabolic Biochemistry, Biomedical Centre (BMC), Faculty of Medicine, Ludwig-Maximilian University, Munich, Germany
- Munich Medical Research School (MMRS), Munich, Germany
| | - Alexander Hruscha
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| | - Chenchen Pan
- Neurology Clinic and National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Martina Schifferer
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Michael K Schmidt
- Zentrum Für Neuropathologie, Ludwig-Maximilians University, Munich, Germany
| | - Brigitte Nuscher
- Metabolic Biochemistry, Biomedical Centre (BMC), Faculty of Medicine, Ludwig-Maximilian University, Munich, Germany
| | - Martin Giera
- Leiden University Medical Center, Leiden, Netherlands
| | | | - Özge Burhan
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| | - Frauke van Bebber
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| | - Dieter Edbauer
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Metabolic Biochemistry, Biomedical Centre (BMC), Faculty of Medicine, Ludwig-Maximilian University, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Thomas Arzberger
- Zentrum Für Neuropathologie, Ludwig-Maximilians University, Munich, Germany
- Department of Psychiatry and Psychotherapy, University Hospital, Ludwig-Maximilians University, Munich, Germany
| | - Christian Haass
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Metabolic Biochemistry, Biomedical Centre (BMC), Faculty of Medicine, Ludwig-Maximilian University, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Bettina Schmid
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany.
| |
Collapse
|
7
|
Amian JG, Fernandez-Portero C, de la Bella R, Arenilla-Villalba MJ, López-Lluch G, Alarcon D. Cognitive Reserve and Frontotemporal Disorders: Exploring the Relationship Between Education, Physical Activity, and Cognitive Dysfunction in Older Adults. Percept Mot Skills 2024; 131:720-736. [PMID: 38523555 DOI: 10.1177/00315125241241358] [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: 03/26/2024]
Abstract
In this study we investigated the relationship between cognitive reserve (CR) proxies, such as education, physical activity (PA), and cognitive dysfunction (CD) in the presence or absence of frontotemporal disorders (FTD). Previous research has suggested that education and PA may delay the onset of CD and reduce the risk of developing dementia. However, it remains unclear whether these CR proxies can protect against CD when FTD is present. We aimed to explore this relationship and determine whether sustained CR may be evident regardless of FTD. We recruited 149 older adults (aged 65-99 years) from community centers where they were voluntarily participating in leisure activities. We used bioelectrical impedance to measure their body composition, and we administered the International PA Questionnaire and the Mini-Mental State Examination to measure their PA and cognitive function, respectively. We used the Frontal Assessment Battery to screen for frontotemporal dementia. Our results showed that people with FTD were older, had lower education, and engaged in less PA, relative to other participants. Regression models revealed that age, education, and PA were significant predictors of FTD. More specifically, FTD was negatively associated with cognitive functioning, and there were significant interaction effects between FTD and education and PA. PA and education were significant predictors of cognitive functioning, and, when values for PA and education were high, they offset the effects of FTD on cognitive function. These findings support impressions that PA and years of education provide an insulating or compensatory effect on cognitive functioning in older adults with executive dysfunction or frontotemporal dementia, highlighting the importance of encouraging both pursuits.
Collapse
Affiliation(s)
- Josue G Amian
- Department of Social Anthropology, Psychology and Public Health, Pablo de Olavide University, Seville, Spain
| | - Cristina Fernandez-Portero
- Department of Social Anthropology, Psychology and Public Health, Pablo de Olavide University, Seville, Spain
| | - Rocío de la Bella
- Department of Physiology, Anatomy and Cell Biology, Andalusian Centre of Developmental Biology, Universidad Pablo de Olavide, Seville, Spain
| | | | - Guillermo López-Lluch
- Department of Physiology, Anatomy and Cell Biology, Andalusian Centre of Developmental Biology, Universidad Pablo de Olavide, Seville, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER, U729), IICS-Madrid, Madrid, Spain
- Centro de investigación en Rendimiento Físico y Deportivo, Universidad Pablo de Olavide, Sevilla, Spain
| | - David Alarcon
- Department of Social Anthropology, Psychology and Public Health, Pablo de Olavide University, Seville, Spain
| |
Collapse
|
8
|
Eisen A, Nedergaard M, Gray E, Kiernan MC. The glymphatic system and Amyotrophic lateral sclerosis. Prog Neurobiol 2024; 234:102571. [PMID: 38266701 DOI: 10.1016/j.pneurobio.2024.102571] [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: 08/07/2023] [Revised: 11/18/2023] [Accepted: 01/15/2024] [Indexed: 01/26/2024]
Abstract
The glymphatic system and the meningeal lymphatic vessels provide a pathway for transport of solutes and clearance of toxic material from the brain. Of specific relevance to ALS, this is applicable for TDP-43 and glutamate, both major elements in disease pathogenesis. Flow is propelled by arterial pulsation, respiration, posture, as well as the positioning and proportion of aquaporin-4 channels (AQP4). Non-REM slow wave sleep is the is key to glymphatic drainage which discontinues during wakefulness. In Parkinson's disease and Alzheimer's disease, sleep impairment is known to predate the development of characteristic clinical features by several years and is associated with progressive accumulation of toxic proteinaceous products. While sleep issues are well described in ALS, consideration of preclinical sleep impairment or the potential of a failing glymphatic system in ALS has rarely been considered. Here we review how the glymphatic system may impact ALS. Preclinical sleep impairment as an unrecognized major risk factor for ALS is considered, while potential therapeutic options to improve glymphatic flow are explored.
Collapse
Affiliation(s)
- Andrew Eisen
- Department of Neurology, University of British Columbia, Vancouver, Canada.
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, University of Rochester Medical School and Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Emma Gray
- Department of Neurology, Royal Prince Alfred Hospital and University of Sydney, NSW 2050, Australia
| | | |
Collapse
|
9
|
Alnaaim SA, Al-Kuraishy HM, Alexiou A, Papadakis M, Saad HM, Batiha GES. Role of Brain Liver X Receptor in Parkinson's Disease: Hidden Treasure and Emerging Opportunities. Mol Neurobiol 2024; 61:341-357. [PMID: 37606719 PMCID: PMC10791998 DOI: 10.1007/s12035-023-03561-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 08/01/2023] [Indexed: 08/23/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease due to the degeneration of dopaminergic neurons (DNs) in the substantia nigra (SN). The liver X receptor (LXR) is involved in different neurodegenerative diseases. Therefore, the objective of the present review was to clarify the possible role of LXR in PD neuropathology. LXRs are the most common nuclear receptors of transcription factors that regulate cholesterol metabolism and have pleiotropic effects, including anti-inflammatory effects and reducing intracellular cholesterol accumulation. LXRs are highly expressed in the adult brain and act as endogenous sensors for intracellular cholesterol. LXRs have neuroprotective effects against the development of neuroinflammation in different neurodegenerative diseases by inhibiting the expression of pro-inflammatory cytokines. LXRs play an essential role in mitigating PD neuropathology by reducing the expression of inflammatory signaling pathways, neuroinflammation, oxidative stress, mitochondrial dysfunction, and enhancement of BDNF signaling.In conclusion, LXRs, through regulating brain cholesterol homeostasis, may be effectual in PD. Also, inhibition of node-like receptor pyrin 3 (NLRP3) inflammasome and nuclear factor kappa B (NF-κB) by LXRs could effectively prevent neuroinflammation in PD. Taken together, LXRs play a crucial role in PD neuropathology by inhibiting neuroinflammation and associated degeneration of DNs.
Collapse
Affiliation(s)
- Saud A Alnaaim
- Clinical Neurosciences Department, College of Medicine, King Faisal University, Hofuf, Saudi Arabia
| | - Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Therapeutic Medicine, College of Medicine, ALmustansiriyiah University, Baghdad, 14132, Iraq
| | - Athanasios Alexiou
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW, 2770, Australia
- AFNP Med, 1030, Wien, Austria
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, Heusnerstrasse 40, University of Witten-Herdecke, 42283, Wuppertal, Germany.
| | - Hebatallah M Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Marsa Matruh, 51744, Egypt
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, AlBeheira, 22511, Egypt
| |
Collapse
|
10
|
Aquilani R, Cotta Ramusino M, Maestri R, Iadarola P, Boselli M, Perini G, Boschi F, Dossena M, Bellini A, Buonocore D, Doria E, Costa A, Verri M. Several dementia subtypes and mild cognitive impairment share brain reduction of neurotransmitter precursor amino acids, impaired energy metabolism, and lipid hyperoxidation. Front Aging Neurosci 2023; 15:1237469. [PMID: 37655338 PMCID: PMC10466813 DOI: 10.3389/fnagi.2023.1237469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 08/03/2023] [Indexed: 09/02/2023] Open
Abstract
Objective Dementias and mild cognitive impairment (MCI) are associated with variously combined changes in the neurotransmitter system and signaling, from neurotransmitter synthesis to synaptic binding. The study tested the hypothesis that different dementia subtypes and MCI may share similar reductions of brain availability in amino acid precursors (AAPs) of neurotransmitter synthesis and concomitant similar impairment in energy production and increase of oxidative stress, i.e., two important metabolic alterations that impact neurotransmission. Materials and methods Sixty-five demented patients (Alzheimer's disease, AD, n = 44; frontotemporal disease, FTD, n = 13; vascular disease, VaD, n = 8), 10 subjects with MCI and 15 control subjects (CTRL) were recruited for this study. Cerebrospinal fluid (CSF) and plasma levels of AAPs, energy substrates (lactate, pyruvate), and an oxidative stress marker (malondialdehyde, MDA) were measured in all participants. Results Demented patients and subjects with MCI were similar for age, anthropometric parameters, biohumoral variables, insulin resistance (HOMA index model), and CSF neuropathology markers. Compared to age-matched CTRL, both demented patients and MCI subjects showed low CSF AAP tyrosine (precursor of dopamine and catecholamines), tryptophan (precursor of serotonin), methionine (precursor of acetylcholine) limited to AD and FTD, and phenylalanine (an essential amino acid largely used for protein synthesis) (p = 0.03 to <0.0001). No significant differences were found among dementia subtypes or between each dementia subtype and MCI subjects. In addition, demented patients and MCI subjects, compared to CTRL, had similar increases in CSF and plasma levels of pyruvate (CSF: p = 0.023 to <0.0001; plasma: p < 0.002 to <0.0001) and MDA (CSF: p < 0.035 to 0.002; plasma: p < 0.0001). Only in AD patients was the CSF level of lactate higher than in CTRL (p = 0.003). Lactate/pyruvate ratios were lower in all experimental groups than in CTRL. Conclusion AD, FTD, and VaD dementia patients and MCI subjects may share similar deficits in AAPs, partly in energy substrates, and similar increases in oxidative stress. These metabolic alterations may be due to AAP overconsumption following high brain protein turnover (leading to phenylalanine reductions), altered mitochondrial structure and function, and an excess of free radical production. All these metabolic alterations may have a negative impact on synaptic plasticity and activity.
Collapse
Affiliation(s)
- Roberto Aquilani
- Department of Biology and Biotechnology, “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| | - Matteo Cotta Ramusino
- Unit of Behavioral Neurology and Center for Cognitive Disorders and Dementia, IRCCS C. Mondino Foundation, Pavia, Italy
- Dementia Research Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Roberto Maestri
- Department of Biomedical Engineering of the Montescano Institute, Istituti Clinici Scientifici Maugeri IRCCS, Montescano, Italy
| | - Paolo Iadarola
- Department of Biology and Biotechnology, “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| | - Mirella Boselli
- Neurorehabilitation Unit of the Montescano Institute, Istituti Clinici Scientifici Maugeri IRCCS, Montescano, Italy
| | - Giulia Perini
- Unit of Behavioral Neurology and Center for Cognitive Disorders and Dementia, IRCCS C. Mondino Foundation, Pavia, Italy
- Dementia Research Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Federica Boschi
- Department of Drug Sciences, University of Pavia, Pavia, Italy
| | - Maurizia Dossena
- Department of Biology and Biotechnology, “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| | - Anna Bellini
- Department of Biology and Biotechnology, “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| | - Daniela Buonocore
- Department of Biology and Biotechnology, “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| | - Enrico Doria
- Department of Biology and Biotechnology, “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| | - Alfredo Costa
- Unit of Behavioral Neurology and Center for Cognitive Disorders and Dementia, IRCCS C. Mondino Foundation, Pavia, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Manuela Verri
- Department of Biology and Biotechnology, “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| |
Collapse
|
11
|
Liu X, Cui C, Sun W, Meng J, Guo J, Wu L, Chen B, Liao D, Jiang P. Paclitaxel Induces Neurotoxicity by Disrupting Tricarboxylic Acid Cycle Metabolic Balance in the Mouse Hippocampus. J Toxicol 2023; 2023:5660481. [PMID: 37575636 PMCID: PMC10423086 DOI: 10.1155/2023/5660481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 06/25/2023] [Accepted: 07/07/2023] [Indexed: 08/15/2023] Open
Abstract
Objective It is well known that paclitaxel (PTX)-induced neurotoxicity seriously affects the quality of life of patients and is the main reason for reducing the dose of chemotherapy or even stopping chemotherapy. The current data are limited, and further information is required for practice and verification. The aims of this study were to clarify the molecular mechanism underlying PTX-induced neurotoxicity by combining in vivo and in vitro metabolomics studies and provide new targets for the prevention and treatment of PTX-induced neurotoxicity. Methods In the in vivo study, a PTX-induced neurotoxicity mouse model was established by intraperitoneal injection of PTX (6 mg/kg every three days) for two consecutive weeks. After verification by water maze tests and HE staining of pathological sections, hippocampal metabolites were measured and the differential metabolites and related metabolic pathways were identified by multivariate statistical analysis. In the in vitro study, we investigated the effects of PTX on mouse hippocampal neuron cells, assessing the concentration and time of administration by MTT assays. After modeling, the relevant metabolites in the TCA cycle were quantified by targeted metabolomics using stable isotope labeling. Finally, the key enzymes of the TCA cycle in tissues and cells were verified by RT-PCR. Results Administration of PTX to model mice resulted in neurological damage, shown by both water-maze tests and hippocampal tissue sections. Twenty-four metabolites and five associated metabolic pathways were found to differ significantly between the hippocampal tissues of the model and control groups. These included metabolites and pathways related to the TCA cycle and pyruvate metabolism. Metabolomics analysis using stable isotope labeling showed significant changes in metabolites associated with the TCA cycle compared with the control group (P < 0.05). Finally, RT-PCR verified that the expression of key enzymes in the TCA cycle was changed to different degrees in both hippocampal tissues and cells. Conclusion Our results showed that PTX neurotoxicity in hippocampal tissue and neuron cells was associated with inhibition of the TCA cycle. This inhibition leads to brain insufficiency and impaired metabolism, resulting in various neurotoxic symptoms.
