1
|
Heydari R, Khosravifar M, Abiri S, Dashtbin S, Alvandi A, Nedaei SE, Salimi Z, Zarei F, Abiri R. A domestic strain of Lactobacillus rhamnosus attenuates cognitive deficit and pro-inflammatory cytokine expression in an animal model of Alzheimer's disease. Behav Brain Res 2025; 476:115277. [PMID: 39343242 DOI: 10.1016/j.bbr.2024.115277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 08/30/2024] [Accepted: 09/27/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND Microbiome dysbiosis plays a significant role in neuroinflammation and Alzheimer's disease (AD). Therefore, gut microbiome restoration using appropriate probiotics may be beneficial in alleviating AD features. In this study, we investigated the effects of a domestic strain of Lactobacillus rhamnosus (L. rhamnosus) on spatial memory, and cytokines expression in an inflammation-based AD model. METHOD Male Wistar rats were randomly divided into four groups (six animals per group) of control, L. rhamnosus-only, D-galactose (D-gal)-only, and D-gal + L. rhamnosus. Spatial learning and memory were assessed using the Morris water maze test. IL-1β, IL-6, and TNF-α expression levels were measured using Real-Time qPCR. A significance level of 0.05 was used for statistical analysis. RESULTS In contrast to the D-gal + L. rhamnosus-treated group, D-gal only treated group showed impaired memory in MWM test compared to the control group. Additionally, D-gal treatment resulted in an increase in IL-1β and TNF-α levels and a decrease in IL-6 levels, which was not statistically significant. However, the TNF-α level was significantly decreased in D-gal + L. rhamnosus-treated group compared to D-gal-only treated group (P < 0.05). Also, IL-6 level was significantly lower in D-gal + L. rhamnosus-treated group compared to control group (P < 0.05). CONCLUSION These results suggest that the domestic L. rhamnosus might positively impact cognitive deficit and neuroinflammation. Further studies are suggested to investigate the specific mechanisms mediating the effects of L. rhamnosus on cognitive functions and neuroinflammation in animal models of AD.
Collapse
Affiliation(s)
- Ruhollah Heydari
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mina Khosravifar
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Shervin Abiri
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Shirin Dashtbin
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Amirhoushang Alvandi
- Department of Microbiology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Seyed Ershad Nedaei
- Department of Physiology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zahra Salimi
- Department of Biology, Faculty of Science, University of Qom, Qom, Iran
| | - Fatemeh Zarei
- Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran
| | - Ramin Abiri
- Department of Microbiology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran; Fertility and Infertility Research Center, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
2
|
Jobin B, Magdamo C, Delphus D, Runde A, Reineke S, Soto AA, Ergun B, Albers AD, Albers MW. AROMHA Brain Health Test: A Remote Olfactory Assessment as a Screen for Cognitive Impairment. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.03.24311283. [PMID: 39211882 PMCID: PMC11361214 DOI: 10.1101/2024.08.03.24311283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Cost-effective, noninvasive screening methods for preclinical Alzheimer's disease (AD) and other neurocognitive disorders remain an unmet need. The olfactory neural circuits develop AD pathological changes prior to symptom onset. To probe these vulnerable circuits, we developed the digital remote AROMHA Brain Health Test (ABHT), an at-home odor identification, discrimination, memory, and intensity assessment. The ABHT was self-administered among cognitively normal (CN) English and Spanish speakers (n=127), participants with subjective cognitive complaints (SCC; n=34), and mild cognitive impairment (MCI; n=19). Self-administered tests took place remotely at home under unobserved (among interested CN participants) and observed modalities (CN, SCC, and MCI), as well as in-person with a research assistant present (CN, SCC, and MCI). Olfactory performance was similar across observed and unobserved remote self-administration and between English and Spanish speakers. Odor memory, identification, and discrimination scores decreased with age, and olfactory identification and discrimination were lower in the MCI group compared to CN and SCC groups, independent of age, sex, and education. The ABHT revealed age-related olfactory decline, and discriminated CN older adults from those with cognitive impairment. Replication of our results in other populations would support the use of the ABHT to identify and monitor individuals at risk for developing dementia.
