1
|
Webeck JA, Laing K, Andrews DM. Improvement in gait and functional abilities in an adult with posterior cortical atrophy after translingual neuromodulation with neurorehabilitation physical therapy: a case report. Physiother Theory Pract 2025:1-9. [PMID: 39922688 DOI: 10.1080/09593985.2025.2464849] [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: 03/23/2024] [Revised: 01/20/2025] [Accepted: 02/04/2025] [Indexed: 02/10/2025]
Abstract
BACKGROUND Posterior cortical atrophy (PCA) is a neurodegenerative syndrome characterized by progressive damage to the brain's visual and association areas, resulting in impaired spatial awareness, visual processing, and functional independence. PURPOSE This report examines the effects of a 14-week treatment protocol consisting of translingual neuromodulation via a portable neuromodulation stimulator (PoNS®1) in conjunction with physical therapy on balance, gait, and functional mobility in an adult male with PCA. CASE DESCRIPTION Assessments included objective and subjective measures of balance and gait - the 10-Metre Walk Test, Functional Gait Assessment (FGA), Dynamic Gait Index (DGI), Community Balance and Mobility Scale (CB&M), Neuro-Quality of Life (Neuro-QoL), and Activities-specific Balance Confidence (ABC) Scale. These were performed at baseline and weeks 4, 8, and 14 to evaluate the protocol's efficacy in improving balance, stability, and gait. OUTCOMES Postural stability, balance, gait patterning, and gait speed improved, enhancing daily functioning abilities and self-confidence. Gait speed improved by 0.48 m/s (comfortable) and 0.46 m/s (fast), exceeding MDC thresholds. The participant's FGA score increased 21 points and DGI increased 17 points, both exceeding their respective MDC thresholds (6 points for FGA, 3.2 points for DGI), reflecting marked gait improvements. The CB&M score rose 24 points, exceeding the MDC of 9.6 points. Despite these gains, gait speed remained below age-related norms. CONCLUSION Given the largely positive response to the protocol, further investigation should be undertaken to continue to explore the efficacy of PoNS® and physical therapy to determine its viability as a treatment for symptoms of PCA.
Collapse
Affiliation(s)
- Julie Ann Webeck
- Department of Kinesiology, University of Windsor, Ontario, Canada
| | - Katherine Laing
- Enable Physiotherapy, Neurological Physiotherapist, Ontario, Canada
| | - David M Andrews
- Department of Kinesiology, University of Windsor, Ontario, Canada
| |
Collapse
|
2
|
Takahashi H, Perez-Canamas A, Lee CW, Ye H, Han X, Strittmatter SM. Lysosomal TMEM106B interacts with galactosylceramidase to regulate myelin lipid metabolism. Commun Biol 2024; 7:1088. [PMID: 39237682 PMCID: PMC11377756 DOI: 10.1038/s42003-024-06810-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 08/30/2024] [Indexed: 09/07/2024] Open
Abstract
TMEM106B is an endolysosomal transmembrane protein not only associated with multiple neurological disorders including frontotemporal dementia, Alzheimer's disease, and hypomyelinating leukodystrophy but also potentially involved in COVID-19. Additionally, recent studies have identified amyloid fibrils of C-terminal TMEM106B in both aged healthy and neurodegenerative brains. However, so far little is known about physiological functions of TMEM106B in the endolysosome and how TMEM106B is involved in a wide range of human conditions at molecular levels. Here, we performed lipidomic analysis of the brain of TMEM106B-deficient mice. We found that TMEM106B deficiency significantly decreases levels of two major classes of myelin lipids, galactosylceramide and its sulfated derivative sulfatide. Subsequent co-immunoprecipitation assay showed that TMEM106B physically interacts with galactosylceramidase. We also found that galactosylceramidase activity was significantly increased in TMEM106B-deficient brains. Thus, our results suggest that TMEM106B interacts with galactosylceramidase to regulate myelin lipid metabolism and have implications for TMEM106B-associated diseases.
Collapse
Affiliation(s)
- Hideyuki Takahashi
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT, 06536, USA
| | - Azucena Perez-Canamas
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT, 06536, USA
| | - Chris W Lee
- Biomedical Research Institute of New Jersey (BRInj), Cedar Knolls, NJ, 07927, USA
- MidAtlantic Neonatology Associates (MANA), Morristown, NJ, 07960, USA
- Atlantic Health System, Morristown, NJ, 07960, USA
| | - Hongping Ye
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center At San Antonio, San Antonio, TX, 78229, USA
| | - Xianlin Han
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center At San Antonio, San Antonio, TX, 78229, USA
- Department of Medicine, University of Texas Health Science Center At San Antonio, San Antonio, TX, 78229, USA
| | - Stephen M Strittmatter
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT, 06536, USA.
| |
Collapse
|
3
|
Zota I, Chanoumidou K, Charalampopoulos I, Gravanis A. Dynamics of myelin deficits in the 5xFAD mouse model for Alzheimer's disease and the protective role of BDNF. Glia 2024; 72:809-827. [PMID: 38205694 DOI: 10.1002/glia.24505] [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: 06/11/2023] [Revised: 12/08/2023] [Accepted: 12/14/2023] [Indexed: 01/12/2024]
Abstract
Recent findings highlight myelin breakdown as a decisive early event in Alzheimer's Disease (AD) acting as aggravating factor of its progression. However, it is still unclear whether myelin loss is attributed to increased oligodendrocyte vulnerability, reduced repairing capacity or toxic stimuli. In the present study, we sought to clarify the starting point of myelin disruption accompanied with Oligodendrocyte Progenitor Cell (OPC) elimination in the brain of the 5xFAD mouse model of AD at 6 months of age in Dentate Gyrus of the hippocampus in relation to neurotrophin system. Prominent inflammation presence was detected since the age of 6 months playing a key role in myelin disturbance and AD progression. Expression analysis of neurotrophin receptors in OPCs was performed to identify new targets that could increase myelination in health and disease. OPCs in both control and 5xFAD mice express TrkB, TrkC and p75 receptors but not TrkA. Brain-derived neurotrophic factor (BDNF) that binds to TrkB receptor is well-known about its pro-myelination effect, promoting oligodendrocytes proliferation and differentiation, so we focused our investigation on its effects in OPCs under neurodegenerative conditions. Our in vitro results showed that BDNF rescues OPCs from death and promotes their proliferation and differentiation in presence of the toxic Amyloid-β 1-42. Collectively, our results indicate that BDNF possess an additional neuroprotective role through its actions on oligodendrocytic component and its use could be proposed as a drug-based myelin-enhancing strategy, complementary to amyloid and tau centered therapies in AD.
Collapse
Affiliation(s)
- Ioanna Zota
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas (IMBB-FORTH), Heraklion, Crete, Greece
| | - Konstantina Chanoumidou
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas (IMBB-FORTH), Heraklion, Crete, Greece
| | - Ioannis Charalampopoulos
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas (IMBB-FORTH), Heraklion, Crete, Greece
| | - Achille Gravanis
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas (IMBB-FORTH), Heraklion, Crete, Greece
| |
Collapse
|
4
|
Da X, Hempel E, Ou Y, Rowe OE, Malchano Z, Hajós M, Kern R, Megerian JT, Cimenser A. Noninvasive Gamma Sensory Stimulation May Reduce White Matter and Myelin Loss in Alzheimer's Disease. J Alzheimers Dis 2024; 97:359-372. [PMID: 38073386 PMCID: PMC10789351 DOI: 10.3233/jad-230506] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/27/2023] [Indexed: 12/19/2023]
Abstract
BACKGROUND Patients with Alzheimer's disease (AD) demonstrate progressive white matter atrophy and myelin loss. Restoring myelin content or preventing demyelination has been suggested as a therapeutic approach for AD. OBJECTIVE Herein, we investigate the effects of non-invasive, combined visual and auditory gamma-sensory stimulation on white matter atrophy and myelin content loss in patients with AD. METHODS In this study, we used the magnetic resonance imaging (MRI) data from the OVERTURE study (NCT03556280), a randomized, controlled, clinical trial in which active treatment participants received daily, non-invasive, combined visual and auditory, 40 Hz stimulation for six months. A subset of OVERTURE participants who meet the inclusion criteria for detailed white matter (N = 38) and myelin content (N = 36) assessments are included in the analysis. White matter volume assessments were performed using T1-weighted MRI, and myelin content assessments were performed using T1-weighted/T2-weighted MRI. Treatment effects on white matter atrophy and myelin content loss were assessed. RESULTS Combined visual and auditory gamma-sensory stimulation treatment is associated with reduced total and regional white matter atrophy and myelin content loss in active treatment participants compared to sham treatment participants. Across white matter structures evaluated, the most significant changes were observed in the entorhinal region. CONCLUSIONS The study results suggest that combined visual and auditory gamma-sensory stimulation may modulate neuronal network function in AD in part by reducing white matter atrophy and myelin content loss. Furthermore, the entorhinal region MRI outcomes may have significant implications for early disease intervention, considering the crucial afferent connections to the hippocampus and entorhinal cortex.
Collapse
Affiliation(s)
- Xiao Da
- Cognito Therapeutics, Inc., Cambridge, MA, USA
| | - Evan Hempel
- Cognito Therapeutics, Inc., Cambridge, MA, USA
| | - Yangming Ou
- Department of Radiology, Harvard Medical School, Boston, MA, USA
| | | | | | - Mihály Hajós
- Cognito Therapeutics, Inc., Cambridge, MA, USA
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Ralph Kern
- Cognito Therapeutics, Inc., Cambridge, MA, USA
| | | | | |
Collapse
|
5
|
Takahashi H, Perez-Canamas A, Ye H, Han X, Strittmatter SM. Lysosomal TMEM106B interacts with galactosylceramidase to regulate myelin lipid metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.14.557804. [PMID: 37745346 PMCID: PMC10515910 DOI: 10.1101/2023.09.14.557804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
TMEM106B is an endolysosomal transmembrane protein not only associated with multiple neurological disorders including frontotemporal dementia, Alzheimer's disease, and hypomyelinating leukodystrophy but also potentially involved in COVID-19. Additionally, recent studies have identified amyloid fibrils of C-terminal TMEM106B in both aged healthy and neurodegenerative brains. However, so far little is known about physiological functions of TMEM106B in the endolysosome and how TMEM106B is involved in a wide range of human conditions at molecular levels. Here, we performed lipidomic analysis of the brain of TMEM106B-deficient mice. We found that TMEM106B deficiency significantly decreases levels of two major classes of myelin lipids, galactosylceramide and its sulfated derivative sulfatide. Subsequent co-immunoprecipitation assay showed that TMEM106B physically interacts with galactosylceramidase. We also found that galactosyceramidase activity was significantly increased in TMEM106B-deficient brains. Thus, our results reveal a novel function of TMEM106B interacting with galactosyceramidase to regulate myelin lipid metabolism and have implications for TMEM106B-associated diseases.
Collapse
Affiliation(s)
- Hideyuki Takahashi
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Azucena Perez-Canamas
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Hongping Ye
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center At San Antonio, San Antonio, TX, 78229, USA
| | - Xianlin Han
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center At San Antonio, San Antonio, TX, 78229, USA
- Department of Medicine, University of Texas Health Science Center At San Antonio, San Antonio, TX, 78229, USA
| | - Stephen M. Strittmatter
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT 06536, USA
| |
Collapse
|
6
|
Azargoonjahromi A. Dual role of nitric oxide in Alzheimer's Disease. Nitric Oxide 2023; 134-135:23-37. [PMID: 37019299 DOI: 10.1016/j.niox.2023.03.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 03/02/2023] [Accepted: 03/31/2023] [Indexed: 04/05/2023]
Abstract
Nitric oxide (NO), an enzymatic product of nitric oxide synthase (NOS), has been associated with a variety of neurological diseases such as Alzheimer's disease (AD). NO has long been thought to contribute to neurotoxic insults caused by neuroinflammation in AD. This perception shifts as more attention is paid to the early stages before cognitive problems manifest. However, it has revealed a compensatory neuroprotective role for NO that protects synapses by increasing neuronal excitability. NO can positively affect neurons by inducing neuroplasticity, neuroprotection, and myelination, as well as having cytolytic activity to reduce inflammation. NO can also induce long-term potentiation (LTP), a process by which synaptic connections among neurons become more potent. Not to mention that such functions give rise to AD protection. Notably, it is unquestionably necessary to conduct more research to clarify NO pathways in neurodegenerative dementias because doing so could help us better understand their pathophysiology and develop more effective treatment options. All these findings bring us to the prevailing notion that NO can be used either as a therapeutic agent in patients afflicted with AD and other memory impairment disorders or as a contributor to the neurotoxic and aggressive factor in AD. In this review, after presenting a general background on AD and NO, various factors that have a pivotal role in both protecting and exacerbating AD and their correlation with NO will be elucidated. Following this, both the neuroprotective and neurotoxic effects of NO on neurons and glial cells among AD cases will be discussed in detail.
Collapse
|
7
|
Sharp FR, DeCarli CS, Jin LW, Zhan X. White matter injury, cholesterol dysmetabolism, and APP/Abeta dysmetabolism interact to produce Alzheimer's disease (AD) neuropathology: A hypothesis and review. Front Aging Neurosci 2023; 15:1096206. [PMID: 36845656 PMCID: PMC9950279 DOI: 10.3389/fnagi.2023.1096206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 01/30/2023] [Indexed: 02/12/2023] Open
Abstract
We postulate that myelin injury contributes to cholesterol release from myelin and cholesterol dysmetabolism which contributes to Abeta dysmetabolism, and combined with genetic and AD risk factors, leads to increased Abeta and amyloid plaques. Increased Abeta damages myelin to form a vicious injury cycle. Thus, white matter injury, cholesterol dysmetabolism and Abeta dysmetabolism interact to produce or worsen AD neuropathology. The amyloid cascade is the leading hypothesis for the cause of Alzheimer's disease (AD). The failure of clinical trials based on this hypothesis has raised other possibilities. Even with a possible new success (Lecanemab), it is not clear whether this is a cause or a result of the disease. With the discovery in 1993 that the apolipoprotein E type 4 allele (APOE4) was the major risk factor for sporadic, late-onset AD (LOAD), there has been increasing interest in cholesterol in AD since APOE is a major cholesterol transporter. Recent studies show that cholesterol metabolism is intricately involved with Abeta (Aβ)/amyloid transport and metabolism, with cholesterol down-regulating the Aβ LRP1 transporter and upregulating the Aβ RAGE receptor, both of which would increase brain Aβ. Moreover, manipulating cholesterol transport and metabolism in rodent AD models can ameliorate pathology and cognitive deficits, or worsen them depending upon the manipulation. Though white matter (WM) injury has been noted in AD brain since Alzheimer's initial observations, recent studies have shown abnormal white matter in every AD brain. Moreover, there is age-related WM injury in normal individuals that occurs earlier and is worse with the APOE4 genotype. Moreover, WM injury precedes formation of plaques and tangles in human Familial Alzheimer's disease (FAD) and precedes plaque formation in rodent AD models. Restoring WM in rodent AD models improves cognition without affecting AD pathology. Thus, we postulate that the amyloid cascade, cholesterol dysmetabolism and white matter injury interact to produce and/or worsen AD pathology. We further postulate that the primary initiating event could be related to any of the three, with age a major factor for WM injury, diet and APOE4 and other genes a factor for cholesterol dysmetabolism, and FAD and other genes for Abeta dysmetabolism.
Collapse
Affiliation(s)
- Frank R. Sharp
- Department of Neurology, The MIND Institute, University of California at Davis Medical Center, Sacramento, CA, United States
| | | | | | | |
Collapse
|
8
|
Singh NA, Graff-Radford J, Machulda MM, Thu NT, Schwarz CG, Reid RI, Lowe VJ, Petersen RC, Jack CR, Josephs KA, Whitwell JL. Diffusivity Changes in Posterior Cortical Atrophy and Logopenic Progressive Aphasia: A Longitudinal Diffusion Tensor Imaging Study. J Alzheimers Dis 2023; 94:709-725. [PMID: 37302032 PMCID: PMC10785680 DOI: 10.3233/jad-221217] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
BACKGROUND Posterior cortical atrophy (PCA) and logopenic progressive aphasia (LPA) are associated with characteristic patterns of structural network degeneration. Little is known about longitudinal patterns of white matter tract degeneration in these phenotypes. OBJECTIVE To assess longitudinal patterns of white matter degeneration and identify phenotype specific cross-sectional and longitudinal diffusion tensor imaging (DTI) biomarkers in PCA and LPA. METHODS Twenty-five PCA, 22 LPA and 25 cognitively unimpaired (CU) individuals were recruited and underwent structural MRI that included a DTI sequence with a follow-up one year later. Cross-sectional and longitudinal mixed effects models were fit to assess the effects of diagnosis on baseline and annualized change in regional DTI metrics. Discriminatory power was investigated using the area under the receiver operating characteristic curves (AUROC). RESULTS PCA and LPA showed overlapping white matter degeneration profiles predominantly in the left occipital and temporal lobes, the posterior thalamic radiation and sagittal stratum at baseline, as well as the parietal lobe longitudinally. PCA showed degeneration in the occipital and parietal white matter, cross-sectionally and longitudinally, compared to CU, while LPA showed greater degeneration in the temporal and inferior parietal white matter and the inferior fronto-occipital fasciculus cross-sectionally, and in parietal white matter longitudinally compared to CU. Cross-sectionally, integrity of the inferior occipital white matter was best able to differentiate PCA from LPA, with an AUROC of 0.82. CONCLUSION These findings contribute to our understanding of white matter degeneration and support usage of DTI as a useful additional diagnostic biomarker for PCA and LPA.