Collapse
Affiliation(s)
- Xi Liu
- Department of Pharmacy, Linfen People's Hospital, Linfen, China
| | - Changmeng Cui
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University, Jining, China
| | - Wenxue Sun
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Jining Medical University, Jining, China
| | - Junjun Meng
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Jining Medical University, Jining, China
| | - Jinxiu Guo
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Jining Medical University, Jining, China
| | - Linlin Wu
- Department of Oncology, Tengzhou Central People's Hospital, Affiliated to Jining Medical College, Tengzhou, China
| | - Beibei Chen
- ADFA School of Science, University of New South Wales, Canberra, Australia
| | - Dehua Liao
- Department of Pharmacy, Hunan Cancer Hospital, Changsha, China
| | - Pei Jiang
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, China
| |
Collapse
|
12
|
Morrow CB, Leoutsakos J, Yan H, Onyike C, Kamath V. Weight Change and Neuropsychiatric Symptoms in Alzheimer's Disease and Frontotemporal Dementia: Associations with Cognitive Decline. J Alzheimers Dis Rep 2023; 7:767-774. [PMID: 37662607 PMCID: PMC10473120 DOI: 10.3233/adr-230034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 06/29/2023] [Indexed: 09/05/2023] Open
Abstract
Weight changes, neuropsychiatric symptoms (NPS), and cognitive decline often coincide in Alzheimer's disease (AD) and frontotemporal dementia (FTD); however, the direction of their relationship remains unclear. This study aims to clarify the connection between weight changes, NPS, and cognition in AD and FTD. We found that cognitive decline was associated with decreased body mass index (BMI) in AD, while BMI gain was associated with increased conversion to FTD. Elevated NPS were associated with decreased BMI in AD and increased BMI in FTD. Identifying early changes in NPS and BMI may facilitate the detection of cognitive decline, providing an opportunity for early intervention.
Collapse
Affiliation(s)
- Christopher B. Morrow
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jeannie Leoutsakos
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Haijuan Yan
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Chiadi Onyike
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Vidyulata Kamath
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
13
|
Pamphlett R, Bishop DP. The toxic metal hypothesis for neurological disorders. Front Neurol 2023; 14:1173779. [PMID: 37426441 PMCID: PMC10328356 DOI: 10.3389/fneur.2023.1173779] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 05/30/2023] [Indexed: 07/11/2023] Open
Abstract
Multiple sclerosis and the major sporadic neurogenerative disorders, amyotrophic lateral sclerosis, Parkinson disease, and Alzheimer disease are considered to have both genetic and environmental components. Advances have been made in finding genetic predispositions to these disorders, but it has been difficult to pin down environmental agents that trigger them. Environmental toxic metals have been implicated in neurological disorders, since human exposure to toxic metals is common from anthropogenic and natural sources, and toxic metals have damaging properties that are suspected to underlie many of these disorders. Questions remain, however, as to how toxic metals enter the nervous system, if one or combinations of metals are sufficient to precipitate disease, and how toxic metal exposure results in different patterns of neuronal and white matter loss. The hypothesis presented here is that damage to selective locus ceruleus neurons from toxic metals causes dysfunction of the blood-brain barrier. This allows circulating toxicants to enter astrocytes, from where they are transferred to, and damage, oligodendrocytes, and neurons. The type of neurological disorder that arises depends on (i) which locus ceruleus neurons are damaged, (ii) genetic variants that give rise to susceptibility to toxic metal uptake, cytotoxicity, or clearance, (iii) the age, frequency, and duration of toxicant exposure, and (iv) the uptake of various mixtures of toxic metals. Evidence supporting this hypothesis is presented, concentrating on studies that have examined the distribution of toxic metals in the human nervous system. Clinicopathological features shared between neurological disorders are listed that can be linked to toxic metals. Details are provided on how the hypothesis applies to multiple sclerosis and the major neurodegenerative disorders. Further avenues to explore the toxic metal hypothesis for neurological disorders are suggested. In conclusion, environmental toxic metals may play a part in several common neurological disorders. While further evidence to support this hypothesis is needed, to protect the nervous system it would be prudent to take steps to reduce environmental toxic metal pollution from industrial, mining, and manufacturing sources, and from the burning of fossil fuels.
Collapse
Affiliation(s)
- Roger Pamphlett
- Department of Pathology, Brain and Mind Centre, School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia
- Department of Neuropathology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
- Hyphenated Mass Spectrometry Laboratory, School of Mathematical and Physical Sciences, University of Technology Sydney, Sydney, NSW, Australia
| | - David P. Bishop
- Hyphenated Mass Spectrometry Laboratory, School of Mathematical and Physical Sciences, University of Technology Sydney, Sydney, NSW, Australia
| |
Collapse
|
14
|
Wang W, Zhang L, Xia K, Huang T, Fan D. Mendelian Randomization Analysis Reveals Statins Potentially Increase Amyotrophic Lateral Sclerosis Risk Independent of Peripheral Cholesterol-Lowering Effects. Biomedicines 2023; 11:biomedicines11051359. [PMID: 37239030 DOI: 10.3390/biomedicines11051359] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 04/28/2023] [Accepted: 05/03/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND Observational studies suggest that statins may affect amyotrophic lateral sclerosis (ALS). However, they are limited by confounding and reverse causality biases. Therefore, we aimed to investigate the potential causal associations between statins and ALS using a mendelian randomization (MR) approach. METHODS Two-sample MR and drug-target MR were performed. Exposure sources included GWAS summary statistics of statin use, low-density-lipoprotein cholesterol (LDL-C), HMGCR-mediated LDL-C and LDL-C response to statins. RESULTS Genetic predisposition to statin medication was associated with increased ALS risk (OR = 1.085, 95% CI = 1.025-1.148, p = 0.005). After removing SNPs significantly associated with statin use from the instrumental variables (IVs), LDL-C-related higher ALS risk was absent (before removing: OR = 1.075, 95% CI = 1.013-1.141, p = 0.017; after removing: OR = 1.036, 95% CI = 0.949-1.131, p = 0.432). HMGCR-mediated LDL-C (OR = 1.033, 95% CI = 0.823-1.296, p = 0.779) and blood LDL-C response to statins (OR = 0.998, 95% CI = 0.991-1.005, p = 0.538) had no association with ALS. CONCLUSIONS Here, we show that statins may be a risky exposure that increases ALS risk independent of the lowering effect of LDL-C in peripheral circulation. This provides insights into ALS development and prevention.
Collapse
Affiliation(s)
- Wenjing Wang
- Department of Neurology, Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing 100191, China
- Key Laboratory for Neuroscience, National Health Commission/Ministry of Education, Peking University, Beijing 100191, China
| | - Linjing Zhang
- Department of Neurology, Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing 100191, China
- Key Laboratory for Neuroscience, National Health Commission/Ministry of Education, Peking University, Beijing 100191, China
| | - Kailin Xia
- Department of Neurology, Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing 100191, China
- Key Laboratory for Neuroscience, National Health Commission/Ministry of Education, Peking University, Beijing 100191, China
| | - Tao Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, China
- Key Laboratory of Molecular Cardiovascular Sciences, Peking University, Ministry of Education, Beijing 100191, China
| | - Dongsheng Fan
- Department of Neurology, Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing 100191, China
- Key Laboratory for Neuroscience, National Health Commission/Ministry of Education, Peking University, Beijing 100191, China
| |
Collapse
|
15
|
Yang L, Cheng Y, Zhu Y, Cui L, Li X. The Serotonergic System and Amyotrophic Lateral Sclerosis: A Review of Current Evidence. Cell Mol Neurobiol 2023:10.1007/s10571-023-01320-0. [PMID: 36729314 DOI: 10.1007/s10571-023-01320-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 01/18/2023] [Indexed: 02/03/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder characterized by the premature death of motor neurons. Serotonin (5-HT) is a crucial neurotransmitter, and its dysfunction, whether as a contributor or by-product, has been implicated in ALS pathogenesis. Here, we summarize current evidence linking serotonergic alterations to ALS, including results from post-mortem and neuroimaging studies, biofluid testing, and studies of ALS animal models. We also discuss the possible role of 5-HT in modulating some important mechanisms of ALS (i.e. glutamate excitotoxity and neuroinflammation) and in regulating ALS phenotypes (i.e. breathing dysfunction and metabolic defects). Finally, we discuss the promise and limitations of the serotonergic system as a target for the development of ALS biomarkers and therapeutic approaches. However, due to a relative paucity of data and standardized methodologies in previous studies, proper interpretation of existing results remains a challenge. Future research is needed to unravel the mechanisms linking serotonergic pathways and ALS and to provide valid, reproducible, and translatable findings.
Collapse
Affiliation(s)
- Lu Yang
- Department of Neurology, Peking Union Medical College Hospital (PUMCH), The Transformation Medical Center of PUMC, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, 100005, China
| | - Yanfei Cheng
- Department of Neurology, Peking Union Medical College Hospital (PUMCH), The Transformation Medical Center of PUMC, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, 100005, China
| | - Yicheng Zhu
- Department of Neurology, Peking Union Medical College Hospital (PUMCH), The Transformation Medical Center of PUMC, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, 100005, China.,Neuroscience Center, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Liying Cui
- Department of Neurology, Peking Union Medical College Hospital (PUMCH), The Transformation Medical Center of PUMC, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, 100005, China.,Neuroscience Center, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Xiaoguang Li
- Department of Neurology, Peking Union Medical College Hospital (PUMCH), The Transformation Medical Center of PUMC, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, 100005, China. .,Neuroscience Center, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, China.
| |
Collapse
|
16
|
Chang J, Shaw TB, Holdom CJ, McCombe PA, Henderson RD, Fripp J, Barth M, Guo CC, Ngo ST, Steyn FJ. Lower hypothalamic volume with lower body mass index is associated with shorter survival in patients with amyotrophic lateral sclerosis. Eur J Neurol 2023; 30:57-68. [PMID: 36214080 PMCID: PMC10099625 DOI: 10.1111/ene.15589] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/15/2022] [Accepted: 09/30/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND AND PURPOSE Weight loss in patients with amyotrophic lateral sclerosis (ALS) is associated with faster disease progression and shorter survival. Decreased hypothalamic volume is proposed to contribute to weight loss due to loss of appetite and/or hypermetabolism. We aimed to investigate the relationship between hypothalamic volume and body mass index (BMI) in ALS and Alzheimer's disease (AD), and the associations of hypothalamic volume with weight loss, appetite, metabolism and survival in patients with ALS. METHODS We compared hypothalamic volumes from magnetic resonance imaging scans with BMI for patients with ALS (n = 42), patients with AD (n = 167) and non-neurodegenerative disease controls (n = 527). Hypothalamic volumes from patients with ALS were correlated with measures of appetite and metabolism, and change in anthropomorphic measures and disease outcomes. RESULTS Lower hypothalamic volume was associated with lower and higher BMI in ALS (quadratic association; probability of direction = 0.96). This was not observed in AD patients or controls. Hypothalamic volume was not associated with loss of appetite (p = 0.58) or hypermetabolism (p = 0.49). Patients with lower BMI and lower hypothalamic volume tended to lose weight (p = 0.08) and fat mass (p = 0.06) over the course of their disease, and presented with an increased risk of earlier death (hazard ratio [HR] 3.16, p = 0.03). Lower hypothalamic volume alone trended for greater risk of earlier death (HR 2.61, p = 0.07). CONCLUSION These observations suggest that lower hypothalamic volume in ALS contributes to positive and negative energy balance, and is not universally associated with loss of appetite or hypermetabolism. Critically, lower hypothalamic volume with lower BMI was associated with weight loss and earlier death.
Collapse
Affiliation(s)
- Jeryn Chang
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Saint Lucia, Australia
| | - Thomas B Shaw
- Department of Neurology, Royal Brisbane and Women's Hospital, Herston, Australia.,Centre for Advanced Imaging, The University of Queensland, Saint Lucia, Australia.,School of Information Technology and Electrical Engineering, The University of Queensland, Saint Lucia, Australia
| | - Cory J Holdom
- Australian Institute of Bioengineering and Nanotechnology, The University of Queensland, Saint Lucia, Australia
| | - Pamela A McCombe
- Department of Neurology, Royal Brisbane and Women's Hospital, Herston, Australia.,UQ Centre for Clinical Research, The University of Queensland, Herston, Australia.,Wesley Medical Research, The Wesley Hospital, Auchenflower, Australia
| | - Robert D Henderson
- Department of Neurology, Royal Brisbane and Women's Hospital, Herston, Australia.,UQ Centre for Clinical Research, The University of Queensland, Herston, Australia.,Wesley Medical Research, The Wesley Hospital, Auchenflower, Australia
| | - Jurgen Fripp
- CSIRO Health and Biosecurity, Herston, Australia
| | - Markus Barth
- Centre for Advanced Imaging, The University of Queensland, Saint Lucia, Australia.,School of Information Technology and Electrical Engineering, The University of Queensland, Saint Lucia, Australia
| | | | - Shyuan T Ngo
- Department of Neurology, Royal Brisbane and Women's Hospital, Herston, Australia.,Australian Institute of Bioengineering and Nanotechnology, The University of Queensland, Saint Lucia, Australia.,Wesley Medical Research, The Wesley Hospital, Auchenflower, Australia
| | - Frederik J Steyn
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Saint Lucia, Australia.,Department of Neurology, Royal Brisbane and Women's Hospital, Herston, Australia.,Wesley Medical Research, The Wesley Hospital, Auchenflower, Australia
| | | |
Collapse
|
17
|
Fatty acids derived from the probiotic Lacticaseibacillus rhamnosus HA-114 suppress age-dependent neurodegeneration. Commun Biol 2022; 5:1340. [PMID: 36477191 PMCID: PMC9729297 DOI: 10.1038/s42003-022-04295-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 11/24/2022] [Indexed: 12/12/2022] Open
Abstract
The human microbiota is believed to influence health. Microbiome dysbiosis may be linked to neurological conditions like Alzheimer's disease, amyotrophic lateral sclerosis, and Huntington's disease. We report the ability of a probiotic bacterial strain in halting neurodegeneration phenotypes. We show that Lacticaseibacillus rhamnosus HA-114 is neuroprotective in C. elegans models of amyotrophic lateral sclerosis and Huntington's disease. Our results show that neuroprotection from L. rhamnosus HA-114 is unique from other L. rhamnosus strains and resides in its fatty acid content. Neuroprotection by L. rhamnosus HA-114 requires acdh-1/ACADSB, kat-1/ACAT1 and elo-6/ELOVL3/6, which are associated with fatty acid metabolism and mitochondrial β-oxidation. Our data suggest that disrupted lipid metabolism contributes to neurodegeneration and that dietary intervention with L. rhamnosus HA-114 restores lipid homeostasis and energy balance through mitochondrial β-oxidation. Our findings encourage the exploration of L. rhamnosus HA-114 derived interventions to modify the progression of neurodegenerative diseases.
Collapse
|
18
|
Identification of potentially functional modules and diagnostic genes related to amyotrophic lateral sclerosis based on the WGCNA and LASSO algorithms. Sci Rep 2022; 12:20144. [PMID: 36418457 PMCID: PMC9684499 DOI: 10.1038/s41598-022-24306-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 11/14/2022] [Indexed: 11/24/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a genetically and phenotypically heterogeneous disease results in the loss of motor neurons. Mounting information points to involvement of other systems including cognitive impairment. However, neither the valid biomarker for diagnosis nor effective therapeutic intervention is available for ALS. The present study is aimed at identifying potentially genetic biomarker that improves the diagnosis and treatment of ALS patients based on the data of the Gene Expression Omnibus. We retrieved datasets and conducted a weighted gene co-expression network analysis (WGCNA) to identify ALS-related co-expression genes. Functional enrichment analysis was performed to determine the features and pathways of the main modules. We then constructed an ALS-related model using the least absolute shrinkage and selection operator (LASSO) regression analysis and verified the model by the receiver operating characteristic (ROC) curve. Besides we screened the non-preserved gene modules in FTD and ALS-mimic disorders to distinct ALS-related genes from disorders with overlapping genes and features. Altogether, 4198 common genes between datasets with the most variation were analyzed and 16 distinct modules were identified through WGCNA. Blue module had the most correlation with ALS and functionally enriched in pathways of neurodegeneration-multiple diseases', 'amyotrophic lateral sclerosis', and 'endocytosis' KEGG terms. Further, some of other modules related to ALS were enriched in 'autophagy' and 'amyotrophic lateral sclerosis'. The 30 top of hub genes were recruited to a LASSO regression model and 5 genes (BCLAF1, GNA13, ARL6IP5, ARGLU1, and YPEL5) were identified as potentially diagnostic ALS biomarkers with validating of the ROC curve and AUC value.
