1
|
Wang S, Huang J, Chen Y, Liang Y, Chen L, Ye D, Yang H, Hui Z, Wang X, Zhang Z, Zhu X. Qifu-yin activates the Keap1/Nrf2/ARE signaling and ameliorates synaptic injury and oxidative stress in APP/PS1 mice. JOURNAL OF ETHNOPHARMACOLOGY 2024; 333:118497. [PMID: 38942156 DOI: 10.1016/j.jep.2024.118497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 06/13/2024] [Accepted: 06/25/2024] [Indexed: 06/30/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The traditional medicinal formulation, Qifu-yin (QFY), has been widely prescribed for Alzheimer's disease (AD) treatment in China, yet the comprehensive mechanisms through which QFY mitigates AD pathology remain to be fully delineated. AIM OF THE STUDY This study aimed to explore the therapeutic implications of QFY on the synaptic injury and oxidative stress in the hippocampus of APPswe/PS1dE9 (APP/PS1) mice, with a concerted effort to elucidate the molecular mechanisms related to synaptic preservation and memory improvement. MATERIALS AND METHODS The components of QFY were identified by ultra-high-performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS). The neuroprotective effects of QFY was evaluated using six-month-old male APP/PS1 mice. Subsequent to a 15 days of QFY regimen, spatial memory was assessed utilizing the Morris water maze (MWM) test. Amyloid-beta (Aβ) aggregation was detected via immunostaining, while the quantification of Aβ1-40 and Aβ1-42 was achieved through enzyme-linked immunosorbent assay (ELISA). Transmission electron microscopy (TEM) was used to investigate the synaptic structure and mitochondrial morphology. Golgi staining was applied to examine dendritic spine density. Reactive oxygen species (ROS), 3-nitrotyrosine (3-NT) and 4-hydroxy-nonenal (4-HNE) assays were employed to assess oxidative stress. The expression profiles of Aβ metabolism-associated enzymes and the Keap1/Nrf2/ARE signaling pathway were determined by Western blot. RESULTS A total of 20 principal compounds in QFY were identified. QFY mitigated memory deficits of APP/PS1 mice, including reducing escape latency and search distance and increasing the time and distance spent in the target quadrant. In addition, QFY increased platform crossings of APP/PS1 mice in the probe trial of MWM tests. TEM analysis showed that QFY increased synapse number in the CA1 region of APP/PS1 mice. Further studies indicated that QFY elevated the expression levels of Post synaptic density protein 95 (PSD95) and synaptophysin, and mitigated the loss of dendritic spine density in the hippocampus of APP/PS1 mice. QFY has been shown to ameliorated the structural abnormalities of mitochondria, including mitochondrial dissolution and degradation, up-regulate ATP synthesis and membrane potential in the hippocampus of APP/PS1 mice. Moreover, QFY activated the Keap1/Nrf2/ARE signaling pathway in the hippocampus of APP/PS1 mice, which might contribute to the neuroprotective effects of QFY. CONCLUSION QFY activates the Keap1/Nrf2/ARE signaling, and protects against synaptic and mitochondrial dysfunction in APP/PS1 mice, proposing a potential alternative therapeutic strategy for AD management.
Collapse
Affiliation(s)
- Sulei Wang
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210001, China; Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China.
| | - Jing Huang
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210001, China; Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China.
| | - Yanting Chen
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210001, China; Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China.
| | - Ying Liang
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210001, China; Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China.
| | - Liqiu Chen
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210001, China; Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China.
| | - Dan Ye
- Department of Neurology, The Third Affiliated Hospital of Soochow University, The First People's Hospital of Changzhou, Changzhou, 213000, China.
| | - Hui Yang
- School of Nursing, Nanjing University of Chinese Medicine, Nanjing, 210046, China.
| | - Zhen Hui
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210001, China; Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China.
| | - Xiaomian Wang
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210001, China; Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China.
| | - Zhennian Zhang
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210001, China; Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China.
| | - Xiaolei Zhu
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210001, China; Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China.
| |
Collapse
|
2
|
Pecher H, Storch M, Beyer F, Witte V, Baasner CF, Schönknecht P, Weise CM. Hypothalamic atrophy and structural covariance in amnestic mild cognitive impairment and Alzheimer's dementia. Neuroimage Clin 2024; 44:103687. [PMID: 39406040 PMCID: PMC11525751 DOI: 10.1016/j.nicl.2024.103687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 09/10/2024] [Accepted: 10/08/2024] [Indexed: 11/03/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) is characterized by progressive cognitive decline and specific brain atrophy patterns, primarily involving the medial temporal lobes. A number of studies have discussed hypothalamic involvement in AD with consecutive metabolic and/or autonomic disturbances yet only few studies have investigated hypothalamic atrophy in AD and its early stages in particular. METHODS We applied semi-automated volumetry of the hypothalamus (HTH) in 3 T MRI in a sample N = 175 participants [age 74.9 ± 7.22; gender 85 m/90f; cognitively normal controls (CN; N = 56); amnestic mild cognitive impairment (MCI; N = 78); AD (N = 41)] from the Alzheimer's Disease Neuroimaging Initiative (ADNI). In addition, we used voxel-based morphometry (VBM), cortical thickness (CTH) analyses and source-based morphometry (SBM) derived networks of structural covariance to investigate brain structural covariance patterns of the HTH under consideration of diagnostic groups, β-amyloid (AB) positivity and apolipoprotein E (APOE) ε4 status. RESULTS Hypothalamic atrophy was observed in both early and advanced disease stages (i.e. hypothalamic volume CN > MCI > AD). VBM, CTH analysis and SBM revealed positive associations between hypothalamic volume (HV) and AD-vulnerable regions, largely corresponding to the Papez circuit and brain regions implicated in autonomic regulation, however, group differences regarding HTH structural covariance were not observed. Similar observations were made in carriers and non-carriers of the ε4 allele, yet more pronounced in ε4 carriers. Although not reaching significance, comparisons of AB positive vs. negative subjects indicated stronger HTH atrophy in biomarker positive participants. HV was not associated with body mass index or longitudinal weight change. CONCLUSIONS Our findings support early structural changes of the HTH in AD. HV covaries with regional volumes of AD-vulnerable regions. This could point to secondary atrophy of the HTH following atrophy of the hippocampus and other structures of the Papez circuit in AD.
