1
|
Nagori K, Pradhan M, Nakhate KT. Ethyl gallate ameliorates diabetes-induced Alzheimer's disease-like phenotype in rats via activation of α7 nicotinic receptors and mitigation of oxidative stress. Biochem Biophys Res Commun 2024; 737:150925. [PMID: 39492127 DOI: 10.1016/j.bbrc.2024.150925] [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: 07/28/2024] [Revised: 09/30/2024] [Accepted: 10/28/2024] [Indexed: 11/05/2024]
Abstract
Cognitive decline, an important comorbidity of type 2 diabetes (T2D), is attributed to oxidative stress and impaired cholinergic signaling in the brain. The α7 nicotinic acetylcholine receptor (α7nAChR) is densely distributed in the hippocampus and cortex, and exerts neuroprotective and procognitive actions. Ethyl gallate (EG), a natural phenolic antioxidant compound, showed high in-silico binding affinity towards α7nAChR and brain penetrability. Therefore, the present study aimed to evaluate the involvement of α7nAChR in the potential of EG to ameliorate T2D-induced Alzheimer's disease-like condition. T2D was induced by intraperitoneal (i.p.) injection of streptozotocin (35 mg/kg) in rats on high-fat diet. Diabetic animals were treated with EG (10 and 20 mg/kg, i.p.) for four weeks, and their learning and memory performance was evaluated by the Morris water maze (MWM). Further, the brains were subjected to biochemical analysis of antioxidants like glutathione (GSH), superoxide dismutase (SOD), and catalase (CAT), and oxidative stress marker malonaldehyde (MDA). While diabetic rats showed a significant decline in cognitive performance in the MWM, a substantial improvement was noticed following EG treatment. Further, the diabetes-associated reductions in GSH, SOD, and CAT levels, along with increased MDA contents in the brain, were effectively restored by EG. Interestingly, pre-treatment with α7nAChR antagonist methyllycaconitine (1 mg/kg, i.p.) attenuated the effects of EG on behavioral and biochemical parameters. The results suggest that EG may augment cholinergic signaling in the brain via α7nAChR to mitigate oxidative stress, consequently alleviating T2D-associated dementia. Therefore, EG could be a potential candidate for addressing cognitive impairment comorbid with T2D.
Collapse
Affiliation(s)
- Kushagra Nagori
- Department of Pharmaceutical Chemistry, Rungta College of Pharmaceutical Sciences and Research, Bhilai, 490024, Chhattisgarh, India
| | - Madhulika Pradhan
- Department of Pharmaceutical Technology, Gracious College of Pharmacy, Abhanpur, 493661, Chhattisgarh, India
| | - Kartik T Nakhate
- Department of Pharmacology, Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, 424001, Maharashtra, India.
| |
Collapse
|
2
|
Kaboutari M, Asle-Rousta M, Mahmazi S. Protective effect of menthol against thioacetamide-induced hepatic encephalopathy by suppressing oxidative stress and inflammation, augmenting expression of BDNF and α7-nACh receptor, and improving spatial memory. Eur J Pharmacol 2024; 981:176916. [PMID: 39154831 DOI: 10.1016/j.ejphar.2024.176916] [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: 06/07/2024] [Revised: 08/02/2024] [Accepted: 08/15/2024] [Indexed: 08/20/2024]
Abstract
Hepatic encephalopathy (HE) is a neuropsychiatric syndrome that can occur in people with acute or chronic liver disease. Here, we investigated the effects of menthol, a natural monoterpene, on HE induced by thioacetamide (TA) in male Wistar rats. The rats received 200 mg/kg of TA twice a week for four weeks and were administered 10 mg/kg of menthol intraperitoneally daily for the same period. The results showed that menthol treatment reduced oxidative stress and inflammation in the livers and hippocampi of the rats that received TA. It also lowered the levels of ammonium and liver enzymes AST, ALT, ALP, and GGT in the serum of these animals and prevented liver histopathological damage. In addition, the expression and activity of acetylcholinesterase in the hippocampus of HE model rats were decreased by menthol. Likewise, this monoterpene reduced the expression of TLR4, MyD88, and NF-κB in the hippocampus while increasing the expression of BDNF and α7-nACh receptor. Menthol also reduced neuronal death in the hippocampal cornu ammonis-1 and dentate gyrus regions and reduced astrocyte swelling, which led to improved learning and spatial memory in rats with HE. In conclusion, the study suggests that menthol may have strong protective effects on the liver and brain, making it a potential treatment for HE and neurodegenerative diseases.
