1
|
Liu D, Lu J, Wei L, Yao M, Yang H, Lv P, Wang H, Zhu Y, Zhu Z, Zhang X, Chen J, Yang QX, Zhang B. Olfactory deficit: a potential functional marker across the Alzheimer's disease continuum. Front Neurosci 2024; 18:1309482. [PMID: 38435057 PMCID: PMC10907997 DOI: 10.3389/fnins.2024.1309482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 02/02/2024] [Indexed: 03/05/2024] Open
Abstract
Alzheimer's disease (AD) is a prevalent form of dementia that affects an estimated 32 million individuals globally. Identifying early indicators is vital for screening at-risk populations and implementing timely interventions. At present, there is an urgent need for early and sensitive biomarkers to screen individuals at risk of AD. Among all sensory biomarkers, olfaction is currently one of the most promising indicators for AD. Olfactory dysfunction signifies a decline in the ability to detect, identify, or remember odors. Within the spectrum of AD, impairment in olfactory identification precedes detectable cognitive impairments, including mild cognitive impairment (MCI) and even the stage of subjective cognitive decline (SCD), by several years. Olfactory impairment is closely linked to the clinical symptoms and neuropathological biomarkers of AD, accompanied by significant structural and functional abnormalities in the brain. Olfactory behavior examination can subjectively evaluate the abilities of olfactory identification, threshold, and discrimination. Olfactory functional magnetic resonance imaging (fMRI) can provide a relatively objective assessment of olfactory capabilities, with the potential to become a promising tool for exploring the neural mechanisms of olfactory damage in AD. Here, we provide a timely review of recent literature on the characteristics, neuropathology, and examination of olfactory dysfunction in the AD continuum. We focus on the early changes in olfactory indicators detected by behavioral and fMRI assessments and discuss the potential of these techniques in MCI and preclinical AD. Despite the challenges and limitations of existing research, olfactory dysfunction has demonstrated its value in assessing neurodegenerative diseases and may serve as an early indicator of AD in the future.
Collapse
Affiliation(s)
- Dongming Liu
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Institute of Medical Imaging and Artificial Intelligence, Nanjing University, Nanjing, China
- Medical Imaging Center, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jiaming Lu
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Institute of Medical Imaging and Artificial Intelligence, Nanjing University, Nanjing, China
- Medical Imaging Center, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Liangpeng Wei
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Institute of Medical Imaging and Artificial Intelligence, Nanjing University, Nanjing, China
- Medical Imaging Center, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Mei Yao
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Institute of Medical Imaging and Artificial Intelligence, Nanjing University, Nanjing, China
- Medical Imaging Center, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Huiquan Yang
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Institute of Medical Imaging and Artificial Intelligence, Nanjing University, Nanjing, China
- Medical Imaging Center, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Pin Lv
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Institute of Medical Imaging and Artificial Intelligence, Nanjing University, Nanjing, China
- Medical Imaging Center, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Haoyao Wang
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Institute of Medical Imaging and Artificial Intelligence, Nanjing University, Nanjing, China
- Medical Imaging Center, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yajing Zhu
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zhengyang Zhu
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xin Zhang
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Institute of Medical Imaging and Artificial Intelligence, Nanjing University, Nanjing, China
- Medical Imaging Center, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jiu Chen
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Institute of Medical Imaging and Artificial Intelligence, Nanjing University, Nanjing, China
- Medical Imaging Center, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Qing X. Yang
- Department of Radiology, Center for NMR Research, Penn State University College of Medicine, Hershey, PA, United States
| | - Bing Zhang
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Institute of Medical Imaging and Artificial Intelligence, Nanjing University, Nanjing, China
- Medical Imaging Center, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing, China
- Institute of Brain Science, Nanjing University, Nanjing, China
- Jiangsu Provincial Medical Key Discipline (Laboratory), Nanjing, China
| |
Collapse
|
2
|
Alotaibi M, Lessard-Beaudoin M, Busch K, Loudghi A, Gaudreau P, Graham RK. Olfactory Dysfunction Associated with Cognitive Decline in an Elderly Population. Exp Aging Res 2024; 50:1-16. [PMID: 36545820 DOI: 10.1080/0361073x.2022.2160597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND In many neurological disorders, including Alzheimer disease, early olfactory dysfunction is observed. OBJECTIVE In order to determine if deficits in olfactory memory are present in the elderly and if olfactory dysfunction correlates with cognitive impairment in the aging population, olfactory testing has been done on seniors from the NuAge cohort accepting to participate in the Olfactory Response Cognition and Aging (ORCA) secondary sub-study. The t-Mini Mental Statement Examination and the Telephone Interview for Cognitive Status tests were done to assess cognition levels. RESULTS Overall, 94% of the ORCA cohort displayed olfactory dysfunction. Deficits in olfactory memory were also present. A correlation was observed between olfactory function and cognitive test scores. Moreover, in women who smoked, there was an association between olfactory memory and cognitive scores. CONCLUSION Our results suggest that olfactory dysfunction may predict impending cognitive decline and highlights the need for olfactory training in seniors to improve olfaction and overall well-being.
Collapse
Affiliation(s)
- Majed Alotaibi
- King Abdullah International Medical Research Center, KSAU-HS, Riyadh, Saudi Arabia
- Department of Neuroscience, The University of Sheffield, Sheffiled, UK
- Research Centre on Aging CIUSSS de l'Estrie - CHUS, Sherbrooke, Quebec, Canada
| | - Melissa Lessard-Beaudoin
- Research Centre on Aging CIUSSS de l'Estrie - CHUS, Sherbrooke, Quebec, Canada
- Department of Pharmacology and Physiology, University of Sherbrooke, Sherbrooke, Canada
| | - Kate Busch
- Research Centre on Aging CIUSSS de l'Estrie - CHUS, Sherbrooke, Quebec, Canada
| | - Amal Loudghi
- Research Centre on Aging CIUSSS de l'Estrie - CHUS, Sherbrooke, Quebec, Canada
- Department of Pharmacology and Physiology, University of Sherbrooke, Sherbrooke, Canada
| | - Pierrette Gaudreau
- Department of Medicine, University of Montreal, Quebec, Canada
- Department of Biochemistry and Molecular Medicine, Centre Hospitalier de l'Université de Montréal Research Center, Montreal, Quebec, Canada
| | - Rona K Graham
- Research Centre on Aging CIUSSS de l'Estrie - CHUS, Sherbrooke, Quebec, Canada
- Department of Pharmacology and Physiology, University of Sherbrooke, Sherbrooke, Canada
| |
Collapse
|
3
|
Woo CC, Miranda B, Sathishkumar M, Dehkordi-Vakil F, Yassa MA, Leon M. Overnight olfactory enrichment using an odorant diffuser improves memory and modifies the uncinate fasciculus in older adults. Front Neurosci 2023; 17:1200448. [PMID: 37554295 PMCID: PMC10405466 DOI: 10.3389/fnins.2023.1200448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 07/07/2023] [Indexed: 08/10/2023] Open
Abstract
OBJECTIVE Cognitive loss in older adults is a growing issue in our society, and there is a need to develop inexpensive, simple, effective in-home treatments. This study was conducted to explore the use of olfactory enrichment at night to improve cognitive ability in healthy older adults. METHODS Male and female older adults (N = 43), age 60-85, were enrolled in the study and randomly assigned to an Olfactory Enriched or Control group. Individuals in the enriched group were exposed to 7 different odorants a week, one per night, for 2 h, using an odorant diffuser. Individuals in the control group had the same experience with de minimis amounts of odorant. Neuropsychological assessments and fMRI scans were administered at the beginning of the study and after 6 months. RESULTS A statistically significant 226% improvement was observed in the enriched group compared to the control group on the Rey Auditory Verbal Learning Test and improved functioning was observed in the left uncinate fasciculus, as assessed by mean diffusivity. CONCLUSION Minimal olfactory enrichment administered at night produces improvements in both cognitive and neural functioning. Thus, olfactory enrichment may provide an effective and low-effort pathway to improved brain health.
Collapse
Affiliation(s)
- Cynthia C. Woo
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, United States
| | - Blake Miranda
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, United States
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, CA, United States
| | - Mithra Sathishkumar
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, United States
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, CA, United States
| | | | - Michael A. Yassa
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, United States
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, CA, United States
| | - Michael Leon
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, United States
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, CA, United States
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
4
|
Brunert D, Quintela RM, Rothermel M. The anterior olfactory nucleus revisited - an emerging role for neuropathological conditions? Prog Neurobiol 2023:102486. [PMID: 37343762 DOI: 10.1016/j.pneurobio.2023.102486] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 06/14/2023] [Accepted: 06/16/2023] [Indexed: 06/23/2023]
Abstract
Olfaction is an important sensory modality for many species and greatly influences animal and human behavior. Still, much about olfactory perception remains unknown. The anterior olfactory nucleus is one of the brain's central early olfactory processing areas. Located directly posterior to the olfactory bulb in the olfactory peduncle with extensive in- and output connections and unique cellular composition, it connects olfactory processing centers of the left and right hemispheres. Almost 20 years have passed since the last comprehensive review on the anterior olfactory nucleus has been published and significant advances regarding its anatomy, function, and pathophysiology have been made in the meantime. Here we briefly summarize previous knowledge on the anterior olfactory nucleus, give detailed insights into the progress that has been made in recent years, and map out its emerging importance in translational research of neurological diseases.