Collapse
Affiliation(s)
- Hannah Pecher
- Department of Neurology, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), German; Department of Radiology, Bundeswehrkrankenhaus Berlin, Scharnhorststr. 13, 10115 Berlin, Germany.
| | - Melanie Storch
- Department of Psychiatry and Psychotherapy, University Hospital Leipzig, Semmelweisstr. 10, 04103 Leipzig, Germany; Department of Biology, University of Leipzig, 04103 Leipzig, Germany
| | - Frauke Beyer
- Department of Neurology, Max Planck-Institute for Human Cognitive and Brain Sciences, and Cognitive Neurology, University of Leipzig Medical Center, Leipzig, Germany
| | - Veronica Witte
- Department of Neurology, Max Planck-Institute for Human Cognitive and Brain Sciences, and Cognitive Neurology, University of Leipzig Medical Center, Leipzig, Germany
| | - Christian-Frank Baasner
- Department of Neurology, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), German
| | - Peter Schönknecht
- Medical Faculty, Department of Psychiatry and Psychotherapy, University Hospital Leipzig, 04103 Leipzig, Germany; Out-Patient Department for Sexual-Therapeutic Prevention and Forensic Psychiatry, University Hospital Leipzig, 04103, Leipzig, Germany; Academic Saxon State Hospital Altscherbitz, 04435 Schkeuditz, Germany
| | - Christopher M Weise
- Department of Neurology, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), German
| |
Collapse
|
3
|
Qi G, Tang H, Gong P, Liu Y, He C, Hu J, Kang S, Chen L, Qin S. Sex-specific hypothalamic neuropathology and glucose metabolism in an amyloidosis transgenic mouse model of Alzheimer's disease. Cell Biosci 2024; 14:120. [PMID: 39272160 PMCID: PMC11395863 DOI: 10.1186/s13578-024-01295-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 08/21/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND Amyloid toxicity and glucose metabolic disorders are key pathological features during the progression of Alzheimer's disease (AD). While the hypothalamus plays a crucial role in regulating systemic energy balance, the distribution of amyloid plaques in the preoptic, anterior, tuberal, and mammillary regions of the hypothalamus in AD mice, particularly across both sexes, remains largely unclear. Our ongoing research aims to explore hypothalamic neuropathology and glucose metabolic disturbances in a well-described APP/PS1 mouse model of AD. RESULTS Immunocytochemical staining revealed that Old-AD-Female mice exhibited a greater hypothalamic Amyloid β (Aβ) burden than their Old-AD-Male counterparts, with the mammillary bodies showing the most severe accumulation. Analysis of ionized calcium binding adaptor molecule 1 (IBA1) immunoreactivity and Iba1 mRNA indicated differential microgliosis based on sex, while tanycytic territory and ZO-1 tight junction protein expression remained stable in AD mice. Moreover, sex-specific peripheral glucose metabolic parameters (random and fasting blood glucose) seemed to be exacerbated by age. Old AD mice of both sexes exhibited limited hypothalamic activation (c-Fos + cells) in response to blood glucose fluctuations. Hypothalamic Glut 1 expression decreased in young but increased in old female AD mice compared with age-matched male AD mice. Pearson correlation analysis further supported a negative correlation between hypothalamic Aβ load and random blood glucose in old AD groups of both genders, shedding light on the mechanisms underlying this amyloidosis mouse model. CONCLUSION Aged APP/PS1 mice exhibit sex-specific hypothalamic neuropathology and differential glucose metabolism, highlighting distinct pathological mechanisms within each gender.
Collapse
Affiliation(s)
- Guibo Qi
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Han Tang
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Pifang Gong
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Yitong Liu
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Chenzhao He
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Jianian Hu
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Siying Kang
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Liang Chen
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
| | - Song Qin
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
4
|
Arjmand S, Ilaghi M, Sisakht AK, Guldager MB, Wegener G, Landau AM, Gjedde A. Regulation of mitochondrial dysfunction by estrogens and estrogen receptors in Alzheimer's disease: A focused review. Basic Clin Pharmacol Toxicol 2024; 135:115-132. [PMID: 38801027 DOI: 10.1111/bcpt.14035] [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: 03/01/2024] [Revised: 05/02/2024] [Accepted: 05/07/2024] [Indexed: 05/29/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that primarily manifests itself by progressive memory loss and cognitive decline, thus significantly affecting memory functions and quality of life. In this review, we proceed from the understanding that the canonical amyloid-β hypothesis, while significant, has faced setbacks, highlighting the need to adopt a broader perspective considering the intricate interplay of diverse pathological pathways for effective AD treatments. Sex differences in AD offer valuable insights into a better understanding of its pathophysiology. Fluctuation of the levels of ovarian sex hormones during perimenopause is associated with changes in glucose metabolism, as a possible window of opportunity to further understand the roles of sex steroid hormones and their associated receptors in the pathophysiology of AD. We review these dimensions, emphasizing the potential of estrogen receptors (ERs) to reveal mitochondrial functions in the search for further research and therapeutic strategies for AD pharmacotherapy. Understanding and addressing the intricate interactions of mitochondrial dysfunction and ERs potentially pave the way for more effective approaches to AD therapy.