Collapse
|
3
|
Murai T, Bailey L, Schultz L, Mongeau L, DeSana A, Silva AC, Roberts AC, Sukoff Rizzo SJ. Improving preclinical to clinical translation of cognitive function for aging-related disorders: the utility of comprehensive touchscreen testing batteries in common marmosets. COGNITIVE, AFFECTIVE & BEHAVIORAL NEUROSCIENCE 2024; 24:325-348. [PMID: 38200282 PMCID: PMC11039501 DOI: 10.3758/s13415-023-01144-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 12/04/2023] [Indexed: 01/12/2024]
Abstract
Concerns about poor animal to human translation have come increasingly to the fore, in particular with regards to cognitive improvements in rodent models, which have failed to translate to meaningful clinical benefit in humans. This problem has been widely acknowledged, most recently in the field of Alzheimer's disease, although this issue pervades the spectrum of central nervous system (CNS) disorders, including neurodevelopmental, neuropsychiatric, and neurodegenerative diseases. Consequently, recent efforts have focused on improving preclinical to clinical translation by incorporating more clinically analogous outcome measures of cognition, such as touchscreen-based assays, which can be employed across species, and have great potential to minimize the translational gap. For aging-related research, it also is important to incorporate model systems that facilitate the study of the long prodromal phase in which cognitive decline begins to emerge and which is a major limitation of short-lived species, such as laboratory rodents. We posit that to improve translation of cognitive function and dysfunction, nonhuman primate models, which have conserved anatomical and functional organization of the primate brain, are necessary to move the field of translational research forward and to bridge the translational gaps. The present studies describe the establishment of a comprehensive battery of touchscreen-based tasks that capture a spectrum of domains sensitive to detecting aging-related cognitive decline, which will provide the greatest benefit through longitudinal evaluation throughout the prolonged lifespan of the marmoset.
Collapse
Affiliation(s)
- Takeshi Murai
- Aging Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Lauren Bailey
- Aging Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Laura Schultz
- Aging Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Lauren Mongeau
- Aging Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Andrew DeSana
- Aging Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Afonso C Silva
- Department of Neurobiology, University of Pittsburgh School of Medicine, 514A Bridgeside Point 1, 100 Technology Drive, Pittsburgh, PA, 15219, USA
| | - Angela C Roberts
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Stacey J Sukoff Rizzo
- Aging Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Department of Neurobiology, University of Pittsburgh School of Medicine, 514A Bridgeside Point 1, 100 Technology Drive, Pittsburgh, PA, 15219, USA.
| |
Collapse
|
4
|
Pluta R. A Look at the Etiology of Alzheimer's Disease based on the Brain Ischemia Model. Curr Alzheimer Res 2024; 21:166-182. [PMID: 38963100 DOI: 10.2174/0115672050320921240627050736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 05/18/2024] [Accepted: 05/30/2024] [Indexed: 07/05/2024]
Abstract
Alzheimer's disease (AD) is the frequent form of dementia in the world. Despite over 100 years of research into the causes of AD, including amyloid and tau protein, the research has stalled and has not led to any conclusions. Moreover, numerous projects aimed at finding a cure for AD have also failed to achieve a breakthrough. Thus, the failure of anti-amyloid and anti-tau protein therapy to treat AD significantly influenced the way we began to think about the etiology of the disease. This situation prompted a group of researchers to focus on ischemic brain episodes, which, like AD, mostly present alterations in the hippocampus. In this context, it has been proposed that cerebral ischemic incidents may play a major role in promoting amyloid and tau protein in neurodegeneration in AD. In this review, we summarized the experimental and clinical research conducted over several years on the role of ischemic brain episodes in the development of AD. Studies have shown changes typical of AD in the course of brain neurodegeneration post-ischemia, i.e., progressive brain and hippocampal atrophy, increased amyloid production, and modification of tau protein. In the post-ischemic brain, the diffuse and senile amyloid plaques and the development of neurofibrillary tangles characteristic of AD were revealed. The above data evidently showed that after brain ischemia, there are modifications in protein folding, leading to massive neuronal death and damage to the neuronal network, which triggers dementia with the AD phenotype.
Collapse
Affiliation(s)
- Ryszard Pluta
- Department of Pathophysiology, Medical University of Lublin, 20-090 Lublin, Poland
| |
Collapse
|
5
|
Stecker MM, Srivastava A, Reiss AB. Amyloid-β Effects on Peripheral Nerve: A New Model System. Int J Mol Sci 2023; 24:14488. [PMID: 37833938 PMCID: PMC10572603 DOI: 10.3390/ijms241914488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 09/22/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023] Open
Abstract
Although there are many biochemical methods to measure amyloid-β (Aβ)42 concentration, one of the critical issues in the study of the effects of Aβ42 on the nervous system is a simple physiological measurement. The in vitro rat sciatic nerve model is employed and the nerve action potential (NAP) is quantified with different stimuli while exposed to different concentrations of Aβ42. Aβ42 predominantly reduces the NAP amplitude with minimal effects on other parameters except at low stimulus currents and short inter-stimulus intervals. The effects of Aβ42 are significantly concentration-dependent, with a maximum reduction in NAP amplitude at a concentration of 70 nM and smaller effects on the NAP amplitude at higher and lower concentrations. However, even physiologic concentrations in the range of 70 pM did reduce the NAP amplitude. The effects of Aβ42 became maximal 5-8 h after exposure and did not reverse during a 30 min washout period. The in vitro rat sciatic nerve model is sensitive to the effects of physiologic concentrations of Aβ42. These experiments suggest that the effect of Aβ42 is a very complex function of concentration that may be the result of amyloid-related changes in membrane properties or sodium channels.