Collapse
Affiliation(s)
- Daniela Brunert
- Institute of Physiology, Medical Faculty, Otto-von-Guericke-University, 39120 Magdeburg, Germany
| | | | - Markus Rothermel
- Institute of Physiology, Medical Faculty, Otto-von-Guericke-University, 39120 Magdeburg, Germany.
| |
Collapse
|
5
|
Neurons, Nose, and Neurodegenerative Diseases: Olfactory Function and Cognitive Impairment. Int J Mol Sci 2023; 24:ijms24032117. [PMID: 36768440 PMCID: PMC9916823 DOI: 10.3390/ijms24032117] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/16/2023] [Accepted: 01/17/2023] [Indexed: 01/24/2023] Open
Abstract
Olfactory capacity declines with aging, but increasing evidence shows that smell dysfunction is one of the early signs of prodromal neurodegenerative diseases such as Alzheimer's and Parkinson's disease. The study of olfactory ability and its role in neurodegenerative diseases arouses much interest in the scientific community. In neurology, olfactory impairment is a potential early marker for the onset of neurodegenerative diseases, but the underlying mechanism is poorly understood. The loss of smell is considered a clinical sign of early-stage disease and a marker of the disease's progression and cognitive impairment. Highlighting the importance of biological bases of smell and molecular pathways could be fundamental to improve neuroprotective and therapeutic strategies. We focused on the review articles and meta-analyses on olfactory and cognitive impairment. We depicted the neurobiology of olfaction and the most common olfactory tests in neurodegenerative diseases. In addition, we underlined the close relationship between the olfactory and cognitive deficit due to nasal neuroepithelium, which is a direct extension of the CNS in communication with the external environment. Neurons, Nose, and Neurodegenerative diseases highlights the role of olfactory dysfunction as a clinical marker for early stages of neurodegenerative diseases when it is associated with molecular, clinical, and neuropathological correlations.
Collapse
|
6
|
Tsui KC, Roy J, Chau SC, Wong KH, Shi L, Poon CH, Wang Y, Strekalova T, Aquili L, Chang RCC, Fung ML, Song YQ, Lim LW. Distribution and inter-regional relationship of amyloid-beta plaque deposition in a 5xFAD mouse model of Alzheimer’s disease. Front Aging Neurosci 2022; 14:964336. [PMID: 35966777 PMCID: PMC9371463 DOI: 10.3389/fnagi.2022.964336] [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: 06/08/2022] [Accepted: 06/28/2022] [Indexed: 11/13/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common form of dementia. Although previous studies have selectively investigated the localization of amyloid-beta (Aβ) deposition in certain brain regions, a comprehensive characterization of the rostro-caudal distribution of Aβ plaques in the brain and their inter-regional correlation remain unexplored. Our results demonstrated remarkable working and spatial memory deficits in 9-month-old 5xFAD mice compared to wildtype mice. High Aβ plaque load was detected in the somatosensory cortex, piriform cortex, thalamus, and dorsal/ventral hippocampus; moderate levels of Aβ plaques were observed in the motor cortex, orbital cortex, visual cortex, and retrosplenial dysgranular cortex; and low levels of Aβ plaques were located in the amygdala, and the cerebellum; but no Aβ plaques were found in the hypothalamus, raphe nuclei, vestibular nucleus, and cuneate nucleus. Interestingly, the deposition of Aβ plaques was positively associated with brain inter-regions including the prefrontal cortex, somatosensory cortex, medial amygdala, thalamus, and the hippocampus. In conclusion, this study provides a comprehensive morphological profile of Aβ deposition in the brain and its inter-regional correlation. This suggests an association between Aβ plaque deposition and specific brain regions in AD pathogenesis.
Collapse
Affiliation(s)
- Ka Chun Tsui
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Jaydeep Roy
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Sze Chun Chau
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Kah Hui Wong
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Department of Anatomy, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Lei Shi
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Chi Him Poon
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Yingyi Wang
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Tatyana Strekalova
- Department of Neuroscience, Maastricht University, Maastricht, Netherlands
- Department of Normal Physiology and Laboratory of Psychiatric Neurobiology, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Luca Aquili
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Discipline of Psychology, College of Science, Health, Engineering, and Education, Murdoch University, Perth, WA, Australia
| | - Raymond Chuen-Chung Chang
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Man-Lung Fung
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- *Correspondence: Man-Lung Fung,
| | - You-qiang Song
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- The State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- You-qiang Song,
| | - Lee Wei Lim
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Lee Wei Lim,
| |
Collapse
|
7
|
Non-Invasive Nasal Discharge Fluid and Other Body Fluid Biomarkers in Alzheimer’s Disease. Pharmaceutics 2022; 14:pharmaceutics14081532. [PMID: 35893788 PMCID: PMC9330777 DOI: 10.3390/pharmaceutics14081532] [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: 06/21/2022] [Revised: 07/12/2022] [Accepted: 07/19/2022] [Indexed: 02/04/2023] Open
Abstract
The key to current Alzheimer’s disease (AD) therapy is the early diagnosis for prompt intervention, since available treatments only slow the disease progression. Therefore, this lack of promising therapies has called for diagnostic screening tests to identify those likely to develop full-blown AD. Recent AD diagnosis guidelines incorporated core biomarker analyses into criteria, including amyloid-β (Aβ), total-tau (T-tau), and phosphorylated tau (P-tau). Though effective, the accessibility of screening tests involving conventional cerebrospinal fluid (CSF)- and blood-based analyses is often hindered by the invasiveness and high cost. In an attempt to overcome these shortcomings, biomarker profiling research using non-invasive body fluid has shown the potential to capture the pathological changes in the patients’ bodies. These novel non-invasive body fluid biomarkers for AD have emerged as diagnostic and pathological targets. Here, we review the potential peripheral biomarkers, including non-invasive peripheral body fluids of nasal discharge, tear, saliva, and urine for AD.
Collapse
|
8
|
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
|
9
|
Salimi M, Tabasi F, Abdolsamadi M, Dehghan S, Dehdar K, Nazari M, Javan M, Mirnajafi-Zadeh J, Raoufy MR. Disrupted connectivity in the olfactory bulb-entorhinal cortex-dorsal hippocampus circuit is associated with recognition memory deficit in Alzheimer's disease model. Sci Rep 2022; 12:4394. [PMID: 35292712 PMCID: PMC8924156 DOI: 10.1038/s41598-022-08528-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 03/02/2022] [Indexed: 12/18/2022] Open
Abstract
Neural synchrony in brain circuits is the mainstay of cognition, including memory processes. Alzheimer's disease (AD) is a progressive neurodegenerative disorder that disrupts neural synchrony in specific circuits, associated with memory dysfunction before a substantial neural loss. Recognition memory impairment is a prominent cognitive symptom in the early stages of AD. The entorhinal-hippocampal circuit is critically engaged in recognition memory and is known as one of the earliest circuits involved due to AD pathology. Notably, the olfactory bulb is closely connected with the entorhinal-hippocampal circuit and is suggested as one of the earliest regions affected by AD. Therefore, we recorded simultaneous local field potential from the olfactory bulb (OB), entorhinal cortex (EC), and dorsal hippocampus (dHPC) to explore the functional connectivity in the OB-EC-dHPC circuit during novel object recognition (NOR) task performance in a rat model of AD. Animals that received amyloid-beta (Aβ) showed a significant impairment in task performance and a marked reduction in OB survived cells. We revealed that Aβ reduced coherence and synchrony in the OB-EC-dHPC circuit at theta and gamma bands during NOR performance. Importantly, our results exhibit that disrupted functional connectivity in the OB-EC-dHPC circuit was correlated with impaired recognition memory induced by Aβ. These findings can elucidate dynamic changes in neural activities underlying AD, helping to find novel diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Morteza Salimi
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, 1411713116, Iran
| | - Farhad Tabasi
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, 1411713116, Iran
- Faculty of Medical Sciences, Institute for Brain Sciences and Cognition, Tarbiat Modares University, Tehran, Iran
| | - Maryam Abdolsamadi
- Department of Mathematics, Faculty of Science, Islamic Azad University, North Tehran Branch, Tehran, Iran
| | - Samaneh Dehghan
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
- Eye Research Center, The Five Senses Institute, Rassoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Kolsoum Dehdar
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, 1411713116, Iran
- Faculty of Medical Sciences, Institute for Brain Sciences and Cognition, Tarbiat Modares University, Tehran, Iran
| | - Milad Nazari
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
- DANDRITE, The Danish Research Institute of Translational Neuroscience, Aarhus University, Aarhus, Denmark
| | - Mohammad Javan
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, 1411713116, Iran
- Faculty of Medical Sciences, Institute for Brain Sciences and Cognition, Tarbiat Modares University, Tehran, Iran
| | - Javad Mirnajafi-Zadeh
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, 1411713116, Iran
- Faculty of Medical Sciences, Institute for Brain Sciences and Cognition, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Reza Raoufy
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, 1411713116, Iran.