Collapse
Affiliation(s)
- Shokouh Arjmand
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Mehran Ilaghi
- Institute of Neuropharmacology, Kerman Neuroscience Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Ali Karimi Sisakht
- Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Matti Bock Guldager
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Gregers Wegener
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Anne M Landau
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Albert Gjedde
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
5
|
Akhgari A, Michel TM, Vafaee MS. Dendritic spines and their role in the pathogenesis of neurodevelopmental and neurological disorders. Rev Neurosci 2024; 35:489-502. [PMID: 38440811 DOI: 10.1515/revneuro-2023-0151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 02/02/2024] [Indexed: 03/06/2024]
Abstract
Since Cajal introduced dendritic spines in the 19th century, they have attained considerable attention, especially in neuropsychiatric and neurologic disorders. Multiple roles of dendritic spine malfunction and pathology in the progression of various diseases have been reported. Thus, it is inevitable to consider these structures as new therapeutic targets for treating neuropsychiatric and neurologic disorders such as autism spectrum disorders, schizophrenia, dementia, Down syndrome, etc. Therefore, we attempted to prepare a narrative review of the literature regarding the role of dendritic spines in the pathogenesis of aforementioned diseases and to shed new light on their pathophysiology.
Collapse
Affiliation(s)
- Aisan Akhgari
- Student Research Committee, Tabriz University of Medical Sciences, Golgasht Street, Tabriz 5166616471, Iran
| | - Tanja Maria Michel
- Research Unit for Psychiatry, Odense University Hospital, J. B. Winsløws Vej 4, Odense 5000, Denmark
- Clinical Institute, University of Southern Denmark, Campusvej 55, Odense 5230, Denmark
| | - Manouchehr Seyedi Vafaee
- Research Unit for Psychiatry, Odense University Hospital, J. B. Winsløws Vej 4, Odense 5000, Denmark
- Clinical Institute, University of Southern Denmark, Campusvej 55, Odense 5230, Denmark
| |
Collapse
|
6
|
Ferrer I. Historical review: The golden age of the Golgi method in human neuropathology. J Neuropathol Exp Neurol 2024; 83:375-395. [PMID: 38622902 DOI: 10.1093/jnen/nlae031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2024] Open
Abstract
Golgi methods were used to study human neuropathology in the 1970s, 1980s, and 1990s of the last century. Although a relatively small number of laboratories applied these methods, their impact was crucial by increasing knowledge about: (1) the morphology, orientation, and localization of neurons in human cerebral and cerebellar malformations and ganglionic tumors, and (2) the presence of abnormal structures including large and thin spines (spine dysgenesis) in several disorders linked to mental retardation, focal enlargements of the axon hillock and dendrites (meganeurites) in neuronal storage diseases, growth cone-like appendages in Alzheimer disease, as well as abnormal structures in other dementias. Although there were initial concerns about their reliability, reduced dendritic branches and dendritic spines were identified as common alterations in mental retardation, dementia, and other pathological conditions. Similar observations in appropriate experimental models have supported many abnormalities that were first identified using Golgi methods in human material. Moreover, electron microscopy, immunohistochemistry, fluorescent tracers, and combined methods have proven the accuracy of pioneering observations uniquely visualized as 3D images of fully stained individual neurons. Although Golgi methods had their golden age many years ago, these methods may still be useful complementary tools in human neuropathology.
Collapse
Affiliation(s)
- Isidro Ferrer
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Hospitalet de LLobregat, Spain
| |
Collapse
|
7
|
Rapaka D, Tebogo MO, Mathew EM, Adiukwu PC, Bitra VR. Targeting papez circuit for cognitive dysfunction- insights into deep brain stimulation for Alzheimer's disease. Heliyon 2024; 10:e30574. [PMID: 38726200 PMCID: PMC11079300 DOI: 10.1016/j.heliyon.2024.e30574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 04/28/2024] [Accepted: 04/29/2024] [Indexed: 05/12/2024] Open
Abstract
Hippocampus is the most widely studied brain area coupled with impairment of memory in a variety of neurological diseases and Alzheimer's disease (AD). The limbic structures within the Papez circuit have been linked to various aspects of cognition. Unfortunately, the brain regions that include this memory circuit are often ignored in terms of understanding cognitive decline in these diseases. To properly comprehend where cognition problems originate, it is crucial to clarify any aberrant contributions from all components of a specific circuit -on both a local and a global level. The pharmacological treatments currently available are not long lasting. Deep Brain Stimulation (DBS) emerged as a new powerful therapeutic approach for alleviation of the cognitive dysfunctions. Metabolic, functional, electrophysiological, and imaging studies helped to find out the crucial nodes that can be accessible for DBS. Targeting these nodes within the memory circuit produced significant improvement in learning and memory by disrupting abnormal circuit activity and restoring the physiological network. Here, we provide an overview of the neuroanatomy of the circuit of Papez along with the mechanisms and various deep brain stimulation targets of the circuit structures which could be significant for improving cognitive dysfunctions in AD.