Collapse
Affiliation(s)
- Masoud Kaboutari
- Department of Genetics, Zanjan Branch, Islamic Azad University, Zanjan, Iran
| | | | - Sanaz Mahmazi
- Department of Genetics, Zanjan Branch, Islamic Azad University, Zanjan, Iran
| |
Collapse
|
3
|
DeMercy HM, Brenner CA. The Relationship Between Antipsychotics, Cognitive Enhancers, and Major Adverse Cardiovascular/Cerebrovascular Events (MACCE) in Older Adults with Behavioral and Psychological Symptoms of Dementia. Drugs Aging 2024; 41:847-858. [PMID: 39120787 PMCID: PMC11480141 DOI: 10.1007/s40266-024-01134-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/15/2024] [Indexed: 08/10/2024]
Abstract
BACKGROUND AND OBJECTIVES Antipsychotics and cognitive enhancers are often used to treat psychosis and behavioral disturbances in individuals with dementia; however, these drugs have been linked with various adverse events including both metabolic and cerebro/cardiovascular events. Thus, this study sought to estimate the risk of major adverse cardiovascular/cerebrovascular events (MACCE) across four behavioral and psychological symptoms of dementia (BPSD) treatment models by exploring potential associations between antipsychotics (APs), cognitive-enhancing medications, dosage, and earlier MACCE onset. METHODS Patients were obtained from the Loma Linda University Medical Center database who were age ≥ 50 or older and who were diagnosed with dementia and BPSD symptoms. Treatment group and drug dosing were analyzed using Cox regression analyses to predict time until MACCE onset. Patient age at dementia diagnosis, sex, smoking status, race/ethnicity, and previous MACCE diagnoses were included as covariate variables. RESULTS The final study population consisted of 1162 individuals. Results indicated a significant effect of medication type on duration until MACCE, (p < 0.001), with the odds of experiencing a MACCE being 96.3% higher for individuals treated with both APs and cognitive enhancers (p < 0.001). There was also a significant effect of AP dosage on duration until MACCE (p < 0.001) and a significant effect of cognitive enhancer dosage on duration until a MACCE, (p < 0.001). The odds of experiencing a MACCE sooner were 238% higher for those on high doses of APs (p < 0.001) and 76% higher for individuals on high doses of cognitive enhancers (p < 0.010). CONCLUSION The use of APs at high doses was associated with the greatest risk of an adverse medical outcome in older adults with dementia with concurrent behavioral symptoms. Use of AP medications in this population should include close monitoring for cardiovascular/cerebrovascular events.
Collapse
Affiliation(s)
- Haylie M DeMercy
- Department of Psychology, Loma Linda University, 11130 Anderson Street, Loma Linda, CA, 92350, USA
| | - Colleen A Brenner
- Department of Psychology, Loma Linda University, 11130 Anderson Street, Loma Linda, CA, 92350, USA.