- Faculty of Medical Sciences, Institute for Brain Sciences and Cognition, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
10
|
Dong Y, Wang Y, Liu K, Hou T, Han X, Cong L, Ren Y, Zhang Q, Tang S, Ekström I, Laukka EJ, Du Y, Qiu C. Dementia screening in rural-dwelling Chinese older adults: The utility of a smell test and the self-rated AD8. J Am Geriatr Soc 2021; 70:1106-1116. [PMID: 34874063 DOI: 10.1111/jgs.17586] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 10/05/2021] [Accepted: 11/03/2021] [Indexed: 02/01/2023]
Abstract
BACKGROUND Olfactory impairment is associated with dementia in clinical settings. We examined the relationship of olfactory identification function with all-cause dementia, Alzheimer's disease (AD), and vascular dementia (VaD) and assessed the discriminative ability of the Sniffin' Sticks Identification Test (SSIT), the self-rated Ascertain Dementia 8-item Questionnaire (AD8), and their combination for dementia detection among rural-dwelling older adults in China. METHODS This population-based cross-sectional study included 4481 participants (age ≥ 65 years; 56.8% women; 38.1% illiteracy) living in rural communities. The 16-item SSIT was performed to assess olfactory identification function. The self-rated AD8 was administered to participants for cognitive status. We diagnosed dementia, AD, and VaD following the international criteria. Data were analyzed with logistic regression models and receiver operating characteristic curve. RESULTS Of the 4481 participants, dementia was diagnosed in 139 persons (3.1%), including 92 with AD and 42 with VaD. The SSIT score (range, 0-16) was associated with multiadjusted odds ratios of 0.83 (95% CI: 0.79-0.88) for dementia, 0.84 (0.79-0.90) for AD, and 0.79 (0.71-0.87) for VaD. The area under the curve for the discrimination between participants with and without dementia was 0.73 (95% CI: 0.69-0.77) for SSIT score ≤ 8 alone, 0.86 (0.82-0.89) for self-rated AD8 score ≥ 3 alone, and 0.89 (0.86-0.92) for their combination using a logistic model. CONCLUSIONS Olfactory impairment is a clinical marker for all-cause dementia, AD, and VaD. The smell identification test, in combination with the brief self-rated cognitive screening tool, is accurate for screening dementia among rural-dwelling Chinese older adults with no or limited education.
Collapse
Affiliation(s)
- Yi Dong
- Department of Neurology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yongxiang Wang
- Department of Neurology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Keke Liu
- Department of Neurology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Tingting Hou
- Department of Neurology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xiaolei Han
- Department of Neurology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lin Cong
- Department of Neurology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yifei Ren
- Department of Neurology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qinghua Zhang
- Department of Neurology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Shi Tang
- Department of Neurology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Ingrid Ekström
- Aging Research Center and Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet-Stockholm University, Stockholm, Sweden
| | - Erika J Laukka
- Aging Research Center and Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet-Stockholm University, Stockholm, Sweden
| | - Yifeng Du
- Department of Neurology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Chengxuan Qiu
- Department of Neurology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Aging Research Center and Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet-Stockholm University, Stockholm, Sweden
| |
Collapse
|
11
|
The Olfactory System as Marker of Neurodegeneration in Aging, Neurological and Neuropsychiatric Disorders. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18136976. [PMID: 34209997 PMCID: PMC8297221 DOI: 10.3390/ijerph18136976] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 06/19/2021] [Accepted: 06/24/2021] [Indexed: 12/14/2022]
Abstract
Research studies that focus on understanding the onset of neurodegenerative pathology and therapeutic interventions to inhibit its causative factors, have shown a crucial role of olfactory bulb neurons as they transmit and propagate nerve impulses to higher cortical and limbic structures. In rodent models, removal of the olfactory bulb results in pathology of the frontal cortex that shows striking similarity with frontal cortex features of patients diagnosed with neurodegenerative disorders. Widely different approaches involving behavioral symptom analysis, histopathological and molecular alterations, genetic and environmental influences, along with age-related alterations in cellular pathways, indicate a strong correlation of olfactory dysfunction and neurodegeneration. Indeed, declining olfactory acuity and olfactory deficits emerge either as the very first symptoms or as prodromal symptoms of progressing neurodegeneration of classical conditions. Olfactory dysfunction has been associated with most neurodegenerative, neuropsychiatric, and communication disorders. Evidence revealing the dual molecular function of the olfactory receptor neurons at dendritic and axonal ends indicates the significance of olfactory processing pathways that come under environmental pressure right from the onset. Here, we review findings that olfactory bulb neuronal processing serves as a marker of neuropsychiatric and neurodegenerative disorders.
Collapse
|
12
|
Heinbockel T, Bhatia-Dey N, Shields VDC. Endocannabinoid-mediated neuromodulation in the main olfactory bulb at the interface of environmental stimuli and central neural processing. Eur J Neurosci 2021; 55:1002-1014. [PMID: 33724578 DOI: 10.1111/ejn.15186] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 02/10/2021] [Accepted: 03/05/2021] [Indexed: 12/16/2022]
Abstract
The olfactory system has become an important functional gateway to understand and analyze neuromodulation since olfactory dysfunction and deficits have emerged as prodromal and, at other times, as first symptoms of many of neurodegenerative, neuropsychiatric and communication disorders. Considering olfactory dysfunction as outcome of altered, damaged and/or inefficient olfactory processing, in the current review, we analyze how olfactory processing interacts with the endocannabinoid signaling system. In the human body, endocannabinoid synthesis is a natural and on-demand response to a wide range of physiological and environmental stimuli. Our current understanding of the response dynamics of the endocannabinoid system is based in large part on research advances in limbic system areas, such as the hippocampus and the amygdala. Functional interactions of this signaling system with olfactory processing and associated pathways are just emerging but appear to grow rapidly with multidimensional approaches. Recent work analyzing the crystal structure of endocannabinoid receptors bound to their agonists in a signaling complex has opened avenues for developing specific therapeutic drugs that could help with neuroinflammation, neurodegeneration, and alleviation/reduction of pain. We discuss the role of endocannabinoids as signaling molecules in the olfactory system and the relevance of the endocannabinoid system for synaptic plasticity.
Collapse
Affiliation(s)
- Thomas Heinbockel
- Department of Anatomy, Howard University College of Medicine, Washington, DC, USA
| | - Naina Bhatia-Dey
- Department of Anatomy, Howard University College of Medicine, Washington, DC, USA
| | - Vonnie D C Shields
- Biological Sciences Department, Fisher College of Science and Mathematics, Towson University, Towson, MD, USA
| |
Collapse
|
13
|
Gao K, Gao F, Li J, He C, Liu M, Zhu Q, Qian Z, Ma T, Wang P. Biomimetic integrated olfactory sensory and olfactory bulb systems in vitro based on a chip. Biosens Bioelectron 2021; 171:112739. [PMID: 33096431 DOI: 10.1016/j.bios.2020.112739] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 10/14/2020] [Accepted: 10/16/2020] [Indexed: 11/28/2022]
Abstract
A variety of mammalian or insect behaviors rely on the recognition of relevant odor stimuli. The olfactory system detects and translates complex olfactory stimuli (odors) through the unique and reproducible dynamic ensembles of neuronal activities. This process is involved in various types of neurons of olfactory parts, thereby encoding olfactory information or predicting progression in some neuropsychiatric diseases. In this paper, we constructed a biomimetic model including olfactory sensing system and olfactory bulb processing system to map olfactory-associated ensembles of neuronal activity. The olfactory receptor neurons (ORNs) and olfactory bulb (OB) neurons were primarily cultured and the immunofluorescence images were performed to identify the types of neurons. Diacetyl solution was used as an odor stimulus, and the spike bursts and random spike firing patterns of concentration-dependent excitatory responses were obtained from the ORNs network. The spike waveform and feature parameters were extracted including the spike number and interval in per burst to program the stimulation unit and sequences. The sequences containing odor information were applied to the OB neuronal network for the simulation of the primary olfactory processing. The response pattern and change rule of the OB neuronal network were consistent with the OB neurons affected by the neurotransmitter, which is the carrier of olfactory information transmission in vivo. This biomimetic integrated olfactory sensory and processing system can serve as a novel model for studying the physiological and pathological mechanisms of olfaction, and the pharmacological application in vitro.