Collapse
Affiliation(s)
| | - Motshegwana O. Tebogo
- School of Pharmacy, Faculty of Health Sciences, University of Botswana, Gaborone, Botswana, P/Bag-0022
| | - Elizabeth M. Mathew
- School of Pharmacy, Faculty of Health Sciences, University of Botswana, Gaborone, Botswana, P/Bag-0022
| | | | - Veera Raghavulu Bitra
- School of Pharmacy, Faculty of Health Sciences, University of Botswana, Gaborone, Botswana, P/Bag-0022
| |
Collapse
|
8
|
Mar KD, So C, Hou Y, Kim JC. Age dependent path integration deficit in 5xFAD mice. Behav Brain Res 2024; 463:114919. [PMID: 38408521 DOI: 10.1016/j.bbr.2024.114919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 02/16/2024] [Accepted: 02/20/2024] [Indexed: 02/28/2024]
Abstract
Alzheimer's disease (AD) is a severe neurodegenerative disorder and the most common form of dementia in elderly individuals, characterized by memory deficits, cognitive decline, and neuropathology. The identification of preclinical markers for AD remains elusive. We employed an ultrasound-evoked spatial memory assay to investigate path integration (PI) in wild type C57BL/6 J and 5xFAD mice. We observed significant recruitment of the mammillary bodies (MB) and subiculum (Sub) - core regions of the Papez circuit during PI, as indicated by increased expression of the immediate early gene c-Fos in C57BL/6 J mice. In 5xFAD mice, amyloid-beta (Aβ) vulnerability in the MB and Sub was evident at 3-months of age, preceding widespread pathology at 5-months of age. In parallel, we detected significant behavioral deficits in PI in the 5XFAD mice at 5- but not 3-months of age. Sex based analysis revealed a more profound deficit in males compared to females at 5-months of age. Our data suggest PI may be as an early indicator of AD, potentially associated with dysfunction within the Papez circuit.
Collapse
Affiliation(s)
- Kendall D Mar
- Department of Psychology, University of Toronto, 100 St. George Street, Sidney Smith Hall, Toronto, Ontario M5S 3G3, Canada.
| | - Chanbee So
- Department of Cell and Systems Biology, University of Toronto, 25 Harbord Street, Toronto, Ontario M5S 3G5, Canada.
| | - Yixin Hou
- Department of Cell and Systems Biology, University of Toronto, 25 Harbord Street, Toronto, Ontario M5S 3G5, Canada.
| | - Jun Chul Kim
- Department of Psychology, University of Toronto, 100 St. George Street, Sidney Smith Hall, Toronto, Ontario M5S 3G3, Canada; Department of Cell and Systems Biology, University of Toronto, 25 Harbord Street, Toronto, Ontario M5S 3G5, Canada.
| |
Collapse
|
9
|
Huang WC, Peng Z, Murdock MH, Liu L, Mathys H, Davila-Velderrain J, Jiang X, Chen M, Ng AP, Kim T, Abdurrob F, Gao F, Bennett DA, Kellis M, Tsai LH. Lateral mammillary body neurons in mouse brain are disproportionately vulnerable in Alzheimer's disease. Sci Transl Med 2023; 15:eabq1019. [PMID: 37075128 PMCID: PMC10511020 DOI: 10.1126/scitranslmed.abq1019] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 03/31/2023] [Indexed: 04/21/2023]
Abstract
The neural circuits governing the induction and progression of neurodegeneration and memory impairment in Alzheimer's disease (AD) are incompletely understood. The mammillary body (MB), a subcortical node of the medial limbic circuit, is one of the first brain regions to exhibit amyloid deposition in the 5xFAD mouse model of AD. Amyloid burden in the MB correlates with pathological diagnosis of AD in human postmortem brain tissue. Whether and how MB neuronal circuitry contributes to neurodegeneration and memory deficits in AD are unknown. Using 5xFAD mice and postmortem MB samples from individuals with varying degrees of AD pathology, we identified two neuronal cell types in the MB harboring distinct electrophysiological properties and long-range projections: lateral neurons and medial neurons. lateral MB neurons harbored aberrant hyperactivity and exhibited early neurodegeneration in 5xFAD mice compared with lateral MB neurons in wild-type littermates. Inducing hyperactivity in lateral MB neurons in wild-type mice impaired performance on memory tasks, whereas attenuating aberrant hyperactivity in lateral MB neurons ameliorated memory deficits in 5xFAD mice. Our findings suggest that neurodegeneration may be a result of genetically distinct, projection-specific cellular dysfunction and that dysregulated lateral MB neurons may be causally linked to memory deficits in AD.
Collapse
Affiliation(s)
- Wen-Chin Huang
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
| | - Zhuyu Peng
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
| | - Mitchell H. Murdock
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
| | - Liwang Liu
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
| | - Hansruedi Mathys
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Broad Institute of MIT and Harvard; Cambridge, MA, 02139, USA
| | - Jose Davila-Velderrain
- Broad Institute of MIT and Harvard; Cambridge, MA, 02139, USA
- MIT Computer Science and Artificial Intelligence Laboratory; Cambridge, MA 02139, USA
| | - Xueqiao Jiang
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
| | - Maggie Chen
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
| | - Ayesha P. Ng
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
| | - TaeHyun Kim
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
| | - Fatema Abdurrob
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
| | - Fan Gao
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
| | - David A. Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center; Chicago, IL 60612, USA
| | - Manolis Kellis
- Broad Institute of MIT and Harvard; Cambridge, MA, 02139, USA
- MIT Computer Science and Artificial Intelligence Laboratory; Cambridge, MA 02139, USA
| | - Li-Huei Tsai
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology; Cambridge, MA, 02139, USA
- Broad Institute of MIT and Harvard; Cambridge, MA, 02139, USA
| |
Collapse
|
10
|
Cheron G, Ristori D, Marquez-Ruiz J, Cebolla AM, Ris L. Electrophysiological alterations of the Purkinje cells and deep cerebellar neurons in a mouse model of Alzheimer disease (electrophysiology on cerebellum of AD mice). Eur J Neurosci 2022; 56:5547-5563. [PMID: 35141975 DOI: 10.1111/ejn.15621] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 12/16/2021] [Accepted: 12/19/2021] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease is histopathologically well defined by the presence of amyloid deposits and tau-related neurofibrillary tangles in crucial regions of the brain. Interest is growing in revealing and determining possible pathological markers also in the cerebellum as its involvement in cognitive functions is now well supported. Despite the central position of the Purkinje cell in the cerebellum, its electrophysiological behaviour in mouse models of Alzheimer's disease is scarce in the literature. Our first aim was here to focus on the electrophysiological behaviour of the cerebellum in awake mouse model of Alzheimer's disease (APPswe/PSEN1dE9) and the related performance on the water-maze test classically used in behavioural studies. We found prevalent signs of electrophysiological alterations in both Purkinje cells and deep cerebellar nuclei neurons which might explain the behavioural deficits reported during the water-maze test. The alterations of neurons firing were accompanied by a dual (~16 and ~228 Hz) local field potential's oscillation in the Purkinje cell layer of Alzheimer's disease mice which was concomitant to an important increase of both the simple and the complex spikes. In addition, β-amyloid deposits were present in the molecular layer of the cerebellum. These results highlight the importance of the output firing modification of the AD cerebellum that may indirectly impact the activity of its subcortical and cortical targets.