| |
Collapse
|
4
|
Sahoo M, Thakor JC, Kumar P, Singh R, Kumar P, Singh K, Puvvala B, Kumar A, Gopinathan A, Palai S, Patra S, Tripathy JP, Acharya R, Sahoo NR, Behera P. AFB1 induced free radicals cause encephalopathy in goat kids via intrinsic pathway of apoptosis: pathological and immunohistochemical confirmation of non-hepatic neuroaflatoxicosis. Vet Res Commun 2024; 48:317-327. [PMID: 37684400 DOI: 10.1007/s11259-023-10216-9] [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: 05/19/2023] [Accepted: 09/03/2023] [Indexed: 09/10/2023]
Abstract
Aflatoxins, particularly AFB1, are the most common feed contaminants worldwide, causing significant economic losses to the livestock sector. The current paper describes an outbreak of aflatoxicosis in a herd of 160 male young goat kids (3-4 months), of which 68 young kids succumbed over a period of 25 days after showing neurological signs of abnormal gait, progressive paralysis and head pressing. The haematobiochemical investigation showed reduced haemoglobin, leucocyte count, PCV level, increased levels of AST, ALT, glucose, BUN, creatinine and reduced level of total protein. Grossly, kids had pale mucous membranes, pale and swollen liver; right apical lobe consolidation, and petechiation of the synovial membrane of the hock joints. The microscopic changes were characterized by multifocal hemorrhages, status spongiosus/ vacuolation, vasculitis, focal to diffuse gliosis, satellitosis, and ischemic apoptotic neurons in different parts of the brain and spinal cord. These changes corresponded well with strong immunoreactivity for AFB1 in neurons, glia cells (oligodendrocytes, astrocytes, and ependymal cells) in various anatomical sites of the brain. The higher values of LPO and reduced levels of antioxidant enzymes (Catalase, SOD, GSH) with strong immunoreactivity of 8-OHdG in the brain indicating high level of oxidative stress. Further, the higher immunosignaling of caspase-3 and caspase-9 in the brain points towards the association with intrinsic pathway of apoptosis. The toxicological analysis of feed samples detected high amounts of AFB1 (0.38ppm). These findings suggest that AFB1 in younger goat kids has more of neurotoxic effect mediated through caspase dependent intrinsic pathway.
Collapse
Affiliation(s)
- Monalisa Sahoo
- Division of Pathology, ICAR-Indian Veterinary Research Institute (IVRI), Izatnagar, 243112, India.
- ICAR-ICFMD-National Institute on Foot and Mouth Disease, Arugul, Jatni, Bhubaneswar, Odisha, 752050, India.
| | - Jigarji Chaturji Thakor
- Division of Pathology, ICAR-Indian Veterinary Research Institute (IVRI), Izatnagar, 243112, India
| | - Pradeep Kumar
- Division of Pathology, ICAR-Indian Veterinary Research Institute (IVRI), Izatnagar, 243112, India
| | - Rajendra Singh
- Division of Pathology, ICAR-Indian Veterinary Research Institute (IVRI), Izatnagar, 243112, India
| | - Pawan Kumar
- Division of Pathology, ICAR-Indian Veterinary Research Institute (IVRI), Izatnagar, 243112, India
| | - Karampal Singh
- ICAR-CADRAD, Indian Veterinary Research Institute (IVRI), Izatnagar, India
| | - Bhavani Puvvala
- Division of Bacteriology & Mycology, ICAR-Indian Veterinary Research Institute (IVRI), Izatnagar, India
| | - Ajay Kumar
- ICAR- Division of Biochemistry, Indian Veterinary Research Institute (IVRI), Izatnagar, India
| | - Aswathy Gopinathan
- ICAR- Division of Surgery, Indian Veterinary Research Institute (IVRI), Izatnagar, India
| | - Santwana Palai
- ICAR- Department of Veterinary Pharmacology & Toxicology, College of Veterinary Science and Animal Husbandry, OUAT, Bhubaneswar, India
| | - Sushmita Patra
- Advance Centre for Treatment, Research & Education in Cancer, Tata Memorial Centre, Navi Mumbai, India
| | - Jagannath Prasad Tripathy
- ICAR-ICFMD-National Institute on Foot and Mouth Disease, Arugul, Jatni, Bhubaneswar, Odisha, 752050, India
| | - Ramakant Acharya
- ICAR-ICFMD-National Institute on Foot and Mouth Disease, Arugul, Jatni, Bhubaneswar, Odisha, 752050, India
| | - Nihar Ranjan Sahoo