Collapse
Affiliation(s)
| | | | - Mary M. Machulda
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, MN, USA
| | - Nha Trang Thu
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | | | - Robert I. Reid
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
- Department of Information Technology, Mayo Clinic, Rochester, MN, USA
| | - Val J. Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | |
Collapse
|
9
|
Rahmani F, Ghezzi L, Tosti V, Liu J, Song SK, Wu AT, Rajamanickam J, Obert KA, Benzinger TL, Mittendorfer B, Piccio L, Raji CA. Twelve Weeks of Intermittent Caloric Restriction Diet Mitigates Neuroinflammation in Midlife Individuals with Multiple Sclerosis: A Pilot Study with Implications for Prevention of Alzheimer's Disease. J Alzheimers Dis 2023; 93:263-273. [PMID: 37005885 PMCID: PMC10460547 DOI: 10.3233/jad-221007] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
BACKGROUND Multiple sclerosis (MS) is a prototype neuroinflammatory disorder with increasingly recognized role for neurodegeneration. Most first-line treatments cannot prevent the progression of neurodegeneration and the resultant disability. Interventions can improve symptoms of MS and might provide insights into the underlying pathology. OBJECTIVE To investigate the effect of intermittent caloric restriction on neuroimaging markers of MS. METHODS We randomized ten participants with relapsing remitting MS to either a 12-week intermittent calorie restriction (iCR) diet (n = 5) or control (n = 5). Cortical thickness and volumes were measured through FreeSurfer, cortical perfusion was measured by arterial spin labeling and neuroinflammation through diffusion basis spectrum imaging. RESULTS After 12 weeks of iCR, brain volume increased in the left superior and inferior parietal gyri (p: 0.050 and 0.049, respectively) and the banks of the superior temporal sulcus (p: 0.01). Similarly in the iCR group, cortical thickness improved in the bilateral medial orbitofrontal gyri (p: 0.04 and 0.05 in right and left, respectively), the left superior temporal gyrus (p: 0.03), and the frontal pole (p: 0.008) among others. Cerebral perfusion decreased in the bilateral fusiform gyri (p: 0.047 and 0.02 in right and left, respectively) and increased in the bilateral deep anterior white matter (p: 0.03 and 0.013 in right and left, respectively). Neuroinflammation, demonstrated through hindered and restricted water fractions (HF and RF), decreased in the left optic tract (HF p: 0.02), and the right extreme capsule (RF p: 0.007 and HF p: 0.003). CONCLUSION These pilot data suggest therapeutic effects of iCR in improving cortical volume and thickness and mitigating neuroinflammation in midlife adults with MS.
Collapse
Affiliation(s)
- Farzaneh Rahmani
- Mallinckrodt Institute of Radiology, Division of Neuroradiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Laura Ghezzi
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Valeria Tosti
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Jingxia Liu
- Mallinckrodt Institute of Radiology, Division of Neuroradiology, Washington University in St. Louis, St. Louis, MO, USA
- Department of Surgery, Division of Public Health Sciences, Washington University in St. Louis, St. Louis, MO, USA
| | - Sheng-Kwei Song
- Department of Physics, Washington University in St. Louis, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University in St. Louis, St. Louis, MO, USA
| | - Anthony T. Wu
- Department of Physics, Washington University in St. Louis, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University in St. Louis, St. Louis, MO, USA
| | - Jayashree Rajamanickam
- Mallinckrodt Institute of Radiology, Division of Neuroradiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Kathleen A. Obert
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Tammie L.S. Benzinger
- Mallinckrodt Institute of Radiology, Division of Neuroradiology, Washington University in St. Louis, St. Louis, MO, USA
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University in St Louis, St. Louis, MO, USA
| | - Bettina Mittendorfer
- Department of Medicine, Division of Geriatrics and Nutritional Science, Washington University in St. Louis, St. Louis, MO, USA
| | - Laura Piccio
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University in St. Louis, St. Louis, MO, USA
- Brain and Mind Centre, School of Medical Sciences, The University of Sydney, NSW, Australia
- Charles Perkin Centre, The University of Sydney NSW, Australia
| | - Cyrus A. Raji
- Mallinckrodt Institute of Radiology, Division of Neuroradiology, Washington University in St. Louis, St. Louis, MO, USA
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University in St Louis, St. Louis, MO, USA
| |
Collapse
|
10
|
Alenius M, Hokkanen L, Koskinen S, Hallikainen I, Hänninen T, Karrasch M, Raivio MM, Laakkonen ML, Krüger J, Suhonen NM, Kivipelto M, Ngandu T. Cognitive Performance at Time of AD Diagnosis: A Clinically Augmented Register-Based Study. Front Psychol 2022; 13:901945. [PMID: 35846684 PMCID: PMC9284003 DOI: 10.3389/fpsyg.2022.901945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/02/2022] [Indexed: 12/02/2022] Open
Abstract
We aimed to evaluate the feasibility of using real-world register data for identifying persons with mild Alzheimer’s disease (AD) and to describe their cognitive performance at the time of diagnosis. Patients diagnosed with AD during 2010–2013 (aged 60–81 years) were identified from the Finnish national health registers and enlarged with a smaller private sector sample (total n = 1,268). Patients with other disorders impacting cognition were excluded. Detailed clinical and cognitive screening data (the Consortium to Establish a Registry for Alzheimer’s Disease neuropsychological battery [CERAD-nb]) were obtained from local health records. Adequate cognitive data were available for 389 patients with mild AD (31%) of the entire AD group. The main reasons for not including patients in analyses of cognitive performance were AD diagnosis at a moderate/severe stage (n = 266, 21%), AD diagnosis given before full register coverage (n = 152, 12%), and missing CERAD-nb data (n = 139, 11%). The cognitive performance of persons with late-onset AD (n = 284), mixed cerebrovascular disease and AD (n = 51), and other AD subtypes (n = 54) was compared with that of a non-demented sample (n = 1980) from the general population. Compared with the other AD groups, patients with late-onset AD performed the worst in word list recognition, while patients with mixed cerebrovascular disease and AD performed the worst in constructional praxis and clock drawing tests. A combination of national registers and local health records can be used to collect data relevant for cognitive screening; today, the process is laborious, but it could be improved in the future with refined search algorithms and electronic data.
Collapse
Affiliation(s)
- Minna Alenius
- Department of Psychology and Logopedics, University of Helsinki, Helsinki, Finland
- Population Health Unit, Finnish Institute for Health and Welfare, Helsinki, Finland
- *Correspondence: Minna Alenius,
| | - Laura Hokkanen
- Department of Psychology and Logopedics, University of Helsinki, Helsinki, Finland
| | - Sanna Koskinen
- Department of Psychology and Logopedics, University of Helsinki, Helsinki, Finland
| | - Ilona Hallikainen
- Unit of Neurology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Tuomo Hänninen
- Unit of Neurology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
- Neurology of Neuro Center, Kuopio University Hospital, Kuopio, Finland
| | - Mira Karrasch
- Department of Psychology, Abo Akademi University, Turku, Finland
| | - Minna M. Raivio
- Department of General Practice, University of Helsinki, Helsinki, Finland
- Unit of Primary Health Care, Helsinki University Hospital, Helsinki, Finland
| | - Marja-Liisa Laakkonen
- Department of General Practice, University of Helsinki, Helsinki, Finland
- Unit of Primary Health Care, Helsinki University Hospital, Helsinki, Finland
- Geriatric Clinic, Department of Social Services and Health Care, Laakso Hospital, Helsinki, Finland
| | - Johanna Krüger
- Research Unit of Clinical Neuroscience, Neurology, University of Oulu, Oulu, Finland
- MRC, Oulu University Hospital, Oulu, Finland
| | | | - Miia Kivipelto
- Population Health Unit, Finnish Institute for Health and Welfare, Helsinki, Finland
- Division of Clinical Geriatrics, Center for Alzheimer Research, NVS, Karolinska Institutet, Stockholm, Sweden
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
- Ageing Epidemiology Research Unit, School of Public Health, Imperial College London, London, United Kingdom
| | - Tiia Ngandu
- Population Health Unit, Finnish Institute for Health and Welfare, Helsinki, Finland
- Division of Clinical Geriatrics, Center for Alzheimer Research, NVS, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
11
|
Volumetric Assessment of Hippocampus and Subcortical Gray Matter Regions in Alzheimer Disease and Amnestic Mild Cognitive Impairment. Cogn Behav Neurol 2022; 35:95-103. [PMID: 35639010 DOI: 10.1097/wnn.0000000000000296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 07/23/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Quantitative MRI assessment methods have limited utility due to a lack of standardized methods and measures for Alzheimer disease (AD) and amnestic mild cognitive impairment (aMCI). OBJECTIVE To employ a relatively new and easy-to-use quantitative assessment method to reveal volumetric changes in subcortical gray matter (GM) regions, hippocampus, and global intracranial structures as well as the diagnostic performance and best thresholds of total hippocampal volumetry in individuals with AD and those with aMCI. METHOD A total of 74 individuals-37 with mild to moderate AD, 19 with aMCI, and 18 with normal cognition (NC)-underwent a 3T MRI. Fully automated segmentation and volumetric measurements were performed. RESULTS The AD and aMCI groups had smaller volumes of amygdala, nucleus accumbens, and hippocampus compared with the NC group. These same two groups had significantly smaller total white matter volume than the NC group. The AD group had smaller total GM volume compared with the aMCI and NC groups. The thalamus in the AD group showed a subtle atrophy. There were no significant volumetric differences in the caudate nucleus, putamen, or globus pallidus between the groups. CONCLUSION The amygdala and nucleus accumbens showed atrophy comparable to the hippocampal atrophy in both the AD and aMCI groups, which may contribute to cognitive impairment. Hippocampal volumetry is a reliable tool for differentiating between AD and NC groups but has substantially less power in differentiating between AD and aMCI groups. The loss of total GM volume differentiates AD from aMCI and NC.
Collapse
|
12
|
Migliaccio R, Cacciamani F. The temporal lobe in typical and atypical Alzheimer disease. HANDBOOK OF CLINICAL NEUROLOGY 2022; 187:449-466. [PMID: 35964987 DOI: 10.1016/b978-0-12-823493-8.00004-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Alzheimer disease (AD) is defined neuropathologically by abnormal extra-cellular β-amyloid plaques combined with intraneuronal tau aggregation. Patients sharing the same neuropathological features but presenting different clinical manifestations and evolutions have led to the notion of AD spectrum. This spectrum encompasses typical and atypical forms of AD. For all of them, specific parts of the temporal lobes, as well as their structural and functional connections with other brain regions, are affected. In typical amnestic late-onset Alzheimer's disease (>65 years old; LOAD), tau pathology gradually spreads to the brain from the medial temporal lobe (MTL). MTL is an inhomogeneous structure consisting of several subregions densely connected to each other and to other cortical and subcortical brain regions. These regions play a crucial role in the storage of information in episodic memory. In less common early-onset AD (<65 years old; EOAD), a large proportion of patients presents atypical clinical manifestations, in which memory impairment is not inaugural and predominant. Instead, these patients have predominant and/or isolated deficits in language, visuospatial, motor, or executive/behavioral functions. In atypical variants, brain damage is mainly centered on the posterior regions, with relative sparing of the MTL. However, the temporal lobe also appears to be variably and specifically damaged in some subtypes of EOAD. For example, the left superior temporal gyrus is the core of brain damage in the language variant, as well as the ventral regions of the temporal lobe play an important role in the clinic of the visual variant.
Collapse
Affiliation(s)
- Raffaella Migliaccio
- Paris Brain Institute, INSERM U1127, Hôpital de la Pitié-Salpêtrière, Paris, France; Department of Neurology, Institut de la mémoire et de la maladie d'Alzheimer, Hôpital de la Pitié-Salpêtrière, Paris, France.
| | | |
Collapse
|
13
|
Bandyopadhyay S. Role of Neuron and Glia in Alzheimer's Disease and Associated Vascular Dysfunction. Front Aging Neurosci 2021; 13:653334. [PMID: 34211387 PMCID: PMC8239194 DOI: 10.3389/fnagi.2021.653334] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 05/05/2021] [Indexed: 12/14/2022] Open
Abstract
Amyloidogenicity and vascular dysfunction are the key players in the pathogenesis of Alzheimer’s disease (AD), involving dysregulated cellular interactions. An intricate balance between neurons, astrocytes, microglia, oligodendrocytes and vascular cells sustains the normal neuronal circuits. Conversely, cerebrovascular diseases overlap neuropathologically with AD, and glial dyshomeostasis promotes AD-associated neurodegenerative cascade. While pathological hallmarks of AD primarily include amyloid-β (Aβ) plaques and neurofibrillary tangles, microvascular disorders, altered cerebral blood flow (CBF), and blood-brain barrier (BBB) permeability induce neuronal loss and synaptic atrophy. Accordingly, microglia-mediated inflammation and astrogliosis disrupt the homeostasis of the neuro-vascular unit and stimulate infiltration of circulating leukocytes into the brain. Large-scale genetic and epidemiological studies demonstrate a critical role of cellular crosstalk for altered immune response, metabolism, and vasculature in AD. The glia associated genetic risk factors include APOE, TREM2, CD33, PGRN, CR1, and NLRP3, which correlate with the deposition and altered phagocytosis of Aβ. Moreover, aging-dependent downregulation of astrocyte and microglial Aβ-degrading enzymes limits the neurotrophic and neurogenic role of glial cells and inhibits lysosomal degradation and clearance of Aβ. Microglial cells secrete IGF-1, and neurons show a reduced responsiveness to the neurotrophic IGF-1R/IRS-2/PI3K signaling pathway, generating amyloidogenic and vascular dyshomeostasis in AD. Glial signals connect to neural stem cells, and a shift in glial phenotype over the AD trajectory even affects adult neurogenesis and the neurovascular niche. Overall, the current review informs about the interaction of neuronal and glial cell types in AD pathogenesis and its critical association with cerebrovascular dysfunction.
Collapse
Affiliation(s)
- Sanghamitra Bandyopadhyay
- Developmental Toxicology Laboratory, Systems Toxicology & Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Lucknow, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
14
|
Chen JF, Liu K, Hu B, Li RR, Xin W, Chen H, Wang F, Chen L, Li RX, Ren SY, Xiao L, Chan JR, Mei F. Enhancing myelin renewal reverses cognitive dysfunction in a murine model of Alzheimer's disease. Neuron 2021; 109:2292-2307.e5. [PMID: 34102111 DOI: 10.1016/j.neuron.2021.05.012] [Citation(s) in RCA: 210] [Impact Index Per Article: 52.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 04/15/2021] [Accepted: 05/10/2021] [Indexed: 12/13/2022]
Abstract
Severe cognitive decline is a hallmark of Alzheimer's disease (AD). In addition to gray matter loss, significant white matter pathology has been identified in AD patients. Here, we characterized the dynamics of myelin generation and loss in the APP/PS1 mouse model of AD. Unexpectedly, we observed a dramatic increase in the rate of new myelin formation in APP/PS1 mice, reminiscent of the robust oligodendroglial response to demyelination. Despite this increase, overall levels of myelination are decreased in the cortex and hippocampus of APP/PS1 mice and postmortem AD tissue. Genetically or pharmacologically enhancing myelin renewal, by oligodendroglial deletion of the muscarinic M1 receptor or systemic administration of the pro-myelinating drug clemastine, improved the performance of APP/PS1 mice in memory-related tasks and increased hippocampal sharp wave ripples. Taken together, these results demonstrate the potential of enhancing myelination as a therapeutic strategy to alleviate AD-related cognitive impairment.