Collapse
Affiliation(s)
- Keqiang Gao
- School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Fan Gao
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, China
| | - Jiaxin Li
- School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Chuanjiang He
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou, Zhejiang, 310027, China
| | - Mengxue Liu
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou, Zhejiang, 310027, China
| | - Qiaoqiao Zhu
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, China
| | - Zhiyu Qian
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, China
| | - Tengfei Ma
- School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China.
| | - Ping Wang
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou, Zhejiang, 310027, China.
| |
Collapse
|
14
|
Tu L, Lv X, Fan Z, Zhang M, Wang H, Yu X. Association of Odor Identification Ability With Amyloid-β and Tau Burden: A Systematic Review and Meta-Analysis. Front Neurosci 2020; 14:586330. [PMID: 33324151 PMCID: PMC7726324 DOI: 10.3389/fnins.2020.586330] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 10/27/2020] [Indexed: 01/09/2023] Open
Abstract
Background: The associations between olfactory identification (OI) ability and the Alzheimer's disease biomarkers were not clear. Objective: This meta-analysis aimed to examine the associations between OI and Aβ and tau burden. Methods: Electronic databases (PubMed, Embase, PsycINFO, and Google Scholar) were searched until June 2019 to identify studies that reported correlation coefficients or regression coefficients between OI and Aβ or tau levels measured by positron emission tomography (PET) or cerebrospinal fluid (CSF). Pooled Pearson correlation coefficients were computed for the PET imaging and CSF biomarkers, with subgroup analysis for subjects classified into different groups. Results: Nine studies met the inclusion criteria. Of these, five studies (N = 494) involved Aβ PET, one involved tau PET (N = 26), and four involved CSF Aβ or tau (N = 345). OI was negatively associated with Aβ PET in the mixed (r = -0.25, P = 0.008) and cognitively normal groups (r = -0.15, P = 0.004) but not in the mild cognitive impairment group. A similar association with CSF total tau in the mixed group was also observed. No association was found between OI and CSF phosphorylated tau or Aβ42 in the subgroup analysis of the CSF biomarkers. Due to a lack of data, no pooled r value could be computed for the association between the OI and tau PET. Conclusion: The associations between OI ability and Aβ and CSF tau burden in older adults are negligible. While current evidence does not support the association, further studies using PET tau imaging are warranted.
Collapse
Affiliation(s)
- Lihui Tu
- Dementia Care and Research Center, Clinical Research Division, Peking University Institute of Mental Health (Sixth Hospital), Beijing, China
- Beijing Dementia Key Lab, National Clinical Research Center for Mental Disorders, Key Laboratory of Mental Health, Ministry of Health Peking University, Beijing, China
| | - Xiaozhen Lv
- Dementia Care and Research Center, Clinical Research Division, Peking University Institute of Mental Health (Sixth Hospital), Beijing, China
- Beijing Dementia Key Lab, National Clinical Research Center for Mental Disorders, Key Laboratory of Mental Health, Ministry of Health Peking University, Beijing, China
| | - Zili Fan
- Dementia Care and Research Center, Clinical Research Division, Peking University Institute of Mental Health (Sixth Hospital), Beijing, China
- Beijing Dementia Key Lab, National Clinical Research Center for Mental Disorders, Key Laboratory of Mental Health, Ministry of Health Peking University, Beijing, China
| | - Ming Zhang
- Dementia Care and Research Center, Clinical Research Division, Peking University Institute of Mental Health (Sixth Hospital), Beijing, China
- Beijing Dementia Key Lab, National Clinical Research Center for Mental Disorders, Key Laboratory of Mental Health, Ministry of Health Peking University, Beijing, China
- Department of Psychiatry, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Huali Wang
- Dementia Care and Research Center, Clinical Research Division, Peking University Institute of Mental Health (Sixth Hospital), Beijing, China
- Beijing Dementia Key Lab, National Clinical Research Center for Mental Disorders, Key Laboratory of Mental Health, Ministry of Health Peking University, Beijing, China
| | - Xin Yu
- Dementia Care and Research Center, Clinical Research Division, Peking University Institute of Mental Health (Sixth Hospital), Beijing, China
- Beijing Dementia Key Lab, National Clinical Research Center for Mental Disorders, Key Laboratory of Mental Health, Ministry of Health Peking University, Beijing, China
| |
Collapse
|
15
|
Chuang HC, Chen HC, Chai PJ, Liao HT, Wu CF, Chen CL, Jhan MK, Hsieh HI, Wu KY, Chen TF, Cheng TJ. Neuropathology changed by 3- and 6-months low-level PM 2.5 inhalation exposure in spontaneously hypertensive rats. Part Fibre Toxicol 2020; 17:59. [PMID: 33243264 PMCID: PMC7691081 DOI: 10.1186/s12989-020-00388-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 10/30/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Epidemiological evidence has linked fine particulate matter (PM2.5) to neurodegenerative diseases; however, the toxicological evidence remains unclear. The objective of this study was to investigate the effects of PM2.5 on neuropathophysiology in a hypertensive animal model. We examined behavioral alterations (Morris water maze), lipid peroxidation (malondialdehyde (MDA)), tau and autophagy expressions, neuron death, and caspase-3 levels after 3 and 6 months of whole-body exposure to urban PM2.5 in spontaneously hypertensive (SH) rats. RESULTS SH rats were exposed to S-, K-, Si-, and Fe-dominated PM2.5 at 8.6 ± 2.5 and 10.8 ± 3.8 μg/m3 for 3 and 6 months, respectively. We observed no significant alterations in the escape latency, distance moved, mean area crossing, mean time spent, or mean swimming velocity after PM2.5 exposure. Notably, levels of MDA had significantly increased in the olfactory bulb, hippocampus, and cortex after 6 months of PM2.5 exposure (p < 0.05). We observed that 3 months of exposure to PM2.5 caused significantly higher expressions of t-tau and p-tau in the olfactory bulb (p < 0.05) but not in other brain regions. Beclin 1 was overexpressed in the hippocampus with 3 months of PM2.5 exposure, but significantly decreased in the cortex with 6 months exposure to PM2.5. Neuron numbers had decreased with caspase-3 activation in the cerebellum, hippocampus, and cortex after 6 months of PM2.5 exposure. CONCLUSIONS Chronic exposure to low-level PM2.5 could accelerate the development of neurodegenerative pathologies in subjects with hypertension.
Collapse
Affiliation(s)
- Hsiao-Chi Chuang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Hsin-Chang Chen
- Institute of Food Safety and Health, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Pei-Jui Chai
- Institute of Environmental and Occupational Health Science, College of Public Health, National Taiwan University, 17 Xu-Zhou Road, Taipei, 100 Taiwan
| | - Ho-Tang Liao
- Institute of Environmental and Occupational Health Science, College of Public Health, National Taiwan University, 17 Xu-Zhou Road, Taipei, 100 Taiwan
| | - Chang-Fu Wu
- Institute of Environmental and Occupational Health Science, College of Public Health, National Taiwan University, 17 Xu-Zhou Road, Taipei, 100 Taiwan
- Department of Public Health, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Chia-Ling Chen
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ming-Kai Jhan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hui-I Hsieh
- Department of Occupational Medicine, Cathay General Hospital, Taipei, Taiwan
| | - Kuen-Yuh Wu
- Institute of Environmental and Occupational Health Science, College of Public Health, National Taiwan University, 17 Xu-Zhou Road, Taipei, 100 Taiwan
| | - Ta-Fu Chen
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, No. 1, Changde Street, Taipei, 10048 Taiwan
| | - Tsun-Jen Cheng
- Institute of Environmental and Occupational Health Science, College of Public Health, National Taiwan University, 17 Xu-Zhou Road, Taipei, 100 Taiwan
- Department of Public Health, College of Public Health, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
16
|
Mathew D. Loss of Smell in COVID-19 Patients: Lessons and Opportunities. Front Hum Neurosci 2020; 14:598465. [PMID: 33328942 PMCID: PMC7732576 DOI: 10.3389/fnhum.2020.598465] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 11/04/2020] [Indexed: 11/25/2022] Open
Affiliation(s)
- Dennis Mathew
- Department of Biology, University of Nevada, Reno, NV, United States
| |
Collapse
|
17
|
Rebholz H, Braun RJ, Ladage D, Knoll W, Kleber C, Hassel AW. Loss of Olfactory Function-Early Indicator for Covid-19, Other Viral Infections and Neurodegenerative Disorders. Front Neurol 2020; 11:569333. [PMID: 33193009 PMCID: PMC7649754 DOI: 10.3389/fneur.2020.569333] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 09/15/2020] [Indexed: 12/15/2022] Open
Abstract
The loss of the senses of smell (anosmia) and taste (ageusia) are rather common disorders, affecting up to 20% of the adult population. Yet, this condition has not received the attention it deserves, most probably because per se such a disorder is not life threatening. However, loss of olfactory function significantly reduces the quality of life of the affected patients, leading to dislike in food and insufficient, exaggerated or unbalanced food intake, unintentional exposure to toxins such as household gas, social isolation, depression, and an overall insecurity. Not only is olfactory dysfunction rather prevalent in the healthy population, it is, in many instances, also a correlate or an early indicator of a panoply of diseases. Importantly, olfactory dysfunction is linked to the two most prominent neurodegenerative disorders, Parkinson's disease and Alzheimer's disease. Anosmia and hyposmia (reduced sense of smell) affect a majority of patients years before the onset of cognitive or motor symptoms, establishing olfactory dysfunction as early biomarker that can enable earlier diagnosis and preventative treatments. In the current health crisis caused by SARS-CoV2, anosmia and dysgeusia as early-onset symptoms in virus-positive patients may prove to be highly relevant and crucial for pre-symptomatic Covid-19 detection from a public health perspective, preceding by days the more classical respiratory tract symptoms such as cough, tightness of the chest or fever. Thus, the olfactory system seems to be at the frontline of pathologic assault, be it through pathogens or insults that can lead to or at least associate with neurodegeneration. The aim of this review is to assemble current knowledge from different medical fields that all share a common denominator, olfactory/gustatory dysfunction, and to distill overarching etiologies and disease progression mechanisms.