- ICAR-ICFMD-National Institute on Foot and Mouth Disease, Arugul, Jatni, Bhubaneswar, Odisha, 752050, India
| | - Pratima Behera
- Animal Disease Research Institute, Phulnakhara, Cuttack, Odisha, India
| |
Collapse
|
5
|
Chu KT, Lei WC, Wu MH, Fuh JL, Wang SJ, French IT, Chang WS, Chang CF, Huang NE, Liang WK, Juan CH. A holo-spectral EEG analysis provides an early detection of cognitive decline and predicts the progression to Alzheimer's disease. Front Aging Neurosci 2023; 15:1195424. [PMID: 37674782 PMCID: PMC10477374 DOI: 10.3389/fnagi.2023.1195424] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 07/25/2023] [Indexed: 09/08/2023] Open
Abstract
Aims Our aim was to differentiate patients with mild cognitive impairment (MCI) and Alzheimer's disease (AD) from cognitively normal (CN) individuals and predict the progression from MCI to AD within a 3-year longitudinal follow-up. A newly developed Holo-Hilbert Spectral Analysis (HHSA) was applied to resting state EEG (rsEEG), and features were extracted and subjected to machine learning algorithms. Methods A total of 205 participants were recruited from three hospitals, with CN (n = 51, MMSE > 26), MCI (n = 42, CDR = 0.5, MMSE ≥ 25), AD1 (n = 61, CDR = 1, MMSE < 25), AD2 (n = 35, CDR = 2, MMSE < 16), and AD3 (n = 16, CDR = 3, MMSE < 16). rsEEG was also acquired from all subjects. Seventy-two MCI patients (CDR = 0.5) were longitudinally followed up with two rsEEG recordings within 3 years and further subdivided into an MCI-stable group (MCI-S, n = 36) and an MCI-converted group (MCI-C, n = 36). The HHSA was then applied to the rsEEG data, and features were extracted and subjected to machine-learning algorithms. Results (a) At the group level analysis, the HHSA contrast of MCI and different stages of AD showed augmented amplitude modulation (AM) power of lower-frequency oscillations (LFO; delta and theta bands) with attenuated AM power of higher-frequency oscillations (HFO; beta and gamma bands) compared with cognitively normal elderly controls. The alpha frequency oscillation showed augmented AM power across MCI to AD1 with a reverse trend at AD2. (b) At the individual level of cross-sectional analysis, implementation of machine learning algorithms discriminated between groups with good sensitivity (Sen) and specificity (Spec) as follows: CN elderly vs. MCI: 0.82 (Sen)/0.80 (Spec), CN vs. AD1: 0.94 (Sen)/0.80 (Spec), CN vs. AD2: 0.93 (Sen)/0.90 (Spec), and CN vs. AD3: 0.75 (Sen)/1.00 (Spec). (c) In the longitudinal MCI follow-up, the initial contrasted HHSA between MCI-S and MCI-C groups showed significantly attenuated AM power of alpha and beta band oscillations. (d) At the individual level analysis of longitudinal MCI groups, deploying machine learning algorithms with the best seven features resulted in a sensitivity of 0.9 by the support vector machine (SVM) classifier, with a specificity of 0.8 yielded by the decision tree classifier. Conclusion Integrating HHSA into EEG signals and machine learning algorithms can differentiate between CN and MCI as well as also predict AD progression at the MCI stage.
Collapse
Affiliation(s)
- Kwo-Ta Chu
- Institute of Cognitive Neuroscience, National Central University, Taoyuan, Taiwan
- Yang-Ming Hospital, Taoyuan, Taiwan
| | - Weng-Chi Lei
- Institute of Cognitive Neuroscience, National Central University, Taoyuan, Taiwan
- Cognitive Intelligence and Precision Healthcare Center, National Central University, Taoyuan, Taiwan
| | - Ming-Hsiu Wu
- Division of Neurology, Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan
- Department of Long-Term Care and Health Promotion, Min-Hwei Junior College of Health Care Management, Tainan, Taiwan
| | - Jong-Ling Fuh
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Shuu-Jiun Wang
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Isobel T. French
- Institute of Cognitive Neuroscience, National Central University, Taoyuan, Taiwan
- Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Central University and Academia Sinica, Taipei, Taiwan
| | - Wen-Sheng Chang
- Institute of Cognitive Neuroscience, National Central University, Taoyuan, Taiwan