Collapse
Affiliation(s)
- Jing-Fei Chen
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University, Chongqing 400038, China
| | - Kun Liu
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University, Chongqing 400038, China
| | - Bo Hu
- Department of Physiology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing 400038, China
| | - Rong-Rong Li
- Department of Physiology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing 400038, China
| | - Wendy Xin
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Hao Chen
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, China
| | - Fei Wang
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University, Chongqing 400038, China
| | - Lin Chen
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University, Chongqing 400038, China
| | - Rui-Xue Li
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University, Chongqing 400038, China
| | - Shu-Yu Ren
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University, Chongqing 400038, China
| | - Lan Xiao
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University, Chongqing 400038, China.
| | - Jonah R Chan
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA.
| | - Feng Mei
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University, Chongqing 400038, China.
| |
Collapse
|
15
|
Shea YF, Pan Y, Mak HKF, Bao Y, Lee SC, Chiu PKC, Chan HWF. A systematic review of atypical Alzheimer's disease including behavioural and psychological symptoms. Psychogeriatrics 2021; 21:396-406. [PMID: 33594793 DOI: 10.1111/psyg.12665] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/06/2021] [Accepted: 01/25/2021] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD) is the commonest cause of dementia, characterized by the clinical presentation of progressive anterograde episodic memory impairment. However, atypical presentation of patients is increasingly recognized. These atypical AD include logopenic aphasia, behavioural variant AD, posterior cortical atrophy, and corticobasal syndrome. These atypical AD are more common in patients with young onset AD before the age of 65 years old. Since medical needs (including the behavioural and psychological symptoms of dementia) of atypical AD patients could be different from typical AD patients, it is important for clinicians to be aware of these atypical forms of AD. In addition, disease modifying treatment may be available in the future. This review aims at providing an update on various important subtypes of atypical AD including behavioural and psychological symptoms.
Collapse
Affiliation(s)
- Yat-Fung Shea
- Department of Medicine, LKS Faculty of Medicine, University of Hong Kong, Queen Mary Hospital, Pok Fu Lam, Hong Kong
| | - Yining Pan
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Henry Ka-Fung Mak
- Department of Diagnostic Radiology, LKS Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Yiwen Bao
- Department of Diagnostic Radiology, LKS Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Shui-Ching Lee
- Department of Medicine, LKS Faculty of Medicine, University of Hong Kong, Queen Mary Hospital, Pok Fu Lam, Hong Kong
| | - Patrick Ka-Chun Chiu
- Department of Medicine, LKS Faculty of Medicine, University of Hong Kong, Queen Mary Hospital, Pok Fu Lam, Hong Kong
| | - Hon-Wai Felix Chan
- Department of Medicine, LKS Faculty of Medicine, University of Hong Kong, Queen Mary Hospital, Pok Fu Lam, Hong Kong
| |
Collapse
|
16
|
Overman MJ, Zamboni G, Butler C, Ahmed S. Splenial white matter integrity is associated with memory impairments in posterior cortical atrophy. Brain Commun 2021; 3:fcab060. [PMID: 34007964 PMCID: PMC8112963 DOI: 10.1093/braincomms/fcab060] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 12/09/2020] [Accepted: 02/23/2021] [Indexed: 11/22/2022] Open
Abstract
Posterior cortical atrophy is an atypical form of Alzheimer’s disease characterized by visuospatial impairments and predominant tissue loss in the posterior parieto-occipital and temporo-occipital cortex. Whilst episodic memory is traditionally thought to be relatively preserved in posterior cortical atrophy, recent work indicates that memory impairments form a common clinical symptom in the early stages of the disease. Neuroimaging studies suggest that memory dysfunction in posterior cortical atrophy may originate from atrophy and functional hypoconnectivity of parietal cortex. The structural connectivity patterns underpinning these memory impairments, however, have not been investigated. This line of inquiry is of particular interest, as changes in white matter tracts of posterior cortical atrophy patients have been shown to be more extensive than expected based on posterior atrophy of grey matter. In this cross-sectional diffusion tensor imaging MRI study, we examine the relationship between white matter microstructure and verbal episodic memory in posterior cortical atrophy. We assessed episodic memory performance in a group of posterior cortical atrophy patients (n = 14) and a group of matched healthy control participants (n = 19) using the Free and Cued Selective Reminding Test with Immediate Recall. Diffusion tensor imaging measures were obtained for 13 of the posterior cortical atrophy patients and a second control group of 18 healthy adults. Patients and healthy controls demonstrated similar memory encoding performance, indicating that learning of verbal information was preserved in posterior cortical atrophy. However, retrieval of verbal items was significantly impaired in the patient group compared with control participants. As expected, tract-based spatial statistics analyses showed widespread reductions of white matter integrity in posterior cortical regions of patients compared with healthy adults. Correlation analyses indicated that poor verbal retrieval in the patient group was specifically associated with microstructural damage of the splenium of the corpus callosum. Post-hoc tractography analyses in healthy controls demonstrated that this splenial region was connected to thalamic radiations and the retrolenticular part of the internal capsule. These results provide insight into the brain circuits that underlie memory impairments in posterior cortical atrophy. From a cognitive perspective, we propose that the association between splenial integrity and memory dysfunction could arise indirectly via disruption of attentional processes. We discuss implications for the clinical phenotype and development of therapeutic aids for cognitive impairment in posterior cortical atrophy.
Collapse
Affiliation(s)
- Margot Juliëtte Overman
- Research Institute for the Care of Older People (RICE), Bath BA1 3NG, UK.,MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge CB2 7EF, UK
| | - Giovanna Zamboni
- Dipartimento di Scienze Biomediche, Metaboliche e Neuroscienze, Università di Modena e Reggio Emilia, Modena, Italy.,Center for Neuroscience and Neurotechnology, Università di Modena e Reggio Emilia, Modena, Italy.,Nuffield Department of Clinical Neuroscience, University of Oxford, Oxfordshire OX3 9DU, UK
| | - Christopher Butler
- Nuffield Department of Clinical Neuroscience, University of Oxford, Oxfordshire OX3 9DU, UK.,Department of Brain Sciences, Imperial College London, London SW7 2AZ, UK.,Departamento de Neurología, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Samrah Ahmed
- Nuffield Department of Clinical Neuroscience, University of Oxford, Oxfordshire OX3 9DU, UK.,School of Psychology and Clinical Language Sciences, University of Reading, Reading RG6 6ES, UK
| |
Collapse
|
17
|
Benedet AL, Leuzy A, Pascoal TA, Ashton NJ, Mathotaarachchi S, Savard M, Therriault J, Kang MS, Chamoun M, Schöll M, Zimmer ER, Gauthier S, Labbe A, Zetterberg H, Rosa-Neto P, Blennow K. Stage-specific links between plasma neurofilament light and imaging biomarkers of Alzheimer's disease. Brain 2021; 143:3793-3804. [PMID: 33210117 DOI: 10.1093/brain/awaa342] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/28/2020] [Accepted: 08/17/2020] [Indexed: 11/12/2022] Open
Abstract
Neurofilament light (NfL) is a marker of neuroaxonal injury, a prominent feature of Alzheimer's disease. It remains uncertain, however, how it relates to amyloid and tau pathology or neurodegeneration across the Alzheimer's disease continuum. The aim of this study was to investigate how plasma NfL relates to amyloid and tau PET and MRI measures of brain atrophy in participants with and without cognitive impairment. We retrospectively examined the association between plasma NfL and MRI measures of grey/white matter volumes in the Alzheimer's Disease Neuroimaging Initiative [ADNI: n = 1149; 382 cognitively unimpaired control subjects and 767 cognitively impaired participants (mild cognitive impairment n = 420, Alzheimer's disease dementia n = 347)]. Longitudinal plasma NfL was measured using single molecule array (Simoa) technology. Cross-sectional associations between plasma NfL and PET amyloid and tau measures were independently assessed in two cohorts: ADNI [n = 198; 110 cognitively unimpaired, 88 cognitively impaired (MCI n = 67, Alzheimer's disease dementia n = 21), data accessed October 2018]; and Translational Biomarkers in Aging and Dementia [TRIAD, n = 116; 74 cognitively unimpaired, 42 cognitively impaired (MCI n = 16, Alzheimer's disease dementia n = 26), data obtained November 2017 to January 2019]. Associations between plasma NfL and imaging-derived measures were examined voxel-wise using linear regression (cross-sectional) and linear mixed effect models (longitudinal). Cross-sectional analyses in both cohorts showed that plasma NfL was associated with PET findings in brain regions typically affected by Alzheimer's disease; associations were specific to amyloid PET in cognitively unimpaired and tau PET in cognitively impaired (P < 0.05). Longitudinal analyses showed that NfL levels were associated with grey/white matter volume loss; grey matter atrophy in cognitively unimpaired was specific to APOE ε4 carriers (P < 0.05). These findings suggest that plasma NfL increases in response to amyloid-related neuronal injury in preclinical stages of Alzheimer's disease, but is related to tau-mediated neurodegeneration in symptomatic patients. As such, plasma NfL may a useful measure to monitor effects in disease-modifying drug trials.
Collapse
Affiliation(s)
- Andréa L Benedet
- Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada.,CAPES Foundation, Ministry of Education of Brazil, Brasília, Brazil
| | - Antoine Leuzy
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - Tharick A Pascoal
- Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.,King's College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, UK.,NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK
| | - Sulantha Mathotaarachchi
- Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada
| | - Melissa Savard
- Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada
| | - Min Su Kang
- Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada
| | - Mira Chamoun
- Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada
| | - Michael Schöll
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | - Eduardo R Zimmer
- Alzheimer's Disease Research Unit, The McGill University Research Centre for Studies in Aging, Montreal, McGill University, Montreal, QC, Canada.,Departament of Pharmacology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Serge Gauthier
- Alzheimer's Disease Research Unit, The McGill University Research Centre for Studies in Aging, Montreal, McGill University, Montreal, QC, Canada
| | - Aurélie Labbe
- Department of Decision Sciences, HEC Montreal, Montreal, QC, Canada
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,UK Dementia Research Institute at UCL, London, UK
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill Centre for Studies in Aging, McGill University, Montreal, QC, Canada.,Alzheimer's Disease Research Unit, The McGill University Research Centre for Studies in Aging, Montreal, McGill University, Montreal, QC, Canada.,Montreal Neurological Institute, Montreal, QC, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | | |
Collapse
|
18
|
Ohm DT, Fought AJ, Martersteck A, Coventry C, Sridhar J, Gefen T, Weintraub S, Bigio E, Mesulam M, Rogalski E, Geula C. Accumulation of neurofibrillary tangles and activated microglia is associated with lower neuron densities in the aphasic variant of Alzheimer's disease. Brain Pathol 2021; 31:189-204. [PMID: 33010092 PMCID: PMC7855834 DOI: 10.1111/bpa.12902] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/27/2020] [Accepted: 09/28/2020] [Indexed: 12/15/2022] Open
Abstract
The neurofibrillary tangles (NFT) and amyloid-ß plaques (AP) that comprise Alzheimer's disease (AD) neuropathology are associated with neurodegeneration and microglial activation. Activated microglia exist on a dynamic spectrum of morphologic subtypes that include resting, surveillant microglia capable of converting to activated, hypertrophic microglia closely linked to neuroinflammatory processes and AD neuropathology in amnestic AD. However, quantitative analyses of microglial subtypes and neurons are lacking in non-amnestic clinical AD variants, including primary progressive aphasia (PPA-AD). PPA-AD is a language disorder characterized by cortical atrophy and NFT densities concentrated to the language-dominant hemisphere. Here, a stereologic investigation of five PPA-AD participants determined the densities and distributions of neurons and microglial subtypes to examine how cellular changes relate to AD neuropathology and may contribute to cortical atrophy. Adjacent series of sections were immunostained for neurons (NeuN) and microglia (HLA-DR) from bilateral language and non-language regions where in vivo cortical atrophy and Thioflavin-S-positive APs and NFTs were previously quantified. NeuN-positive neurons and morphologic subtypes of HLA-DR-positive microglia (i.e., resting [ramified] microglia and activated [hypertrophic] microglia) were quantified using unbiased stereology. Relationships between neurons, microglia, AD neuropathology, and cortical atrophy were determined using linear mixed models. NFT densities were positively associated with hypertrophic microglia densities (P < 0.01) and inversely related to neuron densities (P = 0.01). Hypertrophic microglia densities were inversely related to densities of neurons (P < 0.01) and ramified microglia (P < 0.01). Ramified microglia densities were positively associated with neuron densities (P = 0.02) and inversely related to cortical atrophy (P = 0.03). Our findings provide converging evidence of divergent roles for microglial subtypes in patterns of neurodegeneration, which includes hypertrophic microglia likely driving a neuroinflammatory response more sensitive to NFTs than APs in PPA-AD. Moreover, the accumulation of both NFTs and activated hypertrophic microglia in association with low neuron densities suggest they may collectively contribute to focal neurodegeneration characteristic of PPA-AD.
Collapse
Affiliation(s)
- Daniel T. Ohm
- Mesulam Center for Cognitive Neurology and Alzheimer’s DiseaseNorthwestern University Feinberg School of MedicineChicagoIL
| | - Angela J. Fought
- Department of Preventive MedicineNorthwestern University Feinberg School of MedicineChicagoIL
| | - Adam Martersteck
- Mesulam Center for Cognitive Neurology and Alzheimer’s DiseaseNorthwestern University Feinberg School of MedicineChicagoIL
| | - Christina Coventry
- Mesulam Center for Cognitive Neurology and Alzheimer’s DiseaseNorthwestern University Feinberg School of MedicineChicagoIL
| | - Jaiashre Sridhar
- Mesulam Center for Cognitive Neurology and Alzheimer’s DiseaseNorthwestern University Feinberg School of MedicineChicagoIL
| | - Tamar Gefen
- Mesulam Center for Cognitive Neurology and Alzheimer’s DiseaseNorthwestern University Feinberg School of MedicineChicagoIL
- Department of Psychiatry and Behavioral SciencesNorthwestern University Feinberg School of MedicineChicagoIL
| | - Sandra Weintraub
- Mesulam Center for Cognitive Neurology and Alzheimer’s DiseaseNorthwestern University Feinberg School of MedicineChicagoIL
- Department of Psychiatry and Behavioral SciencesNorthwestern University Feinberg School of MedicineChicagoIL
| | - Eileen Bigio
- Mesulam Center for Cognitive Neurology and Alzheimer’s DiseaseNorthwestern University Feinberg School of MedicineChicagoIL
- Department of PathologyNorthwestern University Feinberg School of MedicineChicagoIL
| | - M.‐Marsel Mesulam
- Mesulam Center for Cognitive Neurology and Alzheimer’s DiseaseNorthwestern University Feinberg School of MedicineChicagoIL
- Department of NeurologyNorthwestern University Feinberg School of MedicineChicagoIL
| | - Emily Rogalski
- Mesulam Center for Cognitive Neurology and Alzheimer’s DiseaseNorthwestern University Feinberg School of MedicineChicagoIL
- Department of Psychiatry and Behavioral SciencesNorthwestern University Feinberg School of MedicineChicagoIL
| | - Changiz Geula
- Mesulam Center for Cognitive Neurology and Alzheimer’s DiseaseNorthwestern University Feinberg School of MedicineChicagoIL
| |
Collapse
|
19
|
Abbate C, Trimarchi PD, Inglese S, Damanti S, Dolci GAM, Ciccone S, Rossi PD, Mari D, Arosio B, Bagarolo R, Giunco F, Cesari M. Does the Right Focal Variant of Alzheimer's Disease Really Exist? A Literature Analysis. J Alzheimers Dis 2020; 71:405-420. [PMID: 31381515 DOI: 10.3233/jad-190338] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is a clinically heterogeneous disease. Multiple atypical syndromes, distinct from the usual amnesic phenotype, have been described. In this context, the existence of a right variant of AD (RAD), characterized by enduring visuospatial impairment associated with right-sided asymmetric brain damage, has been proposed. However, to date, this phenotype remains controversial. In particular, its peculiar characteristics and the independence from more prevalent cases (especially the posterior cortical atrophy syndrome) have to be demonstrated. OBJECTIVE To explore the existence of focal RAD on the basis of existing literature. METHODS We performed a literature search for the description of atypical AD presentations, potentially evoking cases of focal RAD. To be considered as affected by RAD, the described cases had to present: 1) well documented right-sided asymmetry at neuroimaging; 2) predominant cognitive deficits localizable on the right hemisphere; 3) no specific diagnosis of a known variant of AD. RESULTS Twenty-one cases were found in the literature, but some of them were subsequently excluded because some features of a different clinical syndrome were overlapped with the clinical features of RAD. Thirteen positive cases, three of them with pathologically confirmed AD, remained. A common right clinical-radiological syndrome, characterized by memory and visuospatial impairment with temporal and parietal involvement, consistently emerged. However, the heterogeneity among the reports prevented a definitive and univocal description of the syndrome. CONCLUSION Even if sporadic observations strongly support the existence of a focal RAD, no definitive conclusions can still be drawn about it as an independent condition.