Collapse
Affiliation(s)
| | - Ankita Srivastava
- Department of Medicine and Biomedical Research Institute, NYU Grossman Long Island School of Medicine, Long Island, NY 11501, USA; (A.S.); (A.B.R.)
| | - Allison B. Reiss
- Department of Medicine and Biomedical Research Institute, NYU Grossman Long Island School of Medicine, Long Island, NY 11501, USA; (A.S.); (A.B.R.)
| |
Collapse
|
6
|
Leitner DF, Kanshin E, Faustin A, Thierry M, Friedman D, Devore S, Ueberheide B, Devinsky O, Wisniewski T. Localized proteomic differences in the choroid plexus of Alzheimer's disease and epilepsy patients. Front Neurol 2023; 14:1221775. [PMID: 37521285 PMCID: PMC10379643 DOI: 10.3389/fneur.2023.1221775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 06/22/2023] [Indexed: 08/01/2023] Open
Abstract
Introduction Alzheimer's disease (AD) and epilepsy are reciprocally related. Among sporadic AD patients, clinical seizures occur in 10-22% and subclinical epileptiform abnormalities occur in 22-54%. Cognitive deficits, especially short-term memory impairments, occur in most epilepsy patients. Common neurophysiological and molecular mechanisms occur in AD and epilepsy. The choroid plexus undergoes pathological changes in aging, AD, and epilepsy, including decreased CSF turnover, amyloid beta (Aβ), and tau accumulation due to impaired clearance and disrupted CSF amino acid homeostasis. This pathology may contribute to synaptic dysfunction in AD and epilepsy. Methods We evaluated control (n = 8), severe AD (n = 8; A3, B3, C3 neuropathology), and epilepsy autopsy cases (n = 12) using laser capture microdissection (LCM) followed by label-free quantitative mass spectrometry on the choroid plexus adjacent to the hippocampus at the lateral geniculate nucleus level. Results Proteomics identified 2,459 proteins in the choroid plexus. At a 5% false discovery rate (FDR), 616 proteins were differentially expressed in AD vs. control, 1 protein in epilepsy vs. control, and 438 proteins in AD vs. epilepsy. There was more variability in the epilepsy group across syndromes. The top 20 signaling pathways associated with differentially expressed proteins in AD vs. control included cell metabolism pathways; activated fatty acid beta-oxidation (p = 2.00 x 10-7, z = 3.00), and inhibited glycolysis (p = 1.00 x 10-12, z = -3.46). For AD vs. epilepsy, the altered pathways included cell metabolism pathways, activated complement system (p = 5.62 x 10-5, z = 2.00), and pathogen-induced cytokine storm (p = 2.19 x 10-2, z = 3.61). Of the 617 altered proteins in AD and epilepsy vs. controls, 497 (81%) were positively correlated (p < 0.0001, R2 = 0.27). Discussion We found altered signaling pathways in the choroid plexus of severe AD cases and many correlated changes in the protein expression of cell metabolism pathways in AD and epilepsy cases. The shared molecular mechanisms should be investigated further to distinguish primary pathogenic changes from the secondary ones. These mechanisms could inform novel therapeutic strategies to prevent disease progression or restore normal function. A focus on dual-diagnosed AD/epilepsy cases, specific epilepsy syndromes, such as temporal lobe epilepsy, and changes across different severity levels in AD and epilepsy would add to our understanding.
Collapse
Affiliation(s)
- Dominique F. Leitner
- Comprehensive Epilepsy Center, New York University Grossman School of Medicine, New York, NY, United States
- Center for Cognitive Neurology, Department of Neurology, New York University Grossman School of Medicine, New York, NY, United States
- Department of Neurology, New York University Grossman School of Medicine, New York, NY, United States
| | - Evgeny Kanshin
- Proteomics Laboratory, Division of Advanced Research Technologies, New York University Grossman School of Medicine, New York, NY, United States
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, United States
| | - Arline Faustin
- Center for Cognitive Neurology, Department of Neurology, New York University Grossman School of Medicine, New York, NY, United States
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, United States
| | - Manon Thierry
- Center for Cognitive Neurology, Department of Neurology, New York University Grossman School of Medicine, New York, NY, United States
- Department of Neurology, New York University Grossman School of Medicine, New York, NY, United States
| | - Daniel Friedman
- Comprehensive Epilepsy Center, New York University Grossman School of Medicine, New York, NY, United States
- Department of Neurology, New York University Grossman School of Medicine, New York, NY, United States
| | - Sasha Devore
- Comprehensive Epilepsy Center, New York University Grossman School of Medicine, New York, NY, United States
- Department of Neurology, New York University Grossman School of Medicine, New York, NY, United States
| | - Beatrix Ueberheide
- Center for Cognitive Neurology, Department of Neurology, New York University Grossman School of Medicine, New York, NY, United States
- Proteomics Laboratory, Division of Advanced Research Technologies, New York University Grossman School of Medicine, New York, NY, United States
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, United States
| | - Orrin Devinsky
- Comprehensive Epilepsy Center, New York University Grossman School of Medicine, New York, NY, United States