Collapse
|
19
|
Fast Versus Slow Disease Progression in Amyotrophic Lateral Sclerosis – Clinical and Genetic Factors at the Edges of the Survival Spectrum. Neurobiol Aging 2022; 119:117-126. [DOI: 10.1016/j.neurobiolaging.2022.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 06/07/2022] [Accepted: 07/11/2022] [Indexed: 11/30/2022]
|
20
|
Henderson RD, Kepp KP, Eisen A. ALS/FTD: Evolution, Aging, and Cellular Metabolic Exhaustion. Front Neurol 2022; 13:890203. [PMID: 35711269 PMCID: PMC9196861 DOI: 10.3389/fneur.2022.890203] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 04/19/2022] [Indexed: 11/15/2022] Open
Abstract
Amyotrophic lateral sclerosis and frontotemporal dementia (ALS/FTD) are neurodegenerations with evolutionary underpinnings, expansive clinical presentations, and multiple genetic risk factors involving a complex network of pathways. This perspective considers the complex cellular pathology of aging motoneuronal and frontal/prefrontal cortical networks in the context of evolutionary, clinical, and biochemical features of the disease. We emphasize the importance of evolution in the development of the higher cortical function, within the influence of increasing lifespan. Particularly, the role of aging on the metabolic competence of delicately optimized neurons, age-related increased proteostatic costs, and specific genetic risk factors that gradually reduce the energy available for neuronal function leading to neuronal failure and disease.
Collapse
Affiliation(s)
| | - Kasper Planeta Kepp
- Department of Chemistry, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Andrew Eisen
- Division of Neurology, Department of Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
21
|
Baba D, Jingami N, Minami T, Park K, Takahashi R, Ohtsuru S. [A case of amyotrophic lateral sclerosis presenting with rapid progression of respiratory deterioration due to severe obesity]. Rinsho Shinkeigaku 2022; 62:602-608. [PMID: 35613859 DOI: 10.5692/clinicalneurol.cn-001723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
A 55-year-old woman with extreme obesity presenting with limb weakness since 1 year was diagnosed with amyotrophic lateral sclerosis (ALS) based on clinical findings and needle electromyography. She had a habit of overeating, and her body mass index (BMI) was 38.2. MRI showed an enlargement of the right central cerebral sulcus, and N-isopropyl-p-[123I]-iodoamphetamine single-photon emission computed tomography demonstrated reduced blood flow predominantly in the right frontal lobes, suggesting overlapping frontotemporal dementia (FTD). She maintained adequate dietary intake, and her BMI was stable at 38.2 until 3 months after diagnosis. However, over the next 2 months, her dietary intake decreased owing to pronounced bulbar palsy and BMI decreased to 34.5. At this point, forced vital capacity decreased from 69.3% to 39.0%, while forced expiratory volume in 1 second decreased from 75.3% to 47.7%. Consequently, noninvasive ventilation at night was initiated, followed by tracheostomy invasive ventilation at the emergency department after 2 months. We assume that the frontotemporal lobar degeneration pathology progressed to the frontal lobe and hypothalamus over time, which increased the patient's excessive appetite and body weight. Her obesity reduced the compliance of the thorax and increased the workload of the respiratory muscles, resulting in rapid respiratory deterioration. Additionally, the extensive neurodegeneration, extending to the area other than the primary motor cortex, might have played a pivotal role in rapid ALS progression. High-calorie nutritional management is generally recommended in patients with ALS. Although the prognosis of patients with ALS having BMI under 27 can be improved via high calorie intake and BMI maintenance, the nutritional management strategy for patients with ALS and high obesity (BMI ≥ 35) remains unclear. Through this case we emphasize that in patients with ALS and FTD excessive appetite and obesity can lead to rapid respiratory deterioration, and therefore, prudent calorie management is recommended.
Collapse
Affiliation(s)
- Daisuke Baba
- Department of Primary Care and Emergency Medicine, Graduate School of Medicine, Kyoto University
| | - Naoto Jingami
- Department of Primary Care and Emergency Medicine, Graduate School of Medicine, Kyoto University.,Department of Neurology, Graduate School of Medicine, Kyoto University
| | - Takuma Minami
- Department of Primary Care and Emergency Medicine, Graduate School of Medicine, Kyoto University.,Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University
| | - Kwiyoung Park
- Department of Neurology and Clinical Research Center, Utano National Hospital
| | - Ryosuke Takahashi
- Department of Neurology, Graduate School of Medicine, Kyoto University
| | - Shigeru Ohtsuru
- Department of Primary Care and Emergency Medicine, Graduate School of Medicine, Kyoto University
| |
Collapse
|
22
|
Body Weight Gain Is Associated with the Disease Stage in Advanced Amyotrophic Lateral Sclerosis with Invasive Ventilation. Metabolites 2022; 12:metabo12020191. [PMID: 35208264 PMCID: PMC8874426 DOI: 10.3390/metabo12020191] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/05/2022] [Accepted: 02/15/2022] [Indexed: 02/04/2023] Open
Abstract
We investigated the incidence of weight gain and its related factors in patients with amyotrophic lateral sclerosis (ALS) who underwent tracheostomy and invasive ventilation (TIV). Seventy-eight patients with ALS and TIV were enrolled and followed up prospectively. We clarified the clinical profiles of patients with increased weight following TIV and examined chronological variations in their body mass index (BMI), energy intake, and serum albumin levels. Post follow-up, we determined their disease stage according to their communication impairment (stage I to V) and investigated factors associated with BMI increase following TIV. Patients with a post-TIV BMI increase ≥1.86 kg/m2 demonstrated a higher incidence of ophthalmoplegia (76.2%), total quadriplegia (61.9%), severe communication impairment (stage V; 33.3%), and hypoalbuminemia than those with a BMI increase <1.86 kg/m2. Patients with stage V communication impairment exhibited a larger and faster BMI decrease before TIV (mean −4.2 kg/m2 and −2.5 kg/m2/year, respectively); a larger BMI increase (mean +4.6 kg/m2) following TIV, despite lower energy intake; and lower albumin levels post follow-up than those with lower-stage communication impairment. Multilevel linear regression analysis demonstrated an independent association between communication impairment stages (stage V) and a post-TIV BMI increase (p = 0.030). Weight gain and hypoalbuminemia during TIV in patients with ALS were associated with the disease stage and may be attributable to the neurodegenerative processes that are peculiar to ALS.
Collapse
|
23
|
Ye S, Luo Y, Jin P, Wang Y, Zhang N, Zhang G, Chen L, Shi L, Fan D. MRI Volumetric Analysis of the Thalamus and Hypothalamus in Amyotrophic Lateral Sclerosis. Front Aging Neurosci 2022; 13:610332. [PMID: 35046789 PMCID: PMC8763328 DOI: 10.3389/fnagi.2021.610332] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 11/16/2021] [Indexed: 11/17/2022] Open
Abstract
Background: Increasing evidence has shown that amyotrophic lateral sclerosis (ALS) can result in abnormal energy metabolism and sleep disorders, even before motor dysfunction. Although the hypothalamus and thalamus are important structures in these processes, few ALS studies have reported abnormal MRI structural findings in the hypothalamus and thalamus. Purpose: We aimed to investigate volumetric changes in the thalamus and hypothalamus by using the automatic brain structure volumetry tool AccuBrain®. Methods: 3D T1-weighted magnetization-prepared gradient echo imaging (MPRAGE) scans were acquired from 16 patients with ALS with normal cognitive scores and 16 age-, sex- and education-matched healthy controls. Brain tissue and structure volumes were automatically calculated using AccuBrain®. Results: There were no significant differences in bilateral thalamic (F = 1.31, p = 0.287) or hypothalamic volumes (F = 1.65, p = 0.213) between the ALS and control groups by multivariate analysis of covariance (MANCOVA). Left and right hypothalamic volumes were correlated with whole-brain volume in patients with ALS (t = 3.19, p = 0.036; t = 3.03, p = 0.044), while the correlation between age and bilateral thalamic volumes tended to be significant after Bonferroni correction (t = 2.76, p = 0.068; t = 2.83, p = 0.06). In the control group, left and right thalamic volumes were correlated with whole-brain volume (t = 4.26, p = 0.004; t = 4.52, p = 0.004). Conclusion: Thalamic and hypothalamic volumes did not show differences between patients with normal frontotemporal function ALS and healthy controls, but further studies are still needed.
Collapse
Affiliation(s)
- Shan Ye
- Department of Neurology, Peking University Third Hospital, Beijing, China.,Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China
| | - Yishan Luo
- Brain Research Institute, Shenzhen, China.,Department of Imaging and Interventional Radiology, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Pingping Jin
- Department of Neurology, Peking University Third Hospital, Beijing, China.,Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China
| | - Yajun Wang
- Department of Neurology, Peking University Third Hospital, Beijing, China.,Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China
| | - Nan Zhang
- Department of Neurology, Peking University Third Hospital, Beijing, China.,Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China
| | - Gan Zhang
- Department of Neurology, Peking University Third Hospital, Beijing, China.,Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China
| | - Lu Chen
- Department of Neurology, Peking University Third Hospital, Beijing, China.,Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China
| | - Lin Shi
- Brain Research Institute, Shenzhen, China.,Department of Imaging and Interventional Radiology, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Dongsheng Fan
- Department of Neurology, Peking University Third Hospital, Beijing, China.,Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China
| |
Collapse
|
24
|
Shapiro NL, Todd EG, Billot B, Cash DM, Iglesias JE, Warren JD, Rohrer JD, Bocchetta M. In vivo hypothalamic regional volumetry across the frontotemporal dementia spectrum. Neuroimage Clin 2022; 35:103084. [PMID: 35717886 PMCID: PMC9218583 DOI: 10.1016/j.nicl.2022.103084] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 06/07/2022] [Accepted: 06/11/2022] [Indexed: 11/04/2022]
Abstract
BACKGROUND Frontotemporal dementia (FTD) is a spectrum of diseases characterised by language, behavioural and motor symptoms. Among the different subcortical regions implicated in the FTD symptomatology, the hypothalamus regulates various bodily functions, including eating behaviours which are commonly present across the FTD spectrum. The pattern of specific hypothalamic involvement across the clinical, pathological, and genetic forms of FTD has yet to be fully investigated, and its possible associations with abnormal eating behaviours have yet to be fully explored. METHODS Using an automated segmentation tool for volumetric T1-weighted MR images, we measured hypothalamic regional volumes in a cohort of 439 patients with FTD (197 behavioural variant FTD [bvFTD]; 7 FTD with associated motor neurone disease [FTD-MND]; 99 semantic variant primary progressive aphasia [svPPA]; 117 non-fluent variant PPA [nfvPPA]; 19 PPA not otherwise specified [PPA-NOS]) and 118 age-matched controls. We compared volumes across the clinical, genetic (29 MAPT, 32 C9orf72, 23 GRN), and pathological diagnoses (61 tauopathy, 40 TDP-43opathy, 4 FUSopathy). We correlated the volumes with presence of abnormal eating behaviours assessed with the revised version of the Cambridge Behavioural Inventory (CBI-R). RESULTS On average, FTD patients showed 14% smaller hypothalamic volumes than controls. The groups with the smallest hypothalamic regions were FTD-MND (20%), MAPT (25%) and FUS (33%), with differences mainly localised in the anterior and posterior regions. The inferior tuberal region was only significantly smaller in tauopathies (MAPT and Pick's disease) and in TDP-43 type C compared to controls and was the only regions that did not correlate with eating symptoms. PPA-NOS and nfvPPA were the groups with the least frequent eating behaviours and the least hypothalamic involvement. CONCLUSIONS Abnormal hypothalamic volumes are present in all the FTD forms, but different hypothalamic regions might play a different role in the development of abnormal eating behavioural and metabolic symptoms. These findings might therefore help in the identification of different underlying pathological mechanisms, suggesting the potential use of hypothalamic imaging biomarkers and the research of potential therapeutic targets within the hypothalamic neuropeptides.
Collapse
Affiliation(s)
- Noah L Shapiro
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, UK
| | - Emily G Todd
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, UK
| | - Benjamin Billot
- Centre for Medical Image Computing, Department of Medical Physics and Biomedical Engineering, University College London, UK
| | - David M Cash
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, UK; Centre for Medical Image Computing, Department of Medical Physics and Biomedical Engineering, University College London, UK; UK Dementia Research Institute at UCL, UCL, London, UK
| | - Juan Eugenio Iglesias
- Centre for Medical Image Computing, Department of Medical Physics and Biomedical Engineering, University College London, UK; Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, USA; Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Boston, USA
| | - Jason D Warren
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, UK
| | - Jonathan D Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, UK
| | - Martina Bocchetta
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, UK.
| |
Collapse
|
25
|
Diet and Dementia: A Prospective Study. Nutrients 2021; 13:nu13124500. [PMID: 34960052 PMCID: PMC8705494 DOI: 10.3390/nu13124500] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/07/2021] [Accepted: 12/10/2021] [Indexed: 02/02/2023] Open
Abstract
Whether dietary and nutrition and dietary patterns are associated with the development of dementia is an interesting research question. Participants of a longitudinal cohort study that included European adults who were middle to old aged at baseline and who had not been diagnosed with dementia at baseline (2006–2010) and had not been diagnosed with dementia or died within 5 years after baseline were followed up (until 2018) and analyzed. Associations between intake frequency of each food class measured by the food-frequency questionnaire at baseline and incident dementia 5 years after baseline were analyzed after correcting for confounding variables. A total of approximately 340,000 participants and 900 cases were included in the analysis for each food class. Cox proportional hazard models with self-reported intake level of each food category divided into four mostly equally divided categorical variables revealed a high intake of bread, moderate total meat and total fish intake and low vegetable and fruit intake were thus associated with a small but significant decrease in the onset risk of dementia, while poultry and cereal were not. These findings are mostly inconsistent with the idea that Mediterranean diet is associated with lower risk of subsequent incident dementia.