Collapse
Affiliation(s)
- Carlo Abbate
- Geriatric Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Silvia Inglese
- Geriatric Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Sarah Damanti
- Geriatric Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | | | - Simona Ciccone
- Geriatric Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Paolo D Rossi
- Geriatric Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Daniela Mari
- Geriatric Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Beatrice Arosio
- Geriatric Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | | | | | - Matteo Cesari
- Geriatric Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| |
Collapse
|
20
|
Neurovascular unit dysregulation, white matter disease, and executive dysfunction: the shared triad of vascular cognitive impairment and Alzheimer disease. GeroScience 2020; 42:445-465. [PMID: 32002785 DOI: 10.1007/s11357-020-00164-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 01/22/2020] [Indexed: 01/07/2023] Open
Abstract
Executive dysfunction is the most important predictor for loss of independence in dementia. As executive function involves the coordination of distributed cerebral functions, executive function requires healthy white matter. However, white matter is highly vulnerable to cerebrovascular insults, with executive dysfunction being a core feature of vascular cognitive impairment (VCI). At the same time, cerebrovascular pathology, white matter disease, and executive dysfunction are all increasingly recognized as features of Alzheimer disease (AD). Recent studies have characterized the crucial role of glial cells in the pathological changes observed in both VCI and AD. In comorbid VCI and AD, the glial cells of the neurovascular unit (NVU) emerge as important therapeutic targets for the preservation of white matter integrity and executive function. Our synthesis from current research identifies dysregulation of the NVU, white matter disease, and executive dysfunction as a fundamental triad that is common to both VCI and AD. Further study of this triad will be critical for advancing the prevention and management of dementia.
Collapse
|
21
|
Migliaccio R, Agosta F, Basaia S, Cividini C, Habert MO, Kas A, Montembeault M, Filippi M. Functional brain connectome in posterior cortical atrophy. Neuroimage Clin 2019; 25:102100. [PMID: 31865020 PMCID: PMC6931188 DOI: 10.1016/j.nicl.2019.102100] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 10/17/2019] [Accepted: 11/18/2019] [Indexed: 01/29/2023]
Abstract
This study investigated the functional brain connectome architecture in patients with Posterior Cortical Atrophy (PCA). Eighteen PCA patients and 29 age- and sex- matched healthy controls were consecutively recruited in a specialized referral center. Participants underwent neurologic examination, cerebrospinal fluid (CSF) examination for Alzheimer's disease (AD) biomarkers, cognitive assessment, and brain MRI. For a smaller subset of participants, FDG-PET examination was available. We assessed topological brain network properties and regional functional connectivity as well as intra- and inter-hemispheric connectivity, using graph analysis and connectomics. Supplementary analyses were performed to explore the association between the CSF AD profile and the connectome status, and taking into account hypometabolic, atrophic, and spared regions (nodes). PCA patients showed diffuse functional connectome alterations at both global and regional level, as well as a connectivity breakdown between the posterior brain nodes. They had a widespread loss of both intra- and inter-hemispheric connections, exceeding the structural damage, and including the frontal connections. In PCA, connectome alterations were identified in all the brain nodes irrespectively of their structural and metabolic classification and were associated with a connectivity breakdown between damaged and spared areas. Taken together, these findings suggest the potentially high sensitivity of graph-analysis and connectomic in capturing the progression and maybe early signs of neurodegeneration in PCA patients.
Collapse
Affiliation(s)
- Raffaella Migliaccio
- FrontLab, INSERM U 1127, CNRS UMR 7225, Sorbonne Universités, and Université Pierre et Marie Curie-Paris 6, UMR S1127, Institut du Cerveau et de la Moelle épinière (ICM), Pitié-Salpêtrière hospital, Paris, France; Institut de la mémoire et de la maladie d'Alzheimer, IM2A, Reference Centre for Rare dementias and Early Onset Alzheimer's disease, Neurology Departement, Pitié-Salpêtrière hospital, Paris, France.
| | - Federica Agosta
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Silvia Basaia
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Camilla Cividini
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Marie-Odile Habert
- Department of Nuclear Medicine, Pitié-Salpêtrière hospital, Paris, France; LIB, Inserm U1146, Université Pierre et Marie Curie, Paris 6, Paris, France
| | - Aurélie Kas
- Department of Nuclear Medicine, Pitié-Salpêtrière hospital, Paris, France; LIB, Inserm U1146, Université Pierre et Marie Curie, Paris 6, Paris, France
| | - Maxime Montembeault
- Memory & Aging Center, Deparment of Neurology, University of California in San Francisco, San Francisco, United-States
| | - Massimo Filippi
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy; Department of Neurology, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
22
|
Guerrier L, Cransac C, Pages B, Saint-Aubert L, Payoux P, Péran P, Pariente J. Posterior Cortical Atrophy: Does Complaint Match the Impairment? A Neuropsychological and FDG-PET Study. Front Neurol 2019; 10:1010. [PMID: 31616363 PMCID: PMC6764288 DOI: 10.3389/fneur.2019.01010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 09/04/2019] [Indexed: 12/12/2022] Open
Abstract
Objective: Posterior Cortical Atrophy (PCA) is a neurodegenerative disease characterized predominantly by visual impairment. However, diagnosis of PCA remains complicated with an interval of several years between initial reporting of symptoms and diagnosis. The aim of the present study is to define if patients' visual and gestural complaints are consistent with their clinical profile. Method: An evaluation of daily visual problems as well as a full neuropsychological assessment and FDG-PET were performed in 15 PCA patients. We compared glucose metabolism between these PCA patients and 18 healthy controls. Correlation analyses were conducted in PCA patients between visual and gestural complaint, clinical impairments, and brain glucose metabolism. Results: Major impairment of cognitive functions was detected in PCA patients specifically in visual domains. Positive correlations were found between visual impairments and hypometabolism in the right temporo-parieto-occipital cortices. However, no correlation was found between complaint and visual impairment in PCA patients. Discussion: Our main results suggest a consistent relationship between clinical impairment and brain metabolism. However, the patient's complaint and visual performance are not linked. Combining the literature and our results, it seems that patients are generally aware of difficulties but misinterpret them. This misinterpretation may be responsible for the delayed diagnosis.
Collapse
Affiliation(s)
- Laura Guerrier
- ToNIC, Toulouse NeuroImaging Centre, University of Toulouse, Inserm, UPS, Toulouse, France
| | - Camille Cransac
- Department of Neurology, University Hospital of Toulouse, Toulouse, France
| | - Bérengère Pages
- Department of Neurology, University Hospital of Toulouse, Toulouse, France
| | - Laure Saint-Aubert
- ToNIC, Toulouse NeuroImaging Centre, University of Toulouse, Inserm, UPS, Toulouse, France.,Department of Nuclear Medicine, University Hospital of Toulouse, Toulouse, France
| | - Pierre Payoux
- ToNIC, Toulouse NeuroImaging Centre, University of Toulouse, Inserm, UPS, Toulouse, France.,Department of Nuclear Medicine, University Hospital of Toulouse, Toulouse, France
| | - Patrice Péran
- ToNIC, Toulouse NeuroImaging Centre, University of Toulouse, Inserm, UPS, Toulouse, France
| | - Jérémie Pariente
- ToNIC, Toulouse NeuroImaging Centre, University of Toulouse, Inserm, UPS, Toulouse, France.,Department of Neurology, University Hospital of Toulouse, Toulouse, France
| |
Collapse
|
23
|
Chen Y, Liu P, Wang Y, Peng G. Neural Mechanisms of Visual Dysfunction in Posterior Cortical Atrophy. Front Neurol 2019; 10:670. [PMID: 31293507 PMCID: PMC6603128 DOI: 10.3389/fneur.2019.00670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 06/07/2019] [Indexed: 11/13/2022] Open
Abstract
Posterior cortical atrophy (PCA) is characterized predominantly by visual dysfunction that arises from bilateral impairments in occipital, parietal, and temporal regions of the brain. PCA is clinically identified based primarily on visual symptoms and neuroimaging findings. Region-specific gray and white matter deficits have been discussed in detail, and are associated with clinical manifestations that present with similar patterns of perfusion and metabolic findings. Here, we discuss both structural and functional changes in the ventral and dorsal visual streams along with their underlying relationships. We also discuss the most recent developments in neuroimaging characteristics and summarize correlations between distinct neuroimaging presentations.
Collapse
Affiliation(s)
- Yi Chen
- Department of Neurology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ping Liu
- Department of Neurology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yunyun Wang
- Department of Neurology, Shengzhou People's Hospital, Shengzhou, China
| | - Guoping Peng
- Department of Neurology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Guoping Peng
| |
Collapse
|
24
|
Roby DA, Ruiz F, Kermath BA, Voorhees JR, Niehoff M, Zhang J, Morley JE, Musiek ES, Farr SA, Burris TP. Pharmacological activation of the nuclear receptor REV-ERB reverses cognitive deficits and reduces amyloid-β burden in a mouse model of Alzheimer's disease. PLoS One 2019; 14:e0215004. [PMID: 30973894 PMCID: PMC6459530 DOI: 10.1371/journal.pone.0215004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 03/26/2019] [Indexed: 12/27/2022] Open
Abstract
Alzheimer's disease currently lacks treatment options that effectively reverse the biological/anatomical pathology and cognitive deficits associated with the disease. Loss of function of the nuclear receptor REV-ERB is associated with reduced cognitive function in mouse models. The effect of enhanced REV-ERB activity on cognitive function has not been examined. In this study, we tested the hypothesis that enhanced REV-ERB function may enhance cognitive function in a model of Alzheimer's disease. We utilized the REV-ERB agonist SR9009 to pharmacologically activate the activity of REV-ERB in the SAMP8 mouse model of Alzheimer's disease. SR9009 reversed cognitive dysfunction of an aged SAMP8 mouse in several behavioral assays including novel object recognition, T-maze foot shock avoidance, and lever press operant conditioning task assessments. SR9009 treatment reduced amyloid-β 1-40 and 1-42 levels in the cortex, which is consistent with improved cognitive function. Furthermore, SR9009 treatment led to increased hippocampal PSD-95, cortical synaptophysin expression and the number of synapses suggesting improvement in synaptic function. We conclude that REV-ERB is a potential target for treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Deborah A. Roby
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO, United States of America
| | - Fernanda Ruiz
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO, United States of America
| | - Bailey A. Kermath
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO, United States of America
| | - Jaymie R. Voorhees
- Center for Clinical Pharmacology, Washington University School of Medicine and St. Louis College of Pharmacy, St. Louis, MO, United States of America
| | - Michael Niehoff
- Department of Internal Medicine, Saint Louis University School of Medicine, St. Louis, MO, United States of America
| | - Jinsong Zhang
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO, United States of America
| | - John E. Morley
- Department of Internal Medicine, Saint Louis University School of Medicine, St. Louis, MO, United States of America
| | - Erik S. Musiek
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Susan A. Farr
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO, United States of America
- Department of Internal Medicine, Saint Louis University School of Medicine, St. Louis, MO, United States of America
| | - Thomas P. Burris
- Center for Clinical Pharmacology, Washington University School of Medicine and St. Louis College of Pharmacy, St. Louis, MO, United States of America
| |
Collapse
|
25
|
Sintini I, Schwarz CG, Martin PR, Graff-Radford J, Machulda MM, Senjem ML, Reid RI, Spychalla AJ, Drubach DA, Lowe VJ, Jack CR, Josephs KA, Whitwell JL. Regional multimodal relationships between tau, hypometabolism, atrophy, and fractional anisotropy in atypical Alzheimer's disease. Hum Brain Mapp 2018; 40:1618-1631. [PMID: 30549156 DOI: 10.1002/hbm.24473] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 10/20/2018] [Accepted: 10/25/2018] [Indexed: 11/10/2022] Open
Abstract
Alzheimer's disease (AD) can present with atypical clinical forms where the prominent domain of deficit is not memory, that is, atypical AD. Atypical AD patients show cortical atrophy on MRI, hypometabolism on [18 F]fluorodeoxyglucose (FDG) PET, tau uptake on [18 F]AV-1451 PET, and white matter tract degeneration on diffusion tensor imaging (DTI). How these disease processes relate to each other locally and distantly remains unclear. We aimed to examine multimodal neuroimaging relationships in individuals with atypical AD, using univariate and multivariate techniques at region- and voxel-level. Forty atypical AD patients underwent MRI, FDG-PET, tau-PET, beta-amyloid PET, and DTI. Patients were all beta-amyloid positive. Partial Pearson's correlations were performed between tau and FDG standardized uptake value ratios, gray matter MRI-volumes and white matter tract fractional anisotropy. Sparse canonical correlation analysis was applied to identify multivariate relationships between the same quantities. Voxel-level associations across modalities were also assessed. Tau showed strong local negative correlations with FDG metabolism in the occipital and frontal lobes. Tau in frontal and parietal regions was negatively associated with temporoparietal gray matter MRI-volume. Fractional anisotropy in a set of posterior white matter tracts, including the splenium of the corpus callosum, cingulum, and posterior thalamic radiation, was negatively correlated with parietal and occipital tau, atrophy and, predominantly, with hypometabolism. These results support the view that tau is the driving force behind neurodegeneration in atypical AD, and that a breakdown in structural connectivity is related to cortical neurodegeneration, particularly hypometabolism.
Collapse
Affiliation(s)
- Irene Sintini
- Department of Radiology, Mayo Clinic, Rochester, Minnesota
| | | | - Peter R Martin
- Department of Health Science Research (Biostatistics), Mayo Clinic, Rochester, Minnesota
| | | | - Mary M Machulda
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota
| | - Matthew L Senjem
- Department of Radiology, Mayo Clinic, Rochester, Minnesota.,Department of Information Technology, Mayo Clinic, Rochester, Minnesota
| | - Robert I Reid
- Department of Information Technology, Mayo Clinic, Rochester, Minnesota
| | | | | | - Val J Lowe
- Department of Radiology, Mayo Clinic, Rochester, Minnesota
| | | | | | | |
Collapse
|
26
|
Adamson C, Beare R, Ball G, Walterfang M, Seal M. Callosal thickness profiles for prognosticating conversion from mild cognitive impairment to Alzheimer's disease: A classification approach. Brain Behav 2018; 8:e01142. [PMID: 30565884 PMCID: PMC6305917 DOI: 10.1002/brb3.1142] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 08/31/2018] [Accepted: 09/27/2018] [Indexed: 01/21/2023] Open
Abstract
INTRODUCTION Alzheimer's disease (AD) is the most common form of dementia. Finding biomarkers to prognosticate transition from mild cognitive impairment (MCI) to AD is important to clinical medicine. Promising imaging biomarkers of AD conversion identified so far include atrophy of the cerebral cortex and subcortical gray matter nuclei. METHODS This study introduces thickness and bending angle of the corpus callosum as a putative white matter marker of MCI to AD conversion. The corpus callosum is computationally less demanding to segment automatically compared to more complicated structures and a subject can be processed in a few minutes. We aimed to demonstrate that callosal shape and thickness measures provide a simple, effective, and accurate prognostication tool in ADNI dataset. Using longitudinal datasets, we classified MCI subjects based on conversion to AD assessed via cognitive testing. We evaluated the classification accuracy of callosal shape features in comparison with the existing "gold standard" cortical thickness and subcortical gray matter volume measures. RESULTS The callosal thickness measures were less accurate in classifying conversion status by cognitive scores compared to gray matter measures for AD. CONCLUSIONS While this paper presented a negative result, this method may be more suitable for a disease of the white matter.