| | - Chi-Fu Chang
- Institute of Cognitive Neuroscience, National Central University, Taoyuan, Taiwan
| | - Norden E. Huang
- Cognitive Intelligence and Precision Healthcare Center, National Central University, Taoyuan, Taiwan
- Key Laboratory of Data Analysis and Applications, First Institute of Oceanography, SOA, Qingdao, China
| | - Wei-Kuang Liang
- Institute of Cognitive Neuroscience, National Central University, Taoyuan, Taiwan
- Cognitive Intelligence and Precision Healthcare Center, National Central University, Taoyuan, Taiwan
| | - Chi-Hung Juan
- Institute of Cognitive Neuroscience, National Central University, Taoyuan, Taiwan
- Cognitive Intelligence and Precision Healthcare Center, National Central University, Taoyuan, Taiwan
- Department of Psychology, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
6
|
Sabbir MG, Swanson M, Albensi BC. Loss of cholinergic receptor muscarinic 1 impairs cortical mitochondrial structure and function: implications in Alzheimer's disease. Front Cell Dev Biol 2023; 11:1158604. [PMID: 37274741 PMCID: PMC10233041 DOI: 10.3389/fcell.2023.1158604] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 05/04/2023] [Indexed: 06/06/2023] Open
Abstract
Introduction: Cholinergic Receptor Muscarinic 1 (CHRM1) is a G protein-coupled acetylcholine (ACh) receptor predominantly expressed in the cerebral cortex. In a retrospective postmortem brain tissues-based study, we demonstrated that severely (≥50% decrease) reduced CHRM1 proteins in the temporal cortex of Alzheimer's patients significantly correlated with poor patient outcomes. The G protein-mediated CHRM1 signal transduction cannot sufficiently explain the mechanistic link between cortical CHRM1 loss and the appearance of hallmark Alzheimer's pathophysiologies, particularly mitochondrial structural and functional abnormalities. Therefore, the objective of this study was to analyze the molecular, ultrastructural, and functional properties of cortical mitochondria using CHRM1 knockout (Chrm1-/-) and wild-type mice to identify mitochondrial abnormalities. Methods: Isolated and enriched cortical mitochondrial fractions derived from wild-type and Chrm1-/- mice were assessed for respiratory deficits (oxygen consumption) following the addition of different substrates. The supramolecular assembly of mitochondrial oxidative phosphorylation (OXPHOS)-associated protein complexes (complex I-V) and cortical mitochondrial ultrastructure were investigated by blue native polyacrylamide gel electrophoresis and transmission electron microscopy (TEM), respectively. A cocktail of antibodies, specific to Ndufb8, Sdhb, Uqcrc2, Mtco1, and Atp5a proteins representing different subunits of complexes I-V, respectively was used to characterize different OXPHOS-associated protein complexes. Results: Loss of Chrm1 led to a significant reduction in cortical mitochondrial respiration (oxygen consumption) concomitantly associated with reduced oligomerization of ATP synthase (complex V) and supramolecular assembly of complexes I-IV (Respirasome). Overexpression of Chrm1 in transformed cells (lacking native Chrm1) significantly increased complex V oligomerization and respirasome assembly leading to enhanced respiration. TEM analysis revealed that Chrm1 loss led to mitochondrial ultrastructural defects and alteration in the tinctorial properties of cortical neurons causing a significant increase in the abundance of dark cortical neurons (Chrm1-/- 85% versus wild-type 2%). Discussion: Our findings indicate a hitherto unknown effect of Chrm1 deletion in cortical neurons affecting mitochondrial function by altering multiple interdependent factors including ATP synthase oligomerization, respirasome assembly, and mitochondrial ultrastructure. The appearance of dark neurons in Chrm1-/- cortices implies potentially enhanced glutamatergic signaling in pyramidal neurons under Chrm1 loss condition. The findings provide novel mechanistic insights into Chrm1 loss with the appearance of mitochondrial pathophysiological deficits in Alzheimer's disease.