- Department of Neurology, New York University Grossman School of Medicine, New York, NY, United States
| | - Thomas Wisniewski
- Center for Cognitive Neurology, Department of Neurology, New York University Grossman School of Medicine, New York, NY, United States
- Department of Neurology, New York University Grossman School of Medicine, New York, NY, United States
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, United States
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, United States
| |
Collapse
|
7
|
Reiss AB, Muhieddine D, Jacob B, Mesbah M, Pinkhasov A, Gomolin IH, Stecker MM, Wisniewski T, De Leon J. Alzheimer's Disease Treatment: The Search for a Breakthrough. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1084. [PMID: 37374288 PMCID: PMC10302500 DOI: 10.3390/medicina59061084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 05/22/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023]
Abstract
As the search for modalities to cure Alzheimer's disease (AD) has made slow progress, research has now turned to innovative pathways involving neural and peripheral inflammation and neuro-regeneration. Widely used AD treatments provide only symptomatic relief without changing the disease course. The recently FDA-approved anti-amyloid drugs, aducanumab and lecanemab, have demonstrated unclear real-world efficacy with a substantial side effect profile. Interest is growing in targeting the early stages of AD before irreversible pathologic changes so that cognitive function and neuronal viability can be preserved. Neuroinflammation is a fundamental feature of AD that involves complex relationships among cerebral immune cells and pro-inflammatory cytokines, which could be altered pharmacologically by AD therapy. Here, we provide an overview of the manipulations attempted in pre-clinical experiments. These include inhibition of microglial receptors, attenuation of inflammation and enhancement of toxin-clearing autophagy. In addition, modulation of the microbiome-brain-gut axis, dietary changes, and increased mental and physical exercise are under evaluation as ways to optimize brain health. As the scientific and medical communities work together, new solutions may be on the horizon to slow or halt AD progression.
Collapse
Affiliation(s)
- Allison B. Reiss
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, Mineola, NY 11501, USA; (D.M.); (B.J.); (M.M.); (A.P.); (I.H.G.); (J.D.L.)
| | - Dalia Muhieddine
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, Mineola, NY 11501, USA; (D.M.); (B.J.); (M.M.); (A.P.); (I.H.G.); (J.D.L.)
| | - Berlin Jacob
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, Mineola, NY 11501, USA; (D.M.); (B.J.); (M.M.); (A.P.); (I.H.G.); (J.D.L.)
| | - Michael Mesbah
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, Mineola, NY 11501, USA; (D.M.); (B.J.); (M.M.); (A.P.); (I.H.G.); (J.D.L.)
| | - Aaron Pinkhasov
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, Mineola, NY 11501, USA; (D.M.); (B.J.); (M.M.); (A.P.); (I.H.G.); (J.D.L.)
| | - Irving H. Gomolin
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, Mineola, NY 11501, USA; (D.M.); (B.J.); (M.M.); (A.P.); (I.H.G.); (J.D.L.)
| | | | - Thomas Wisniewski
- Center for Cognitive Neurology, Departments of Neurology, Pathology and Psychiatry, NYU School of Medicine, New York, NY 10016, USA;
| | - Joshua De Leon
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, Mineola, NY 11501, USA; (D.M.); (B.J.); (M.M.); (A.P.); (I.H.G.); (J.D.L.)
| |
Collapse
|
8
|
Xu Y, Pan X, Li H, Cao Q, Xu F, Zhang J. Accuracy of Raman spectroscopy in the diagnosis of Alzheimer's disease. Front Psychiatry 2023; 14:1112615. [PMID: 37009107 PMCID: PMC10060832 DOI: 10.3389/fpsyt.2023.1112615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 03/01/2023] [Indexed: 03/18/2023] Open
Abstract
ObjectiveTo systematically evaluate the accuracy of Raman spectroscopy in the diagnosis of Alzheimer's disease.MethodsDatabases including Web of Science, PubMed, The Cochrane Library, EMbase, CBM, CNKI, Wan Fang Data, and VIP were electronically searched for studies on Raman spectroscopy in diagnosis of Alzheimer's disease from inception to November 2022. Two reviewers independently screened the literature, extracted data, and assessed the risk of bias in the included studies. Then, meta-analysis was performed using Meta-Disc1.4 and Stata 16.0 software.ResultsA total of eight studies were finally included. The pooled sensitivity of Raman spectroscopy was 0.86 [95% CI (0.80–0.91)], specificity was 0.87 [95% CI (0.79–0.92)], positive likelihood ratio was 5.50 [95% CI (3.55–8.51)], negative likelihood ratio was 0.17 [95% CI (0.09–0.34)], diagnosis odds ratio and area under the curve of SROC were 42.44 [95% CI (19.80–90.97)] and 0.931, respectively. Sensitivity analysis was carried out after each study was excluded one by one, and the results showed that pooled sensitivity and specificity had no significant change, indicating that the stability of the meta-analysis results was great.ConclusionsOur findings indicated that Raman spectroscopy had high accuracy in the diagnosis of AD, though it still did not rule out the possibility of misdiagnosis and missed diagnosis. Limited by the quantity and quality of the included studies, the above conclusions need to be verified by more high-quality studies.