Collapse
Affiliation(s)
- Chris Adamson
- Developmental ImagingMurdoch Children’s Research InstituteParkvilleVictoriaAustralia
| | - Richard Beare
- Developmental ImagingMurdoch Children’s Research InstituteParkvilleVictoriaAustralia
- Department of MedicineMonash UniversityMelbourneVictoriaAustralia
| | - Gareth Ball
- Developmental ImagingMurdoch Children’s Research InstituteParkvilleVictoriaAustralia
| | - Mark Walterfang
- Neuropsychiatry UnitRoyal Melbourne HospitalMelbourneVictoriaAustralia
- Department of PsychiatryUniversity of MelbourneMelbourneVictoriaAustralia
- Florey Institute of Neuroscience and Mental HealthMelbourneVictoriaAustralia
| | - Marc Seal
- Developmental ImagingMurdoch Children’s Research InstituteParkvilleVictoriaAustralia
| | | |
Collapse
|
27
|
Montembeault M, Brambati SM, Lamari F, Michon A, Samri D, Epelbaum S, Lacomblez L, Lehéricy S, Habert MO, Dubois B, Kas A, Migliaccio R. Atrophy, metabolism and cognition in the posterior cortical atrophy spectrum based on Alzheimer's disease cerebrospinal fluid biomarkers. Neuroimage Clin 2018; 20:1018-1025. [PMID: 30340200 PMCID: PMC6197495 DOI: 10.1016/j.nicl.2018.10.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Revised: 09/25/2018] [Accepted: 10/08/2018] [Indexed: 12/28/2022]
Abstract
INTRODUCTION In vivo clinical, anatomical and metabolic differences between posterior cortical atrophy (PCA) patients presenting with different Alzheimer's disease (AD) cerebrospinal fluid (CSF) biomarkers profiles are still unknown. METHODS Twenty-seven PCA patients underwent CSF examination and were classified as 1) PCA with a typical CSF AD profile (PCA-tAD; abnormal amyloid and T-tau/P-tau biomarkers, n = 13); 2) PCA with an atypical AD CSF profile (PCA-aAD; abnormal amyloid biomarker only, n = 9); and 3) PCA not associated with AD (PCA-nonAD; normal biomarkers, n = 5). All patients underwent clinical and cognitive assessment, structural MRI, and a subset of them underwent brain 18F-FDG PET. RESULTS All patients' groups showed a common pattern of posterior GM atrophy and hypometabolism typical of PCA, as well as equivalent demographics and clinical/cognitive profiles. PCA-tAD patients showed a group-specific pattern of hypometabolism in the left fusiform gyrus and inferior temporal gyrus. PCA-aAD did not present a group-specific atrophy pattern. Finally, group-specific gray matter atrophy in the right dorsolateral prefrontal cortex, left caudate nucleus and right medial temporal regions and hypometabolism in the right supplementary motor area and paracentral lobule were observed in PCA-nonAD patients. CONCLUSION Our findings suggest that both PCA-tAD and PCA-aAD patients are on the AD continuum, in agreement with the recently suggested A/T/N model. Furthermore, in PCA, the underlying pathology has an impact at least on the anatomo-functional presentation. Brain damage observed in PCA-tAD and PCA-aAD was mostly consistent with the well-described presentation of the disease, although it was more widespread in PCA-tAD group, especially in the left temporal lobe. Additional fronto-temporal (especially dorsolateral prefrontal) damage seems to be a clue to underlying non-AD pathology in PCA, which warrants the need for longitudinal follow-ups to investigate frontal symptoms in these patients.
Collapse
Affiliation(s)
- Maxime Montembeault
- FrontLab, Institut du Cerveau et de la Moelle épinière (ICM), 75013 Paris, France
- INSERM U 1127, CNRS UMR 7225, Sorbonne Universités, Université Pierre et Marie Curie-Paris 6, UMR S1127, Institut du Cerveau et de la Moelle épinière (ICM), Pitié-Salpêtrière hospital, 75013 Paris, France
- Centre de recherche de l'Institut Universitaire de Gériatrie de Montréal, H3W 1W6 Montréal, QC, Canada
- Department of Psychology, University of Montreal, H2V 2S9 Montréal, QC, Canada
| | - Simona M. Brambati
- Centre de recherche de l'Institut Universitaire de Gériatrie de Montréal, H3W 1W6 Montréal, QC, Canada
- Department of Psychology, University of Montreal, H2V 2S9 Montréal, QC, Canada
| | - Foudil Lamari
- Department of Metabolic biochemistry, Pitié-Salpêtrière hospital, 75013 Paris, France
| | - Agnès Michon
- Department of Nervous system diseases, Institut de la mémoire et de la maladie d’Alzheimer (IM2A), Neurology, Pitié-Salpêtrière hospital, 75013 Paris, France
| | - Dalila Samri
- Department of Nervous system diseases, Institut de la mémoire et de la maladie d’Alzheimer (IM2A), Neurology, Pitié-Salpêtrière hospital, 75013 Paris, France
| | - Stéphane Epelbaum
- Department of Nervous system diseases, Institut de la mémoire et de la maladie d’Alzheimer (IM2A), Neurology, Pitié-Salpêtrière hospital, 75013 Paris, France
| | - Lucette Lacomblez
- LIB, Inserm U1146, Université Pierre et Marie Curie, Paris 6, 75006 Paris, France
- Department of Nervous system diseases, CIC-CET, Pitié-Salpêtrière hospital, 75013 Paris, France
- Pharmacology service, Pitié-Salpêtrière hospital, 75013 Paris, France
| | - Stéphane Lehéricy
- INSERM U 1127, CNRS UMR 7225, Sorbonne Universités, Université Pierre et Marie Curie-Paris 6, UMR S1127, Institut du Cerveau et de la Moelle épinière (ICM), Pitié-Salpêtrière hospital, 75013 Paris, France
- Centre de Neuro-imagerie de Recherche (CENIR) de l’Institut du Cerveau et de la Moelle Epiniere (ICM), Hôpital de la Pitié-Salpêtrière, 75013 Paris, France
| | - Marie-Odile Habert
- LIB, Inserm U1146, Université Pierre et Marie Curie, Paris 6, 75006 Paris, France
- Department of Nuclear Medicine, Pitié-Salpêtrière hospital, 75013 Paris, France
| | - Bruno Dubois
- FrontLab, Institut du Cerveau et de la Moelle épinière (ICM), 75013 Paris, France
- INSERM U 1127, CNRS UMR 7225, Sorbonne Universités, Université Pierre et Marie Curie-Paris 6, UMR S1127, Institut du Cerveau et de la Moelle épinière (ICM), Pitié-Salpêtrière hospital, 75013 Paris, France
- Department of Nervous system diseases, Institut de la mémoire et de la maladie d’Alzheimer (IM2A), Neurology, Pitié-Salpêtrière hospital, 75013 Paris, France
| | - Aurélie Kas
- LIB, Inserm U1146, Université Pierre et Marie Curie, Paris 6, 75006 Paris, France
- Department of Nuclear Medicine, Pitié-Salpêtrière hospital, 75013 Paris, France
| | - Raffaella Migliaccio
- FrontLab, Institut du Cerveau et de la Moelle épinière (ICM), 75013 Paris, France
- INSERM U 1127, CNRS UMR 7225, Sorbonne Universités, Université Pierre et Marie Curie-Paris 6, UMR S1127, Institut du Cerveau et de la Moelle épinière (ICM), Pitié-Salpêtrière hospital, 75013 Paris, France
- Department of Nervous system diseases, Institut de la mémoire et de la maladie d’Alzheimer (IM2A), Neurology, Pitié-Salpêtrière hospital, 75013 Paris, France
| |
Collapse
|
28
|
Montembeault M, Brambati SM, Gorno-Tempini ML, Migliaccio R. Clinical, Anatomical, and Pathological Features in the Three Variants of Primary Progressive Aphasia: A Review. Front Neurol 2018; 9:692. [PMID: 30186225 PMCID: PMC6110931 DOI: 10.3389/fneur.2018.00692] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 07/31/2018] [Indexed: 11/22/2022] Open
Abstract
Primary progressive aphasias (PPA) are neurodegenerative diseases clinically characterized by an early and relatively isolated language impairment. Three main clinical variants, namely the nonfluent/agrammatic variant (nfvPPA), the semantic variant (svPPA), and the logopenic variant (lvPPA) have been described, each with specific linguistic/cognitive deficits, corresponding anatomical and most probable pathological features. Since the discovery and the development of diagnostic criteria for the PPA variants by the experts in the field, significant progress has been made in the understanding of these diseases. This review aims to provide an overview of the literature on each of the PPA variant in terms of their clinical, anatomical and pathological features, with a specific focus on recent findings. In terms of clinical advancements, recent studies have allowed a better characterization and differentiation of PPA patients based on both their linguistic and non-linguistic profiles. In terms of neuroimaging, techniques such as diffusion imaging and resting-state fMRI have allowed a deeper understanding of the impact of PPA on structural and functional connectivity alterations beyond the well-defined pattern of regional gray matter atrophy. Finally, in terms of pathology, despite significant advances, clinico-pathological correspondence in PPA remains far from absolute. Nonetheless, the improved characterization of PPA has the potential to have a positive impact on the management of patients. Improved reliability of diagnoses and the development of reliable in vivo biomarkers for underlying neuropathology will also be increasingly important in the future as trials for etiology-specific treatments become available.
Collapse
Affiliation(s)
- Maxime Montembeault
- INSERM U 1127, CNRS UMR 7225, Sorbonne Universités, and Université Pierre et Marie Curie-Paris 6, UMR S 1127, Institut du Cerveau et de la Moelle Épinière (ICM), FrontLab, Paris, France.,Centre de Recherche de l'Institut Universitaire de Gériatrie de Montréal, Montréal, QC, Canada.,Département de Psychologie, Université de Montréal, Montréal, QC, Canada
| | - Simona M Brambati
- Centre de Recherche de l'Institut Universitaire de Gériatrie de Montréal, Montréal, QC, Canada.,Département de Psychologie, Université de Montréal, Montréal, QC, Canada
| | - Maria Luisa Gorno-Tempini
- Memory and Aging Center, University of California at San Francisco, San Francisco, CA, United States
| | - Raffaella Migliaccio
- INSERM U 1127, CNRS UMR 7225, Sorbonne Universités, and Université Pierre et Marie Curie-Paris 6, UMR S 1127, Institut du Cerveau et de la Moelle Épinière (ICM), FrontLab, Paris, France.,Department of Neurology, Institut de la Mémoire et de la Maladie d'Alzheimer (IM2A), Paris, France
| |
Collapse
|
29
|
Agosta F, Mandic-Stojmenovic G, Canu E, Stojkovic T, Imperiale F, Caso F, Stefanova E, Copetti M, Kostic VS, Filippi M. Functional and structural brain networks in posterior cortical atrophy: A two-centre multiparametric MRI study. NEUROIMAGE-CLINICAL 2018; 19:901-910. [PMID: 30013929 PMCID: PMC6019262 DOI: 10.1016/j.nicl.2018.06.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 05/07/2018] [Accepted: 06/12/2018] [Indexed: 11/24/2022]
Abstract
This study identified structural and functional brain connectivity alterations in two independent samples of patients along the posterior cortical atrophy (PCA) disease course. Twenty-one PCA patients and 44 controls were recruited from two expert centres. Microstructural damage of white matter (WM) tracts was assessed using probabilistic tractography; resting state (RS) functional connectivity of brain networks was explored using a model free approach; grey matter (GM) atrophy was investigated using voxel-based morphometry. Compared with controls, common patterns of damage across PCA patients included: GM atrophy in the occipital-temporal-parietal regions; diffusion tensor (DT) MRI alterations of the corpus callosum and superior (SLF) and inferior longitudinal fasciculi (ILF) bilaterally; and decreased functional connectivity of the occipital gyri within the visual network and the precuneus and posterior cingulum within the default mode network (DMN). In PCA patients with longer disease duration and greater disease severity, WM damage extended to the cingulum and RS functional connectivity alterations spread within the frontal, dorsal attentive and salience networks. In PCA, reduced DMN functional connectivity was associated with SLF and ILF structural alterations. PCA patients showed distributed WM damage. Altered RS functional connectivity extends with disease worsening from occipital to temporo-parietal and frontostriatal regions, and this is likely to occur through WM connections. Future longitudinal studies are needed to establish trajectories of damage spreading in PCA and whether a combined DT MRI/RS functional MRI approach is promising in monitoring the disease progression. PCA patients showed distributed WM damage. In PCA, WM damage is associated with longer disease duration ad greater severity. In PCA, altered RS functional connectivity extends with disease worsening.
Collapse
Affiliation(s)
- Federica Agosta
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, via Olgettina 60, 20132 Milan, Italy
| | - Gorana Mandic-Stojmenovic
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, via Olgettina 60, 20132 Milan, Italy; Clinic of Neurology, University of Belgrade, Dr Subotića 6, PO, Box 12, 11129, Belgrade 102, Serbia
| | - Elisa Canu
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, via Olgettina 60, 20132 Milan, Italy
| | - Tanja Stojkovic
- Clinic of Neurology, University of Belgrade, Dr Subotića 6, PO, Box 12, 11129, Belgrade 102, Serbia
| | - Francesca Imperiale
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, via Olgettina 60, 20132 Milan, Italy
| | - Francesca Caso
- Department of Neurology, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, via Olgettina 60, 20132 Milan, Italy
| | - Elka Stefanova
- Clinic of Neurology, University of Belgrade, Dr Subotića 6, PO, Box 12, 11129, Belgrade 102, Serbia
| | - Massimiliano Copetti
- Biostatistics Unit, IRCCS-Ospedale Casa Sollievo della Sofferenza, Viale Cappuccini, San Giovanni Rotondo 71013, Foggia, Italy
| | - Vladimir S Kostic
- Clinic of Neurology, University of Belgrade, Dr Subotića 6, PO, Box 12, 11129, Belgrade 102, Serbia
| | - Massimo Filippi
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, via Olgettina 60, 20132 Milan, Italy; Department of Neurology, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, via Olgettina 60, 20132 Milan, Italy.
| |
Collapse
|
30
|
Serra L, Gabrielli GB, Tuzzi E, Spanò B, Giulietti G, Failoni V, Marra C, Caltagirone C, Koch G, Cercignani M, Bozzali M. Damage to the Frontal Aslant Tract Accounts for Visuo-Constructive Deficits in Alzheimer's Disease. J Alzheimers Dis 2018; 60:1015-1024. [PMID: 28984608 DOI: 10.3233/jad-170638] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The frontal aslant tract (FAT) has been described as a bundle connecting the Broca's area to the supplementary motor area (SMA) and the pre-SMA in both hemispheres. The functional properties of this tract and its role in degenerative dementia, such as Alzheimer's disease (AD), still need to be fully clarified. The aim of this study was to explore the microstructural integrity of the FAT in patients with AD and its potential relationship with cognitive functioning. Twenty-three patients with AD and 25 healthy subjects (HS) were enrolled. All subjects underwent cognitive and MRI examination. MRI, including diffusion sequences, was used for probabilistic tractography analysis. We reconstructed individual FATs bilaterally and assessed their microstructural integrity using fractional anisotropy (FA), computed as both mean tract value and voxel-wise using SPM-8. Mean FA values were then used to test for correlations with cognitive measures. Mean tract FA and voxel-wise analyses revealed that patients with AD, compared to HS, had decreased FA in the FAT bilaterally. In addition, positive associations were found between FA in the FATs and patients' performance at tests for constructional praxis and visuospatial logical reasoning. The present results reveal a bilateral damage of FAT in AD patients. The association between FATs' microscopic abnormalities and constructive abilities fits well with the knowledge of a functional involvement of SMA and pre-SMA in movement sequences when executing constructive praxis tasks. The FAT is an associative bundle critically involved in the network sub-serving constructional praxis in patients with AD.
Collapse
Affiliation(s)
- Laura Serra
- Neuroimaging Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
| | | | - Elisa Tuzzi
- Neuroimaging Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Barbara Spanò
- Neuroimaging Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
| | | | - Virginia Failoni
- Neuroimaging Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Camillo Marra
- Institute of Neurology, Catholic University, Rome, Italy
| | - Carlo Caltagirone
- Department of Clinical and Behavioural Neurology, IRCCS Santa Lucia Foundation, Rome, Italy.,Department of Systems Medicine, University of Rome 'Tor Vergata', Rome, Italy
| | - Giacomo Koch
- Department of Clinical and Behavioural Neurology, IRCCS Santa Lucia Foundation, Rome, Italy.,Department of Systems Medicine, University of Rome 'Tor Vergata', Rome, Italy
| | - Mara Cercignani
- Neuroimaging Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy.,Department of Neuroscience, Brighton and Sussex Medical School, University of Sussex, Brighton, UK
| | - Marco Bozzali
- Neuroimaging Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy.,Department of Neuroscience, Brighton and Sussex Medical School, University of Sussex, Brighton, UK
| |
Collapse
|
31
|
Ohm DT, Kim G, Gefen T, Rademaker A, Weintraub S, Bigio EH, Mesulam MM, Rogalski E, Geula C. Prominent microglial activation in cortical white matter is selectively associated with cortical atrophy in primary progressive aphasia. Neuropathol Appl Neurobiol 2018; 45:216-229. [PMID: 29679378 DOI: 10.1111/nan.12494] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 04/02/2018] [Indexed: 02/04/2023]
Abstract
AIMS Primary progressive aphasia (PPA) is a clinical syndrome characterized by selective language impairments associated with focal cortical atrophy favouring the language dominant hemisphere. PPA is associated with Alzheimer's disease (AD), frontotemporal lobar degeneration (FTLD) and significant accumulation of activated microglia. Activated microglia can initiate an inflammatory cascade that may contribute to neurodegeneration, but their quantitative distribution in cortical white matter and their relationship with cortical atrophy remain unknown. We investigated white matter activated microglia and their association with grey matter atrophy in 10 PPA cases with either AD or FTLD-TDP pathology. METHODS Activated microglia were quantified with optical density measures of HLA-DR immunoreactivity in two regions with peak cortical atrophy, and one nonatrophied region within the language dominant hemisphere of each PPA case. Nonatrophied contralateral homologues of the language dominant regions were examined for hemispheric asymmetry. RESULTS Qualitatively, greater densities of activated microglia were observed in cortical white matter when compared to grey matter. Quantitative analyses revealed significantly greater densities of activated microglia in the white matter of atrophied regions compared to nonatrophied regions in the language dominant hemisphere (P < 0.05). Atrophied regions of the language dominant hemisphere also showed significantly more activated microglia compared to contralateral homologues (P < 0.05). CONCLUSIONS White matter activated microglia accumulate more in atrophied regions in the language dominant hemisphere of PPA. While microglial activation may constitute a response to neurodegenerative processes in white matter, the resultant inflammatory processes may also exacerbate disease progression and contribute to cortical atrophy.