Collapse
Affiliation(s)
- Mohammad Golam Sabbir
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada
- Alzo Biosciences Inc, SanDiego, CA, United States
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada
- Barry & Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Mamiko Swanson
- Alzo Biosciences Inc, SanDiego, CA, United States
- Barry & Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Benedict C. Albensi
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada
- Barry & Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, United States
- Department of Pharmacology & Therapeutics, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
7
|
Wu J, Zhang H, Wang Y, Yin G, Li Q, Zhuo L, Chen H, Wang Z. From tryptamine to the discovery of efficient multi-target directed ligands against cholinesterase-associated neurodegenerative disorders. Front Pharmacol 2022; 13:1036030. [PMID: 36518670 PMCID: PMC9742383 DOI: 10.3389/fphar.2022.1036030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 11/03/2022] [Indexed: 11/26/2023] Open
Abstract
A novel class of benzyl-free and benzyl-substituted carbamylated tryptamine derivatives (CDTs) was designed and synthesized to serve as effective building blocks for the development of novel multi-target directed ligands (MTDLs) for the treatment of neurological disorders linked to cholinesterase (ChE) activity. The majority of them endowed butyrylcholinesterase (BuChE) with more substantial inhibition potency than acetylcholinesterase (AChE), according to the full study of ChE inhibition. Particularly, hybrids with dibenzyl groups (2b-2f, 2j, 2o, and 2q) showed weak or no neuronal toxicity and hepatotoxicity and single-digit nanomolar inhibitory effects against BuChE. Through molecular docking and kinetic analyses, the potential mechanism of action on BuChE was first investigated. In vitro H2O2-induced HT-22 cells assay demonstrated the favorable neuroprotective potency of 2g, 2h, 2j, 2m, 2o, and 2p. Besides, 2g, 2h, 2j, 2m, 2o, and 2p endowed good antioxidant activities and COX-2 inhibitory effects. This study suggested that this series of hybrids can be applied to treat various ChE-associated neurodegenerative disorders such as Alzheimer's disease (AD) and Parkinson's disease (PD), as well as promising building blocks for further structure modification to develop efficient MTDLs.
Collapse
Affiliation(s)
- Junbo Wu
- Department of Colorectal Surgery, The Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Honghua Zhang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yuying Wang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China
| | - Gaofeng Yin
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Qien Li
- Tibetan Medical College, Qinghai University, Xining, Qinghai, China
| | - Linsheng Zhuo
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Hongjin Chen
- Department of Colorectal Surgery, The Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Zhen Wang
- Department of Colorectal Surgery, The Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
8
|
Abdelnour C, Ferreira D, van de Beek M, Cedres N, Oppedal K, Cavallin L, Blanc F, Bousiges O, Wahlund LO, Pilotto A, Padovani A, Boada M, Pagonabarraga J, Kulisevsky J, Aarsland D, Lemstra AW, Westman E. Parsing heterogeneity within dementia with Lewy bodies using clustering of biological, clinical, and demographic data. Alzheimers Res Ther 2022; 14:14. [PMID: 35063023 PMCID: PMC8783432 DOI: 10.1186/s13195-021-00946-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 12/06/2021] [Indexed: 11/10/2022]
Abstract
BACKGROUND Dementia with Lewy bodies (DLB) includes various core clinical features that result in different phenotypes. In addition, Alzheimer's disease (AD) and cerebrovascular pathologies are common in DLB. All this increases the heterogeneity within DLB and hampers clinical diagnosis. We addressed this heterogeneity by investigating subgroups of patients with similar biological, clinical, and demographic features. METHODS We studied 107 extensively phenotyped DLB patients from the European DLB consortium. Factorial analysis of mixed data (FAMD) was used to identify dimensions in the data, based on sex, age, years of education, disease duration, Mini-Mental State Examination (MMSE), cerebrospinal fluid (CSF) levels of AD biomarkers, core features of DLB, and regional brain atrophy. Subsequently, hierarchical clustering analysis was used to subgroup individuals based on the FAMD dimensions. RESULTS We identified 3 dimensions using FAMD that explained 38% of the variance. Subsequent hierarchical clustering identified 4 clusters. Cluster 1 was characterized by amyloid-β and cerebrovascular pathologies, medial temporal atrophy, and cognitive fluctuations. Cluster 2 had posterior atrophy and showed the lowest frequency of visual hallucinations and cognitive fluctuations and the worst cognitive performance. Cluster 3 had the highest frequency of tau pathology, showed posterior atrophy, and had a low frequency of parkinsonism. Cluster 4 had virtually normal AD biomarkers, the least regional brain atrophy and cerebrovascular pathology, and the highest MMSE scores. CONCLUSIONS This study demonstrates that there are subgroups of DLB patients with different biological, clinical, and demographic characteristics. These findings may have implications in the diagnosis and prognosis of DLB, as well as in the treatment response in clinical trials.