Collapse
Affiliation(s)
- Heike Rebholz
- Center of Neurodegeneration, Faculty of Medicine/Dental Medicine, Danube Private University, Krems, Austria
- Institut de Psychiatrie et Neurosciences de Paris (IPNP), UMR S1266, INSERM, Université de Paris, Paris, France
- GHU Psychiatrie et Neurosciences, Paris, France
| | - Ralf J. Braun
- Center of Neurodegeneration, Faculty of Medicine/Dental Medicine, Danube Private University, Krems, Austria
| | - Dennis Ladage
- Center of Chemistry and Physics of Materials, Faculty of Medicine/Dental Medicine, Danube Private University, Krems, Austria
- Universitaetsklinikum Köln, Cologne, Germany
| | | | - Christoph Kleber
- Center of Chemistry and Physics of Materials, Faculty of Medicine/Dental Medicine, Danube Private University, Krems, Austria
- Institute of Chemical Technology of Inorganic Materials, Johannes Kepler University Linz, Linz, Austria
| | - Achim W. Hassel
- Institute of Chemical Technology of Inorganic Materials, Johannes Kepler University Linz, Linz, Austria
| |
Collapse
|
18
|
Dibattista M, Pifferi S, Menini A, Reisert J. Alzheimer's Disease: What Can We Learn From the Peripheral Olfactory System? Front Neurosci 2020; 14:440. [PMID: 32508565 PMCID: PMC7248389 DOI: 10.3389/fnins.2020.00440] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 04/09/2020] [Indexed: 01/01/2023] Open
Abstract
The sense of smell has been shown to deteriorate in patients with some neurodegenerative disorders. In Parkinson's disease (PD) and Alzheimer's disease (AD), decreased ability to smell is associated with early disease stages. Thus, olfactory neurons in the nose and olfactory bulb (OB) may provide a window into brain physiology and pathophysiology to address the pathogenesis of neurodegenerative diseases. Because nasal olfactory receptor neurons regenerate throughout life, the olfactory system offers a broad variety of cellular mechanisms that could be altered in AD, including odorant receptor expression, neurogenesis and neurodegeneration in the olfactory epithelium, axonal targeting to the OB, and synaptogenesis and neurogenesis in the OB. This review focuses on pathophysiological changes in the periphery of the olfactory system during the progression of AD in mice, highlighting how the olfactory epithelium and the OB are particularly sensitive to changes in proteins and enzymes involved in AD pathogenesis. Evidence reviewed here in the context of the emergence of other typical pathological changes in AD suggests that olfactory impairments could be used to understand the molecular mechanisms involved in the early phases of the pathology.
Collapse
Affiliation(s)
- Michele Dibattista
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, University of Bari A. Moro, Bari, Italy
| | - Simone Pifferi
- Neurobiology Group, SISSA, Scuola Internazionale Superiore di Studi Avanzati, Trieste, Italy
| | - Anna Menini
- Neurobiology Group, SISSA, Scuola Internazionale Superiore di Studi Avanzati, Trieste, Italy
| | | |
Collapse
|
19
|
Joshi S, Bayat A, Jones A, Xiao X, Koubeissi MZ. The effects of ammonia stimulation on kainate-induced status epilepticus and anterior piriform cortex electrophysiology. Epilepsy Behav 2020; 104:106885. [PMID: 31935647 DOI: 10.1016/j.yebeh.2019.106885] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 12/19/2019] [Accepted: 12/19/2019] [Indexed: 11/17/2022]
Abstract
OBJECTIVE Strong olfactory stimulation (OS) with such substances as toluene or ammonia has been reported to suppress seizures. We aimed to investigate the role of ammonia stimulation on acute kainic acid (KA)-induced seizures. We also investigated any possible effects of ammonia stimulation on the electrophysiology of the anterior piriform cortex (APC). METHODS Adult male Sprague-Dawley rats were implanted with bilateral hippocampal electrodes and an electrode in the left APC. Animals were exposed to either distilled water (control) or ammonia stimulation for 20 s every 5 min during KA induction of status epilepticus (SE). The electroencephalogram (EEG) was analyzed for seizure frequency, duration, severity, and total KA doses given prior to reaching SE. Seizure-free EEG epochs that coincided with OS were chosen and analyzed via wavelet analysis for any spectral changes. RESULTS We found no significant differences in seizure frequency, duration, severity, or administered KA doses before SE between the groups. In the experimental group, a wavelet analysis of variance (WANOVA) revealed a significant stimulation-induced increase of power in the delta and alpha bands prior to the first KA injection and higher power in the delta and theta bands after KA injection. CONCLUSIONS Whereas the spectral analysis of the APC revealed specific OS-induced changes, our findings suggest that OS with ammonia does not result in altering the threshold of attaining KA-induced SE. This does not rule out a potential role for OS in reducing recurrent seizures in the KA or other epilepsy models.
Collapse
Affiliation(s)
- Sweta Joshi
- Department of Neurology, George Washington University, 2150 Pennsylvania Ave, NW, Washington, DC 20037, USA
| | - Arezou Bayat
- Department of Neurology, George Washington University, 2150 Pennsylvania Ave, NW, Washington, DC 20037, USA
| | - Andrew Jones
- Translational Health Sciences, George Washington University, 2100 Pennsylvania Ave, NW, Washington, DC 20037, USA
| | - Xiao Xiao
- School of Engineering and Applied Science, George Washington University, 800 22nd St NW, Washington, DC 20052, USA
| | - Mohamad Z Koubeissi
- Department of Neurology, George Washington University, 2150 Pennsylvania Ave, NW, Washington, DC 20037, USA.
| |
Collapse
|
20
|
Leandro GS, Evangelista AF, Lobo RR, Xavier DJ, Moriguti JC, Sakamoto-Hojo ET. Changes in Expression Profiles Revealed by Transcriptomic Analysis in Peripheral Blood Mononuclear Cells of Alzheimer's Disease Patients. J Alzheimers Dis 2019; 66:1483-1495. [PMID: 30400085 DOI: 10.3233/jad-170205] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative pathology associated with accumulation of DNA damage. Inflammation and cell cycle alterations seem to be implicated in the pathogenesis of AD, although the molecular mechanisms have not been thoroughly elucidated to date. The aim of the present study was to evaluate whether peripheral blood mononuclear cells (PBMCs) of AD patients display alterations in gene expression profiles, focusing on finding markers that might improve the diagnosis of AD. Blood samples were collected from 22 AD patients and 13 healthy individuals to perform genome-wide mRNA expression. We found 593 differentially expressed genes in AD compared to controls, from which 428 were upregulated, and 165 were downregulated. By performing a gene set enrichment analysis, we observed pathways involved in inflammation, DNA damage response, cell cycle, and neuronal processes. Moreover, functional annotation analyses indicated that differentially expressed genes are strongly related to pathways associated with the cell cycle and the immune system. The results were compared with those of an independent study on hippocampus samples, and a number of genes in common between both studies were identified as potential peripheral biomarkers for AD, including DUSP1, FOS, SLC7A2, RGS1, GFAP, CCL2, ANGPTL4, and SSPN. Taken together, our results demonstrate that PBMCs of AD patients do present alterations in gene expression profiles, and these results are comparable to those previously reported in the literature for AD neurons, supporting the hypothesis that blood peripheral mononuclear cells express molecular changes that occur in the neurons of AD patients.
Collapse
Affiliation(s)
- Giovana Silva Leandro
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo - USP, Ribeirão Preto, SP, Brazil
| | | | - Romulo Rebouças Lobo
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo - USP, Ribeirão Preto, SP, Brazil
| | - Danilo Jordão Xavier
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo - USP, Ribeirão Preto, SP, Brazil
| | - Julio César Moriguti
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo - USP, Ribeirão Preto, SP, Brazil
| | - Elza Tiemi Sakamoto-Hojo
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo - USP, Ribeirão Preto, SP, Brazil.,Department of Biology, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo - USP, Ribeirão Preto, SP, Brazil
| |
Collapse
|
21
|
Pusil S, Dimitriadis SI, López ME, Pereda E, Maestú F. Aberrant MEG multi-frequency phase temporal synchronization predicts conversion from mild cognitive impairment-to-Alzheimer's disease. Neuroimage Clin 2019; 24:101972. [PMID: 31522127 PMCID: PMC6745514 DOI: 10.1016/j.nicl.2019.101972] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/19/2019] [Accepted: 08/03/2019] [Indexed: 11/15/2022]
Abstract
Many neuroimaging studies focus on a frequency-specific or a multi-frequency network analysis showing that functional brain networks are disrupted in patients with Alzheimer's disease (AD). Although those studies enriched our knowledge of the impact of AD in brain's functionality, our goal is to test the effectiveness of combining neuroimaging with network neuroscience to predict with high accuracy subjects with mild cognitive impairment (MCI) that will convert to AD. In this study, eyes-closed resting-state magnetoencephalography (MEG) recordings from 27 stable MCI (sMCI) and 27 progressive MCI (pMCI) from two scan sessions (baseline and follow-up after approximately 3 years) were projected via beamforming onto an atlas-based set of regions of interest (ROIs). Dynamic functional connectivity networks were constructed independently for the five classical frequency bands while a multivariate phase-based coupling metric was adopted. Thus, computing the distance between the fluctuation of functional strength of every pair of ROIs between the two conditions with dynamic time wrapping (DTW), a large set of features was extracted. A machine learning algorithm revealed 30 DTW-based features in the five frequency bands that can distinguish the sMCI from pMCI with absolute accuracy (100%). Further analysis of the selected links revealed that most of the connected ROIs were part of the default mode network (DMN), the cingulo-opercular (CO), the fronto-parietal and the sensorimotor network. Overall, our dynamic network multi-frequency analysis approach provides an effective framework of constructing a sensitive MEG-based connectome biomarker for the prediction of conversion from MCI to Alzheimer's disease.