Collapse
Affiliation(s)
- D T Ohm
- Cognitive Neurology and Alzheimer's Disease Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - G Kim
- Cognitive Neurology and Alzheimer's Disease Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - T Gefen
- Cognitive Neurology and Alzheimer's Disease Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - A Rademaker
- Cognitive Neurology and Alzheimer's Disease Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - S Weintraub
- Cognitive Neurology and Alzheimer's Disease Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - E H Bigio
- Cognitive Neurology and Alzheimer's Disease Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - M-M Mesulam
- Cognitive Neurology and Alzheimer's Disease Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - E Rogalski
- Cognitive Neurology and Alzheimer's Disease Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - C Geula
- Cognitive Neurology and Alzheimer's Disease Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
32
|
Taipa R, Ferreira V, Brochado P, Robinson A, Reis I, Marques F, Mann DM, Melo-Pires M, Sousa N. Inflammatory pathology markers (activated microglia and reactive astrocytes) in early and late onset Alzheimer disease: a post mortem study. Neuropathol Appl Neurobiol 2017; 44:298-313. [PMID: 29044639 DOI: 10.1111/nan.12445] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 09/22/2017] [Indexed: 01/03/2023]
Abstract
AIMS The association between the pathological features of AD and dementia is stronger in younger old persons than in older old persons suggesting that additional factors are involved in the clinical expression of dementia in the oldest old. Cumulative data suggests that neuroinflammation plays a prominent role in Alzheimer's disease (AD) and different studies reported an age-associated dysregulation of the neuroimmune system. Consequently, we sought to characterize the pattern of microglial cell activation and astrogliosis in brain post mortem tissue of pathologically confirmed cases of early and late onset AD (EOAD and LOAD) and determine their relation to age. METHODS Immunohistochemistry (CD68 and glial fibrillary acidic protein) with morphometric analysis of astroglial profiles in 36 cases of AD and 28 similarly aged controls. RESULTS Both EOAD and LOAD groups had higher microglial scores in CA1, entorhinal and temporal cortices, and higher astroglial response in CA1, dentate gyrus, entorhinal and temporal cortices, compared to aged matched controls. Additionally, EOAD had higher microglial scores in subiculum, entorhinal and temporal subcortical white matter, and LOAD higher astrogliosis in CA2 region. CONCLUSIONS Overall, we found that the neuroinflammatory pathological markers in late stage AD human tissue to have a similar pattern in both EOAD and LOAD, though the severity of the pathological markers in the younger group was higher. Understanding the age effect in AD will be important when testing modifying agents that act on the neuroinflammation.
Collapse
Affiliation(s)
- R Taipa
- Neuropathology Unit, Department of Neurosciences, Centro Hospitalar do Porto, Porto, Portugal.,Life and Health Sciences Research Institute, University of Minho, Braga, Portugal.,ICVS/3B's Associate Lab, PT Government Associated Lab, Braga/Guimarães, Portugal
| | - V Ferreira
- Life and Health Sciences Research Institute, University of Minho, Braga, Portugal.,ICVS/3B's Associate Lab, PT Government Associated Lab, Braga/Guimarães, Portugal
| | - P Brochado
- Neuropathology Unit, Department of Neurosciences, Centro Hospitalar do Porto, Porto, Portugal
| | - A Robinson
- Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, School of Biological Sciences, Salford Royal Hospital Foundation NHS Trust, University of Manchester, Salford, UK
| | - I Reis
- Neuropathology Unit, Department of Neurosciences, Centro Hospitalar do Porto, Porto, Portugal
| | - F Marques
- Life and Health Sciences Research Institute, University of Minho, Braga, Portugal.,ICVS/3B's Associate Lab, PT Government Associated Lab, Braga/Guimarães, Portugal
| | - D M Mann
- Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, School of Biological Sciences, Salford Royal Hospital Foundation NHS Trust, University of Manchester, Salford, UK
| | - M Melo-Pires
- Neuropathology Unit, Department of Neurosciences, Centro Hospitalar do Porto, Porto, Portugal
| | - N Sousa
- Life and Health Sciences Research Institute, University of Minho, Braga, Portugal.,ICVS/3B's Associate Lab, PT Government Associated Lab, Braga/Guimarães, Portugal
| |
Collapse
|
33
|
Raj D, Yin Z, Breur M, Doorduin J, Holtman IR, Olah M, Mantingh-Otter IJ, Van Dam D, De Deyn PP, den Dunnen W, Eggen BJL, Amor S, Boddeke E. Increased White Matter Inflammation in Aging- and Alzheimer's Disease Brain. Front Mol Neurosci 2017; 10:206. [PMID: 28713239 PMCID: PMC5492660 DOI: 10.3389/fnmol.2017.00206] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 06/12/2017] [Indexed: 11/13/2022] Open
Abstract
Chronic neuroinflammation, which is primarily mediated by microglia, plays an essential role in aging and neurodegeneration. It is still unclear whether this microglia-induced neuroinflammation occurs globally or is confined to distinct brain regions. In this study, we investigated microglia activity in various brain regions upon healthy aging and Alzheimer's disease (AD)-related pathology in both human and mouse samples. In purified microglia isolated from aging mouse brains, we found a profound gene expression pattern related to pro-inflammatory processes, phagocytosis, and lipid homeostasis. Particularly in white matter microglia of 24-month-old mice, abundant expression of phagocytic markers including Mac-2, Axl, CD16/32, Dectin1, CD11c, and CD36 was detected. Interestingly, in white matter of human brain tissue the first signs of inflammatory activity were already detected during middle age. Thus quantification of microglial proteins, such as CD68 (commonly associated with phagocytosis) and HLA-DR (associated with antigen presentation), in postmortem human white matter brain tissue showed an age-dependent increase in immunoreactivity already in middle-aged people (53.2 ± 2.0 years). This early inflammation was also detectable by non-invasive positron emission tomography imaging using [11C]-(R)-PK11195, a ligand that binds to activated microglia. Increased microglia activity was also prominently present in the white matter of human postmortem early-onset AD (EOAD) brain tissue. Interestingly, microglia activity in the white matter of late-onset AD (LOAD) CNS was similar to that of the aged clinically silent AD cases. These data indicate that microglia-induced neuroinflammation is predominant in the white matter of aging mice and humans as well as in EOAD brains. This white matter inflammation may contribute to the progression of neurodegeneration, and have prognostic value for detecting the onset and progression of aging and neurodegeneration.
Collapse
Affiliation(s)
- Divya Raj
- Department of Neuroscience, Section Medical Physiology, University Medical Center Groningen, University of GroningenGroningen, Netherlands
| | - Zhuoran Yin
- Department of Neuroscience, Section Medical Physiology, University Medical Center Groningen, University of GroningenGroningen, Netherlands.,Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
| | - Marjolein Breur
- Department of Pathology, VU University Medical CenterAmsterdam, Netherlands
| | - Janine Doorduin
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of GroningenGroningen, Netherlands
| | - Inge R Holtman
- Department of Neuroscience, Section Medical Physiology, University Medical Center Groningen, University of GroningenGroningen, Netherlands
| | - Marta Olah
- Department of Neuroscience, Section Medical Physiology, University Medical Center Groningen, University of GroningenGroningen, Netherlands
| | - Ietje J Mantingh-Otter
- Department of Neuroscience, Section Medical Physiology, University Medical Center Groningen, University of GroningenGroningen, Netherlands
| | - Debby Van Dam
- Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, University of AntwerpWilrijk, Belgium.,Department of Neurology and Alzheimer Research Center, University Medical Center Groningen, University of GroningenGroningen, Netherlands
| | - Peter P De Deyn
- Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, University of AntwerpWilrijk, Belgium.,Department of Neurology and Alzheimer Research Center, University Medical Center Groningen, University of GroningenGroningen, Netherlands.,Biobank, Institute Born-BungeWilrijk, Belgium
| | - Wilfred den Dunnen
- Department of Pathology, University Medical Center Groningen, University of GroningenGroningen, Netherlands
| | - Bart J L Eggen
- Department of Neuroscience, Section Medical Physiology, University Medical Center Groningen, University of GroningenGroningen, Netherlands
| | - Sandra Amor
- Department of Pathology, VU University Medical CenterAmsterdam, Netherlands.,Neuroimmunology Unit, Blizard Institute of Cell and Molecular Science, Barts and The London School of Medicine and DentistryLondon, United Kingdom
| | - Erik Boddeke
- Department of Neuroscience, Section Medical Physiology, University Medical Center Groningen, University of GroningenGroningen, Netherlands
| |
Collapse
|
34
|
Amoroso N, Monaco A, Tangaro S, Neuroimaging Initiative AD. Topological Measurements of DWI Tractography for Alzheimer's Disease Detection. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2017; 2017:5271627. [PMID: 28352290 PMCID: PMC5352968 DOI: 10.1155/2017/5271627] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 10/27/2016] [Indexed: 12/20/2022]
Abstract
Neurodegenerative diseases affect brain morphology and connectivity, making complex networks a suitable tool to investigate and model their effects. Because of its stereotyped pattern Alzheimer's disease (AD) is a natural benchmark for the study of novel methodologies. Several studies have investigated the network centrality and segregation changes induced by AD, especially with a single subject approach. In this work, a holistic perspective based on the application of multiplex network concepts is introduced. We define and assess a diagnostic score to characterize the brain topology and measure the disease effects on a mixed cohort of 52 normal controls (NC) and 47 AD patients, from Alzheimer's Disease Neuroimaging Initiative (ADNI). The proposed topological score allows an accurate NC-AD classification: the average area under the curve (AUC) is 95% and the 95% confidence interval is 92%-99%. Besides, the combination of topological information and structural measures, such as the hippocampal volumes, was also investigated. Topology is able to capture the disease signature of AD and, as the methodology is general, it can find interesting applications to enhance our insight into disease with more heterogeneous patterns.
Collapse
Affiliation(s)
- Nicola Amoroso
- Università degli Studi di Bari “A. Moro”, Via Orabona 4, 70123 Bari, Italy
- Istituto Nazionale di Fisica Nucleare, Sezione di Bari, Via Orabona 4, 70123 Bari, Italy
| | - Alfonso Monaco
- Istituto Nazionale di Fisica Nucleare, Sezione di Bari, Via Orabona 4, 70123 Bari, Italy
| | - Sabina Tangaro
- Istituto Nazionale di Fisica Nucleare, Sezione di Bari, Via Orabona 4, 70123 Bari, Italy
| | | |
Collapse
|
35
|
Alterations of myelin morphology and oligodendrocyte development in early stage of Alzheimer’s disease mouse model. Neurosci Lett 2017; 642:102-106. [DOI: 10.1016/j.neulet.2017.02.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 01/23/2017] [Accepted: 02/03/2017] [Indexed: 11/19/2022]
|
36
|
Moon CM, Shin IS, Jeong GW. Alterations in white matter volume and its correlation with neuropsychological scales in patients with Alzheimer's disease: a DARTEL-based voxel-based morphometry study. Acta Radiol 2017; 58:204-210. [PMID: 27081089 DOI: 10.1177/0284185116640162] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Non-invasive imaging markers can be used to diagnose Alzheimer's disease (AD) in its early stages, but an optimized quantification analysis to measure the brain integrity has been less studied. Purpose To evaluate white matter volume change and its correlation with neuropsychological scales in patients with AD using a diffeomorphic anatomical registration through exponentiated lie algebra (DARTEL)-based voxel-based morphometry (VBM). Material and Methods The 21 participants comprised 11 patients with AD and 10 age-matched healthy controls. High-resolution magnetic resonance imaging (MRI) data were processed by VBM analysis based on DARTEL algorithm. Results The patients showed significant white matter volume reductions in the posterior limb of the internal capsule, cerebral peduncle of the midbrain, and parahippocampal gyrus compared to healthy controls. In correlation analysis, the parahippocampal volume was positively correlated with the Korean-mini mental state examination score in AD. Conclusion This study provides an evidence for localized white matter volume deficits in conjunction with cognitive dysfunction in AD. These findings would be helpful to understand the neuroanatomical mechanisms in AD and to robust the diagnostic accuracy for AD.
Collapse
Affiliation(s)
- Chung-Man Moon
- Research Institute for Medical Imaging, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Il-Seon Shin
- Department of Psychiatry, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Gwang-Woo Jeong
- Research Institute for Medical Imaging, Chonnam National University Medical School, Gwangju, Republic of Korea
- Department of Radiology, Chonnam National University Hospital, Chonnam Natioanl University Medical School, Gwangju, Republic of Korea
| |
Collapse
|
37
|
Millington RS, James-Galton M, Maia Da Silva MN, Plant GT, Bridge H. Lateralized occipital degeneration in posterior cortical atrophy predicts visual field deficits. NEUROIMAGE-CLINICAL 2017; 14:242-249. [PMID: 28180083 PMCID: PMC5288489 DOI: 10.1016/j.nicl.2017.01.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 12/19/2016] [Accepted: 01/15/2017] [Indexed: 11/21/2022]
Abstract
Background Posterior cortical atrophy (PCA), the visual variant of Alzheimer's disease, leads to high-level visual deficits such as alexia or agnosia. Visual field deficits have also been identified, but often inconsistently reported. Little is known about the pattern of visual field deficits or the underlying cortical changes leading to this visual loss. Methods Multi-modal magnetic resonance imaging was used to investigate differences in gray matter volume, cortical thickness, white matter microstructure and functional activity in patients with PCA compared to age-matched controls. Additional analyses investigated hemispheric asymmetries in these metrics according to the visual field most affected by the disease. Results Analysis of structural data indicated considerable loss of gray matter in the occipital and parietal cortices, lateralized to the hemisphere contralateral to the visual loss. This lateralized pattern of gray matter loss was also evident in the hippocampus and parahippocampal gyrus. Diffusion-weighted imaging showed considerable effects of PCA on white matter microstructure in the occipital cortex, and in the corpus callosum. The change in white matter was only lateralized in the occipital lobe, however, with greatest change in the optic radiation contralateral to the visual field deficit. Indeed, there was a significant correlation between the laterality of the optic radiation microstructure and visual field loss. Conclusions Detailed brain imaging shows that the asymmetric visual field deficits in patients with PCA reflect the pattern of degeneration of both white and gray matter in the occipital lobe. Understanding the nature of both visual field deficits and the neurodegenerative brain changes in PCA may improve diagnosis and understanding of this disease. Patients with posterior cortical atrophy show asymmetric visual field deficits manifesting as hemianopia. Both gray and white matter show lateralized degeneration corresponding to the most affected visual field. Laterality of microstructure in the optic radiation correlates with visual field loss.
Collapse
Affiliation(s)
- Rebecca S Millington
- Oxford Centre for fMRI of the Brain (FMRIB), Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | | | | | - Gordon T Plant
- National Hospital for Neurology and Neurosurgery, London, UK; Moorfields Eye Hospital, London, UK
| | - Holly Bridge
- Oxford Centre for fMRI of the Brain (FMRIB), Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| |
Collapse
|
38
|
Chang P, Li X, Ma C, Zhang S, Liu Z, Chen K, Ai L, Chang J, Zhang Z. The Effects of an APOE Promoter Polymorphism on Human White Matter Connectivity during Non-Demented Aging. J Alzheimers Dis 2016; 55:77-87. [DOI: 10.3233/jad-160447] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Peifen Chang
- Dongzhimen Hospital, Beijing university of Chinese Medicine, Beijing, P.R. China
| | - Xin Li
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, P.R. China
| | - Chao Ma
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, P.R. China
| | - Sisi Zhang
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, P.R. China
| | - Zhen Liu
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, P.R. China
| | - Kewei Chen
- Banner Alzheimer’s Institute, Phoenix, AZ, USA
| | - Lin Ai
- Department of Radiology, Beijing Tiantan Hospital, Beijing Neurosurgery Institute, Capital Medical University, Beijing, P.R. China
| | - Jingling Chang
- Dongzhimen Hospital, Beijing university of Chinese Medicine, Beijing, P.R. China
| | - Zhanjun Zhang
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, P.R. China
| |
Collapse
|
39
|
Mattsson N, Insel PS, Palmqvist S, Portelius E, Zetterberg H, Weiner M, Blennow K, Hansson O. Cerebrospinal fluid tau, neurogranin, and neurofilament light in Alzheimer's disease. EMBO Mol Med 2016; 8:1184-1196. [PMID: 27534871 PMCID: PMC5048367 DOI: 10.15252/emmm.201606540] [Citation(s) in RCA: 208] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Cerebrospinal fluid (CSF) tau (total tau, T‐tau), neurofilament light (NFL), and neurogranin (Ng) are potential biomarkers for neurodegeneration in Alzheimer's disease (AD). It is unknown whether these biomarkers provide similar or complementary information in AD. We examined 93 patients with AD, 187 patients with mild cognitive impairment, and 109 controls. T‐tau, Ng, and NFL were all predictors of AD diagnosis. Combinations improved the diagnostic accuracy (AUC 85.5% for T‐tau, Ng, and NFL) compared to individual biomarkers (T‐tau 80.8%; Ng 71.4%; NFL 77.7%). T‐tau and Ng were highly correlated (ρ = 0.79, P < 0.001) and strongly associated with β‐amyloid (Aβ) pathology, and with longitudinal deterioration in cognition and brain structure, primarily in people with Aβ pathology. NFL on the other hand was not associated with Aβ pathology and was associated with cognitive decline and brain atrophy independent of Aβ. T‐tau, Ng, and NFL provide partly independent information about neuronal injury and may be combined to improve the diagnostic accuracy for AD. T‐tau and Ng reflect Aβ‐dependent neurodegeneration, while NFL reflects neurodegeneration independently of Aβ pathology.