Collapse
Affiliation(s)
- Carla Abdelnour
- Research Center and Memory Clinic, Ace Alzheimer Center Barcelona, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya-Barcelona, Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain.
- Department of Medicine of the Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - Daniel Ferreira
- Division of Clinical Geriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Stockholm, Sweden
| | - Marleen van de Beek
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Nira Cedres
- Division of Clinical Geriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Stockholm, Sweden
- Department of Psychology, Sensory Cognitive Interaction Laboratory (SCI-lab), Stockholm University, Stockholm, Sweden
| | - Ketil Oppedal
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
- Department of Radiology, Stavanger University Hospital, Stavanger, Norway
- Department of Electrical Engineering and Computer Science, University of Stavanger, Stavanger, Norway
| | - Lena Cavallin
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Radiology Karolinska University Hospital, Stockholm, Sweden
| | - Frédéric Blanc
- Service, Memory Resources and Research Centre, University Hospital of Strasbourg, Strasbourg, France
- Team IMIS/Neurocrypto, French National Center for Scientific Research, ICube Laboratory and Fédération de Médecine Translationnelle de Strasbourg (FMTS), University of Strasbourg, Strasbourg, France
- Centre Mémoire, de Ressources et de Recherche d'Alsace (Strasbourg-Colmar), Strasbourg, France
| | - Olivier Bousiges
- Centre Mémoire, de Ressources et de Recherche d'Alsace (Strasbourg-Colmar), Strasbourg, France
- Laboratory of Biochemistry and Molecular Biology, CNRS, Laboratoire de Neurosciences Cognitives et Adaptatives, UMR7364, University Hospital of Strasbourg, Strasbourg, France
| | - Lars-Olof Wahlund
- Division of Clinical Geriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Stockholm, Sweden
| | - Andrea Pilotto
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Alessandro Padovani
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Mercè Boada
- Research Center and Memory Clinic, Ace Alzheimer Center Barcelona, Institut Català de Neurociències Aplicades, Universitat Internacional de Catalunya-Barcelona, Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Javier Pagonabarraga
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau. Biomedical Research Institute (IIB-Sant Pau), Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Jaime Kulisevsky
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau. Biomedical Research Institute (IIB-Sant Pau), Centro de Investigación en Red-Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Dag Aarsland
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Afina W Lemstra
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Eric Westman
- Division of Clinical Geriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Stockholm, Sweden
- Department of Neuroimaging, Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| |
Collapse
|
9
|
Labban S, Alghamdi BS, Alshehri FS, Kurdi M. Effects of melatonin and resveratrol on recognition memory and passive avoidance performance in a mouse model of Alzheimer's disease. Behav Brain Res 2021; 402:113100. [PMID: 33417994 DOI: 10.1016/j.bbr.2020.113100] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/12/2020] [Accepted: 12/27/2020] [Indexed: 12/25/2022]
Abstract
Alzheimer's disease (AD) is the foremost cause of dementia among other neurodegenerative diseases, leading to memory loss and cognitive deficits. AD has gained extensive attention in research for exploring possible interventions. One promising field is natural substances and compounds that could provide a wide range of neuroprotection against AD. This study aimed to investigate the possible effects of melatonin (MEL) and resveratrol (RES) in improving memory deficits in a sporadic mouse model of AD. Memory deficit was induced using AlCl3 and d-galactose for generating an AD mouse model. Mice were randomly distributed into five groups (n = 13): control, AD, AD + MEL (AD mice treated with 80 mg/kg of MEL), AD + RES (AD mice treated with 40 mg/kg of RES), and AD + Combination)AD mice that received 80 mg/kg MEL and 40 mg/kg RES). A novel object recognition task (NORT) and passive avoidance task (PAT) were used for assessing memory. Moreover, acetylcholinesterase (AChE) level, brain-derived neurotrophic factor (BDNF), and cAMP-response element binding (CREB) protein expression were measured in the prefrontal cortex tissue. Our results showed that MEL significantly improved memory deficits in both the NORT and PAT of the AD model, while RES improved the PAT only in the AD model. Co-treatment with MEL and RES exerted beneficial additive effects on recognition memory impairment in the AD mouse model. Moreover, our results demonstrated that both MEL and RES enhanced the cholinergic system and BDNF and CREB signaling pathways in the prefrontal cortex in an AD mouse model.