Collapse
Affiliation(s)
- Sandra Pusil
- Laboratory of Neuropsychology, University of the Balearic Islands, Spain; Laboratory of Cognitive and Computational Neuroscience, Center for Biomedical Technology, Universidad Complutense and Universidad Politécnica de Madrid, Madrid, Spain.
| | - Stavros I Dimitriadis
- Cardiff University Brain Research Imaging Centre, School of Psychology, Cardiff University, Cardiff, United Kingdom; Neuroinformatics Group, Cardiff University Brain Research Imaging Centre, School of Psychology, Cardiff University, Cardiff, United Kingdom; Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom; School of Psychology, Cardiff University, Cardiff, United Kingdom; Neuroscience and Mental Health Research Institute, School of Medicine, Cardiff University, Cardiff, United Kingdom; MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - María Eugenia López
- Laboratory of Cognitive and Computational Neuroscience, Center for Biomedical Technology, Universidad Complutense and Universidad Politécnica de Madrid, Madrid, Spain; Department of Experimental Psychology, Universidad Complutense de Madrid, Madrid, Spain; Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Ernesto Pereda
- Laboratory of Cognitive and Computational Neuroscience, Center for Biomedical Technology, Universidad Complutense and Universidad Politécnica de Madrid, Madrid, Spain; Electrical Engineering and Bioengineering Lab, Department of Industrial Engineering, IUNE Universidad de La Laguna, Tenerife, Spain
| | - Fernando Maestú
- Laboratory of Cognitive and Computational Neuroscience, Center for Biomedical Technology, Universidad Complutense and Universidad Politécnica de Madrid, Madrid, Spain; Department of Experimental Psychology, Universidad Complutense de Madrid, Madrid, Spain; Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| |
Collapse
|
22
|
Invitto S, Mazzatenta A. Olfactory Event-Related Potentials and Exhaled Organic Volatile Compounds: The Slow Link Between Olfactory Perception and Breath Metabolic Response. A Pilot Study on Phenylethyl Alcohol and Vaseline Oil. Brain Sci 2019; 9:E84. [PMID: 30991670 PMCID: PMC6523942 DOI: 10.3390/brainsci9040084] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 04/08/2019] [Accepted: 04/12/2019] [Indexed: 12/20/2022] Open
Abstract
Olfactory processing starts with the breath and elicits neuronal, metabolic and cortical responses. This process can be investigated centrally via the Olfactory Event-Related Potentials (OERPs) and peripherally via exhaled Volatile Organic Compounds (VOCs). Despite this, the relationship between OERPs (i.e., N1 and Late Positive Component LPC) and exhaled VOCs has not been investigated enough. The aim of this research is to study OERPs and VOCs connection to two different stimuli: phenylethyl alcohol (PEA) and Vaseline Oil (VO). Fifteen healthy subjects performed a perceptual olfactory task with PEA as a smell target stimulus and VO as a neutral stimulus. The results suggest that OERPs and VOCs distributions follow the same amplitude trend and that PEA is highly arousing in both psychophysiological measures. PEA shows ampler and faster N1, a component related to the sensorial aspect of the stimulus. The N1 topographic localization is different between PEA and VO: PEA stimulus evokes greater N1 in the left centroparietal site. LPC, a component elicited by the perceptual characteristic of the stimulus, shows faster latency in the Frontal lobe and decreased amplitude in the Central and Parietal lobe elicited by the PEA smell. Moreover, the delayed time between the onset of N1-LPC and the onset of VOCs seems to be about 3 s. This delay could be identified as the internal metabolic time in which the odorous stimulus, once perceived at the cortical level, is metabolized and subsequently exhaled. Furthermore, the VO stimulus does not allocate the attentive, perceptive and metabolic resource as with PEA.
Collapse
Affiliation(s)
- Sara Invitto
- Department of Biological and Environmental Science and Technologies, University of Salento, Campus Ecotekne, Via per Monteroni, 73100 Lecce, Italy.
- DReAM Laboratory of InterDisciplinary Research Applied to Medicine, University of Salento-Vito Fazzi Hospital, 73100 Lecce, Italy.
| | - Andrea Mazzatenta
- Dipartimento di Neuroscienze, Imaging e Scienze Cliniche, Università "d'Annunzio" di Chieti-Pescara, 66100 Chieti, Italy.
| |
Collapse
|
23
|
Mullane K, Williams M. Preclinical Models of Alzheimer's Disease: Relevance and Translational Validity. ACTA ACUST UNITED AC 2019; 84:e57. [PMID: 30802363 DOI: 10.1002/cpph.57] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The only drugs currently approved for the treatment of Alzheimer's Disease (AD) are four acetylcholinesterase inhibitors and the NMDA antagonist memantine. Apart from these drugs, which have minimal to no clinical benefit, the 40-year search for effective therapeutics to treat AD has resulted in a clinical failure rate of 100% not only for compounds that prevent brain amyloid deposition or remove existing amyloid plaques but also those acting by a variety of other putative disease-associated mechanisms. This indicates that the preclinical data generated from current AD targets to support the selection, optimization, and translation of new chemical entities (NCEs) and biologics to clinical trials is seriously compromised. While many of these failures reflect flawed hypotheses or a lack of adequate characterization of the preclinical pharmacodynamic and pharmacokinetic (PD/PK) properties of lead NCEs-including their bioavailability and toxicity-the conceptualization, validation, and interrogation of the current animal models of AD represent key limitations. The overwhelming majority of these AD models are transgenic, based on aspects of the amyloid hypothesis and the genetics of the familial form of the disease. As a result, these generally lack construct and predictive validity for the sporadic form of the human disease. The 170 or so transgenic models, perhaps the largest number ever focused on a single disease, use rodents, mainly mice, and in addition to amyloid also address aspects of tau causality with more complex multigene models including other presumed causative factors together with amyloid. This overview discusses the current animal models of AD in the context of both the controversies surrounding the causative role of amyloid in the disease and the need to develop validated models of cognitive function/dysfunction that more appropriately reflect the phenotype(s) of human aged-related dementias. © 2019 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
| | - Michael Williams
- Department of Biological Chemistry and Pharmacology, College of Medicine, Ohio State University, Columbus, Ohio
| |
Collapse
|
24
|
Jean L, Brimijoin S, Vaux DJ. In vivo localization of human acetylcholinesterase-derived species in a β-sheet conformation at the core of senile plaques in Alzheimer's disease. J Biol Chem 2019; 294:6253-6272. [PMID: 30787102 DOI: 10.1074/jbc.ra118.006230] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 02/13/2019] [Indexed: 12/22/2022] Open
Abstract
Many neurodegenerative diseases are characterized by amyloid deposition. In Alzheimer's disease (AD), β-amyloid (Aβ) peptides accumulate extracellularly in senile plaques. The AD amyloid cascade hypothesis proposes that Aβ production or reduced clearance leads to toxicity. In contrast, the cholinergic hypothesis argues for a specific pathology of brain cholinergic pathways. However, neither hypothesis in isolation explains the pattern of AD pathogenesis. Evidence suggests that a connection exists between these two scenarios: the synaptic form of human acetylcholinesterase (hAChE-S) associates with plaques in AD brains; among hAChE variants, only hAChE-S enhances Aβ fibrillization in vitro and Aβ deposition and toxicity in vivo Only hAChE-S contains an amphiphilic C-terminal domain (T40, AChE575-614), with AChE586-599 homologous to Aβ and forming amyloid fibrils, which implicates T40 in AD pathology. We previously showed that the amyloid scavenger, insulin-degrading enzyme (IDE), generates T40-derived amyloidogenic species that, as a peptide mixture, seed Aβ fibrillization. Here, we characterized 11 peptides from a T40-IDE digest for β-sheet conformation, surfactant activity, fibrillization, and seeding capability. We identified residues important for amyloidogenicity and raised polyclonal antibodies against the most amyloidogenic peptide. These new antisera, alongside other specific antibodies, labeled sections from control, hAChE-S, hAPPswe, and hAChE-S/hAPPswe transgenic mice. We observed that hAChE-S β-sheet species co-localized with Aβ in mature plaque cores, surrounded by hAChE-S α-helical species. This observation provides the first in vivo evidence of the conformation of hAChE-S species within plaques. Our results may explain the role of hAChE-S in Aβ deposition and aggregation, as amyloidogenic hAChE-S β-sheet species might seed Aβ aggregation.