Collapse
Affiliation(s)
- Guy Cheron
- Laboratory of Neurophysiology and Movement Biomechanics, Université Libre de Bruxelles, Brussels, Belgium.,ULB Neuroscience Institut, Université Libre de Bruxelles, Brussels, Belgium.,Laboratory of Neuroscience, Université de Mons, Mons, Belgium
| | - Dominique Ristori
- Laboratory of Neurophysiology and Movement Biomechanics, Université Libre de Bruxelles, Brussels, Belgium
| | - Javier Marquez-Ruiz
- Department of Physiology, Anatomy and Cell Biology, Pablo de Olavide University, Seville, Spain
| | - Anna-Maria Cebolla
- Laboratory of Neurophysiology and Movement Biomechanics, Université Libre de Bruxelles, Brussels, Belgium
| | - Laurence Ris
- Laboratory of Neuroscience, Université de Mons, Mons, Belgium.,UMONS Research Institut for health and technology, Université de Mons, Mons, Belgium
| |
Collapse
|
11
|
Meys KME, de Vries LS, Groenendaal F, Vann SD, Lequin MH. The Mammillary Bodies: A Review of Causes of Injury in Infants and Children. AJNR Am J Neuroradiol 2022; 43:802-812. [PMID: 35487586 PMCID: PMC9172959 DOI: 10.3174/ajnr.a7463] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 11/22/2021] [Indexed: 12/20/2022]
Abstract
Despite their small size, the mammillary bodies play an important role in supporting recollective memory. However, they have typically been overlooked when assessing neurologic conditions that present with memory impairment. While there is increasing evidence of mammillary body involvement in a wide range of neurologic disorders in adults, very little attention has been given to infants and children. Literature searches of PubMed and EMBASE were performed to identify articles that describe mammillary body pathology on brain MR imaging in children. Mammillary body pathology is present in the pediatric population in several conditions, indicated by signal change and/or atrophy on MR imaging. The main causes of mammillary body pathology are thiamine deficiency, hypoxia-ischemia, direct damage due to masses or hydrocephalus, or deafferentation resulting from pathology within the wider Papez circuit. Optimizing scanning protocols and assessing mammillary body status as a standard procedure are critical, given their role in memory processes.
Collapse
Affiliation(s)
- K M E Meys
- From the Department of Radiology (K.M.E.M., F.G., M.H.L.), Wilhelmina Children's Hospital, University Medical Center Utrecht and Utrecht University, Utrecht, Netherlands
| | - L S de Vries
- Department of Neonatology (L.S.D.V.), Wilhelmina Children's Hospital, University Medical Center Utrecht and Utrecht University, Utrecht, Netherlands
| | - F Groenendaal
- From the Department of Radiology (K.M.E.M., F.G., M.H.L.), Wilhelmina Children's Hospital, University Medical Center Utrecht and Utrecht University, Utrecht, Netherlands
| | - S D Vann
- School of Psychology (S.D.V.), Cardiff University, Cardiff, UK
| | - M H Lequin
- From the Department of Radiology (K.M.E.M., F.G., M.H.L.), Wilhelmina Children's Hospital, University Medical Center Utrecht and Utrecht University, Utrecht, Netherlands
| |
Collapse
|
12
|
Reiss AB, Ahmed S, Dayaramani C, Glass AD, Gomolin IH, Pinkhasov A, Stecker MM, Wisniewski T, De Leon J. The role of mitochondrial dysfunction in Alzheimer's disease: A potential pathway to treatment. Exp Gerontol 2022; 164:111828. [DOI: 10.1016/j.exger.2022.111828] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 04/15/2022] [Accepted: 04/27/2022] [Indexed: 11/17/2022]
|
13
|
Kim S, Nam Y, Kim HS, Jung H, Jeon SG, Hong SB, Moon M. Alteration of Neural Pathways and Its Implications in Alzheimer’s Disease. Biomedicines 2022; 10:biomedicines10040845. [PMID: 35453595 PMCID: PMC9025507 DOI: 10.3390/biomedicines10040845] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/29/2022] [Accepted: 03/31/2022] [Indexed: 02/01/2023] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disease accompanied by cognitive and behavioral symptoms. These AD-related manifestations result from the alteration of neural circuitry by aggregated forms of amyloid-β (Aβ) and hyperphosphorylated tau, which are neurotoxic. From a neuroscience perspective, identifying neural circuits that integrate various inputs and outputs to determine behaviors can provide insight into the principles of behavior. Therefore, it is crucial to understand the alterations in the neural circuits associated with AD-related behavioral and psychological symptoms. Interestingly, it is well known that the alteration of neural circuitry is prominent in the brains of patients with AD. Here, we selected specific regions in the AD brain that are associated with AD-related behavioral and psychological symptoms, and reviewed studies of healthy and altered efferent pathways to the target regions. Moreover, we propose that specific neural circuits that are altered in the AD brain can be potential targets for AD treatment. Furthermore, we provide therapeutic implications for targeting neuronal circuits through various therapeutic approaches and the appropriate timing of treatment for AD.