Collapse
|
26
|
Devenney EM, McErlean K, Tse NY, Caga J, Dharmadasa T, Huynh W, Mahoney CJ, Zoing M, Mazumder S, Dobson-Stone C, Kwok JB, Halliday GM, Hodges JR, Piguet O, Ahmed RM, Kiernan MC. Factors That Influence Non-Motor Impairment Across the ALS-FTD Spectrum: Impact of Phenotype, Sex, Age, Onset and Disease Stage. Front Neurol 2021; 12:743688. [PMID: 34899567 PMCID: PMC8656429 DOI: 10.3389/fneur.2021.743688] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 10/26/2021] [Indexed: 11/13/2022] Open
Abstract
Objective: This study aimed to establish (1) the pattern and severity of neuropsychiatric symptoms and other non-motor symptoms of sleep and mood, across ALS phenotypes in comparison to bvFTD and (2) the contribution of non-modifiable factors including age, sex and disease state to the severity of symptoms experienced by ALS patients. Methods: Consecutive participants were recruited to the study and underwent a detailed clinical, cognitive, behavioral and neuroimaging assessment. Neuropsychiatric and other non-motor symptoms were determined using the Cambridge Behavioral Inventory, the CBI-R. The scores were converted to define impairment in terms of mild, moderate and severe symptoms for each subscale. Rate, severity and contribution of King's staging and modifiable factors were also determined and a regression model identified predictors of symptom severity. Results: In total, 250 participants (115 ALS, 98 bvFTD, and 37 ALS-FTD patients) were recruited. A similar pattern of neuropsychiatric symptom severity was identified (apathy, disinhibition and stereotypic behavior) for all behavioral phenotypes of ALS compared to bvFTD (all p > 0.05). Neuropsychiatric symptoms were also present in cases defined as ALSpure and the cognitive phenotype of ALS (ALSci) although they occurred less frequently and were at the milder end of the spectrum. Disordered sleep and disrupted mood were common across all phenotypes (all p < 0.05). The severity of sleep dysfunction was influenced by both sex and age (all p < 0.05). Neuropsychiatric symptoms, sleep and mood disorders were common early in the disease process and deteriorated in line with progression on the Amyotrophic Lateral Sclerosis Functional Rating Scale-Revised (ALSFRS-R; all p < 0.05). Diagnostic phenotype, disease duration and global cognition scores were the strongest predictors of non-motor and neuropsychiatric impairments. Conclusion: The current findings reveal strikingly similar patterns of changes across the subgroups of ALS and bvFTD, supporting the concept of the ALS-FTD spectrum. The findings further highlight the impact of non-motor and neuropsychiatric symptoms in patients with ALS, that are often as severe as that seen in ALS-FTD and bvFTD. This study advances understanding across the ALS-FTD spectrum that may accelerate the early identification of patient needs, to ensure prompt recognition of symptoms and thereby to improve clinical awareness, patient care and management.
Collapse
Affiliation(s)
- Emma M Devenney
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Kate McErlean
- Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Nga Yan Tse
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Jashelle Caga
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Thanuja Dharmadasa
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.,Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - William Huynh
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.,Prince of Wales Clinical School, University of New South Wales, Kensington, NSW, Australia
| | - Colin J Mahoney
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Margaret Zoing
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Srestha Mazumder
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Carol Dobson-Stone
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - John B Kwok
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Glenda M Halliday
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - John R Hodges
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Olivier Piguet
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia.,School of Psychology, The University of Sydney, Sydney, NSW, Australia
| | - Rebekah M Ahmed
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.,Institute of Clinical Neurosciences, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Matthew C Kiernan
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.,Institute of Clinical Neurosciences, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| |
Collapse
|
27
|
Phan K, He Y, Fu Y, Dzamko N, Bhatia S, Gold J, Rowe D, Ke YD, Ittner LM, Hodges JR, Piguet O, Kiernan MC, Halliday GM, Kim WS. Pathological manifestation of human endogenous retrovirus K in frontotemporal dementia. COMMUNICATIONS MEDICINE 2021; 1:60. [PMID: 35083468 PMCID: PMC8788987 DOI: 10.1038/s43856-021-00060-w] [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: 06/11/2021] [Accepted: 11/16/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Behavioral variant frontotemporal dementia (bvFTD) is a common form of younger-onset dementia with a proportion of cases overlapping pathologically and genetically with amyotrophic lateral sclerosis (ALS). Previous studies have identified that the human endogenous retrovirus K (HERV-K) is elevated in ALS serum and is associated with ALS TDP-43 pathology. In contrast, little is known about HERV-K changes in bvFTD. Here, we investigated the possible role of HERV-K in bvFTD. METHODS We measured the HERV-K env gene in sporadic bvFTD (N = 63), sporadic ALS (N = 89), and control (N = 21) serum by ddPCR. We also analyzed HERV-K env, by qPCR, and the HERV-K reverse transcriptase protein, by confocal immunofluorescence microscopy, in the disease-affected superior frontal cortex of bvFTD with TDP-43 pathology. RESULTS Here, we show that HERV-K env levels are significantly elevated (P = 3.5 × 10-6) in bvFTD compared to control serum, differentiating cases with an AUC value of 0.867. HERV-K env levels are also specifically elevated in the superior frontal cortex of bvFTD with TDP-43 pathology, with the HERV-K reverse transcriptase protein and TDP-43 deposit localized to the neuronal cytoplasm. Furthermore, in a neuronal cell line overexpression of TDP-43 induces HERV-K env transcription. CONCLUSIONS These results suggest that manifestation of HERV-K is associated with bvFTD TDP-43 pathology. Analysis of HERV-K in bvFTD may provide insight into an unrecognized but targetable perturbed pathology.
Collapse
Affiliation(s)
- Katherine Phan
- grid.1013.30000 0004 1936 834XThe University of Sydney, Brain and Mind Centre, Sydney, NSW Australia ,grid.1013.30000 0004 1936 834XThe University of Sydney, School of Medical Sciences, Sydney, NSW Australia
| | - Ying He
- grid.1013.30000 0004 1936 834XThe University of Sydney, Brain and Mind Centre, Sydney, NSW Australia ,grid.1013.30000 0004 1936 834XThe University of Sydney, School of Medical Sciences, Sydney, NSW Australia
| | - YuHong Fu
- grid.1013.30000 0004 1936 834XThe University of Sydney, Brain and Mind Centre, Sydney, NSW Australia ,grid.1013.30000 0004 1936 834XThe University of Sydney, School of Medical Sciences, Sydney, NSW Australia
| | - Nicolas Dzamko
- grid.1013.30000 0004 1936 834XThe University of Sydney, Brain and Mind Centre, Sydney, NSW Australia ,grid.1013.30000 0004 1936 834XThe University of Sydney, School of Medical Sciences, Sydney, NSW Australia
| | - Surabhi Bhatia
- grid.1013.30000 0004 1936 834XThe University of Sydney, Brain and Mind Centre, Sydney, NSW Australia ,grid.1013.30000 0004 1936 834XThe University of Sydney, School of Medical Sciences, Sydney, NSW Australia
| | - Julian Gold
- grid.452312.30000 0004 0644 0381The University of Sydney, The Albion Centre, Sydney, NSW Australia
| | - Dominic Rowe
- grid.1004.50000 0001 2158 5405Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| | - Yazi D. Ke
- grid.1004.50000 0001 2158 5405Dementia Research Centre and Department of Biomedical Sciences, Macquarie University, Sydney, NSW Australia
| | - Lars M. Ittner
- grid.1004.50000 0001 2158 5405Dementia Research Centre and Department of Biomedical Sciences, Macquarie University, Sydney, NSW Australia
| | - John R. Hodges
- grid.1013.30000 0004 1936 834XThe University of Sydney, Brain and Mind Centre, Sydney, NSW Australia
| | - Olivier Piguet
- grid.1013.30000 0004 1936 834XThe University of Sydney, Brain and Mind Centre, Sydney, NSW Australia ,grid.1013.30000 0004 1936 834XThe University of Sydney, School of Psychology, Sydney, NSW Australia
| | - Matthew C. Kiernan
- grid.1013.30000 0004 1936 834XThe University of Sydney, Brain and Mind Centre, Sydney, NSW Australia ,grid.413249.90000 0004 0385 0051Institute of Clinical Neurosciences, Royal Prince Alfred Hospital, Sydney, NSW Australia
| | - Glenda M. Halliday
- grid.1013.30000 0004 1936 834XThe University of Sydney, Brain and Mind Centre, Sydney, NSW Australia ,grid.1013.30000 0004 1936 834XThe University of Sydney, School of Medical Sciences, Sydney, NSW Australia
| | - Woojin Scott Kim
- The University of Sydney, Brain and Mind Centre, Sydney, NSW, Australia. .,The University of Sydney, School of Medical Sciences, Sydney, NSW, Australia.
| |
Collapse
|
28
|
Ahmed RM, Bocchetta M, Todd EG, Tse NY, Devenney EM, Tu S, Caga J, Hodges JR, Halliday GM, Irish M, Kiernan MC, Piguet O, Rohrer JD. Tackling clinical heterogeneity across the amyotrophic lateral sclerosis-frontotemporal dementia spectrum using a transdiagnostic approach. Brain Commun 2021; 3:fcab257. [PMID: 34805999 PMCID: PMC8599039 DOI: 10.1093/braincomms/fcab257] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 07/31/2021] [Accepted: 08/18/2021] [Indexed: 11/28/2022] Open
Abstract
The disease syndromes of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) display considerable clinical, genetic and pathological overlap, yet mounting evidence indicates substantial differences in progression and survival. To date, there has been limited examination of how profiles of brain atrophy might differ between clinical phenotypes. Here, we address this longstanding gap in the literature by assessing cortical and subcortical grey and white matter volumes on structural MRI in a large cohort of 209 participants. Cognitive and behavioural changes were assessed using the Addenbrooke’s Cognitive Examination and the Cambridge Behavioural Inventory. Relative to 58 controls, behavioural variant FTD (n = 58) and ALS–FTD (n = 41) patients displayed extensive atrophy of frontoinsular, cingulate, temporal and motor cortices, with marked subcortical atrophy targeting the hippocampus, amygdala, thalamus and striatum, with atrophy further extended to the brainstem, pons and cerebellum in the latter group. At the other end of the spectrum, pure-ALS patients (n = 52) displayed considerable frontoparietal atrophy, including right insular and motor cortices and pons and brainstem regions. Subcortical regions included the bilateral pallidum and putamen, but to a lesser degree than in the ALS–FTD and behavioural variant FTD groups. Across the spectrum the most affected region in all three groups was the insula, and specifically the anterior part (76–90% lower than controls). Direct comparison of the patient groups revealed disproportionate temporal atrophy and widespread subcortical involvement in ALS–FTD relative to pure-ALS. In contrast, pure-ALS displayed significantly greater parietal atrophy. Both behavioural variant FTD and ALS–FTD were characterized by volume decrease in the frontal lobes relative to pure-ALS. The motor cortex and insula emerged as differentiating structures between clinical syndromes, with bilateral motor cortex atrophy more pronounced in ALS–FTD compared with pure-ALS, and greater left motor cortex and insula atrophy relative to behavioural variant FTD. Taking a transdiagnostic approach, we found significant associations between abnormal behaviour and volume loss in a predominantly frontoinsular network involving the amygdala, striatum and thalamus. Our findings demonstrate the presence of distinct atrophy profiles across the ALS–FTD spectrum, with key structures including the motor cortex and insula. Notably, our results point to subcortical involvement in the origin of behavioural disturbances, potentially accounting for the marked phenotypic variability typically observed across the spectrum.
Collapse
Affiliation(s)
- Rebekah M Ahmed
- Memory and Cognition Clinic, Institute of Clinical Neurosciences, Royal Prince Alfred Hospital, Sydney 2050, Australia.,Brain and Mind Centre, The University of Sydney, Sydney, NSW 2050, Australia
| | - Martina Bocchetta
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London WC1E, UK
| | - Emily G Todd
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London WC1E, UK
| | - Nga Yan Tse
- Brain and Mind Centre, The University of Sydney, Sydney, NSW 2050, Australia
| | - Emma M Devenney
- Brain and Mind Centre, The University of Sydney, Sydney, NSW 2050, Australia
| | - Sicong Tu
- Brain and Mind Centre, The University of Sydney, Sydney, NSW 2050, Australia
| | - Jashelle Caga
- Brain and Mind Centre, The University of Sydney, Sydney, NSW 2050, Australia
| | - John R Hodges
- Brain and Mind Centre, The University of Sydney, Sydney, NSW 2050, Australia.,School of Psychology and Brain and Mind Centre, The University of Sydney, Sydney 2050, Australia
| | - Glenda M Halliday
- Brain and Mind Centre, The University of Sydney, Sydney, NSW 2050, Australia
| | - Muireann Irish
- School of Psychology and Brain and Mind Centre, The University of Sydney, Sydney 2050, Australia
| | - Matthew C Kiernan
- Memory and Cognition Clinic, Institute of Clinical Neurosciences, Royal Prince Alfred Hospital, Sydney 2050, Australia.,Brain and Mind Centre, The University of Sydney, Sydney, NSW 2050, Australia
| | - Olivier Piguet
- School of Psychology and Brain and Mind Centre, The University of Sydney, Sydney 2050, Australia
| | - Jonathan D Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London WC1E, UK
| |
Collapse
|
29
|
Swim Training Ameliorates Hyperlocomotion of ALS Mice and Increases Glutathione Peroxidase Activity in the Spinal Cord. Int J Mol Sci 2021; 22:ijms222111614. [PMID: 34769048 PMCID: PMC8583724 DOI: 10.3390/ijms222111614] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 12/18/2022] Open
Abstract
(1) Background: Amyotrophic lateral sclerosis (ALS) is an incurable, neurodegenerative disease. In some cases, ALS causes behavioral disturbances and cognitive dysfunction. Swimming has revealed a neuroprotective influence on the motor neurons in ALS. (2) Methods: In the present study, a SOD1-G93A mice model of ALS were used, with wild-type B6SJL mice as controls. ALS mice were analyzed before ALS onset (10th week of life), at ALS 1 onset (first symptoms of the disease, ALS 1 onset, and ALS 1 onset SWIM), and at terminal ALS (last stage of the disease, ALS TER, and ALS TER SWIM), and compared with wild-type mice. Swim training was applied 5 times per week for 30 min. All mice underwent behavioral tests. The spinal cord was analyzed for the enzyme activities and oxidative stress markers. (3) Results: Pre-symptomatic ALS mice showed increased locomotor activity versus control mice; the swim training reduced these symptoms. The metabolic changes in the spinal cord were present at the pre-symptomatic stage of the disease with a shift towards glycolytic processes at the terminal stage of ALS. Swim training caused an adaptation, resulting in higher glutathione peroxidase (GPx) and protection against oxidative stress. (4) Conclusion: Therapeutic aquatic activity might slow down the progression of ALS.