Collapse
Affiliation(s)
- Létitia Jean
- From the Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, United Kingdom and
| | - Stephen Brimijoin
- the Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota 55905
| | - David J Vaux
- From the Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, United Kingdom and
| |
Collapse
|
25
|
Xiang Q, Li XH, Fang XX, Jia J, Ren J, Dong YC, Ou-Yang C. Alterations of synaptic plasticity in aged rats: Evidence of functional and morphological studies. Technol Health Care 2018; 26:271-278. [PMID: 29309043 DOI: 10.3233/thc-170978] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND The aging-related disease and associated neurodegenerative complications, such as cognitive impairment, has received increasing attention. OBJECTIVE The aim of this study was to show changes in cognitive behavior and molecular related the synaptic plasticity in aged-induced cognitive deficits rats. METHODS We used novel object recognition testing and morphological staining as well as western blot to detect changes in cognitive behavior and molecular related the synaptic plasticity. RESULTS The morphological changes of synaptic structure and number on hippocampal neurons and learning and memory deficits were shown during natural aging. Moreover, learning and memory improvement was associated with alterations of hippocampal synaptic plasticity-related proteins, such as SNAP-25, synaptophysin, snapsoin-1 and so on, which distributes to cognitive decline in natural aging. CONCLUSIONS Our study provides more behavior and molecular evidence on relationship of cognitive deficits and aging.
Collapse
|
26
|
Lietzau G, Davidsson W, Östenson CG, Chiazza F, Nathanson D, Pintana H, Skogsberg J, Klein T, Nyström T, Darsalia V, Patrone C. Type 2 diabetes impairs odour detection, olfactory memory and olfactory neuroplasticity; effects partly reversed by the DPP-4 inhibitor Linagliptin. Acta Neuropathol Commun 2018; 6:14. [PMID: 29471869 PMCID: PMC5824492 DOI: 10.1186/s40478-018-0517-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 02/12/2018] [Indexed: 12/26/2022] Open
Abstract
Recent data suggest that olfactory deficits could represent an early marker and a pathogenic mechanism at the basis of cognitive decline in type 2 diabetes (T2D). However, research is needed to further characterize olfactory deficits in diabetes, their relation to cognitive decline and underlying mechanisms. The aim of this study was to determine whether T2D impairs odour detection, olfactory memory as well as neuroplasticity in two major brain areas responsible for olfaction and odour coding: the main olfactory bulb (MOB) and the piriform cortex (PC), respectively. Dipeptidyl peptidase-4 inhibitors (DPP-4i) are clinically used T2D drugs exerting also beneficial effects in the brain. Therefore, we aimed to determine whether DPP-4i could reverse the potentially detrimental effects of T2D on the olfactory system. Non-diabetic Wistar and T2D Goto-Kakizaki rats, untreated or treated for 16 weeks with the DPP-4i linagliptin, were employed. Odour detection and olfactory memory were assessed by using the block, the habituation-dishabituation and the buried pellet tests. We assessed neuroplasticity in the MOB by quantifying adult neurogenesis and GABAergic inhibitory interneurons positive for calbindin, parvalbumin and carletinin. In the PC, neuroplasticity was assessed by quantifying the same populations of interneurons and a newly identified form of olfactory neuroplasticity mediated by post-mitotic doublecortin (DCX) + immature neurons. We show that T2D dramatically reduced odour detection and olfactory memory. Moreover, T2D decreased neurogenesis in the MOB, impaired the differentiation of DCX+ immature neurons in the PC and altered GABAergic interneurons protein expression in both olfactory areas. DPP-4i did not improve odour detection and olfactory memory. However, it normalized T2D-induced effects on neuroplasticity. The results provide new knowledge on the detrimental effects of T2D on the olfactory system. This knowledge could constitute essentials for understanding the interplay between T2D and cognitive decline and for designing effective preventive therapies.
Collapse
|
27
|
Reijs BL, Ramakers IH, Elias-Sonnenschein L, Teunissen CE, Koel-Simmelink M, Tsolaki M, Wahlund LO, Waldemar G, Hausner L, Johannsen P, Vanderstichele H, Verhey F, Devanand D, Visser PJ. Relation of Odor Identification with Alzheimer’s Disease Markers in Cerebrospinal Fluid and Cognition. J Alzheimers Dis 2017; 60:1025-1034. [DOI: 10.3233/jad-170564] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Babette L.R. Reijs
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, The Netherlands
| | - Inez H.G.B. Ramakers
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, The Netherlands
| | - Lyzel Elias-Sonnenschein
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, The Netherlands
| | - Charlotte E. Teunissen
- Neurochemistry Laboratory and Biobank, Department of Clinical Chemistry, Neuroscience Campus Amsterdam, VU University Medical Centre Amsterdam, The Netherlands
| | - Marleen Koel-Simmelink
- Neurochemistry Laboratory and Biobank, Department of Clinical Chemistry, Neuroscience Campus Amsterdam, VU University Medical Centre Amsterdam, The Netherlands
| | - Magda Tsolaki
- Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Lars-Olof Wahlund
- Karolinska Institutet, Karolinska University Hospital, Huddinge, Sweden
| | - Gunhild Waldemar
- Danish Dementia Research Centre, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Lucrezia Hausner
- Department of Geriatric Psychiatry, Central Institute of Mental Health, Medical Faculty of Mannheim, Heidelberg University, Germany
| | - Peter Johannsen
- Danish Dementia Research Centre, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | | | - Frans Verhey
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, The Netherlands
| | - D.P. Devanand
- Division of Geriatric Psychiatry, New York State Psychiatric Institute and Columbia University Medical Center, New York, NY, USA
| | - Pieter Jelle Visser
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, The Netherlands
- Department of Neurology and Alzheimer Centre, VU University Medical Centre, Amsterdam, The Netherlands
| |
Collapse
|
28
|
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
|
29
|
Heterogeneity of odorant identification impairment in patients with Alzheimer's Disease. Sci Rep 2017; 7:4798. [PMID: 28684764 PMCID: PMC5500500 DOI: 10.1038/s41598-017-05201-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 05/24/2017] [Indexed: 02/01/2023] Open
Abstract
Alzheimer’s disease (AD) patients exhibit olfactory dysfunction. However, the olfactory declineti precise nature is not fully understood. One hundred patients (60 AD, 28 amnestic mild cognitive impairment (aMCI), 12 Normal) were enrolled. All participants underwent olfactory function testing using an odour stick identification test for Japanese (OSIT-J). OSIT-J scores were significantly correlated with recall. We classified OSIT-J odorants into three groups: Category I, odorants that were difficult for normal aged subjects to identify; Category II, odorants that became harder to accurately identify with cognitive decline; and Category III, odorants that even AD patients could identify. We defined a “cognitive subset” consisting of six Category II OSIT-J odorants (perfume, rose, Japanese cypress, curry, India ink and gas leak odour). The ability to identify “cognitive subset” odours was significantly better indicator of cognitive status than the ability to identify “non-cognitive subset”, which consisted of the six remaining items. The ability to identify the gas leak odorant was decreased early in the aMCI stage, suggesting a need to reconsider the odours used to signal gas leaks. The “cognitive subset” would provide a more convenient and effective biomarker for diagnosing dementia in clinical settings.
Collapse
|
30
|
Lipnicki DM, Crawford J, Kochan NA, Trollor JN, Draper B, Reppermund S, Maston K, Mather KA, Brodaty H, Sachdev PS. Risk Factors for Mild Cognitive Impairment, Dementia and Mortality: The Sydney Memory and Ageing Study. J Am Med Dir Assoc 2016; 18:388-395. [PMID: 28043804 DOI: 10.1016/j.jamda.2016.10.014] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 10/31/2016] [Accepted: 10/31/2016] [Indexed: 01/27/2023]
Abstract
BACKGROUND The nature and commonality of late-life risk factors for mild cognitive impairment (MCI), dementia, and mortality remain unclear. Our aim was to investigate potential risk factors, simultaneously in a single cohort including many individuals initially with normal cognition and followed for 6 years. METHODS We classified 873 community-dwelling individuals (70-90 years old and without dementia at baseline) from the Sydney Memory and Ageing Study as cognitively normal (CN), having MCI or dementia, or deceased 6 years after baseline. Associations with baseline demographic, lifestyle, health, and medical factors were investigated, including apolipoprotein (APOE) genotype, MCI at baseline, and reversion from MCI to CN within 2 years of baseline. RESULTS Eighty-three (9.5%) participants developed dementia and 114 (13%) died within 6 years; nearly 33% had MCI at baseline, of whom 28% reverted to CN within 2 years. A core set of baseline factors was associated with MCI and dementia at 6 years, including older age (per year: odds ratios and 95% confidence intervals = 1.08, 1.01-1.14 for MCI; 1.19, 1.09-1.31 for dementia), MCI at baseline (5.75, 3.49-9.49; 8.23, 3.93-17.22), poorer smelling ability (per extra test point: 0.89, 0.79-1.02; 0.80, 0.68-0.94), slower walking speed (per second: 1.12, 1.00-1.25; 1.21, 1.05-1.39), and being an APOE ε4 carrier (1.84, 1.07-3.14; 3.63, 1.68-7.82). All except APOE genotype were also associated with mortality (age: 1.11, 1.03-1.20; MCI: 3.87, 1.97-7.59; smelling ability: 0.83, 0.70-0.97; walking speed: 1.18, 1.03-1.34). Compared with stable CN participants, individuals reverting from MCI to CN after 2 years were at greater risk of future MCI (3.06, 1.63-5.72). Those who reverted exhibited some different associations between baseline risk factors and 6-year outcomes than individuals with stable MCI. CONCLUSION A core group of late-life risk factors indicative of physical and mental frailty are associated with each of dementia, MCI, and mortality after 6 years. Tests for slower walking speed and poorer smelling ability may help screen for cognitive decline. Individuals with normal cognition are at greater risk of future cognitive impairment if they have a history of MCI.