Collapse
Affiliation(s)
- Sujin Kim
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon 35365, Korea; (S.K.); (Y.N.); (H.s.K.); (H.J.); (S.G.J.); (S.B.H.)
- Research Institute for Dementia Science, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon 35365, Korea
| | - Yunkwon Nam
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon 35365, Korea; (S.K.); (Y.N.); (H.s.K.); (H.J.); (S.G.J.); (S.B.H.)
| | - Hyeon soo Kim
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon 35365, Korea; (S.K.); (Y.N.); (H.s.K.); (H.J.); (S.G.J.); (S.B.H.)
| | - Haram Jung
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon 35365, Korea; (S.K.); (Y.N.); (H.s.K.); (H.J.); (S.G.J.); (S.B.H.)
| | - Seong Gak Jeon
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon 35365, Korea; (S.K.); (Y.N.); (H.s.K.); (H.J.); (S.G.J.); (S.B.H.)
| | - Sang Bum Hong
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon 35365, Korea; (S.K.); (Y.N.); (H.s.K.); (H.J.); (S.G.J.); (S.B.H.)
| | - Minho Moon
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon 35365, Korea; (S.K.); (Y.N.); (H.s.K.); (H.J.); (S.G.J.); (S.B.H.)
- Research Institute for Dementia Science, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon 35365, Korea
- Correspondence:
| |
Collapse
|
14
|
Marterstock DC, Knott MFX, Hoelter P, Lang S, Oberstein T, Kornhuber J, Doerfler A, Schmidt MA. Pulsed Arterial Spin Labeling and Segmented Brain Volumetry in the Diagnostic Evaluation of Frontotemporal Dementia, Alzheimer’s Disease and Mild Cognitive Impairment. Tomography 2022; 8:229-244. [PMID: 35076603 PMCID: PMC8788517 DOI: 10.3390/tomography8010018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 11/21/2022] Open
Abstract
Background: Previous studies suggest that brain atrophy can not only be defined by its morphological extent, but also by the cerebral blood flow (CBF) within a certain area of the brain, including white and gray matter. The aim of this study is to investigate known atrophy patterns in different forms of dementia and to compare segmented brain volumetrics and pulsed arterial spin labeling (pASL) data to explore the correlation between brain maps with atrophy and this non-contrast-enhanced brain-perfusion method. Methods: Our study comprised 17 patients with diagnosed cognitive impairment (five Alzheimer’s disease = AD, five frontotemporal dementia = FTD, seven mild cognitive impairment = MCI) and 19 healthy control subjects (CO). All patients and controls underwent 4D-pASL brain-perfusion MR imaging and T1w MPRAGE. The data were assessed regarding relative brain volume on the basis of 286 brain regions, and absolute and relative cerebral blood flow (CBF/rCBF) were derived from pASL data in the corresponding brain regions. Mini-Mental State Examination (MMSE) was performed to assess cognitive functions. Results: FTD patients demonstrated significant brain atrophy in 43 brain regions compared to CO. Patients with MCI showed significant brain atrophy in 18 brain regions compared to CO, whereas AD patients only showed six brain regions with significant brain atrophy compared to CO. There was good correlation of brain atrophy and pASL perfusion data in five brain regions of patients with diagnosed FTD, especially in the superior temporal gyrus (r = 0.900, p = 0.037), the inferior frontal white matter (pars orbitalis; r = 0.968, p = 0.007) and the thalami (r = 0.810, p = 0.015). Patients with MCI demonstrated a correlation in one brain region (left inferior fronto-occipital fasciculus; r = 0.786, p = 0.036), whereas patients with diagnosed AD revealed no correlation. Conclusions: pASL can detect affected brain regions in cognitive impairment and corresponds with brain atrophy, especially for patients suffering from FTD and MCI. However, there was no correlation of perfusion alterations and brain atrophy in AD. pASL perfusion might thus represent a promising tool for noninvasive brain-perfusion evaluation in specific dementia subtypes as a complimentary imaging-based bio marker in addition to brain volumetry.
Collapse
Affiliation(s)
- Dominique Cornelius Marterstock
- Department of Neuroradiology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Schwabachanlage 6, 91054 Erlangen, Germany
| | - Michael Franz Xaver Knott
- Department of Neuroradiology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Schwabachanlage 6, 91054 Erlangen, Germany
| | - Philip Hoelter
- Department of Neuroradiology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Schwabachanlage 6, 91054 Erlangen, Germany
| | - Stefan Lang
- Department of Neuroradiology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Schwabachanlage 6, 91054 Erlangen, Germany
| | - Timo Oberstein
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Schwabachanlage 6, 91054 Erlangen, Germany
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Schwabachanlage 6, 91054 Erlangen, Germany
| | - Arnd Doerfler
- Department of Neuroradiology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Schwabachanlage 6, 91054 Erlangen, Germany
| | - Manuel A Schmidt
- Department of Neuroradiology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Schwabachanlage 6, 91054 Erlangen, Germany
| |
Collapse
|
15
|
Forno G, Lladó A, Hornberger M. Going round in circles-The Papez circuit in Alzheimer's disease. Eur J Neurosci 2021; 54:7668-7687. [PMID: 34656073 DOI: 10.1111/ejn.15494] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 10/01/2021] [Accepted: 10/12/2021] [Indexed: 11/29/2022]
Abstract
The hippocampus is regarded as the pivotal structure for episodic memory symptoms associated with Alzheimer's disease (AD) pathophysiology. However, what is often overlooked is that the hippocampus is 'only' one part of a network of memory critical regions, the Papez circuit. Other Papez circuit regions are often regarded as less relevant for AD as they are thought to sit 'downstream' of the hippocampus. However, this notion is oversimplistic, and increasing evidence suggests that other Papez regions might be affected before or concurrently with the hippocampus. In addition, AD research has mostly focused on episodic memory deficits, whereas spatial navigation processes are also subserved by the Papez circuit with increasing evidence supporting its valuable potential as a diagnostic measure of incipient AD pathophysiology. In the current review, we take a step forward analysing recent evidence on the structural and functional integrity of the Papez circuit across AD disease stages. Specifically, we will review the integrity of specific Papez regions from at-genetic-risk (APOE4 carriers), to mild cognitive impairment (MCI), to dementia stage of sporadic AD and autosomal dominant AD (ADAD). We related those changes to episodic memory and spatial navigation/orientation deficits in AD. Finally, we provide an overview of how the Papez circuit is affected in AD diseases and their specific symptomology contributions. This overview strengthened the need for moving away from a hippocampal-centric view to a network approach on how the whole Papez circuit is affected in AD and contributes to its symptomology, informing future research and clinical approaches.