Collapse
Affiliation(s)
- Yanmei Xu
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xinyu Pan
- School of Pharmacy, Chengdu Medical College, Chengdu, Sichuan, China
| | - Huan Li
- School of Pharmacy, Chengdu Medical College, Chengdu, Sichuan, China
| | - Qiongfang Cao
- Department of Public Health, Chengdu Medical College, Chengdu, Sichuan, China
| | - Fan Xu
- Department of Public Health, Chengdu Medical College, Chengdu, Sichuan, China
- *Correspondence: Fan Xu
| | - Jianshu Zhang
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Jianshu Zhang
| |
Collapse
|
9
|
Stecker M. A Perspective: Challenges in Dementia Research. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:1368. [PMID: 36295529 PMCID: PMC9609997 DOI: 10.3390/medicina58101368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/21/2022] [Accepted: 09/27/2022] [Indexed: 11/17/2022]
Abstract
Although dementia is a common and devastating disease that has been studied intensely for more than 100 years, no effective disease modifying treatment has been found. At this impasse, new approaches are important. The purpose of this paper is to provide, in the context of current research, one clinician's perspective regarding important challenges in the field in the form of specific challenges. These challenges not only illustrate the scope of the problems inherent in finding treatments for dementia, but can also be specific targets to foster discussion, criticism and new research. One common theme is the need to transform research activities from small projects in individual laboratories/clinics to larger multinational projects, in which each clinician and researcher works as an integral part. This transformation will require collaboration between researchers, large corporations, regulatory/governmental authorities and the general population, as well as significant financial investments. However, the costs of transforming the approach are small in comparison with the cost of dementia.
Collapse
Affiliation(s)
- Mark Stecker
- Fresno Institute of Neuroscience, Fresno, CA 93720, USA
| |
Collapse
|
10
|
Stecker MM, Peltier MR, Reiss AB. The role of massive demographic databases in intractable illnesses: Denomics for dementia. AIMS Public Health 2022; 9:618-629. [PMID: 36330282 PMCID: PMC9581740 DOI: 10.3934/publichealth.2022043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 08/10/2022] [Accepted: 08/14/2022] [Indexed: 07/26/2023] Open
Abstract
Despite intensive research, effective treatments for many common and devastating diseases are lacking. For example, huge efforts and billions of dollars have been invested in Alzheimer's disease (AD), which affects over 50 million people worldwide. However, there is still no effective drug that can slow or cure AD. This relates, in part, to the absence of an animal model or cellular system that incorporates all the relevant features of the disease. Therefore, large scale studies on human populations and tissues will be key to better understanding dementia and developing methods to prevent or treat it. This is especially difficult because the dementia phenotype can result from many different processes and is likely to be affected by multiple personal and environmental variables. We hypothesize that analyzing massive volumes of demographic data that are currently available and combining this with genomic, proteomic, and metabolomic profiles of AD patients and their families, new insights into pathophysiology and treatment of AD may arise. While this requires much coordination and cooperation among large institutions, the potential for advancement would be life-changing for millions of people. In many ways this represents the next step in the information revolution started by the Human Genome Project.
Collapse
Affiliation(s)
| | - Morgan R. Peltier
- Department of Psychiatry, Hackensack Meridian Health, Neptune City, NJ 07753, USA
| | | |
Collapse
|
11
|
Pluta R, Januszewski S, Jabłoński M. Acetylated Tau Protein: A New Piece in the Puzzle between Brain Ischemia and Alzheimer’s Disease. Int J Mol Sci 2022; 23:ijms23169174. [PMID: 36012440 PMCID: PMC9408862 DOI: 10.3390/ijms23169174] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 08/12/2022] [Indexed: 11/29/2022] Open
Affiliation(s)
- Ryszard Pluta
- Laboratory of Ischemic and Neurodegenerative Brain Research, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland
- Correspondence: ; Tel.: +48-22-6086-540
| | - Sławomir Januszewski
- Laboratory of Ischemic and Neurodegenerative Brain Research, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Mirosław Jabłoński
- Department of Rehabilitation and Orthopedics, Medical University of Lublin, 20-090 Lublin, Poland
| |
Collapse
|
12
|
Plants, Plants, and More Plants: Plant-Derived Nutrients and Their Protective Roles in Cognitive Function, Alzheimer’s Disease, and Other Dementias. Medicina (B Aires) 2022; 58:medicina58081025. [PMID: 36013492 PMCID: PMC9414574 DOI: 10.3390/medicina58081025] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/18/2022] [Accepted: 07/25/2022] [Indexed: 11/16/2022] Open
Abstract
Background and Objectives: Alzheimer’s disease (AD) is the most common form of dementia, with the risk of developing it attributed to non-modifiable and modifiable factors. Currently, there is no cure for AD. A plant-based diet may protect against cognitive decline, due to the effects of plant-based nutrients such as vitamins, antioxidants, and fiber. The aim of the review is to summarize current literature on plant-based nutrients and their impact on cognition. Materials and Methods: A search was conducted on PubMed for clinical and murine studies, using combinations of the following words: “Alzheimer’s disease”, “dementia”, “cognition”, “plant-based diet”, “mild cognitive impairment”, “vitamin B”, “vitamin C”, “vitamin E, “beta carotene”, “antioxidants”, “fiber”, “vitamin K”, “Mediterranean diet”, “vitamin D”, and “mushrooms”. Results and Conclusions: A diet rich in vitamin B and antioxidants can benefit the cognitive functions of individuals as shown in randomized clinical trials. Vitamin K is associated with improved cognition, although large randomized controlled trials need to be done. Fiber has been shown to prevent cognitive decline in animal studies. Vitamin D may contribute to cognitive health via anti-inflammatory processes. Several medical organizations have recommended a plant-based diet for optimizing cognitive health and potentially helping to prevent dementia.