Collapse
Affiliation(s)
- Darren M Lipnicki
- CHeBA (Centre for Healthy Brain Ageing), School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | - John Crawford
- CHeBA (Centre for Healthy Brain Ageing), School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | - Nicole A Kochan
- CHeBA (Centre for Healthy Brain Ageing), School of Psychiatry, University of New South Wales, Sydney, NSW, Australia; Neuropsychiatric Institute, Prince of Wales Hospital, Sydney, NSW, Australia
| | - Julian N Trollor
- CHeBA (Centre for Healthy Brain Ageing), School of Psychiatry, University of New South Wales, Sydney, NSW, Australia; Department of Developmental Disability Neuropsychiatry, School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | - Brian Draper
- CHeBA (Centre for Healthy Brain Ageing), School of Psychiatry, University of New South Wales, Sydney, NSW, Australia; Primary Dementia Collaborative Research Centre, School of Psychiatry, University of New South Wales, Sydney, NSW, Australia; Academic Department for Old Age Psychiatry, Prince of Wales Hospital, Sydney, NSW, Australia
| | - Simone Reppermund
- CHeBA (Centre for Healthy Brain Ageing), School of Psychiatry, University of New South Wales, Sydney, NSW, Australia; Department of Developmental Disability Neuropsychiatry, School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | - Kate Maston
- CHeBA (Centre for Healthy Brain Ageing), School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | - Karen A Mather
- CHeBA (Centre for Healthy Brain Ageing), School of Psychiatry, University of New South Wales, Sydney, NSW, Australia; Neuropsychiatric Institute, Prince of Wales Hospital, Sydney, NSW, Australia
| | - Henry Brodaty
- CHeBA (Centre for Healthy Brain Ageing), School of Psychiatry, University of New South Wales, Sydney, NSW, Australia; Primary Dementia Collaborative Research Centre, School of Psychiatry, University of New South Wales, Sydney, NSW, Australia; Academic Department for Old Age Psychiatry, Prince of Wales Hospital, Sydney, NSW, Australia
| | - Perminder S Sachdev
- CHeBA (Centre for Healthy Brain Ageing), School of Psychiatry, University of New South Wales, Sydney, NSW, Australia; Neuropsychiatric Institute, Prince of Wales Hospital, Sydney, NSW, Australia; Primary Dementia Collaborative Research Centre, School of Psychiatry, University of New South Wales, Sydney, NSW, Australia.
| | -
- CHeBA (Centre for Healthy Brain Ageing), School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
31
|
Coronas-Samano G, Baker KL, Tan WJT, Ivanova AV, Verhagen JV. Fus1 KO Mouse As a Model of Oxidative Stress-Mediated Sporadic Alzheimer's Disease: Circadian Disruption and Long-Term Spatial and Olfactory Memory Impairments. Front Aging Neurosci 2016; 8:268. [PMID: 27895577 PMCID: PMC5108791 DOI: 10.3389/fnagi.2016.00268] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 10/24/2016] [Indexed: 12/13/2022] Open
Abstract
Insufficient advances in the development of effective therapeutic treatments of sporadic Alzheimer's Disease (sAD) to date are largely due to the lack of sAD-relevant animal models. While the vast majority of models do recapitulate AD's hallmarks of plaques and tangles by virtue of tau and/or beta amyloid overexpression, these models do not reflect the fact that in sAD (unlike familial AD) these genes are not risk factors per se and that other mechanisms like oxidative stress, metabolic dysregulation and inflammation play key roles in AD etiology. Here we characterize and propose the Fus1 KO mice that lack a mitochondrial protein Fus1/Tusc2 as a new sAD model. To establish sAD relevance, we assessed sAD related deficits in Fus1 KO and WT adult mice of 4-5 months old, the equivalent human age when the earliest cognitive and olfactory sAD symptoms arise. Fus1 KO mice showed oxidative stress (increased levels of ROS, decreased levels of PRDX1), disruption of metabolic homeostasis (decreased levels of ACC2, increased phosphorylation of AMPK), autophagy (decreased levels of LC3-II), PKC (decreased levels of RACK1) and calcium signaling (decreased levels of Calb2) in the olfactory bulb and/or hippocampus. Mice were behaviorally tested using objective and accurate video tracking (Noldus), in which Fus1 KO mice showed clear deficits in olfactory memory (decreased habituation/cross-habituation in the short and long term), olfactory guided navigation memory (inability to reduce their latency to find the hidden cookie), spatial memory (learning impairments on finding the platform in the Morris water maze) and showed more sleep time during the diurnal cycle. Fus1 KO mice did not show clear deficits in olfactory perception (cross-habituation), association memory (passive avoidance) or in species-typical behavior (nest building) and no increased anxiety (open field, light-dark box) or depression/anhedonia (sucrose preference) at this relatively young age. These neurobehavioral deficits of the Fus1 KO mice at this relatively young age are highly relevant to sAD, making them suitable for effective research on pharmacological targets in the context of early intervention of sAD.
Collapse
Affiliation(s)
| | - Keeley L Baker
- The John B. Pierce LaboratoryNew Haven, CT, USA; Department of Neuroscience, Yale University School of MedicineNew Haven, CT, USA
| | - Winston J T Tan
- Department of Surgery, Yale University School of Medicine New Haven, CT, USA
| | - Alla V Ivanova
- Department of Surgery, Yale University School of Medicine New Haven, CT, USA
| | - Justus V Verhagen
- The John B. Pierce LaboratoryNew Haven, CT, USA; Department of Neuroscience, Yale University School of MedicineNew Haven, CT, USA
| |
Collapse
|
32
|
Hadar A, Milanesi E, Squassina A, Niola P, Chillotti C, Pasmanik-Chor M, Yaron O, Martásek P, Rehavi M, Weissglas-Volkov D, Shomron N, Gozes I, Gurwitz D. RGS2 expression predicts amyloid-β sensitivity, MCI and Alzheimer's disease: genome-wide transcriptomic profiling and bioinformatics data mining. Transl Psychiatry 2016; 6:e909. [PMID: 27701409 PMCID: PMC5315547 DOI: 10.1038/tp.2016.179] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Revised: 05/26/2016] [Accepted: 06/15/2016] [Indexed: 12/30/2022] Open
Abstract
Alzheimer's disease (AD) is the most frequent cause of dementia. Misfolded protein pathological hallmarks of AD are brain deposits of amyloid-β (Aβ) plaques and phosphorylated tau neurofibrillary tangles. However, doubts about the role of Aβ in AD pathology have been raised as Aβ is a common component of extracellular brain deposits found, also by in vivo imaging, in non-demented aged individuals. It has been suggested that some individuals are more prone to Aβ neurotoxicity and hence more likely to develop AD when aging brains start accumulating Aβ plaques. Here, we applied genome-wide transcriptomic profiling of lymphoblastoid cells lines (LCLs) from healthy individuals and AD patients for identifying genes that predict sensitivity to Aβ. Real-time PCR validation identified 3.78-fold lower expression of RGS2 (regulator of G-protein signaling 2; P=0.0085) in LCLs from healthy individuals exhibiting high vs low Aβ sensitivity. Furthermore, RGS2 showed 3.3-fold lower expression (P=0.0008) in AD LCLs compared with controls. Notably, RGS2 expression in AD LCLs correlated with the patients' cognitive function. Lower RGS2 expression levels were also discovered in published expression data sets from postmortem AD brain tissues as well as in mild cognitive impairment and AD blood samples compared with controls. In conclusion, Aβ sensitivity phenotyping followed by transcriptomic profiling and published patient data mining identified reduced peripheral and brain expression levels of RGS2, a key regulator of G-protein-coupled receptor signaling and neuronal plasticity. RGS2 is suggested as a novel AD biomarker (alongside other genes) toward early AD detection and future disease modifying therapeutics.
Collapse
Affiliation(s)
- A Hadar
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - E Milanesi
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - A Squassina
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - P Niola
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - C Chillotti
- Unit of Clinical Pharmacology, University Hospital of Cagliari, Cagliari, Italy
| | - M Pasmanik-Chor
- Bioinformatics Unit, George Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - O Yaron
- The Genomic Analysis Laboratory, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - P Martásek
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Prague, Czech Republic
| | - M Rehavi
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv, Israel
| | - D Weissglas-Volkov
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - N Shomron
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel,Adams Super Center for Brain Studies, Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - I Gozes
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel,Adams Super Center for Brain Studies, Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel,Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel. E-mail: or
| | - D Gurwitz
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel,Adams Super Center for Brain Studies, Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel,Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel. E-mail: or
| |
Collapse
|