Collapse
Affiliation(s)
- Gonzalo Forno
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain.,School of Psychology, Universidad de los Andes, Santiago, Chile.,Neuropsychology and Clinical Neuroscience Laboratory (LANNEC), Physiopathology Department, ICBM, Neurosciences Department, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Albert Lladó
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | | |
Collapse
|
16
|
Mavroudis IA, Petridis F, Chatzikonstantinou S, Karantali E, Kazis D. A meta-analysis on the levels of VILIP-1 in the CSF of Alzheimer's disease compared to normal controls and other neurodegenerative conditions. Aging Clin Exp Res 2021; 33:265-272. [PMID: 31939203 DOI: 10.1007/s40520-019-01458-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 12/18/2019] [Indexed: 01/24/2023]
Abstract
Alzheimer's disease (AD) is a progressing neurodegenerative disorder and the main cause of serious irreversible cognitive decline in elderly people. Visinin-like protein 1 (VILIP-1) is a member of the family of calcium-binding proteins and plays a crucial role in AD pathophysiology. Multiple studies have shown that CSF levels of VILIP-1 are increased in AD patients compared to normal controls, or other neurodegenerative conditions. We searched online databases for studies on the levels of VILIP-1 in the CSF of AD patients in comparison to normal controls, mild cognitive impairment (MCI) patients and Dementia with Lewy bodies (DLB) patients. A total of ten studies were used for the comparison between AD and controls, three studies for the comparison between AD and MCI, two studies for AD and DLB and two studies for the comparison between stable MCI and MCI progressed to AD. We found that VILIP-1 levels are significantly higher in AD compared to normal controls, but not to the other groups, and furthermore, they are significantly higher in patient with MCI progressed to AD, than in stable MCI patients.
Collapse
Affiliation(s)
- Ioannis A Mavroudis
- Leeds Teaching Hospitals NHS Trust, Leeds, UK.
- Medical School, University of Cyprus, Nicosia, Cyprus.
| | - Foivos Petridis
- Third Department of Neurology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | - Eleni Karantali
- Third Department of Neurology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Dimitris Kazis
- Third Department of Neurology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
17
|
Mickelsen LE, Flynn WF, Springer K, Wilson L, Beltrami EJ, Bolisetty M, Robson P, Jackson AC. Cellular taxonomy and spatial organization of the murine ventral posterior hypothalamus. eLife 2020; 9:58901. [PMID: 33119507 PMCID: PMC7595735 DOI: 10.7554/elife.58901] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 09/21/2020] [Indexed: 01/02/2023] Open
Abstract
The ventral posterior hypothalamus (VPH) is an anatomically complex brain region implicated in arousal, reproduction, energy balance, and memory processing. However, neuronal cell type diversity within the VPH is poorly understood, an impediment to deconstructing the roles of distinct VPH circuits in physiology and behavior. To address this question, we employed a droplet-based single-cell RNA sequencing (scRNA-seq) approach to systematically classify molecularly distinct cell populations in the mouse VPH. Analysis of >16,000 single cells revealed 20 neuronal and 18 non-neuronal cell populations, defined by suites of discriminatory markers. We validated differentially expressed genes in selected neuronal populations through fluorescence in situ hybridization (FISH). Focusing on the mammillary bodies (MB), we discovered transcriptionally-distinct clusters that exhibit neuroanatomical parcellation within MB subdivisions and topographic projections to the thalamus. This single-cell transcriptomic atlas of VPH cell types provides a resource for interrogating the circuit-level mechanisms underlying the diverse functions of VPH circuits.