Collapse
|
13
|
Singh S, Yang F, Sivils A, Cegielski V, Chu XP. Amylin and Secretases in the Pathology and Treatment of Alzheimer's Disease. Biomolecules 2022; 12:996. [PMID: 35883551 PMCID: PMC9312829 DOI: 10.3390/biom12070996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/11/2022] [Accepted: 07/15/2022] [Indexed: 11/17/2022] Open
Abstract
Alzheimer's disease remains a prevailing neurodegenerative condition which has an array physical, emotional, and financial consequences to patients and society. In the past decade, there has been a greater degree of investigation on therapeutic small peptides. This group of biomolecules have a profile of fundamentally sound characteristics which make them an intriguing area for drug development. Among these biomolecules, there are four modulatory mechanisms of interest in this review: alpha-, beta-, gamma-secretases, and amylin. These protease-based biomolecules all have a contributory role in the amyloid cascade hypothesis. Moreover, the involvement of various biochemical pathways intertwines these peptides to have shared regulators (i.e., retinoids). Further clinical and translational investigation must occur to gain a greater understanding of its potential application in patient care. The aim of this narrative review is to evaluate the contemporary literature on these protease biomolecule modulators and determine its utility in the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
| | | | | | | | - Xiang-Ping Chu
- Department of Biomedical Sciences, School of Medicine, University of Missouri, Kansas City, MO 64108, USA; (S.S.); (F.Y.); (A.S.); (V.C.)
| |
Collapse
|
14
|
Hudák A, Letoha A, Vizler C, Letoha T. Syndecan-3 as a Novel Biomarker in Alzheimer's Disease. Int J Mol Sci 2022; 23:3407. [PMID: 35328830 PMCID: PMC8955174 DOI: 10.3390/ijms23063407] [Citation(s) in RCA: 13] [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: 02/25/2022] [Revised: 03/18/2022] [Accepted: 03/19/2022] [Indexed: 11/17/2022] Open
Abstract
Early diagnosis of Alzheimer's disease (AD) is of paramount importance in preserving the patient's mental and physical health in a fairly manageable condition for a longer period. Reliable AD detection requires novel biomarkers indicating central nervous system (CNS) degeneration in the periphery. Members of the syndecan family of transmembrane proteoglycans are emerging new targets in inflammatory and neurodegenerative disorders. Reviewing the growing scientific evidence on the involvement of syndecans in the pathomechanism of AD, we analyzed the expression of the neuronal syndecan, syndecan-3 (SDC3), in experimental models of neurodegeneration. Initial in vitro studies showed that prolonged treatment of tumor necrosis factor-alpha (TNF-α) increases SDC3 expression in model neuronal and brain microvascular endothelial cell lines. In vivo studies revealed elevated concentrations of TNF-α in the blood and brain of APPSWE-Tau transgenic mice, along with increased SDC3 concentration in the brain and the liver. Primary brain endothelial cells and peripheral blood monocytes isolated from APPSWE-Tau mice exhibited increased SDC3 expression than wild-type controls. SDC3 expression of blood-derived monocytes showed a positive correlation with amyloid plaque load in the brain, demonstrating that SDC3 on monocytes is a good indicator of amyloid pathology in the brain. Given the well-established role of blood tests, the SDC3 expression of monocytes could serve as a novel biomarker for early AD detection.