Collapse
Affiliation(s)
- Samah Labban
- Department of Physiology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Physiology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia.
| | - Badrah S Alghamdi
- Department of Physiology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia; Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Fahad S Alshehri
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia.
| | - Maher Kurdi
- Department of Pathology, Faculty of Medicine, King Abdulaziz University, Rabigh, Saudi Arabia.
| |
Collapse
|
10
|
Shen H, Meng Y, Liu D, Qin Z, Huang H, Pan L, Wang W, Kang J. α7 Nicotinic Acetylcholine Receptor Agonist PNU-282987 Ameliorates Cognitive Impairment Induced by Chronic Intermittent Hypoxia. Nat Sci Sleep 2021; 13:579-590. [PMID: 34007230 PMCID: PMC8123952 DOI: 10.2147/nss.s296701] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 04/13/2021] [Indexed: 12/18/2022] Open
Abstract
PURPOSE Cognitive impairment is an important complication of obstructive sleep apnea (OSA). Chronic intermittent hypoxia (CIH), the main pathophysiological characteristics of OSA, is closely related to cognitive dysfunction and may be mediated by alpha-7 nicotinic acetylcholine receptors (α7nAChR). This study investigated the effects and clarified the mechanisms of α7nAChR on the cognitive function of mice with CIH. METHODS Thirty CD-1 mice were randomly divided into room air (RA), CIH-2 weeks (CIH2W), and CIH-4 weeks (CIH4W) groups. Cognitive function was evaluated by novel object recognition (NOR) and Morris water maze (MWM) tests after exposure. Then, 104 CD-1 mice were exposed to CIH for 4 weeks and randomly divided into four groups: CIH4W (control), with dimethyl sulfoxide (DMSO) (sham), with α7nAChR-specific agonist PNU-282987 (PNU), and with α7nAChR-specific inhibitor methyllycaconitine and PNU-282987 (MLA+PNU). In addition to the evaluation of cognitive function, apoptotic bodies in the hippocampus were detected by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining, changes in p-CREB and BDNF were detected by immunohistochemistry, while those of ERK1/2, CREB, PGC-1α, FNDC5, and BDNF were detected by Western blotting in the hippocampal tissues of the mice. RESULTS Compared to the CIH2W and RA groups, the CIH4W group showed cognitive dysfunction in the NOR and MWM tests. The changes in cognitive dysfunction were alleviated by PNU-282987; furthermore, MLA pretreatment offset the effect. In hippocampal tissues, TUNEL assays showed decreased apoptotic cells, immunohistochemical staining showed increased expressions of p-CREB and BDNF. The expression levels of p-ERK1/2/t-ERK1/2, p-CREB/t-CREB, PGC-1α, FNDC5, and BDNF were increased after PNU-282987 injection. CONCLUSION Four weeks of CIH caused cognitive dysfunction in mice. Activating α7nAChR might ameliorate this dysfunction by upregulating the ERK1/2/CREB signaling pathway; enhancing PGC-1α, FNDC5, and BDNF expression levels; and reducing cell apoptosis in the hippocampal tissue of mice.
Collapse
Affiliation(s)
- Hui Shen
- Institute of Respiratory Disease, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Yanling Meng
- Institute of Respiratory Disease, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Dan Liu
- Institute of Respiratory Disease, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Zheng Qin
- Institute of Respiratory Disease, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Hong Huang
- Institute of Respiratory Disease, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Lei Pan
- Institute of Respiratory Disease, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Wei Wang
- Institute of Respiratory Disease, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Jian Kang
- Institute of Respiratory Disease, The First Hospital of China Medical University, Shenyang, People's Republic of China
| |
Collapse
|