Collapse
|
30
|
Picillo M, Tepedino MF, Russillo MC, Abate F, Savastano M, De Simone A, Erro R, Pellecchia MT, Barone P. Energy expenditure, body composition and dietary habits in progressive supranuclear palsy. J Neurol 2021; 269:2610-2618. [PMID: 34676446 PMCID: PMC8530200 DOI: 10.1007/s00415-021-10846-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/21/2021] [Accepted: 10/11/2021] [Indexed: 12/02/2022]
Abstract
Introduction Little is known about metabolic changes in progressive supranuclear palsy. Goals of the present study are to: (1) investigate whether early progressive supranuclear palsy is associated with changes in energy expenditure, body composition and dietary intake compared with Parkinson’s disease and healthy controls; (2) assess the accuracy of the Harris–Benedict equation to predict measured rest energy expenditure in progressive supranuclear palsy; (3) verify differences according to sex, phenotypes, disease severity and presence of dysphagia in progressive supranuclear palsy. Methods Twenty-one progressive supranuclear palsy, 41 Parkinson’s disease and nine healthy controls were included. Rest energy expenditure was assessed with indirect calorimeter, body composition with bio-impedance analysis and physical activity and dietary intake were estimated with a validated frequency questionnaire. Parametric testing was used to analyze differences between groups. Results Progressive supranuclear palsy showed reduced total daily energy expenditure and physical activity compared to both other cohorts (p < 0.001) and a tendency toward lower fat-free mass compared to Parkinson’s disease (p > 0.05). Limited accuracy was shown for the Harris–Benedict equation (accurate prediction frequency < 60%). Greater disease severity was associated with lower rest energy expenditure (p = 0.030), fat-free mass (p = 0.026) and muscle mass (p = 0.029). Conclusion Greater disease severity is associated with reduction in rest energy expenditure likely due to the reduction in lean mass and muscle mass. Such data may pave the way to clinical trials evaluating the efficacy of muscle-targeted nutritional support and physical therapy in preserving muscle mass and improving motor performances in progressive supranuclear palsy at early stages. Supplementary Information The online version contains supplementary material available at 10.1007/s00415-021-10846-6.
Collapse
Affiliation(s)
- Marina Picillo
- Center for Neurodegenerative Diseases (CEMAND), Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via Allende, 84081, Baronissi, Salerno, Italy.
| | - Maria Francesca Tepedino
- Center for Neurodegenerative Diseases (CEMAND), Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via Allende, 84081, Baronissi, Salerno, Italy
| | - Maria Claudia Russillo
- Center for Neurodegenerative Diseases (CEMAND), Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via Allende, 84081, Baronissi, Salerno, Italy
| | - Filomena Abate
- Center for Neurodegenerative Diseases (CEMAND), Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via Allende, 84081, Baronissi, Salerno, Italy
| | - Marta Savastano
- Center for Neurodegenerative Diseases (CEMAND), Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via Allende, 84081, Baronissi, Salerno, Italy
| | - Antonio De Simone
- Center for Neurodegenerative Diseases (CEMAND), Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via Allende, 84081, Baronissi, Salerno, Italy
| | - Roberto Erro
- Center for Neurodegenerative Diseases (CEMAND), Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via Allende, 84081, Baronissi, Salerno, Italy
| | - Maria Teresa Pellecchia
- Center for Neurodegenerative Diseases (CEMAND), Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via Allende, 84081, Baronissi, Salerno, Italy
| | - Paolo Barone
- Center for Neurodegenerative Diseases (CEMAND), Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via Allende, 84081, Baronissi, Salerno, Italy
| |
Collapse
|
31
|
Chiò A, Canosa A, Calvo A, Moglia C, Cicolin A, Mora G. Developments in the assessment of non-motor disease progression in amyotrophic lateral sclerosis. Expert Rev Neurother 2021; 21:1419-1440. [PMID: 34554894 DOI: 10.1080/14737175.2021.1984883] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION The burden of non-motor symptoms is a major determinant of quality of life and outcome in amyotrophic lateral sclerosis (ALS) and has profound negative effect also on caregivers. AREAS COVERED Non-motor symptoms in ALS include cognitive impairment, neurobehavioral symptoms, depression and anxiety, suicidal ideation, pain, disordered sleep, fatigue, weight loss and reduced appetite, and autonomic dysfunctions. This review summarizes the measures used for the assessment of non-motor symptoms and their properties and recaps the frequency and progression of these symptoms along the course of ALS. EXPERT OPINION Non-motor symptoms in ALS represent a major component of the disease and span over several domains. These symptoms require a high level of medical attention and should be checked at each visit using ad hoc questionnaires and proactively treated. Several instruments assessing non-motor symptoms have been used in ALS. Specific screening questionnaires for non-motor symptoms can be used for monitoring patients during telehealth visits and for remote surveillance through sensors and apps installed on smartphones. Novel trials for non-motor symptoms treatment specifically designed for ALS are necessary to increase and refine the therapeutic armamentarium. Finally, scales assessing the most frequent and burdensome non-motor symptoms should be included in clinical trials.
Collapse
Affiliation(s)
- Adriano Chiò
- 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy.,Neurology I, Azienda Ospedaliera Città Della Salute E Della Scienza of Turin, Turin, Italy
| | - Antonio Canosa
- 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy.,Neurology I, Azienda Ospedaliera Città Della Salute E Della Scienza of Turin, Turin, Italy
| | - Andrea Calvo
- 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy.,Neurology I, Azienda Ospedaliera Città Della Salute E Della Scienza of Turin, Turin, Italy
| | - Cristina Moglia
- 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy.,Neurology I, Azienda Ospedaliera Città Della Salute E Della Scienza of Turin, Turin, Italy
| | - Alessandro Cicolin
- 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy.,Sleep Medicine Center, Azienda Ospedaliera Città Della Salute E Della Scienza of Turin, Turin, Italy
| | - Gabriele Mora
- Neurorehabilitation Department, Ics Maugeri Irccs, Institute of Milan, Milan, Italy
| |
Collapse
|
32
|
Extensive phenotypic characterisation of a human TDP-43 Q331K transgenic mouse model of amyotrophic lateral sclerosis (ALS). Sci Rep 2021; 11:16659. [PMID: 34404845 PMCID: PMC8370970 DOI: 10.1038/s41598-021-96122-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 08/04/2021] [Indexed: 11/29/2022] Open
Abstract
The majority of preclinical studies in ALS have relied on transgenic models with overexpression of mutant human superoxide dismutase 1 (SOD1), widely regarded to have failed in terms of translation of therapeutic effects. However, there are still no widely accepted models of other genetic subtypes of ALS. The majority of patients show ubiquitinated cytoplasmic inclusions of TAR DNA binding protein of 43 kilodaltons (TDP-43) in spinal motor neurons at the end stage of disease and a small proportion have mutations in TARDBP, the gene encoding TDP-43. TDP-43 transgenic mouse models have been produced, but have not been widely adopted. Here, we characterised one of these models available from the Jackson Laboratory in detail. Compared to TDP-43WT mice, TDP-43Q331K mice had 43% less hindlimb muscle mass at 6 months and a 73% reduction in hindlimb compound muscle action potential at 8 months of age. Rotarod and gait analysis indicated motor system decline with elevated weight gain. At the molecular level, the lack of TDP-43 cellular pathology was confirmed with a surprising increase in nuclear TDP-43 in motor neurons. Power analysis indicated group sizes of 12–14 mice are needed to detect 10–20% changes in measured parameters with a power of 80%, providing valid readouts for preclinical testing. Overall, this model may represent a useful component of multi-model pre-clinical therapeutic studies for ALS.
Collapse
|
33
|
Anhedonia in Semantic Dementia-Exploring Right Hemispheric Contributions to the Loss of Pleasure. Brain Sci 2021; 11:brainsci11080998. [PMID: 34439617 PMCID: PMC8392684 DOI: 10.3390/brainsci11080998] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/19/2021] [Accepted: 07/21/2021] [Indexed: 11/24/2022] Open
Abstract
Semantic dementia (SD) is a younger-onset neurodegenerative disease characterised by progressive deterioration of the semantic knowledge base in the context of predominantly left-lateralised anterior temporal lobe (ATL) atrophy. Mounting evidence indicates the emergence of florid socioemotional changes in SD as atrophy encroaches into right temporal regions. How lateralisation of temporal lobe pathology impacts the hedonic experience in SD remains largely unknown yet has important implications for understanding socioemotional and functional impairments in this syndrome. Here, we explored how lateralisation of temporal lobe atrophy impacts anhedonia severity on the Snaith–Hamilton Pleasure Scale in 28 SD patients presenting with variable right- (SD-R) and left-predominant (SD-L) profiles of temporal lobe atrophy compared to that of 30 participants with Alzheimer’s disease and 30 healthy older Control participants. Relative to Controls, SD-R but not SD-L or Alzheimer’s patients showed clinically significant anhedonia, representing a clear departure from premorbid levels. Overall, anhedonia was more strongly associated with functional impairment on the Frontotemporal Dementia Functional Rating Scale and motivational changes on the Cambridge Behavioural Inventory in SD than in Alzheimer’s disease patients. Voxel-based morphometry analyses revealed that anhedonia severity correlated with reduced grey matter intensity in a restricted set of regions centred on right orbitofrontal and temporopolar cortices, bilateral posterior temporal cortices, as well as the anterior cingulate gyrus and parahippocampal gyrus, bilaterally. Finally, regression and mediation analysis indicated a unique role for right temporal lobe structures in modulating anhedonia in SD. Our findings suggest that degeneration of predominantly right-hemisphere structures deleteriously impacts the capacity to experience pleasure in SD. These findings offer important insights into hemispheric lateralisation of motivational disturbances in dementia and suggest that anhedonia may emerge at different timescales in the SD disease trajectory depending on the integrity of the right hemisphere.
Collapse
|
34
|
Sol J, Jové M, Povedano M, Sproviero W, Domínguez R, Piñol-Ripoll G, Romero-Guevara R, Hye A, Al-Chalabi A, Torres P, Andres-Benito P, Area-Gómez E, Pamplona R, Ferrer I, Ayala V, Portero-Otín M. Lipidomic traits of plasma and cerebrospinal fluid in amyotrophic lateral sclerosis correlate with disease progression. Brain Commun 2021; 3:fcab143. [PMID: 34396104 PMCID: PMC8361390 DOI: 10.1093/braincomms/fcab143] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/10/2021] [Indexed: 01/01/2023] Open
Abstract
Since amyotrophic lateral sclerosis cases exhibit significant heterogeneity, we aim to investigate the association of lipid composition of plasma and CSF with amyotrophic lateral sclerosis diagnosis, its progression and clinical characteristics. Lipidome analyses would help to stratify patients on a molecular basis. For this reason, we have analysed the lipid composition of paired plasma and CSF samples from amyotrophic lateral sclerosis cases and age-matched non-amyotrophic lateral sclerosis individuals (controls) by comprehensive liquid chromatography coupled to mass spectrometry. The concentrations of neurofilament light chain-an index of neuronal damage-were also quantified in CSF samples and plasma. Amyotrophic lateral sclerosis versus control comparison, in a moderate stringency mode, showed that plasma from cases contains more differential lipids (n = 122 for raw P < 0.05; n = 27 for P < 0.01) than CSF (n = 17 for raw P < 0.05; n = 4 for P < 0.01), with almost no overlapping differential species, mainly characterized by an increased content of triacylglyceride species in plasma and decreased in CSF. Of note, false discovery rate correction indicated that one of the CSF lipids (monoacylglycerol 18:0) had high statistic robustness (false discovery rate-P < 0.01). Plasma lipidomes also varied significantly with the main involvement at onset (bulbar, spinal or respiratory). Notably, faster progression cases showed particular lipidome fingerprints, featured by decreased triacylclycerides and specific phospholipids in plasma, with 11 lipids with false discovery rate-P < 0.1 (n = 56 lipids in plasma for raw P < 0.01). Lipid species associated with progression rate clustered in a relatively low number of metabolic pathways, mainly triacylglyceride metabolism and glycerophospholipid and sphingolipid biosynthesis. A specific triacylglyceride (68:12), correlated with neurofilament content (r = 0.8, P < 0.008). Thus, the present findings suggest that systemic hypermetabolism-potentially sustained by increased triacylglyceride content-and CNS alterations of specific lipid pathways could be associated as modifiers of disease progression. Furthermore, these results confirm biochemical lipid heterogeneity in amyotrophic lateral sclerosis with different presentations and progression, suggesting the use of specific lipid species as potential disease classifiers.
Collapse
Affiliation(s)
- Joaquim Sol
- Metabolic Physiopathology Research Group, Experimental Medicine Department, Lleida University-Lleida Biochemical Research Institute (UdL-IRBLleida), Lleida, Spain
- Institut Català de la Salut, Atenció Primària, Lleida, Spain
- Research Support Unit Lleida, Fundació Institut Universitari per a la recerca a l'Atenció Primària de Salut Jordi Gol i Gurina (IDIAPJGol), Lleida, Spain
| | - Mariona Jové
- Metabolic Physiopathology Research Group, Experimental Medicine Department, Lleida University-Lleida Biochemical Research Institute (UdL-IRBLleida), Lleida, Spain
| | - Monica Povedano
- Functional Unit of Amyotrophic Lateral Sclerosis (UFELA), Service of Neurology, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
| | - William Sproviero
- Department of Basic and Clinical Neuroscience, King's College London, Maurice Wohl Clinical Neuroscience Institute, London, UK
| | - Raul Domínguez
- Functional Unit of Amyotrophic Lateral Sclerosis (UFELA), Service of Neurology, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Gerard Piñol-Ripoll
- Cognitive Disorders Unit, Clinical Neuroscience Research, IRBLleida-Hospital Universitari Santa Maria Lleida, Lleida, Spain
| | - Ricardo Romero-Guevara
- Metabolic Physiopathology Research Group, Experimental Medicine Department, Lleida University-Lleida Biochemical Research Institute (UdL-IRBLleida), Lleida, Spain
| | - Abdul Hye
- Department of Basic and Clinical Neuroscience, King's College London, Maurice Wohl Clinical Neuroscience Institute, London, UK
| | - Ammar Al-Chalabi
- Department of Basic and Clinical Neuroscience, King's College London, Maurice Wohl Clinical Neuroscience Institute, London, UK
| | - Pascual Torres
- Metabolic Physiopathology Research Group, Experimental Medicine Department, Lleida University-Lleida Biochemical Research Institute (UdL-IRBLleida), Lleida, Spain
| | - Pol Andres-Benito
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Barcelona, Spain
- CIBERNED (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, Ministry of Economy and Competitiveness, Barcelona, Spain
- Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Estela Area-Gómez
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Reinald Pamplona
- Metabolic Physiopathology Research Group, Experimental Medicine Department, Lleida University-Lleida Biochemical Research Institute (UdL-IRBLleida), Lleida, Spain
| | - Isidro Ferrer
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Barcelona, Spain
- CIBERNED (Network Centre of Biomedical Research of Neurodegenerative Diseases), Institute of Health Carlos III, Ministry of Economy and Competitiveness, Barcelona, Spain
- Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
- Senior Consultant, Bellvitge University Hospital, Barcelona, Spain
- Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| | - Victòria Ayala
- Metabolic Physiopathology Research Group, Experimental Medicine Department, Lleida University-Lleida Biochemical Research Institute (UdL-IRBLleida), Lleida, Spain
| | - Manuel Portero-Otín
- Metabolic Physiopathology Research Group, Experimental Medicine Department, Lleida University-Lleida Biochemical Research Institute (UdL-IRBLleida), Lleida, Spain
| |
Collapse
|
35
|
Bayer D, Antonucci S, Müller HP, Saad R, Dupuis L, Rasche V, Böckers TM, Ludolph AC, Kassubek J, Roselli F. Disruption of orbitofrontal-hypothalamic projections in a murine ALS model and in human patients. Transl Neurodegener 2021; 10:17. [PMID: 34059131 PMCID: PMC8168014 DOI: 10.1186/s40035-021-00241-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 05/11/2021] [Indexed: 12/12/2022] Open
Abstract
Background Increased catabolism has recently been recognized as a clinical manifestation of amyotrophic lateral sclerosis (ALS). The hypothalamic systems have been shown to be involved in the metabolic dysfunction in ALS, but the exact extent of hypothalamic circuit alterations in ALS is yet to be determined. Here we explored the integrity of large-scale cortico-hypothalamic circuits involved in energy homeostasis in murine models and in ALS patients. Methods The rAAV2-based large-scale projection mapping and image analysis pipeline based on Wholebrain and Ilastik software suites were used to identify and quantify projections from the forebrain to the lateral hypothalamus in the SOD1(G93A) ALS mouse model (hypermetabolic) and the FusΔNLS ALS mouse model (normo-metabolic). 3 T diffusion tensor imaging (DTI)-magnetic resonance imaging (MRI) was performed on 83 ALS and 65 control cases to investigate cortical projections to the lateral hypothalamus (LHA) in ALS. Results Symptomatic SOD1(G93A) mice displayed an expansion of projections from agranular insula, ventrolateral orbitofrontal and secondary motor cortex to the LHA. These findings were reproduced in an independent cohort by using a different analytic approach. In contrast, in the FusΔNLS ALS mouse model hypothalamic inputs from insula and orbitofrontal cortex were maintained while the projections from motor cortex were lost. The DTI-MRI data confirmed the disruption of the orbitofrontal-hypothalamic tract in ALS patients. Conclusion This study provides converging murine and human data demonstrating the selective structural disruption of hypothalamic inputs in ALS as a promising factor contributing to the origin of the hypermetabolic phenotype. Supplementary Information The online version contains supplementary material available at 10.1186/s40035-021-00241-6.