Collapse
Affiliation(s)
| | - Annamária Letoha
- Albert Szent-Györgyi Clinical Center, Department of Medicine, Faculty of Medicine, University of Szeged, H-6720 Szeged, Hungary;
| | - Csaba Vizler
- Biological Research Centre, Institute of Biochemistry, H-6726 Szeged, Hungary;
| | | |
Collapse
|
15
|
Pachner AR. The Neuroimmunology of Multiple Sclerosis: Fictions and Facts. Front Neurol 2022; 12:796378. [PMID: 35197914 PMCID: PMC8858985 DOI: 10.3389/fneur.2021.796378] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 12/16/2021] [Indexed: 11/13/2022] Open
Abstract
There have been tremendous advances in the neuroimmunology of multiple sclerosis over the past five decades, which have led to improved diagnosis and therapy in the clinic. However, further advances must take into account an understanding of some of the complex issues in the field, particularly an appreciation of "facts" and "fiction." Not surprisingly given the incredible complexity of both the nervous and immune systems, our understanding of the basic biology of the disease is very incomplete. This lack of understanding has led to many controversies in the field. This review identifies some of these controversies and facts/fictions with relation to the basic neuroimmunology of the disease (cells and molecules), and important clinical issues. Fortunately, the field is in a healthy transition from excessive reliance on animal models to a broader understanding of the disease in humans, which will likely lead to many improved treatments especially of the neurodegeneration in multiple sclerosis (MS).
Collapse
Affiliation(s)
- Andrew R. Pachner
- Dartmouth–Hitchcock Medical Center, Lebanon, NH, United States
- Geisel School of Medicine, Dartmouth College, Hanover, NH, United States
| |
Collapse
|
16
|
Wang M, Lv J, Huang X, Wisniewski T, Zhang W. High-fat diet-induced atherosclerosis promotes neurodegeneration in the triple transgenic (3 × Tg) mouse model of Alzheimer's disease associated with chronic platelet activation. ALZHEIMERS RESEARCH & THERAPY 2021; 13:144. [PMID: 34454596 PMCID: PMC8403418 DOI: 10.1186/s13195-021-00890-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 08/18/2021] [Indexed: 01/14/2023]
Abstract
Background Epidemiological studies link vascular disease risk factors such as atherosclerosis, hypertension, and diabetes mellitus with Alzheimer’s disease (AD). Whether there are direct links between these conditions to β-amyloid (Aβ) aggregation and tau pathology is uncertain. Methods To investigate the possible link between atherosclerosis and AD pathology, we subjected triple transgenic (3 × Tg) AD mice to a high-fat diet (HFD) at 3 months of age, which corresponds to early adulthood in humans. Results After 9 months of treatment, HFD-treated 3 × Tg mice exhibited worse memory deficits accompanied by blood hypercoagulation, thrombocytosis, and chronic platelet activation. Procoagulant platelets from HFD-treated 3 × Tg mice actively induced the conversion of soluble Aβ40 into fibrillar Aβ aggregates, associated with increased expression of integrin αIIbβ3 and clusterin. At 9 months and older, platelet-associated fibrillar Aβ aggregates were observed to obstruct the cerebral blood vessels in HFD-treated 3 × Tg mice. HFD-treated 3 × Tg mice exhibited a greater cerebral amyloid angiopathy (CAA) burden and increased cerebral vascular permeability, as well as more extensive neuroinflammation, tau hyperphosphorylation, and neuron loss. Disaggregation of preexisting platelet micro-clots with humanized GPIIIa49-66 scFv Ab (A11) significantly reduced platelet-associated fibrillar Aβ aggregates in vitro and improved vascular permeability in vivo. Conclusions These findings suggest that a major contribution of atherosclerosis to AD pathology is via its effects on blood coagulation and the formation of platelet-mediated Aβ aggregates that compromise cerebral blood flow and therefore neuronal function. This leads to cognitive decline. Supplementary Information The online version contains supplementary material available at 10.1186/s13195-021-00890-9.
Collapse
Affiliation(s)
- Min Wang
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, 3663 North Zhongshan Road, Shanghai, 200062, China
| | - Junyan Lv
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, 3663 North Zhongshan Road, Shanghai, 200062, China
| | - Xiaoshan Huang
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, 3663 North Zhongshan Road, Shanghai, 200062, China
| | - Thomas Wisniewski
- Center for Cognitive Neurology and Departments of Neurology, Pathology and Psychiatry, New York University School of Medicine, Science Building, Rm1017, 435 East 30th Street, New York, NY, 10016, USA.
| | - Wei Zhang
- Key Laboratory of Brain Functional Genomics (Ministry of Education and Shanghai), School of Life Sciences, East China Normal University, 3663 North Zhongshan Road, Shanghai, 200062, China.