Collapse
Affiliation(s)
- Niklas Mattsson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden Department of Neurology, Skåne University Hospital, Lund, Sweden
| | - Philip S Insel
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden Department of Radiology, University of California San Francisco, San Francisco, CA, USA
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden Department of Neurology, Skåne University Hospital, Lund, Sweden
| | - Erik Portelius
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - Michael Weiner
- Department of Radiology, University of California San Francisco, San Francisco, CA, USA
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden Department of Neurology, Skåne University Hospital, Lund, Sweden
| | | |
Collapse
|
40
|
Neitzel J, Ortner M, Haupt M, Redel P, Grimmer T, Yakushev I, Drzezga A, Bublak P, Preul C, Sorg C, Finke K. Neuro-cognitive mechanisms of simultanagnosia in patients with posterior cortical atrophy. Brain 2016; 139:3267-3280. [DOI: 10.1093/brain/aww235] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 07/01/2016] [Accepted: 07/31/2016] [Indexed: 11/13/2022] Open
|
41
|
Migliaccio R, Agosta F, Possin KL, Canu E, Filippi M, Rabinovici GD, Rosen HJ, Miller BL, Gorno-Tempini ML. Mapping the Progression of Atrophy in Early- and Late-Onset Alzheimer's Disease. J Alzheimers Dis 2016; 46:351-64. [PMID: 25737041 DOI: 10.3233/jad-142292] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The term early-onset Alzheimer's disease (EOAD) identifies patients who meet criteria for AD, but show onset of symptoms before the age of 65. We map progression of gray matter atrophy in EOAD patients compared to late-onset AD (LOAD). T1-weighted MRI scans were obtained at diagnosis and one-year follow-up from 15 EOAD, 10 LOAD, and 38 age-matched controls. Voxel-based and tensor-based morphometry were used, respectively, to assess the baseline and progression of atrophy. At baseline, EOAD patients already showed a widespread atrophy in temporal, parietal, occipital, and frontal cortices. After one year, EOAD had atrophy progression in medial temporal and medial parietal cortices. At baseline, LOAD patients showed atrophy in the medial temporal regions only, and, after one year, an extensive pattern of atrophy progression in the same neocortical cortices of EOAD. Although atrophy mainly involved different lateral neocortical or medial temporal hubs at baseline, it eventually progressed along the same brain default-network regions in both groups. The cortical region showing a significant progression in both groups was the medial precuneus/posterior cingulate.
Collapse
|
42
|
Khan TK, Alkon DL. Alzheimer's Disease Cerebrospinal Fluid and Neuroimaging Biomarkers: Diagnostic Accuracy and Relationship to Drug Efficacy. J Alzheimers Dis 2016; 46:817-36. [PMID: 26402622 DOI: 10.3233/jad-150238] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Widely researched Alzheimer's disease (AD) biomarkers include in vivo brain imaging with PET and MRI, imaging of amyloid plaques, and biochemical assays of Aβ 1 - 42, total tau, and phosphorylated tau (p-tau-181) in cerebrospinal fluid (CSF). In this review, we critically evaluate these biomarkers and discuss their clinical utility for the differential diagnosis of AD. Current AD biomarker tests are either highly invasive (requiring CSF collection) or expensive and labor-intensive (neuroimaging), making them unsuitable for use in the primary care, clinical office-based setting, or to assess drug efficacy in clinical trials. In addition, CSF and neuroimaging biomarkers continue to face challenges in achieving required sensitivity and specificity and minimizing center-to-center variability (for CSF-Aβ 1 - 42 biomarkers CV = 26.5% ; http://www.alzforum.org/news/conference-coverage/paris-standardization-hurdle-spinal-fluid-imaging-markers). Although potentially useful for selecting patient populations for inclusion in AD clinical trials, the utility of CSF biomarkers and neuroimaging techniques as surrogate endpoints of drug efficacy needs to be validated. Recent trials of β- and γ-secretase inhibitors and Aβ immunization-based therapies in AD showed no significant cognitive improvements, despite changes in CSF and neuroimaging biomarkers. As we learn more about the dysfunctional cellular and molecular signaling processes that occur in AD, and how these processes are manifested in tissues outside of the brain, new peripheral biomarkers may also be validated as non-invasive tests to diagnose preclinical and clinical AD.
Collapse
|
43
|
Ishiwata A, Kimura K. Homonymous Hemianopsia Associated with Probable Alzheimer's Disease. J NIPPON MED SCH 2016; 83:87-92. [PMID: 27180794 DOI: 10.1272/jnms.83.87] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Posterior cortical atrophy (PCA) is a rare neurodegenerative disorder that has cerebral atrophy in the parietal, occipital, or occipitotemporal cortices and is characterized by visuospatial and visuoperceptual impairments. The most cases are pathologically compatible with Alzheimer's disease (AD). We describe a case of PCA in which a combination of imaging methods, in conjunction with symptoms and neurological and neuropsychological examinations, led to its being diagnosed and to AD being identified as its probable cause. Treatment with donepezil for 6 months mildly improved alexia symptoms, but other symptoms remained unchanged. A 59-year-old Japanese woman with progressive alexia, visual deficit, and mild memory loss was referred to our neurologic clinic for the evaluation of right homonymous hemianopsia. Our neurological examination showed alexia, constructional apraxia, mild disorientation, short-term memory loss, and right homonymous hemianopsia. These findings resulted in a score of 23 (of 30) points on the Mini-Mental State Examination. Occipital atrophy was identified, with magnetic resonance imaging (MRI) showing left-side dominance. The MRI data were quantified with voxel-based morphometry, and PCA was diagnosed on the basis of these findings. Single photon emission computed tomography with (123)I-N-isopropyl-p-iodoamphetamine showed hypoperfusion in the corresponding voxel-based morphometry occipital lobes. Additionally, the finding of hypoperfusion in the posterior associate cortex, posterior cingulate gyrus, and precuneus was consistent with AD. Therefore, the PCA was considered to be a result of AD. We considered Lewy body dementia as a differential diagnosis because of the presence of hypoperfusion in the occipital lobes. However, the patient did not meet the criteria for Lewy body dementia during the course of the disease. We therefore consider including PCA in the differential diagnoses to be important for patients with visual deficit, cognitive impairment, and cerebral atrophy in the parietal, occipital, or occipitotemporal cortices. A combination of imaging methods, including MRI and single photon emission computed tomography, may help identify probable causes of PCA.
Collapse
Affiliation(s)
- Akiko Ishiwata
- Departments of Neurological Science, Graduate School of Medicine, Nippon Medical School
| | | |
Collapse
|
44
|
Zetterberg H, Skillbäck T, Mattsson N, Trojanowski JQ, Portelius E, Shaw LM, Weiner MW, Blennow K. Association of Cerebrospinal Fluid Neurofilament Light Concentration With Alzheimer Disease Progression. JAMA Neurol 2016; 73:60-7. [PMID: 26524180 DOI: 10.1001/jamaneurol.2015.3037] [Citation(s) in RCA: 362] [Impact Index Per Article: 40.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
IMPORTANCE The extent to which large-caliber axonal degeneration contributes to Alzheimer disease (AD) progression is unknown. Cerebrospinal fluid (CSF) neurofilament light (NFL) concentration is a general marker of damage to large-caliber myelinated axons. OBJECTIVE To test whether CSF NFL concentration is associated with cognitive decline and imaging evidence of neurodegeneration and white matter change in AD. DESIGN, SETTING, AND PARTICIPANTS A commercially available immunoassay was used to analyze CSF NFL concentration in a cohort of patients with AD (n = 95) or mild cognitive impairment (MCI) (n = 192) and in cognitively normal individuals (n = 110) from the Alzheimer's Disease Neuroimaging Initiative. The study dates were January 2005 to December 2007. The NFL analysis was performed in November 2014. MAIN OUTCOMES AND MEASURES Correlation was investigated among baseline CSF NFL concentration and longitudinal cognitive impairment, white matter change, and regional brain atrophy within each diagnostic group. RESULTS Cerebrospinal fluid NFL concentration (median [interquartile range]) was higher in the AD dementia group (1479 [1134-1842] pg/mL), stable MCI group (no progression to AD during follow-up; 1182 [923-1687] pg/mL), and progressive MCI group (MCI with progression to AD dementia during follow-up; 1336 [1061-1693] pg/mL) compared with control participants (1047 [809-1265] pg/mL) (P < .001 for all) and in the AD dementia group compared with the stable MCI group (P = .01). In the MCI group, a higher CSF NFL concentration was associated with faster brain atrophy over time as measured by changes in whole-brain volume (β = -4177, P = .003), ventricular volume (β = 1835, P < .001), and hippocampus volume (β = -54.22, P < .001); faster disease progression as reflected by decreased Mini-Mental State Examination scores (β = -1.077, P < .001) and increased Alzheimer Disease Assessment Scale cognitive subscale scores (β = 2.30, P < .001); and faster white matter intensity change (β = 598.7, P < .001). CONCLUSIONS AND RELEVANCE Cerebrospinal fluid NFL concentration is increased by the early clinical stage of AD and is associated with cognitive deterioration and structural brain changes over time. This finding corroborates the contention that degeneration of large-caliber axons is an important feature of AD neurodegeneration.
Collapse
Affiliation(s)
- Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden2Department of Molecular Neuroscience, University College London Institute of Neurology, London, Engla
| | - Tobias Skillbäck
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Niklas Mattsson
- Department of Veterans Affairs Medical Center, University of California, San Francisco4Center for Imaging of Neurodegenerative Diseases, University of California, San Francisco5Department of Radiology and Biomedical Imaging, University of California, San
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, Institute on Aging, University of Pennsylvania School of Medicine, Philadelphia
| | - Erik Portelius
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Leslie M Shaw
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, Institute on Aging, University of Pennsylvania School of Medicine, Philadelphia
| | - Michael W Weiner
- Department of Veterans Affairs Medical Center, University of California, San Francisco
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | | |
Collapse
|
45
|
Moon CM, Kim BC, Jeong GW. Effects of donepezil on brain morphometric and metabolic changes in patients with Alzheimer's disease: A DARTEL-based VBM and (1)H-MRS. Magn Reson Imaging 2016; 34:1008-16. [PMID: 27131829 DOI: 10.1016/j.mri.2016.04.025] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 04/19/2016] [Accepted: 04/22/2016] [Indexed: 11/29/2022]
Abstract
A few studies have performed on the brain morphometric changes over the whole brain structure following donepezil treatment in patients with Alzheimer's disease (AD). We evaluated the gray matter (GM) and white matter (WM) volume alterations and cellular metabolic changes in patients with AD before and after donepezil treatment, and further to reveal the correlations of the scores of various neuropsychological scales with the volumetric and metabolic changes. Twenty-one subjects comprising of 11 patients with AD and 10 age-matched healthy controls participated in this study. All of the patients participated in the follow-up study 24weeks following donepezil treatment. In this study, a combination of voxel-based morphometry (VBM) and proton magnetic resonance spectroscopy ((1)H-MRS) was used to assess the brain morphometric and metabolic alterations in AD. In the GM volumetric analysis, both of the untreated and treated patients with donepezil showed significantly reduced volumes in the hippocampus (Hip), parahippocampal gyrus (PHG), precuneus (PCu) and middle frontal gyrus compared with healthy controls. However, donepezil-treated patients showed significantly increased volumes in the Hip, PCu, fusiform gyrus and caudate nucleus compared to untreated patients. In the WM volumetric analysis, untreated and treated patients showed significant volume reductions in the posterior limb of internal capsule (PLIC), cerebral peduncle of the midbrain and PHG compared to healthy controls. However, there was no significant WM morphological change after donepezil treatment in patients with AD. In MRS study, untreated patients with AD showed decreased N-acetylaspartate/creatine (NAA/Cr) and increased myo-inositol (mI)/Cr compared to healthy controls, while treated patients showed only decreased NAA/Cr in the same comparison. However, the treated patients showed simultaneously increased NAA/Cr and decreased mI/Cr and choline (Cho)/Cr ratios compared to untreated patients. This study shows the regional GM and WM volume changes in combination with metabolic changes following donepezil treatment in AD. These findings would be helpful to aid our understanding of the neuroanatomical mechanisms associated with effects of donepezil on the cognitive function in AD.
Collapse
Affiliation(s)
- Chung-Man Moon
- Research Institute for Medical Imaging, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Byeong-Chae Kim
- Department of Neurology, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Gwang-Woo Jeong
- Research Institute for Medical Imaging, Chonnam National University Medical School, Gwangju, Republic of Korea; Department of Radiology, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju, Republic of Korea.