Collapse
Affiliation(s)
- David Bayer
- Department of Neurology, Ulm University, Ulm, Germany.,CEMMA (Cellular and Molecular Mechanisms in Aging) Research Training Group, Ulm, Germany
| | | | | | - Rami Saad
- Department of Neurology, Ulm University, Ulm, Germany
| | - Luc Dupuis
- University of Strasbourg, Strasbourg, France
| | - Volker Rasche
- Department of Internal Medicine II, Ulm University Medical Centre, Ulm, Germany
| | - Tobias M Böckers
- Institute of Anatomy and Cell Biology, Ulm University, Ulm, Germany.,German Center for Neurodegenerative Diseases-DZNE, Ulm, Germany
| | - Albert C Ludolph
- Department of Neurology, Ulm University, Ulm, Germany.,German Center for Neurodegenerative Diseases-DZNE, Ulm, Germany
| | - Jan Kassubek
- Department of Neurology, Ulm University, Ulm, Germany.,German Center for Neurodegenerative Diseases-DZNE, Ulm, Germany
| | - Francesco Roselli
- Department of Neurology, Ulm University, Ulm, Germany. .,German Center for Neurodegenerative Diseases-DZNE, Ulm, Germany.
| |
Collapse
|
36
|
Metabolic Dysfunction in Spinal Muscular Atrophy. Int J Mol Sci 2021; 22:ijms22115913. [PMID: 34072857 PMCID: PMC8198411 DOI: 10.3390/ijms22115913] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 05/25/2021] [Accepted: 05/29/2021] [Indexed: 12/11/2022] Open
Abstract
Spinal muscular atrophy (SMA) is an autosomal recessive genetic disorder leading to paralysis, muscle atrophy, and death. Significant advances in antisense oligonucleotide treatment and gene therapy have made it possible for SMA patients to benefit from improvements in many aspects of the once devastating natural history of the disease. How the depletion of survival motor neuron (SMN) protein, the product of the gene implicated in the disease, leads to the consequent pathogenic changes remains unresolved. Over the past few years, evidence toward a potential contribution of gastrointestinal, metabolic, and endocrine defects to disease phenotype has surfaced. These findings ranged from disrupted body composition, gastrointestinal tract, fatty acid, glucose, amino acid, and hormonal regulation. Together, these changes could have a meaningful clinical impact on disease traits. However, it is currently unclear whether these findings are secondary to widespread denervation or unique to the SMA phenotype. This review provides an in-depth account of metabolism-related research available to date, with a discussion of unique features compared to other motor neuron and related disorders.
Collapse
|
37
|
Ahmed RM, Tse NY, Chen Y, Henning E, Hodges JR, Kiernan MC, Irish M, Farooqi IS, Piguet O. Neural correlates of fat preference in frontotemporal dementia: translating insights from the obesity literature. Ann Clin Transl Neurol 2021; 8:1318-1329. [PMID: 33973740 PMCID: PMC8164857 DOI: 10.1002/acn3.51369] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/28/2021] [Accepted: 04/11/2021] [Indexed: 11/09/2022] Open
Abstract
OBJECTIVE Alterations in eating behaviour are one of the diagnostic features of behavioural variant frontotemporal dementia (bvFTD). It is hypothesised that underlying brain network disturbances and atrophy to key structures may affect macronutrient preference in bvFTD. We aimed to establish whether a preference for dietary fat exists in bvFTD, its association with cognitive symptoms and the underlying neural mechanisms driving these changes. METHODS Using a test meal paradigm, adapted from the obesity literature, with variable fat content (low 20%, medium 40% and high 60%), preference for fat in 20 bvFTD was compared to 16 Alzheimer's disease (AD) and 13 control participants. MRI brain scans were analysed to determine the neural correlates of fat preference. RESULTS Behavioural variant FTD patients preferred the high-fat meal compared to both AD (U = 61.5; p = 0.001) and controls (U = 41.5; p = 0.001), with 85% of bvFTD participants consistently rating the high-fat content meal as their preferred option. This increased preference for the high-fat meal was associated with total behavioural change (Cambridge Behavioural Inventory: rs = 0.462; p = 0.001), as well as overall functional decline (Frontotemporal Dementia Rating Scale: rs = -0.420; p = 0.03). A preference for high-fat content in bvFTD was associated with atrophy in an extended brain network including frontopolar, anterior cingulate, insular cortices, putamen and amygdala extending into lateral temporal, posteromedial parietal and occipital cortices. CONCLUSIONS Increased preference for fat content is associated with many of the canonical features of bvFTD. These findings offer new insights into markers of disease progression and pathogenesis, providing potential treatment targets.
Collapse
Affiliation(s)
- Rebekah M Ahmed
- Memory and Cognition Clinic, Department of Clinical Neurosciences, Royal Prince Alfred Hospital, Sydney, NSW, Australia.,Central Sydney Medical School and Brain & Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Nga Yan Tse
- Central Sydney Medical School and Brain & Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Yu Chen
- Central Sydney Medical School and Brain & Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Elana Henning
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, the NIHR Cambridge Biomedical Research Centre, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - John R Hodges
- Central Sydney Medical School and Brain & Mind Centre, The University of Sydney, Sydney, NSW, Australia.,ARC Centre of Excellence of Cognition and its Disorders, Sydney, NSW, Australia
| | - Matthew C Kiernan
- Memory and Cognition Clinic, Department of Clinical Neurosciences, Royal Prince Alfred Hospital, Sydney, NSW, Australia.,Central Sydney Medical School and Brain & Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Muireann Irish
- ARC Centre of Excellence of Cognition and its Disorders, Sydney, NSW, Australia.,School of Psychology and Brain & Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - I Sadaf Farooqi
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, the NIHR Cambridge Biomedical Research Centre, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Olivier Piguet
- ARC Centre of Excellence of Cognition and its Disorders, Sydney, NSW, Australia.,School of Psychology and Brain & Mind Centre, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
38
|
Mahoney CJ, Ahmed RM, Huynh W, Tu S, Rohrer JD, Bedlack RS, Hardiman O, Kiernan MC. Pathophysiology and Treatment of Non-motor Dysfunction in Amyotrophic Lateral Sclerosis. CNS Drugs 2021; 35:483-505. [PMID: 33993457 DOI: 10.1007/s40263-021-00820-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/20/2021] [Indexed: 12/21/2022]
Abstract
Amyotrophic lateral sclerosis is a progressive and fatal neurodegenerative disease typically presenting with bulbar or limb weakness. There is increasing evidence that amyotrophic lateral sclerosis is a multisystem disease with early and frequent impacts on cognition, behaviour, sleep, pain and fatigue. Dysfunction of normal physiological and metabolic processes also appears common. Evidence from pre-symptomatic studies and large epidemiological cohorts examining risk factors for the future development of amyotrophic lateral sclerosis have reported a high prevalence of changes in behaviour and mental health before the emergence of motor weakness. This suggests that changes beyond the motor system are underway at an early stage with dysfunction across brain networks regulating a variety of cognitive, behavioural and other homeostatic processes. The full impact of non-motor dysfunction continues to be established but there is now sufficient evidence that the presence of non-motor symptoms impacts overall survival in amyotrophic lateral sclerosis, and with up to 80% reporting non-motor symptoms, there is an urgent need to develop more robust therapeutic approaches. This review provides a contemporary overview of the pathobiology of non-motor dysfunction, offering readers a practical approach with regard to assessment and management. We review the current evidence for pharmacological and non-pharmacological treatment of non-motor dysfunction in amyotrophic lateral sclerosis and highlight the need to further integrate non-motor dysfunction as an important outcome measure for future clinical trial design.
Collapse
Affiliation(s)
- Colin J Mahoney
- Brain and Mind Centre, The University of Sydney, 94 Mallett Street, Camperdown, NSW, Australia.
| | - Rebekah M Ahmed
- Brain and Mind Centre, The University of Sydney, 94 Mallett Street, Camperdown, NSW, Australia.,Department of Neurology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - William Huynh
- Brain and Mind Centre, The University of Sydney, 94 Mallett Street, Camperdown, NSW, Australia
| | - Sicong Tu
- Brain and Mind Centre, The University of Sydney, 94 Mallett Street, Camperdown, NSW, Australia
| | - Jonathan D Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Richard S Bedlack
- Department of Neurology, Duke University Hospital, Durham, North Carolina, USA
| | - Orla Hardiman
- Academic Unit of Neurology, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
| | - Matthew C Kiernan
- Brain and Mind Centre, The University of Sydney, 94 Mallett Street, Camperdown, NSW, Australia.,Department of Neurology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| |
Collapse
|
39
|
Loss of appetite in patients with amyotrophic lateral sclerosis is associated with weight loss and anxiety/depression. Sci Rep 2021; 11:9119. [PMID: 33907295 PMCID: PMC8079393 DOI: 10.1038/s41598-021-88755-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 04/14/2021] [Indexed: 12/29/2022] Open
Abstract
Weight loss is common in patients with Amyotrophic lateral sclerosis (ALS), and associated with disease progression. Loss of appetite has been shown to be a contributor to weight loss in patients with amyotrophic lateral sclerosis (ALS). However, the reason of loss of appetite is not clear. The Council on Nutrition appetite questionnaire (CNAQ) and the simplified nutritional appetite questionnaire (SNAQ) are short and simple appetite assessment tools, which were using in ALS patients. In our study, the CNAQ and SNAQ were translated into Chinese, and their reliability and validity were tested. The Chinese version of the CNAQ (CNAQ-C) presented more appropriate reliability and validity than the SNAQ. Among the 94 ALS patients, 50 patients (53.2%) had loss of appetite, and we found that anxiety and/or depression contributed to the loss of appetite in the ALS patients. We reconfirmed that loss of appetite was associated with greater weight loss but not with clinical features of ALS. The loss of appetite caused by emotional problems in ALS patients should be taken seriously, and early intervention should be implemented to reduce weight loss.
Collapse
|
40
|
Reus LM, Pasaniuc B, Posthuma D, Boltz T, Pijnenburg YA, Ophoff RA. Gene Expression Imputation Across Multiple Tissue Types Provides Insight Into the Genetic Architecture of Frontotemporal Dementia and Its Clinical Subtypes. Biol Psychiatry 2021; 89:825-835. [PMID: 33637304 PMCID: PMC8415425 DOI: 10.1016/j.biopsych.2020.12.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 12/01/2020] [Accepted: 12/29/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND The etiology of frontotemporal dementia (FTD) is poorly understood. To identify genes with predicted expression levels associated with FTD, we integrated summary statistics with external reference gene expression data using a transcriptome-wide association study approach. METHODS FUSION software was used to leverage FTD summary statistics (all FTD: n = 2154 cases, n = 4308 controls; behavioral variant FTD: n = 1337 cases, n = 2754 controls; semantic dementia: n = 308 cases, n = 616 controls; progressive nonfluent aphasia: n = 269 cases, n = 538 controls; FTD with motor neuron disease: n = 200 cases, n = 400 controls) from the International FTD-Genomics Consortium with 53 expression quantitative loci tissue type panels (n = 12,205; 5 consortia). Significance was assessed using a 5% false discovery rate threshold. RESULTS We identified 73 significant gene-tissue associations for FTD, representing 44 unique genes in 34 tissue types. Most significant findings were derived from dorsolateral prefrontal cortex splicing data (n = 19 genes, 26%). The 17q21.31 inversion locus contained 23 significant associations, representing 6 unique genes. Other top hits included SEC22B (a gene involved in vesicle trafficking), TRGV5, and ZNF302. A single gene finding (RAB38) was observed for behavioral variant FTD. For other clinical subtypes, no significant associations were observed. CONCLUSIONS We identified novel candidate genes (e.g., SEC22B) and previously reported risk regions (e.g., 17q21.31) for FTD. Most significant associations were observed in dorsolateral prefrontal cortex splicing data despite the modest sample size of this reference panel. This suggests that our findings are specific to FTD and are likely to be biologically relevant highlights of genes at different FTD risk loci that are contributing to the disease pathology.