| |
Collapse
|
17
|
Boutajangout A, Zhang W, Kim J, Abdali WA, Prelli F, Wisniewski T. Passive Immunization With a Novel Monoclonal Anti-PrP Antibody TW1 in an Alzheimer's Mouse Model With Tau Pathology. Front Aging Neurosci 2021; 13:640677. [PMID: 33716717 PMCID: PMC7947695 DOI: 10.3389/fnagi.2021.640677] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 02/03/2021] [Indexed: 11/13/2022] Open
Abstract
Neurofibrillary tangles (NFTs) are a major pathologic hallmark of Alzheimer’s disease (AD). Several studies have shown that amyloid β oligomers (Aβo) and tau oligomers mediate their toxicity, in part, via binding to cellular prion protein (PrPC) and that some anti-PrP antibodies can block this interaction. We have generated a novel monoclonal anti-PrP antibody (TW1) and assessed the efficacy of passive immunization with it in a mouse model of AD with extensive tau pathology: hTau/PS1 transgenic (Tg) mice. These mice were injected intraperitoneally once a week with TW1 starting at 5 months of age. Behavior was assessed at 8 months of age and brain tissue was subsequently harvested for analysis of treatment efficacy at 9 months. Mice treated with TW1 did not show any significant difference in sensorimotor testing including traverse beam, rotarod, and locomotor activity compared to controls. Significant cognitive benefits were observed with the novel object recognition test (ORT) in the immunized mice (two-tailed, t-test p = 0.0019). Immunized mice also showed cognitive benefits on the closed field symmetrical maze (day 1 two-tailed t-test p = 0.0001; day 2 two-tailed t-test p = 0.0015; day 3 two-tailed t-test p = 0.0002). Reduction of tau pathology was observed with PHF-1 immunohistochemistry in the piriform cortex by 60% (two-tailed t-test p = 0.01) and in the dentate gyrus by 50% (two-tailed t-test p = 0.02) in animals treated with TW1 compared to controls. There were no significant differences in astrogliosis or microgliosis observed between treated and control mice. As assessed by Western blots using PHF-1, the TW1 therapy reduced phosphorylated tau pathology (two-tailed t-test p = 0.03) and improved the ratio of pathological soluble tau to tubulin (PHF1/tubulin; two-tailed t-test p = 0.0006). Reduction of tau pathology also was observed using the CP13 antibody (two-tailed t-test p = 0.0007). These results indicate that passive immunization with the TW1 antibody can significantly decrease tau pathology as assessed by immunohistochemical and biochemical methods, resulting in improved cognitive function in a tau transgenic mouse model of AD.
Collapse
Affiliation(s)
- Allal Boutajangout
- Center for Cognitive Neurology, New York University Langone Health, New York, NY, United States.,Department of Neurology, New York University Langone Health, New York, NY, United States.,Department of Pathology, New York University Langone Health, New York, NY, United States.,Department of Physiology and Neuroscience, New York University Langone Health, New York, NY, United States
| | - Wei Zhang
- Key Laboratory of Brain Functional Genomics (Ministry of Education) Shanghai, School of Life Sciences, East China Normal University, Shanghai, China
| | - Justin Kim
- Center for Cognitive Neurology, New York University Langone Health, New York, NY, United States.,Department of Neurology, New York University Langone Health, New York, NY, United States
| | - Wed Ali Abdali
- Center for Cognitive Neurology, New York University Langone Health, New York, NY, United States.,Department of Neurology, New York University Langone Health, New York, NY, United States
| | - Frances Prelli
- Center for Cognitive Neurology, New York University Langone Health, New York, NY, United States.,Department of Neurology, New York University Langone Health, New York, NY, United States
| | - Thomas Wisniewski
- Center for Cognitive Neurology, New York University Langone Health, New York, NY, United States.,Department of Neurology, New York University Langone Health, New York, NY, United States.,Department of Pathology, New York University Langone Health, New York, NY, United States.,Department of Psychiatry, New York University Langone Health, New York, NY, United States
| |
Collapse
|
18
|
Zaretsky DV, Zaretskaia MV. Flow cytometry method to quantify the formation of beta-amyloid membrane ion channels. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1863:183506. [PMID: 33171157 DOI: 10.1016/j.bbamem.2020.183506] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 10/31/2020] [Accepted: 11/05/2020] [Indexed: 11/17/2022]
Abstract
It is accepted that the cytotoxicity of beta-amyloid is mediated by its oligomers. Amyloid peptides can form ion channels in cell membranes and allow calcium and other ions to enter cells. In this project, we developed a technique to quantify the appearance of calcium in liposomes and applied this technique to study the effect of amyloid peptides on the permeability of membranes. Calcium influx was monitored in liposomes made of phosphatidylcholine (PC) or phosphatidylserine (PS) with an addition of a lipid-soluble dye DiD and containing fluorescent calcium-sensitive probe Fluo-3. The intensity of fluorescence of individual liposomes was measured using a flow cytometer. Calcium ionophore ionomycin served as a positive control. The addition of micromolar concentrations of short fragments of amyloid-beta (Aβ25-35) permeabilized a significant number of PS liposomes. This effect was not observed in PC liposomes. Our data supports the hypothesis that the ion channel formation by amyloid peptide is dependent on electrostatic interactions. High concentrations of Aβ25-35 (above 20 μM) increased signal intensity in a recording channel corresponding to the calcium-sensing probe. However, this phenomenon was also observed in Ca2+-free conditions and even in liposomes without Fluo-3, so we interpreted it as an artifact. Using the described technique, we were not able to detect the formation of calcium channels by several other amyloid peptides. Considering that liposomes appeared resistant to reasonable concentrations of solvents, we expect that described flowmetric technique can be used in high-throughput screening applications.
Collapse
|