| |
Collapse
|
46
|
Migliaccio R, Gallea C, Kas A, Perlbarg V, Samri D, Trotta L, Michon A, Lacomblez L, Dubois B, Lehericy S, Bartolomeo P. Functional Connectivity of Ventral and Dorsal Visual Streams in Posterior Cortical Atrophy. J Alzheimers Dis 2016; 51:1119-30. [DOI: 10.3233/jad-150934] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Raffaella Migliaccio
- INSERM U 1127, CNRS UMR 7225, Sorbonne Universités, and Université Pierre et Marie Curie-Paris 6, UMR S 1127, Institut du Cerveau et de la Moelle épinière (ICM), F-75013 Paris, France
- Department of Neurology, Institut de la mémoire et de la maladie d’Alzheimer, Hôpital de la Pitié-Salpêtrière, AP-HP, Paris, France
| | - Cécile Gallea
- INSERM U 1127, CNRS UMR 7225, Sorbonne Universités, and Université Pierre et Marie Curie-Paris 6, UMR S 1127, Institut du Cerveau et de la Moelle épinière (ICM), F-75013 Paris, France
- Centre de Neuro-imagerie de Recherche (CENIR) de l’Institut du Cerveau et de la Moelle Epiniere (ICM), Hôpital de la Pitié-Salpêtrière, Paris, France
- Equipe “Mouvements Anormaux et Ganglions de la Base”, Institut du Cerveau et de la Moëlle Epinière, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Aurélie Kas
- Service de médecine nucléaire, Hôpital Pitié-Salpêtrière, APHP, Paris, France
- INSERM U1146, CNRS UMR7371, laboratoire d’imagerie biomédicale, Sorbonne université, UPMC université, Paris 60 UMCR2, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Vincent Perlbarg
- INSERM U 1127, CNRS UMR 7225, Sorbonne Universités, and Université Pierre et Marie Curie-Paris 6, UMR S 1127, Institut du Cerveau et de la Moelle épinière (ICM), F-75013 Paris, France
- INSERM U1146, CNRS UMR7371, laboratoire d’imagerie biomédicale, Sorbonne université, UPMC université, Paris 60 UMCR2, Hôpital de la Pitié-Salpêtrière, Paris, France
- IHU-A-ICM, Bioinformatics/Biostatistis Plateform, Paris, France
| | - Dalila Samri
- Department of Neurology, Institut de la mémoire et de la maladie d’Alzheimer, Hôpital de la Pitié-Salpêtrière, AP-HP, Paris, France
| | - Laura Trotta
- INSERM U 1127, CNRS UMR 7225, Sorbonne Universités, and Université Pierre et Marie Curie-Paris 6, UMR S 1127, Institut du Cerveau et de la Moelle épinière (ICM), F-75013 Paris, France
- Department of Neurology, Institut de la mémoire et de la maladie d’Alzheimer, Hôpital de la Pitié-Salpêtrière, AP-HP, Paris, France
| | - Agnès Michon
- Department of Neurology, Institut de la mémoire et de la maladie d’Alzheimer, Hôpital de la Pitié-Salpêtrière, AP-HP, Paris, France
| | - Lucette Lacomblez
- INSERM U1146, CNRS UMR7371, laboratoire d’imagerie biomédicale, Sorbonne université, UPMC université, Paris 60 UMCR2, Hôpital de la Pitié-Salpêtrière, Paris, France
- Department des maladies du système nerveux, CIC-CET, Hôpital de la Pitié-Salpêtrière, AP-HP, Paris, France
- Service de pharmacologie, Hôpital de la Pitié-Salpêtrière, AP-HP, Paris, France
| | - Bruno Dubois
- INSERM U 1127, CNRS UMR 7225, Sorbonne Universités, and Université Pierre et Marie Curie-Paris 6, UMR S 1127, Institut du Cerveau et de la Moelle épinière (ICM), F-75013 Paris, France
- Department of Neurology, Institut de la mémoire et de la maladie d’Alzheimer, Hôpital de la Pitié-Salpêtrière, AP-HP, Paris, France
| | - Stéphane Lehericy
- INSERM U 1127, CNRS UMR 7225, Sorbonne Universités, and Université Pierre et Marie Curie-Paris 6, UMR S 1127, Institut du Cerveau et de la Moelle épinière (ICM), F-75013 Paris, France
- Centre de Neuro-imagerie de Recherche (CENIR) de l’Institut du Cerveau et de la Moelle Epiniere (ICM), Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Paolo Bartolomeo
- INSERM U 1127, CNRS UMR 7225, Sorbonne Universités, and Université Pierre et Marie Curie-Paris 6, UMR S 1127, Institut du Cerveau et de la Moelle épinière (ICM), F-75013 Paris, France
- Department of Psychology, Catholic University, Milan, Italy
| |
Collapse
|
47
|
Marceglia S, Mrakic-Sposta S, Rosa M, Ferrucci R, Mameli F, Vergari M, Arlotti M, Ruggiero F, Scarpini E, Galimberti D, Barbieri S, Priori A. Transcranial Direct Current Stimulation Modulates Cortical Neuronal Activity in Alzheimer's Disease. Front Neurosci 2016; 10:134. [PMID: 27065792 PMCID: PMC4814712 DOI: 10.3389/fnins.2016.00134] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 03/16/2016] [Indexed: 02/02/2023] Open
Abstract
Quantitative electroencephalography (qEEG) showed that Alzheimer's disease (AD) is characterized by increased theta power, decreased alpha and beta power, and decreased coherence in the alpha and theta band in posterior regions. These abnormalities are thought to be associated with functional disconnections among cortical areas, death of cortical neurons, axonal pathology, and cholinergic deficits. Since transcranial Direct Current Stimulation (tDCS) over the temporo-parietal area is thought to have beneficial effects in patients with AD, in this study we aimed to investigate whether tDCS benefits are related to tDCS-induced changes in cortical activity, as represented by qEEG. A weak anodal current (1.5 mA, 15 min) was delivered bilaterally over the temporal-parietal lobe to seven subjects with probable AD (Mini-Mental State Examination, MMSE score >20). EEG (21 electrodes, 10–20 international system) was recorded for 5 min with eyes closed before (baseline, t0) and 30 min after anodal and cathodal tDCS ended (t1). At the same time points, patients performed a Word Recognition Task (WRT) to assess working memory functions. The spectral power and the inter- and intra-hemispheric EEG coherence in different frequency bands (e.g., low frequencies, including delta and theta; high frequencies, including alpha and beta) were calculated for each subject at t0 and t1. tDCS-induced changes in EEG neurophysiological markers were correlated with the performance of patients at the WRT. At baseline, qEEG features in AD patients confirmed that the decreased high frequency power was correlated with lower MMSE. After anodal tDCS, we observed an increase in the high-frequency power in the temporo-parietal area and an increase in the temporo-parieto-occipital coherence that correlated with the improvement at the WRT. In addition, cathodal tDCS produced a non-specific effect of decreased theta power all over the scalp that was not correlated with the clinical observation at the WRT. Our findings disclosed that tDCS induces significant modulations in the cortical EEG activity in AD patients. The abnormal pattern of EEG activity observed in AD during memory processing is partially reversed by applying anodal tDCS, suggesting that anodal tDCS benefits in AD patients during working memory tasks are supported by the modulation of cortical activity.
Collapse
Affiliation(s)
- Sara Marceglia
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Clinical Center for Neurostimulation, Neurotechnology, and Movement DisordersMilano, Italia; Dipartimento di Ingegneria e Architettura, Università degli Studi di TriesteTrieste, Italia
| | - Simona Mrakic-Sposta
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Clinical Center for Neurostimulation, Neurotechnology, and Movement DisordersMilano, Italia; Istituto di Bioimmagini e di Fisiologia Molecolare, Consiglio Nazionale delle RicercheSegrate, Italia
| | - Manuela Rosa
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Clinical Center for Neurostimulation, Neurotechnology, and Movement Disorders Milano, Italia
| | - Roberta Ferrucci
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Clinical Center for Neurostimulation, Neurotechnology, and Movement Disorders Milano, Italia
| | - Francesca Mameli
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Clinical Center for Neurostimulation, Neurotechnology, and Movement Disorders Milano, Italia
| | - Maurizio Vergari
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Clinical Center for Neurostimulation, Neurotechnology, and Movement Disorders Milano, Italia
| | - Mattia Arlotti
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Clinical Center for Neurostimulation, Neurotechnology, and Movement DisordersMilano, Italia; Dipartimento di Ingegneria Elettrica e dell'Informazione "Guglielmo Marconi," Università di BolognaCesena, Italia
| | - Fabiana Ruggiero
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Clinical Center for Neurostimulation, Neurotechnology, and Movement Disorders Milano, Italia
| | - Elio Scarpini
- Unità di Neurologia, Dipartimento di Fisiologia Medico-Chirurgica e dei Trapianti, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico Milano, Italia
| | - Daniela Galimberti
- Unità di Neurologia, Dipartimento di Fisiologia Medico-Chirurgica e dei Trapianti, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico Milano, Italia
| | - Sergio Barbieri
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Clinical Center for Neurostimulation, Neurotechnology, and Movement Disorders Milano, Italia
| | - Alberto Priori
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Clinical Center for Neurostimulation, Neurotechnology, and Movement DisordersMilano, Italia; Dipartimento di Scienze della Salute, Università degli Studi di Milano, Polo Ospedaliero San PaoloMilano, Italia
| |
Collapse
|
48
|
Migliaccio R, Boutet C, Valabregue R, Ferrieux S, Nogues M, Lehéricy S, Dormont D, Levy R, Dubois B, Teichmann M. The Brain Network of Naming: A Lesson from Primary Progressive Aphasia. PLoS One 2016; 11:e0148707. [PMID: 26901052 PMCID: PMC4764674 DOI: 10.1371/journal.pone.0148707] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 01/20/2016] [Indexed: 12/23/2022] Open
Abstract
OBJECTIVE Word finding depends on the processing of semantic and lexical information, and it involves an intermediate level for mapping semantic-to-lexical information which also subserves lexical-to-semantic mapping during word comprehension. However, the brain regions implementing these components are still controversial and have not been clarified via a comprehensive lesion model encompassing the whole range of language-related cortices. Primary progressive aphasia (PPA), for which anomia is thought to be the most common sign, provides such a model, but the exploration of cortical areas impacting naming in its three main variants and the underlying processing mechanisms is still lacking. METHODS We addressed this double issue, related to language structure and PPA, with thirty patients (11 semantic, 12 logopenic, 7 agrammatic variant) using a picture-naming task and voxel-based morphometry for anatomo-functional correlation. First, we analyzed correlations for each of the three variants to identify the regions impacting naming in PPA and to disentangle the core regions of word finding. We then combined the three variants and correlation analyses for naming (semantic-to-lexical mapping) and single-word comprehension (lexical-to-semantic mapping), predicting an overlap zone corresponding to a bidirectional lexical-semantic hub. RESULTS AND CONCLUSIONS Our results showed that superior portions of the left temporal pole and left posterior temporal cortices impact semantic and lexical naming mechanisms in semantic and logopenic PPA, respectively. In agrammatic PPA naming deficits were rare, and did not correlate with any cortical region. Combined analyses revealed a cortical overlap zone in superior/middle mid-temporal cortices, distinct from the two former regions, impacting bidirectional binding of lexical and semantic information. Altogether, our findings indicate that lexical/semantic word processing depends on an anterior-posterior axis within lateral-temporal cortices, including an anatomically intermediate hub dedicated to lexical-semantic integration. Within this axis our data reveal the underpinnings of anomia in the PPA variants, which is of relevance for both diagnosis and future therapy strategies.
Collapse
Affiliation(s)
- Raffaella Migliaccio
- Institut du Cerveau et de la Moelle Epinière, UMR INSERM-CNRS-UPMC 1127, Frontlab, Paris, France
- Department of Neurology, National Reference Center for « PPA and rare dementias », Pitié Salpêtrière Hospital, AP-HP, Paris, France
| | - Claire Boutet
- Institut du Cerveau et de la Moelle Epinière, UMR INSERM-CNRS-UPMC 1127, Frontlab, Paris, France
- Centre de Neuro-imagerie de Recherche (CENIR), Institut du Cerveau et de la Moëlle Epinière, Paris, France
| | - Romain Valabregue
- Institut du Cerveau et de la Moelle Epinière, UMR INSERM-CNRS-UPMC 1127, Frontlab, Paris, France
- Centre de Neuro-imagerie de Recherche (CENIR), Institut du Cerveau et de la Moëlle Epinière, Paris, France
| | - Sophie Ferrieux
- Department of Neurology, National Reference Center for « PPA and rare dementias », Pitié Salpêtrière Hospital, AP-HP, Paris, France
| | - Marie Nogues
- Department of Neurology, National Reference Center for « PPA and rare dementias », Pitié Salpêtrière Hospital, AP-HP, Paris, France
| | - Stéphane Lehéricy
- Institut du Cerveau et de la Moelle Epinière, UMR INSERM-CNRS-UPMC 1127, Frontlab, Paris, France
- Centre de Neuro-imagerie de Recherche (CENIR), Institut du Cerveau et de la Moëlle Epinière, Paris, France
| | - Didier Dormont
- Université Pierre et Marie Curie, INSERM, UMR-S 678, Paris, France
- Service de Neuroradiologie Diagnostique et Fonctionnelle, Hôpital de la Pitié-Salpêtrière, AP-HP, Paris, France
| | - Richard Levy
- Institut du Cerveau et de la Moelle Epinière, UMR INSERM-CNRS-UPMC 1127, Frontlab, Paris, France
- Department of Neurology, Hôpital Saint Antoine, AP-HP, Paris, France
| | - Bruno Dubois
- Institut du Cerveau et de la Moelle Epinière, UMR INSERM-CNRS-UPMC 1127, Frontlab, Paris, France
- Department of Neurology, National Reference Center for « PPA and rare dementias », Pitié Salpêtrière Hospital, AP-HP, Paris, France
| | - Marc Teichmann
- Institut du Cerveau et de la Moelle Epinière, UMR INSERM-CNRS-UPMC 1127, Frontlab, Paris, France
- Department of Neurology, National Reference Center for « PPA and rare dementias », Pitié Salpêtrière Hospital, AP-HP, Paris, France
- * E-mail:
| |
Collapse
|
49
|
Madhavan A, Schwarz CG, Duffy JR, Strand EA, Machulda MM, Drubach DA, Kantarci K, Przybelski SA, Reid RI, Senjem ML, Gunter JL, Apostolova LG, Lowe VJ, Petersen RC, Jack CR, Josephs KA, Whitwell JL. Characterizing White Matter Tract Degeneration in Syndromic Variants of Alzheimer's Disease: A Diffusion Tensor Imaging Study. J Alzheimers Dis 2016; 49:633-43. [PMID: 26484918 PMCID: PMC10038690 DOI: 10.3233/jad-150502] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Different clinical syndromes can arise from Alzheimer's disease (AD) neuropathology, including dementia of the Alzheimer's type (DAT), logopenic primary progressive aphasia (lvPPA), and posterior cortical atrophy (PCA). OBJECTIVE To assess similarities and differences in patterns of white matter tract degeneration across these syndromic variants of AD. METHODS Sixty-four subjects (22 DAT, 24 lvPPA, and 18 PCA) that had diffusion tensor imaging and showed amyloid-β deposition on PET were assessed in this case-control study. A whole-brain voxel-based analysis was performed to assess differences in fractional anisotropy, mean diffusivity, axial diffusivity, and radial diffusivity across groups. RESULTS All three groups showed overlapping diffusion abnormalities in a network of tracts, including fornix, corpus callosum, posterior thalamic radiations, superior longitudinal fasciculus, inferior longitudinal fasciculus, inferior fronto-occipital fasciculus, and uncinate fasciculus. Subtle regional differences were also observed across groups, with DAT particularly associated with degeneration of fornix and cingulum, lvPPA with left inferior fronto-occipital fasciculus and uncinate fasciculus, and PCA with posterior thalamic radiations, superior longitudinal fasciculus, posterior cingulate, and splenium of the corpus callosum. CONCLUSION These findings show that while each AD phenotype is associated with degeneration of a specific structural network of white matter tracts, striking spatial overlap exists among the three network patterns that may be related to AD pathology.
Collapse
Affiliation(s)
- Ajay Madhavan
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | | | - Joseph R. Duffy
- Department of Neurology (Speech Pathology), Mayo Clinic, Rochester, MN, USA
| | - Edythe A. Strand
- Department of Neurology (Speech Pathology), Mayo Clinic, Rochester, MN, USA
| | - Mary M. Machulda
- Department of Psychiatry and Psychology (Neuropsychology), Mayo Clinic, Rochester, MN, USA
| | - Daniel A. Drubach
- Department of Neurology (Behavioral Neurology), Mayo Clinic, Rochester, MN, USA
| | - Kejal Kantarci
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Scott A. Przybelski
- Department of Health Sciences Research (Biostatistics), Mayo Clinic, Rochester, MN, USA
| | - Robert I. Reid
- Department of Information Technology, Mayo Clinic, Rochester, MN, USA
| | - Matthew L. Senjem
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
- Department of Information Technology, Mayo Clinic, Rochester, MN, USA
| | - Jeffrey L. Gunter
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
- Department of Information Technology, Mayo Clinic, Rochester, MN, USA
| | - Liana G. Apostolova
- Department of Neurology, University of California Los Angeles, Los Angeles, CA, USA
| | - Val J. Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Ronald C. Petersen
- Department of Neurology (Behavioral Neurology), Mayo Clinic, Rochester, MN, USA
| | | | - Keith A. Josephs
- Department of Neurology (Behavioral Neurology), Mayo Clinic, Rochester, MN, USA
| | - Jennifer L. Whitwell
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
- Correspondence to: Jennifer L. Whitwell, PhD, Associate Professor of Radiology, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA. Tel.: +1 507 284 5576; Fax: +1 507 284 9778;
| |
Collapse
|
50
|
In-vivo imaging of grey and white matter neuroinflammation in Alzheimer's disease: a positron emission tomography study with a novel radioligand, [18F]-FEPPA. Mol Psychiatry 2015; 20:1579-87. [PMID: 25707397 PMCID: PMC8026116 DOI: 10.1038/mp.2015.1] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 12/16/2014] [Accepted: 12/19/2014] [Indexed: 11/09/2022]
Abstract
Our primary aim was to compare neuroinflammation in cognitively intact control subjects and patients with Alzheimer's disease (AD) by using positron emission tomography (PET) with translocator protein 18 kDa (TSPO)-specific radioligand [(18)F]-FEPPA. [(18)F]-FEPPA PET scans were acquired on a high-resolution research tomograph in 21 patients with AD (47- 81 years) and 21 control subjects (49-82 years). They were analyzed by using a 2-tissue compartment model with arterial plasma input function. Differences in neuroinflammation, indexed as [(18)F]-FEPPA binding were compared, adjusting for differences in binding affinity class as determined by a single polymorphism in the TSPO gene (rs6971). In grey matter areas, [(18)F]-FEPPA was significantly higher in AD compared with healthy control subjects. Large increases were seen in the hippocampus, prefrontal, temporal, parietal and occipital cortex (average Cohen's d= 0.89). Voxel-based analyses confirmed significant clusters of neuroinflammation in the frontal, temporal and parietal cortex in patients with AD. In white matter, [(18)F]-FEPPA binding was elevated in the posterior limb of the internal capsule, and the cingulum bundle. Higher neuroinflammation in the parietal cortex (r= -0.7, P= 0.005), and posterior limb of the internal capsule (r= -0.8, P=0.001) was associated with poorer visuospatial function. In addition, a higher [(18)F]-FEPPA binding in the posterior limb of the internal capsule was associated with a greater impairment in language ability (r= -0.7, P=0.004). Elevated neuroinflammation can be detected in AD patients throughout the brain grey and white matter by using [(18)F]-FEPPA PET. Our results also suggest that neuroinflammation is associated with some cognitive deficits.
Collapse
|