Collapse
Affiliation(s)
- Lianne M. Reus
- Alzheimer Center Amsterdam, Department of Neurology,
Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The
Netherlands
| | - Bogdan Pasaniuc
- Department of Human Genetics, David Geffen School of
Medicine, University of California Los Angeles, Los Angeles, California.,Department of Pathology and Laboratory Medicine, David
Geffen School of Medicine, University of California Los Angeles, Los Angeles,
California.,Department of Computational Medicine, David Geffen School
of Medicine, University of California Los Angeles, Los Angeles, California
| | - Danielle Posthuma
- Department of Complex Trait Genetics, Center for
Neurogenomics and Cognitive research, VU University Amsterdam, The
Netherlands
| | - Toni Boltz
- Department of Human Genetics, David Geffen School of
Medicine, University of California Los Angeles, Los Angeles, California
| | | | - Yolande A.L. Pijnenburg
- Alzheimer Center Amsterdam, Department of Neurology,
Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The
Netherlands
| | - Roel A Ophoff
- Department of Human Genetics, David Geffen School of
Medicine, University of California Los Angeles, Los Angeles, California.,Center for Neurobehavioral Genetics, University of
California Los Angeles, Los Angeles, California.,Department of Psychiatry, Erasmus University Medical Center
Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
41
|
Shaw SR, El-Omar H, Roquet D, Hodges JR, Piguet O, Ahmed RM, Whitton AE, Irish M. Uncovering the prevalence and neural substrates of anhedonia in frontotemporal dementia. Brain 2021; 144:1551-1564. [PMID: 33843983 DOI: 10.1093/brain/awab032] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 10/21/2020] [Accepted: 11/30/2020] [Indexed: 12/14/2022] Open
Abstract
Much of human behaviour is motivated by the drive to experience pleasure. The capacity to envisage pleasurable outcomes and to engage in goal-directed behaviour to secure these outcomes depends upon the integrity of frontostriatal circuits in the brain. Anhedonia refers to the diminished ability to experience, and to pursue, pleasurable outcomes, and represents a prominent motivational disturbance in neuropsychiatric disorders. Despite increasing evidence of motivational disturbances in frontotemporal dementia (FTD), no study to date has explored the hedonic experience in these syndromes. Here, we present the first study to document the prevalence and neural correlates of anhedonia in FTD in comparison with Alzheimer's disease, and its potential overlap with related motivational symptoms including apathy and depression. A total of 172 participants were recruited, including 87 FTD, 34 Alzheimer's disease, and 51 healthy older control participants. Within the FTD group, 55 cases were diagnosed with clinically probable behavioural variant FTD, 24 presented with semantic dementia, and eight cases had progressive non-fluent aphasia (PNFA). Premorbid and current anhedonia was measured using the Snaith-Hamilton Pleasure Scale, while apathy was assessed using the Dimensional Apathy Scale, and depression was indexed via the Depression, Anxiety and Stress Scale. Whole-brain voxel-based morphometry analysis was used to examine associations between grey matter atrophy and levels of anhedonia, apathy, and depression in patients. Relative to controls, behavioural variant FTD and semantic dementia, but not PNFA or Alzheimer's disease, patients showed clinically significant anhedonia, representing a clear departure from pre-morbid levels. Voxel-based morphometry analyses revealed that anhedonia was associated with atrophy in an extended frontostriatal network including orbitofrontal and medial prefrontal, paracingulate and insular cortices, as well as the putamen. Although correlated on the behavioural level, the neural correlates of anhedonia were largely dissociable from that of apathy, with only a small region of overlap detected in the right orbitofrontal cortices whilst no overlapping regions were found between anhedonia and depression. This is the first study, to our knowledge, to demonstrate profound anhedonia in FTD syndromes, reflecting atrophy of predominantly frontostriatal brain regions specialized for hedonic tone. Our findings point to the importance of considering anhedonia as a primary presenting feature of behavioural variant FTD and semantic dementia, with distinct neural drivers to that of apathy or depression. Future studies will be essential to address the impact of anhedonia on everyday activities, and to inform the development of targeted interventions to improve quality of life in patients and their families.
Collapse
Affiliation(s)
- Siobhán R Shaw
- The University of Sydney, Brain and Mind Centre, Sydney, New South Wales, Australia.,The University of Sydney, School of Psychology, Sydney, New South Wales, Australia
| | - Hashim El-Omar
- The University of Sydney, Brain and Mind Centre, Sydney, New South Wales, Australia.,The University of Sydney, School of Psychology, Sydney, New South Wales, Australia
| | - Daniel Roquet
- The University of Sydney, Brain and Mind Centre, Sydney, New South Wales, Australia.,The University of Sydney, School of Psychology, Sydney, New South Wales, Australia
| | - John R Hodges
- The University of Sydney, Brain and Mind Centre, Sydney, New South Wales, Australia.,ARC Centre of Excellence in Cognition and its Disorders, Sydney, New South Wales, Australia.,The University of Sydney, School of Medical Sciences, Sydney, New South Wales, Australia
| | - Olivier Piguet
- The University of Sydney, Brain and Mind Centre, Sydney, New South Wales, Australia.,The University of Sydney, School of Psychology, Sydney, New South Wales, Australia.,ARC Centre of Excellence in Cognition and its Disorders, Sydney, New South Wales, Australia
| | - Rebekah M Ahmed
- The University of Sydney, Brain and Mind Centre, Sydney, New South Wales, Australia.,The University of Sydney, School of Medical Sciences, Sydney, New South Wales, Australia.,Memory and Cognition Clinic, Department of Clinical Neurosciences, Royal Prince Alfred Hospital, Sydney, Australia
| | - Alexis E Whitton
- Black Dog Institute, University of New South Wales, Sydney, New South Wales, Australia
| | - Muireann Irish
- The University of Sydney, Brain and Mind Centre, Sydney, New South Wales, Australia.,The University of Sydney, School of Psychology, Sydney, New South Wales, Australia.,ARC Centre of Excellence in Cognition and its Disorders, Sydney, New South Wales, Australia
| |
Collapse
|
42
|
Dai Z, Kalra S, Mah D, Seres P, Sun H, Wu R, Wilman AH. Amide signal intensities may be reduced in the motor cortex and the corticospinal tract of ALS patients. Eur Radiol 2021; 31:1401-1409. [PMID: 32909054 DOI: 10.1007/s00330-020-07243-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 07/21/2020] [Accepted: 08/28/2020] [Indexed: 02/05/2023]
Abstract
OBJECTIVES The aim of the study is to assess amide concentration changes in ALS patients compared with healthy controls by using quantitative amide proton transfer (APT) and multiparameter magnetic resonance imaging, and testing its correlation with clinical scores. METHODS Sixteen ALS patients and sixteen healthy controls were recruited as part of the Canadian ALS Neuroimaging Consortium, and multimodal magnetic resonance imaging was performed at 3 T, including APT and diffusion imaging. Lorentz fitting was used to quantify the amide effect. Clinical disability was evaluated using the revised ALS functional rating scale (ALSFRS-R), and its correlation with image characteristics was assessed. The diagnostic performance of different imaging parameters was evaluated with receiver operating characteristic analysis. RESULTS Our results showed that the amide peak was significantly different between the motor cortex and other gray matter territories within the brain of ALS patients (p < 0.001). Compared with controls, amide signal intensities in ALS were significantly reduced in the motor cortex (p < 0.001) and corticospinal tract (p = 0.046), while abnormalities were not detected using routine imaging methods. There was no significant correlation between amide and ALSFRS-R score. The diagnostic accuracy of the amide peak was superior to that of diffusion imaging. CONCLUSIONS This study demonstrated changes of amide signal intensities in the motor cortex and corticospinal tract of ALS patients. KEY POINTS • The neurodegenerative disease amyotrophic lateral sclerosis (ALS) has a lack of objective imaging indicators for diagnosis and assessment. • Analysis of amide proton transfer imaging revealed changes in the motor cortex and corticospinal tract of ALS patients that were not visible on standard magnetic resonance imaging. • The diagnostic accuracy of the amide peak was superior to that of diffusion imaging.
Collapse
Affiliation(s)
- Zhuozhi Dai
- Department of Radiology, 2nd Affiliated Hospital of Shantou University Medical College, Shantou, 515041, Guangdong, China
- Department of Biomedical Engineering, University of Alberta, Edmonton, Alberta, T6G 2V2, Canada
| | - Sanjay Kalra
- Department of Medicine (Neurology), University of Alberta, Edmonton, Alberta, T6G 2S2, Canada
| | - Dennell Mah
- Department of Medicine (Neurology), University of Alberta, Edmonton, Alberta, T6G 2S2, Canada
| | - Peter Seres
- Department of Biomedical Engineering, University of Alberta, Edmonton, Alberta, T6G 2V2, Canada
| | - Hongfu Sun
- School of Information Technology and Electrical Engineering, University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Renhua Wu
- Department of Radiology, 2nd Affiliated Hospital of Shantou University Medical College, Shantou, 515041, Guangdong, China.
| | - Alan H Wilman
- Department of Biomedical Engineering, University of Alberta, Edmonton, Alberta, T6G 2V2, Canada.
| |
Collapse
|
43
|
Wei QQ, Ou R, Cao B, Chen Y, Hou Y, Zhang L, Wu F, Shang H. Early weight instability is associated with cognitive decline and poor survival in amyotrophic lateral sclerosis. Brain Res Bull 2021; 171:10-15. [PMID: 33636227 DOI: 10.1016/j.brainresbull.2021.02.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 02/19/2021] [Accepted: 02/21/2021] [Indexed: 02/08/2023]
Abstract
OBJECTIVE Our aim was to measure the monthly rate of weight loss during 6 months prior to a diagnosis of amyotrophic lateral sclerosis (ALS) and to explore the effect on prognosis. METHODS We enrolled 522 patients free from eating difficulties and with short diagnostic delay between June 2014 to June 2019. The calculating formula for the monthly rate of weight loss=[(weight at baseline-weight at diagnosis)/(weight at baseline*100 %)]/time interval. We employed logistic regression analysis to reveal any association between weight loss and cognitive dysfunction. Survival analysis was performed using the Kaplan-Meier curves and Cox proportional hazard models. RESULTS Weight loss was observed in 272 patients (52.1 %). Patients with severe weight loss had an older age of onset, a lower ALS Functional Rating Scale-Revised score, a faster disease progression rate, and higher frequencies of executive dysfunction and cognitive decline. The monthly rate of weight loss was associated with executive dysfunction and cognitive decline after adjusting for the emotional state. The stratified monthly rate of weight loss was strongly and independently related to ALS survival after adjusting for confounding factors (HR = 1.473, P trend<0.001). Each upper ladder of the rate of weight loss was correlated with worse survival and a 47.3 % (95 % CI: 25.0-73.6 %) increased risk of mortality. CONCLUSIONS Weight loss is very common in patients with ALS and is associated with poor survival. It is also associated with executive dysfunction and cognitive decline. An important mechanism of weight loss in the early stage of this disease may be hypermetabolism.
Collapse
Affiliation(s)
- Qian-Qian Wei
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Diseases Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ruwei Ou
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Diseases Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Bei Cao
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Diseases Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yongping Chen
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Diseases Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yanbing Hou
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Diseases Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lingyu Zhang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Diseases Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Fanyi Wu
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Diseases Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Huifang Shang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Diseases Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
44
|
Agarwal S, Highton-Williamson E, Caga J, Howells J, Dharmadasa T, Matamala JM, Ma Y, Shibuya K, Hodges JR, Ahmed RM, Vucic S, Kiernan MC. Motor cortical excitability predicts cognitive phenotypes in amyotrophic lateral sclerosis. Sci Rep 2021; 11:2172. [PMID: 33500476 PMCID: PMC7838179 DOI: 10.1038/s41598-021-81612-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 12/30/2020] [Indexed: 12/31/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are well-recognised as an extended disease spectrum. This study hypothesised that cortical hyperexcitability, an early pathophysiological abnormality in ALS, would distinguish cognitive phenotypes, as a surrogate marker of pathological disease burden. 61 patients with ALS, matched for disease duration (pure motor ALS, n = 39; ALS with coexistent FTD, ALS-FTD, n = 12; ALS with cognitive/behavioural abnormalities not meeting FTD criteria, ALS-Cog, n = 10) and 30 age-matched healthy controls. Cognitive function on the Addenbrooke's cognitive examination (ACE) scale, behavioural function on the motor neuron disease behavior scale (MiND-B) and cortical excitability using transcranial magnetic stimulation (TMS) were documented. Cortical resting motor threshold (RMT), lower threshold indicating hyperexcitability, was lower in ALS-FTD (50.2 ± 6.9) compared to controls (64.3 ± 12.6, p < 0.005), while ALS-Cog (63.3 ± 12.7) and ALS (60.8 ± 13.9, not significant) were similar to controls. Short interval intracortical inhibition (SICI) was reduced across all ALS groups compared to controls, indicating hyperexcitability. On receiver operating characteristic curve analysis, RMT differentiated ALS-FTD from ALS (area under the curve AUC = 0.745, p = 0.011). The present study has identified a distinct pattern of cortical excitability across cognitive phenotypes in ALS. As such, assessment of cortical physiology may provide more precise clinical prognostication in ALS.
Collapse
Affiliation(s)
- Smriti Agarwal
- Brain and Mind Centre and Sydney Medical School, University of Sydney, Sydney, Australia. .,Neurology Unit, A5, Box 165, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK.
| | | | - Jashelle Caga
- Brain and Mind Centre and Sydney Medical School, University of Sydney, Sydney, Australia
| | - James Howells
- Brain and Mind Centre and Sydney Medical School, University of Sydney, Sydney, Australia
| | - Thanuja Dharmadasa
- Brain and Mind Centre and Sydney Medical School, University of Sydney, Sydney, Australia
| | - José M Matamala
- Brain and Mind Centre and Sydney Medical School, University of Sydney, Sydney, Australia
| | - Yan Ma
- Brain and Mind Centre and Sydney Medical School, University of Sydney, Sydney, Australia
| | - Kazumoto Shibuya
- Brain and Mind Centre and Sydney Medical School, University of Sydney, Sydney, Australia
| | - John R Hodges
- Brain and Mind Centre and Sydney Medical School, University of Sydney, Sydney, Australia
| | - Rebekah M Ahmed
- Brain and Mind Centre and Sydney Medical School, University of Sydney, Sydney, Australia.,Institute of Clinical Neurosciences, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Steve Vucic
- Brain and Mind Centre and Sydney Medical School, University of Sydney, Sydney, Australia
| | - Matthew C Kiernan
- Brain and Mind Centre and Sydney Medical School, University of Sydney, Sydney, Australia.,Institute of Clinical Neurosciences, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| |
Collapse
|
45
|
El-Wahsh S, Finger EC, Piguet O, Mok V, Rohrer JD, Kiernan MC, Ahmed RM. Predictors of survival in frontotemporal lobar degeneration syndromes. J Neurol Neurosurg Psychiatry 2021; 92:jnnp-2020-324349. [PMID: 33441385 DOI: 10.1136/jnnp-2020-324349] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/26/2020] [Accepted: 12/14/2020] [Indexed: 12/11/2022]
Abstract
After decades of research, large-scale clinical trials in patients diagnosed with frontotemporal lobar degeneration (FTLD) are now underway across multiple centres worldwide. As such, refining the determinants of survival in FTLD represents a timely and important challenge. Specifically, disease outcome measures need greater clarity of definition to enable accurate tracking of therapeutic interventions in both clinical and research settings. Multiple factors potentially determine survival, including the clinical phenotype at presentation; radiological patterns of atrophy including markers on both structural and functional imaging; metabolic factors including eating behaviour and lipid metabolism; biomarkers including both serum and cerebrospinal fluid markers of underlying pathology; as well as genetic factors, including both dominantly inherited genes, but also genetic modifiers. The present review synthesises the effect of these factors on disease survival across the syndromes of frontotemporal dementia, with comparison to amyotrophic lateral sclerosis, progressive supranuclear palsy and corticobasal syndrome. A pathway is presented that outlines the utility of these varied survival factors for future clinical trials and drug development. Given the complexity of the FTLD spectrum, it seems unlikely that any single factor may predict overall survival in individual patients, further suggesting that a precision medicine approach will need to be developed in predicting disease survival in FTLD, to enhance drug target development and future clinical trial methodologies.