Collapse
Affiliation(s)
- Laura E Mickelsen
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, United States.,Connecticut Institute for the Brain and Cognitive Sciences, Storrs, United States
| | - William F Flynn
- The Jackson Laboratory for Genomic Medicine, Farmington, United States
| | - Kristen Springer
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, United States
| | - Lydia Wilson
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, United States
| | - Eric J Beltrami
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, United States
| | - Mohan Bolisetty
- The Jackson Laboratory for Genomic Medicine, Farmington, United States
| | - Paul Robson
- The Jackson Laboratory for Genomic Medicine, Farmington, United States.,Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, United States.,Institute for Systems Genomics, University of Connecticut, Farmington, United States
| | - Alexander C Jackson
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, United States.,Connecticut Institute for the Brain and Cognitive Sciences, Storrs, United States.,Institute for Systems Genomics, University of Connecticut, Farmington, United States
| |
Collapse
|
18
|
Jin Z, Sethi SK, Li B, Tang R, Li Y, Hsu CCT, He N, Haacke EM, Yan F. Susceptibility and Volume Measures of the Mammillary Bodies Between Mild Cognitively Impaired Patients and Healthy Controls. Front Neurosci 2020; 14:572595. [PMID: 33041764 PMCID: PMC7522522 DOI: 10.3389/fnins.2020.572595] [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: 06/15/2020] [Accepted: 08/24/2020] [Indexed: 12/11/2022] Open
Abstract
Purpose To investigate the baseline values and differences for susceptibility and volume of the mammillary bodies between mild cognitively impaired (MCI) patients and healthy controls (HCs), and further explore their differences in relation to gender, MCI subtypes and apolipoprotein E (APOE) genotypes. Methods T1-weighted and multi-echo gradient echo imaging sequences were acquired on a 3T MR scanner to evaluate the T1W based volume and susceptibility differences in the mammillary body for 47 MCI and 47 HCs. T-tests were performed to compare volume and susceptibility between groups, and right and left hemispheres. Correlation analysis was used to relate the volume and mean susceptibility as a function of age in MCI and HC groups separately, and to investigate the relationship of susceptibility with the neuro-psychological scales in the MCI group. Results Susceptibility was found to be elevated within the right mammillary body in MCI patients compared to HCs (p < 0.05). There were no differences for the mammillary body volumes between the MCI and HC groups, although there was a reduction in volume with age for the MCI group (p = 0.007). Women showed decreased mammillary body volume compared to men in the HC group (p = 0.004). No significant differences were found in relation to MCI subtypes and APOE genotypes. No significant correlations were observed between mammillary body susceptibility with neuro-psychological scales. Conclusion This work provides a quantitative baseline for both the volume and susceptibility of the mammillary body which can be used for future studies of cognitive impairment patients underlying the pathology of the Papez circuit.
Collapse
Affiliation(s)
- Zhijia Jin
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Sean K Sethi
- Magnetic Resonance Innovations, Inc., Bingham Farms, MI, United States.,Department of Radiology, Wayne State University, Detroit, MI, United States
| | - Binyin Li
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rongbiao Tang
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yufei Li
- Institute for Medical Imaging Technology, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Charlie Chia-Tsong Hsu
- Department of Medical Imaging, Gold Coast University Hospital, Southport, QLD, Australia
| | - Naying He
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - E Mark Haacke
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Magnetic Resonance Innovations, Inc., Bingham Farms, MI, United States.,Department of Radiology, Wayne State University, Detroit, MI, United States.,Department of Biomedical Engineering, Wayne State University, Detroit, MI, United States
| | - Fuhua Yan
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
19
|
Ishunina TA, Bogolepova IN, Swaab DF. Increased Neuronal Nuclear and Perikaryal Size in the Medial Mamillary Nucleus of Vascular Dementia and Alzheimer's Disease Patients: Relation to Nuclear Estrogen Receptor α. Dement Geriatr Cogn Disord 2020; 47:274-280. [PMID: 31319413 DOI: 10.1159/000500244] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 04/09/2019] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The hypothalamic medial mamillary (MMN) and the tuberomamillary (TMN) nuclei are important hubs in memory circuits. Previous studies determining the neuronal Golgi complex size showed decreased metabolic activity of the TMN neurons in both Alzhei-mer's disease (AD) and vascular dementia (VD), and no obvious decline in the MMN of these patients. OBJECTIVES In the present study, we aimed at determining whether other morphometric parameters that are informative about the neuronal metabolic activity are changed in the MMN of AD and VD patients and whether they can be related to the expression of the nuclear estrogen receptor α (ERα) that can mediate neurotrophic effects of estrogens in the brain. METHOD The size of neuronal nuclei and perikarya was determined in AD, VD, and nondemented control patients, in relation to the expression of the nuclear ERα. RESULTS We found that neuronal nuclear and perikaryal sizes were significantly larger in the MMN in VD than in control patients (p < 0.01). Neuronal nuclei (p < 0.05), but not perikarya were larger in AD than in control patients. Neuronal nuclei and perikarya were larger if nuclear ERα staining was present. The intensity of ERα in the neuronal nuclei was significantly correlated with both nuclear and perikaryal sizes (p < 0.007). CONCLUSIONS The human MMN shows a remarkable activation in aging and extra activation in dementias (AD and VD) that may be mediated by nuclear ERα. This makes it so far a unique brain area to study compensatory mechanisms that may prevent neurodegeneration.
Collapse
Affiliation(s)
- Tatjana A Ishunina
- Department of Histology, Embryology, Cytology, Kursk State Medical University, Kursk, Russian Federation, .,Netherlands Institute for Neuroscience, Amsterdam, The Netherlands,
| | - Irina N Bogolepova
- Department of Brain Research, Federal State Budget Scientific Institution "Research Center of Neurology", Moscow, Russian Federation
| | - Dick F Swaab
- Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| |
Collapse
|
20
|
Cognitive behavioral therapy (CBT) for preventing Alzheimer's disease. Behav Brain Res 2017; 334:163-177. [PMID: 28743599 DOI: 10.1016/j.bbr.2017.07.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 07/15/2017] [Accepted: 07/18/2017] [Indexed: 12/13/2022]
Abstract
This review provides the rationale for implementing cognitive behavioral therapy (CBT) for the prevention of Alzheimer's disease (AD). There are known risk factors associated with the development of AD, some of which may be ameliorated with CBT. We posit that treating the risk factors of inactivity, poor diet, hyposmia and anosmia, sleep disorders and lack of regularly engaged challenging cognitive activity will modify the physiology of the brain sufficiently to avoid the accumulation of excess proteins, including amyloid beta, causal events in the development of AD. Further, the successful treatment of the listed risk factors is well within our technology to do so and, even further, it is cost effective. Also, there is considerable scientific literature to support the proposition that, if implemented by well-established practices, CBT will be effective and will be engaged by those of retirement age. That is, we present a biologically informed CBT for the prevention of the development of AD, i.e., an aspect of applied behavioral neuroscience.
Collapse
|