Collapse
Affiliation(s)
- Shadi El-Wahsh
- Department of Neurology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Elizabeth C Finger
- Department of Clinicial Neurological Sciences, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Olivier Piguet
- Brain and Mind Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Vincent Mok
- Gerald Choa Neuroscience Centre, Lui Che Woo Institute of Innovative Medicine, Margaret K.L. Cheung Research Centre for Management of Parkinsonism, Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Jonathan D Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Matthew C Kiernan
- Department of Neurology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
- Brain and Mind Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Rebekah M Ahmed
- Department of Neurology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
- Brain and Mind Centre, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
46
|
Vucic S, Kiernan MC, Menon P, Huynh W, Rynders A, Ho KS, Glanzman R, Hotchkin MT. Study protocol of RESCUE-ALS: A Phase 2, randomised, double-blind, placebo-controlled study in early symptomatic amyotrophic lateral sclerosis patients to assess bioenergetic catalysis with CNM-A u8 as a mechanism to slow diseas e progression. BMJ Open 2021; 11:e041479. [PMID: 33431491 PMCID: PMC7802642 DOI: 10.1136/bmjopen-2020-041479] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Amyotrophic lateral sclerosis (ALS) is an adult-onset, progressive and universally fatal neurodegenerative disorder. In Europe, Australia and Canada, riluzole is the only approved therapeutic agent for the treatment of ALS, while in the USA, riluzole and edaravone have been approved by the Food and Drug Administration (FDA) . Neither riluzole nor edaravone treatment has resulted in substantial disease-modifying effects. There is, therefore, an urgent need for drugs that result in safe and effective treatment. Here, we present the design and rationale for the phase 2 RESCUE-ALS study, investigating the novel nanocatalytic drug, CNM-Au8, as a therapeutic intervention that enhances the metabolic and energetic capacity of motor neurones. CNM-Au8 is an aqueous suspension of clean-surfaced, faceted gold nanocrystals that have extraordinary catalytic capabilities, that enhance efficiencies of key metabolic reactions, while simultaneously reducing levels of reactive oxygen species. This trial utilises a novel design by employing motor unit number index (MUNIX), measured by electromyography, as a quantitative measure of lower motor neurone loss and as an early marker of ALS disease progression. METHODS AND ANALYSIS This is a multicentre, randomised, double-blind, parallel group, placebo-controlled study of the efficacy, safety, pharmacokinetics and pharmacodynamics of CNM-Au8 in ALS patients. Patients will be randomised 1:1 to either receive 30 mg of CNM-Au8 once daily or matching placebo over a 36-week double-blind treatment period. Efficacy will be assessed as the change in motor neurone loss as measured by electromyography (eg, MUNIX, the primary endpoint; and secondary endpoints including MScanFit, motor unit size index, Split Hand Index, Neurophysiology Index). Exploratory endpoints include standard clinical and quality of life assessments. ETHICS AND DISSEMINATION RESCUE-ALS was approved by the Western Sydney Local Health District Human Research Ethics Committee (Ethics Ref: 2019/ETH12107). Results of the study will be submitted for publication in a peer-reviewed journal. TRIAL REGISTRATION NUMBER NCT04098406.
Collapse
Affiliation(s)
- Steve Vucic
- Department of Neurology, Westmead Hospital and Western Clinical School, University of Sydney, Sydney, New South Wales, Australia
| | - Matthew C Kiernan
- Brain and Mind Centre, University of Sydney, Sydney, New South Wales, Australia
- Department of Neurology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Parvathi Menon
- Department of Neurology, Westmead Hospital and Western Clinical School, University of Sydney, Sydney, New South Wales, Australia
| | - William Huynh
- Brain and Mind Centre, University of Sydney, Sydney, New South Wales, Australia
- Prince of Wales Clinical School, University of New South Wales, Sydney, New South Wales, Australia
| | | | - Karen S Ho
- Clene Nanomedicine, Salt Lake City, Utah, USA
| | | | | |
Collapse
|
47
|
Medinas DB, Hazari Y, Hetz C. Disruption of Endoplasmic Reticulum Proteostasis in Age-Related Nervous System Disorders. PROGRESS IN MOLECULAR AND SUBCELLULAR BIOLOGY 2021; 59:239-278. [PMID: 34050870 DOI: 10.1007/978-3-030-67696-4_12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Endoplasmic reticulum (ER) stress is a prominent cellular alteration of diseases impacting the nervous system that are associated to the accumulation of misfolded and aggregated protein species during aging. The unfolded protein response (UPR) is the main pathway mediating adaptation to ER stress, but it can also trigger deleterious cascades of inflammation and cell death leading to cell dysfunction and neurodegeneration. Genetic and pharmacological studies in experimental models shed light into molecular pathways possibly contributing to ER stress and the UPR activation in human neuropathies. Most of experimental models are, however, based on the overexpression of mutant proteins causing familial forms of these diseases or the administration of neurotoxins that induce pathology in young animals. Whether the mechanisms uncovered in these models are relevant for the etiology of the vast majority of age-related sporadic forms of neurodegenerative diseases is an open question. Here, we provide a systematic analysis of the current evidence linking ER stress to human pathology and the main mechanisms elucidated in experimental models. Furthermore, we highlight the recent association of metabolic syndrome to increased risk to undergo neurodegeneration, where ER stress arises as a common denominator in the pathogenic crosstalk between peripheral organs and the nervous system.
Collapse
Affiliation(s)
- Danilo B Medinas
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile. .,Program of Cellular and Molecular Biology, Center for Molecular Studies of the Cell, Institute of Biomedical Sciences, University of Chile, Santiago, Chile. .,Center for Geroscience, Brain Health and Metabolism, Santiago, Chile.
| | - Younis Hazari
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile.,Program of Cellular and Molecular Biology, Center for Molecular Studies of the Cell, Institute of Biomedical Sciences, University of Chile, Santiago, Chile.,Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Claudio Hetz
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile. .,Program of Cellular and Molecular Biology, Center for Molecular Studies of the Cell, Institute of Biomedical Sciences, University of Chile, Santiago, Chile. .,Center for Geroscience, Brain Health and Metabolism, Santiago, Chile. .,Buck Institute for Research on Aging, Novato, CA, USA.
| |
Collapse
|
48
|
Fostinelli S, De Amicis R, Leone A, Giustizieri V, Binetti G, Bertoli S, Battezzati A, Cappa SF. Eating Behavior in Aging and Dementia: The Need for a Comprehensive Assessment. Front Nutr 2020; 7:604488. [PMID: 33392240 PMCID: PMC7772186 DOI: 10.3389/fnut.2020.604488] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 11/23/2020] [Indexed: 01/01/2023] Open
Abstract
Eating behavior can change during aging due to physiological, psychological, and social changes. Modifications can occur at different levels: (1) in food choice, (2) in eating habits, and (3) in dietary intake. A good dietary behavior, like the Mediterranean dietary pattern, can be a protective factor for some aging related pathologies, such as dementia, while a worse eating behavior can lead to pathological conditions such as malnutrition. Changes in eating behavior can also be linked to the onset of dementia: for some types of dementia, such as frontotemporal dementia, dietary changes are one of the key clinical diagnostic feature, for others, like Alzheimer's disease, weight loss is a clinical reported feature. For these reasons, it is important to be able to assess eating behavior in a proper way, considering that there are normal age-related changes. An adequate assessment of dietary behavior can help to plan preventive intervention strategies for heathy aging or can help to identify abnormal behaviors that underline aging related-diseases. In this review, we have analyzed normal age-related and dementia-related changes and the tools that can be used to assess eating behavior. Thus, we make recommendations to screening and monitoring eating behavior in aging and dementia, and to adopt these tools in clinical practice.
Collapse
Affiliation(s)
- Silvia Fostinelli
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Ramona De Amicis
- Department of Food, Environmental and Nutritional Sciences, International Center for the Assessment of Nutritional Status, University of Milan, Milan, Italy
| | - Alessandro Leone
- Department of Food, Environmental and Nutritional Sciences, International Center for the Assessment of Nutritional Status, University of Milan, Milan, Italy
| | - Valentina Giustizieri
- Department of Food, Environmental and Nutritional Sciences, International Center for the Assessment of Nutritional Status, University of Milan, Milan, Italy
| | - Giuliano Binetti
- Memory Clinic, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Simona Bertoli
- Department of Food, Environmental and Nutritional Sciences, International Center for the Assessment of Nutritional Status, University of Milan, Milan, Italy.,Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Obesity Unit and Laboratory of Nutrition and Obesity Research, Milan, Italy
| | - Alberto Battezzati
- Department of Food, Environmental and Nutritional Sciences, International Center for the Assessment of Nutritional Status, University of Milan, Milan, Italy
| | - Stefano F Cappa
- University School for Advanced Studies, Pavia, Italy.,IRCCS Mondino Foundation, Pavia, Italy
| |
Collapse
|
49
|
Jääskeläinen O, Solje E, Hall A, Katisko K, Korhonen V, Tiainen M, Kangas AJ, Helisalmi S, Pikkarainen M, Koivisto A, Hartikainen P, Hiltunen M, Ala-Korpela M, Soininen H, Soininen P, Haapasalo A, Remes AM, Herukka SK. Low Serum High-Density Lipoprotein Cholesterol Levels Associate with the C9orf72 Repeat Expansion in Frontotemporal Lobar Degeneration Patients. J Alzheimers Dis 2020; 72:127-137. [PMID: 31561355 PMCID: PMC6839456 DOI: 10.3233/jad-190132] [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] [Indexed: 02/07/2023]
Abstract
Decreased levels of serum high-density lipoprotein (HDL) cholesterol have previously been linked to systemic inflammation and neurodegenerative diseases, such as Alzheimer’s disease. Here, we aimed to analyze the lipoprotein profile and inflammatory indicators, the high-sensitivity C-reactive peptide (hs-CRP) and glycoprotein acetyls (GlycA), in sporadic and C9orf72 repeat expansion-associated frontotemporal lobar degeneration (FTLD) patients. The C9orf72 hexanucleotide repeat expansion is the most frequent genetic etiology underlying FTLD. The concentrations of different lipid measures in the sera of 67 FTLD patients (15 C9orf72 repeat expansion carriers), including GlycA, were analyzed by nuclear magnetic resonance spectroscopy. To verify the state of systemic inflammation, hs-CRP was also quantified from patient sera. We found that the total serum HDL concentration was decreased in C9orf72 repeat expansion carriers when compared to non-carriers. Moreover, decreased concentrations of HDL particles of different sizes and subclass were consistently observed. No differences were detected in the very low- and low-density lipoprotein subclasses between the C9orf72 repeat expansion carriers and non-carriers. Furthermore, hs-CRP and GlycA levels did not differ between the C9orf72 repeat expansion carriers and non-carriers. In conclusion, the HDL-related changes were linked with C9orf72 repeat expansion associated FTLD but were not seen to associate with systemic inflammation. The underlying reason for the HDL changes remains unclear.
Collapse
Affiliation(s)
- Olli Jääskeläinen
- Institute of Clinical Medicine-Neurology, University of Eastern Finland, Kuopio, Finland
| | - Eino Solje
- Institute of Clinical Medicine-Neurology, University of Eastern Finland, Kuopio, Finland.,Neuro Center, Kuopio University Hospital, Kuopio, Finland
| | - Anette Hall
- Institute of Clinical Medicine-Neurology, University of Eastern Finland, Kuopio, Finland
| | - Kasper Katisko
- Institute of Clinical Medicine-Neurology, University of Eastern Finland, Kuopio, Finland
| | - Ville Korhonen
- Neuro Center, Kuopio University Hospital, Kuopio, Finland.,Institute of Clinical Medicine - Neurosurgery, University of Eastern Finland, Kuopio, Finland
| | - Mika Tiainen
- NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Antti J Kangas
- Computational Medicine, Faculty of Medicine, University of Oulu and Biocenter Oulu, Oulu, Finland
| | - Seppo Helisalmi
- Institute of Clinical Medicine-Neurology, University of Eastern Finland, Kuopio, Finland
| | - Maria Pikkarainen
- Institute of Clinical Medicine-Neurology, University of Eastern Finland, Kuopio, Finland
| | - Anne Koivisto
- Institute of Clinical Medicine-Neurology, University of Eastern Finland, Kuopio, Finland.,Neuro Center, Kuopio University Hospital, Kuopio, Finland
| | | | - Mikko Hiltunen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Mika Ala-Korpela
- NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland.,Computational Medicine, Faculty of Medicine, University of Oulu and Biocenter Oulu, Oulu, Finland.,Medical Research Council Integrative Epidemiology Unit at the University of Bristol, Bristol, UK.,Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK.,Systems Epidemiology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, The Alfred Hospital, Monash University, Melbourne, VIC, Australia
| | - Hilkka Soininen
- Institute of Clinical Medicine-Neurology, University of Eastern Finland, Kuopio, Finland.,Neuro Center, Kuopio University Hospital, Kuopio, Finland
| | - Pasi Soininen
- NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Annakaisa Haapasalo
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Anne M Remes
- Medical Research Center, Oulu University Hospital, Oulu, Finland.,Research Unit of Clinical Neuroscience, Neurology, University of Oulu, Oulu, Finland
| | - Sanna-Kaisa Herukka
- Institute of Clinical Medicine-Neurology, University of Eastern Finland, Kuopio, Finland.,Neuro Center, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
50
|
Bouscary A, Quessada C, René F, Spedding M, Turner BJ, Henriques A, Ngo ST, Loeffler JP. Sphingolipids metabolism alteration in the central nervous system: Amyotrophic lateral sclerosis (ALS) and other neurodegenerative diseases. Semin Cell Dev Biol 2020; 112:82-91. [PMID: 33160824 DOI: 10.1016/j.semcdb.2020.10.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 10/14/2020] [Accepted: 10/19/2020] [Indexed: 12/11/2022]
Abstract
Sphingolipids are complex lipids. They play a structural role in neurons, but are also involved in regulating cellular communication, and neuronal differentiation and maturation. There is increasing evidence to suggest that dysregulated metabolism of sphingolipids is linked to neurodegenerative processes in amyotrophic lateral sclerosis (ALS), Parkinson's disease and Gaucher's disease. In this review, we provide an overview of the role of sphingolipids in the development and maintenance of the nervous system. We describe the implications of altered metabolism of sphingolipids in the pathophysiology of certain neurodegenerative diseases, with a primary focus on ALS. Finally, we provide an update of potential treatments that could be used to target the metabolism of sphingolipids in neurodegenerative diseases.
Collapse
Affiliation(s)
- Alexandra Bouscary
- Université de Strasbourg, UMR_S 1118, Fédération de Médecine Translationnelle, CRBS, 1 rue Eugène Boeckel, 67000 Strasbourg, France; INSERM, U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, CRBS, 1 rue Eugène Boeckel, 67000 Strasbourg, France
| | - Cyril Quessada
- Université de Strasbourg, UMR_S 1118, Fédération de Médecine Translationnelle, CRBS, 1 rue Eugène Boeckel, 67000 Strasbourg, France; INSERM, U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, CRBS, 1 rue Eugène Boeckel, 67000 Strasbourg, France
| | - Frédérique René
- Université de Strasbourg, UMR_S 1118, Fédération de Médecine Translationnelle, CRBS, 1 rue Eugène Boeckel, 67000 Strasbourg, France; INSERM, U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, CRBS, 1 rue Eugène Boeckel, 67000 Strasbourg, France
| | - Michael Spedding
- Spedding Research Solutions SAS, 6 rue Ampere, 78650 Le Vesinet, France
| | - Bradley J Turner
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, 30 Royal Parade, Parkville, VIC 3052, Australia
| | | | - Shyuan T Ngo
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Building 75, Cnr College Rd & Cooper Rd, Brisbane city, QLD 4072, Australia; Centre for Clinical Research, The University of Queensland, Building 71/918, Royal Brisbane & Women's Hospital Campus, Herston, QLD 4029, Australia; Queensland Brain Institute Building 79, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Jean-Philippe Loeffler
- Université de Strasbourg, UMR_S 1118, Fédération de Médecine Translationnelle, CRBS, 1 rue Eugène Boeckel, 67000 Strasbourg, France; INSERM, U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, CRBS, 1 rue Eugène Boeckel, 67000 Strasbourg, France.
| |
Collapse
|